1
|
Ye X, Chen S, Xiong W, Wang F, Chan HF, Lai H, Guo X, Yang T, Shen S, Chen H, Wang W, Liu GS, Guo Y, Chen J. Magnetic-Guided Delivery of Antisense Oligonucleotides for Targeted Transduction in Multiple Retinal Explant and Organoid Models. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025:e2417363. [PMID: 40278802 DOI: 10.1002/advs.202417363] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/22/2024] [Revised: 03/21/2025] [Indexed: 04/26/2025]
Abstract
Antisense oligonucleotide (ASO) therapy holds promise in gene therapy but faces challenges due to poor delivery efficiency and limited evaluation models. This investigation employs magnetic nanoparticles (MNPs) to augment the delivery efficiency of ASOs. It assesses their distribution and therapeutic efficacy across various models, including retinal explants from mice and macaques or human retinal and inner ear organoids. Retinal explants from both mice and monkeys are methodically arranged to expose the ganglion cell layer (GCL) or the photoreceptor layer (PL). MNPs markedly enhanced the penetration and targeting of ASOs, resulting in a 60% accumulation in the GCL or 72% in the photoreceptors. Furthermore, an in vitro biomimetic model of the neuroretina-RPE/choroid-sclera complex is developed to examine ASO distribution under dynamic flow conditions. Moreover, the utilization of MNP-assisted ASO-Cy3 markedly enhanced transfection efficiency within human retinal and inner ear organoids, resulting in an increase in positively transfected cells to 60% and 70%, respectively. Here, for the first time, an MNP-explant-organoid platform is carried out for the promotion of ASO transfection efficiency, therapeutic screening and targeted delivery. This development paves the way for investigating novel gene therapy strategies targeting retinal diseases.
Collapse
Affiliation(s)
- Xiuhong Ye
- Department of Ophthalmology, The First Affiliated Hospital of Jinan University, Jinan University, Guangzhou, 510632, China
- Institute of Ophthalmology, Medical College, Jinan University, Guangzhou, 510632, China
| | - Sihui Chen
- Department of Ophthalmology, The First Affiliated Hospital of Jinan University, Jinan University, Guangzhou, 510632, China
- Institute of Ophthalmology, Medical College, Jinan University, Guangzhou, 510632, China
| | - Wei Xiong
- Key Laboratory for Regenerative Medicine, Ministry of Education, Jinan University, Guangzhou, 510632, China
| | - Fan Wang
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China
| | - Hon Fai Chan
- Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Hong Kong, 999077, China
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, 999077, China
| | - Haocheng Lai
- Division of Life Science, Hong Kong University of Science and Technology, Hong Kong, 999077, China
| | - Xiangyu Guo
- Guangdong Key Laboratory of Non-human Primate Research, Guangdong-Hongkong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou, 510000, China
| | - Tingting Yang
- Department of Ophthalmology, The First Affiliated Hospital of Jinan University, Jinan University, Guangzhou, 510632, China
- Institute of Ophthalmology, Medical College, Jinan University, Guangzhou, 510632, China
| | - Shuhao Shen
- Department of Ophthalmology, The First Affiliated Hospital of Jinan University, Jinan University, Guangzhou, 510632, China
- Institute of Ophthalmology, Medical College, Jinan University, Guangzhou, 510632, China
| | - Hang Chen
- Department of Ophthalmology, The First Affiliated Hospital of Jinan University, Jinan University, Guangzhou, 510632, China
- Institute of Ophthalmology, Medical College, Jinan University, Guangzhou, 510632, China
| | - Wenxuan Wang
- Department of Ophthalmology, The First Affiliated Hospital of Jinan University, Jinan University, Guangzhou, 510632, China
- Institute of Ophthalmology, Medical College, Jinan University, Guangzhou, 510632, China
| | - Guei-Sheung Liu
- Aier Eye Institute, Aier Eye Hospital Group Co., Ltd., Changsha, 410000, China
- Centre for Eye Research Australia, Royal Victorian Eye and Ear Hospital, East Melbourne, VIC, 3002, Australia
- Ophthalmology, Department of Surgery, University of Melbourne, East Melbourne, VIC, 3010, Australia
- Menzies Institute for Medical Research, University of Tasmania, Hobart, TAS, 7001, Australia
| | - Yonglong Guo
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China
| | - Jiansu Chen
- Department of Ophthalmology, The First Affiliated Hospital of Jinan University, Jinan University, Guangzhou, 510632, China
- Institute of Ophthalmology, Medical College, Jinan University, Guangzhou, 510632, China
- Key Laboratory for Regenerative Medicine, Ministry of Education, Jinan University, Guangzhou, 510632, China
- Aier Eye Institute, Aier Eye Hospital Group Co., Ltd., Changsha, 410000, China
| |
Collapse
|
2
|
Birkle TJ, Willems HM, Skidmore J, Brown GC. Disease phenotypic screening in neuron-glia cocultures identifies blockers of inflammatory neurodegeneration. iScience 2024; 27:109454. [PMID: 38550989 PMCID: PMC10973195 DOI: 10.1016/j.isci.2024.109454] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Revised: 10/09/2023] [Accepted: 03/06/2024] [Indexed: 01/30/2025] Open
Abstract
Neuropathology is often mediated by interactions between neurons and glia that cannot be modeled by monocultures. However, cocultures are difficult to use and analyze for high-content screening. Here, we perform compound screening using primary neuron-glia cultures to model inflammatory neurodegeneration, live-cell stains, and automated classification of neurons, astrocytes or microglia using open-source software. Out of 227 compounds with known bioactivities, 29 protected against lipopolysaccharide-induced neuronal loss, including drugs affecting adrenergic, steroid, inflammatory and MAP kinase signaling. The screen also identified physiological compounds, such as noradrenaline and progesterone, that protected and identified neurotoxic compounds, such as a TLR7 agonist, that induced microglial proliferation. Most compounds used here have not been tested in a neuron-glia coculture neurodegeneration assay previously. Thus, combining a complex cellular disease model with high-content screening of known compounds and automated image analysis allows identification of important biology, as well as potential targets and drugs for treatment.
Collapse
Affiliation(s)
| | | | - John Skidmore
- ALBORADA Drug Discovery Institute, Cambridge CB2 0AH, UK
| | - Guy C. Brown
- Department of Biochemistry, University of Cambridge, Cambridge CB2 1QW, UK
| |
Collapse
|
3
|
Morshedi Rad D, Alsadat Rad M, Razavi Bazaz S, Kashaninejad N, Jin D, Ebrahimi Warkiani M. A Comprehensive Review on Intracellular Delivery. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2021; 33:e2005363. [PMID: 33594744 DOI: 10.1002/adma.202005363] [Citation(s) in RCA: 72] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Revised: 09/22/2020] [Indexed: 05/22/2023]
Abstract
Intracellular delivery is considered an indispensable process for various studies, ranging from medical applications (cell-based therapy) to fundamental (genome-editing) and industrial (biomanufacture) approaches. Conventional macroscale delivery systems critically suffer from such issues as low cell viability, cytotoxicity, and inconsistent material delivery, which have opened up an interest in the development of more efficient intracellular delivery systems. In line with the advances in microfluidics and nanotechnology, intracellular delivery based on micro- and nanoengineered platforms has progressed rapidly and held great promises owing to their unique features. These approaches have been advanced to introduce a smorgasbord of diverse cargoes into various cell types with the maximum efficiency and the highest precision. This review differentiates macro-, micro-, and nanoengineered approaches for intracellular delivery. The macroengineered delivery platforms are first summarized and then each method is categorized based on whether it employs a carrier- or membrane-disruption-mediated mechanism to load cargoes inside the cells. Second, particular emphasis is placed on the micro- and nanoengineered advances in the delivery of biomolecules inside the cells. Furthermore, the applications and challenges of the established and emerging delivery approaches are summarized. The topic is concluded by evaluating the future perspective of intracellular delivery toward the micro- and nanoengineered approaches.
Collapse
Affiliation(s)
- Dorsa Morshedi Rad
- School of Biomedical Engineering, University of Technology Sydney, Sydney, NSW, 2007, Australia
- Institute for Biomedical Materials & Devices (IBMD), Faculty of Science, University of Technology Sydney, Sydney, NSW, 2007, Australia
| | - Maryam Alsadat Rad
- School of Biomedical Engineering, University of Technology Sydney, Sydney, NSW, 2007, Australia
- Institute for Biomedical Materials & Devices (IBMD), Faculty of Science, University of Technology Sydney, Sydney, NSW, 2007, Australia
| | - Sajad Razavi Bazaz
- School of Biomedical Engineering, University of Technology Sydney, Sydney, NSW, 2007, Australia
- Institute for Biomedical Materials & Devices (IBMD), Faculty of Science, University of Technology Sydney, Sydney, NSW, 2007, Australia
| | - Navid Kashaninejad
- School of Biomedical Engineering, University of Technology Sydney, Sydney, NSW, 2007, Australia
- Institute for Biomedical Materials & Devices (IBMD), Faculty of Science, University of Technology Sydney, Sydney, NSW, 2007, Australia
| | - Dayong Jin
- Institute for Biomedical Materials & Devices (IBMD), Faculty of Science, University of Technology Sydney, Sydney, NSW, 2007, Australia
- School of Life Sciences, Faculty of Science, University of Technology Sydney, Sydney, NSW, 2007, Australia
| | - Majid Ebrahimi Warkiani
- School of Biomedical Engineering, University of Technology Sydney, Sydney, NSW, 2007, Australia
- Institute for Biomedical Materials & Devices (IBMD), Faculty of Science, University of Technology Sydney, Sydney, NSW, 2007, Australia
- Institute of Molecular Medicine, Sechenov University, Moscow, 119991, Russia
| |
Collapse
|
4
|
Belete TM. The Current Status of Gene Therapy for the Treatment of Cancer. Biologics 2021; 15:67-77. [PMID: 33776419 PMCID: PMC7987258 DOI: 10.2147/btt.s302095] [Citation(s) in RCA: 60] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Accepted: 03/04/2021] [Indexed: 02/06/2023]
Abstract
Gene therapy is the administration of foreign genomic material into the host tissue to modify the expression of a gene product or to change the biological properties of cells for therapeutic use. Initially, the major objective of gene therapy was to manage genetic diseases, but now different disorders with several patterns of acquired and inherited disorders are targets of gene therapy. Over three decades, the advancement of Genome engineering technologies facilitated gene therapy for the prevention and management of intractable diseases. Researchers are advancing with cautious optimism that safe and effective treatment will give to patients with single-gene disorders and complex acquired disorders. To date, over 3000 genes associates with disease-causing mutations, and about 2600 gene therapy trials are undergoing for the management of various disorders. This review summarizes the principles of genome-editing approaches, such as zinc finger nucleases, transcription activator-like effector nucleases, meganucleases, and the CRISPR/Cas9 system with the underlying mechanisms. This review also explains the types of gene delivery systems as viral [adenoviral, adeno association, herpes simplex virus] and nonviral delivery systems (physical: DNA bombardment, electroporation) and (chemical: Cationic lipids, cationic polymers). Finally, this review summarizes gene therapy medicines approved to treat cancer in detail, including names, indications, vectors, and mode of gene therapy. Gene therapy becomes an alternative to an existing management for different diseases. Therefore, gene products with safe vectors and better biotechnologies play a significant role in the prophylaxis and management of various disorders in the future.
Collapse
Affiliation(s)
- Tafere Mulaw Belete
- Department of Pharmacology, College of Medicine and Health Sciences, University of Gondar, Gondar, Amhara Region, Ethiopia
| |
Collapse
|
5
|
Blokpoel Ferreras LA, Chan SY, Vazquez Reina S, Dixon JE. Rapidly Transducing and Spatially Localized Magnetofection Using Peptide-Mediated Non-Viral Gene Delivery Based on Iron Oxide Nanoparticles. ACS APPLIED NANO MATERIALS 2021; 4:167-181. [PMID: 33763629 PMCID: PMC7978400 DOI: 10.1021/acsanm.0c02465] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Accepted: 12/06/2020] [Indexed: 05/03/2023]
Abstract
Non-viral delivery systems are generally of low efficiency, which limits their use in gene therapy and editing applications. We previously developed a technology termed glycosaminoglycan (GAG)-binding enhanced transduction (GET) to efficiently deliver a variety of cargos intracellularly; our system employs GAG-binding peptides, which promote cell targeting, and cell penetrating peptides (CPPs), which enhance endocytotic cell internalization. Herein, we describe a further modification by combining gene delivery and magnetic targeting with the GET technology. We associated GET peptides, plasmid (p)DNA, and iron oxide superparamagnetic nanoparticles (MNPs), allowing rapid and targeted GET-mediated uptake by application of static magnetic fields in NIH3T3 cells. This produced effective transfection levels (significantly higher than the control) with seconds to minutes of exposure and localized gene delivery two orders of magnitude higher in targeted over non-targeted cell monolayers using magnetic fields (in 15 min exposure delivering GFP reporter pDNA). More importantly, high cell membrane targeting by GET-DNA and MNP co-complexes and magnetic fields allowed further enhancement to endocytotic uptake, meaning that the nucleic acid cargo was rapidly internalized beyond that of GET complexes alone (GET-DNA). Magnetofection by MNPs combined with GET-mediated delivery allows magnetic field-guided local transfection in vitro and could facilitate focused gene delivery for future regenerative and disease-targeted therapies in vivo.
Collapse
Affiliation(s)
- Lia A. Blokpoel Ferreras
- Regenerative
Medicine & Cellular Therapies Division, The University of Nottingham
Biodiscovery Institute (BDI), School of Pharmacy, University of Nottingham, Nottingham NG7 2RD, U.K.
| | - Sze Yan Chan
- Regenerative
Medicine & Cellular Therapies Division, The University of Nottingham
Biodiscovery Institute (BDI), School of Pharmacy, University of Nottingham, Nottingham NG7 2RD, U.K.
| | - Saul Vazquez Reina
- School
of Veterinary Sciences, University of Nottingham, Nottingham NG7 2RD, U.K.
| | - James E. Dixon
- Regenerative
Medicine & Cellular Therapies Division, The University of Nottingham
Biodiscovery Institute (BDI), School of Pharmacy, University of Nottingham, Nottingham NG7 2RD, U.K.
| |
Collapse
|
6
|
Mashel TV, Tarakanchikova YV, Muslimov AR, Zyuzin MV, Timin AS, Lepik KV, Fehse B. Overcoming the delivery problem for therapeutic genome editing: Current status and perspective of non-viral methods. Biomaterials 2020; 258:120282. [PMID: 32798742 DOI: 10.1016/j.biomaterials.2020.120282] [Citation(s) in RCA: 71] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Revised: 07/22/2020] [Accepted: 08/01/2020] [Indexed: 12/11/2022]
|
7
|
Lehmann M, Canatelli-Mallat M, Chiavellini P, Cónsole GM, Gallardo MD, Goya RG. Partial Reprogramming As An Emerging Strategy for Safe Induced Cell Generation and Rejuvenation. Curr Gene Ther 2020; 19:248-254. [PMID: 31475896 DOI: 10.2174/1566523219666190902154511] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Revised: 08/05/2019] [Accepted: 08/08/2019] [Indexed: 11/22/2022]
Abstract
BACKGROUND Conventional cell reprogramming involves converting a somatic cell line into induced pluripotent stem cells (iPSC), which subsequently can be re-differentiated to specific somatic cell types. Alternatively, partial cell reprogramming converts somatic cells into other somatic cell types by transient expression of pluripotency genes thus generating intermediates that retain their original cell identity, but are responsive to appropriate cocktails of specific differentiation factors. Additionally, biological rejuvenation by partial cell reprogramming is an emerging avenue of research. OBJECTIVE Here, we will briefly review the emerging information pointing to partial reprogramming as a suitable strategy to achieve cell reprogramming and rejuvenation, bypassing cell dedifferentiation. METHODS In this context, regulatable pluripotency gene expression systems are the most widely used at present to implement partial cell reprogramming. For instance, we have constructed a regulatable bidirectional adenovector expressing Green Fluorescent Protein and oct4, sox2, klf4 and c-myc genes (known as the Yamanaka genes or OSKM). RESULTS Partial cell reprogramming has been used to reprogram fibroblasts to cardiomyocytes, neural progenitors and neural stem cells. Rejuvenation by cyclic partial reprogramming has been achieved both in vivo and in cell culture using transgenic mice and cells expressing the OSKM genes, respectively, controlled by a regulatable promoter. CONCLUSION Partial reprogramming emerges as a powerful tool for the genesis of iPSC-free induced somatic cells of therapeutic value and for the implementation of in vitro and in vivo rejuvenation keeping cell type identity unchanged.
Collapse
Affiliation(s)
- Marianne Lehmann
- INIBIOLP-Pathology B, Faculty of Medicine, UNLP, CC 455 (zip 1900) La Plata, Argentina.,Department of Histology, Cytology and of Embryology B, School of Medicine, University of La Plata, La Plata, Argentina
| | - Martina Canatelli-Mallat
- INIBIOLP-Pathology B, Faculty of Medicine, UNLP, CC 455 (zip 1900) La Plata, Argentina.,Department of Histology, Cytology and of Embryology B, School of Medicine, University of La Plata, La Plata, Argentina
| | - Priscila Chiavellini
- INIBIOLP-Pathology B, Faculty of Medicine, UNLP, CC 455 (zip 1900) La Plata, Argentina.,Department of Histology, Cytology and of Embryology B, School of Medicine, University of La Plata, La Plata, Argentina
| | - Gloria M Cónsole
- Department of Histology, Cytology and of Embryology B, School of Medicine, University of La Plata, La Plata, Argentina
| | - Maria D Gallardo
- INIBIOLP-Pathology B, Faculty of Medicine, UNLP, CC 455 (zip 1900) La Plata, Argentina.,Department of Histology, Cytology and of Embryology B, School of Medicine, University of La Plata, La Plata, Argentina
| | - Rodolfo G Goya
- INIBIOLP-Pathology B, Faculty of Medicine, UNLP, CC 455 (zip 1900) La Plata, Argentina.,Department of Histology, Cytology and of Embryology B, School of Medicine, University of La Plata, La Plata, Argentina
| |
Collapse
|
8
|
Sosa-Acosta JR, Iriarte-Mesa C, Ortega GA, Díaz-García AM. DNA–Iron Oxide Nanoparticles Conjugates: Functional Magnetic Nanoplatforms in Biomedical Applications. Top Curr Chem (Cham) 2020; 378:13. [DOI: 10.1007/s41061-019-0277-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Accepted: 12/20/2019] [Indexed: 02/08/2023]
|
9
|
Nuzhina JV, Shtil AA, Prilepskii AY, Vinogradov VV. Preclinical Evaluation and Clinical Translation of Magnetite-Based Nanomedicines. J Drug Deliv Sci Technol 2019. [DOI: 10.1016/j.jddst.2019.101282] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
10
|
Zusso M, Lunardi V, Franceschini D, Pagetta A, Lo R, Stifani S, Frigo AC, Giusti P, Moro S. Ciprofloxacin and levofloxacin attenuate microglia inflammatory response via TLR4/NF-kB pathway. J Neuroinflammation 2019; 16:148. [PMID: 31319868 PMCID: PMC6637517 DOI: 10.1186/s12974-019-1538-9] [Citation(s) in RCA: 351] [Impact Index Per Article: 58.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Accepted: 07/08/2019] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Neuroinflammation is the response of the central nervous system to events that interfere with tissue homeostasis and represents a common denominator in virtually all neurological diseases. Activation of microglia, the principal immune effector cells of the brain, contributes to neuronal injury by release of neurotoxic products. Toll-like receptor 4 (TLR4), expressed on the surface of microglia, plays an important role in mediating lipopolysaccharide (LPS)-induced microglia activation and inflammatory responses. We have previously shown that curcumin and some of its analogues harboring an α,β-unsaturated 1,3-diketone moiety, able to coordinate the magnesium ion, can interfere with LPS-mediated TLR4-myeloid differentiation protein-2 (MD-2) signaling. Fluoroquinolone (FQ) antibiotics are compounds that contain a keto-carbonyl group that binds divalent ions, including magnesium. In addition to their antimicrobial activity, FQs are endowed with immunomodulatory properties, but the mechanism underlying their anti-inflammatory activity remains to be defined. The aim of the current study was to elucidate the molecular mechanism of these compounds in the TLR4/NF-κB inflammatory signaling pathway. METHODS The putative binding mode of five FQs [ciprofloxacin (CPFX), levofloxacin (LVFX), moxifloxacin, ofloxacin, and delafloxacin] to TLR4-MD-2 was determined using molecular docking simulations. The effect of CPFX and LVFX on LPS-induced release of IL-1β and TNF-α and NF-κB activation was investigated in primary microglia by ELISA and fluorescence staining. The interaction of CPFX and LVFX with TLR4-MD-2 complex was assessed by immunoprecipitation followed by Western blotting using Ba/F3 cells. RESULTS CPFX and LVFX bound to the hydrophobic region of the MD-2 pocket and inhibited LPS-induced secretion of pro-inflammatory cytokines and activation of NF-κB in primary microglia. Furthermore, these FQs diminished the binding of LPS to TLR4-MD-2 complex and decreased the resulting TLR4-MD-2 dimerization in Ba/F3 cells. CONCLUSIONS These results provide new insight into the mechanism of the anti-inflammatory activity of CPFX and LVFX, which involves, at least in part, the activation of TLR4/NF-κB signaling pathway. Our findings might facilitate the development of new molecules directed at the TLR4-MD-2 complex, a potential key target for controlling neuroinflammation.
Collapse
Affiliation(s)
- Morena Zusso
- Department of Pharmaceutical and Pharmacological Sciences, University of Padua, Largo E. Meneghetti 2, 35131, Padua, Italy
| | - Valentina Lunardi
- Department of Pharmaceutical and Pharmacological Sciences, University of Padua, Largo E. Meneghetti 2, 35131, Padua, Italy
| | - Davide Franceschini
- Department of Pharmaceutical and Pharmacological Sciences, University of Padua, Largo E. Meneghetti 2, 35131, Padua, Italy.,Present address: Selvita S.A., Park Life Science ul, Bobrzyńskiego 14, 30-348, Kraków, Poland
| | - Andrea Pagetta
- Department of Pharmaceutical and Pharmacological Sciences, University of Padua, Largo E. Meneghetti 2, 35131, Padua, Italy
| | - Rita Lo
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, QC, H3A 2B4, Canada
| | - Stefano Stifani
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, QC, H3A 2B4, Canada
| | - Anna Chiara Frigo
- Unit of Biostatistics, Epidemiology and Public Health, Department of Cardiac, Thoracic and Vascular Sciences, University of Padua, Padua, Italy
| | - Pietro Giusti
- Department of Pharmaceutical and Pharmacological Sciences, University of Padua, Largo E. Meneghetti 2, 35131, Padua, Italy.
| | - Stefano Moro
- Department of Pharmaceutical and Pharmacological Sciences, University of Padua, Largo E. Meneghetti 2, 35131, Padua, Italy
| |
Collapse
|
11
|
Venero JL, Burguillos MA. Magnetofection as a new tool to study microglia biology. Neural Regen Res 2019; 14:767-768. [PMID: 30688259 PMCID: PMC6375049 DOI: 10.4103/1673-5374.249221] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Affiliation(s)
- Jose Luis Venero
- Instituto de Biomedicina de Sevilla (IBiS)/Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla and Departamento de Bioquímica y Biología Molecular, Facultad de Farmacia, Universidad de Sevilla and, 41012, Sevilla, Spain
| | | |
Collapse
|
12
|
Di Scala C, Tessier M, Sapet C, Poulhes F, Sicard F, Zelphati O, Pellegrino C. A new polymer-based approach for in vivo transfection in postnatal brain. J Neurosci Methods 2019; 311:295-306. [PMID: 30408559 DOI: 10.1016/j.jneumeth.2018.11.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Revised: 10/26/2018] [Accepted: 11/04/2018] [Indexed: 10/27/2022]
Abstract
BACKGROUND Gene delivery within the central nervous system at postnatal age is one of the most challenging tasks in neuroscience and currently only a few effective methods are available. COMPARISON WITH EXISTING METHODS For postnatal central nervous system cells, viral approaches are commonly used for genetic engineering but they face several biosafety requirements for production and use making them less accessible to the community. Conversely, lipid-based methods are widely used in cell culture but face limitation in vivo mainly due to the inflammatory responses they induce. To this aspect, the use of a transgenic mouse line can represent a credible answer to the community working on rat models still requires an effective and successful solution to circumvent these difficulties. NEW METHOD We describe a new polymer-based gene delivery system allowing persistent and robust in vivo transfection with low DNA amount, reduced inflammation and high diffusion. The expression profile along the brain, the stability, the diffusion of the DNA together with the quantity of cells transfected were evaluated through in vivo approaches. RESULTS With a single low-volume injection, we targeted different cell types within the rat brain. We measured the diffusion rate ranging from 1 to 5 mm based on the injected volume, in the three-dimensions axis. Finally, we modified brain susceptibility to epileptic seizures using a specific knock-down of the neuronal specific potassium-chloride transporter 2. CONCLUSIONS This safe and easy system opens perspectives for non viral gene delivery in the rat brain with perspectives to study brain function in vivo.
Collapse
Affiliation(s)
- C Di Scala
- INMED, INSERM, Aix-Marseille Univ, 163 route de luminy, BP13, Marseille, France
| | - M Tessier
- INMED, INSERM, Aix-Marseille Univ, 163 route de luminy, BP13, Marseille, France
| | - C Sapet
- OZ Biosciences, Parc Scientifique de Luminy, 163 Avenue de Luminy case 922, 13288 Marseille cedex 9, France
| | - F Poulhes
- OZ Biosciences, Parc Scientifique de Luminy, 163 Avenue de Luminy case 922, 13288 Marseille cedex 9, France
| | - F Sicard
- OZ Biosciences, Parc Scientifique de Luminy, 163 Avenue de Luminy case 922, 13288 Marseille cedex 9, France
| | - O Zelphati
- OZ Biosciences, Parc Scientifique de Luminy, 163 Avenue de Luminy case 922, 13288 Marseille cedex 9, France.
| | - C Pellegrino
- INMED, INSERM, Aix-Marseille Univ, 163 route de luminy, BP13, Marseille, France.
| |
Collapse
|
13
|
Gahl TJ, Kunze A. Force-Mediating Magnetic Nanoparticles to Engineer Neuronal Cell Function. Front Neurosci 2018; 12:299. [PMID: 29867315 PMCID: PMC5962660 DOI: 10.3389/fnins.2018.00299] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Accepted: 04/18/2018] [Indexed: 12/12/2022] Open
Abstract
Cellular processes like membrane deformation, cell migration, and transport of organelles are sensitive to mechanical forces. Technically, these cellular processes can be manipulated through operating forces at a spatial precision in the range of nanometers up to a few micrometers through chaperoning force-mediating nanoparticles in electrical, magnetic, or optical field gradients. But which force-mediating tool is more suitable to manipulate cell migration, and which, to manipulate cell signaling? We review here the differences in forces sensation to control and engineer cellular processes inside and outside the cell, with a special focus on neuronal cells. In addition, we discuss technical details and limitations of different force-mediating approaches and highlight recent advancements of nanomagnetics in cell organization, communication, signaling, and intracellular trafficking. Finally, we give suggestions about how force-mediating nanoparticles can be used to our advantage in next-generation neurotherapeutic devices.
Collapse
Affiliation(s)
| | - Anja Kunze
- Department of Electrical and Computer Engineering, Montana State University, Bozeman, MT, United States
| |
Collapse
|