1
|
Abasi M, Kianmehr A, Variji A, Sangali P, Mahrooz A. microRNAs as molecular tools for brain health: Neuroprotective potential in neurodegenerative disorders. Neuroscience 2025; 574:83-103. [PMID: 40210196 DOI: 10.1016/j.neuroscience.2025.04.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2024] [Revised: 03/09/2025] [Accepted: 04/05/2025] [Indexed: 04/12/2025]
Abstract
As research on microRNAs (miRNAs) advances, it is becoming increasingly clear that these small molecules play crucial roles in the central nervous system (CNS). They are involved in various essential neuronal functions, with specific miRNAs preferentially expressed in different cell types within the nervous system. Notably, certain miRNAs are found at higher levels in the brain and spinal cord compared to other tissues, suggesting they may have specialized functions in the CNS. miRNAs associated with long-term neurodegenerative changes could serve as valuable tools for early treatment decisions and disease monitoring. The significance of miRNAs such as miR-320, miR-146 and miR-29 in the early diagnosis of neurodegenerative disorders becomes evident, especially considering that many neurological and physical symptoms manifest only after substantial degeneration of specific neurons. Interestingly, serum miRNA levels such as miR-92 and miR-486 may correlate with various MRI parameters in multiple sclerosis. Targeting miRNAs using antisense strategies, such as antisense miR-146 and miR-485, may provide advantages over targeting mRNAs, as a single anti-miRNA can regulate multiple disease-related genes. In the future, anti-miRNA-based therapeutic approaches could be integrated into the clinical management of neurological diseases. Certain miRNAs, including miR-223, miR-106, miR-181, and miR-146, contribute to the pathogenesis of various neurodegenerative diseases and thus warrant greater attention. This knowledge could pave the way for the identification of new diagnostic, prognostic, and theranostic biomarkers, and potentially guiding the development of RNA-based therapeutic strategies. This review highlights recent research on the roles of miRNAs in the nervous system, particularly their protective functions in neurodegenerative disorders.
Collapse
Affiliation(s)
- Mozhgan Abasi
- Department of Tissue Engineering and Applied Cell Sciences, Faculty of Advanced Technologies in Medicine, Mazandaran University of Medical Sciences, Sari, Iran; Immunogenetics Research Center, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Anvarsadat Kianmehr
- Medical Cellular and Molecular Research Center, Golestan University of Medical Sciences, Gorgan, Iran
| | - Athena Variji
- Adelaide Medical School, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, Australia; Lifelong Health Theme, South Australian Health and Medical Research Institute, Adelaide, Australia
| | - Parisa Sangali
- Department of Clinical Biochemistry and Medical Genetics, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Abdolkarim Mahrooz
- Immunogenetics Research Center, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran.
| |
Collapse
|
2
|
Wang L, Yang Z, Satoshi F, Prasanna X, Yan Z, Vihinen H, Chen Y, Zhao Y, He X, Bu Q, Li H, Zhao Y, Jiang L, Qin F, Dai Y, Zhang N, Qin M, Kuang W, Zhao Y, Jokitalo E, Vattulainen I, Kajander T, Zhao H, Cen X. Membrane remodeling by FAM92A1 during brain development regulates neuronal morphology, synaptic function, and cognition. Nat Commun 2024; 15:6209. [PMID: 39043703 PMCID: PMC11266426 DOI: 10.1038/s41467-024-50565-w] [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: 09/18/2023] [Accepted: 07/12/2024] [Indexed: 07/25/2024] Open
Abstract
The Bin/Amphiphysin/Rvs (BAR) domain protein FAM92A1 is a multifunctional protein engaged in regulating mitochondrial ultrastructure and ciliogenesis, but its physiological role in the brain remains unclear. Here, we show that FAM92A1 is expressed in neurons starting from embryonic development. FAM92A1 knockout in mice results in altered brain morphology and age-associated cognitive deficits, potentially due to neuronal degeneration and disrupted synaptic plasticity. Specifically, FAM92A1 deficiency impairs diverse neuronal membrane morphology, including the mitochondrial inner membrane, myelin sheath, and synapses, indicating its roles in membrane remodeling and maintenance. By determining the crystal structure of the FAM92A1 BAR domain, combined with atomistic molecular dynamics simulations, we uncover that FAM92A1 interacts with phosphoinositide- and cardiolipin-containing membranes to induce lipid-clustering and membrane curvature. Altogether, these findings reveal the physiological role of FAM92A1 in the brain, highlighting its impact on synaptic plasticity and neural function through the regulation of membrane remodeling and endocytic processes.
Collapse
Affiliation(s)
- Liang Wang
- Mental Health Center and National Chengdu Center for Safety Evaluation of Drugs, State Key Laboratory of Biotherapy, West China Hospital of Sichuan University, Chengdu, 610041, China
- Faculty of Biological and Environmental Sciences, University of Helsinki, 00014, Helsinki, Finland
| | - Ziyun Yang
- Mental Health Center and National Chengdu Center for Safety Evaluation of Drugs, State Key Laboratory of Biotherapy, West China Hospital of Sichuan University, Chengdu, 610041, China
| | - Fudo Satoshi
- Helsinki Institute of Life Science - Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| | - Xavier Prasanna
- Department of Physics, University of Helsinki, Helsinki, Finland
| | - Ziyi Yan
- Faculty of Biological and Environmental Sciences, University of Helsinki, 00014, Helsinki, Finland
| | - Helena Vihinen
- Helsinki Institute of Life Science (HiLIFE) - Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| | - Yaxing Chen
- Mental Health Center and National Chengdu Center for Safety Evaluation of Drugs, State Key Laboratory of Biotherapy, West China Hospital of Sichuan University, Chengdu, 610041, China
| | - Yue Zhao
- Mental Health Center and National Chengdu Center for Safety Evaluation of Drugs, State Key Laboratory of Biotherapy, West China Hospital of Sichuan University, Chengdu, 610041, China
| | - Xiumei He
- Mental Health Center and National Chengdu Center for Safety Evaluation of Drugs, State Key Laboratory of Biotherapy, West China Hospital of Sichuan University, Chengdu, 610041, China
- School of Life Sciences, Guangxi Normal University, Guilin, China
- Guangxi Universities Key Laboratory of Stem Cell and Biopharmaceutical Technology, Guangxi Normal University, Guilin, 541004, China
| | - Qian Bu
- Mental Health Center and National Chengdu Center for Safety Evaluation of Drugs, State Key Laboratory of Biotherapy, West China Hospital of Sichuan University, Chengdu, 610041, China
| | - Hongchun Li
- Mental Health Center and National Chengdu Center for Safety Evaluation of Drugs, State Key Laboratory of Biotherapy, West China Hospital of Sichuan University, Chengdu, 610041, China
| | - Ying Zhao
- Mental Health Center and National Chengdu Center for Safety Evaluation of Drugs, State Key Laboratory of Biotherapy, West China Hospital of Sichuan University, Chengdu, 610041, China
| | - Linhong Jiang
- Mental Health Center and National Chengdu Center for Safety Evaluation of Drugs, State Key Laboratory of Biotherapy, West China Hospital of Sichuan University, Chengdu, 610041, China
| | - Feng Qin
- Mental Health Center and National Chengdu Center for Safety Evaluation of Drugs, State Key Laboratory of Biotherapy, West China Hospital of Sichuan University, Chengdu, 610041, China
| | - Yanping Dai
- Mental Health Center and National Chengdu Center for Safety Evaluation of Drugs, State Key Laboratory of Biotherapy, West China Hospital of Sichuan University, Chengdu, 610041, China
| | - Ni Zhang
- Mental Health Center, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Meng Qin
- Mental Health Center and National Chengdu Center for Safety Evaluation of Drugs, State Key Laboratory of Biotherapy, West China Hospital of Sichuan University, Chengdu, 610041, China
| | - Weihong Kuang
- Mental Health Center, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Yinglan Zhao
- Mental Health Center and National Chengdu Center for Safety Evaluation of Drugs, State Key Laboratory of Biotherapy, West China Hospital of Sichuan University, Chengdu, 610041, China
| | - Eija Jokitalo
- Helsinki Institute of Life Science (HiLIFE) - Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| | - Ilpo Vattulainen
- Department of Physics, University of Helsinki, Helsinki, Finland
| | - Tommi Kajander
- Helsinki Institute of Life Science - Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| | - Hongxia Zhao
- Faculty of Biological and Environmental Sciences, University of Helsinki, 00014, Helsinki, Finland.
- School of Life Sciences, Guangxi Normal University, Guilin, China.
- Guangxi Universities Key Laboratory of Stem Cell and Biopharmaceutical Technology, Guangxi Normal University, Guilin, 541004, China.
| | - Xiaobo Cen
- Mental Health Center and National Chengdu Center for Safety Evaluation of Drugs, State Key Laboratory of Biotherapy, West China Hospital of Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
3
|
Padmakumar S, D'Souza A, Parayath NN, Bleier BS, Amiji MM. Nucleic acid therapies for CNS diseases: Pathophysiology, targets, barriers, and delivery strategies. J Control Release 2022; 352:121-145. [PMID: 36252748 DOI: 10.1016/j.jconrel.2022.10.018] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 09/10/2022] [Accepted: 10/10/2022] [Indexed: 11/08/2022]
Abstract
Nucleic acid therapeutics have emerged as one of the very advanced and efficacious treatment approaches for debilitating health conditions, including those diseases affecting the central nervous system (CNS). Precise targeting with an optimal control over gene regulation confers long-lasting benefits through the administration of nucleic acid payloads via viral, non-viral, and engineered vectors. The current review majorly focuses on the development and clinical translational potential of non-viral vectors for treating CNS diseases with a focus on their specific design and targeting approaches. These carriers must be able to surmount the various intracellular and extracellular barriers, to ensure successful neuronal transfection and ultimately attain higher therapeutic efficacies. Additionally, the specific challenges associated with CNS administration also include the presence of blood-brain barrier (BBB), the complex pathophysiological and biochemical changes associated with different disease conditions and the existence of non-dividing cells. The advantages offered by lipid-based or polymeric systems, engineered proteins, particle-based systems coupled with various approaches of neuronal targeting have been discussed in the context of a variety of CNS diseases. The possibilities of rapid yet highly efficient gene modifications rendered by the breakthrough methodologies for gene editing and gene manipulation have also opened vast avenues of research in neuroscience and CNS disease therapy. The current review also underscores the extensive scientific efforts to optimize specialized, efficacious yet non-invasive and safer administration approaches to overcome the therapeutic delivery challenges specifically posed by the CNS transport barriers and the overall obstacles to clinical translation.
Collapse
Affiliation(s)
- Smrithi Padmakumar
- Department of Pharmaceutical Sciences, School of Pharmacy, Northeastern University, Boston, MA 02115, USA
| | - Anisha D'Souza
- Department of Pharmaceutical Sciences, School of Pharmacy, Northeastern University, Boston, MA 02115, USA; Massachusetts Eye and Ear Infirmary, Harvard Medical School, Boston, MA 20115, USA
| | - Neha N Parayath
- Department of Pharmaceutical Sciences, School of Pharmacy, Northeastern University, Boston, MA 02115, USA
| | - Benjamin S Bleier
- Massachusetts Eye and Ear Infirmary, Harvard Medical School, Boston, MA 20115, USA
| | - Mansoor M Amiji
- Department of Pharmaceutical Sciences, School of Pharmacy, Northeastern University, Boston, MA 02115, USA; Department of Chemical Engineering, College of Engineering, Northeastern University, Boston, MA 02115, USA.
| |
Collapse
|
4
|
Ma L, Gilani A, Yi Q, Tang L. MicroRNAs as Mediators of Adipose Thermogenesis and Potential Therapeutic Targets for Obesity. BIOLOGY 2022; 11:1657. [PMID: 36421371 PMCID: PMC9687157 DOI: 10.3390/biology11111657] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/09/2022] [Revised: 11/09/2022] [Accepted: 11/11/2022] [Indexed: 07/30/2023]
Abstract
Obesity is a growing health problem worldwide, associated with an increased risk of multiple chronic diseases. The thermogenic activity of brown adipose tissue (BAT) correlates with leanness in adults. Understanding the mechanisms behind BAT activation and the process of white fat "browning" has important implications for developing new treatments to combat obesity. MicroRNAs (miRNAs) are small transcriptional regulators that control gene expression in various tissues, including adipose tissue. Recent studies show that miRNAs are involved in adipogenesis and adipose tissue thermogenesis. In this review, we discuss recent advances in the role of miRNAs in adipocyte thermogenesis and obesity. The potential for miRNA-based therapies for obesity and recommendations for future research are highlighted, which may help provide new targets for treating obesity and obesity-related diseases.
Collapse
Affiliation(s)
- Lunkun Ma
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400044, China
| | - Ankit Gilani
- Weill Center for Metabolic Health, Cardiovascular Research Institute, Division of Cardiology, Department of Medicine, Weill Cornell Medicine, New York, NY 10021, USA
| | - Qian Yi
- Department of Physiology, School of Basic Medical Science, Southwest Medical University, Luzhou 646099, China
| | - Liling Tang
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400044, China
| |
Collapse
|
5
|
Emerging trends in the nanomedicine applications of functionalized magnetic nanoparticles as novel therapies for acute and chronic diseases. J Nanobiotechnology 2022; 20:393. [PMID: 36045375 PMCID: PMC9428876 DOI: 10.1186/s12951-022-01595-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Accepted: 08/13/2022] [Indexed: 11/10/2022] Open
Abstract
High-quality point-of-care is critical for timely decision of disease diagnosis and healthcare management. In this regard, biosensors have revolutionized the field of rapid testing and screening, however, are confounded by several technical challenges including material cost, half-life, stability, site-specific targeting, analytes specificity, and detection sensitivity that affect the overall diagnostic potential and therapeutic profile. Despite their advances in point-of-care testing, very few classical biosensors have proven effective and commercially viable in situations of healthcare emergency including the recent COVID-19 pandemic. To overcome these challenges functionalized magnetic nanoparticles (MNPs) have emerged as key players in advancing the biomedical and healthcare sector with promising applications during the ongoing healthcare crises. This critical review focus on understanding recent developments in theranostic applications of functionalized magnetic nanoparticles (MNPs). Given the profound global economic and health burden, we discuss the therapeutic impact of functionalized MNPs in acute and chronic diseases like small RNA therapeutics, vascular diseases, neurological disorders, and cancer, as well as for COVID-19 testing. Lastly, we culminate with a futuristic perspective on the scope of this field and provide an insight into the emerging opportunities whose impact is anticipated to disrupt the healthcare industry.
Collapse
|
6
|
Bansal A, Shikha S, Zhang Y. Towards translational optogenetics. Nat Biomed Eng 2022; 7:349-369. [PMID: 35027688 DOI: 10.1038/s41551-021-00829-3] [Citation(s) in RCA: 74] [Impact Index Per Article: 24.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2018] [Accepted: 10/21/2021] [Indexed: 02/07/2023]
Abstract
Optogenetics is widely used to interrogate the neural circuits underlying disease and has most recently been harnessed for therapeutic applications. The optogenetic toolkit consists of light-responsive proteins that modulate specific cellular functions, vectors for the delivery of the transgenes that encode the light-responsive proteins to targeted cellular populations, and devices for the delivery of light of suitable wavelengths at effective fluence rates. A refined toolkit with a focus towards translational uses would include efficient and safer viral and non-viral gene-delivery vectors, increasingly red-shifted photoresponsive proteins, nanomaterials that efficiently transduce near-infrared light deep into tissue, and wireless implantable light-delivery devices that allow for spatiotemporally precise interventions at clinically relevant tissue depths. In this Review, we examine the current optogenetics toolkit and the most notable preclinical and translational uses of optogenetics, and discuss future methodological and translational developments and bottlenecks.
Collapse
Affiliation(s)
- Akshaya Bansal
- Department of Biomedical Engineering, Faculty of Engineering, National University of Singapore, Singapore, Singapore
| | - Swati Shikha
- Department of Biomedical Engineering, Faculty of Engineering, National University of Singapore, Singapore, Singapore
| | - Yong Zhang
- Department of Biomedical Engineering, Faculty of Engineering, National University of Singapore, Singapore, Singapore. .,NUS Graduate School for Integrative Sciences and Engineering, National University of Singapore, Singapore, Singapore. .,NUS Suzhou Research Institute, Suzhou, Jiangsu, P. R. China.
| |
Collapse
|
7
|
Al Qtaish N, Gallego I, Villate-Beitia I, Sainz-Ramos M, Martínez-Navarrete G, Soto-Sánchez C, Fernández E, Gálvez-Martín P, Lopez-Mendez TB, Puras G, Luis Pedraz J. Sphingolipid extracts enhance gene delivery of cationic lipid vesicles into retina and brain. Eur J Pharm Biopharm 2021; 169:103-112. [PMID: 34606927 DOI: 10.1016/j.ejpb.2021.09.011] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Revised: 09/23/2021] [Accepted: 09/27/2021] [Indexed: 11/15/2022]
Abstract
The aim was to evaluate relevant biophysic processes related to the physicochemical features and gene transfection mechanism when sphingolipids are incorporated into a cationic niosome formulation for non-viral gene delivery to central nervous system. For that, two formulations named niosphingosomes and niosomes devoid of sphingolipid extracts, as control, were developed by the oil-in water emulsion technique. Both formulations and the corresponding complexes, obtained upon the addition of the reporter EGFP plasmid, were physicochemically and biologically characterized and evaluated. Compared to niosomes, niosphingosomes, and the corresponding complexes decreased particle size and increased superficial charge. Although there were not significant differences in the cellular uptake, cell viability and transfection efficiency increased when human retinal pigment epithelial (ARPE-19) cells were exposed to niosphingoplexes. Endocytosis via caveolae decreased in the case of niosphingoplexes, which showed higher co-localization with lysosomal compartment, and endosomal escape properties. Moreover, niosphingoplexes transfected not only primary central nervous system cells, but also different cells in mouse retina, depending on the administration route, and brain cortex. These preliminary results suggest that niosphingosomes represent a promising non-viral vector formulation purposed for the treatment of both retinal and brain diseases by gene therapy approach.
Collapse
Affiliation(s)
- Nuseibah Al Qtaish
- NanoBioCel Research Group, Laboratory of Pharmacy and Pharmaceutical Technology. Faculty of Pharmacy, University of the Basque Country (UPV/EHU), Paseo de la Universidad 7, 01006 Vitoria-Gasteiz, Spain; Networking Research Centre of Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Institute of Health Carlos III, Av Monforte de Lemos 3-5, 28029 Madrid, Spain.
| | - Idoia Gallego
- NanoBioCel Research Group, Laboratory of Pharmacy and Pharmaceutical Technology. Faculty of Pharmacy, University of the Basque Country (UPV/EHU), Paseo de la Universidad 7, 01006 Vitoria-Gasteiz, Spain; Networking Research Centre of Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Institute of Health Carlos III, Av Monforte de Lemos 3-5, 28029 Madrid, Spain; Bioaraba, NanoBioCel Research Group, Calle José Achotegui s/n, 01009 Vitoria-Gasteiz, Spain.
| | - Ilia Villate-Beitia
- NanoBioCel Research Group, Laboratory of Pharmacy and Pharmaceutical Technology. Faculty of Pharmacy, University of the Basque Country (UPV/EHU), Paseo de la Universidad 7, 01006 Vitoria-Gasteiz, Spain; Networking Research Centre of Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Institute of Health Carlos III, Av Monforte de Lemos 3-5, 28029 Madrid, Spain; Bioaraba, NanoBioCel Research Group, Calle José Achotegui s/n, 01009 Vitoria-Gasteiz, Spain.
| | - Myriam Sainz-Ramos
- NanoBioCel Research Group, Laboratory of Pharmacy and Pharmaceutical Technology. Faculty of Pharmacy, University of the Basque Country (UPV/EHU), Paseo de la Universidad 7, 01006 Vitoria-Gasteiz, Spain; Networking Research Centre of Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Institute of Health Carlos III, Av Monforte de Lemos 3-5, 28029 Madrid, Spain; Bioaraba, NanoBioCel Research Group, Calle José Achotegui s/n, 01009 Vitoria-Gasteiz, Spain.
| | - Gema Martínez-Navarrete
- Networking Research Centre of Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Institute of Health Carlos III, Av Monforte de Lemos 3-5, 28029 Madrid, Spain; Neuroprothesis and Neuroengineering Research Group, Institute of Bioengineering, Miguel Hernández University, Avenida de la Universidad, 03202 Elche, Spain.
| | - Cristina Soto-Sánchez
- Networking Research Centre of Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Institute of Health Carlos III, Av Monforte de Lemos 3-5, 28029 Madrid, Spain; Neuroprothesis and Neuroengineering Research Group, Institute of Bioengineering, Miguel Hernández University, Avenida de la Universidad, 03202 Elche, Spain.
| | - Eduardo Fernández
- Networking Research Centre of Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Institute of Health Carlos III, Av Monforte de Lemos 3-5, 28029 Madrid, Spain; Neuroprothesis and Neuroengineering Research Group, Institute of Bioengineering, Miguel Hernández University, Avenida de la Universidad, 03202 Elche, Spain.
| | | | - Tania B Lopez-Mendez
- NanoBioCel Research Group, Laboratory of Pharmacy and Pharmaceutical Technology. Faculty of Pharmacy, University of the Basque Country (UPV/EHU), Paseo de la Universidad 7, 01006 Vitoria-Gasteiz, Spain; Networking Research Centre of Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Institute of Health Carlos III, Av Monforte de Lemos 3-5, 28029 Madrid, Spain; Bioaraba, NanoBioCel Research Group, Calle José Achotegui s/n, 01009 Vitoria-Gasteiz, Spain.
| | - Gustavo Puras
- NanoBioCel Research Group, Laboratory of Pharmacy and Pharmaceutical Technology. Faculty of Pharmacy, University of the Basque Country (UPV/EHU), Paseo de la Universidad 7, 01006 Vitoria-Gasteiz, Spain; Networking Research Centre of Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Institute of Health Carlos III, Av Monforte de Lemos 3-5, 28029 Madrid, Spain; Bioaraba, NanoBioCel Research Group, Calle José Achotegui s/n, 01009 Vitoria-Gasteiz, Spain.
| | - José Luis Pedraz
- NanoBioCel Research Group, Laboratory of Pharmacy and Pharmaceutical Technology. Faculty of Pharmacy, University of the Basque Country (UPV/EHU), Paseo de la Universidad 7, 01006 Vitoria-Gasteiz, Spain; Networking Research Centre of Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Institute of Health Carlos III, Av Monforte de Lemos 3-5, 28029 Madrid, Spain; Bioaraba, NanoBioCel Research Group, Calle José Achotegui s/n, 01009 Vitoria-Gasteiz, Spain.
| |
Collapse
|
8
|
Rinoldi C, Zargarian SS, Nakielski P, Li X, Liguori A, Petronella F, Presutti D, Wang Q, Costantini M, De Sio L, Gualandi C, Ding B, Pierini F. Nanotechnology-Assisted RNA Delivery: From Nucleic Acid Therapeutics to COVID-19 Vaccines. SMALL METHODS 2021; 5:e2100402. [PMID: 34514087 PMCID: PMC8420172 DOI: 10.1002/smtd.202100402] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 07/04/2021] [Indexed: 05/07/2023]
Abstract
In recent years, the main quest of science has been the pioneering of the groundbreaking biomedical strategies needed for achieving a personalized medicine. Ribonucleic acids (RNAs) are outstanding bioactive macromolecules identified as pivotal actors in regulating a wide range of biochemical pathways. The ability to intimately control the cell fate and tissue activities makes RNA-based drugs the most fascinating family of bioactive agents. However, achieving a widespread application of RNA therapeutics in humans is still a challenging feat, due to both the instability of naked RNA and the presence of biological barriers aimed at hindering the entrance of RNA into cells. Recently, material scientists' enormous efforts have led to the development of various classes of nanostructured carriers customized to overcome these limitations. This work systematically reviews the current advances in developing the next generation of drugs based on nanotechnology-assisted RNA delivery. The features of the most used RNA molecules are presented, together with the development strategies and properties of nanostructured vehicles. Also provided is an in-depth overview of various therapeutic applications of the presented systems, including coronavirus disease vaccines and the newest trends in the field. Lastly, emerging challenges and future perspectives for nanotechnology-mediated RNA therapies are discussed.
Collapse
Affiliation(s)
- Chiara Rinoldi
- Department of Biosystems and Soft MatterInstitute of Fundamental Technological ResearchPolish Academy of Sciencesul. Pawińskiego 5BWarsaw02‐106Poland
| | - Seyed Shahrooz Zargarian
- Department of Biosystems and Soft MatterInstitute of Fundamental Technological ResearchPolish Academy of Sciencesul. Pawińskiego 5BWarsaw02‐106Poland
| | - Pawel Nakielski
- Department of Biosystems and Soft MatterInstitute of Fundamental Technological ResearchPolish Academy of Sciencesul. Pawińskiego 5BWarsaw02‐106Poland
| | - Xiaoran Li
- Innovation Center for Textile Science and TechnologyDonghua UniversityWest Yan'an Road 1882Shanghai200051China
| | - Anna Liguori
- Department of Chemistry “Giacomo Ciamician” and INSTM UdR of BolognaUniversity of BolognaVia Selmi 2Bologna40126Italy
| | - Francesca Petronella
- Institute of Crystallography CNR‐ICNational Research Council of ItalyVia Salaria Km 29.300Monterotondo – Rome00015Italy
| | - Dario Presutti
- Institute of Physical ChemistryPolish Academy of Sciencesul. M. Kasprzaka 44/52Warsaw01‐224Poland
| | - Qiusheng Wang
- Innovation Center for Textile Science and TechnologyDonghua UniversityWest Yan'an Road 1882Shanghai200051China
| | - Marco Costantini
- Institute of Physical ChemistryPolish Academy of Sciencesul. M. Kasprzaka 44/52Warsaw01‐224Poland
| | - Luciano De Sio
- Department of Medico‐Surgical Sciences and BiotechnologiesResearch Center for BiophotonicsSapienza University of RomeCorso della Repubblica 79Latina04100Italy
- CNR‐Lab. LicrylInstitute NANOTECArcavacata di Rende87036Italy
| | - Chiara Gualandi
- Department of Chemistry “Giacomo Ciamician” and INSTM UdR of BolognaUniversity of BolognaVia Selmi 2Bologna40126Italy
- Interdepartmental Center for Industrial Research on Advanced Applications in Mechanical Engineering and Materials TechnologyCIRI‐MAMUniversity of BolognaViale Risorgimento 2Bologna40136Italy
| | - Bin Ding
- Innovation Center for Textile Science and TechnologyDonghua UniversityWest Yan'an Road 1882Shanghai200051China
| | - Filippo Pierini
- Department of Biosystems and Soft MatterInstitute of Fundamental Technological ResearchPolish Academy of Sciencesul. Pawińskiego 5BWarsaw02‐106Poland
| |
Collapse
|
9
|
Understanding MNPs Behaviour in Response to AMF in Biological Milieus and the Effects at the Cellular Level: Implications for a Rational Design That Drives Magnetic Hyperthermia Therapy toward Clinical Implementation. Cancers (Basel) 2021; 13:cancers13184583. [PMID: 34572810 PMCID: PMC8465027 DOI: 10.3390/cancers13184583] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Revised: 09/03/2021] [Accepted: 09/07/2021] [Indexed: 12/11/2022] Open
Abstract
Simple Summary Magnetic hyperthermia therapy is an alternative treatment for cancer that complements traditional therapies and that has shown great promise in recent years. In this review, we assess the current applications of this therapy in order to understand why its translation from the laboratory to the clinic has been less smooth than was anticipated, identifying the possible bottlenecks and proposing solutions to the problems encountered. Abstract Hyperthermia has emerged as a promising alternative to conventional cancer therapies and in fact, traditional hyperthermia is now commonly used in combination with chemotherapy or surgery during cancer treatment. Nevertheless, non-specific application of hyperthermia generates various undesirable side-effects, such that nano-magnetic hyperthermia has arisen a possible solution to this problem. This technique to induce hyperthermia is based on the intrinsic capacity of magnetic nanoparticles to accumulate in a given target area and to respond to alternating magnetic fields (AMFs) by releasing heat, based on different principles of physics. Unfortunately, the clinical implementation of nano-magnetic hyperthermia has not been fluid and few clinical trials have been carried out. In this review, we want to demonstrate the need for more systematic and basic research in this area, as many of the sub-cellular and molecular mechanisms associated with this approach remain unclear. As such, we shall consider here the biological effects that occur and why this theoretically well-designed nano-system fails in physiological conditions. Moreover, we will offer some guidelines that may help establish successful strategies through the rational design of magnetic nanoparticles for magnetic hyperthermia.
Collapse
|
10
|
Sizikov AA, Kharlamova MV, Nikitin MP, Nikitin PI, Kolychev EL. Nonviral Locally Injected Magnetic Vectors for In Vivo Gene Delivery: A Review of Studies on Magnetofection. NANOMATERIALS (BASEL, SWITZERLAND) 2021; 11:1078. [PMID: 33922066 PMCID: PMC8143545 DOI: 10.3390/nano11051078] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Revised: 04/19/2021] [Accepted: 04/19/2021] [Indexed: 12/20/2022]
Abstract
Magnetic nanoparticles have been widely used in nanobiomedicine for diagnostics and the treatment of diseases, and as carriers for various drugs. The unique magnetic properties of "magnetic" drugs allow their delivery in a targeted tumor or tissue upon application of a magnetic field. The approach of combining magnetic drug targeting and gene delivery is called magnetofection, and it is very promising. This method is simple and efficient for the delivery of genetic material to cells using magnetic nanoparticles controlled by an external magnetic field. However, magnetofection in vivo has been studied insufficiently both for local and systemic routes of magnetic vector injection, and the relevant data available in the literature are often merely descriptive and contradictory. In this review, we collected and systematized the data on the efficiency of the local injections of magnetic nanoparticles that carry genetic information upon application of external magnetic fields. We also investigated the efficiency of magnetofection in vivo, depending on the structure and coverage of magnetic vectors. The perspectives of the development of the method were also considered.
Collapse
Affiliation(s)
- Artem A. Sizikov
- Moscow Institute of Physics and Technology, 141701 Dolgoprudny, Russia; (A.A.S.); (M.V.K.); (M.P.N.)
| | - Marianna V. Kharlamova
- Moscow Institute of Physics and Technology, 141701 Dolgoprudny, Russia; (A.A.S.); (M.V.K.); (M.P.N.)
| | - Maxim P. Nikitin
- Moscow Institute of Physics and Technology, 141701 Dolgoprudny, Russia; (A.A.S.); (M.V.K.); (M.P.N.)
- Sirius University of Science and Technology, 354340 Sochi, Russia
| | - Petr I. Nikitin
- Prokhorov General Physics Institute of the Russian Academy of Sciences, 117942 Moscow, Russia
| | - Eugene L. Kolychev
- Moscow Institute of Physics and Technology, 141701 Dolgoprudny, Russia; (A.A.S.); (M.V.K.); (M.P.N.)
| |
Collapse
|
11
|
Dasgupta I, Chatterjee A. Recent Advances in miRNA Delivery Systems. Methods Protoc 2021; 4:mps4010010. [PMID: 33498244 PMCID: PMC7839010 DOI: 10.3390/mps4010010] [Citation(s) in RCA: 186] [Impact Index Per Article: 46.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2020] [Revised: 01/14/2021] [Accepted: 01/15/2021] [Indexed: 12/27/2022] Open
Abstract
MicroRNAs (miRNAs) represent a family of short non-coding regulatory RNA molecules that are produced in a tissue and time-specific manner to orchestrate gene expression post-transcription. MiRNAs hybridize to target mRNA(s) to induce translation repression or mRNA degradation. Functional studies have demonstrated that miRNAs are engaged in virtually every physiological process and, consequently, miRNA dysregulations have been linked to multiple human pathologies. Thus, miRNA mimics and anti-miRNAs that restore miRNA expression or downregulate aberrantly expressed miRNAs, respectively, are highly sought-after therapeutic strategies for effective manipulation of miRNA levels. In this regard, carrier vehicles that facilitate proficient and safe delivery of miRNA-based therapeutics are fundamental to the clinical success of these pharmaceuticals. Here, we highlight the strengths and weaknesses of current state-of-the-art viral and non-viral miRNA delivery systems and provide perspective on how these tools can be exploited to improve the outcomes of miRNA-based therapeutics.
Collapse
Affiliation(s)
- Ishani Dasgupta
- Horae Gene Therapy Center, Department of Pediatrics, University of Massachusetts Medical School, Worcester, MA 01605, USA;
| | - Anushila Chatterjee
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, CO 80045, USA
- Correspondence:
| |
Collapse
|
12
|
Huang RY, Liu ZH, Weng WH, Chang CW. Magnetic nanocomplexes for gene delivery applications. J Mater Chem B 2021; 9:4267-4286. [PMID: 33942822 DOI: 10.1039/d0tb02713h] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Gene delivery is an indispensable technique for various biomedical applications such as gene therapy, stem cell engineering and gene editing. Recently, magnetic nanoparticles (MNPs) have received increasing attention for their use in promoting gene delivery efficiency. Under magnetic attraction, gene delivery efficiency using viral or nonviral gene carriers could be universally enhanced. Besides, magnetic nanoparticles could be utilized in magnetic resonance imaging or magnetic hyperthermia therapy, providing extra theranostic opportunities. In this review, recent research integrating MNPs with a viral or nonviral gene vector is summarized from both technical and application perspectives. Applications of MNPs in cutting-edge research technologies, such as biomimetic cell membrane nano-gene carriers, exosome-based gene delivery, cell-based drug delivery systems or CRISPR/Cas9 gene editing, are also discussed.
Collapse
Affiliation(s)
- Rih-Yang Huang
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu, Taiwan.
| | - Zhuo-Hao Liu
- Department of Neurosurgery, Chang Gung Memorial Hospital at Linkou, Chang Gung Medical College and University, Taiwan.
| | - Wei-Han Weng
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu, Taiwan.
| | - Chien-Wen Chang
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu, Taiwan.
| |
Collapse
|
13
|
Hurst J, Fietz A, Tsai T, Joachim SC, Schnichels S. Organ Cultures for Retinal Diseases. Front Neurosci 2020; 14:583392. [PMID: 33324149 PMCID: PMC7724035 DOI: 10.3389/fnins.2020.583392] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Accepted: 10/13/2020] [Indexed: 12/18/2022] Open
Abstract
The successful development of novel therapies is closely linked with understanding the underlying pathomechanisms of a disease. To do so, model systems that reflect human diseases and allow for the evaluation of new therapeutic approaches are needed. Yet, preclinical animal studies often have limited success in predicting human physiology, pathology, and therapeutic responses. Moreover, animal testing is facing increasing ethical and bureaucratic hurdles, while human cell cultures are limited in their ability to represent in vivo situations due to the lack of the tissue microenvironment, which may alter cellular responses. To overcome these struggles, organ cultures, especially those of complex organs such as the retina, can be used to study physiological reactions to substances or stressors. Human and animal organ cultures are now well established and recognized. This mini-review discusses how retinal organ cultures can be used to preserve tissue architecture more realistically and therefore better represent disease-related changes. It also shows how molecular biological, biochemical, and histological techniques can be combined to investigate how anatomical localization may alter cellular responses. Examples for the use of retinal organ cultures, including models to study age-related macular degeneration (AMD), retinitis pigmentosa (RP), central artery occlusion (CRAO), and glaucoma are presented, and their advantages and disadvantages are discussed. We conclude that organ cultures significantly improve our understanding of complex retinal diseases and may advance treatment testing without the need for animal testing.
Collapse
Affiliation(s)
- José Hurst
- Center for Ophthalmology, University Eye Hospital, University of Tübingen, Tübingen, Germany
| | - Agnes Fietz
- Center for Ophthalmology, University Eye Hospital, University of Tübingen, Tübingen, Germany
| | - Teresa Tsai
- Experimental Eye Research Institute, University Eye Hospital, Ruhr-University Bochum, Bochum, Germany
| | - Stephanie C. Joachim
- Experimental Eye Research Institute, University Eye Hospital, Ruhr-University Bochum, Bochum, Germany
| | - Sven Schnichels
- Center for Ophthalmology, University Eye Hospital, University of Tübingen, Tübingen, Germany
| |
Collapse
|
14
|
Gallego I, Villate-Beitia I, Soto-Sánchez C, Menéndez M, Grijalvo S, Eritja R, Martínez-Navarrete G, Humphreys L, López-Méndez T, Puras G, Fernández E, Pedraz JL. Brain Angiogenesis Induced by Nonviral Gene Therapy with Potential Therapeutic Benefits for Central Nervous System Diseases. Mol Pharm 2020; 17:1848-1858. [PMID: 32293897 DOI: 10.1021/acs.molpharmaceut.9b01213] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Gene therapy employing nanocarriers represents a promising strategy to treat central nervous system (CNS) diseases, where brain microvasculature is frequently compromised. Vascular endothelial growth factor (VEGF) is a key angiogenic molecule; however, its in vivo administration to the CNS by nonviral gene therapy has not been conducted. Hence, we prepared and physicochemically characterized four cationic niosome formulations (1-4), which were combined with pVEGF-GFP to explore their capacity to transfer the VEGF gene to CNS cells and achieve angiogenesis in the brain. Experiments in primary neuronal cells showed successful and safe transfection with niosome 4, producing double levels of biologically active VEGF in comparison to the rest of the formulations. Intracortical administration of niosome 4 based nioplexes in mouse brain validated the ability of this nonviral vector to deliver the VEGF gene to CNS cells, inducing brain angiogenesis and emerging as a promising therapeutic approach for the treatment of CNS diseases.
Collapse
Affiliation(s)
- Idoia Gallego
- NanoBioCel Group, University of the Basque Country (UPV/EHU), Vitoria-Gasteiz 01006, Spain.,Biomedical Research Networking Center in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Madrid 28029, Spain
| | - Ilia Villate-Beitia
- NanoBioCel Group, University of the Basque Country (UPV/EHU), Vitoria-Gasteiz 01006, Spain.,Biomedical Research Networking Center in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Madrid 28029, Spain
| | - Cristina Soto-Sánchez
- Biomedical Research Networking Center in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Madrid 28029, Spain.,Neuroprothesis and Neuroengineering Research Group, Miguel Hernández University, Elche 03202, Spain
| | - Margarita Menéndez
- Rocasolano Physical Chemistry Institute, Superior Council of Scientific Investigations (IQFR-CSIC), Madrid 28006, Spain.,Biomedical Research Networking Center in Respiratory Diseases (CIBERES), Madrid 28029, Spain
| | - Santiago Grijalvo
- Biomedical Research Networking Center in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Madrid 28029, Spain.,Institute of Advanced Chemistry of Catalonia (IQAC-CSIC), Barcelona 08034, Spain
| | - Ramón Eritja
- Biomedical Research Networking Center in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Madrid 28029, Spain.,Institute of Advanced Chemistry of Catalonia (IQAC-CSIC), Barcelona 08034, Spain
| | - Gema Martínez-Navarrete
- Biomedical Research Networking Center in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Madrid 28029, Spain.,Neuroprothesis and Neuroengineering Research Group, Miguel Hernández University, Elche 03202, Spain
| | - Lawrence Humphreys
- Biomedical Research Networking Center in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Madrid 28029, Spain.,Neuroprothesis and Neuroengineering Research Group, Miguel Hernández University, Elche 03202, Spain
| | - Tania López-Méndez
- NanoBioCel Group, University of the Basque Country (UPV/EHU), Vitoria-Gasteiz 01006, Spain.,Biomedical Research Networking Center in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Madrid 28029, Spain
| | - Gustavo Puras
- NanoBioCel Group, University of the Basque Country (UPV/EHU), Vitoria-Gasteiz 01006, Spain.,Biomedical Research Networking Center in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Madrid 28029, Spain
| | - Eduardo Fernández
- Biomedical Research Networking Center in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Madrid 28029, Spain.,Neuroprothesis and Neuroengineering Research Group, Miguel Hernández University, Elche 03202, Spain
| | - José Luis Pedraz
- NanoBioCel Group, University of the Basque Country (UPV/EHU), Vitoria-Gasteiz 01006, Spain.,Biomedical Research Networking Center in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Madrid 28029, Spain
| |
Collapse
|
15
|
Titze-de-Almeida SS, Soto-Sánchez C, Fernandez E, Koprich JB, Brotchie JM, Titze-de-Almeida R. The Promise and Challenges of Developing miRNA-Based Therapeutics for Parkinson's Disease. Cells 2020; 9:cells9040841. [PMID: 32244357 PMCID: PMC7226753 DOI: 10.3390/cells9040841] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Revised: 03/16/2020] [Accepted: 03/18/2020] [Indexed: 02/07/2023] Open
Abstract
MicroRNAs (miRNAs) are small double-stranded RNAs that exert a fine-tuning sequence-specific regulation of cell transcriptome. While one unique miRNA regulates hundreds of mRNAs, each mRNA molecule is commonly regulated by various miRNAs that bind to complementary sequences at 3’-untranslated regions for triggering the mechanism of RNA interference. Unfortunately, dysregulated miRNAs play critical roles in many disorders, including Parkinson’s disease (PD), the second most prevalent neurodegenerative disease in the world. Treatment of this slowly, progressive, and yet incurable pathology challenges neurologists. In addition to L-DOPA that restores dopaminergic transmission and ameliorate motor signs (i.e., bradykinesia, rigidity, tremors), patients commonly receive medication for mood disorders and autonomic dysfunctions. However, the effectiveness of L-DOPA declines over time, and the L-DOPA-induced dyskinesias commonly appear and become highly disabling. The discovery of more effective therapies capable of slowing disease progression –a neuroprotective agent–remains a critical need in PD. The present review focus on miRNAs as promising drug targets for PD, examining their role in underlying mechanisms of the disease, the strategies for controlling aberrant expressions, and, finally, the current technologies for translating these small molecules from bench to clinics.
Collapse
Affiliation(s)
- Simoneide S. Titze-de-Almeida
- Technology for Gene Therapy Laboratory, Central Institute of Sciences, FAV, University of Brasilia, Brasília 70910-900, Brazil;
| | - Cristina Soto-Sánchez
- Neuroprosthetics and Visual Rehabilitation Research Unit, Bioengineering Institute, Miguel Hernández University, 03202 Alicante, Spain; (C.S.-S.); (E.F.)
| | - Eduardo Fernandez
- Neuroprosthetics and Visual Rehabilitation Research Unit, Bioengineering Institute, Miguel Hernández University, 03202 Alicante, Spain; (C.S.-S.); (E.F.)
- Biomedical Research Networking Center in Bioengineering, Biomaterials and Nanomedicine—CIBER-BBN, 28029 Madrid, Spain
| | - James B. Koprich
- Krembil Neuroscience Centre, Toronto Western Hospital, University Health Network, Toronto, Ontario M5T 2S8, Canada; (J.B.K.); (J.M.B.)
| | - Jonathan M. Brotchie
- Krembil Neuroscience Centre, Toronto Western Hospital, University Health Network, Toronto, Ontario M5T 2S8, Canada; (J.B.K.); (J.M.B.)
| | - Ricardo Titze-de-Almeida
- Technology for Gene Therapy Laboratory, Central Institute of Sciences, FAV, University of Brasilia, Brasília 70910-900, Brazil;
- Correspondence: ; Tel.: +55-61-3107-7222
| |
Collapse
|
16
|
Funnell JL, Balouch B, Gilbert RJ. Magnetic Composite Biomaterials for Neural Regeneration. Front Bioeng Biotechnol 2019; 7:179. [PMID: 31404143 PMCID: PMC6669379 DOI: 10.3389/fbioe.2019.00179] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2019] [Accepted: 07/10/2019] [Indexed: 12/11/2022] Open
Abstract
Nervous system damage caused by physical trauma or degenerative diseases can result in loss of sensory and motor function for patients. Biomaterial interventions have shown promise in animal studies, providing contact guidance for extending neurites or sustained release of various drugs and growth factors; however, these approaches often target only one aspect of the regeneration process. More recent studies investigate hybrid approaches, creating complex materials that can reduce inflammation or provide neuroprotection in addition to stimulating growth and regeneration. Magnetic materials have shown promise in this field, as they can be manipulated non-invasively, are easily functionalized, and can be used to mechanically stimulate cells. By combining different types of biomaterials (hydrogels, nanoparticles, electrospun fibers) and incorporating magnetic elements, magnetic materials can provide multiple physical and chemical cues to promote regeneration. This review, for the first time, will provide an overview of design strategies for promoting regeneration after neural injury with magnetic biomaterials.
Collapse
Affiliation(s)
| | | | - Ryan J. Gilbert
- Department of Biomedical Engineering, Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, NY, United States
| |
Collapse
|
17
|
Jiang C, Li HT, Zhou YM, Wang X, Wang L, Liu ZQ. Cardiac optogenetics: a novel approach to cardiovascular disease therapy. Europace 2019; 20:1741-1749. [PMID: 29253159 DOI: 10.1093/europace/eux345] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2017] [Accepted: 10/24/2017] [Indexed: 12/13/2022] Open
Abstract
Optogenetics is a cell-type specific and high spatial-temporal resolution method that combines genetic encoding of light-sensitive proteins and optical manipulation techniques. Optogenetics technology provides a novel approach for research on cardiac arrhythmia treatment, including pacing, recovering the conduction system, and achieving cardiac resynchronization with precise and low-energy optical control. Photosensitive proteins, which usually act as ion channels, pumps, or receptors, are delivered to target cells, where they respond to light pulses of specific wavelengths, evoke transient flows of transmembrane ion currents, and induce signal transmission. With the development of gene technology, the in vivo efficiency of optogenetics in cardiology has been trialed, and in vitro experiments have been performed to test its potential in cardiac electrophysiology. Challenges for applying optogenetics in large animals and humans include the effectiveness, safety, and long-term expression of photosensitive proteins, unscattered and unattenuated exogenous light stimulation, and the need for implantable miniature light stimulators. Photosensitive proteins, genetic engineering technology, and light equipment are essential for experiments in cardiac optogenetics. Optogenetics may provide an alternative method for evaluating the mechanism of cardiac arrhythmias, testing hypotheses, and treating cardiovascular diseases.
Collapse
Affiliation(s)
- Chan Jiang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, PR China.,Cardiovascular Research Institute, Wuhan University, Wuhan, PR China.,Hubei Key Laboratory of Cardiology, Wuhan, PR China
| | - Hai Tao Li
- Department of Cardiology, Hainan General Hospital, Haikou, PR China
| | - Yong Ming Zhou
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, PR China.,Cardiovascular Research Institute, Wuhan University, Wuhan, PR China.,Hubei Key Laboratory of Cardiology, Wuhan, PR China
| | - Xi Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, PR China.,Cardiovascular Research Institute, Wuhan University, Wuhan, PR China.,Hubei Key Laboratory of Cardiology, Wuhan, PR China
| | - Long Wang
- Cardiovascular Research Institute, Wuhan University, Wuhan, PR China.,Hubei Key Laboratory of Cardiology, Wuhan, PR China.,Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, PR China
| | - Zi Qiang Liu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, PR China.,Cardiovascular Research Institute, Wuhan University, Wuhan, PR China.,Hubei Key Laboratory of Cardiology, Wuhan, PR China
| |
Collapse
|
18
|
Titze-de-Almeida R, Titze-de-Almeida SS, Ferreira NR, Fontanari C, Faccioli LH, Del Bel E. Suppressing nNOS Enzyme by Small-Interfering RNAs Protects SH-SY5Y Cells and Nigral Dopaminergic Neurons from 6-OHDA Injury. Neurotox Res 2019; 36:117-131. [PMID: 31041676 DOI: 10.1007/s12640-019-00043-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Revised: 03/27/2019] [Accepted: 04/04/2019] [Indexed: 01/11/2023]
Abstract
Nitric oxide (NO) has chemical properties that make it uniquely suitable as an intracellular and intercellular messenger. NO is produced by the activity of the enzyme nitric oxide synthases (NOS). There is substantial and mounting evidence that slight abnormalities of NO may underlie a wide range of neurodegenerative disorders. NO participates of the oxidative stress and inflammatory processes that contribute to the progressive dopaminergic loss in Parkinson's disease (PD). The present study aimed to evaluate in vitro and in vivo the effects of neuronal NOS-targeted siRNAs on the injury caused in dopaminergic neurons by the toxin 6-hidroxydopamine (6-OHDA). First, we confirmed (immunohistochemistry and Western blotting) that SH-SY5Y cell lineage expresses the dopaminergic marker tyrosine hydroxylase (TH) and the protein under analysis, neuronal NOS (nNOS). We designed four siRNAs by using the BIOPREDsi algorithm choosing the one providing the highest knockdown of nNOS mRNA in SH-SY5Y cells, as determined by qPCR. siRNA 4400 carried by liposomes was internalized into cells, caused a concentration-dependent knockdown on nNOS, and reduced the toxicity induced by 6-OHDA (p < 0.05). Regarding in vivo action in the dopamine-depleted animals, intra-striatal injection of siRNA 4400 at 4 days prior 6-OHDA produced a decrease in the rotational behavior induced by apomorphine. Finally, siRNA 4400 mitigated the loss of TH(+) cells in substantia nigra dorsal and ventral part. In conclusion, the suppression of nNOS enzyme by targeted siRNAs modified the progressive death of dopaminergic cells induced by 6-OHDA and merits further pre-clinical investigations as a neuroprotective approach for PD.
Collapse
Affiliation(s)
- Ricardo Titze-de-Almeida
- Technology for Gene Therapy Laboratory, Central Institute of Sciences, University of Brasília-FAV, Brasília, DF, 70910-900, Brazil
| | - Simoneide S Titze-de-Almeida
- Technology for Gene Therapy Laboratory, Central Institute of Sciences, University of Brasília-FAV, Brasília, DF, 70910-900, Brazil
| | - Nadia Rubia Ferreira
- Department of Basic and Oral Biology, Dental School, University of São Paulo (USP), Ribeirão Preto, SP, 14040-904, Brazil
| | - Caroline Fontanari
- Department of Clinical Analyses, Toxicology and Bromatology, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, 14040-903, Brazil
| | - Lúcia Helena Faccioli
- Department of Clinical Analyses, Toxicology and Bromatology, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, 14040-903, Brazil
| | - Elaine Del Bel
- Department of Basic and Oral Biology, Dental School, University of São Paulo (USP), Ribeirão Preto, SP, 14040-904, Brazil.
- Center for Interdisciplinary Research on Applied Neurosciences (NAPNA), University of São Paulo (USP), São Paulo, SP, Brazil.
| |
Collapse
|
19
|
Huang WC, Chi HS, Lee YC, Lo YC, Liu TC, Chiang MY, Chen HY, Li SJ, Chen YY, Chen SY. Gene-Embedded Nanostructural Biotic-Abiotic Optoelectrode Arrays Applied for Synchronous Brain Optogenetics and Neural Signal Recording. ACS APPLIED MATERIALS & INTERFACES 2019; 11:11270-11282. [PMID: 30844235 DOI: 10.1021/acsami.9b03264] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Optogenetics is a recently established neuromodulation technique in which photostimulation is used to manipulate neurons with high temporal and spatial precision. However, sequential genetic and optical insertion with double brain implantation tends to cause excessive tissue damage. In addition, the incorporation of light-sensitive genes requires the utilization of viral vectors, which remains a safety concern. Here, by combining device fabrication design, nanotechnology, and cell targeting technology, we developed a new gene-embedded optoelectrode array for neural implantation to enable spatiotemporal electroporation (EP) for gene delivery/transfection, photomodulation, and synchronous electrical monitoring of neural signals in the brain via one-time implantation. A biotic-abiotic neural interface (called PG) composed of reduced graphene oxide and conductive polyelectrolyte 3,4-ethylenedioxythiophene-modified amphiphilic chitosan was developed to form a nanostructural hydrogel with assembled nanodomains for encapsulating nonviral gene vectors (called PEI-NT-pDNA) formulated by neurotensin (NT) and polyethylenimine (PEI)-coupled plasmid DNA (pDNA). The PG can maintain high charge storage ability to respond to a minimal current of 125 μA for controllable gene delivery. The in vitro analysis of PG-PEI-NT-pDNA on the microelectrode array chip showed that the microelectrodes provided electrically inductive electropermeabilization, which permitted gene transfection into localized rat adrenal pheochromocytoma cells with a strong green fluorescent protein expression that was up to 8-fold higher than that in nontreated cells. Furthermore, the in vivo implantation enabled on-demand spatiotemporal gene transfection to neurons with 10-fold enhancement of targeting ability compared with astrocytes. Finally, using the real optogenetic opsin channelrhodopsin-2, the flexible neural probe incorporated with an optical waveguide fiber displayed photoevoked extracellular spikes in the thalamic ventrobasal region after focal EP for only 7 days, which provided a proof of concept for the use of photomodulation to facilitate neural therapies.
Collapse
Affiliation(s)
| | - Hui-Shang Chi
- Department of Materials Science and Engineering , National Chiao Tung University , No. 1001, Ta-Hsueh Road , Hsinchu 30010 , Taiwan , R.O.C
| | | | | | - Ta-Chung Liu
- Department of Materials Science and Engineering , National Chiao Tung University , No. 1001, Ta-Hsueh Road , Hsinchu 30010 , Taiwan , R.O.C
| | - Min-Yu Chiang
- Department of Materials Science and Engineering , National Chiao Tung University , No. 1001, Ta-Hsueh Road , Hsinchu 30010 , Taiwan , R.O.C
| | - Hsu-Yan Chen
- Department of Biomedical Engineering , National Yang Ming University , No. 155, Section 2, Linong Street , Taipei 11221 , Taiwan , R.O.C
| | - Ssu-Ju Li
- Department of Biomedical Engineering , National Yang Ming University , No. 155, Section 2, Linong Street , Taipei 11221 , Taiwan , R.O.C
| | - You-Yin Chen
- Department of Biomedical Engineering , National Yang Ming University , No. 155, Section 2, Linong Street , Taipei 11221 , Taiwan , R.O.C
| | - San-Yuan Chen
- Department of Materials Science and Engineering , National Chiao Tung University , No. 1001, Ta-Hsueh Road , Hsinchu 30010 , Taiwan , R.O.C
- Frontier Research Center on Fundamental and Applied Sciences of Matters , National Tsing Hua University , No. 101, Section 2, Kuang-Fu Road , Hsinchu 30013 , Taiwan , R.O.C
| |
Collapse
|
20
|
Du X, Wang J, Zhou Q, Zhang L, Wang S, Zhang Z, Yao C. Advanced physical techniques for gene delivery based on membrane perforation. Drug Deliv 2018; 25:1516-1525. [PMID: 29968512 PMCID: PMC6058615 DOI: 10.1080/10717544.2018.1480674] [Citation(s) in RCA: 81] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Gene delivery as a promising and valid tool has been used for treating many serious diseases that conventional drug therapies cannot cure. Due to the advancement of physical technology and nanotechnology, advanced physical gene delivery methods such as electroporation, magnetoporation, sonoporation and optoporation have been extensively developed and are receiving increasing attention, which have the advantages of briefness and nontoxicity. This review introduces the technique detail of membrane perforation, with a brief discussion for future development, with special emphasis on nanoparticles mediated optoporation that have developed as an new alternative transfection technique in the last two decades. In particular, the advanced physical approaches development and new technology are highlighted, which intends to stimulate rapid advancement of perforation techniques, develop new delivery strategies and accelerate application of these techniques in clinic.
Collapse
Affiliation(s)
- Xiaofan Du
- a Key Laboratory of Biomedical Information Engineering of Ministry of Education, Institute of Biomedical Analytical Technology and Instrumentation , School of Life Science and Technology, Xi'an Jiaotong University , Xi'an , People's Republic of China
| | - Jing Wang
- a Key Laboratory of Biomedical Information Engineering of Ministry of Education, Institute of Biomedical Analytical Technology and Instrumentation , School of Life Science and Technology, Xi'an Jiaotong University , Xi'an , People's Republic of China
| | - Quan Zhou
- a Key Laboratory of Biomedical Information Engineering of Ministry of Education, Institute of Biomedical Analytical Technology and Instrumentation , School of Life Science and Technology, Xi'an Jiaotong University , Xi'an , People's Republic of China
| | - Luwei Zhang
- a Key Laboratory of Biomedical Information Engineering of Ministry of Education, Institute of Biomedical Analytical Technology and Instrumentation , School of Life Science and Technology, Xi'an Jiaotong University , Xi'an , People's Republic of China
| | - Sijia Wang
- a Key Laboratory of Biomedical Information Engineering of Ministry of Education, Institute of Biomedical Analytical Technology and Instrumentation , School of Life Science and Technology, Xi'an Jiaotong University , Xi'an , People's Republic of China
| | - Zhenxi Zhang
- a Key Laboratory of Biomedical Information Engineering of Ministry of Education, Institute of Biomedical Analytical Technology and Instrumentation , School of Life Science and Technology, Xi'an Jiaotong University , Xi'an , People's Republic of China
| | - Cuiping Yao
- a Key Laboratory of Biomedical Information Engineering of Ministry of Education, Institute of Biomedical Analytical Technology and Instrumentation , School of Life Science and Technology, Xi'an Jiaotong University , Xi'an , People's Republic of China
| |
Collapse
|
21
|
Cwetsch AW, Pinto B, Savardi A, Cancedda L. In vivo methods for acute modulation of gene expression in the central nervous system. Prog Neurobiol 2018; 168:69-85. [PMID: 29694844 PMCID: PMC6080705 DOI: 10.1016/j.pneurobio.2018.04.008] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2017] [Revised: 04/17/2018] [Accepted: 04/20/2018] [Indexed: 12/17/2022]
Abstract
Accurate and timely expression of specific genes guarantees the healthy development and function of the brain. Indeed, variations in the correct amount or timing of gene expression lead to improper development and/or pathological conditions. Almost forty years after the first successful gene transfection in in vitro cell cultures, it is currently possible to regulate gene expression in an area-specific manner at any step of central nervous system development and in adulthood in experimental animals in vivo, even overcoming the very poor accessibility of the brain. Here, we will review the diverse approaches for acute gene transfer in vivo, highlighting their advantages and disadvantages with respect to the efficiency and specificity of transfection as well as to brain accessibility. In particular, we will present well-established chemical, physical and virus-based approaches suitable for different animal models, pointing out their current and future possible applications in basic and translational research as well as in gene therapy.
Collapse
Affiliation(s)
- Andrzej W Cwetsch
- Local Micro-environment and Brain Development Laboratory, Istituto Italiano di Tecnologia, via Morego, 30, 16163 Genova, Italy; Università degli Studi di Genova, Via Balbi, 5, 16126 Genova, Italy
| | - Bruno Pinto
- Local Micro-environment and Brain Development Laboratory, Istituto Italiano di Tecnologia, via Morego, 30, 16163 Genova, Italy; Bio@SNS, Scuola Normale Superiore, Piazza dei Cavalieri 7, 56126, Pisa, Italy
| | - Annalisa Savardi
- Local Micro-environment and Brain Development Laboratory, Istituto Italiano di Tecnologia, via Morego, 30, 16163 Genova, Italy; Università degli Studi di Genova, Via Balbi, 5, 16126 Genova, Italy
| | - Laura Cancedda
- Local Micro-environment and Brain Development Laboratory, Istituto Italiano di Tecnologia, via Morego, 30, 16163 Genova, Italy; DulbeccoTelethon Institute, Italy.
| |
Collapse
|
22
|
Delivery of miRNA-Targeted Oligonucleotides in the Rat Striatum by Magnetofection with Neuromag ®. Molecules 2018; 23:molecules23071825. [PMID: 30041414 PMCID: PMC6099620 DOI: 10.3390/molecules23071825] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2018] [Revised: 07/17/2018] [Accepted: 07/21/2018] [Indexed: 12/15/2022] Open
Abstract
MicroRNAs (miRNAs) regulate gene expression at posttranscriptional level by triggering RNA interference. In such a sense, aberrant expressions of miRNAs play critical roles in the pathogenesis of many disorders, including Parkinson’s disease (PD). Controlling the level of specific miRNAs in the brain is thus a promising therapeutic strategy for neuroprotection. A fundamental need for miRNA regulation (either replacing or inhibition) is a carrier capable of delivering oligonucleotides into brain cells. This study aimed to examine a polymeric magnetic particle, Neuromag®, for delivery of synthetic miRNA inhibitors in the rat central nervous system. We injected the miRNA inhibitor complexed with Neuromag® into the lateral ventricles next to the striatum, by stereotaxic surgery. Neuromag efficiently delivered oligonucleotides in the striatum and septum areas, as shown by microscopy imaging of fluorescein isothiocyanate (FITC)-labeled oligos in astrocytes and neurons. Transfected oligos showed efficacy concerning miRNA inhibition. Neuromag®-structured miR-134 antimiR (0.36 nmol) caused a significant 0.35 fold decrease of striatal miR-134, as revealed by real-time quantitative polymerase chain reaction (RT-qPCR). In conclusion, the polymeric magnetic particle Neuromag® efficiently delivered functional miRNA inhibitors in brain regions surrounding lateral ventricles, particularly the striatum. This delivery system holds potential as a promising miRNA-based disease-modifying drug and merits further pre-clinical studies using animal models of PD.
Collapse
|
23
|
|
24
|
Magnetic Nanoparticles in the Central Nervous System: Targeting Principles, Applications and Safety Issues. Molecules 2017; 23:molecules23010009. [PMID: 29267188 PMCID: PMC5943969 DOI: 10.3390/molecules23010009] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2017] [Revised: 12/12/2017] [Accepted: 12/19/2017] [Indexed: 02/07/2023] Open
Abstract
One of the most challenging goals in pharmacological research is overcoming the Blood Brain Barrier (BBB) to deliver drugs to the Central Nervous System (CNS). The use of physical means, such as steady and alternating magnetic fields to drive nanocarriers with proper magnetic characteristics may prove to be a useful strategy. The present review aims at providing an up-to-date picture of the applications of magnetic-driven nanotheranostics agents to the CNS. Although well consolidated on physical ground, some of the techniques described herein are still under investigation on in vitro or in silico models, while others have already entered in—or are close to—clinical validation. The review provides a concise overview of the physical principles underlying the behavior of magnetic nanoparticles (MNPs) interacting with an external magnetic field. Thereafter we describe the physiological pathways by which a substance can reach the brain from the bloodstream and then we focus on those MNP applications that aim at a nondestructive crossing of the BBB such as static magnetic fields to facilitate the passage of drugs and alternating magnetic fields to increment BBB permeability by magnetic heating. In conclusion, we briefly cite the most notable biomedical applications of MNPs and some relevant remarks about their safety and potential toxicity.
Collapse
|
25
|
Villate-Beitia I, Puras G, Soto-Sánchez C, Agirre M, Ojeda E, Zarate J, Fernández E, Pedraz JL. Non-viral vectors based on magnetoplexes, lipoplexes and polyplexes for VEGF gene delivery into central nervous system cells. Int J Pharm 2017; 521:130-140. [DOI: 10.1016/j.ijpharm.2017.02.016] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2016] [Revised: 02/02/2017] [Accepted: 02/05/2017] [Indexed: 12/19/2022]
|
26
|
Megías R, Arco M, Ciriza J, Saenz del Burgo L, Puras G, López-Viota M, Delgado ÁV, Dobson JP, Arias JL, Pedraz JL. Design and characterization of a magnetite/PEI multifunctional nanohybrid as non-viral vector and cell isolation system. Int J Pharm 2017; 518:270-280. [DOI: 10.1016/j.ijpharm.2016.12.042] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2016] [Revised: 12/14/2016] [Accepted: 12/17/2016] [Indexed: 12/20/2022]
|
27
|
Huang F, Zhao F, Liang LP, Zhou M, Qu ZL, Cao YZ, Lin C. Optomizing Transfection Efficiency of Cervical Cancer Cells Transfected by Cationic Liposomes LipofectamineTM2000. Asian Pac J Cancer Prev 2016; 16:7749-54. [PMID: 26625792 DOI: 10.7314/apjcp.2015.16.17.7749] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Currently, cationic liposome has become the commonly used vehicles for gene transfection. Furthermore, one of the most significant steps in microRNAs expression studies is transferring microRNAs into cell cultures successfully. In this study we aim to approach the feasibility of transfection of cervical cancer cell lines mediated by liposome and to obtain the optimized transfection condition for cervical cancer cell lines. MATERIALS AND METHODS Lipofectamine(TM)2000 as the carrier, miR-101 mimic was transfected into Hela cells and Siha cells. Using green fluorescent protein as reporter gene, to set different groups according to cell seeding density, the amount of miRNA , miRNA and the proportion of Liposomes, Whether to add serum into medium to study their impact on the liposomal transfection efficiency. Finally, MTT assay was used to analyze the relative minimal cell toxicity of liposome reagents. RESULTS The seeding density of Hela cell line and Siha are 1.5 x 10(4) (per well of 24 well plates), miRNA amount is 1ul of both, the ratio of miRNA and liposome is 1:0.5 of Hela cell line; 1:0.7 of Siha cell line respectively, after 24 hours we can get the highest transfection efficiency. Compared with serum medium, only Siha cells cultured with serum-free medium obtained higher transfection efficiency before transfection (P<0.01).MTT assay showed that according to the above conditions which has the lowest cytotoxicity. CONCLUSIONS The method of Liposome to transfected is a suitable way and it can be an efficient reagent for miRNA delivery for Hela cells and Siha cells in vitro. It may serve as a reference for the further research or application.
Collapse
Affiliation(s)
- Fei Huang
- Department of Pathology, The Affiliated Tumor Hospital Of Xinjiang Medical University, Urumqi, China E-mail :
| | | | | | | | | | | | | |
Collapse
|
28
|
Pereyra AS, Mykhaylyk O, Lockhart EF, Taylor JR, Delbono O, Goya RG, Plank C, Hereñu CB. Magnetofection Enhances Adenoviral Vector-based Gene Delivery in Skeletal Muscle Cells. ACTA ACUST UNITED AC 2016; 7. [PMID: 27274908 PMCID: PMC4888903 DOI: 10.4172/2157-7439.1000364] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The goal of magnetic field-assisted gene transfer is to enhance internalization of exogenous nucleic acids by association with magnetic nanoparticles (MNPs). This technique named magnetofection is particularly useful in difficult-to-transfect cells. It is well known that human, mouse, and rat skeletal muscle cells suffer a maturation-dependent loss of susceptibility to Recombinant Adenoviral vector (RAd) uptake. In postnatal, fully differentiated myofibers, the expression of the primary Coxsackie and Adenoviral membrane receptor (CAR) is severely downregulated representing a main hurdle for the use of these vectors in gene transfer/therapy. Here we demonstrate that assembling of Recombinant Adenoviral vectors with suitable iron oxide MNPs into magneto-adenovectors (RAd-MNP) and further exposure to a gradient magnetic field enables to efficiently overcome transduction resistance in skeletal muscle cells. Expression of Green Fluorescent Protein and Insulin-like Growth Factor 1 was significantly enhanced after magnetofection with RAd-MNPs complexes in C2C12 myotubes in vitro and mouse skeletal muscle in vivo when compared to transduction with naked virus. These results provide evidence that magnetofection, mainly due to its membrane-receptor independent mechanism, constitutes a simple and effective alternative to current methods for gene transfer into traditionally hard-to-transfect biological models.
Collapse
Affiliation(s)
- Andrea Soledad Pereyra
- Biochemistry Research Institute of La Plata (INIBIOLP)/National Scientific and Technical Research Council (CONICET), School of Medicine, National University of La Plata, La Plata, BA, Argentina (ZC 1900)
| | - Olga Mykhaylyk
- Ismaninger Street 22, Institute of Immunology and Experimental Klinikum rechts der Isar, Technical University of Munich, Munich, Germany (ZC 81675)
| | - Eugenia Falomir Lockhart
- Biochemistry Research Institute of La Plata (INIBIOLP)/National Scientific and Technical Research Council (CONICET), School of Medicine, National University of La Plata, La Plata, BA, Argentina (ZC 1900)
| | - Jackson Richard Taylor
- Department of Internal Medicine, Section on Gerontology and Geriatric Medicine, Wake Forest School of Medicine, Winston-Salem, NC, USA (ZC 27157)
| | - Osvaldo Delbono
- Department of Internal Medicine, Section on Gerontology and Geriatric Medicine, Wake Forest School of Medicine, Winston-Salem, NC, USA (ZC 27157)
| | - Rodolfo Gustavo Goya
- Biochemistry Research Institute of La Plata (INIBIOLP)/National Scientific and Technical Research Council (CONICET), School of Medicine, National University of La Plata, La Plata, BA, Argentina (ZC 1900)
| | - Christian Plank
- Ismaninger Street 22, Institute of Immunology and Experimental Klinikum rechts der Isar, Technical University of Munich, Munich, Germany (ZC 81675)
| | - Claudia Beatriz Hereñu
- Biochemistry Research Institute of La Plata (INIBIOLP)/National Scientific and Technical Research Council (CONICET), School of Medicine, National University of La Plata, La Plata, BA, Argentina (ZC 1900); IFEC-CONICET, Farmacology Department, School of Chemistry, National University of Cordoba, (ZC 5000) Córdoba, Argentina
| |
Collapse
|
29
|
Ojeda E, Puras G, Agirre M, Zarate J, Grijalvo S, Eritja R, Martinez-Navarrete G, Soto-Sánchez C, Diaz-Tahoces A, Aviles-Trigueros M, Fernández E, Pedraz JL. The influence of the polar head-group of synthetic cationic lipids on the transfection efficiency mediated by niosomes in rat retina and brain. Biomaterials 2015; 77:267-79. [PMID: 26610076 DOI: 10.1016/j.biomaterials.2015.11.017] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2015] [Revised: 11/02/2015] [Accepted: 11/06/2015] [Indexed: 01/28/2023]
Abstract
The development of novel non-viral delivery vehicles is essential in the search of more efficient strategies for retina and brain diseases. Herein, optimized niosome formulations prepared by oil-in water (o/w) and film-hydration techniques were characterized in terms of size, PDI, zeta potential, morphology and stability. Three ionizable glycerol-based cationic lipids containing a primary amine group (lipid 1), a triglycine group (lipid 2) and a dimethylamino ethyl pendent group (lipid 3) as polar head-groups were part of such niosomes. Upon the addition of pCMS-EGFP plasmid, nioplexes were obtained at different cationic lipid/DNA ratios (w/w). The resultant nioplexes were further physicochemically characterized and evaluated to condense, release and protect the DNA against enzymatic digestion. In vitro experiments were performed to evaluate transfection efficiency and cell viability in HEK-293, ARPE-19 and PECC cells. Interestingly, niosome formulations based on lipid 3 showed better transfection efficiencies in ARPE-19 and PECC cells than the rest of cationic lipids showed in this study. In vivo experiments in rat retina after intravitreal and subretinal injections together with in rat brain after cerebral cortex administration showed promising transfection efficiencies when niosome formulations based on lipid 3 were used. These results provide new insights for the development of non-viral vectors based on cationic lipids and their applications for efficient delivery of genetic material to the retina and brain.
Collapse
Affiliation(s)
- E Ojeda
- NanoBioCel Group, University of Basque Country (UPV/EHU), Vitoria-Gasteiz, Spain; Biomedical Research Networking Center in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Vitoria-Gasteiz, Spain
| | - G Puras
- NanoBioCel Group, University of Basque Country (UPV/EHU), Vitoria-Gasteiz, Spain; Biomedical Research Networking Center in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Vitoria-Gasteiz, Spain
| | - M Agirre
- NanoBioCel Group, University of Basque Country (UPV/EHU), Vitoria-Gasteiz, Spain; Biomedical Research Networking Center in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Vitoria-Gasteiz, Spain
| | - J Zarate
- NanoBioCel Group, University of Basque Country (UPV/EHU), Vitoria-Gasteiz, Spain; Biomedical Research Networking Center in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Vitoria-Gasteiz, Spain
| | - S Grijalvo
- Biomedical Research Networking Center in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Vitoria-Gasteiz, Spain; Institute of Advanced Chemistry of Catalonia, IQAC-CSIC, Barcelona, Spain
| | - R Eritja
- Biomedical Research Networking Center in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Vitoria-Gasteiz, Spain; Institute of Advanced Chemistry of Catalonia, IQAC-CSIC, Barcelona, Spain
| | - G Martinez-Navarrete
- Biomedical Research Networking Center in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Vitoria-Gasteiz, Spain; Neuroprothesis and Neuroengineering Research Group, Miguel Hernández University, Spain
| | - C Soto-Sánchez
- Biomedical Research Networking Center in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Vitoria-Gasteiz, Spain; Neuroprothesis and Neuroengineering Research Group, Miguel Hernández University, Spain
| | - A Diaz-Tahoces
- Biomedical Research Networking Center in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Vitoria-Gasteiz, Spain; Neuroprothesis and Neuroengineering Research Group, Miguel Hernández University, Spain
| | - M Aviles-Trigueros
- Laboratory of Experimental Ophthalmology, Faculty of Medicine, University of Murcia, Regional Campus of International Excellence "Campus Mare Nostrum", Murcia, Spain
| | - E Fernández
- Biomedical Research Networking Center in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Vitoria-Gasteiz, Spain; Neuroprothesis and Neuroengineering Research Group, Miguel Hernández University, Spain
| | - J L Pedraz
- NanoBioCel Group, University of Basque Country (UPV/EHU), Vitoria-Gasteiz, Spain; Biomedical Research Networking Center in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Vitoria-Gasteiz, Spain.
| |
Collapse
|