Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 397
Filter
2.
Biomed Pharmacother ; 175: 116691, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38713941

ABSTRACT

Neurodegenerative diseases affect many people worldwide, and as the population ages, the incidence of these conditions increases. Alzheimer's disease (AD) and Parkinson's disease (PD) are the most prevalent neurodegenerative disorders worldwide. Different medicines are being used to control symptoms related to these conditions, but no treatment has yet been approved. Both genetic and environmental factors are involved in disease pathogenesis, and research on the pathophysiological pathways is still ongoing. The role of subcellular pathways and dysregulation in RNA pathways has been highlighted in pathophysiological studies, and treatment strategies focused on these pathways can be a promising approach. Many experiments have been conducted on delivering RNA cargo to the CNS to modulate various pathways involved. Yet another challenge to be faced is the effective transport of desired molecules to targets, which can be greatly hindered by distinct barriers limiting transport to the CNS, most noticeably the blood-brain barrier (BBB). Nanotechnology and the use of different nano-carriers for the delivery of nucleotides, peptides, proteins, and drug molecules are currently of great interest as these carriers help with better delivery and protection and, as a result, improve the effectiveness of the cargo. Nanocarriers can protect susceptible RNA molecules from possible degradation or destruction and improve their ability to reach the brain by enhancing BBB penetration. Different mechanisms for this process have been hypothesized. This review will go through the therapeutic application of RNA molecules in the treatment of AD and PD and the role of nanocarriers in overcoming delivery challenges and enhancing efficacy.


Subject(s)
Blood-Brain Barrier , Neurodegenerative Diseases , RNA , Humans , Neurodegenerative Diseases/drug therapy , Neurodegenerative Diseases/metabolism , Neurodegenerative Diseases/genetics , Animals , RNA/genetics , RNA/administration & dosage , Blood-Brain Barrier/metabolism , Blood-Brain Barrier/drug effects , Nanoparticles , Nanoparticle Drug Delivery System , Drug Delivery Systems/methods
3.
Int J Pharm ; 657: 124151, 2024 May 25.
Article in English | MEDLINE | ID: mdl-38657717

ABSTRACT

Neovascularization contributes to various posterior eye segment diseases such as age-related macular degeneration and diabetic retinopathy. RNA nanoparticles were demonstrated previously to enter the corneal and retinal cells after subconjunctival injection for ocular delivery. In the present study, antiangiogenic aptamers (anti-vascular endothelial growth factor (VEGF) and anti-angiopoietin-2 (Ang2) aptamers) were conjugated to RNA nanoparticles. The objectives were to investigate the clearance and distribution of these angiogenesis-inhibiting RNA nanoparticles after subconjunctival injection in vivo and their antiangiogenic effects for inhibiting ocular neovascularization in vitro. The results in the whole-body fluorescence imaging study showed that the clearance of RNA nanoparticles was size-dependent with no significant differences between RNA nanoparticles with and without the aptamers except for pRNA-3WJ. The distribution study of RNA nanoparticles by confocal microscopy of the dissected eye tissues in vivo indicated cell internalization of the larger RNA nanoparticles in the retina and retinal pigment epithelium after subconjunctival injection, and the larger nanoparticles with aptamers showed higher levels of cell internalization than those without. In the cell proliferation assay in vitro, RNA nanoparticles with multiple aptamers had higher antiangiogenic effects. With both longer retention time and high antiangiogenic effect, SQR-VEGF-Ang2 could be a promising RNA nanoparticle for posterior eye delivery.


Subject(s)
Angiogenesis Inhibitors , Nanoparticles , RNA , Vascular Endothelial Growth Factor A , Animals , Nanoparticles/chemistry , Angiogenesis Inhibitors/administration & dosage , Angiogenesis Inhibitors/pharmacology , Angiogenesis Inhibitors/chemistry , RNA/administration & dosage , Aptamers, Nucleotide/administration & dosage , Aptamers, Nucleotide/chemistry , Humans , Angiopoietin-2 , Male , Mice , Conjunctiva/metabolism , Injections, Intraocular , Cell Proliferation/drug effects , Neovascularization, Pathologic/drug therapy , Human Umbilical Vein Endothelial Cells/drug effects , Retina/metabolism , Retina/drug effects , Drug Delivery Systems/methods , Mice, Inbred C57BL , Angiogenesis
5.
Cell ; 186(8): 1535-1540, 2023 04 13.
Article in English | MEDLINE | ID: mdl-37059063

ABSTRACT

Decades of oncologic clinical use have demonstrated that cancer immunotherapy provides unprecedented therapeutic benefits. Tragically, only a minority of patients respond to existing immunotherapies. RNA lipid nanoparticles have recently emerged as modular tools for immune stimulation. Here, we discuss advancements in RNA-based cancer immunotherapies and opportunities for improvement.


Subject(s)
Immunotherapy , Neoplasms , RNA , Humans , Neoplasms/therapy , RNA/administration & dosage
6.
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
8.
Int Immunopharmacol ; 101(Pt B): 108231, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34655852

ABSTRACT

The aim of this study was to evaluate self-replicating RNA lipid nanoparticles (saRNA LNPs) to neutralize SARS-CoV-2 variants delta (B.1.617 lineage) and alpha (B.1.1.7 lineage). Before immunization of mice with saRNA LNPs, we saw high expression of S-protein at both mRNA and protein levels after transfection of HEK293T/17 cells with saRNA LNPs. After oral immunization of BALB/c mice with 0.1 - 10 µg saRNA LNPs , a high quantity of SARS-CoV-2 specific IgG and IgA antibodies were seen with a dose-dependent pattern. Importantly, the ratio of IgG2a/IgG1 in serum of vaccinated mice showed Th1/Th2 skewing response. We also found that the secreted antibodies could neutralize SARS-CoV-2 variants delta (B.1.617 lineage) and alpha (B.1.1.7 lineage). Re-stimulated splenocytes of vaccinated mice showed high secretion of IFN-γ, IL-6, and TNF- α . The authors think that although the preclinical study confirmed the efficacy of saRNA LNPs against SARS-CoV-2, the actual efficacy and safety of the oral vaccine must be evaluated in clinical trials.


Subject(s)
COVID-19 Vaccines/administration & dosage , COVID-19/prevention & control , Liposomes/administration & dosage , Nanoparticles/administration & dosage , RNA/administration & dosage , SARS-CoV-2/immunology , Administration, Oral , Animals , Antibodies, Neutralizing/blood , Antibodies, Viral/blood , COVID-19/blood , COVID-19/immunology , Caco-2 Cells , Cytokines/blood , Cytokines/immunology , HEK293 Cells , Humans , Immunoglobulin A/blood , Immunoglobulin G/blood , Mice, Inbred BALB C , Neutralization Tests , Spike Glycoprotein, Coronavirus/genetics , Spike Glycoprotein, Coronavirus/immunology
9.
Oxid Med Cell Longev ; 2021: 4539453, 2021.
Article in English | MEDLINE | ID: mdl-34621464

ABSTRACT

Mesenchymal stem cells (MSCs) are the most exploited stem cells with multilineage differentiation potential and immunomodulatory properties. Numerous lines of findings have reported their successful applications in a multitude of inflammatory conditions and immune disorders. However, it is currently discovered that these effects are mainly mediated in a paracrine manner by MSC-exosomes. Moreover, MSC-exosomes have been implicated in a wide variety of biological responses including immunomodulation, oxidative stress, tumor progression, and tissue regeneration. Meanwhile, they are reported to actively participate in various hematological diseases by the means of transferring different types of exosomal components to the target cells. Therefore, in this review, we briefly discuss the sources and biological features of MSCs and then illustrate the biogenesis and biological processes of MSC-exosomes. Of note, this paper especially highlights the latest research progress of MSC-exosomes in hematological diseases.


Subject(s)
Cell- and Tissue-Based Therapy/methods , Cytokines/administration & dosage , Drug Delivery Systems/methods , Exosomes/immunology , Exosomes/metabolism , Hematologic Diseases/drug therapy , Immunomodulation , Mesenchymal Stem Cells/cytology , RNA/administration & dosage , Animals , Hematologic Diseases/immunology , Humans
10.
Adv Drug Deliv Rev ; 179: 113994, 2021 12.
Article in English | MEDLINE | ID: mdl-34619287

ABSTRACT

Traditional nanoparticle carriers such as liposomes, micelles, and polymeric vehicles improve drug delivery by protecting, stabilizing, and increasing the circulatory half-life of the encapsulated drugs. However, traditional drug delivery systems frequently suffer from poor drug loading and require an excess of carrier materials. This carrier material excess poses an additional systemic burden through accumulation, if not degradable the need for metabolism, and potential toxicity. To address these shortcomings, minimal-carrier nanoparticle systems and pharmacoactive carrier materials have been developed. Both solutions provide drug delivery systems in which the majority of the nanoparticle is pharmacologically active. While minimal-carrier and pharmacoactive drug delivery systems can improve drug loading, they can also suffer from poor stability. Here, we review minimal-carrier and pharmacoactive delivery systems, discuss ongoing challenges and outline opportunities to translate minimal-carrier and pharmacoactive drug delivery systems into the clinic.


Subject(s)
Nanoparticle Drug Delivery System/chemistry , Nanoparticle Drug Delivery System/therapeutic use , DNA/administration & dosage , Drug Carriers/therapeutic use , Drug Stability , Humans , Nanoparticle Drug Delivery System/administration & dosage , Particle Size , Prodrugs , Proteins/administration & dosage , RNA/administration & dosage
11.
Adv Drug Deliv Rev ; 179: 113999, 2021 12.
Article in English | MEDLINE | ID: mdl-34715258

ABSTRACT

Glioblastoma (GBM) is an aggressive central nervous system cancer with a dismal prognosis. The standard of care involves surgical resection followed by radiotherapy and chemotherapy, but five-year survival is only 5.6% despite these measures. Novel therapeutic approaches, such as immunotherapies, targeted therapies, and gene therapies, have been explored to attempt to extend survival for patients. Nanoparticles have been receiving increasing attention as promising vehicles for non-viral nucleic acid delivery in the context of GBM, though delivery is often limited by low blood-brain barrier permeability, particle instability, and low trafficking to target brain structures and cells. In this review, nanoparticle design considerations and new advances to overcome nucleic acid delivery challenges to treat brain cancer are summarized and discussed.


Subject(s)
Brain Neoplasms/drug therapy , Glioblastoma/drug therapy , Nanoparticle Drug Delivery System/chemistry , Nanoparticle Drug Delivery System/pharmacokinetics , RNA/administration & dosage , Antineoplastic Agents, Immunological/pharmacology , Biological Transport/physiology , Blood-Brain Barrier/metabolism , Drug Administration Routes , Drug Carriers , Drug Stability , Gene Transfer Techniques , Humans , MicroRNAs/administration & dosage , RNA, Messenger/administration & dosage , RNA, Small Interfering/administration & dosage
12.
Mol Pharm ; 18(8): 2924-2934, 2021 08 02.
Article in English | MEDLINE | ID: mdl-34212728

ABSTRACT

Therapeutic efficiency and toxicity are two of the three critical factors in molecular therapy and pharmaceutical drug development. Specific tumor targeting and rapid renal excretion contribute to improving efficiency and reducing toxicity. We recently found that RNA nanoparticles display rubber-like properties, enabling them to deliver therapeutics to cancer with high efficiency. Off-target RNA nanoparticles were rapidly cleared by renal excretion, resulting in nontoxicity. However, previous biodistribution studies relied mainly on fluorescent markers, which can cause interference from fluorophore quenching and autofluorescence. Thus, the quantification of biodistribution requires further scrutiny. In this study, radionuclide [3H] markers were used for quantitative pharmacokinetic (PK) studies to elucidate the favorable PK profile of RNA nanoparticles. Approximately 5% of [3H]-RNA nanoparticles accumulated in tumors, in contrast to the 0.7% tumor accumulation reported in the literature for other kinds of nanoparticles. The amount of [3H]-RNA nanoparticles accumulated in tumors was higher than that in the liver, heart, lung, spleen, and brain throughout the entire process after IV injection. [3H]-RNA nanoparticles rapidly reached the tumor vasculature within 30 min and remained in tumors for more than 2 days. Nontargeting [3H]-RNA nanoparticles were found in the urine 30 min after IV injection without degradation and processing, and more than 55% of the IV-injected radiolabeled RNA nanoparticles were cleared from the body within 12 h, while the other 45% includes the radiative counts that cannot be recovered due to whole-body distribution and blood dilution after intravenous injection. The high specificity of tumor targeting, fast renal excretion, and low organ accumulation illustrate the high therapeutic potential of RNA nanoparticles in cancer treatment as efficient cancer-targeting carriers with low toxicity and side effects.


Subject(s)
Breast Neoplasms/drug therapy , Nanoparticle Drug Delivery System/chemistry , Nanoparticles/chemistry , RNA/administration & dosage , RNA/pharmacokinetics , Tritium/administration & dosage , Animals , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Drug Stability , Female , Humans , Injections, Intravenous , Mice , Tissue Distribution , Xenograft Model Antitumor Assays
13.
Drug Deliv ; 28(1): 1548-1561, 2021 12.
Article in English | MEDLINE | ID: mdl-34286631

ABSTRACT

PURPOSE: Tumor RNA vaccines can activate dendritic cells to generate systemic anti-tumor immune response. However, due to easily degraded of RNA, direct RNA vaccine is less effective. In this study, we optimized the method for preparing PEGylated liposom-polycationic DNA complex (LPD) nanoliposomes, increased encapsulate amount of total RNA derived from CT-26 colorectal cancer cells. Tumor RNA LPD nanoliposomes vaccines improved anti-tumor immune response ability of tumor RNA and can effectively promote anti-tumor therapeutic effect of oxaliplatin. METHODS: Total tumor-derived RNA was extracted from colorectal cancer cells (CT-26 cells), and loaded to our optimized the LPD complex, resulting in the LPD nanoliposomes. We evaluated the characteristics (size, zeta potential, and stability), cytotoxicity, transfection ability, and tumor-growth inhibitory efficacy of LPD nanoliposomes. RESULTS: The improved LPD nanoliposomes exhibited a spherical shape, RNA loading efficiency of 9.07%, the average size of 120.37 ± 2.949 nm and zeta potential was 3.34 ± 0.056 mV. Also, the improved LPD nanoliposomes showed high stability at 4 °C, with a low toxicity and high cell transfection efficacy toward CT-26 colorectal cancer cells. Notably, the improved LPD nanoliposomes showed tumor growth inhibition by activating anti-tumor immune response in CT-26 colorectal cancer bearing mice, with mini side effects toward the normal organs of mice. Furthermore, the effect of the improved LPD nanoliposomes in combination with oxaliplatin can be better than that of oxaliplatin alone. CONCLUSION: The improved LPD nanoliposomes may serve as an effective vaccine to induce antitumor immunity, presenting a new treatment option for colorectal cancer.


Subject(s)
Antineoplastic Agents/pharmacology , Cancer Vaccines/administration & dosage , Colorectal Neoplasms/drug therapy , Nanoparticles/chemistry , Oxaliplatin/pharmacology , RNA/administration & dosage , Animals , CD8-Positive T-Lymphocytes/cytology , Cancer Vaccines/pharmacology , Cell Line, Tumor , Cell Survival , Chemistry, Pharmaceutical , Drug Carriers/chemistry , Drug Stability , Drug Therapy, Combination , Liposomes/chemistry , Male , Mice , Mice, Inbred BALB C , Particle Size , Polyethylene Glycols/chemistry , RNA/pharmacology , Surface Properties , Transfection
14.
Methods Mol Biol ; 2277: 49-67, 2021.
Article in English | MEDLINE | ID: mdl-34080144

ABSTRACT

Defects in human mitochondrial genome can cause a wide range of clinical disorders that still do not have efficient therapies. The natural pathway of small noncoding RNA import can be exploited to address therapeutic RNAs into the mitochondria. To create an approach of carrier-free targeting of RNA into living human cells, we designed conjugates containing a cholesterol residue and developed the protocols of chemical synthesis of oligoribonucleotides conjugated with cholesterol residue through cleavable pH-triggered hydrazone bond. The biodegradable conjugates of importable RNA with cholesterol can be internalized by cells in a carrier-free manner; RNA can then be released in the late endosomes due to a change in pH and partially targeted into mitochondria. Here we provide detailed protocols for solid-phase and "in solution" chemical synthesis of oligoribonucleotides conjugated to a cholesterol residue through a hydrazone bond. We describe the optimization of the carrier-free cell transfection with these conjugated RNA molecules and methods for evaluating the cellular and mitochondrial uptake of lipophilic conjugates.


Subject(s)
Mitochondria/genetics , Oligoribonucleotides/chemical synthesis , RNA/chemistry , Transfection/methods , Cells, Cultured , Cholesterol/chemistry , Humans , Hydrazones/chemistry , Hydrogen-Ion Concentration , RNA/administration & dosage
15.
Expert Opin Investig Drugs ; 30(7): 721-736, 2021 Jul.
Article in English | MEDLINE | ID: mdl-33966550

ABSTRACT

INTRODUCTION: Mood disorders are severe yet frequent psychiatric disorders worldwide, comprising major depressive disorder (MDD) and bipolar disorders (BD). Their treatment remains poorly effective. Recently, growing evidence for epigenetic mechanisms has emerged. Consequently, a great interest in a novel pharmacological class arose: RNA therapeutics. AREAS COVERED: We conducted a systematic review of RNA therapeutics -antisense oligonucleotides (ASOs), small interfering RNAs (siRNAs), short hairpin RNAs (shRNAs), and micro-RNA (miRNA) therapeutics- for the treatment of mood disorders studied in pre-clinical animal models listed in PubMed, in clinical trials registered in ClinicalTrials.gov and available on the market by combining literature search and Food and Drug Administration and European Medicine Agency online databases. Eighteen pre-clinical studies investigated the antidepressant effects of RNA therapeutics. However, even though there is an increasing number of marketing authorizations and clinical trials for the past twenty years, no RNA therapeutic has reached the clinical development pipeline for the treatment of psychiatric disorders yet. EXPERT OPINION: Several promising RNA therapeutics have been tested in pre-clinical studies for MDD, whereas no molecule has been developed for BD. There are several issues to address before reaching clinical development and new challenges include stratifying patient population and predicting therapeutic response.


Subject(s)
Bipolar Disorder/therapy , Depressive Disorder, Major/therapy , RNA/administration & dosage , Animals , Bipolar Disorder/genetics , Bipolar Disorder/physiopathology , Clinical Trials as Topic , Depressive Disorder, Major/genetics , Depressive Disorder, Major/physiopathology , Humans , Mood Disorders/genetics , Mood Disorders/physiopathology , Mood Disorders/therapy
16.
Adv Drug Deliv Rev ; 174: 250-264, 2021 07.
Article in English | MEDLINE | ID: mdl-33894328

ABSTRACT

RNA-based therapeutics are highly promising for the treatment of numerous diseases, by their ability to tackle the genetic origin in multiple possible ways. RNA molecules are, however, incapable of crossing cell membranes, hence a safe and efficient delivery vehicle is pivotal. Extracellular vesicles (EVs) are endogenously derived nano-sized particles and possess several characteristics which make them excellent candidates as therapeutic RNA delivery agent. This includes the inherent capability to functionally transfer RNAs in a selective manner and an enhanced safety profile compared to synthetic particles. Nonetheless, the fundamental mechanisms underlying this selective inter- and intracellular trafficking and functional transfer of RNAs by EVs are poorly understood. Improving our understanding of these systems is a key element of working towards an EV-based or EV-mimicking system for the functional delivery of therapeutic RNA. In this review, state-of-the-art approaches to detect and visualize RNA in situ and in live cells are discussed, as well as strategies to assess functional RNA transfer, highlighting their potential in studying EV-RNA trafficking mechanisms.


Subject(s)
Extracellular Vesicles/metabolism , Gene Transfer Techniques , RNA/administration & dosage , Animals , Biological Transport , Genetic Therapy/methods , Humans , Nanoparticles , RNA/metabolism
17.
Eur J Pharm Biopharm ; 163: 252-265, 2021 Jun.
Article in English | MEDLINE | ID: mdl-33745980

ABSTRACT

Lipid-based nanoparticles for RNA delivery (LNP-RNA) are revolutionizing the nanomedicine field, with one approved gene therapy formulation and two approved vaccines against COVID-19, as well as multiple ongoing clinical trials. As for other innovative nanopharmaceuticals (NPhs), the advancement of robust methods to assess their quality and safety profiles-in line with regulatory needs-is critical for facilitating their development and clinical translation. Asymmetric-flow field-flow fractionation coupled to multiple online optical detectors (MD-AF4) is considered a very versatile and robust approach for the physical characterisation of nanocarriers, and has been used successfully for measuring particle size, polydispersity and physical stability of lipid-based systems, including liposomes and solid lipid nanoparticles. However, the unique core structure of LNP-RNA, composed of ionizable lipids electrostatically complexed with RNA, and the relatively labile lipid-monolayer coating, is more prone to destabilization during focusing in MD-AF4 than previously characterised nanoparticles, resulting in particle aggregation and sample loss. Hence characterisation of LNP-RNA by MD-AF4 needs significant adaptation of the methods developed for liposomes. To improve the performance of MD-AF4 applied to LNP-RNA in a systematic and comprehensive manner, we have explored the use of the frit-inlet channel where, differently from the standard AF4 channel, the particles are relaxed hydrodynamically as they are injected. The absence of a focusing step minimizes contact between the particle and the membrane, reducing artefacts (e.g. sample loss, particle aggregation). Separation in a frit-inlet channel enables satisfactory reproducibility and acceptable sample recovery in the commercially available MD-AF4 instruments. In addition to slice-by-slice measurements of particle size, MD-AF4 also allows to determine particle concentration and the particle size distribution, demonstrating enhanced versatility beyond standard sizing measurements.


Subject(s)
Drug Carriers/chemistry , Lipids/chemistry , Nanoparticles/chemistry , RNA/administration & dosage , RNA/chemistry , Fractionation, Field Flow/methods , Humans , Nanomedicine/methods , Particle Size , Pharmaceutical Preparations/administration & dosage , Pharmaceutical Preparations/chemistry
18.
Mol Pharm ; 18(3): 1431-1443, 2021 03 01.
Article in English | MEDLINE | ID: mdl-33522823

ABSTRACT

MicroRNAs can regulate a variety of physiological and pathological processes and are increasingly recognized as being involved in regulating the malignant progression of cancer, which is an important direction for the study and treatment of cancer. In addition, the tumor microenvironment has gradually become an important direction of study for combating cancer. Researchers can inhibit tumor growth by remodeling and suppressing an immunosuppressive phenotype in the tumor microenvironment. Therefore, the combination of microRNA delivery and tumor microenvironment remodeling may be a potential research direction. In a previous study, we developed a novel cationic and hydrophilic antimicrobial peptide, DP7, by computer simulation. It was found that cholesterol-modified DP7 (DP7-C) has dual functions as a carrier and an immune adjuvant. In this experiment, we used DP7-C to deliver microRNAs or inhibitors intratumorally, where it played a dual role as a carrier and an immune adjuvant. As a delivery vector, DP7-C has more advantages in terms of transfection efficiency and cytotoxicity than Lipo2000 and PEI25K. Components of the DP7-C/RNA complex can effectively escape endosomes after uptake via caveolin- and clathrin-dependent pathways. As an immune adjuvant, DP7-C can activate dendritic cells and promote macrophage polarization. Moreover, it can transform the immunosuppressive tumor microenvironment into an immune-activated tumor microenvironment, indicating its potential as an anticancer therapy. In conclusion, this study identifies a novel microRNA and inhibitor delivery system that can remodel the tumor microenvironment and introduces an alternative scheme for antitumor treatment.


Subject(s)
Neoplasms/therapy , Peptides/administration & dosage , RNA/administration & dosage , Tumor Microenvironment/drug effects , Animals , Antineoplastic Agents/administration & dosage , Caveolins/genetics , Cell Line , Clathrin/genetics , Computer Simulation , Endosomes/drug effects , Female , HEK293 Cells , Human Umbilical Vein Endothelial Cells , Humans , Macrophages/drug effects , Mice , Mice, Inbred BALB C , MicroRNAs/genetics , Neoplasms/genetics
19.
Genes (Basel) ; 12(2)2021 01 27.
Article in English | MEDLINE | ID: mdl-33513776

ABSTRACT

Exosomes are packaged with a variety of cellular cargo including RNA, DNA, lipids and proteins. For several decades now there has been ongoing debate as to what extent exosomes are the garbage bin of the cell or if these entities function as a distributer of cellular cargo which acts in a meaningful mechanistic way on target cells. Are the contents of exosomes unwanted excess cellular produce or are they selective nucleic acid packaged nanoparticles used to communicate in a paracrine fashion? Overexpressed RNAs and fragments of DNA have been shown to collect into exosomes which are jettisoned from cells in response to particular stimuli to maintain homeostasis suggesting exosomes are functional trash bins of the cell. Other studies however have deciphered selective packaging of particular nucleic acids into exosomes. Nucleic acids packaged into exosomes are increasingly reported to exert transcriptional control on recipient cells, supporting the notion that exosomes may provide a role in signaling and intracellular communication. We survey the literature and conclude that exosomes are multifunctional entities, with a plethora of roles that can each be taken advantage to functionally modulate cells. We also note that the potential utility of developing exosomes as a next generation genetic therapy may in future transform cellular therapies. We also depict three models of methodologies which can be adopted by researchers intending to package nucleic acid in exosomes for developing gene and cell therapy.


Subject(s)
Cell- and Tissue-Based Therapy , Exosomes/metabolism , Genetic Therapy , Animals , Bioengineering/methods , Cell- and Tissue-Based Therapy/methods , Cell- and Tissue-Based Therapy/trends , Cell-Derived Microparticles/metabolism , Cell-Derived Microparticles/transplantation , DNA/administration & dosage , DNA/genetics , Drug Carriers , Exosomes/transplantation , Gene Transfer Techniques , Genetic Therapy/methods , Genetic Therapy/trends , Humans , Nanoparticles , RNA/administration & dosage , RNA/genetics
20.
J Mol Med (Berl) ; 98(12): 1767-1779, 2020 12.
Article in English | MEDLINE | ID: mdl-33146744

ABSTRACT

Advanced non-viral gene delivery experiments often require co-delivery of multiple nucleic acids. Therefore, the availability of reliable and robust co-transfection methods and defined selection criteria for their use in, e.g., expression of multimeric proteins or mixed RNA/DNA delivery is of utmost importance. Here, we investigated different co- and successive transfection approaches, with particular focus on in vitro transcribed messenger RNA (IVT-mRNA). Expression levels and patterns of two fluorescent protein reporters were determined, using different IVT-mRNA doses, carriers, and cell types. Quantitative parameters determining the efficiency of co-delivery were analyzed for IVT-mRNAs premixed before nanocarrier formation (integrated co-transfection) and when simultaneously transfecting cells with separately formed nanocarriers (parallel co-transfection), which resulted in a much higher level of expression heterogeneity for the two reporters. Successive delivery of mRNA revealed a lower transfection efficiency in the second transfection round. All these differences proved to be more pronounced for low mRNA doses. Concurrent delivery of siRNA with mRNA also indicated the highest co-transfection efficiency for integrated method. However, the maximum efficacy was shown for successive delivery, due to the kinetically different peak output for the two discretely operating entities. Our findings provide guidance for selection of the co-delivery method best suited to accommodate experimental requirements, highlighting in particular the nucleic acid dose-response dependence on co-delivery on the single-cell level.


Subject(s)
Gene Transfer Techniques , RNA/genetics , Cell Line , Cells, Cultured , Fluorescent Antibody Technique , Gene Expression , Humans , Macrophages/metabolism , Monocytes/metabolism , RNA/administration & dosage , RNA, Messenger/genetics , Transfection/methods
SELECTION OF CITATIONS
SEARCH DETAIL
...