Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 69
Filter
1.
Mol Pharm ; 20(5): 2513-2526, 2023 05 01.
Article in English | MEDLINE | ID: mdl-37071030

ABSTRACT

Docetaxel (DTX), a semisynthetic analogue of paclitaxel, is often used to treat cancers. Owing to its poor aqueous solubility, the current formulation of DTX for clinical applications involves using high surfactant and ethanol concentrations, causing hypersensitivity reactions. To overcome this issue, we developed a reduction-responsive DTX prodrug encapsulated within human serum albumin (HSA) nanoparticles (DTX-SS-COOH/HSA NPs). First, the DTX prodrug was conjugated to undecanoic acid through a disulfide bond (DTX-SS-COOH) via a four-step reaction. Subsequently, DTX-SS-COOH/HSA NPs were prepared via the desolvation method. The NPs exhibited a spherical structure with a diameter range of 140-220 nm, as revealed by dynamic light scattering and transmission electron microscopy. Fluorescence quenching analysis confirmed the formation of DTX-SS-COOH/HSA, which was ascribed to electrostatic interactions and hydrophobic forces. Notably, NPs with a feed mole ratio corresponding to DTX-SS-COOH/HSA = 9:1 demonstrated high drug-loading and encapsulation efficiency of 12.84 and 93.11%, respectively, alongside good stability. Moreover, the reduced responsiveness experiment revealed an accelerated DTX release in the presence of glutathione. An in vivo pharmacokinetic study indicated that DTX-SS-COOH/HSA NPs demonstrated considerably a prolonged circulation time (6.2-fold) compared to that of free DTX. Ultimately, the antitumor test of MDA-MB-231 tumor-bearing mice revealed that DTX-SS-COOH/HSA NPs were superior to DTX/HSA NPs for tumor growth inhibition. Thus, DTX-SS-COOH/HSA NPs represent a promising DTX nanoformulation for clinical application.


Subject(s)
Antineoplastic Agents , Nanoparticles , Neoplasms , Prodrugs , Humans , Mice , Animals , Docetaxel , Serum Albumin, Human , Paclitaxel , Drug Carriers/chemistry , Nanoparticles/chemistry , Cell Line, Tumor , Antineoplastic Agents/therapeutic use , Antineoplastic Agents/chemistry , Neoplasms/drug therapy
2.
Mol Pharm ; 20(10): 4971-4983, 2023 10 02.
Article in English | MEDLINE | ID: mdl-37699256

ABSTRACT

mRNA vaccines encoding a single spike protein effectively prevent severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection. However, the emergence of SARS-CoV-2 variants leads to a wide range of immune evasion. Herein, a unique trivalent mRNA vaccine based on ancestral SARS-CoV-2, Delta, and Omicron variant spike receptor-binding domain (RBD) mRNAs was developed to tackle the immune evasion of the variants. First, three RBD mRNAs of SARS-CoV-2, Delta, and Omicron were coencapsulated into lipid nanoparticles (LNPs) by using microfluidic technology. After that, the physicochemical properties and time-dependent storage stability of the trivalent mRNA vaccine nanoformulation were tested by using dynamic light scattering (DLS). In vitro, the trivalent mRNA vaccine exhibited better lysosomal escape ability, transfection efficiency, and biocompatibility than did the commercial transfection reagent Lipo3000. In addition, Western blot analyses confirmed that the three RBD proteins can be detected in cells transfected with the trivalent mRNA vaccine. Furthermore, ex vivo imaging analysis indicated that the livers of BALB/c mice had the strongest protein expression levels after intramuscular (IM) injection. Using a prime-boost strategy, this trivalent vaccine elicited robust humoral and T-cell immune responses in both the high-dose and low-dose groups and showed no toxicity in BALB/c mice. Three specific IgG antibodies in the high-dose group against SARS-CoV-2, Delta, and Omicron variants approached ∼1/1,833,333, ∼1/1,866,667, and ∼1/925,000, respectively. Taken together, two doses of inoculation with the trivalent mRNA vaccine may provide broad and effective immunization responses against SARS-CoV-2 and variants.


Subject(s)
COVID-19 , SARS-CoV-2 , Animals , Mice , Humans , SARS-CoV-2/genetics , COVID-19 Vaccines , COVID-19/prevention & control , Immunization , mRNA Vaccines , Antibodies, Neutralizing
3.
Bioorg Med Chem ; 78: 117135, 2023 01 15.
Article in English | MEDLINE | ID: mdl-36577327

ABSTRACT

Vascular endothelial growth factor A (VEGFA) plays an important role in the healing of skin wound. However, the application of VEGFA protein in clinic is limited because of its high cost manufacturing, complicated purification and poor pharmacokinetic profile. Herein, we developed nucleoside-modified mRNA encoding VEGFA encapsulated ionizable lipid nanoparticles (LNP) to improve angiogenesis and increase wound healing rate. First, VEGFA mRNA was synthesized by an in vitro transcription (IVT) method. After that, VEGFA mRNA-LNP was prepared by encapsulating mRNA in ionizable lipid based nanoparticles via a microfluidic mixer. The physicochemical properties of VEGFA mRNA-LNP were investigated via dynamic light scattering (DLS) and transmission electron microscopy (TEM). The results showed that the VEGFA mRNA-LNP possessed regular spherical morphology with an average size of 112.67 nm and a negative Zeta potential of -3.43 mV. The LNP delivery system had excellent lysosome escape capability and high transfection efficiency. ELISA and Western Blot analysis indicated that the mRNA-LNP could express VEGFA protein in Human umbilical vein endothelial cells (HUVECs). Besides, endothelial tube formation, cell proliferation and scratch assays were performed. The results revealed VEGFA mRNA-LNP boosted angiogenesis, cell proliferation and cell migration by expressing VEGFA protein. Finally, C57BL/6 mouse model of skin wound was established and intradermally treated with VEGFA mRNA-LNP. The VEGFA mRNA-LNP treated wounds were almost healed with an average wound size of 1.56 mm2 compared with the blank of 18.66 mm2 after 9 days. The results indicated that the VEGFA mRNA-LNP was able to significantly expedite wound healing. Histological analysis further demonstrated tissue epithelialization, collagen deposition and enhancement of vascular density after treatment. Taken together, VEGFA mRNA-LNP can be uptaken by cells to express protein effectively and promote wound healing, which may provide a promising strategy for clinical remedy.


Subject(s)
Nanoparticles , Vascular Endothelial Growth Factor A , Mice , Animals , Humans , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism , RNA, Messenger/genetics , Mice, Inbred C57BL , Wound Healing , Nanoparticles/chemistry , Human Umbilical Vein Endothelial Cells/metabolism
4.
Med Sci Monit ; 29: e939972, 2023 Jul 30.
Article in English | MEDLINE | ID: mdl-37516906

ABSTRACT

Bone defects and dysfunctions are prevalent among patients, resulting from various causes such as trauma, tumors, congenital malformations, inflammation, and infection. The demand for bone defect repair materials is second only to blood transfusions. Artificial bone composites offer numerous advantages for bone damage repair, including their availability, absence of rejection or immune reactions, high malleability, exceptional mechanical strength, and outstanding biocompatibility. However, bacterial infections frequently occur during bone transplantation or on graft material structures, leading to severe complications such as osteomyelitis and osteoporosis. Moreover, existing osteogenic materials alone are inadequate to address the challenges posed by traumatic infections, presenting a significant hurdle for clinicians in reconstructing infectious bone defects. Consequently, it is crucial to functionalize artificial bone composites to facilitate effective bone repair and regeneration. Notably, antibacterial capabilities play a critical role in preventing and treating infectious bone defects, and current research is focusing on the interface between artificial bone composites and antibacterial treatments. This article provides an extensive review of the current state of artificial composite bone scaffolds with antibacterial properties for infection prevention in bone grafting.


Subject(s)
Arthrodesis , Bone Transplantation , Humans , Osteogenesis , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/therapeutic use , Inflammation
5.
Antimicrob Agents Chemother ; 66(5): e0206521, 2022 05 17.
Article in English | MEDLINE | ID: mdl-35416709

ABSTRACT

Current best practice for the treatment of malaria relies on short half-life artemisinins that are failing against emerging Kelch 13 mutant parasite strains. Here, we introduce a liposome-like self-assembly of a dimeric artesunate glycerophosphocholine conjugate (dAPC-S) as an amphiphilic prodrug for the short-lived antimalarial drug, dihydroartemisinin (DHA), with enhanced killing of Kelch 13 mutant artemisinin-resistant parasites. Cryo-electron microscopy (cryoEM) images and the dynamic light scattering (DLS) technique show that dAPC-S typically exhibits a multilamellar liposomal structure with a size distribution similar to that of the liposomes generated using thin-film dispersion (dAPC-L). Liquid chromatography-mass spectrometry (LCMS) was used to monitor the release of DHA. Sustainable release of DHA from dAPC-S and dAPC-L assemblies increased the effective dose and thus efficacy against Kelch 13 mutant artemisinin-resistant parasites in an in vitro assay. To better understand the enhanced killing effect, we investigated processes for deactivation of both the assemblies and DHA, including the roles of serum components and trace levels of iron. Analysis of parasite proteostasis pathways revealed that dAPC assemblies exert their activity via the same mechanism as DHA. We conclude that this easily prepared multilamellar liposome-like dAPC-S with long-acting efficacy shows potential for the treatment of severe and artemisinin-resistant malaria.


Subject(s)
Antimalarials , Artemisinins , Malaria, Falciparum , Malaria , Antimalarials/pharmacology , Antimalarials/therapeutic use , Artemisinins/pharmacology , Artemisinins/therapeutic use , Artesunate/pharmacology , Artesunate/therapeutic use , Cryoelectron Microscopy , Drug Resistance/genetics , Humans , Liposomes/chemistry , Malaria/drug therapy , Malaria, Falciparum/drug therapy , Malaria, Falciparum/parasitology , Plasmodium falciparum/genetics
6.
Mol Pharm ; 19(12): 4588-4600, 2022 12 05.
Article in English | MEDLINE | ID: mdl-35731922

ABSTRACT

Protein kinase N3 (PKN3), by virtue of its abnormal expression in prostate cells, has been widely used as a target of RNAi (shRNA, siRNA, miRNA) therapy. The major challenges of PKN3 RNAi therapy lie in how to design effective interference sequences and delivery systems. Herein, new PKN3 shRNA sequences (shPKN3-2459 and shPKN3-3357) were designed, and bioreducible, biodegradable, ionizable lipid-based nanoparticles were developed for shPKN3 delivery. First, an ionizable lipid (DDA-SS-DMA) bridged with disulfide bond and ester bonds was synthesized by a three-step reaction and confirmed by MS, 1H NMR, and 13C NMR. The ionizable lipid was mixed with cholesterol, DSPC, PEG-lipid, and shPKN3 by a microfluidic mixer to prepare lipid nanoparticles (LNP-shPKN3) which were characterized by DLS and TEM. Afterward, the pH and glutathione (GSH)-responsiveness of the DDA-SS-DMA based LNP delivery system were investigated by lysosome escape and gel electrophoresis assays. Compared with the commercial transfection reagent Lipo2000, the DDA-SS-DMA based delivery system showed higher transfection efficiency and lower toxicity. Western blot analysis, invasion tests, and migration assays were performed to evaluate the silencing effect of shPKN3 in vitro. In in vivo studies, high tumor suppression (65.8%) and treatment safety were evident in the LNP-shPKN3-2459 treatment group. Taken together, the DDA-SS-DMA based delivery system encapsulating shPKN3-2459 showed significant antitumor efficacy and might be a promising formulation for the treatment of prostate cancer.


Subject(s)
Nanoparticles , Prostatic Neoplasms , Humans , Male , Lipids/chemistry , Nanoparticles/chemistry , Prostate , Prostatic Neoplasms/genetics , Prostatic Neoplasms/therapy , RNA, Small Interfering
7.
Bioorg Med Chem ; 69: 116884, 2022 09 01.
Article in English | MEDLINE | ID: mdl-35752145

ABSTRACT

Protein kinase N3 (PKN3), an AGC-family member, is often overexpressed in breast tumor cells. RNAi therapy is a promising approach to inhibit tumor growth by reducing the expression of PKN3. In this report, lipid nanoparticles encapsulated with new shRNA PKN3 (SS-LNP/shPKN3) with redox-responsiveness were developed in order to specifically down-regulate the expression of PKN3 for breast cancer treatment. The SS-LNP/shPKN3 was prepared by microfluidic method using disulfide bonds based ionizable lipid as main component. The as-prepared SS-LNP/shPKN3 lipid nanoparticles were characterized via using dynamic light scattering (DLS) and transmission electron microscopy (TEM). The results indicated that the obtained SS-LNP/shPKN3 exhibited uniform particle size and regular spherical morphology. Moreover, glutathione (GSH) triggered release of shPKN3 confirmed the redox-responsiveness of the SS-LNP/shPKN3. Finally, the anti-tumor effect of SS-LNP/shPKN3 was evaluated against MDA-MB-231 cells and derived xenograft tumor bearing mice. It was found that the SS-LNP/shPKN3-2 had the highest PKN3 protein inhibition rate of 60.8% and tumor inhibition rate of 62.3%. Taken together, the SS-LNP/shPKN3 might be a potential therapeutic strategy for breast cancer.


Subject(s)
Breast Neoplasms , Nanoparticles , Animals , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Female , Humans , Liposomes , Mice , Nanoparticles/chemistry , Protein Kinase C , RNA, Small Interfering/chemistry
8.
Mol Pharm ; 18(10): 3862-3870, 2021 10 04.
Article in English | MEDLINE | ID: mdl-34470216

ABSTRACT

In this work, dimeric artesunate-phosphatidylcholine conjugate (dARTPC)-based liposomes encapsulated with irinotecan (Ir) were developed for anticancer combination therapy. First, dARTPC featured with unique amphipathic properties formed liposomes by classical thin-film methods. After that, Ir was encapsulated into dARTPC-based liposomes (Ir/dARTPC-LP) by the triethylammonium sucrose octasulfate gradient method. Physicochemical characterization indicated that Ir/dARTPC-LP had a mean size of around 140 nm and a negative ζ potential of approximately -30 mV. Most noticeably, liposomes displayed an encapsulation efficiency of greater than 98% with a controllable drug loading of 4-22%. The in vitro release of dihydroartemisinin (DHA) and Ir from Ir/dARTPC-LP was investigated by dialysis in different media. It was found that effective release of both DHA (65.42%) and Ir (77.28%) in a weakly acidic medium (pH 5.0) after 48 h was achieved in comparison to very slow release under a neutral environment (DHA 9.90% and Ir 8.72%), indicating the controllable release of both drugs. Confocal laser scanning microscopy confirmed the improved cellular internalization of Ir/dARTPC-LP. The cytotoxicity of Ir/dARTPC-LP was evaluated in the MCF-7, A549, and HepG2 cell lines. The results showed that Ir/dARTPC-LP had significant synergistic efficacy in the loss of cell growth. In vivo anticancer evaluation was performed using a 4T1 xenograft tumor model. Ir/dARTPC-LP had a high tumor inhibition rate of 62.7% without significant toxicity in comparison with the injection of Ir solution. Taken together, dARTPC encapsulated with Ir has great potential for anticancer combination therapy.


Subject(s)
Artesunate/administration & dosage , Drug Delivery Systems , Irinotecan/administration & dosage , Liposomes/administration & dosage , Animals , Antineoplastic Combined Chemotherapy Protocols , Apoptosis/drug effects , Artesunate/pharmacokinetics , Artesunate/therapeutic use , Cell Line, Tumor , Drug Combinations , Drug Delivery Systems/methods , Female , Irinotecan/pharmacokinetics , Irinotecan/therapeutic use , Mice, Inbred BALB C , Mice, Nude , Neoplasm Transplantation , Neoplasms, Experimental/drug therapy , Phosphatidylcholines
9.
Mol Pharm ; 17(1): 262-273, 2020 01 06.
Article in English | MEDLINE | ID: mdl-31747284

ABSTRACT

The microtubule inhibitor paclitaxel (PTX) is used to treat a wide range of solid tumors. Due to the poor aqueous solubility of PTX, a continuous demand for safe, efficient PTX formulations with improved antitumor activity exists. Here, we report a novel form of redox-sensitive paclitaxel (PTX)-encapsulated liposomes based on the previously developed disulfide phosphatidylcholine (SS-PC). PTX-loaded stealth liposomes (PTX/SS-LP) composed of SS-PC, 1,2-distearoyl-sn-glycero-3-phosphoethanolamine-PEG2000 (DSPE-PEG2000), and cholesterol were prepared using the reverse-phase evaporation method. The characterization of the PTX/SS-LP liposomes using dynamic light scattering and transmission electron microscopy confirmed their uniform particle size and typical unilamellar vesicle structure with an average bilayer thickness of approximately 4 nm. Changes in the size and morphology as well as the rapid release of PTX triggered by the addition of dithiothreitol revealed the redox sensitivity of PTX/SS-LP. Finally, evaluations in MCF-7 and A549 cells in vitro and in BALB/c mice in vivo revealed the improved anticancer efficiency, biodistribution, and safety of PTX/SS-LP compared with those of Taxol and nonredox-sensitive PTX/LP. In conclusion, PTX/SS-LP displays a redox-responsive release of paclitaxel with improved antitumor activity and has great potential as a next-generation stealth liposomal PTX delivery system.


Subject(s)
Antineoplastic Agents, Phytogenic/administration & dosage , Liposomes/chemistry , Neoplasms/drug therapy , Paclitaxel/administration & dosage , Phosphatidylcholines/chemistry , Animals , Antineoplastic Agents, Phytogenic/pharmacology , Apoptosis/drug effects , Cell Line, Tumor , Cholesterol/chemistry , Dithiothreitol , Drug Liberation , Dynamic Light Scattering , Humans , Liposomes/pharmacology , Liposomes/toxicity , Liposomes/ultrastructure , Mass Spectrometry , Mice , Mice, Inbred BALB C , Microscopy, Electron, Transmission , Oxidation-Reduction/drug effects , Paclitaxel/chemistry , Paclitaxel/pharmacology , Phosphatidylcholines/chemical synthesis , Phosphatidylethanolamines/chemistry , Polyethylene Glycols/chemistry , Tissue Distribution , Xenograft Model Antitumor Assays
10.
Bioorg Med Chem ; 28(19): 115682, 2020 10 01.
Article in English | MEDLINE | ID: mdl-32912428

ABSTRACT

RIWVIWRR-NH2 (Bac8c) is a natural antimicrobial peptide (AMP) exhibiting great antibacterial activity against Gram-negative and Gram-positive bacteria. In this work, lipoic acid was used as a fatty acid hydrophobic ligand to modify Bac8c (LA-Bac8c) to further improve its antimicrobial properties. Minimal inhibitory concentration (MIC) and minimal bactericidal concentration (MBC) assays showed that LA-Bac8c exhibited lower MIC (MBC) values against Staphylococcus aureus (S. aureus) and methicillin-resistant Staphylococcus aureus (MRSA) than Bac8c. Similar results were reflected in the antibiofilm activity towards S. aureus and MRSA, and LA-Bac8c showed better activity to the biofilm which has been formed or is being formed. In addition to this, the obvious interaction between bacteria/biofilm and LA-Bac8c was observed by microscopy. LA-Bac8c displayed strong membrane depolarization and outer membrane permeabilizing ability, and the cell membrane treated with LA-Bac8c was destroyed to the leakage of bacteria cellular components. All these data indicated LA-Bac8c could be used as a useful antimicrobial peptide with wide application prospect.


Subject(s)
Anti-Bacterial Agents/pharmacology , Antimicrobial Cationic Peptides/pharmacology , Staphylococcus aureus/drug effects , Thioctic Acid/pharmacology , Animals , Anti-Bacterial Agents/chemical synthesis , Anti-Bacterial Agents/chemistry , Antimicrobial Cationic Peptides/chemical synthesis , Antimicrobial Cationic Peptides/chemistry , Biofilms/drug effects , Cell Line , Cell Survival/drug effects , Dose-Response Relationship, Drug , Hydrophobic and Hydrophilic Interactions , Mice , Microbial Sensitivity Tests , Molecular Structure , Structure-Activity Relationship , Thioctic Acid/chemistry
11.
Langmuir ; 35(40): 13031-13039, 2019 10 08.
Article in English | MEDLINE | ID: mdl-31537058

ABSTRACT

The increasing application of gold nanoparticles (AuNPs) in biomedicine requires extensive investigation of surface modification and stabilization to maximize their advantages for the diversity of more challenging biological utilization. Herein, a thiol-mediated multifunctional phospholipid ligand was designed while disclosing a zwitterionic nature to AuNPs. The ligand was synthesized by attachment to two bidentate lipoic acid (LA) anchor groups and incorporation of a zwitterionic phosphatidylcholine (PC) group, allowing for excellent hydrophilicity. As demonstrated through ultraviolet-visible spectroscopy, appropriate 7 nm diameter AuNPs modified with a 1,2-dilipoyl-sn-glycero-3-phosphorylcholine (di-LA-PC) compact ligand exhibited the best colloidal stability in a high NaCl concentration of up to 217 mM, different temperatures, and a wide range of pH values from 3 to 11 when compared to the traditional surfactants or thiol-contained amino acid surface modification cases. These AuNPs are also stable without specific interaction to positively/negatively charged proteins, possibly leading to prolonged blood circulation after in vivo administration. Moreover, much more resistance to ligand competition of dithiothreitol was found than other thiol-coated AuNPs, which further highlighted their affinity in an aqueous system. Biocompatibility of the zwitterionic ligand di-LA-PC-modified AuNPs was finally evaluated by hemolysis and cytotoxicity tests. Cumulatively, the remarkable stability and biocompatibility of AuNPs, multicoordinated with a di-LA-PC ligand, potentially motivated them as a practical alternative for surface tailoring in biotechnology.

12.
Mol Pharm ; 15(12): 5479-5492, 2018 12 03.
Article in English | MEDLINE | ID: mdl-30376635

ABSTRACT

Nanomedicines in polymeric therapeutics present a potential treatment for cancers. However, their clinical effectiveness still has room to be improved. Herein, reduction-responsive reversibly core-cross-linked micelles based on the poly(ethylene glycol)-dihydrolipoic acid (MeO-PEG2k-DHLA) conjugate were developed for triggered intracellular release of camptothecin (CPT). Coupling two molecules of dihydrolipoic acid (DHLA) to methyl-terminated PEG (Mw 2000) through a labile ester bond was performed by solution-phase condensation reaction. Due to the amphiphilic property, the MeO-PEG2k-DHLA conjugate formed micelles that were readily cross-linked with disulfide formation dispersed in water. These sole cross-linked micelles were 74.9 nm in hydrodiameter, as analyzed by dynamic light scattering (DLS). The nanostructures demonstrated excellent stability against extensive dilution, while rapidly dissociating under 10 mM glutathione (GSH), highlighting their potential for drug delivery. Interestingly, CPT was modified with a disulfide linkage and subsequently conjugated to the MeO-PEG2k-DHLA polymer scaffold. Core-cross-linking of the micelles achieved high drug loading of CPT (31.81%, wt %) and demonstrated that CPT release at pH 7.4 was significantly declined by cross-linking (i.e., less than 15% release in 24 h), whereas more than 90% of CPT was released under 10 mM GSH condition. In vitro cellular uptake and MTT assays showed that CPT-conjugated MeO-PEG2k-DHLA micelles were effectively internalized into tumor cells to induce the cytotoxic effects against HepG-2 and MCF-7 cells. Importantly, in vivo pharmacokinetics analysis demonstrated the nanoscale feature of micelles makes CPT to present longer retention time, resulting in a higher accumulation at tumor sites. Taken together, the disulfide core-cross-linked MeO-PEG2k-DHLA multifunctional micelles with high drug loading and excellent stability are potential candidates for tumor-targeting drug delivery.


Subject(s)
Antineoplastic Agents, Phytogenic/administration & dosage , Camptothecin/administration & dosage , Drug Carriers/chemistry , Neoplasms/drug therapy , Antineoplastic Agents, Phytogenic/pharmacokinetics , Camptothecin/pharmacokinetics , Cross-Linking Reagents/chemistry , Drug Compounding/methods , Drug Liberation , Drug Screening Assays, Antitumor , Drug Stability , Glutathione/metabolism , Hep G2 Cells , Humans , MCF-7 Cells , Micelles , Nanoparticles/chemistry , Neoplasms/pathology , Particle Size , Polyethylene Glycols , Thioctic Acid/analogs & derivatives
13.
J Nanosci Nanotechnol ; 18(12): 8124-8132, 2018 12 01.
Article in English | MEDLINE | ID: mdl-30189929

ABSTRACT

To develop new antimicrobial synthetic lipopeptides with optimizing peptide length, cationic tripeptides RWR/WRR were N-terminal fatty acylated, and self-assembled with 1-dodecanethiol-anchored gold nanoparticles (Au-DT NPs) via hydrophobic interaction. The ultrashort lipopeptides and their nano-assemblies were effective against a variety of microorganisms, with minimal inhibitory concentrations ranging from 0.5 to 8 µg/mL. Hemolysis analysis and in vitro cytotoxicity assay revealed that self-assembling with Au-DT NPs would improve biological toxicity of lipopeptides, especially for the most active lipopeptides (Palmitoyl-RWR, Palmitoyl-WRR) with long lipid tails. As lipopeptides/ Au-DT NPs displayed slower bactericidal kinetics compared with free lipopeptides, the mode of action was further investigated. Difference in membrane targeting mechanism between lipopeptides and their nano-assemblies may be attributed to the structural architecture. The simple composition and diverse specificities of lipopeptides/Au-DT NPs, as well as their biocompatibility could make them as economically available potent antimicrobial agents for various applications.


Subject(s)
Anti-Bacterial Agents , Anti-Infective Agents , Metal Nanoparticles , Anti-Bacterial Agents/pharmacology , Anti-Infective Agents/pharmacology , Gold/pharmacology , Lipopeptides/pharmacology , Microbial Sensitivity Tests
14.
Bioorg Med Chem ; 25(12): 3247-3258, 2017 06 15.
Article in English | MEDLINE | ID: mdl-28465086

ABSTRACT

7-Ethyl-10-hydroxycamptothecin (SN38), as a highly active topoisomerase I inhibitor, is 200-2000-fold more cytotoxic than irinotecan (CPT-11) commercially available as Camptosar®. However, poor solubility and low stability extensively restricted its clinical utility. In this report, dual SN38 phospholipid conjugate (Di-SN38-PC) prodrug based liposomes were developed in order to compact these drawbacks. Di-SN38-PC prodrug was first synthesized by inhomogeneous conjugation of two SN38-20-O-succinic acid molecules with L-α-glycerophosphorylcholine (GPC). The assembly of the prodrug was carried out without any excipient by using thin film method. Dynamic light scattering (DLS), transmission electron microscope (TEM) and cryogenic transmission electron microscopy (cyro-TEM) characterization indicated that Di-SN38-PC can form spherical liposomes with narrow particle size (<200nm) and negatively charged surface (-21.6±3.5mV). The loading efficiency of SN38 is 65.2 wt.% after a simple calculation. In vitro release test was further performed in detail. The results demonstrated that Di-SN38-PC liposomes were stable in neutral environment but degraded in a weakly acidic condition thereby released parent drug SN38 effectively. Cellular uptake studies reflected that the liposomes could be internalized into cells more significantly than SN38. In vitro antitumor activities were finally evaluated by MTT assay, colony formation assay, flow cytometry, RT-PCR analysis and Western Blot. The results showed that Di-SN38-PC liposomes had a comparable cytotoxicity with SN38 against MCF-7 and HBL-100, and a selective promotion of apoptosis of tumor cells. Furthermore, a pharmacokinetics test showed that Di-SN38-PC liposomes had a longer circulating time in blood compared with the parent drug. All the results indicate that Di-SN38-PC liposomes are an effective delivery system of SN38.


Subject(s)
Antineoplastic Agents, Phytogenic/chemistry , Antineoplastic Agents, Phytogenic/pharmacology , Camptothecin/analogs & derivatives , Prodrugs/chemistry , Prodrugs/pharmacology , Animals , Antineoplastic Agents, Phytogenic/administration & dosage , Antineoplastic Agents, Phytogenic/pharmacokinetics , Camptothecin/administration & dosage , Camptothecin/chemistry , Camptothecin/pharmacokinetics , Camptothecin/pharmacology , Cell Line, Tumor , Cell Survival/drug effects , Humans , Liposomes , Mice, Inbred BALB C , Mice, Nude , Neoplasms/drug therapy , Neoplasms/metabolism , Phospholipids/administration & dosage , Phospholipids/chemistry , Phospholipids/pharmacokinetics , Phospholipids/pharmacology , Prodrugs/administration & dosage , Prodrugs/pharmacokinetics , Solubility , Succinic Acid/administration & dosage , Succinic Acid/chemistry , Succinic Acid/pharmacokinetics , Succinic Acid/pharmacology
15.
Small ; 12(4): 524-33, 2016 Jan 27.
Article in English | MEDLINE | ID: mdl-26641209

ABSTRACT

Metal nanocrystals (NCs) are grown directly on the surface of reduced graphene oxide (rGO), which can maximize the rGO-NCs contact/interaction to achieve the enhanced catalytic activity. However, it is difficult to control the size and morphology of metal NCs by in situ method due to the effects of functional groups on the surface of GO, and as a result, the metal NCs/rGO hybrids are conventionally synthesized by two-step method. Herein, one-pot synthesis of Pt-Co alloy NCs is demonstrated with concave-polyhedrons and concave-nanocubes bounded by {hkl} and {hk0} high-index facets (HIFs) distributed on rGO. GO can affect the geometry and electronic structure of Pt-Co NCs. Thanks to the synergy of the HIFs and the electronic effect of the intimate contact/interaction between Pt-Co alloy and rGO, these as-prepared Pt-Co NCs/rGO hybrids presents enhanced catalytic properties for the electrooxidation of formic acid, as well as for the oxygen reduction reaction.

16.
Bioorg Med Chem Lett ; 25(2): 188-91, 2015 Jan 15.
Article in English | MEDLINE | ID: mdl-25515557

ABSTRACT

In this Letter, a new type of nitrogen mustard conjugate vesicles is developed to improve the stability and efficiency of anticancer drug. Benzoic acid nitrogen mustard-peptide (AAAK) conjugate was designed and synthesized, which was found to self-assemble into vesicles in water. The formation of the vesicles was confirmed by dynamic light scattering (DLS), transmission electron microscopy (TEM) and circular dichroism (CD). The degradation data revealed that the benzoic acid nitrogen mustard peptide (AAAK) conjugate vesicles are more stable than the parent drug in aqueous solution. Furthermore, MTT assay revealed that the free drug conjugate has similar antitumor activity against MCF-7, Hela, HepG-2 cell lines compared with the parent drug. The benzoic acid nitrogen mustard-peptide conjugate vesicles may have potential in the treatment of cancers.


Subject(s)
Antineoplastic Agents/chemical synthesis , Benzoic Acid/chemical synthesis , Drug Carriers/chemical synthesis , Mechlorethamine/chemical synthesis , Antineoplastic Agents/pharmacology , Benzoic Acid/pharmacology , Cell Survival/drug effects , Cell Survival/physiology , Dose-Response Relationship, Drug , Drug Carriers/pharmacology , HeLa Cells , Hep G2 Cells , Humans , MCF-7 Cells , Mechlorethamine/pharmacology
17.
J Gene Med ; 16(5-6): 109-21, 2014.
Article in English | MEDLINE | ID: mdl-24981025

ABSTRACT

BACKGROUND: In the present study, a well-defined glucose and guanidine based copolymer, galactosylated 2-hydroxypropyl methacrylamide-s-3-guanidinopropyl methacrylamide (HPMA-s-GPMA) abbreviated as GGH was prepared and self-assembled with small hairpin RNA (shRNA) to inhibit human telomerase reverse transcriptase (hTERT) gene expression in vitro to develop a shRNA carrier. METHODS: First, HPMA-s-APMA copolymers were synthesized by aqueous reversible addition-fragmentation chain transfer polymerization, followed by galactosylation and guanidinylation. Then, three target shRNAs containing green fluorescent protein gene as a reporter were combined with GGH to form shRNA/GGH polyplexes. RESULTS: GGH copolymers could condense shRNA to form shRNA/GGH polyplex particles with a diameter in the range 122.8-331.6 nm in phosphate-buffered saline, and zeta potential values ranging from +3.7 to +16.5 mV at various charge ratios (N/P). That the cytotoxicity of GGH copolymers was significantly lower than that of PEI in human hepatocellular liver carcinoma cells (HepG2) and human cervix epithelial carcinoma cells. The transfection efficiency of shRNA/GGH polyplexes was higher than that of PEI at a charge ratio of 12 in the HepG2 cell line. Furthermore, shRNA/GGH polyplexes could effectively silence hTERT mRNA expression in serum-free medium (p < 0.01) and decrease the aggregation of protein in the medium with the presence of 10% serum. In addition, hTERT mRNA expression in HepG2 cells demosntrate a significant difference between siRNA/GGH polyplexes and blank samples (p < 0.05). CONCLUSIONS: GGH copolymers could integrate advantages relating to galactose content for hepatocyte targeting, guanidino groups for cell penetration and HPMA component for shielding, showing great potential for effective hepatocyte targeting gene delivery.


Subject(s)
Acrylic Resins/chemistry , Drug Carriers/chemistry , Gene Expression , Gene Transfer Techniques , Guanidines/chemistry , RNA Interference , RNA, Small Interfering/genetics , Telomerase/genetics , Acrylic Resins/chemical synthesis , Cell Line, Tumor , Drug Carriers/chemical synthesis , Guanidines/chemical synthesis , HeLa Cells , Hep G2 Cells , Humans , Particle Size , RNA, Messenger/genetics , RNA, Small Interfering/administration & dosage , RNA, Small Interfering/chemistry , Transfection
18.
Biofouling ; 30(3): 313-22, 2014.
Article in English | MEDLINE | ID: mdl-24558981

ABSTRACT

In this work, antibacterial and anti-adhesive polymeric thin films were constructed on polyacrylonitrile (PAN) nanofibrous membranes in order to extend their applications. Polyhexamethylene guanidine hydrochloride (PHGH) as an antibacterial agent and heparin (HP) as an anti-adhesive agent have been successfully coated onto the membranes via a layer-by-layer (LBL) assembly technique confirmed by attenuated total reflectance Fourier transform infrared spectroscopy (ATR-FTIR), energy-dispersive spectroscopy (EDS) and scanning electron microscopy (SEM). The antibacterial properties of LBL-functionalized PAN nanofibrous membranes were evaluated using the Gram-positive bacterium Staphylococcus aureus and the Gram-negative Escherichia coli. Furthermore, the dependence of the antibacterial activity and anti-biofouling performance on the number of layers in the LBL films was investigated quantitatively. It was found that these LBL-modified nanofibrous membranes possessed high antibacterial activities, easy-cleaning properties and stability under physiological conditions, thus qualifying them as candidates for anti-biofouling coatings.


Subject(s)
Biofouling/prevention & control , Membranes, Artificial , Nanofibers/chemistry , Biofilms/drug effects , Cell Adhesion/drug effects , Microbial Sensitivity Tests , Staphylococcus aureus/drug effects
19.
J Control Release ; 373: 319-335, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38986911

ABSTRACT

Diabetic foot ulcer (DFU), which is characterised by damage to minute blood vessels or capillaries around wounds, is one of the most serious and dreaded complications of diabetes. It is challenging to repair chronic non-healing DFU wounds. Vascular endothelial growth factor (VEGF) plays an important role in angiogenesis and promotes wound healing in DFU. However, it is difficult to sustainably deliver VEGF to the wound site owing to its poor stability and easy degradation. To overcome this challenge, lipid nanoparticles (LNP) encapsulating circular RNA (circRNA) encoding VEGF-A have been developed to continuously generate and release VEGF-A and accelerate diabetic wound healing. First, VEGF-A circRNA was synthesized using group I intron autocatalysis strategy and confirmed by enzyme digestion, polymerase chain reaction, and sequencing assay. VEGF-A circRNA was encapsulated in ionizable lipid U-105-derived LNP (U-LNP) using microfluidic technology to fabricate U-LNP/VEGF-A circRNA. For comparison, a commercially ionizable lipid ALC-0315-derived LNP (A-LNP) encapsulating circRNA (A-LNP/circRNA) was used. Dynamic light scattering and transmission electron microscopy characterization indicated that U-LNP/circRNA had spherical structure with an average diameter of 108.5 nm, a polydispersity index of 0.22, and a zeta potential of -3.31 mV. The messenger RNA (mRNA) encapsulation efficiency (EE%) of U-LNP was 87.12%. In vitro transfection data confirmed better stability and long-term VEGF-A expression of circRNA compared with linear mRNA. Assessment of cytotoxicity and innate immunity further revealed that U-LNP/circRNA was biocompatible and induced a weak congenital immune response. Cell scratch and angiogenesis tests demonstrated the bioactivity of U-LNP/VEGF-A circRNA owing to its VEGF-A expression. In situ bioluminescence imaging of firefly luciferase (F-Luc) probe and ELISA demonstrated that circRNA had long-term and strong expression of VEGF-A in the first week, and a gradual decrease in the next week at the wound site and surrounding areas. Finally, a diabetic mouse model was used to validate the healing effect of U-LNP/VEGF-A circRNA formulation. The results showed that a single dose of U-LNP/VEGF-A circRNA administered by dripping resulted in almost complete wound recovery on day 12, which was significantly superior to that of U-LNP/VEGF-A linear mRNA, and it also outperformed recombinant human vascular endothelial growth factor (rhVEGF) injection and A-LNP/circRNA dripping. Histological analysis confirmed the healing efficiency and low toxicity of U-LNP/VEGF-A circRNA formulation. Together, VEGF-A circRNA delivered by U-105-derived LNP showed good performance in wound healing, which was ascribed to the long-term expression and continuous release of VEGF-A, and has potential applications for the treatment of diabetic foot ulcer wounds.


Subject(s)
Diabetic Foot , Nanoparticles , RNA, Circular , Vascular Endothelial Growth Factor A , Wound Healing , RNA, Circular/genetics , Vascular Endothelial Growth Factor A/genetics , Wound Healing/drug effects , Animals , Male , Diabetic Foot/genetics , Humans , Diabetes Mellitus, Experimental/metabolism , Mice , Lipids/chemistry , Human Umbilical Vein Endothelial Cells , Mice, Inbred C57BL , Neovascularization, Physiologic/drug effects , Liposomes
20.
J Control Release ; 368: 663-675, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38492862

ABSTRACT

Interleukin-2 (IL-2) exhibits the unique capacity to modulate immune functions, potentially exerting antitumor effects by stimulating immune responses, making it highly promising for immunotherapy. However, the clinical use of recombinant IL-2 protein faces significant limitations due to its short half-life and systemic toxicity. To overcome these challenges and fully exploit IL-2's potential in tumor immunotherapy, this study reports the development of a tumor-activated IL-2 mRNA, delivered via lipid nanoparticles (LNPs). Initially, ionizable lipid U-101 derived nanoparticles (U-101-LNP) were prepared using microfluidic technology. Subsequent in vitro and in vivo delivery tests demonstrated that U-101-LNP achieved more effective transfection than the approved ALC-0315-LNP. Following this, IL-2F mRNAs, encoding fusion proteins comprising IL-2, a linker, and CD25 (IL-2Rα), were designed and synthesized through in vitro transcription. A cleavable linker, consisting of the peptide sequence SGRSEN↓IRTA, was selected for cleavage by matrix metalloproteinase-14 (MMP-14). IL-2F mRNA was then encapsulated in U-101-LNP to create U-101-LNP/IL-2F mRNA complexes. After optimization, assessments of expression efficiency, masking, and release characteristics revealed that IL-2F with linker C4 demonstrated superior performance. Finally, the antitumor activity of IL-2F mRNA was evaluated. The results indicated that U-101-LNP/IL-2F mRNA achieved the strongest antitumor effect, with an inhibition rate of 70.3%. Immunohistochemistry observations revealed significant expressions of IL-2, IFN-γ, and CD8, suggesting an up-regulation of immunomodulation in tumor tissues. This effect could be ascribed to the expression of IL-2F, followed by the cleavage of the linker under the action of MMP-14 in tumor tissue, which sustainably releases IL-2. H&E staining of tissues treated with U-101-LNP/IL-2F mRNA showed no abnormalities. Further evaluations indicated that the U-101-LNP/IL-2F mRNA group maintained proper levels of inflammatory factors without obvious alterations in liver and renal functions. Taken together, the U-101-LNP/IL-2F mRNA formulation demonstrated effective antitumor activity and safety, which suggests potential applicability in clinical immunotherapy.


Subject(s)
Liposomes , Nanoparticles , Neoplasms , Humans , Interleukin-2/genetics , Matrix Metalloproteinase 14 , Immunotherapy , Neoplasms/therapy
SELECTION OF CITATIONS
SEARCH DETAIL