1
|
Ning L, Zanella S, Tomov ML, Amoli MS, Jin L, Hwang B, Saadeh M, Chen H, Neelakantan S, Dasi LP, Avazmohammadi R, Mahmoudi M, Bauser‐Heaton HD, Serpooshan V. Targeted Rapamycin Delivery via Magnetic Nanoparticles to Address Stenosis in a 3D Bioprinted in Vitro Model of Pulmonary Veins. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2400476. [PMID: 38696618 PMCID: PMC11234432 DOI: 10.1002/advs.202400476] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Revised: 04/09/2024] [Indexed: 05/04/2024]
Abstract
Vascular cell overgrowth and lumen size reduction in pulmonary vein stenosis (PVS) can result in elevated PV pressure, pulmonary hypertension, cardiac failure, and death. Administration of chemotherapies such as rapamycin have shown promise by inhibiting the vascular cell proliferation; yet clinical success is limited due to complications such as restenosis and off-target effects. The lack of in vitro models to recapitulate the complex pathophysiology of PVS has hindered the identification of disease mechanisms and therapies. This study integrated 3D bioprinting, functional nanoparticles, and perfusion bioreactors to develop a novel in vitro model of PVS. Bioprinted bifurcated PV constructs are seeded with endothelial cells (ECs) and perfused, demonstrating the formation of a uniform and viable endothelium. Computational modeling identified the bifurcation point at high risk of EC overgrowth. Application of an external magnetic field enabled targeting of the rapamycin-loaded superparamagnetic iron oxide nanoparticles at the bifurcation site, leading to a significant reduction in EC proliferation with no adverse side effects. These results establish a 3D bioprinted in vitro model to study PV homeostasis and diseases, offering the potential for increased throughput, tunability, and patient specificity, to test new or more effective therapies for PVS and other vascular diseases.
Collapse
Affiliation(s)
- Liqun Ning
- Wallace H. Coulter Department of Biomedical EngineeringEmory University School of Medicine and Georgia Institute of TechnologyAtlantaGA30322USA
- Department of Mechanical EngineeringCleveland State UniversityClevelandOH44115USA
| | - Stefano Zanella
- Wallace H. Coulter Department of Biomedical EngineeringEmory University School of Medicine and Georgia Institute of TechnologyAtlantaGA30322USA
| | - Martin L. Tomov
- Wallace H. Coulter Department of Biomedical EngineeringEmory University School of Medicine and Georgia Institute of TechnologyAtlantaGA30322USA
| | - Mehdi Salar Amoli
- Wallace H. Coulter Department of Biomedical EngineeringEmory University School of Medicine and Georgia Institute of TechnologyAtlantaGA30322USA
| | - Linqi Jin
- Wallace H. Coulter Department of Biomedical EngineeringEmory University School of Medicine and Georgia Institute of TechnologyAtlantaGA30322USA
| | - Boeun Hwang
- Wallace H. Coulter Department of Biomedical EngineeringEmory University School of Medicine and Georgia Institute of TechnologyAtlantaGA30322USA
| | - Maher Saadeh
- Wallace H. Coulter Department of Biomedical EngineeringEmory University School of Medicine and Georgia Institute of TechnologyAtlantaGA30322USA
| | - Huang Chen
- Wallace H. Coulter Department of Biomedical EngineeringEmory University School of Medicine and Georgia Institute of TechnologyAtlantaGA30322USA
| | - Sunder Neelakantan
- Department of Biomedical EngineeringTexas A&M UniversityCollege StationTX77843USA
| | - Lakshmi Prasad Dasi
- Wallace H. Coulter Department of Biomedical EngineeringEmory University School of Medicine and Georgia Institute of TechnologyAtlantaGA30322USA
| | - Reza Avazmohammadi
- Department of Biomedical EngineeringTexas A&M UniversityCollege StationTX77843USA
- J. Mike Walker ’66 Department of Mechanical EngineeringTexas A&M UniversityCollege StationTX77840USA
| | - Morteza Mahmoudi
- Department of Radiology and Precision Health ProgramMichigan State UniversityEast LandingMI48824USA
| | - Holly D. Bauser‐Heaton
- Wallace H. Coulter Department of Biomedical EngineeringEmory University School of Medicine and Georgia Institute of TechnologyAtlantaGA30322USA
- Department of PediatricsEmory University School of MedicineAtlantaGA30322USA
- Children's Healthcare of AtlantaAtlantaGA30322USA
- Sibley Heart Center at Children's Healthcare of AtlantaAtlantaGA30322USA
| | - Vahid Serpooshan
- Wallace H. Coulter Department of Biomedical EngineeringEmory University School of Medicine and Georgia Institute of TechnologyAtlantaGA30322USA
- Department of PediatricsEmory University School of MedicineAtlantaGA30322USA
- Children's Healthcare of AtlantaAtlantaGA30322USA
| |
Collapse
|
2
|
Ren J, Zhang X, Zhang Z, Pan J, Hao Z, Li J, Liu J. Apoptosis inhibition enhances induced pluripotent stem cell generation during T cell reprogramming. Biochem Biophys Res Commun 2023; 656:30-37. [PMID: 36947964 DOI: 10.1016/j.bbrc.2023.03.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Accepted: 03/09/2023] [Indexed: 03/12/2023]
Abstract
The widespread adoption of chimeric antigen receptor (CAR)-T cell therapy has been hindered by its complex and costly manufacturing process. Induced pluripotent stem cells (iPSCs) have shown promise as a cellular immunotherapy alternative, due to their unlimited self-renewal potential in culture and capacity to differentiate into functional immune cell types. However, it is imperative to carefully select the original cell for iPSC seed preparation, as iPSCs have been found to retain the epigenetic imprint of the original somatic cells. Additionally, the efficiency of reprogramming terminal differentiated cells for immunotherapy must be addressed. Our research highlights the superiority of lymphocyte-origin cells over embryonic stem cells in functional immune cell differentiation. Furthermore, blocking Fas-FasL induced apoptosis in T cells significantly improves iPSC generation. Interestingly, transient Fas suppression in T cells does not alter the expression of Fas in the resulting iPSCs or affect their differentiation potential. This finding brings up new avenues in the field of cellular immunotherapy and provides a solution for creating high-quality and suitable iPSCs for lymphocyte differentiation for immunotherapy purposes.
Collapse
Affiliation(s)
- Jiangtao Ren
- Department of Oncology, State Key Laboratory of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, National Clinical Research Center for Respiratory Disease, Guangzhou Medical University, Guangzhou, 510530, China; Nanjing Bioheng Biotech Co., Ltd, Nanjing, Jiangsu, China
| | - Xuhua Zhang
- The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, 450008, China
| | - Zhenhui Zhang
- Department of Oncology, State Key Laboratory of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, National Clinical Research Center for Respiratory Disease, Guangzhou Medical University, Guangzhou, 510530, China; Anshun People's Hospital, Anshun, 561000, China
| | - Jiafeng Pan
- Department of Oncology, State Key Laboratory of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, National Clinical Research Center for Respiratory Disease, Guangzhou Medical University, Guangzhou, 510530, China; Anshun People's Hospital, Anshun, 561000, China
| | - Zhexue Hao
- Department of Oncology, State Key Laboratory of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, National Clinical Research Center for Respiratory Disease, Guangzhou Medical University, Guangzhou, 510530, China; Anshun People's Hospital, Anshun, 561000, China
| | - Jin Li
- Department of Oncology, State Key Laboratory of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, National Clinical Research Center for Respiratory Disease, Guangzhou Medical University, Guangzhou, 510530, China; Anshun People's Hospital, Anshun, 561000, China.
| | - Jun Liu
- Department of Oncology, State Key Laboratory of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, National Clinical Research Center for Respiratory Disease, Guangzhou Medical University, Guangzhou, 510530, China.
| |
Collapse
|
3
|
Hu N, Li W, Jiang W, Wen J, Gu S. Creating a Microenvironment to Give Wings to Dental Pulp Regeneration-Bioactive Scaffolds. Pharmaceutics 2023; 15:158. [PMID: 36678787 PMCID: PMC9861529 DOI: 10.3390/pharmaceutics15010158] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 12/13/2022] [Accepted: 12/23/2022] [Indexed: 01/05/2023] Open
Abstract
Dental pulp and periapical diseases make patients suffer from acute pain and economic loss. Although root canal therapies, as demonstrated through evidence-based medicine, can relieve symptoms and are commonly employed by dentists, it is still difficult to fully restore a dental pulp's nutrition, sensory, and immune-regulation functions. In recent years, researchers have made significant progress in tissue engineering to regenerate dental pulp in a desired microenvironment. With breakthroughs in regenerative medicine and material science, bioactive scaffolds play a pivotal role in creating a suitable microenvironment for cell survival, proliferation, and differentiation, following dental restoration and regeneration. This article focuses on current challenges and novel perspectives about bioactive scaffolds in creating a microenvironment to promote dental pulp regeneration. We hope our readers will gain a deeper understanding and new inspiration of dental pulp regeneration through our summary.
Collapse
Affiliation(s)
- Nan Hu
- Department of Endodontics, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, College of Stomatology, Shanghai Jiao Tong University, Shanghai 200011, China
- National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of Stomatology, Shanghai 200011, China
| | - Weiping Li
- National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of Stomatology, Shanghai 200011, China
- Department of Oral and Maxillofacial Head & Neck Oncology, Shanghai Ninth People’s Hospital, College of Stomatology, Shanghai Jiao Tong University, Shanghai 200011, China
| | - Wentao Jiang
- Department of Endodontics, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, College of Stomatology, Shanghai Jiao Tong University, Shanghai 200011, China
- National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of Stomatology, Shanghai 200011, China
| | - Jin Wen
- National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of Stomatology, Shanghai 200011, China
- Department of Prosthodontics, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, College of Stomatology, Shanghai Jiao Tong University, Shanghai 200011, China
- Shanghai Key Laboratory of Stomatology, Shanghai Engineering Research Center of Advanced Dental Technology and Materials, Shanghai 200125, China
| | - Shensheng Gu
- Department of Endodontics, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, College of Stomatology, Shanghai Jiao Tong University, Shanghai 200011, China
- National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of Stomatology, Shanghai 200011, China
| |
Collapse
|
4
|
Marcano L, Orue I, Gandia D, Gandarias L, Weigand M, Abrudan RM, García-Prieto A, García-Arribas A, Muela A, Fdez-Gubieda ML, Valencia S. Magnetic Anisotropy of Individual Nanomagnets Embedded in Biological Systems Determined by Axi-asymmetric X-ray Transmission Microscopy. ACS NANO 2022; 16:7398-7408. [PMID: 35472296 PMCID: PMC9878725 DOI: 10.1021/acsnano.1c09559] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/19/2023]
Abstract
Over the past few years, the use of nanomagnets in biomedical applications has increased. Among others, magnetic nanostructures can be used as diagnostic and therapeutic agents in cardiovascular diseases, to locally destroy cancer cells, to deliver drugs at specific positions, and to guide (and track) stem cells to damaged body locations in regenerative medicine and tissue engineering. All these applications rely on the magnetic properties of the nanomagnets which are mostly determined by their magnetic anisotropy. Despite its importance, the magnetic anisotropy of the individual magnetic nanostructures is unknown. Currently available magnetic sensitive microscopic methods are either limited in spatial resolution or in magnetic field strength or, more relevant, do not allow one to measure magnetic signals of nanomagnets embedded in biological systems. Hence, the use of nanomagnets in biomedical applications must rely on mean values obtained after averaging samples containing thousands of dissimilar entities. Here we present a hybrid experimental/theoretical method capable of working out the magnetic anisotropy constant and the magnetic easy axis of individual magnetic nanostructures embedded in biological systems. The method combines scanning transmission X-ray microscopy using an axi-asymmetric magnetic field with theoretical simulations based on the Stoner-Wohlfarth model. The validity of the method is demonstrated by determining the magnetic anisotropy constant and magnetic easy axis direction of 15 intracellular magnetite nanoparticles (50 nm in size) biosynthesized inside a magnetotactic bacterium.
Collapse
Affiliation(s)
- Lourdes Marcano
- Helmholtz-Zentrum
Berlin für Materialien und Energie, Albert-Einstein-Str. 15, 12489 Berlin, Germany
- Dpto.
Electricidad y Electrónica, Universidad
del País Vasco - UPV/EHU, 48940 Leioa, Spain
| | - Iñaki Orue
- SGIker, Universidad del País Vasco - UPV/EHU, 48940 Leioa, Spain
| | - David Gandia
- BCMaterials, Bld. Martina Casiano third floor, 48940 Leioa, Spain
| | - Lucía Gandarias
- Dpto.
Inmunología, Microbiología y Parasitología, Universidad del País Vasco - UPV/EHU, 48940 Leioa, Spain
| | - Markus Weigand
- Helmholtz-Zentrum
Berlin für Materialien und Energie, Albert-Einstein-Str. 15, 12489 Berlin, Germany
| | - Radu Marius Abrudan
- Helmholtz-Zentrum
Berlin für Materialien und Energie, Albert-Einstein-Str. 15, 12489 Berlin, Germany
| | - Ana García-Prieto
- Dpto. Física
Aplicada, Universidad del País Vasco - UPV/EHU, 48013 Bilbao, Spain
| | - Alfredo García-Arribas
- Dpto.
Electricidad y Electrónica, Universidad
del País Vasco - UPV/EHU, 48940 Leioa, Spain
- BCMaterials, Bld. Martina Casiano third floor, 48940 Leioa, Spain
| | - Alicia Muela
- Dpto.
Inmunología, Microbiología y Parasitología, Universidad del País Vasco - UPV/EHU, 48940 Leioa, Spain
| | - M. Luisa Fdez-Gubieda
- Dpto.
Electricidad y Electrónica, Universidad
del País Vasco - UPV/EHU, 48940 Leioa, Spain
- BCMaterials, Bld. Martina Casiano third floor, 48940 Leioa, Spain
| | - Sergio Valencia
- Helmholtz-Zentrum
Berlin für Materialien und Energie, Albert-Einstein-Str. 15, 12489 Berlin, Germany
| |
Collapse
|
5
|
Filippi M, Garello F, Yasa O, Kasamkattil J, Scherberich A, Katzschmann RK. Engineered Magnetic Nanocomposites to Modulate Cellular Function. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2022; 18:e2104079. [PMID: 34741417 DOI: 10.1002/smll.202104079] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 09/13/2021] [Indexed: 06/13/2023]
Abstract
Magnetic nanoparticles (MNPs) have various applications in biomedicine, including imaging, drug delivery and release, genetic modification, cell guidance, and patterning. By combining MNPs with polymers, magnetic nanocomposites (MNCs) with diverse morphologies (core-shell particles, matrix-dispersed particles, microspheres, etc.) can be generated. These MNCs retain the ability of MNPs to be controlled remotely using external magnetic fields. While the effects of these biomaterials on the cell biology are still poorly understood, such information can help the biophysical modulation of various cellular functions, including proliferation, adhesion, and differentiation. After recalling the basic properties of MNPs and polymers, and describing their coassembly into nanocomposites, this review focuses on how polymeric MNCs can be used in several ways to affect cell behavior. A special emphasis is given to 3D cell culture models and transplantable grafts, which are used for regenerative medicine, underlining the impact of MNCs in regulating stem cell differentiation and engineering living tissues. Recent advances in the use of MNCs for tissue regeneration are critically discussed, particularly with regard to their prospective involvement in human therapy and in the construction of advanced functional materials such as magnetically operated biomedical robots.
Collapse
Affiliation(s)
- Miriam Filippi
- Soft Robotics Laboratory, ETH Zurich, Tannenstrasse 3, Zurich, 8092, Switzerland
| | - Francesca Garello
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Via Nizza 52, Torino, 10126, Italy
| | - Oncay Yasa
- Soft Robotics Laboratory, ETH Zurich, Tannenstrasse 3, Zurich, 8092, Switzerland
| | - Jesil Kasamkattil
- Department of Biomedicine, University Hospital Basel, Hebelstrasse 20, Basel, 4031, Switzerland
| | - Arnaud Scherberich
- Department of Biomedicine, University Hospital Basel, Hebelstrasse 20, Basel, 4031, Switzerland
- Department of Biomedical Engineering, University of Basel, Gewerbestrasse 14, Allschwil, 4123, Switzerland
| | - Robert K Katzschmann
- Soft Robotics Laboratory, ETH Zurich, Tannenstrasse 3, Zurich, 8092, Switzerland
| |
Collapse
|
6
|
Kiyotake EA, Martin MD, Detamore MS. Regenerative rehabilitation with conductive biomaterials for spinal cord injury. Acta Biomater 2022; 139:43-64. [PMID: 33326879 DOI: 10.1016/j.actbio.2020.12.021] [Citation(s) in RCA: 46] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 11/24/2020] [Accepted: 12/09/2020] [Indexed: 02/07/2023]
Abstract
The individual approaches of regenerative medicine efforts alone and rehabilitation efforts alone have not yet fully restored function after severe spinal cord injury (SCI). Regenerative rehabilitation may be leveraged to promote regeneration of the spinal cord tissue, and promote reorganization of the regenerated neural pathways and intact spinal circuits for better functional recovery for SCI. Conductive biomaterials may be a linchpin that empowers the synergy between regenerative medicine and rehabilitation approaches, as electrical stimulation applied to the spinal cord could facilitate neural reorganization. In this review, we discuss current regenerative medicine approaches in clinical trials and the rehabilitation, or neuromodulation, approaches for SCI, along with their respective translational limitations. Furthermore, we review the translational potential, in a surgical context, of conductive biomaterials (e.g., conductive polymers, carbon-based materials, metallic nanoparticle-based materials) as they pertain to SCI. While pre-formed scaffolds may be difficult to translate to human contusion SCIs, injectable composites that contain blended conductive components and can form within the injury may be more translational. However, given that there are currently no in vivo SCI studies that evaluated conductive materials combined with rehabilitation approaches, we discuss several limitations of conductive biomaterials, including demonstrating safety and efficacy, that will need to be addressed in the future for conductive biomaterials to become SCI therapeutics. Even so, the use of conductive biomaterials creates a synergistic opportunity to merge the fields of regenerative medicine and rehabilitation and redefine what regenerative rehabilitation means for the spinal cord. STATEMENT OF SIGNIFICANCE: For spinal cord injury (SCI), the individual approaches of regenerative medicine and rehabilitation are insufficient to fully restore functional recovery; however, the goal of regenerative rehabilitation is to combine these two disparate fields to maximize the functional outcomes. Concepts similar to regenerative rehabilitation for SCI have been discussed in several reviews, but for the first time, this review considers how conductive biomaterials may synergize the two approaches. We cover current regenerative medicine and rehabilitation approaches for SCI, and the translational advantages and disadvantages, in a surgical context, of conductive biomaterials used in biomedical applications that may be additionally applied to SCI. Furthermore, we identify the current limitations and translational challenges for conductive biomaterials before they may become therapeutics for SCI.
Collapse
|
7
|
Go S, Kang M, Kwon SP, Jung M, Jeon OH, Kim B. The Senolytic Drug JQ1 Removes Senescent Cells via Ferroptosis. Tissue Eng Regen Med 2021; 18:841-850. [PMID: 34003467 PMCID: PMC8440740 DOI: 10.1007/s13770-021-00346-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Revised: 04/14/2021] [Accepted: 04/19/2021] [Indexed: 01/18/2023] Open
Abstract
BACKGROUND Ferroptosis is an iron-dependent, non-apoptotic programmed cell death. Cellular senescence contributes to aging and various age-related diseases through the expression of a senescence-associated secretory phenotype (SASP). Senescent cells are often resistant to ferroptosis via increased ferritin and impaired ferritinophagy. In this study, we investigated whether treatment with JQ1 could remove senescent cells by inducing ferroptosis. METHODS Senescence of human dermal fibroblasts was induced in vitro by treating the cells with bleomycin. The senolytic effects of JQ1 were evaluated using a SA-β gal assay, annexin V analysis, cell counting kit-8 assay, and qRT-PCR. Ferroptosis following JQ1 treatment was evaluated with qRT-PCR and BODIPY staining. RESULTS At a certain range of JQ1 concentrations, JQ1 treatment reduced the viability of bleomycin-treated cells (senescent cells) but did not reduce that of untreated cells (non-senescent cells), indicating that JQ1 treatment can selectively eliminate senescent cells. JQ1 treatment also decreased SASP expression only in senescent cells. Subsequently, JQ1 treatment reduced the expression of ferroptosis-resistance genes in senescent cells. JQ1 treatment induced lipid peroxidation in senescent cells but not in non-senescent cells. CONCLUSION The data indicate that JQ1 can eliminate senescent cells via ferroptosis. This study suggests ferroptosis as a new mechanism of senolytic therapy.
Collapse
Affiliation(s)
- Seokhyeong Go
- Interdisciplinary Program for Bioengineering, Seoul National University, Seoul, 08826 Republic of Korea
| | - Mikyung Kang
- Interdisciplinary Program for Bioengineering, Seoul National University, Seoul, 08826 Republic of Korea
| | - Sung Pil Kwon
- School of Chemical and Biological Engineering, Seoul National University, Seoul, 08826 Republic of Korea
| | - Mungyo Jung
- School of Chemical and Biological Engineering, Seoul National University, Seoul, 08826 Republic of Korea
| | - Ok Hee Jeon
- Department of Biomedical Sciences, BK21 Graduate Program, Korea University of College of Medicine, Seoul, 02841, Republic of Korea.
| | - Byung‐Soo Kim
- Interdisciplinary Program for Bioengineering, Seoul National University, Seoul, 08826 Republic of Korea ,School of Chemical and Biological Engineering, Seoul National University, Seoul, 08826 Republic of Korea ,Institute of Chemical Processes, Institute of Engineering Research, and BioMAX, Seoul National University, Seoul, 08826 Republic of Korea
| |
Collapse
|
8
|
Kono Y, Takegaki J, Ohba T, Matsuda K, Negoro R, Fujita S, Fujita T. Magnetization of mesenchymal stem cells using magnetic liposomes enhances their retention and immunomodulatory efficacy in mouse inflamed skeletal muscle. Int J Pharm 2021; 596:120298. [PMID: 33529784 DOI: 10.1016/j.ijpharm.2021.120298] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Revised: 01/07/2021] [Accepted: 01/15/2021] [Indexed: 12/24/2022]
Abstract
Sarcopenia, an age-related reduction in skeletal muscle mass and strength, is mainly caused by chronic inflammation. Because mesenchymal stem cells (MSCs) have the capacity to both promote myogenic cell differentiation and suppress inflammation, they are a promising candidate for sarcopenia treatment. In this study, to achieve the long-term retention of MSCs in skeletal muscle, we prepared magnetized MSCs using magnetic anionic liposome/atelocollagen complexes that we had previously developed, and evaluated their retention efficiency and immunomodulatory effects in mouse inflamed skeletal muscle. Mouse MSCs were efficiently magnetized by incubation with magnetic anionic liposome/atelocollagen complexes for 30 min under a magnetic field. The magnetized MSCs differentiated normally into osteoblasts and adipocytes. Additionally, non-magnetized MSCs and magnetized MSCs increased IL-6 and inducible nitric oxide synthase mRNA expression and decreased TNF-α and IL-1β mRNA expression in C2C12 mouse skeletal muscle myotubes through paracrine effects. Moreover, magnetized MSCs were significantly retained in cell culture plates and mouse skeletal muscle after their local injection in the presence of a magnetic field. Furthermore, magnetized MSCs significantly increased IL-6 and IL-10 mRNA expression and decreased TNF-α and IL-1β mRNA expression in inflamed skeletal muscle. These results suggest that magnetized MSCs may be useful for effective sarcopenia treatment.
Collapse
Affiliation(s)
- Yusuke Kono
- Ritsumeikan-Global Innovation Research Organization, Ritsumeikan University, 1-1-1 Noji-Higashi, Kusatsu 525-8577, Japan.
| | - Junya Takegaki
- Ritsumeikan-Global Innovation Research Organization, Ritsumeikan University, 1-1-1 Noji-Higashi, Kusatsu 525-8577, Japan; Faculty of Sport and Health Science, Ritsumeikan University, 1-1-1 Noji-Higashi, Kusatsu 525-8577, Japan
| | - Takeshi Ohba
- Laboratory of Molecular Pharmacokinetics, College of Pharmaceutical Sciences, Ritsumeikan University, 1-1-1 Noji-Higashi, Kusatsu 525-8577, Japan
| | - Koji Matsuda
- Laboratory of Molecular Pharmacokinetics, College of Pharmaceutical Sciences, Ritsumeikan University, 1-1-1 Noji-Higashi, Kusatsu 525-8577, Japan
| | - Ryosuke Negoro
- Laboratory of Molecular Pharmacokinetics, College of Pharmaceutical Sciences, Ritsumeikan University, 1-1-1 Noji-Higashi, Kusatsu 525-8577, Japan
| | - Satoshi Fujita
- Ritsumeikan-Global Innovation Research Organization, Ritsumeikan University, 1-1-1 Noji-Higashi, Kusatsu 525-8577, Japan; Faculty of Sport and Health Science, Ritsumeikan University, 1-1-1 Noji-Higashi, Kusatsu 525-8577, Japan
| | - Takuya Fujita
- Ritsumeikan-Global Innovation Research Organization, Ritsumeikan University, 1-1-1 Noji-Higashi, Kusatsu 525-8577, Japan; Laboratory of Molecular Pharmacokinetics, College of Pharmaceutical Sciences, Ritsumeikan University, 1-1-1 Noji-Higashi, Kusatsu 525-8577, Japan; Research Center for Drug Discovery and Development, Ritsumeikan University, 1-1-1 Noji-Higashi, Kusatsu 525-8577, Japan
| |
Collapse
|
9
|
Sanjurjo-Rodríguez C, Castro-Viñuelas R, Piñeiro-Ramil M, Rodríguez-Fernández S, Fuentes-Boquete I, Blanco FJ, Díaz-Prado S. Versatility of Induced Pluripotent Stem Cells (iPSCs) for Improving the Knowledge on Musculoskeletal Diseases. Int J Mol Sci 2020; 21:ijms21176124. [PMID: 32854405 PMCID: PMC7504376 DOI: 10.3390/ijms21176124] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Revised: 08/06/2020] [Accepted: 08/20/2020] [Indexed: 12/13/2022] Open
Abstract
Induced pluripotent stem cells (iPSCs) represent an unlimited source of pluripotent cells capable of differentiating into any cell type of the body. Several studies have demonstrated the valuable use of iPSCs as a tool for studying the molecular and cellular mechanisms underlying disorders affecting bone, cartilage and muscle, as well as their potential for tissue repair. Musculoskeletal diseases are one of the major causes of disability worldwide and impose an important socio-economic burden. To date there is neither cure nor proven approach for effectively treating most of these conditions and therefore new strategies involving the use of cells have been increasingly investigated in the recent years. Nevertheless, some limitations related to the safety and differentiation protocols among others remain, which humpers the translational application of these strategies. Nonetheless, the potential is indisputable and iPSCs are likely to be a source of different types of cells useful in the musculoskeletal field, for either disease modeling or regenerative medicine. In this review, we aim to illustrate the great potential of iPSCs by summarizing and discussing the in vitro tissue regeneration preclinical studies that have been carried out in the musculoskeletal field by using iPSCs.
Collapse
Affiliation(s)
- Clara Sanjurjo-Rodríguez
- Cell Therapy and Regenerative Medicine Group, Department of Physiotherapy, Medicine and Biomedical Sciences, Faculty of Health Sciences, University of A Coruña (UDC), 15006 A Coruña, Galicia, Spain; (R.C.-V.); (M.P.-R.); (S.R.-F.); (I.F.-B.)
- Institute of Biomedical Research of A Coruña (INIBIC), University Hospital Complex A Coruña (CHUAC), Galician Health Service (SERGAS), 15006 A Coruña, Galicia, Spain;
- Centro de Investigación Biomédica en Red (CIBER) de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), 28029 Madrid, Spain
- Centro de Investigaciones Científicas Avanzadas (CICA), Agrupación estratégica CICA-INIBIC, University of A Coruña, 15008 A Coruña, Galicia, Spain
- Correspondence: (C.S.-R.); (S.D.-P.)
| | - Rocío Castro-Viñuelas
- Cell Therapy and Regenerative Medicine Group, Department of Physiotherapy, Medicine and Biomedical Sciences, Faculty of Health Sciences, University of A Coruña (UDC), 15006 A Coruña, Galicia, Spain; (R.C.-V.); (M.P.-R.); (S.R.-F.); (I.F.-B.)
- Institute of Biomedical Research of A Coruña (INIBIC), University Hospital Complex A Coruña (CHUAC), Galician Health Service (SERGAS), 15006 A Coruña, Galicia, Spain;
- Centro de Investigaciones Científicas Avanzadas (CICA), Agrupación estratégica CICA-INIBIC, University of A Coruña, 15008 A Coruña, Galicia, Spain
| | - María Piñeiro-Ramil
- Cell Therapy and Regenerative Medicine Group, Department of Physiotherapy, Medicine and Biomedical Sciences, Faculty of Health Sciences, University of A Coruña (UDC), 15006 A Coruña, Galicia, Spain; (R.C.-V.); (M.P.-R.); (S.R.-F.); (I.F.-B.)
- Institute of Biomedical Research of A Coruña (INIBIC), University Hospital Complex A Coruña (CHUAC), Galician Health Service (SERGAS), 15006 A Coruña, Galicia, Spain;
- Centro de Investigaciones Científicas Avanzadas (CICA), Agrupación estratégica CICA-INIBIC, University of A Coruña, 15008 A Coruña, Galicia, Spain
| | - Silvia Rodríguez-Fernández
- Cell Therapy and Regenerative Medicine Group, Department of Physiotherapy, Medicine and Biomedical Sciences, Faculty of Health Sciences, University of A Coruña (UDC), 15006 A Coruña, Galicia, Spain; (R.C.-V.); (M.P.-R.); (S.R.-F.); (I.F.-B.)
- Institute of Biomedical Research of A Coruña (INIBIC), University Hospital Complex A Coruña (CHUAC), Galician Health Service (SERGAS), 15006 A Coruña, Galicia, Spain;
- Centro de Investigaciones Científicas Avanzadas (CICA), Agrupación estratégica CICA-INIBIC, University of A Coruña, 15008 A Coruña, Galicia, Spain
| | - Isaac Fuentes-Boquete
- Cell Therapy and Regenerative Medicine Group, Department of Physiotherapy, Medicine and Biomedical Sciences, Faculty of Health Sciences, University of A Coruña (UDC), 15006 A Coruña, Galicia, Spain; (R.C.-V.); (M.P.-R.); (S.R.-F.); (I.F.-B.)
- Institute of Biomedical Research of A Coruña (INIBIC), University Hospital Complex A Coruña (CHUAC), Galician Health Service (SERGAS), 15006 A Coruña, Galicia, Spain;
- Centro de Investigaciones Científicas Avanzadas (CICA), Agrupación estratégica CICA-INIBIC, University of A Coruña, 15008 A Coruña, Galicia, Spain
| | - Francisco J. Blanco
- Institute of Biomedical Research of A Coruña (INIBIC), University Hospital Complex A Coruña (CHUAC), Galician Health Service (SERGAS), 15006 A Coruña, Galicia, Spain;
- Centro de Investigación Biomédica en Red (CIBER) de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), 28029 Madrid, Spain
- Centro de Investigaciones Científicas Avanzadas (CICA), Agrupación estratégica CICA-INIBIC, University of A Coruña, 15008 A Coruña, Galicia, Spain
- Tissular Bioengineering and Cell Therapy Unit (GBTTC-CHUAC), Rheumatology Group, 15006 A Coruña, Galicia, Spain
| | - Silvia Díaz-Prado
- Cell Therapy and Regenerative Medicine Group, Department of Physiotherapy, Medicine and Biomedical Sciences, Faculty of Health Sciences, University of A Coruña (UDC), 15006 A Coruña, Galicia, Spain; (R.C.-V.); (M.P.-R.); (S.R.-F.); (I.F.-B.)
- Institute of Biomedical Research of A Coruña (INIBIC), University Hospital Complex A Coruña (CHUAC), Galician Health Service (SERGAS), 15006 A Coruña, Galicia, Spain;
- Centro de Investigación Biomédica en Red (CIBER) de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), 28029 Madrid, Spain
- Centro de Investigaciones Científicas Avanzadas (CICA), Agrupación estratégica CICA-INIBIC, University of A Coruña, 15008 A Coruña, Galicia, Spain
- Correspondence: (C.S.-R.); (S.D.-P.)
| |
Collapse
|