1
|
Fernandes F, Talukdar I, Kowshik M. Cysteamine functionalized gold nanoparticles exhibit high efficiency delivery of genetic materials in embryonic stem cells majorly via clathrin mediated endocytosis. Int J Pharm 2024; 667:124928. [PMID: 39521158 DOI: 10.1016/j.ijpharm.2024.124928] [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: 05/30/2024] [Revised: 10/31/2024] [Accepted: 11/06/2024] [Indexed: 11/16/2024]
Abstract
Efficient and safe gene delivery is vital for genetic manipulation of stem cells for regenerative medicine. Gold nanoparticles have been used for various biomedical applications in the past, and are currently being researched as transfection agents. In this study, we report a simple one-pot synthesis of positively charged gold nanoparticles functionalized with cysteamine. The nanoparticles exhibit no cytotoxicity and can bind to both plasmid DNA (pDNA) as well as small interference RNA (siRNA). We observed that a five fold lower concentration of pDNA was sufficient for achieving comparable overexpression as that of a commercial transfection agent. We also observed that about 70 % transient silencing of the target gene was achieved with only 25 nM siRNA delivered by our nano-vehicle. To better understand the fate of the nanoparticle, we attempted to identify its uptake mechanism. The results indicate that while all the mechanisms contribute to the uptake, the clathrin-dependent pathway plays a major role. This is the first study on understanding the mechanism of uptake of CA-AuNPs conjugated to pDNA by embryonic stem cells. This is also the first study, where a successful transfection using gold based nanoparticles has been achieved in ESCs at a concentration as low as 0.5 µg/ml for pDNA and 25ƞM siRNA.
Collapse
Affiliation(s)
- Fiona Fernandes
- Dept. of Biological Sciences, BITS Pilani, K K Birla Goa Campus, Goa, India
| | - Indrani Talukdar
- Dept. of Biological Sciences, BITS Pilani, K K Birla Goa Campus, Goa, India.
| | - Meenal Kowshik
- Dept. of Biological Sciences, BITS Pilani, K K Birla Goa Campus, Goa, India.
| |
Collapse
|
2
|
Léger L, De Clercq C, Aalders J, Van Acker-Verberckt K, Braeckmans K, van Hengel J. Photoporation-mediated spatial intracellular delivery of stem cell-derived cardiomyocytes. MethodsX 2024; 12:102548. [PMID: 38292311 PMCID: PMC10825475 DOI: 10.1016/j.mex.2024.102548] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Accepted: 01/03/2024] [Indexed: 02/01/2024] Open
Abstract
Human induced pluripotent stem cell-derived cardiomyocytes (iPSC-CMs) are promising candidates for disease modeling and therapeutic purposes, however, non-viral intracellular delivery in these cells remains challenging. Gold nanoparticle (AuNP)-sensitized photoporation creates transient pores in the cell membrane by vapor nanobubble formation, allowing diffusion of extracellular biomolecules. This non-viral technique was employed to test and optimize its distinct physical mode of action in iPSC-CMs. Photoporation optimization was aimed at achieving high delivery rates while minimizing cell death. Various AuNP concentrations, in conjunction with different laser fluences, were explored to facilitate the intracellular delivery of 10 kDa and 150 kDa FITC-labelled dextran as model macromolecules. Cardiomyocyte viability was assessed using the CellTiter-Glo® viability assay, while the delivery efficiency was quantified through flow cytometry. On 30 day-old cardiomyocytes, AuNP photoporation was able to yield ∼60 % delivery efficiency while maintaining a high cell viability (∼70 %). Overall, higher AuNP concentrations resulted in greater delivery efficiencies, albeit at the expense of lower cell viability. Finally, photoporation was capable of patterning a geometric shape, demonstrating its exceptional selective resolution in delivering molecules to spatially restricted regions of the cell culture. In conclusion, AuNP-photoporation exhibits considerable potential as an effective and gentle non-viral method for intracellular delivery in iPSC-CMs.•AuNP-photoporation is a non-viral intracellular delivery method suitable for iPSC-CMs with high efficiency and cell viability•This method is capable of spatially resolved intracellular delivery with excellent resolution.
Collapse
Affiliation(s)
- Laurens Léger
- Medical Cell Biology Group, Department of Human Structure and Repair, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
| | - Chloë De Clercq
- Laboratory of General Biochemistry and Physical Pharmacy, Faculty of Pharmaceutical Sciences, Ghent University, Ghent, Belgium
| | - Jeffrey Aalders
- Medical Cell Biology Group, Department of Human Structure and Repair, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
| | - Kiara Van Acker-Verberckt
- Medical Cell Biology Group, Department of Human Structure and Repair, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
| | - Kevin Braeckmans
- Laboratory of General Biochemistry and Physical Pharmacy, Faculty of Pharmaceutical Sciences, Ghent University, Ghent, Belgium
| | - Jolanda van Hengel
- Medical Cell Biology Group, Department of Human Structure and Repair, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
| |
Collapse
|
3
|
Murru C, Duvert L, Magdinier F, Casanova A, Alloncle AP, Testa S, Al-Kattan A. Assessment of laser-synthesized Si nanoparticle effects on myoblast motility, proliferation and differentiation: towards potential tissue engineering applications. NANOSCALE ADVANCES 2024; 6:2104-2112. [PMID: 38633050 PMCID: PMC11019504 DOI: 10.1039/d3na01020a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Accepted: 02/23/2024] [Indexed: 04/19/2024]
Abstract
Due to their biocompatibility and biodegradability and their unique structural and physicochemical properties, laser-synthesized silicon nanoparticles (Si-NPs) are one of the nanomaterials which have been most studied as potential theragnostic tools for non-invasive therapeutic modalities. However, their ability to modulate cell behavior and to promote proliferation and differentiation is still very little investigated or unknown. In this work, ultrapure ligand free Si-NPs of 50 ± 11.5 nm were prepared by femtosecond (fs) laser ablation in liquid. After showing the ability of Si-NPs to be internalized by murine C2C12 myoblasts, the cytotoxicity of the Si-NPs on these cells was evaluated at concentrations ranging from 14 to 224 μg mL-1. Based on these findings, three concentrations of 14, 28 and 56 μg mL-1 were thus considered to study the effect on myoblast differentiation, proliferation and motility at the molecular and phenotypical levels. It was demonstrated that up to 28 μg mL-1, the Si-NPs are able to promote the proliferation of myoblasts and their subsequent differentiation. Scratch tests were also performed revealing the positive Si-NP effect on cellular motility at 14 and 28 μg mL-1. Finally, gene expression analysis confirmed the ability of Si-NPs to promote proliferation, differentiation and motility of myoblasts even at very low concentration. This work opens up novel exciting prospects for Si-NPs made by the laser process as innovative tools for skeletal muscle tissue engineering in view of developing novel therapeutic protocols for regenerative medicine.
Collapse
Affiliation(s)
- Clarissa Murru
- Aix-Marseille University, CNRS, LP3 UMR 7341 Campus de Luminy C13288 Marseille France
| | - Lucas Duvert
- Aix-Marseille University, CNRS, LP3 UMR 7341 Campus de Luminy C13288 Marseille France
- Aix-Marseille University, INSERM, MMG, Marseille Medical Genetics 13385 Marseille France
| | - Frederique Magdinier
- Aix-Marseille University, INSERM, MMG, Marseille Medical Genetics 13385 Marseille France
| | - Adrien Casanova
- Aix-Marseille University, CNRS, LP3 UMR 7341 Campus de Luminy C13288 Marseille France
| | | | - Stefano Testa
- Aix-Marseille University, INSERM, MMG, Marseille Medical Genetics 13385 Marseille France
| | - Ahmed Al-Kattan
- Aix-Marseille University, CNRS, LP3 UMR 7341 Campus de Luminy C13288 Marseille France
| |
Collapse
|
4
|
Chen G, Obal D. Detecting and measuring of GPCR signaling - comparison of human induced pluripotent stem cells and immortal cell lines. Front Endocrinol (Lausanne) 2023; 14:1179600. [PMID: 37293485 PMCID: PMC10244570 DOI: 10.3389/fendo.2023.1179600] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2023] [Accepted: 04/12/2023] [Indexed: 06/10/2023] Open
Abstract
G protein-coupled receptors (GPCRs) are a large family of transmembrane proteins that play a major role in many physiological processes, and thus GPCR-targeted drug development has been widely promoted. Although research findings generated in immortal cell lines have contributed to the advancement of the GPCR field, the homogenous genetic backgrounds, and the overexpression of GPCRs in these cell lines make it difficult to correlate the results with clinical patients. Human induced pluripotent stem cells (hiPSCs) have the potential to overcome these limitations, because they contain patient specific genetic information and can differentiate into numerous cell types. To detect GPCRs in hiPSCs, highly selective labeling and sensitive imaging techniques are required. This review summarizes existing resonance energy transfer and protein complementation assay technologies, as well as existing and new labeling methods. The difficulties of extending existing detection methods to hiPSCs are discussed, as well as the potential of hiPSCs to expand GPCR research towards personalized medicine.
Collapse
Affiliation(s)
- Gaoxian Chen
- Department of Anesthesiology, Perioperative, and Pain Medicine, Stanford University, Stanford, CA, United States
- Stanford Cardiovascular Institute, Stanford University, Stanford, CA, United States
| | - Detlef Obal
- Department of Anesthesiology, Perioperative, and Pain Medicine, Stanford University, Stanford, CA, United States
- Stanford Cardiovascular Institute, Stanford University, Stanford, CA, United States
| |
Collapse
|
5
|
Karami Z, Moradi S, Eidi A, Soleimani M, Jafarian A. Induced pluripotent stem cells: Generation methods and a new perspective in COVID-19 research. Front Cell Dev Biol 2023; 10:1050856. [PMID: 36733338 PMCID: PMC9887183 DOI: 10.3389/fcell.2022.1050856] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Accepted: 11/22/2022] [Indexed: 01/18/2023] Open
Abstract
Induced pluripotent stem cells (iPSCs) exhibit an unlimited ability to self-renew and produce various differentiated cell types, thereby creating high hopes for both scientists and patients as a great tool for basic research as well as for regenerative medicine purposes. The availability and safety of iPSCs for therapeutic purposes require safe and highly efficient methods for production of these cells. Different methods have been used to produce iPSCs, each of which has advantages and disadvantages. Studying these methods would be very helpful in developing an easy, safe, and efficient method for the generation of iPSCs. Since iPSCs can be generated from somatic cells, they can be considered as valuable cellular resources available for important research needs and various therapeutic purposes. Coronavirus disease 2019 (COVID-19) is a disease that has endangered numerous human lives worldwide and currently has no definitive cure. Therefore, researchers have been rigorously studying and examining all aspects of COVID-19 and potential treatment modalities and various drugs in order to enable the treatment, control, and prevention of COVID-19. iPSCs have become one of the most attractive and promising tools in this field by providing the ability to study COVID-19 and the effectiveness of drugs on this disease outside the human body. In this study, we discuss the different methods of generation of iPSCs as well as their respective advantages and disadvantages. We also present recent applications of iPSCs in the study and treatment of COVID-19.
Collapse
Affiliation(s)
- Zahra Karami
- 1Department of Biology, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Sharif Moradi
- 2Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Akram Eidi
- 1Department of Biology, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Masoud Soleimani
- 3Hematology and Cell Therapy Department, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran,4Department of Tissue Engineering and Applied Cell Science, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Arefeh Jafarian
- 5Iranian Tissue Bank and Research Center, Tehran University of Medical Sciences, Tehran, Iran,*Correspondence: Arefeh Jafarian,
| |
Collapse
|
6
|
Virus-Like Particles as Nanocarriers for Intracellular Delivery of Biomolecules and Compounds. Viruses 2022; 14:v14091905. [PMID: 36146711 PMCID: PMC9503347 DOI: 10.3390/v14091905] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Revised: 08/24/2022] [Accepted: 08/25/2022] [Indexed: 11/16/2022] Open
Abstract
Virus-like particles (VLPs) are nanostructures assemble from viral proteins. Besides widely used for vaccine development, VLPs have also been explored as nanocarriers for cargo delivery as they combine the key advantages of viral and non-viral vectors. While it protects cargo molecules from degradation, the VLP has good cell penetrating property to mediate cargo passing the cell membrane and released into cells, making the VLP an ideal tool for intracellular delivery of biomolecules and drugs. Great progresses have been achieved and multiple challenges are still on the way for broad applications of VLP as delivery vectors. Here we summarize current advances and applications in VLP as a delivery vector. Progresses on delivery of different types of biomolecules as well as drugs by VLPs are introduced, and the strategies for cargo packaging are highlighted which is one of the key steps for VLP mediated intracellular delivery. Production and applications of VLPs are also briefly reviewed, with a discussion on future challenges in this rapidly developing field.
Collapse
|
7
|
Rong L, Chen D, Huang X, Sun L. Delivery of Cas9-guided ABE8e into stem cells using poly(l-lysine) polypeptides for correction of the hemophilia-associated FIX missense mutation. Biochem Biophys Res Commun 2022; 628:49-56. [DOI: 10.1016/j.bbrc.2022.08.076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2022] [Accepted: 08/22/2022] [Indexed: 11/15/2022]
|
8
|
Cheng J, Zheng Z, Tang W, Shao J, Jiang H, Lin H. A new strategy for stem cells therapy for erectile dysfunction: Adipose-derived stem cells transfect Neuregulin-1 gene through superparamagnetic iron oxide nanoparticles. Investig Clin Urol 2022; 63:359-367. [PMID: 35534221 PMCID: PMC9091825 DOI: 10.4111/icu.20220016] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Revised: 02/05/2022] [Accepted: 02/09/2022] [Indexed: 11/18/2022] Open
Abstract
PURPOSE Our previous studies showed that nanotechnology improves derived adipose-derived stem cells (ADSCs) therapy for erectile dysfunction (ED). In this study, the Neuregulin-1(NRG1) gene was transfected into ADSCs with superparamagnetic iron oxide nanoparticles (SPION) further to improve the therapeutic effect of ADSCs on ED. MATERIALS AND METHODS ADSCs were isolated from epididymal adipose tissue of Sprague-Dawley rats. The optimal concentration of PEI-SPION (SPION modified with polyethyleneimine) was selected to construct the gene complex. After electrostatic binding of PEI-SPION and DNA, a PEI layer was wrapped to make the PEI-SPION-NRG1-PEI gene transfection complex. Different groups were set up for transfection tests. Lipo2000 transfection reagent was used as the control. PEI-SPION-NRG1-PEI in the experimental group was transfected under an external magnetic field. RESULTS When the concentration of PEI-SPION was 10 µg/mL, it had little cytotoxicity, and cell activity was not significantly affected. PEI-SPION-NRG1-PEI forms positively charged nanocomposites with a particle size of 72.6±14.9 nm when N/P ≥8. The PEI-SPION-NRG1-PEI gene complex can significantly improve the transfection efficiency of ADSCs, reaching 26.74%±4.62%, under the action of the external magnetic field. PCR and Western blot showed that the expression level of the NRG1 gene increased significantly, which proved that the transfection was effective. CONCLUSIONS PEI-SPION can be used as a vector for NRG1 gene transfection into ADSCs. PEI-SPION-NRG1-PEI packaging has the highest transfection efficiency under the external magnetic field than the other groups. These findings may provide a new strategy for ADSCs therapy for ED.
Collapse
Affiliation(s)
- Jianxing Cheng
- Department of Urology, Peking University Third Hospital, Peking University, Beijing, China
- Reproductive Medicine Center, Peking University Third Hospital, Peking University, Beijing, China
- Department of Andrology, Peking University Third Hospital, Peking University, Beijing, China
| | - Zhongjie Zheng
- Department of Urology, Peking University Third Hospital, Peking University, Beijing, China
- Reproductive Medicine Center, Peking University Third Hospital, Peking University, Beijing, China
- Department of Andrology, Peking University Third Hospital, Peking University, Beijing, China
| | - Wenhao Tang
- Department of Urology, Peking University Third Hospital, Peking University, Beijing, China
- Reproductive Medicine Center, Peking University Third Hospital, Peking University, Beijing, China
- Department of Andrology, Peking University Third Hospital, Peking University, Beijing, China
| | - Jichun Shao
- Department of Urology, Second Affiliated Hospital of Chengdu Medical College (China National Nuclear Corporation 416 Hospital), Chengdu, Sichuan, China
| | - Hui Jiang
- Department of Urology, Peking University Third Hospital, Peking University, Beijing, China
- Reproductive Medicine Center, Peking University Third Hospital, Peking University, Beijing, China
- Department of Andrology, Peking University Third Hospital, Peking University, Beijing, China
- Department of Human Sperm Bank, Peking University Third Hospital, Peking University, Beijing, China.
| | - Haocheng Lin
- Department of Urology, Peking University Third Hospital, Peking University, Beijing, China
- Reproductive Medicine Center, Peking University Third Hospital, Peking University, Beijing, China
- Department of Andrology, Peking University Third Hospital, Peking University, Beijing, China.
| |
Collapse
|
9
|
Alzate-Correa D, Lawrence WR, Salazar-Puerta A, Higuita-Castro N, Gallego-Perez D. Nanotechnology-Driven Cell-Based Therapies in Regenerative Medicine. AAPS J 2022; 24:43. [PMID: 35292878 PMCID: PMC9074705 DOI: 10.1208/s12248-022-00692-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Accepted: 02/10/2022] [Indexed: 12/23/2022] Open
Abstract
The administration of cells as therapeutic agents has emerged as a novel approach to complement the use of small molecule drugs and other biologics for the treatment of numerous conditions. Although the use of cells for structural and/or functional tissue repair and regeneration provides new avenues to address increasingly complex disease processes, it also faces numerous challenges related to efficacy, safety, and translational potential. Recent advances in nanotechnology-driven cell therapies have the potential to overcome many of these issues through precise modulation of cellular behavior. Here, we describe several approaches that illustrate the use of different nanotechnologies for the optimization of cell therapies and discuss some of the obstacles that need to be overcome to allow for the widespread implementation of nanotechnology-based cell therapies in regenerative medicine.
Collapse
Affiliation(s)
- D Alzate-Correa
- Department of Biomedical Engineering, The Ohio State University, Columbus, Ohio, 43210, USA
| | - W R Lawrence
- Department of Biomedical Engineering, The Ohio State University, Columbus, Ohio, 43210, USA.,Biomedical Sciences Graduate Program, The Ohio State University, Columbus, Ohio, 43210, USA
| | - A Salazar-Puerta
- Department of Biomedical Engineering, The Ohio State University, Columbus, Ohio, 43210, USA
| | - N Higuita-Castro
- Department of Biomedical Engineering, The Ohio State University, Columbus, Ohio, 43210, USA.,Interdisciplinary Biophysics Graduate Program, The Ohio State University, Columbus, Ohio, 43210, USA.,Department of Surgery, The Ohio State University, 140 W. 19th Ave, room 3018, Columbus, Ohio, 43210, USA
| | - D Gallego-Perez
- Department of Biomedical Engineering, The Ohio State University, Columbus, Ohio, 43210, USA. .,Department of Surgery, The Ohio State University, 140 W. 19th Ave, room 3018, Columbus, Ohio, 43210, USA.
| |
Collapse
|
10
|
Friedrich RP, Cicha I, Alexiou C. Iron Oxide Nanoparticles in Regenerative Medicine and Tissue Engineering. NANOMATERIALS 2021; 11:nano11092337. [PMID: 34578651 PMCID: PMC8466586 DOI: 10.3390/nano11092337] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Revised: 09/03/2021] [Accepted: 09/06/2021] [Indexed: 12/13/2022]
Abstract
In recent years, many promising nanotechnological approaches to biomedical research have been developed in order to increase implementation of regenerative medicine and tissue engineering in clinical practice. In the meantime, the use of nanomaterials for the regeneration of diseased or injured tissues is considered advantageous in most areas of medicine. In particular, for the treatment of cardiovascular, osteochondral and neurological defects, but also for the recovery of functions of other organs such as kidney, liver, pancreas, bladder, urethra and for wound healing, nanomaterials are increasingly being developed that serve as scaffolds, mimic the extracellular matrix and promote adhesion or differentiation of cells. This review focuses on the latest developments in regenerative medicine, in which iron oxide nanoparticles (IONPs) play a crucial role for tissue engineering and cell therapy. IONPs are not only enabling the use of non-invasive observation methods to monitor the therapy, but can also accelerate and enhance regeneration, either thanks to their inherent magnetic properties or by functionalization with bioactive or therapeutic compounds, such as drugs, enzymes and growth factors. In addition, the presence of magnetic fields can direct IONP-labeled cells specifically to the site of action or induce cell differentiation into a specific cell type through mechanotransduction.
Collapse
|
11
|
Ledford HA, Park S, Muir D, Woltz RL, Ren L, Nguyen PT, Sirish P, Wang W, Sihn CR, George AL, Knollmann BC, Yamoah EN, Yarov-Yarovoy V, Zhang XD, Chiamvimonvat N. Different arrhythmia-associated calmodulin mutations have distinct effects on cardiac SK channel regulation. J Gen Physiol 2021; 152:211546. [PMID: 33211795 PMCID: PMC7681919 DOI: 10.1085/jgp.202012667] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2020] [Revised: 08/25/2020] [Accepted: 10/19/2020] [Indexed: 12/21/2022] Open
Abstract
Calmodulin (CaM) plays a critical role in intracellular signaling and regulation of Ca2+-dependent proteins and ion channels. Mutations in CaM cause life-threatening cardiac arrhythmias. Among the known CaM targets, small-conductance Ca2+-activated K+ (SK) channels are unique, since they are gated solely by beat-to-beat changes in intracellular Ca2+. However, the molecular mechanisms of how CaM mutations may affect the function of SK channels remain incompletely understood. To address the structural and functional effects of these mutations, we introduced prototypical human CaM mutations in human induced pluripotent stem cell–derived cardiomyocyte-like cells (hiPSC-CMs). Using structural modeling and molecular dynamics simulation, we demonstrate that human calmodulinopathy-associated CaM mutations disrupt cardiac SK channel function via distinct mechanisms. CaMD96V and CaMD130G mutants reduce SK currents through a dominant-negative fashion. By contrast, specific mutations replacing phenylalanine with leucine result in conformational changes that affect helix packing in the C-lobe, which disengage the interactions between apo-CaM and the CaM-binding domain of SK channels. Distinct mutant CaMs may result in a significant reduction in the activation of the SK channels, leading to a decrease in the key Ca2+-dependent repolarization currents these channels mediate. The findings in this study may be generalizable to other interactions of mutant CaMs with Ca2+-dependent proteins within cardiac myocytes.
Collapse
Affiliation(s)
- Hannah A Ledford
- Division of Cardiovascular Medicine, Department of Internal Medicine, School of Medicine, University of California, Davis, Davis, CA
| | - Seojin Park
- Department of Physiology and Cell Biology, University of Nevada, Reno, Reno, NV
| | - Duncan Muir
- Division of Cardiovascular Medicine, Department of Internal Medicine, School of Medicine, University of California, Davis, Davis, CA
| | - Ryan L Woltz
- Division of Cardiovascular Medicine, Department of Internal Medicine, School of Medicine, University of California, Davis, Davis, CA
| | - Lu Ren
- Division of Cardiovascular Medicine, Department of Internal Medicine, School of Medicine, University of California, Davis, Davis, CA
| | - Phuong T Nguyen
- Department of Physiology and Membrane Biology, School of Medicine, University of California, Davis, Davis, CA
| | - Padmini Sirish
- Division of Cardiovascular Medicine, Department of Internal Medicine, School of Medicine, University of California, Davis, Davis, CA
| | - Wenying Wang
- Department of Physiology and Cell Biology, University of Nevada, Reno, Reno, NV
| | - Choong-Ryoul Sihn
- Department of Physiology and Cell Biology, University of Nevada, Reno, Reno, NV
| | - Alfred L George
- Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL
| | - Björn C Knollmann
- Vanderbilt Center for Arrhythmia Research and Therapeutics, Department of Medicine, School of Medicine, Vanderbilt University, Nashville, TN
| | - Ebenezer N Yamoah
- Department of Physiology and Cell Biology, University of Nevada, Reno, Reno, NV
| | - Vladimir Yarov-Yarovoy
- Department of Physiology and Membrane Biology, School of Medicine, University of California, Davis, Davis, CA
| | - Xiao-Dong Zhang
- Division of Cardiovascular Medicine, Department of Internal Medicine, School of Medicine, University of California, Davis, Davis, CA.,Department of Veterans Affairs, Northern California Health Care System, Mather, CA
| | - Nipavan Chiamvimonvat
- Division of Cardiovascular Medicine, Department of Internal Medicine, School of Medicine, University of California, Davis, Davis, CA.,Department of Veterans Affairs, Northern California Health Care System, Mather, CA
| |
Collapse
|
12
|
Yamoah MA, Thai PN, Zhang XD. Transgene Delivery to Human Induced Pluripotent Stem Cells Using Nanoparticles. Pharmaceuticals (Basel) 2021; 14:334. [PMID: 33917388 PMCID: PMC8067386 DOI: 10.3390/ph14040334] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2021] [Revised: 03/25/2021] [Accepted: 04/02/2021] [Indexed: 11/25/2022] Open
Abstract
Human induced pluripotent stem cells (hiPSCs) and hiPSCs-derived cells have the potential to revolutionize regenerative and precision medicine. Genetically reprograming somatic cells to generate hiPSCs and genetic modification of hiPSCs are considered the key procedures for the study and application of hiPSCs. However, there are significant technical challenges for transgene delivery into somatic cells and hiPSCs since these cells are known to be difficult to transfect. The existing methods, such as viral transduction and chemical transfection, may introduce significant alternations to hiPSC culture which affect the potency, purity, consistency, safety, and functional capacity of hiPSCs. Therefore, generation and genetic modification of hiPSCs through non-viral approaches are necessary and desirable. Nanotechnology has revolutionized fields from astrophysics to biology over the past two decades. Increasingly, nanoparticles have been used in biomedicine as powerful tools for transgene and drug delivery, imaging, diagnostics, and therapeutics. The most successful example is the recent development of SARS-CoV-2 vaccines at warp speed to combat the 2019 coronavirus disease (COVID-19), which brought nanoparticles to the center stage of biomedicine and demonstrated the efficient nanoparticle-mediated transgene delivery into human body. Nanoparticles have the potential to facilitate the transgene delivery into the hiPSCs and offer a simple and robust approach. Nanoparticle-mediated transgene delivery has significant advantages over other methods, such as high efficiency, low cytotoxicity, biodegradability, low cost, directional and distal controllability, efficient in vivo applications, and lack of immune responses. Our recent study using magnetic nanoparticles for transfection of hiPSCs provided an example of the successful applications, supporting the potential roles of nanoparticles in hiPSC biology. This review discusses the principle, applications, and significance of nanoparticles in the transgene delivery to hiPSCs and their successful application in the development of COVID-19 vaccines.
Collapse
Affiliation(s)
- Megan A. Yamoah
- Department of Economics, University of Oxford, Oxford OX1 3UQ, UK;
| | - Phung N. Thai
- Department of Internal Medicine, School of Medicine, University of California, Davis, CA 95616, USA;
| | - Xiao-Dong Zhang
- Department of Internal Medicine, School of Medicine, University of California, Davis, CA 95616, USA;
- Department of Veterans Affairs, Northern California Health Care System, Mather, CA 95655, USA
| |
Collapse
|
13
|
Tang Y, Wu J, Zhang Y, Ju L, Qu X, Jiang D. Magnetic transfection with superparamagnetic chitosan-loaded IGFBP 5 nanoparticles and their in vitro biosafety. ROYAL SOCIETY OPEN SCIENCE 2021; 8:201331. [PMID: 33614075 PMCID: PMC7890493 DOI: 10.1098/rsos.201331] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/15/2020] [Accepted: 11/24/2020] [Indexed: 06/12/2023]
Abstract
We prepared the superparamagnetic chitosan nanoparticles (SPCIONPs) to study the application of them as gene vectors using a magnetic transfection system for the targeted treatment of lung metastasis of osteosarcoma. The SPCIONPs were characterized by transmission electron microscopy, Fourier transform infrared spectrometry, superconducting quantum interference device and atomic force microscopy. Their biosafety was determined by cell counting kit-8 (CCK8) and live-dead staining assays. The transfection in vitro was detected by laser confocal microscopy. SPCIONPs, which can bind closely to plasmids and protect them from DNA enzyme degradation, were prepared with an average particle size of approximately 22 nm and zeta potential of 11.3 mV. The results of the CCK8 and live-dead staining assays showed that superparamagnetic chitosan nanoparticles loaded with insulin-like growth factor-binding protein 5 (SPCIONPs/pIGFBP5) induced no significant cytotoxicity compared to the control group. The result of transfection in vitro suggested that pIGFBP5 emitted a greater amount of red fluorescence in the SPCIONPs/pIGFBP5 group than that in the chitosan-loaded IGFBP5 (CS/pIGFBP5) group. In conclusion, the prepared SPCIONPs had good biosafety and could be effectively used to transfer pIGFBP5 into 143B cells, and they thus have good application prospects for the treatment of lung metastasis of osteosarcoma.
Collapse
Affiliation(s)
- Yue Tang
- Department of Traumatic Joint Center, The Third Affiliated Hospital of Chongqing Medical University (General Hospital), No 1 Shuanghu Road, Yubei District, Chongqing 401120, People's Republic of China
- Department of Orthopedics, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, Laboratory of Biomaterials, 136# Zhongshan 2 road, Yuzhong District, Chongqing 400014, People's Republic of China
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, No 1 Medicine Road, Yuzhong District, Chongqing 400016, People's Republic of China
| | - Jun Wu
- Department of Orthopedics, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, Laboratory of Biomaterials, 136# Zhongshan 2 road, Yuzhong District, Chongqing 400014, People's Republic of China
| | - Yuan Zhang
- Department of Orthopedics, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, Laboratory of Biomaterials, 136# Zhongshan 2 road, Yuzhong District, Chongqing 400014, People's Republic of China
| | - Lingpeng Ju
- Department of Orthopedics, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, Laboratory of Biomaterials, 136# Zhongshan 2 road, Yuzhong District, Chongqing 400014, People's Republic of China
| | - Xiangyang Qu
- Department of Orthopedics, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, Laboratory of Biomaterials, 136# Zhongshan 2 road, Yuzhong District, Chongqing 400014, People's Republic of China
| | - Dianming Jiang
- Department of Traumatic Joint Center, The Third Affiliated Hospital of Chongqing Medical University (General Hospital), No 1 Shuanghu Road, Yubei District, Chongqing 401120, People's Republic of China
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, No 1 Medicine Road, Yuzhong District, Chongqing 400016, People's Republic of China
| |
Collapse
|
14
|
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
|
15
|
Zhang T, Xu Q, Huang T, Ling D, Gao J. New Insights into Biocompatible Iron Oxide Nanoparticles: A Potential Booster of Gene Delivery to Stem Cells. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2020; 16:e2001588. [PMID: 32725792 DOI: 10.1002/smll.202001588] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Revised: 05/10/2020] [Indexed: 06/11/2023]
Abstract
Gene delivery to stem cells is a critical issue of stem cells-based therapies, still facing ongoing challenges regarding efficiency and safety. Recent advances in the controlled synthesis of biocompatible magnetic iron oxide nanoparticles (IONPs) have provided a powerful nanotool for assisting gene delivery to stem cells. However, this field is still at an early stage, with well-designed and scalable IONPs synthesis highly desired. Furthermore, the potential risks or bioeffects of IONPs on stem cells are not completely figured out. Therefore, in this review, the updated researches focused on the gene delivery to stem cells using various designed IONPs are highlighted. Additionally, the impacts of the physicochemical properties of IONPs, as well as the magnetofection systems on the gene delivery performance and biocompatibility are summarized. Finally, challenges attributed to the potential impacts of IONPs on the biologic behaviors of stem cells and the large-scale productions of uniform IONPs are emphasized. The principles and challenges summarized in this review provide a general guidance for the rational design of IONPs-assisted gene delivery to stem cells.
Collapse
Affiliation(s)
- Tianyuan Zhang
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Regenerative Medicine, Zhejiang University, Hangzhou, 310058, China
| | - Qianhao Xu
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Ting Huang
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Daishun Ling
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Jianqing Gao
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Regenerative Medicine, Zhejiang University, Hangzhou, 310058, China
| |
Collapse
|
16
|
Bodbin SE, Denning C, Mosqueira D. Transfection of hPSC-Cardiomyocytes Using Viafect™ Transfection Reagent. Methods Protoc 2020; 3:E57. [PMID: 32784848 PMCID: PMC7564709 DOI: 10.3390/mps3030057] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Revised: 08/05/2020] [Accepted: 08/06/2020] [Indexed: 02/06/2023] Open
Abstract
Twenty years since their first derivation, human pluripotent stem cell-derived cardiomyocytes (hPSC-CMs) have shown promise in disease modelling research, while their potential for cardiac repair is being investigated. However, low transfection efficiency is a barrier to wider realisation of the potential this model system has to offer. We endeavoured to produce a protocol for improved transfection of hPSC-CMs using the ViafectTM reagent by Promega. Through optimisation of four essential parameters: (i) serum supplementation, (ii) time between replating and transfection, (iii) reagent to DNA ratio and (iv) cell density, we were able to successfully transfect hPSC-CMs to ~95% efficiencies. Transfected hPSC-CMs retained high purity and structural integrity despite a mild reduction in viability, and preserved compatibility with phenotyping assays of hypertrophy. This protocol greatly adds value to the field by overcoming limited transfection efficiencies of hPSC-CMs in a simple and quick approach that ensures sustained expression of transfected genes for at least 14 days, opening new opportunities in mechanistic discovery for cardiac-related diseases.
Collapse
Affiliation(s)
- Sara E. Bodbin
- Division of Cancer & Stem Cells, Biodiscovery Institute, University of Nottingham, Nottingham NG7 2RD, UK
| | - Chris Denning
- Division of Cancer & Stem Cells, Biodiscovery Institute, University of Nottingham, Nottingham NG7 2RD, UK
| | - Diogo Mosqueira
- Division of Cancer & Stem Cells, Biodiscovery Institute, University of Nottingham, Nottingham NG7 2RD, UK
| |
Collapse
|
17
|
Li S, Wei C, Lv Y. Preparation and Application of Magnetic Responsive Materials in Bone Tissue Engineering. Curr Stem Cell Res Ther 2020; 15:428-440. [PMID: 31893995 DOI: 10.2174/1574888x15666200101122505] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Revised: 11/01/2019] [Accepted: 12/06/2019] [Indexed: 11/22/2022]
Abstract
At present, many kinds of materials are used for bone tissue engineering, such as polymer materials, metals, etc., which in general have good biocompatibility and mechanical properties. However, these materials cannot be controlled artificially after implantation, which may result in poor repair performance. The appearance of the magnetic response material enables the scaffolds to have the corresponding ability to the external magnetic field. Within the magnetic field, the magnetic response material can achieve the targeted release of the drug, improve the performance of the scaffold, and further have a positive impact on bone formation. This paper first reviewed the preparation methods of magnetic responsive materials such as magnetic nanoparticles, magnetic polymers, magnetic bioceramic materials and magnetic alloys in recent years, and then introduced its main applications in the field of bone tissue engineering, including promoting osteogenic differentiation, targets release, bioimaging, cell patterning, etc. Finally, the mechanism of magnetic response materials to promote bone regeneration was introduced. The combination of magnetic field treatment methods will bring significant progress to regenerative medicine and help to improve the treatment of bone defects and promote bone tissue repair.
Collapse
Affiliation(s)
- Song Li
- Mechanobiology and Regenerative Medicine Laboratory, Bioengineering College, Chongqing University, Chongqing, China
| | - Changling Wei
- Mechanobiology and Regenerative Medicine Laboratory, Bioengineering College, Chongqing University, Chongqing, China
| | - Yonggang Lv
- Mechanobiology and Regenerative Medicine Laboratory, Bioengineering College, Chongqing University, Chongqing, China
| |
Collapse
|