Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 61
Filter
Add more filters











Publication year range
1.
Molecules ; 29(13)2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38999083

ABSTRACT

The delivery of therapeutic agents faces significant hurdles posed by the endo-lysosomal pathway, a bottleneck that hampers clinical effectiveness. This comprehensive review addresses the urgent need to enhance cellular delivery mechanisms to overcome these obstacles. It focuses on the potential of smart nanomaterials, delving into their unique characteristics and mechanisms in detail. Special attention is given to their ability to strategically evade endosomal entrapment, thereby enhancing therapeutic efficacy. The manuscript thoroughly examines assays crucial for understanding endosomal escape and cellular uptake dynamics. By analyzing various assessment methods, we offer nuanced insights into these investigative approaches' multifaceted aspects. We meticulously analyze the use of smart nanocarriers, exploring diverse mechanisms such as pore formation, proton sponge effects, membrane destabilization, photochemical disruption, and the strategic use of endosomal escape agents. Each mechanism's effectiveness and potential application in mitigating endosomal entrapment are scrutinized. This paper provides a critical overview of the current landscape, emphasizing the need for advanced delivery systems to navigate the complexities of cellular uptake. Importantly, it underscores the transformative role of smart nanomaterials in revolutionizing cellular delivery strategies, leading to a paradigm shift towards improved therapeutic outcomes.


Subject(s)
Endosomes , Lysosomes , Lysosomes/metabolism , Humans , Endosomes/metabolism , Drug Delivery Systems , Drug Carriers/chemistry , Nanostructures/chemistry , Animals , Nanoparticles/chemistry
2.
Chemistry ; 30(28): e202400174, 2024 May 17.
Article in English | MEDLINE | ID: mdl-38456376

ABSTRACT

We report the synthesis of a series of amphiphilic p-sulfonatocalix[4]arenes with varying alkyl chain lengths (CX4-Cn) and their application as efficient counterion activators for membrane transport of cell-penetrating peptides (CPPs). The enhanced membrane activity is confirmed with the carboxyfluorescein (CF) assay in vesicles and by the direct cytosolic delivery of CPPs into CHO-K1, HCT 116, and KTC-1 cells enabling excellent cellular uptake of the CPPs into two cancer cell lines. Intracellular delivery was confirmed by fluorescence microscopy after CPP entry into live cells mediated by CX4-Cn, which was also quantified after cell lysis by fluorescence spectroscopy. The results present the first systematic exploration of structure-activity relationships for calixarene-based counterion activators and show that CX4-Cn are exceptionally effective in cellular delivery of CPPs. The dodecyl derivative, CX4-C12, serves as best activator. A first mechanistic insight is provided by efficient CPP uptake at 4 °C and in the presence of the endocytosis inhibitor dynasore, which indicates a direct translocation of the CPP-counterion complexes into the cytosol and highlights the potential benefits of CX4-Cn for efficient and direct translocation of CPPs and CPP-conjugated cargo molecules into the cytosol of live cells.


Subject(s)
Calixarenes , Cell-Penetrating Peptides , Cricetulus , Calixarenes/chemistry , Cell-Penetrating Peptides/chemistry , Cell-Penetrating Peptides/metabolism , Humans , CHO Cells , Animals , Structure-Activity Relationship , Cell Line, Tumor , Phenols/chemistry , Endocytosis , Surface-Active Agents/chemistry
3.
Article in English | MEDLINE | ID: mdl-38405370

ABSTRACT

Nitric Oxide (NO) photocleavable donors are useful tools for interrogating nitric oxide signalling and have potential use in photopharmacological applications. There is currently intensive research into newer methods to improve NO release and kinetic profiles. Herein, we report the design and synthesis of a solid-supported photocleavable NO donor synthesized by ligating an N-nitroso photocleavable nitric oxide derivative to a TentaGel® polymer resin bead. Illumination with 365 nm light released nitric oxide that could be tracked via a turn-on fluorescence response (λex = 450 nm, λem = 545 nm) and measured using the Griess assay and diaminorhodamine derivatives. These beads were further shown to be compatible with living A549 cells and had the ability to deliver greater concentrations of nitric oxide to cells proximal to a bead versus cells at more distal locations within the same well.

4.
J Pept Sci ; 29(7): e3477, 2023 Jul.
Article in English | MEDLINE | ID: mdl-36606596

ABSTRACT

Nucleic acid-templated chemistry opens the intriguing prospect of triggering the synthesis of drugs only in diseased cells. Herein, we explore the feasibility of using RNA-templated chemical reactions for the activation of a known Smac peptidomimetic compound (SMC), which has proapoptotic activity. Two peptide nucleic acid (PNA) conjugates were used to enable conditional activation of a masked SMC by reduction of an azide either by Staudinger reduction or catalytic photoreduction using a ruthenium complex. The latter provided ~135 nM SMC-PNA on as little as 10 nM (0.01 eq.) template. For the evaluation of the templated azido-SMC reduction system in cellulo, a stable HEK 293 cell line was generated, which overexpressed a truncated, non-functional form of the XIAP mRNA target. We furthermore describe the development of electroporation protocols that enable a robust delivery of PNA conjugates into HEK 293 cells. The action of the reactive PNA conjugates was evaluated by viability and flow cytometric apoptosis assays. In addition, electroporated probes were re-isolated and analyzed by ultra-high performance liquid chromatography (UPLC). Unfortunately, the ruthenium-PNA conjugate proved phototoxic, and treatment of cells with PNA-linked reducing agent and the azido-masked SMC conjugate did not result in a greater viability loss than treatment with scrambled sequence controls. Intracellular product formation was not detectable. A control experiment in total cellular RNA isolate indicated that the templated reaction can in principle proceed in a complex system. The results of this first-of-its-kind study reveal the numerous hurdles that must be overcome if RNA molecules are to trigger the synthesis of pro-apoptotic drugs inside cells.


Subject(s)
Peptide Nucleic Acids , Ruthenium , Humans , Peptide Nucleic Acids/pharmacology , Peptide Nucleic Acids/chemistry , RNA , HEK293 Cells , Ruthenium/pharmacology , Ruthenium/chemistry , Peptides
5.
Small ; 19(2): e2205318, 2023 01.
Article in English | MEDLINE | ID: mdl-36399647

ABSTRACT

The clustered regularly interspaced short palindromic repeats (CRISPR)/CRISPR-associated protein 9 (Cas9) system offers great opportunities for the treatment of numerous diseases by precise modification of the genome. The functional unit of the system is represented by Cas9/sgRNA ribonucleoproteins (RNP), which mediate sequence-specific cleavage of DNA. For therapeutic applications, efficient and cell-specific transport into target cells is essential. Here, Cas9 RNP nanocarriers are described, which are based on lipid-modified oligoamino amides and folic acid (FolA)-PEG to realize receptor-mediated uptake and gene editing in cancer cells. In vitro studies confirm strongly enhanced potency of receptor-mediated delivery, and the nanocarriers enable efficient knockout of GFP and two immune checkpoint genes, PD-L1 and PVR, at low nanomolar concentrations. Compared with non-targeted nanoparticles, FolA-modified nanocarriers achieve substantially higher gene editing including dual PD-L1/PVR gene disruption after injection into CT26 tumors in vivo. In the syngeneic mouse model, dual disruption of PD-L1 and PVR leads to CD8+ T cell recruitment and distinct CT26 tumor growth inhibition, clearly superior to the individual knockouts alone. The reported Cas9 RNP nanocarriers represent a versatile platform for potent and receptor-specific gene editing. In addition, the study demonstrates a promising strategy for cancer immunotherapy by permanent and combined immune checkpoint disruption.


Subject(s)
CRISPR-Cas Systems , Neoplasms , Animals , Mice , CRISPR-Cas Systems/genetics , B7-H1 Antigen/metabolism , Ribonucleoproteins/genetics , Gene Editing , DNA , Neoplasms/therapy , Neoplasms/genetics
6.
J Control Release ; 352: 411-421, 2022 12.
Article in English | MEDLINE | ID: mdl-36272662

ABSTRACT

Crystallins, small heat shock chaperone proteins that prevent protein aggregation, are of potential value in treating protein aggregation disorders. However, their therapeutic use is limited by their low potency and poor intracellular delivery. One approach to facilitate the development of crystallins is to improve their activity, stability, and delivery. In this study, zinc addition to αB-crystallin-D3 (αB-D3) formed supramolecular nano- and micro- assemblies, induced dose-dependent changes in structure (beta-sheet to alpha-helix) and increased surface hydrophobicity and chemical stability. Further, crystallin assemblies exhibited a size-dependent chaperone activity, with the nano-assemblies being superior to micro-assemblies and 4.3-fold more effective than the native protein in preventing ß-mercaptoethanol induced aggregation of insulin. Insulin rescued by crystallin assemblies retained the activity as evidenced by glucose uptake in 3T3-L1 cells. The most active nano-assemblies enhanced protein stability, in the presence of urea, by 1.6-fold, whereas intracellular delivery was enhanced by 3.0-fold. The αB-D3 crystallin nano-assemblies exhibit uniquely enhanced stability, activity, and delivery compared to the native protein.


Subject(s)
Insulins , alpha-Crystallin B Chain , Protein Aggregates , Molecular Chaperones/chemistry , Molecular Chaperones/metabolism
7.
J Control Release ; 350: 256-270, 2022 10.
Article in English | MEDLINE | ID: mdl-35963467

ABSTRACT

Since the recent clinical approval of siRNA-based drugs and COVID-19 mRNA vaccines, the potential of RNA therapeutics for patient healthcare has become widely accepted. Lipid nanoparticles (LNPs) are currently the most advanced nanocarriers for RNA packaging and delivery. Nevertheless, the intracellular delivery efficiency of state-of-the-art LNPs remains relatively low and safety and immunogenicity concerns with synthetic lipid components persist, altogether rationalizing the exploration of alternative LNP compositions. In addition, there is an interest in exploiting LNP technology for simultaneous encapsulation of small molecule drugs and RNA in a single nanocarrier. Here, we describe how well-known tricyclic cationic amphiphilic drugs (CADs) can be repurposed as both structural and functional components of lipid-based NPs for mRNA formulation, further referred to as CADosomes. We demonstrate that selected CADs, such as tricyclic antidepressants and antihistamines, self-assemble with the widely-used helper lipid DOPE to form cationic lipid vesicles for subsequent mRNA complexation and delivery, without the need for prior lipophilic derivatization. Selected CADosomes enabled efficient mRNA delivery in various in vitro cell models, including easy-to-transfect cancer cells (e.g. human cervical carcinoma HeLa cell line) as well as hard-to-transfect primary cells (e.g. primary bovine corneal epithelial cells), outperforming commercially available cationic liposomes and state-of-the-art LNPs. In addition, using the antidepressant nortriptyline as a model compound, we show that CADs can maintain their pharmacological activity upon CADosome incorporation. Furthermore, in vivo proof-of-concept was obtained, demonstrating CADosome-mediated mRNA delivery in the corneal epithelial cells of rabbit eyes, which could pave the way for future applications in ophthalmology. Based on our results, the co-formulation of CADs, helper lipids and mRNA into lipid-based nanocarriers is proposed as a versatile and straightforward approach for the rational development of drug combination therapies.


Subject(s)
COVID-19 Drug Treatment , Nanoparticles , Animals , Antidepressive Agents, Tricyclic , Cations , Cattle , Drug Combinations , Drug Repositioning , HeLa Cells , Humans , Lipids/chemistry , Liposomes , Nanoparticles/chemistry , Nortriptyline , RNA, Messenger/genetics , RNA, Small Interfering/genetics , Rabbits
8.
J Colloid Interface Sci ; 627: 270-282, 2022 Dec.
Article in English | MEDLINE | ID: mdl-35849860

ABSTRACT

HYPOTHESIS: Most experimental procedures applied in modern biology involve cargo delivering into cells. One of the ways to cargo introduction is osmotic-mediated intracellular vesicle swelling. However, its widespread use was hindered due to cargo size (<10 nm) and cell-type-related restrictions. We addressed the issue of the composition of colloidal loading solution to enhance the efficiency of cellular delivery. EXPERIMENTS: We examined the effectiveness of colloidal loading solutions of varied compositions, including various types and sizes of polymers building osmotic pressure. We used confocal imaging coupled with fluorescence correlation spectroscopy to evaluate the introduction of polymers, proteins, nanoparticles, and DNA plasmids (cargos of sizes 1-175 nm) to cells representing eight cell lines: cancer, normal, epithelial, and mesenchymal ones. FINDINGS: We found that cellular delivery effectiveness strongly correlates with the size and concentration of osmotic pressure building polymers and not with the high value of the osmotic pressure itself. We show that polymer solutions at the entangled regime of concentrations enhance the delivery of large biomacromolecules even of size 200 nm (DNA plasmids) into cells, including MDA-MB-231 cells - so far resistant to the osmotic procedure. We show that the colloid loading medium based on entangled polymer chains is a versatile cargo delivery tool for molecular biology.


Subject(s)
Nanoparticles , Polymers , Colloids , DNA , Nanoparticles/chemistry , Plasmids , Polymers/chemistry
9.
Chemistry ; 28(45): e202201164, 2022 Aug 10.
Article in English | MEDLINE | ID: mdl-35699671

ABSTRACT

Liposomes are effective therapeutic delivery nanocarriers due to their ability to encapsulate and enhance the pharmacokinetic properties of a wide range of therapeutics. Two primary areas in which improvement is needed for liposomal drug delivery is to enhance the ability to infiltrate cells and to facilitate derivatization of the liposome surface. Herein, we report a liposome platform incorporating a cyclic disulfide lipid (CDL) for the dual purpose of enhancing cell entry and functionalizing the liposome membrane through thiol-disulfide exchange. In order to accomplish this, CDL-1 and CDL-2, composed of lipoic acid (LA) or asparagusic acid (AA) appended to a lipid scaffold, were designed and synthesized. A fluorescence-based microplate immobilization assay was implemented to show that these compounds enable convenient membrane decoration through reaction with thiol-functionalized small molecules. Additionally, fluorescence microscopy experiments indicated dramatic enhancements in cellular delivery when CDLs were incorporated within liposomes. These results demonstrate that multifunctional CDLs serve as an exciting liposome system for surface decoration and enhanced cellular delivery.


Subject(s)
Drug Delivery Systems , Liposomes , Disulfides , Drug Delivery Systems/methods , Lipids , Liposomes/metabolism , Sulfhydryl Compounds
10.
Chemistry ; 28(46): e202201057, 2022 Aug 16.
Article in English | MEDLINE | ID: mdl-35639353

ABSTRACT

We report boronate-caged guanidine-lipid 1 that activates liposomes for cellular delivery only upon uncaging of this compound by reactive oxygen species (ROS) to produce cationic lipid products. These liposomes are designed to mimic the exceptional cell delivery properties of cell-penetrating peptides (CPPs), while the inclusion of the boronate cage is designed to enhance selectivity such that cell entry will only be activated in the presence of ROS. Boronate uncaging by hydrogen peroxide was verified by mass spectrometry and zeta potential (ZP) measurements. A microplate-based fluorescence assay was developed to study the ROS-mediated vesicle interactions between 1-liposomes and anionic membranes, which were further elucidated via dynamic light scattering (DLS) analysis. Cellular delivery studies utilizing fluorescence microscopy demonstrated significant enhancements in cellular delivery only when 1-liposomes were incubated with hydrogen peroxide. Our results showcase that lipid 1 exhibits strong potential as an ROS-responsive liposomal platform for targeted drug delivery applications.


Subject(s)
Hydrogen Peroxide , Liposomes , Guanidine , Lipids/chemistry , Liposomes/chemistry , Reactive Oxygen Species/metabolism
11.
Talanta ; 245: 123462, 2022 Aug 01.
Article in English | MEDLINE | ID: mdl-35436732

ABSTRACT

Currently, researches on nanomaterials have been restricted by slow and multistep synthesis procedures. Herein, we demonstrate an ultrafast, one step method of purification and delivery of quantum dots into living cells, actuated by the acoustic streaming (AS) produced through a gigahertz resonator. Results demonstrate that the impurities in the carbon dots (CDs) can be extracted immediately aided by the acoustic forcing, with extra high purification efficiency of 93%. The system can also efficiently deliver the CDs into cells, showing excellent nucleus and mitochondria uptake under 3 min of AS treatment, and making the organelles of cells to be recorded more easily and simultaneously. More importantly, the AS is found to further accelerate the bioreaction inside the cells, thus realizes the enhanced biosensing of Fe3+ in single living cells. This work develops a novel type of multifunctional method for effective purification, intracellular delivery and biosensing of nanomaterials, inspiring the biological/medical nanotechnology researches at subcellular level.


Subject(s)
Nanostructures , Quantum Dots , Acoustics , Carbon , Fluorescent Dyes , Mitochondria
12.
ACS Biomater Sci Eng ; 8(4): 1566-1572, 2022 04 11.
Article in English | MEDLINE | ID: mdl-35263989

ABSTRACT

Cellular delivery of therapeutic macromolecules such as proteins, peptides, and nucleic acids remains limited due to inefficient transport across the cellular plasma membrane. Gap junction channels, composed of connexin proteins, provide a mechanism for direct transfer of small molecules across membranes, and recent evidence suggests that the transfer of larger, polymer-like molecules such as microRNAs may be possible. Here, we report direct evidence of gap junction-mediated transfer of polymeric macromolecules. Specifically, we examined the transport of dextran chains with molecular weights ranging from 10 to 70 kDa. We found that dextran chains of up to 40 kDa can diffuse through at least five cell layers in a gap junction-dependent manner within a 30 min time frame. Further, we evaluated the ability of connectosomes, cell-derived vesicles containing functional connexin proteins, to be loaded with dextran chains. By opening connexon hemichannel pores within the membranes of connectosomes, we found that 10 kDa dextran was loaded into more than 90% of vesicles, with reduced levels of loading for dextran chains of larger molecular weight. Upon delivering 10 kDa dextran-loaded connectosomes to cells, we further found that connectosomes transferred these membrane-impermeable molecules to the cellular cytosol with dramatically improved efficiency in comparison to the delivery of free, unencapsulated dextran. Collectively, these results reveal that polymeric macromolecules can be delivered to cells via gap junctions, suggesting that the gap junction route may be useful for the delivery of polymeric therapeutic molecules, such as nucleic acids and peptides.


Subject(s)
Dextrans , Nucleic Acids , Connexins/chemistry , Connexins/metabolism , Dextrans/metabolism , Gap Junctions/metabolism , Nucleic Acids/metabolism , Polymers/metabolism
13.
Pharm Res ; 39(6): 1035-1045, 2022 Jun.
Article in English | MEDLINE | ID: mdl-35112228

ABSTRACT

Endosome escape is a key process for intracellular uptake of intact biomolecules and therapeutics, such as nucleic acids. Lysosome escape is a more common pathway during endocytosis, while some biomolecular, organic and inorganic materials are found to enhance the endosome escape, and several mechanisms have been proposed accordingly. Specifically, some inorganic nanomaterials show their unique mechanisms of action for enhanced endosome escape, including salt osmotic effect and gas blast effect. These inorganic nanomaterials are basically weakly alkaline and are naturally featured with the anti-acidification capacity, with limited solubility in neutral solutions. This review paper has briefly presented the strategies in the design of inorganic nanoparticle-based cellular delivery vehicles with endosome escapability and discussed a few typical inorganic nanomaterials that are currently widely examined for delivery purpose. A brief summary and prospect for this kind of inorganic nanomaterials are provided.


Subject(s)
Nanoparticles , Nanostructures , Endocytosis , Endosomes/metabolism , Lysosomes , Nanoparticles/metabolism
14.
Mater Today Bio ; 13: 100193, 2022 Jan.
Article in English | MEDLINE | ID: mdl-35005598

ABSTRACT

Highly efficient intracellular delivery strategies are essential for developing therapeutic, diagnostic, biological, and various biomedical applications. The recent advancement of micro/nanotechnology has focused numerous researches towards developing microfluidic device-based strategies due to the associated high throughput delivery, cost-effectiveness, robustness, and biocompatible nature. The delivery strategies can be carrier-mediated or membrane disruption-based, where membrane disruption methods find popularity due to reduced toxicity, enhanced delivery efficiency, and cell viability. Among all of the membrane disruption techniques, the mechanoporation strategies are advantageous because of no external energy source required for membrane deformation, thereby achieving high delivery efficiencies and increased cell viability into different cell types with negligible toxicity. The past two decades have consequently seen a tremendous boost in mechanoporation-based research for intracellular delivery and cellular analysis. This article provides a brief review of the most recent developments on microfluidic-based mechanoporation strategies such as microinjection, nanoneedle arrays, cell-squeezing, and hydroporation techniques with their working principle, device fabrication, cellular delivery, and analysis. Moreover, a brief discussion of the different mechanoporation strategies integrated with other delivery methods has also been provided. Finally, the advantages, limitations, and future prospects of this technique are discussed compared to other intracellular delivery techniques.

15.
Adv Drug Deliv Rev ; 181: 114041, 2022 02.
Article in English | MEDLINE | ID: mdl-34763002

ABSTRACT

RNA therapeutics (e.g. siRNA, oligonucleotides, mRNA, etc.) show great potential for the treatment of a myriad of diseases. However, to reach their site of action in the cytosol or nucleus of target cells, multiple intra- and extracellular barriers have to be surmounted. Several non-viral delivery systems, such as nanoparticles and conjugates, have been successfully developed to meet this requirement. Unfortunately, despite these clear advances, state-of-the-art delivery agents still suffer from relatively low intracellular delivery efficiencies. Notably, our current understanding of the intracellular delivery process is largely oversimplified. Gaining mechanistic insight into how RNA formulations are processed by cells will fuel rational design of the next generation of delivery carriers. In addition, identifying which intracellular pathways contribute to productive RNA delivery could provide opportunities to boost the delivery performance of existing nanoformulations. In this review, we discuss both established as well as emerging techniques that can be used to assess the impact of different intracellular barriers on RNA transfection performance. Next, we highlight how several modulators, including small molecules but also genetic perturbation technologies, can boost RNA delivery by intervening at differing stages of the intracellular delivery process, such as cellular uptake, intracellular trafficking, endosomal escape, autophagy and exocytosis.


Subject(s)
Nanoparticle Drug Delivery System , RNA/administration & dosage , Transfection/methods , Cell Communication/physiology , Cell Membrane/metabolism , Clustered Regularly Interspaced Short Palindromic Repeats , Drug Evaluation, Preclinical , Humans , MicroRNAs/administration & dosage , Oligonucleotides/administration & dosage , RNA, Messenger/administration & dosage , RNA, Small Interfering/administration & dosage , RNAi Therapeutics
16.
Natl Sci Rev ; 8(8): nwaa268, 2021 Aug.
Article in English | MEDLINE | ID: mdl-34691708

ABSTRACT

Zeolitic imidazolate framework-8 (ZIF-8) and its composites have diverse applications. However, ZIF-8-based nanocomposites are mainly used as carriers in biomolecular delivery, with the functions of metal ions and ligands rarely used to modulate the biofunctions. In this work, dendritic mesoporous organosilica nanoparticles (DMONs) with tetrasulfide bond were used to confine ZIF-8 growth partially inside mesopores as a novel nanocomposite for mRNA delivery. Each component in the resultant DMONs-ZIF-8 contributed to mRNA delivery applications, including high loading benefitting from positively charged ZIF-8 and large mesopores of DMONs, endosomal escape promoted by the imidazole ring of ZIF-8, and long-term glutathione depletion mediated by both zinc ions and tetrasulfide bond. Combined together, DMONs-ZIF-8 demonstrated enhanced mRNA translation and better transfection efficiency than commercial products and toxic polymer-modified DMONs in vitro and in vivo.

17.
Methods Mol Biol ; 2355: 265-273, 2021.
Article in English | MEDLINE | ID: mdl-34386964

ABSTRACT

Based on the exceptionally high stability of γPNA (Gamma-modified peptide nucleic acid) duplexes, we designed a peptide/γPNA chimera in which a cell-penetrating TAT (HIV Tat-derived) peptide is flanked by two short complementary γPNA segments. Intramolecular hybridization of the γPNA segments results in a stable hairpin conformation in which the TAT peptide is constrained to form the loop. The TAT/γPNA hairpin (self-cyclized TAT peptide) enters cells at least tenfold more efficiently than its nonhairpin analog in which the two γPNA segments are noncomplementary. Extending one of the γPNA segments in the hairpin results in an overhang that can be used for binding and delivering a variety of nucleic acid-conjugated molecules into cells via hybridization to the overhang. We demonstrated efficient cellular delivery of an anti-telomerase γPNA that specifically reduced telomerase activity of A549 cells by over 97%.


Subject(s)
Peptides/chemistry , A549 Cells , Humans , Nucleic Acid Hybridization , Nucleic Acids , Peptide Nucleic Acids
18.
ACS Nano ; 15(5): 8095-8109, 2021 05 25.
Article in English | MEDLINE | ID: mdl-33724778

ABSTRACT

RNA therapeutics are poised to revolutionize medicine. To unlock the full potential of RNA drugs, safe and efficient (nano)formulations to deliver them inside target cells are required. Endosomal sequestration of nanocarriers represents a major bottleneck in nucleic acid delivery. Gaining more detailed information on the intracellular behavior of RNA nanocarriers is crucial to rationally develop delivery systems with improved therapeutic efficiency. Surfactant protein B (SP-B) is a key component of pulmonary surfactant (PS), essential for mammalian breathing. In contrast to the general belief that PS should be regarded as a barrier for inhaled nanomedicines, we recently discovered the ability of SP-B to promote gene silencing by siRNA-loaded and lipid-coated nanogels. However, the mechanisms governing this process are poorly understood. The major objective of this work was to obtain mechanistic insights into the SP-B-mediated cellular delivery of siRNA. To this end, we combined siRNA knockdown experiments, confocal microscopy, and focused ion beam scanning electron microscopy imaging in an in vitro non-small-cell lung carcinoma model with lipid mixing assays on vesicles that mimic the composition of (intra)cellular membranes. Our work highlights a strong correlation between SP-B-mediated fusion with anionic endosomal membranes and cytosolic siRNA delivery, a mode of action resembling that of certain viruses and virus-derived cell-penetrating peptides. Building on these gained insights, we optimized the SP-B proteolipid composition, which dramatically improved delivery efficiency. Altogether, our work provides a mechanistic understanding of SP-B-induced perturbation of intracellular membranes, offering opportunities to fuel the rational design of SP-B-inspired RNA nanoformulations for inhalation therapy.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Pulmonary Surfactant-Associated Protein B , Animals , Cell Line, Tumor , RNA, Small Interfering/genetics
19.
ACS Appl Mater Interfaces ; 13(9): 10760-10767, 2021 Mar 10.
Article in English | MEDLINE | ID: mdl-33621039

ABSTRACT

Exosomes are natural delivery vehicles because of their original feature such as low immunogenicity, excellent biocompatibility, and migration capability. Engineering exosomes with appropriate ligands are effective approaches to improve the low cellular uptake efficiency of exosomes. However, current strategies face considerable challenges due to the tedious and labor-intensive operational process. Here, we designed a novel peptides-equipped exosomes platform which can be assembled under convenient and mild reaction condition. Cell-penetrating peptides (CPPs) was conjugated on HepG2 cells-derived exosomes surface which can not only enhance the penetrating capacity of exosomes but also assist exosomes in loading antisense oligonucleotides (ASOs). The cellular uptake mechanism was investigated and we compared the difference between natural exosomes and modified exosomes. The resulting nanosystem demonstrated a preferential tropism for cells that are parented to their source tumor cells and could remarkably increase the cellular delivery of G3139 with efficient downregulation of antiapoptotic Bcl-2. This work developed a rapid strategy for intracellular delivery of nucleic acids, thus providing more possibilities toward personalized cancer medicine.


Subject(s)
Cell-Penetrating Peptides/chemistry , Drug Carriers/chemistry , Exosomes/chemistry , Oligodeoxyribonucleotides, Antisense/pharmacology , Thionucleotides/pharmacology , Cell-Penetrating Peptides/metabolism , Down-Regulation/drug effects , Drug Carriers/metabolism , Exosomes/metabolism , Gene Silencing/drug effects , Hep G2 Cells , Humans , Oligodeoxyribonucleotides, Antisense/genetics , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , Thionucleotides/genetics
20.
Bioorg Med Chem ; 29: 115906, 2021 01 01.
Article in English | MEDLINE | ID: mdl-33310547

ABSTRACT

Inhibitors of protein-protein interactions can be developed through a number of technologies to provide leads that include cell-impermeable molecules. Redesign of these impermeable leads to provide cell-permeable derivatives can be challenging and costly. We hypothesised that intracellular toxicity of leads could be assessed by microinjection prior to investing in the redesign process. We demonstrate this approach for our development of inhibitors of the protein-protein interaction between inducible nitric-oxide synthase (iNOS) and SPRY domain-containing SOCS box proteins (SPSBs). We microinjected a lead molecule into AD-293 cells and were able to perform an intracellular toxicity assessment. We also investigated the intracellular distribution and localisation of injected inhibitor using a fluorescently-labelled analogue. Our findings show that a lead peptide inhibitor, CP2, had no toxicity even at intracellular concentrations four orders of magnitude higher than its Kd for binding to SPSB2. This early toxicity assessment justifies further development of this cell-impermeable lead to confer cell permeability. Our investigation highlights the utility of microinjection as a tool for assessing toxicity during development of drugs targeting protein-protein interactions.


Subject(s)
Cytoplasm/metabolism , Enzyme Inhibitors/chemistry , Nitric Oxide Synthase Type II/metabolism , Peptides/chemistry , Suppressor of Cytokine Signaling Proteins/metabolism , Amino Acid Sequence , Cell Line , Cell Membrane Permeability , Cytoplasm/ultrastructure , Drug Development , Enzyme Inhibitors/administration & dosage , Enzyme Inhibitors/adverse effects , Humans , Microinjections , Models, Molecular , Optical Imaging , Peptides/administration & dosage , Peptides/adverse effects , Protein Binding , Structure-Activity Relationship
SELECTION OF CITATIONS
SEARCH DETAIL