1
|
Yasir Khan H, Tabassum S, Arjmand F. Preparation and characterization of ionic metal-based anticancer formulations of the type [bis-DACH tolfenamate metal {Co(II), Cu(II) and Zn(II)}] complexes: Binding studies with ct-DNA/tRNA, cleavage and cytotoxic activity against chemoresistant cancer cells. Inorganica Chim Acta 2023; 558:121725. [DOI: 10.1016/j.ica.2023.121725] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/12/2023]
|
2
|
Casadidio C, Hartman JEM, Mesquita B, Haegebaert R, Remaut K, Neumann M, Hak J, Censi R, Di Martino P, Hennink WE, Vermonden T. Effect of Polyplex Size on Penetration into Tumor Spheroids. Mol Pharm 2023; 20:5515-5531. [PMID: 37811785 PMCID: PMC10630948 DOI: 10.1021/acs.molpharmaceut.3c00397] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Revised: 09/27/2023] [Accepted: 09/27/2023] [Indexed: 10/10/2023]
Abstract
Ovarian cancer is one of the most lethal gynecological cancers in the world. In recent years, nucleic acid (NA)-based formulations have been shown to be promising treatments for ovarian cancer, including tumor nodules. However, gene therapy is not that far advanced in clinical reality due to unfavorable physicochemical properties of the NAs, such as high molecular weight, poor cellular uptake, rapid degradation by nucleases, etc. One of the strategies used to overcome these drawbacks is the complexation of anionic NAs via electrostatic interactions with cationic polymers, resulting in the formation of so-called polyplexes. In this work, the role of the size of pDNA and siRNA polyplexes on their penetration into ovarian-cancer-based tumor spheroids was investigated. For this, a methoxypoly(ethylene glycol) poly(2-(dimethylamino)ethyl methacrylate) (mPEG-pDMAEMA) diblock copolymer was synthesized as a polymeric carrier for NA binding and condensation with either plasmid DNA (pDNA) or short interfering RNA (siRNA). When prepared in HEPES buffer (10 mM, pH 7.4) at a nitrogen/phosphate (N/P) charge ratio of 5 and pDNA polyplexes were formed with a size of 162 ± 11 nm, while siRNA-based polyplexes displayed a size of 25 ± 2 nm. The polyplexes had a slightly positive zeta potential of +7-8 mV in the same buffer. SiRNA and pDNA polyplexes were tracked in vitro into tumor spheroids, resembling in vivo avascular ovarian tumor nodules. For this purpose, reproducible spheroids were obtained by coculturing ovarian carcinoma cells with primary mouse embryonic fibroblasts in different ratios (5:2, 1:1, and 2:5). Penetration studies revealed that after 24 h of incubation, siRNA polyplexes were able to penetrate deeper into the homospheroids (composed of only cancer cells) and heterospheroids (cancer cells cocultured with fibroblasts) compared to pDNA polyplexes which were mainly located in the rim. The penetration of the polyplexes was slowed when increasing the fraction of fibroblasts present in the spheroids. Furthermore, in the presence of serum siRNA polyplexes encoding for luciferase showed a high cellular uptake in 2D cells resulting in ∼50% silencing of luciferase expression. Taken together, these findings show that self-assembled small siRNA polyplexes have good potential as a platform to test ovarian tumor nodulus penetration..
Collapse
Affiliation(s)
- Cristina Casadidio
- Department
of Pharmaceutical Sciences, Division of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences (UIPS),
Utrecht University 99, 3508 TB Utrecht, The Netherlands
- School
of Pharmacy, Drug Delivery Division, University
of Camerino, CHiP Research Center, Via Madonna delle Carceri, 62032 Camerino, Macerata, Italy
| | - Jet E. M. Hartman
- Department
of Pharmaceutical Sciences, Division of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences (UIPS),
Utrecht University 99, 3508 TB Utrecht, The Netherlands
| | - Bárbara
S. Mesquita
- Department
of Pharmaceutical Sciences, Division of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences (UIPS),
Utrecht University 99, 3508 TB Utrecht, The Netherlands
| | - Ragna Haegebaert
- Laboratory
of General Biochemistry and Physical Pharmacy, Faculty of Pharmaceutical
Sciences, Ghent University, 9000 Ghent, Belgium
| | - Katrien Remaut
- Laboratory
of General Biochemistry and Physical Pharmacy, Faculty of Pharmaceutical
Sciences, Ghent University, 9000 Ghent, Belgium
| | - Myriam Neumann
- Department
of Pharmaceutical Sciences, Division of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences (UIPS),
Utrecht University 99, 3508 TB Utrecht, The Netherlands
| | - Jaimie Hak
- Department
of Pharmaceutical Sciences, Division of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences (UIPS),
Utrecht University 99, 3508 TB Utrecht, The Netherlands
| | - Roberta Censi
- School
of Pharmacy, Drug Delivery Division, University
of Camerino, CHiP Research Center, Via Madonna delle Carceri, 62032 Camerino, Macerata, Italy
- Recusol
Srl, Via del Bastione
16, 62032 Camerino, Macerata, Italy
| | - Piera Di Martino
- Department
of Pharmacy, “G. D’Annunzio”
University of Chieti and Pescara, Via dei Vestini 1, 66100 Chieti, Chieti, Italy
- Recusol
Srl, Via del Bastione
16, 62032 Camerino, Macerata, Italy
| | - Wim E. Hennink
- Department
of Pharmaceutical Sciences, Division of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences (UIPS),
Utrecht University 99, 3508 TB Utrecht, The Netherlands
| | - Tina Vermonden
- Department
of Pharmaceutical Sciences, Division of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences (UIPS),
Utrecht University 99, 3508 TB Utrecht, The Netherlands
| |
Collapse
|
3
|
Baptista B, Oliveira ASR, Mendonça P, Serra AC, Coelho JFJ, Sousa F. pH-responsive nanoparticles based on POEOMA-b-PDPA block copolymers for RNA encapsulation, protection and cell delivery. BIOMATERIALS ADVANCES 2023; 145:213267. [PMID: 36599197 DOI: 10.1016/j.bioadv.2022.213267] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Revised: 12/13/2022] [Accepted: 12/18/2022] [Indexed: 12/24/2022]
Abstract
The use of gene-based products, such as DNA or RNA, is increasingly being explored for various innovative therapies. However, the success of these strategies is highly dependent on the effective delivery of these biomolecules to target cells. Therefore, the development of pH-responsive nanoparticles comprises the creation of intelligent delivery systems with high therapeutic efficiency. In this work, the pH-responsiveness of the poly(2-(diisopropylamino)ethyl methacrylate)) (PDPA) block was investigated for the encapsulation and delivery of small RNAs (sRNA) to cancer cells. The pH responsiveness was dependent on the protonation profile of the tertiary amines of PDPA, which directly affected the electrostatic interactions established with RNA. Thus, block copolymers based on poly(oligo(ethylene oxide) methyl ether methacrylate) (POEOMA) and PDPA, POEOMA-b-PDPA, were synthesized by supplemental activator and reducing agent atom transfer radical polymerization (SARA ATRP). The structure of the block copolymers was characterized by size exclusion chromatography and 1H NMR spectroscopy. The copolymers allowed effective complexation of model sRNAs and a pre-miRNA with efficiencies of about 89 % and 91 %, respectively. The characterization by dynamic light scattering revealed that these systems had sizes between 76 and 1375 nm. It was also found that the morphology of the polyplexes depended on the pH, since the preparation at a pH lower than the pKa of the copolymers resulted in spherical but polydisperse particles, while higher pH values resulted in nanoparticles with more homogeneous size, but altered morphology. Moreover, due to pH-responsiveness, it was achieved the release of RNA at pH higher than the pKa of the copolymers, while maintaining its integrity. The polyplexes also showed a high potential to protect RNA from RNases. The transfection of a lung cancer model (A549) and fibroblast cell lines showed that these polyplexes did not cause cell toxicity. In addition, the polyplexes enabled the effective transfection of the A549 cell line with pre-miRNA-29b and miRNA-29b, resulting in a decrease of expression levels of the target DNMT3B gene by approximately 51 % and 47 %, respectively. Overall, the POEOMA-b-PDPA copolymers proved to be a promising strategy for developing responsive delivery systems, that can play a critical role in some diseases, such as cancer, where pH varies between the intra and extracellular environments.
Collapse
Affiliation(s)
- Bruno Baptista
- CICS-UBI - Health Sciences Research Centre, University of Beira Interior, Av. Infante D. Henrique, 6200-506 Covilhã, Portugal
| | - Andreia S R Oliveira
- University of Coimbra, Centre for Mechanical Engineering, Materials and Processes, Department of Chemical Engineering, Rua Sílvio Lima-Polo II, 3030-790 Coimbra, Portugal
| | - Patrícia Mendonça
- University of Coimbra, Centre for Mechanical Engineering, Materials and Processes, Department of Chemical Engineering, Rua Sílvio Lima-Polo II, 3030-790 Coimbra, Portugal
| | - Arménio C Serra
- University of Coimbra, Centre for Mechanical Engineering, Materials and Processes, Department of Chemical Engineering, Rua Sílvio Lima-Polo II, 3030-790 Coimbra, Portugal
| | - Jorge F J Coelho
- University of Coimbra, Centre for Mechanical Engineering, Materials and Processes, Department of Chemical Engineering, Rua Sílvio Lima-Polo II, 3030-790 Coimbra, Portugal
| | - Fani Sousa
- CICS-UBI - Health Sciences Research Centre, University of Beira Interior, Av. Infante D. Henrique, 6200-506 Covilhã, Portugal.
| |
Collapse
|
4
|
Coelho F, Salonen LM, Silva BFB. Hemiacetal-linked pH-sensitive PEG-lipids for non-viral gene delivery. NEW J CHEM 2022. [DOI: 10.1039/d2nj02217f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Cationic lipid–DNA complexes containing a novel hemiacetal PEG-lipid for endosomal escape were characterized in terms of pH-response, stability, and biological activity.
Collapse
Affiliation(s)
- Filipe Coelho
- International Iberian Nanotechnology Laboratory (INL), Av. Mestre José Veiga, 4715-330 Braga, Portugal
| | - Laura M. Salonen
- International Iberian Nanotechnology Laboratory (INL), Av. Mestre José Veiga, 4715-330 Braga, Portugal
| | - Bruno F. B. Silva
- International Iberian Nanotechnology Laboratory (INL), Av. Mestre José Veiga, 4715-330 Braga, Portugal
| |
Collapse
|
5
|
Coelho F, Botelho C, Paris JL, Marques EF, Silva BF. Influence of the media ionic strength on the formation and in vitro biological performance of polycation-DNA complexes. J Mol Liq 2021. [DOI: 10.1016/j.molliq.2021.117930] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
|
6
|
Vasiliu T, Craciun BF, Neamtu A, Clima L, Isac DL, Maier SS, Pinteala M, Mocci F, Laaksonen A. In silico study of PEI-PEG-squalene-dsDNA polyplex formation: the delicate role of the PEG length in the binding of PEI to DNA. Biomater Sci 2021; 9:6623-6640. [PMID: 34582532 DOI: 10.1039/d1bm00973g] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Biocompatible hydrophilic polyethylene glycol (PEG) is widely used in biomedical applications, such as drug or gene delivery, tissue engineering or as an antifouling component in biomedical devices. Experimental studies have shown that the size of PEG can weaken polycation-polyanion interactions, like those between branched polyethyleneimine (b-PEI) and DNA in gene carriers, but details of its cause and underlying interactions on the atomic scale are still not clear. To better understand the interaction mechanisms in the formation of polyplexes between b-PEI-PEG based carriers and DNA, we have used a combination of in silico tools and experiments on three multicomponent systems differing in PEG MW. Using the PEI-PEG-squalene-dsDNA systems of the same size, both in the all-atom MD simulations and in experimental in-gel electrophoresis measurements, we found that the binding between DNA and the vectors is highly influenced by the size of PEG, with the binding efficiency increasing with a shorter PEG length. The mechanism of how PEG interferes with the binding between PEI and DNA is explained using a two-step MD simulation protocol that showed that the DNA-vector interactions are influenced by the PEG length due to the hydrogen bond formation between PEI and PEG. Although computationally demanding we find it important to study molecular systems of the same size both in silico and in a laboratory and to simulate the behaviour of the carrier prior to the addition of bioactive molecules to understand the molecular mechanisms involved in the formation of the polyplex.
Collapse
Affiliation(s)
- Tudor Vasiliu
- Center of Advanced Research in Bionanoconjugates and Biopolymers, "Petru Poni" Institute of Macromolecular Chemistry, Iasi 700487, Romania.
| | - Bogdan Florin Craciun
- Center of Advanced Research in Bionanoconjugates and Biopolymers, "Petru Poni" Institute of Macromolecular Chemistry, Iasi 700487, Romania.
| | - Andrei Neamtu
- Bioinformatics Laboratory, TRANSCEND IRO, Iaşi 700843, Romania
| | - Lilia Clima
- Center of Advanced Research in Bionanoconjugates and Biopolymers, "Petru Poni" Institute of Macromolecular Chemistry, Iasi 700487, Romania.
| | - Dragos Lucian Isac
- Center of Advanced Research in Bionanoconjugates and Biopolymers, "Petru Poni" Institute of Macromolecular Chemistry, Iasi 700487, Romania.
| | - Stelian S Maier
- Center of Advanced Research in Bionanoconjugates and Biopolymers, "Petru Poni" Institute of Macromolecular Chemistry, Iasi 700487, Romania. .,Polymers Research Center, "Gheorghe Asachi" Technical University of Iasi, Iasi, 700487, Romania
| | - Mariana Pinteala
- Center of Advanced Research in Bionanoconjugates and Biopolymers, "Petru Poni" Institute of Macromolecular Chemistry, Iasi 700487, Romania.
| | - Francesca Mocci
- Center of Advanced Research in Bionanoconjugates and Biopolymers, "Petru Poni" Institute of Macromolecular Chemistry, Iasi 700487, Romania. .,Dipartimento di Scienze Chimiche e Geologiche, Università di Cagliari, Monserrato, 09042 Cagliari, Italy
| | - Aatto Laaksonen
- Center of Advanced Research in Bionanoconjugates and Biopolymers, "Petru Poni" Institute of Macromolecular Chemistry, Iasi 700487, Romania. .,Department of Materials and Environmental Chemistry, Division of Physical Chemistry, Arrhenius Laboratory, Stockholm University, 106 91 Stockholm, Sweden.,State Key Laboratory of Materials-Oriented and Chemical Engineering, Nanjing Tech University, 210009 Nanjing, PR China.,Department of Engineering Sciences and Mathematics, Division of Energy Science, Luleå University of Technology, 97187 Luleå, Sweden
| |
Collapse
|
7
|
Fundamental and Practical Aspects in the Formulation of Colloidal Polyelectrolyte Complexes of Chitosan and siRNA. Methods Mol Biol 2021. [PMID: 33928582 DOI: 10.1007/978-1-0716-1298-9_17] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register]
Abstract
The formation of electrostatic interactions between polyanionic siRNA and polycations gives an easy access to the formation of colloidal particles capable of delivering siRNA in vitro or in vivo. Among the polycations used for siRNA delivery, chitosan occupies a special place due to its unique physicochemical and biological properties. In this chapter we describe the fundamental and practical aspects of the formation of colloidal complexes between chitosan and siRNA. The basis of the electrostatic complexation between oppositely charged polyelectrolytes is first introduced with a focus on the specific conditions to obtain stable colloid complex particles. Subsequent, the properties that make chitosan so special are described. In a third part, the main parameters influencing the colloidal properties and stability of siRNA/chitosan complexes are reviewed with emphasis on some practical aspects to consider in the preparation of complexes.
Collapse
|
8
|
Brain-Targeted Delivery of Pre-miR-29b Using Lactoferrin-Stearic Acid-Modified-Chitosan/Polyethyleneimine Polyplexes. Pharmaceuticals (Basel) 2020; 13:ph13100314. [PMID: 33076502 PMCID: PMC7602608 DOI: 10.3390/ph13100314] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Revised: 10/03/2020] [Accepted: 10/12/2020] [Indexed: 02/07/2023] Open
Abstract
The efficacy of brain therapeutics is largely hampered by the presence of the blood–brain barrier (BBB), mainly due to the failure of most (bio) pharmaceuticals to cross it. Accordingly, this study aims to develop nanocarriers for targeted delivery of recombinant precursor microRNA (pre-miR-29b), foreseeing a decrease in the expression of the BACE1 protein, with potential implications in Alzheimer’s disease (AD) treatment. Stearic acid (SA) and lactoferrin (Lf) were successfully exploited as brain-targeting ligands to modify cationic polymers (chitosan (CS) or polyethyleneimine (PEI)), and its BBB penetration behavior was evaluated. The intracellular uptake of the dual-targeting drug delivery systems by neuronal cell models, as well as the gene silencing efficiency of recombinant pre-miR-29b, was analyzed in vitro. Labeled pre-miR-29b-CS/PEI-SA-Lf systems showed very strong fluorescence in the cytoplasm and nucleus of RBE4 cells, being verified the delivery of pre-miR-29b to neuronal cells after 1 h transfection. The experiment of transport across the BBB showed that CS-SA-Lf delivered 65% of recombinant pre-miR-29b in a period of 4 h, a significantly higher transport ratio than the 42% found for PEI-SA-Lf in the same time frame. Overall, a novel procedure for the dual targeting of DDS is disclosed, opening new perspectives in nanomedicines delivery, whereby a novel drug delivery system harvests the merits and properties of the different immobilized ligands.
Collapse
|
9
|
miR-29b and retinoic acid co-delivery: a promising tool to induce a synergistic antitumoral effect in non-small cell lung cancer cells. Drug Deliv Transl Res 2020; 10:1367-1380. [PMID: 32358723 DOI: 10.1007/s13346-020-00768-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
The high incidence, late diagnosis, and aggressive profile of lung cancer limit the treatment options, causing a reduced survival rate. Consequently, RNAi-based therapy appears as a potential approach to treat non-small cell lung cancer (NSCLC). This approach is based on the delivery of small RNAs, involved in the regulation of key cell pathways, to treat complex diseases among others. Concerning that, the aim of this work was focused on the co-delivery of miR-29b and retinoic acid (RA) into NSCLC cells by multifunctional micellar nanosystems (Pluronic® P123 or Pluronic® P103 linked to polyethyleneimine (PEI)). The developed P103-PEI-RA/miR-29b (10/1) presented better results and most attractive properties, promoting efficient delivery of miR-29b, as well as revealing a significant antitumoral activity promoted by a synergistic effect between miR-29b expression and RA deliver. Furthermore, the developed therapeutic approach was able to significantly decrease cell viability and migration, as well as induce cell cycle arrest and epigenetic regulation in NSCLC cells. Thus, this work outcome enables to discover a hopeful system to deliver therapeutic miRNAs, crafting a novel RNAi-based therapy combined with RA to treat NSCLC. Graphical abstract.
Collapse
|
10
|
Blakney AK, McKay PF, Yus BI, Aldon Y, Shattock RJ. Inside out: optimization of lipid nanoparticle formulations for exterior complexation and in vivo delivery of saRNA. Gene Ther 2019; 26:363-372. [PMID: 31300730 PMCID: PMC6760535 DOI: 10.1038/s41434-019-0095-2] [Citation(s) in RCA: 146] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2019] [Revised: 05/18/2019] [Accepted: 06/28/2019] [Indexed: 12/17/2022]
Abstract
Self-amplifying RNA (saRNA) is a promising biotherapeutic tool that has been used as a vaccine against both infectious diseases and cancer. saRNA has been shown to induce protein expression for up to 60 days and elicit immune responses with lower dosing than messenger RNA (mRNA). Because saRNA is a large (~9500 nt), negatively charged molecule, it requires a delivery vehicle for efficient cellular uptake and degradation protection. Lipid nanoparticles (LNPs) have been widely used for RNA formulations, where the prevailing paradigm is to encapsulate RNA within the particle, including the first FDA-approved small-interfering siRNA therapy. Here, we compared LNP formulations with cationic and ionizable lipids with saRNA either on the interior or exterior of the particle. We show that LNPs formulated with cationic lipids protect saRNA from RNAse degradation, even when it is adsorbed to the surface. Furthermore, cationic LNPs deliver saRNA equivalently to particles formulated with saRNA encapsulated in an ionizable lipid particle, both in vitro and in vivo. Finally, we show that cationic and ionizable LNP formulations induce equivalent antibodies against HIV-1 Env gp140 as a model antigen. These studies establish formulating saRNA on the surface of cationic LNPs as an alternative to the paradigm of encapsulating RNA.
Collapse
Affiliation(s)
- Anna K Blakney
- Department of Medicine, Imperial College London, London, UK
| | - Paul F McKay
- Department of Medicine, Imperial College London, London, UK
| | | | - Yoann Aldon
- Department of Medicine, Imperial College London, London, UK
| | | |
Collapse
|
11
|
Domingues CSDC, Serambeque BP, Laranjo Cândido MS, Marto CMM, Veiga FJDB, Sarmento Antunes Cruz Ribeiro AB, Figueiras ARR, Botelho MFR, Dourado MDARF. Epithelial-mesenchymal transition and microRNAs: Challenges and future perspectives in oral cancer. Head Neck 2018; 40:2304-2313. [PMID: 30120853 DOI: 10.1002/hed.25381] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2017] [Revised: 04/23/2018] [Accepted: 05/28/2018] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND Head and neck cancer is the sixth most common cancer worldwide, with oral squamous cell carcinoma (OSCC) being the most representative type. OSCC is a public health problem with high morbidity and poor survival rate. Epithelial-mesenchymal transition is emerging as a hallmark in OSCC. METHODS In this study, we described the role of microRNAs in epithelial-mesenchymal transition regulation in OSCC based on a PubMed search using articles published in English between January 1, 2010, and January 31, 2018. RESULTS MicroRNA's regulatory networks seem to be a hallmark of epithelial-mesenchymal transition in OSCC pathophysiology becoming a growing challenge to design new studies and strategies from biology to clinical applications. CONCLUSION Therefore, we propose that targeting therapies to epithelial-mesenchymal transition-type cells, namely, coordinating microRNAs and/or hydrophobic drugs, such as conventional therapy, could be a promising strategy to improve the outcomes of patients with OSCC.
Collapse
Affiliation(s)
- Cátia Sofia da Costa Domingues
- Faculty of Pharmacy, University of Coimbra, Coimbra, Portugal.,REQUIMTE/LAQV, Group of Pharmaceutical Technology, Faculty of Pharmacy, University of Coimbra, Coimbra, Portugal.,CIMAGO, Center of Investigation on Environment Genetics and Oncobiology, Faculty of Medicine, University of Coimbra, Coimbra, Portugal.,Laboratory of Oncobiology and Hematology (LOH) and University Clinic of Hematology, Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| | - Beatriz Prazeres Serambeque
- CIMAGO, Center of Investigation on Environment Genetics and Oncobiology, Faculty of Medicine, University of Coimbra, Coimbra, Portugal.,Biophysics Institute, Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| | - Mafalda Sofia Laranjo Cândido
- CIMAGO, Center of Investigation on Environment Genetics and Oncobiology, Faculty of Medicine, University of Coimbra, Coimbra, Portugal.,Biophysics Institute, Faculty of Medicine, University of Coimbra, Coimbra, Portugal.,CNC/IBILI, Center for Neuroscience and Cell Biology, Institute for Biomedical Imaging and Life Sciences, University of Coimbra, Coimbra, Portugal
| | - Carlos Miguel Machado Marto
- CIMAGO, Center of Investigation on Environment Genetics and Oncobiology, Faculty of Medicine, University of Coimbra, Coimbra, Portugal.,Biophysics Institute, Faculty of Medicine, University of Coimbra, Coimbra, Portugal.,CNC/IBILI, Center for Neuroscience and Cell Biology, Institute for Biomedical Imaging and Life Sciences, University of Coimbra, Coimbra, Portugal.,Experimental Pathology Institute, Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| | - Francisco José de Baptista Veiga
- Faculty of Pharmacy, University of Coimbra, Coimbra, Portugal.,REQUIMTE/LAQV, Group of Pharmaceutical Technology, Faculty of Pharmacy, University of Coimbra, Coimbra, Portugal.,CNC/IBILI, Center for Neuroscience and Cell Biology, Institute for Biomedical Imaging and Life Sciences, University of Coimbra, Coimbra, Portugal
| | - Ana Bela Sarmento Antunes Cruz Ribeiro
- CIMAGO, Center of Investigation on Environment Genetics and Oncobiology, Faculty of Medicine, University of Coimbra, Coimbra, Portugal.,Laboratory of Oncobiology and Hematology (LOH) and University Clinic of Hematology, Faculty of Medicine, University of Coimbra, Coimbra, Portugal.,CNC/IBILI, Center for Neuroscience and Cell Biology, Institute for Biomedical Imaging and Life Sciences, University of Coimbra, Coimbra, Portugal
| | - Ana Rita Ramalho Figueiras
- Faculty of Pharmacy, University of Coimbra, Coimbra, Portugal.,REQUIMTE/LAQV, Group of Pharmaceutical Technology, Faculty of Pharmacy, University of Coimbra, Coimbra, Portugal
| | - Maria Filomena Roque Botelho
- CIMAGO, Center of Investigation on Environment Genetics and Oncobiology, Faculty of Medicine, University of Coimbra, Coimbra, Portugal.,Biophysics Institute, Faculty of Medicine, University of Coimbra, Coimbra, Portugal.,CNC/IBILI, Center for Neuroscience and Cell Biology, Institute for Biomedical Imaging and Life Sciences, University of Coimbra, Coimbra, Portugal
| | - Marília de Assunção Rodrigues Ferreira Dourado
- CIMAGO, Center of Investigation on Environment Genetics and Oncobiology, Faculty of Medicine, University of Coimbra, Coimbra, Portugal.,Laboratory of Oncobiology and Hematology (LOH) and University Clinic of Hematology, Faculty of Medicine, University of Coimbra, Coimbra, Portugal.,Pathophysiology Course Unit, Dentistry Area, Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| |
Collapse
|
12
|
miR-145-loaded micelleplexes as a novel therapeutic strategy to inhibit proliferation and migration of osteosarcoma cells. Eur J Pharm Sci 2018; 123:28-42. [PMID: 30010029 DOI: 10.1016/j.ejps.2018.07.021] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2018] [Revised: 06/12/2018] [Accepted: 07/09/2018] [Indexed: 12/20/2022]
Abstract
Osteosarcoma (OS), the main primary malignancy of bone, is the second leading cause of cancer in children and young adults. Despite the advances in modern treatments, the 5-year survival rate is retained in 60-70%, since the conventional treatment options available are associated with relapse, chemoresistance, and development of metastases, which frequently lead to patients death. In this regard, there is an increasing need to search and develop novel and alternative therapeutic approaches. Concerning this, gene therapy appears as an innovative and promising treatment option. This therapeutic option aims to deliver genetic material, through nanosystems, to repress or replace the expression of mutated genes involved in important regulatory pathways. To attain this goal, gene therapy is decidedly dependent on the efficiency of utilized vectors, constituting such a very important parameter to take in consideration. In this work, the main goal was centered on the development and full characterization of an efficient micellar nanosystem, based on the chemical conjugation between the amphiphilic copolymer Pluronic® L64 and the cationic polymer polyethyleneimine (PEI), to deliver the therapeutic miRNA-145 into OS cells leading to inhibition of cell proliferation and migration, and ultimately inducing cell death, crafting a novel anticancer therapeutic approach to OS.
Collapse
|
13
|
Grijalvo S, Alagia A, Jorge AF, Eritja R. Covalent Strategies for Targeting Messenger and Non-Coding RNAs: An Updated Review on siRNA, miRNA and antimiR Conjugates. Genes (Basel) 2018; 9:E74. [PMID: 29415514 PMCID: PMC5852570 DOI: 10.3390/genes9020074] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2018] [Revised: 01/26/2018] [Accepted: 01/26/2018] [Indexed: 12/11/2022] Open
Abstract
Oligonucleotide-based therapy has become an alternative to classical approaches in the search of novel therapeutics involving gene-related diseases. Several mechanisms have been described in which demonstrate the pivotal role of oligonucleotide for modulating gene expression. Antisense oligonucleotides (ASOs) and more recently siRNAs and miRNAs have made important contributions either in reducing aberrant protein levels by sequence-specific targeting messenger RNAs (mRNAs) or restoring the anomalous levels of non-coding RNAs (ncRNAs) that are involved in a good number of diseases including cancer. In addition to formulation approaches which have contributed to accelerate the presence of ASOs, siRNAs and miRNAs in clinical trials; the covalent linkage between non-viral vectors and nucleic acids has also added value and opened new perspectives to the development of promising nucleic acid-based therapeutics. This review article is mainly focused on the strategies carried out for covalently modifying siRNA and miRNA molecules. Examples involving cell-penetrating peptides (CPPs), carbohydrates, polymers, lipids and aptamers are discussed for the synthesis of siRNA conjugates whereas in the case of miRNA-based drugs, this review article makes special emphasis in using antagomiRs, locked nucleic acids (LNAs), peptide nucleic acids (PNAs) as well as nanoparticles. The biomedical applications of siRNA and miRNA conjugates are also discussed.
Collapse
Affiliation(s)
- Santiago Grijalvo
- Institute of Advanced Chemistry of Catalonia (IQAC, CSIC), Jordi Girona 18-26, 08034 Barcelona, Spain.
- Networking Centre in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Jordi Girona 18-26, 08034 Barcelona, Spain.
| | - Adele Alagia
- Networking Centre in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Jordi Girona 18-26, 08034 Barcelona, Spain.
| | - Andreia F Jorge
- Coimbra Chemistry Centre, (CQC), Department of Chemistry, University of Coimbra, Rua Larga, 3004-535 Coimbra, Portugal.
| | - Ramon Eritja
- Institute of Advanced Chemistry of Catalonia (IQAC, CSIC), Jordi Girona 18-26, 08034 Barcelona, Spain.
- Networking Centre in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Jordi Girona 18-26, 08034 Barcelona, Spain.
| |
Collapse
|
14
|
Yang Z, Sun Y, Xian L, Xun Z, Yu J, Yang T, Zhao X, Cai C, Wang D, Ding P. Disulfide‐bond‐containing agamatine‐cystaminebisacrylamide polymer demonstrates better transfection efficiency and lower cytotoxicity than polyethylenimine in NIH/3T3 cells. J Cell Biochem 2017; 119:1767-1779. [DOI: 10.1002/jcb.26338] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2017] [Accepted: 08/08/2017] [Indexed: 01/01/2023]
Affiliation(s)
- Zhen Yang
- School of PharmacyShenyang Pharmaceutical UniversityShenyangChina
| | - Yanping Sun
- School of PharmacyShenyang Pharmaceutical UniversityShenyangChina
| | - Lei Xian
- School of PharmacyShenyang Pharmaceutical UniversityShenyangChina
| | - Zhe Xun
- Institute of Metabolic Disease Research and Drug DevelopmentChina Medical UniversityShenyangChina
| | - Jiankun Yu
- Department of Ion Channel PharmacologySchool of PharmacyChina Medical UniversityShenyangChina
| | - Tianzhi Yang
- Department of Basic Pharmaceutical SciencesSchool of PharmacyHusson UniversityBangorMaine
| | - Xiaoyun Zhao
- Department of Microbiology and Cell BiologySchool of Life Science and BiopharmaceuticsShenyang Pharmaceutical UniversityShenyangChina
| | - Cuifang Cai
- School of PharmacyShenyang Pharmaceutical UniversityShenyangChina
| | - Dongkai Wang
- School of PharmacyShenyang Pharmaceutical UniversityShenyangChina
| | - Pingtian Ding
- School of PharmacyShenyang Pharmaceutical UniversityShenyangChina
| |
Collapse
|
15
|
Pereira P, Pedro AQ, Queiroz JA, Figueiras AR, Sousa F. New insights for therapeutic recombinant human miRNAs heterologous production: Rhodovolum sulfidophilum vs Escherichia coli. Bioengineered 2017; 8:670-677. [PMID: 28282262 DOI: 10.1080/21655979.2017.1284710] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
RNA interference-based technologies have emerged as an attractive and effective therapeutic option with potential application in diverse human diseases. These tools rely on the development of efficient strategies to obtain homogeneous non-coding RNA samples with adequate integrity and purity, thus avoiding non-targeted gene-silencing and related side-effects that impair their application onto pre-clinical practice. These RNAs have been preferentially obtained by in vitro transcription using DNA templates or via chemical synthesis. As an alternative to overcome the limitations presented by these methods, in vivo recombinant production of RNA biomolecules has become the focus in RNA synthesis research. Therefore, using pre-miR-29b as a model, here it is evaluated the time-course profile of Escherichia coli and Rhodovolum sulfidophilum microfactories to produce this microRNA. As the presence of major host contaminants arising from the biosynthesis process may have important implications in the subsequent downstream processing, it is also evaluated the production of genomic DNA and host total proteins. Considering the rapidly growing interest on these innovative biopharmaceuticals, novel, more cost-effective, simple and easily scaled-up technologies are highly desirable. As microRNA recombinant expression fulfills those requirements, it may take the leading edge in the methodologies currently available to obtain microRNAs for clinical or structural studies.
Collapse
Affiliation(s)
- Patrícia Pereira
- a CICS-UBI - Health Sciences Research Centre, Universidade da Beira Interior, Avenida Infante D. Henrique , Covilhã , Portugal
| | - Augusto Q Pedro
- a CICS-UBI - Health Sciences Research Centre, Universidade da Beira Interior, Avenida Infante D. Henrique , Covilhã , Portugal
| | - João A Queiroz
- a CICS-UBI - Health Sciences Research Centre, Universidade da Beira Interior, Avenida Infante D. Henrique , Covilhã , Portugal
| | - Ana R Figueiras
- b Faculty of Pharmacy, University of Coimbra, Azinhaga Sta. Comba , Coimbra , Portugal.,c REQUIMTE/LAQV, Group of Pharmaceutical Technology, Faculty of Pharmacy, University of Coimbra , Coimbra , Portugal
| | - Fani Sousa
- a CICS-UBI - Health Sciences Research Centre, Universidade da Beira Interior, Avenida Infante D. Henrique , Covilhã , Portugal
| |
Collapse
|
16
|
Timin AS, Muslimov AR, Petrova AV, Lepik KV, Okilova MV, Vasin AV, Afanasyev BV, Sukhorukov GB. Hybrid inorganic-organic capsules for efficient intracellular delivery of novel siRNAs against influenza A (H1N1) virus infection. Sci Rep 2017; 7:102. [PMID: 28273907 PMCID: PMC5427965 DOI: 10.1038/s41598-017-00200-0] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2016] [Accepted: 02/14/2017] [Indexed: 12/17/2022] Open
Abstract
The implementation of RNAi technology into the clinical practice has been significantly postponing due to the issues regarding to the delivery of naked siRNA predominantly to target cells. Here we report the approach to enhance the efficiency of siRNA delivery by encapsulating the siRNA into new carrier systems which are obtained via the combination of widely used layer-by-layer technique and in situ modification by sol-gel chemistry. We used three types of siRNAs (NP-717, NP-1155 and NP-1496) in encapsulated form as new therapeutic agents against H1N1 influenza virus infection. By employing the hybrid microcontainers for the siRNA encapsulation we demonstrate the reduction of viral nucleoprotein (NP) level and inhibition of influenza virus production in infected cell lines (MDCK and A549). The obtained hybrid carriers based on assembled biodegradable polyelectrolytes and sol-gel coating possess several advantages such as a high cell uptake efficiency, low toxicity, efficient intracellular delivery of siRNAs and the protection of siRNAs from premature degradation before reaching the target cells. These findings underpin a great potential of versatile microencapsulation technology for the development of anti-viral RNAi delivery systems against influenza virus infection.
Collapse
Affiliation(s)
- Alexander S Timin
- RASA center in Tomsk, Tomsk Polytechnic University, Lenin Avenue, 30, 634050, Tomsk, Russian Federation.
| | - Albert R Muslimov
- First I. P. Pavlov State Medical University of St. Petersburg, Lev Tolstoy str., 6/8, 197022, Saint-Petersburg, Russian Federation
- RASA center in St. Petersburg, Peter The Great St. Petersburg Polytechnic University, Polytechnicheskaya, 29, 195251, St. Petersburg, Russian Federation
| | - Aleksandra V Petrova
- Research Institute of Influenza, Popova str., 15/17, 197376, Saint-Petersburg, Russian Federation
| | - Kirill V Lepik
- First I. P. Pavlov State Medical University of St. Petersburg, Lev Tolstoy str., 6/8, 197022, Saint-Petersburg, Russian Federation
| | - Maria V Okilova
- First I. P. Pavlov State Medical University of St. Petersburg, Lev Tolstoy str., 6/8, 197022, Saint-Petersburg, Russian Federation
| | - Andrey V Vasin
- Research Institute of Influenza, Popova str., 15/17, 197376, Saint-Petersburg, Russian Federation
- Department of Molecular Biology, Peter The Great St. Petersburg Polytechnic University, Polytechnicheskaya, 29, 195251, St. Petersburg, Russian Federation
| | - Boris V Afanasyev
- First I. P. Pavlov State Medical University of St. Petersburg, Lev Tolstoy str., 6/8, 197022, Saint-Petersburg, Russian Federation
| | - Gleb B Sukhorukov
- School of Engineering and Materials Science, Queen Mary University of London, Mile End Road, E1 4NS, London, UK.
| |
Collapse
|
17
|
Ziebarth JD, Kennetz DR, Walker NJ, Wang Y. Structural Comparisons of PEI/DNA and PEI/siRNA Complexes Revealed with Molecular Dynamics Simulations. J Phys Chem B 2017; 121:1941-1952. [PMID: 28145711 PMCID: PMC5677264 DOI: 10.1021/acs.jpcb.6b10775] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Polyplexes composed of polyethyleneimine (PEI) and DNA or siRNA have attracted great attention for their use in gene therapy. Although many physicochemical characteristics of these polyplexes remain unknown, PEI/DNA complexes have been repeatedly shown to be more stable than their PEI/siRNA counterparts. Here, we examine potential causes for this difference using atomistic molecular dynamics simulations of complexation between linear PEI and DNA or siRNA duplexes containing the same number of bases. The two types of polyplexes are stabilized by similar interactions, as PEI amines primarily interact with nucleic acid phosphate groups but also occasionally interact with groove atoms of both nucleic acids. However, the number of interactions in PEI/DNA complexes is greater than in comparable PEI/siRNA complexes, with interactions between protonated PEI amines and DNA being particularly enhanced. These results indicate that structural differences between DNA and siRNA may play a role in the increased stability of PEI/DNA complexes. In addition, we investigate the binding of PEI chains to polyplexes that have a net positive charge. The binding of PEI to these overcharged complexes involves interactions between PEI and areas on the nucleic acid surface that have maintained a negative electrostatic potential and is facilitated by the release of water from the nucleic acid.
Collapse
Affiliation(s)
- Jesse D Ziebarth
- Department of Chemistry, The University of Memphis , Memphis, Tennessee 38152, United States
| | - Dennis R Kennetz
- Department of Chemistry, The University of Memphis , Memphis, Tennessee 38152, United States
| | - Nyles J Walker
- Department of Chemistry, The University of Memphis , Memphis, Tennessee 38152, United States
| | - Yongmei Wang
- Department of Chemistry, The University of Memphis , Memphis, Tennessee 38152, United States
| |
Collapse
|
18
|
Pereira P, Barreira M, Queiroz JA, Veiga F, Sousa F, Figueiras A. Smart micelleplexes as a new therapeutic approach for RNA delivery. Expert Opin Drug Deliv 2016; 14:353-371. [DOI: 10.1080/17425247.2016.1214567] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
|
19
|
Badwaik VD, Aicart E, Mondjinou YA, Johnson MA, Bowman VD, Thompson DH. Structure-property relationship for in vitro siRNA delivery performance of cationic 2-hydroxypropyl-β-cyclodextrin: PEG-PPG-PEG polyrotaxane vectors. Biomaterials 2016; 84:86-98. [PMID: 26826298 PMCID: PMC4755830 DOI: 10.1016/j.biomaterials.2015.11.032] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2015] [Revised: 11/22/2015] [Accepted: 11/25/2015] [Indexed: 11/20/2022]
Abstract
Nanoparticle-mediated siRNA delivery is a promising therapeutic approach, however, the processes required for transport of these materials across the numerous extracellular and intracellular barriers are poorly understood. Efficient delivery of siRNA-containing nanoparticles would ultimately benefit from an improved understanding of how parameters associated with these barriers relate to the physicochemical properties of the nanoparticle vectors. We report the synthesis of three Pluronic(®)-based, cholesterol end-capped cationic polyrotaxanes (PR(+)) threaded with 2-hydroxypropyl-β-cyclodextrin (HPβCD) for siRNA delivery. The biological data showed that PR(+):siRNA complexes were well tolerated (∼90% cell viability) and produced efficient silencing (>80%) in HeLa-GFP and NIH 3T3-GFP cell lines. We further used a multi-parametric approach to identify relationships between the PR(+) structure, PR(+):siRNA complex physical properties, and biological activity. Small angle X-ray scattering and cryoelectron microscopy studies reveal periodicity and lamellar architectures for PR(+):siRNA complexes, whereas the biological assays, ζ potential measurements, and imaging studies suggest that silencing efficiency is influenced by the effective charge ratio (ρeff), polypropylene oxide (PO) block length, and central PO block coverage (i.e., rigidity) of the PR(+) core. We infer from our findings that more compact PR(+):siRNA nanostructures arising from lower molecular weight, rigid rod-like PR(+) polymer cores produce improved silencing efficiency relative to higher molecular weight, more flexible PR(+) vectors of similar effective charge. This study demonstrates that PR(+):siRNA complex formulations can be produced having higher performance than Lipofectamine(®) 2000, while maintaining good cell viability and siRNA sequence protection in cell culture.
Collapse
Affiliation(s)
- Vivek D Badwaik
- Department of Chemistry, Multi-disciplinary Cancer Research Facility, Bindley Bioscience Center, 1203 W. State Street, West Lafayette, IN 47907, USA
| | - Emilio Aicart
- Department of Chemistry, Multi-disciplinary Cancer Research Facility, Bindley Bioscience Center, 1203 W. State Street, West Lafayette, IN 47907, USA
| | - Yawo A Mondjinou
- Department of Chemistry, Multi-disciplinary Cancer Research Facility, Bindley Bioscience Center, 1203 W. State Street, West Lafayette, IN 47907, USA
| | - Merrell A Johnson
- Department of Physics, Indiana University-Purdue University Indianapolis, IN 46202, USA
| | - Valorie D Bowman
- Discovery Park, Hockmeyer Hall of Structural Biology, Purdue University, West Lafayette, IN 47907, USA
| | - David H Thompson
- Department of Chemistry, Multi-disciplinary Cancer Research Facility, Bindley Bioscience Center, 1203 W. State Street, West Lafayette, IN 47907, USA.
| |
Collapse
|
20
|
Ramezanpour M, Leung SSW, Delgado-Magnero KH, Bashe BYM, Thewalt J, Tieleman DP. Computational and experimental approaches for investigating nanoparticle-based drug delivery systems. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2016; 1858:1688-709. [PMID: 26930298 DOI: 10.1016/j.bbamem.2016.02.028] [Citation(s) in RCA: 108] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/23/2016] [Revised: 02/20/2016] [Accepted: 02/23/2016] [Indexed: 12/21/2022]
Abstract
Most therapeutic agents suffer from poor solubility, rapid clearance from the blood stream, a lack of targeting, and often poor translocation ability across cell membranes. Drug/gene delivery systems (DDSs) are capable of overcoming some of these barriers to enhance delivery of drugs to their right place of action, e.g. inside cancer cells. In this review, we focus on nanoparticles as DDSs. Complementary experimental and computational studies have enhanced our understanding of the mechanism of action of nanocarriers and their underlying interactions with drugs, biomembranes and other biological molecules. We review key biophysical aspects of DDSs and discuss how computer modeling can assist in rational design of DDSs with improved and optimized properties. We summarize commonly used experimental techniques for the study of DDSs. Then we review computational studies for several major categories of nanocarriers, including dendrimers and dendrons, polymer-, peptide-, nucleic acid-, lipid-, and carbon-based DDSs, and gold nanoparticles. This article is part of a Special Issue entitled: Membrane Proteins edited by J.C. Gumbart and Sergei Noskov.
Collapse
Affiliation(s)
- M Ramezanpour
- Centre for Molecular Simulation, Department of Biological Sciences, University of Calgary, Calgary, AB T2N 1N4, Canada
| | - S S W Leung
- Department of Physics, Simon Fraser University, Burnaby, BC V5A 1S6, Canada
| | - K H Delgado-Magnero
- Centre for Molecular Simulation, Department of Biological Sciences, University of Calgary, Calgary, AB T2N 1N4, Canada
| | - B Y M Bashe
- Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, BC V5A 1S6, Canada
| | - J Thewalt
- Department of Physics, Simon Fraser University, Burnaby, BC V5A 1S6, Canada; Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, BC V5A 1S6, Canada
| | - D P Tieleman
- Centre for Molecular Simulation, Department of Biological Sciences, University of Calgary, Calgary, AB T2N 1N4, Canada
| |
Collapse
|
21
|
Pereira PA, Tomás JF, Queiroz JA, Figueiras AR, Sousa F. Recombinant pre-miR-29b for Alzheimer´s disease therapeutics. Sci Rep 2016; 6:19946. [PMID: 26818210 PMCID: PMC4730146 DOI: 10.1038/srep19946] [Citation(s) in RCA: 74] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2015] [Accepted: 11/11/2015] [Indexed: 12/16/2022] Open
Abstract
MicroRNAs are arising as the next generation of diagnostic and therapeutic tools for gene silencing. Studies demonstrated that the miR-29 expression is decreased in Alzheimer’s disease (AD) patients displaying high levels of human β-secretase (hBACE1). Recent advances toward an effective therapy for AD intend to employ miR-29 to suppress hBACE1 expression and subsequent Amyloid-β (Aβ) peptide. However, delivery of mature miRNA has demonstrated modest efficacy in vitro; therefore, the preparation of highly pure and biologically active pre-miRNA arises as one of the most important challenges in the development of these therapeutic strategies. Recently, we described a new strategy based arginine-affinity chromatography to specifically purify the recombinant pre-miR-29b. Following this strategy, the purified pre-miR-29b was successfully encapsulated into polyplexes that were further delivered in cytoplasm. It was verified that Chitosan/pre-miR-29b and Polyethylenimine/pre-miR-29b systems efficiently delivered pre-miR-29b to N2a695 cells, thus reducing the hBACE1 protein expression (around 78% and 86%, respectively) and Aβ42 levels (approximately 44% and 47%, respectively). Furthermore, pre-miR-29b downregulates the hBACE1 mRNA expression in 80%. Overall, it was demonstrated that the recombinant pre-miR-29b using polyplexes allowed to decrease the hBACE1 and Aβ42 expression levels, improving the currently available methodologies of miRNA-based therapeutics.
Collapse
Affiliation(s)
- Patrícia A Pereira
- CICS-UBI - Health Sciences Research Centre, University of Beira Interior, Avenida Infante D. Henrique, Covilhã, 6200-506, Portugal
| | - Joana F Tomás
- CICS-UBI - Health Sciences Research Centre, University of Beira Interior, Avenida Infante D. Henrique, Covilhã, 6200-506, Portugal
| | - João A Queiroz
- CICS-UBI - Health Sciences Research Centre, University of Beira Interior, Avenida Infante D. Henrique, Covilhã, 6200-506, Portugal
| | - Ana R Figueiras
- CICS-UBI - Health Sciences Research Centre, University of Beira Interior, Avenida Infante D. Henrique, Covilhã, 6200-506, Portugal.,CNC - Center of Neuroscience and Cell Biology, University of Coimbra, Largo Marquês de Pombal, Coimbra, 3004-517, Portugal
| | - Fani Sousa
- CICS-UBI - Health Sciences Research Centre, University of Beira Interior, Avenida Infante D. Henrique, Covilhã, 6200-506, Portugal
| |
Collapse
|
22
|
Agudelo D, Kreplak L, Tajmir-Riahi HA. tRNA conjugation with chitosan nanoparticles: An AFM imaging study. Int J Biol Macromol 2015; 85:150-6. [PMID: 26723249 DOI: 10.1016/j.ijbiomac.2015.12.057] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2015] [Revised: 12/15/2015] [Accepted: 12/17/2015] [Indexed: 02/06/2023]
Abstract
The conjugation of tRNA with chitosan nanoparticles of different sizes 15,100 and 200 kDa was investigated in aqueous solution using multiple spectroscopic methods and atomic force microscopy (AFM). Structural analysis showed that chitosan binds tRNA via G-C and A-U base pairs as well as backbone PO2 group, through electrostatic, hydrophilic and H-bonding contacts with overall binding constants of KCh-15-tRNA=4.1 (±0.60)×10(3)M(-1), KCh-100-tRNA=5.7 (±0.8)×10(3)M(-1) and KCh-200-tRNA=1.2 (±0.3)×10(4)M(-1). As chitosan size increases more stable polymer-tRNA conjugate is formed. AFM images showed major tRNA aggregation and particle formation occurred as chitosan concentration increased. Even though chitosan induced major biopolymer structural changes, tRNA remains in A-family structure.
Collapse
Affiliation(s)
- D Agudelo
- Department of Chemistry-Biochemistry and Physics, University of Québec at Trois-Rivières, C. P. 500, Trois-Rivières (Québec), G9A 5H7, Canada
| | - L Kreplak
- Department of Physics and Atmospheric Science, Sir James Dunn Building Dalhousie University, Lord Dalhousie Drive, Halifax, NS B3H4R2, Canada
| | - H A Tajmir-Riahi
- Department of Chemistry-Biochemistry and Physics, University of Québec at Trois-Rivières, C. P. 500, Trois-Rivières (Québec), G9A 5H7, Canada.
| |
Collapse
|
23
|
New Techniques to Assess In Vitro Release of siRNA from Nanoscale Polyplexes. Pharm Res 2014; 32:1957-74. [DOI: 10.1007/s11095-014-1589-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2014] [Accepted: 11/24/2014] [Indexed: 12/29/2022]
|
24
|
Ganas C, Weiß A, Nazarenus M, Rösler S, Kissel T, Rivera_Gil P, Parak WJ. Biodegradable capsules as non-viral vectors for in vitro delivery of PEI/siRNA polyplexes for efficient gene silencing. J Control Release 2014; 196:132-8. [DOI: 10.1016/j.jconrel.2014.10.006] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2014] [Revised: 09/23/2014] [Accepted: 10/03/2014] [Indexed: 11/27/2022]
|
25
|
Gene delivery efficiency and intracellular trafficking of novel poly(allylamine) derivatives. Int J Pharm 2014; 478:372-382. [PMID: 25433199 DOI: 10.1016/j.ijpharm.2014.11.053] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2014] [Revised: 10/26/2014] [Accepted: 11/25/2014] [Indexed: 12/12/2022]
Abstract
Non-viral gene carriers for safe and efficient gene transfection have become of particular interest among researchers of different disciplines ranging from physical chemistry to biotechnology. Recently polymeric vectors have been extensively studied as potentially new gene transfer agents. Until now most of the research efforts were made to optimize the gene-to-polymer weight ratio of polyplexes for safe and efficient gene transfection. In this work, we report on the development of novel poly(allylamine) derivatives with different balance of the primary, secondary, tertiary, and quaternary amino groups. All derivatives were able to complex pDNA into polyplexes at low gene-to-polymer weight ratios i.e., 1:1 or 1:2. Moreover, the examined polyplexes were less cytotoxic and showed better transfection efficiency when compared to linear poly(ethyleneimine). These results indicate that the presence of quaternary ammonium groups is important in the formation of stable polyplexes. Polymers with all types of amino groups showed large potential for gene delivery. Furthermore, polyplexes with such derivatives were well internalized by cells and ended up into acidic late endosomes.
Collapse
|
26
|
Costa D, Valente AJM, Miguel MG, Queiroz J. Plasmid DNA hydrogels for biomedical applications. Adv Colloid Interface Sci 2014; 205:257-64. [PMID: 24011472 DOI: 10.1016/j.cis.2013.08.002] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2013] [Revised: 08/05/2013] [Accepted: 08/05/2013] [Indexed: 01/05/2023]
Abstract
In the last few years, our research group has focused on the design and development of plasmid DNA (pDNA) based systems as devices to be used therapeutically in the biomedical field. Biocompatible macro and micro plasmid DNA gels were prepared by a cross-linking reaction. For the first time, the pDNA gels have been investigated with respect to their swelling in aqueous solution containing different additives. Furthermore, we clarified the fundamental and basic aspects of the solute release mechanism from pDNA hydrogels and the significance of this information is enormous as a basic tool for the formulation of pDNA carriers for drug/gene delivery applications. The co-delivery of a specific gene and anticancer drugs, combining chemical and gene therapies in the treatment of cancer was the main challenge of our research. Significant progresses have been made with a new p53 encoding pDNA microgel that is suitable for the loading and release of pDNA and doxorubicin. This represents a strong valuable finding in the strategic development of systems to improve cancer cure through the synergetic effect of chemical and gene therapy.
Collapse
Affiliation(s)
- Diana Costa
- CICS - Centro de Investigação em Ciências da Saúde, Universidade da Beira Interior, 6201-001 Covilhã, Portugal.
| | | | - M Graça Miguel
- Department of Chemistry, University of Coimbra, Coimbra, Portugal
| | - João Queiroz
- CICS - Centro de Investigação em Ciências da Saúde, Universidade da Beira Interior, 6201-001 Covilhã, Portugal
| |
Collapse
|
27
|
Pereira P, Sousa Â, Queiroz J, Figueiras A, Sousa F. New approach for purification of pre-miR-29 using lysine-affinity chromatography. J Chromatogr A 2014; 1331:129-32. [DOI: 10.1016/j.chroma.2014.01.033] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2013] [Revised: 01/06/2014] [Accepted: 01/10/2014] [Indexed: 11/29/2022]
|
28
|
Rinkenauer AC, Vollrath A, Schallon A, Tauhardt L, Kempe K, Schubert S, Fischer D, Schubert US. Parallel high-throughput screening of polymer vectors for nonviral gene delivery: evaluation of structure-property relationships of transfection. ACS COMBINATORIAL SCIENCE 2013; 15:475-82. [PMID: 23886244 DOI: 10.1021/co400025u] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
In recent years, "high-throughput" (HT) has turned into a keyword in polymer research. In this study, we present a novel HT workflow for the investigation of cationic polymers for gene delivery applications. For this purpose, various poly(ethylene imine)s (PEI) were used as representative vectors and investigated via HT-assays in a 96-well plate format, starting from polyplex preparation up to the examination of the transfection process. In detail, automated polyplex preparation, complex size determination, DNA binding affinity, polyplex stability, cytotoxicity, and transfection efficiency were performed in the well plate format. With standard techniques, investigation of the biological properties of polymers is quite time-consuming, so only a limited number of materials and conditions (such as pH, buffer composition, and concentration) can be examined. The approach described here allows many different polymers and parameters to be tested for transfection properties and cytotoxicity, giving faster insights into structure-activity relationships for biological activity.
Collapse
Affiliation(s)
- Alexandra C. Rinkenauer
- Laboratory of Organic and Macromolecular
Chemistry (IOMC), Friedrich Schiller University Jena, Humboldtstraße 10, 07743 Jena, Germany
- Jena Center for Soft Matter
(JCSM), Friedrich Schiller University Jena, Philosophenweg 7, 07743 Jena, Germany
| | - Antje Vollrath
- Laboratory of Organic and Macromolecular
Chemistry (IOMC), Friedrich Schiller University Jena, Humboldtstraße 10, 07743 Jena, Germany
- Jena Center for Soft Matter
(JCSM), Friedrich Schiller University Jena, Philosophenweg 7, 07743 Jena, Germany
| | - Anja Schallon
- Laboratory of Organic and Macromolecular
Chemistry (IOMC), Friedrich Schiller University Jena, Humboldtstraße 10, 07743 Jena, Germany
- Jena Center for Soft Matter
(JCSM), Friedrich Schiller University Jena, Philosophenweg 7, 07743 Jena, Germany
| | - Lutz Tauhardt
- Laboratory of Organic and Macromolecular
Chemistry (IOMC), Friedrich Schiller University Jena, Humboldtstraße 10, 07743 Jena, Germany
- Jena Center for Soft Matter
(JCSM), Friedrich Schiller University Jena, Philosophenweg 7, 07743 Jena, Germany
| | - Kristian Kempe
- Laboratory of Organic and Macromolecular
Chemistry (IOMC), Friedrich Schiller University Jena, Humboldtstraße 10, 07743 Jena, Germany
- Jena Center for Soft Matter
(JCSM), Friedrich Schiller University Jena, Philosophenweg 7, 07743 Jena, Germany
| | - Stephanie Schubert
- Jena Center for Soft Matter
(JCSM), Friedrich Schiller University Jena, Philosophenweg 7, 07743 Jena, Germany
- Institute of Pharmacy, Department
of Pharmaceutical Technology, Friedrich Schiller University Jena, Otto-Schott-Straße 41, 07745 Jena, Germany
| | - Dagmar Fischer
- Institute of Pharmacy, Department
of Pharmaceutical Technology, Friedrich Schiller University Jena, Otto-Schott-Straße 41, 07745 Jena, Germany
| | - Ulrich S. Schubert
- Laboratory of Organic and Macromolecular
Chemistry (IOMC), Friedrich Schiller University Jena, Humboldtstraße 10, 07743 Jena, Germany
- Jena Center for Soft Matter
(JCSM), Friedrich Schiller University Jena, Philosophenweg 7, 07743 Jena, Germany
- Dutch Polymer Institute, P.O. Box 902, 5600 AX Eindhoven, the Netherlands
| |
Collapse
|