1
|
Sun Q, Mu X, Gao Q, Wang J, Hu M, Liu H. Influences of physical stimulations on the migration and differentiation of Schwann cells involved in peripheral nerve repair. Cell Adh Migr 2025; 19:2450311. [PMID: 39817348 PMCID: PMC11740713 DOI: 10.1080/19336918.2025.2450311] [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: 07/02/2024] [Revised: 12/23/2024] [Accepted: 01/02/2025] [Indexed: 01/18/2025] Open
Abstract
Peripheral nerve injury repair has always been a research concern of scientists. At the tissue level, axonal regeneration has become a research spotlight in peripheral nerve repair. Through transplantation of autologous nerve grafts or other emerging biomaterials functional recovery after facial nerve injury is not ideal in clinical scenarios. Great strides have been made to improve facial nerve repair at the micro-cellular level. Physical stimulation techniques can trigger Schwann cells (SCs) to migrate and differentiate into cells required for peripheral nerve repair. Classified by the sources of physical stimulations, SCs repair peripheral nerves through galvanotaxis, magnetotaxis and durotaxis. This article summarized the activation, directional migration and differentiation of SCs induced by physical stimulations, thus providing new ideas for the research of peripheral nerve repair.
Collapse
Affiliation(s)
- Qingyan Sun
- Department of Stomatology, The First Medical Center, Chinese PLA General Hospital, Beijing, China
- Chinese People’s Liberation Army (PLA) Medical School, Beijing, China
| | - Xiaodan Mu
- Department of Stomatology, The First Medical Center, Chinese PLA General Hospital, Beijing, China
- Chinese People’s Liberation Army (PLA) Medical School, Beijing, China
- Department of Stomatology of Air Force Hospital in the Southern Theater, Guangzhou, Guangdong Province, China
| | - Qi Gao
- Department of Stomatology of Air Force Hospital in the Southern Theater, Guangzhou, Guangdong Province, China
| | - Juncheng Wang
- Department of Stomatology, The First Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Min Hu
- Department of Stomatology, The First Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Huawei Liu
- Department of Stomatology, The First Medical Center, Chinese PLA General Hospital, Beijing, China
| |
Collapse
|
2
|
Dogan AB, Marsh SR, Tschetter RJ, E Beard C, Amin MR, Jane Jourdan L, Gourdie RG. Stabilizing milk-derived extracellular vesicles (mEVs) through lyophilization: a novel trehalose and tryptophan formulation for maintaining structure and Bioactivity during long-term storage. J Biol Eng 2025; 19:4. [PMID: 39806456 PMCID: PMC11727230 DOI: 10.1186/s13036-024-00470-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Accepted: 11/26/2024] [Indexed: 01/16/2025] Open
Abstract
Extracellular vesicles (EVs) are widely investigated for their implications in cell-cell signaling, immune modulation, disease pathogenesis, cancer, regenerative medicine, and as a potential drug delivery vector. However, maintaining integrity and bioactivity of EVs between Good Manufacturing Practice separation/filtration and end-user application remains a consistent bottleneck towards commercialization. Milk-derived extracellular vesicles (mEVs), separated from bovine milk, could provide a relatively low-cost, scalable platform for large-scale mEV production; however, the reliance on cold supply chain for storage remains a logistical and financial burden for biologics that are unstable at room temperature. Herein, we aim to characterize and engineer a freeze-dried, mEV formulation that can be stored at room temperature without sacrificing structure/bioactivity and can be reconstituted before delivery. In addition to undertaking established mEV assays of structure and function on our preparations, we introduce a novel, efficient, high throughput assay of mEV bioactivity based on Electric Cell Substrate Impedance Sensing (ECIS) in Human dermal fibroblast monolayers. By adding appropriate excipients, such as trehalose and tryptophan, we describe a protective formulation that preserves mEV bioactivity during long-term, room temperature storage. Our identification of the efficacy of tryptophan as a novel additive to mEV lyophilization solutions could represent a significant advancement in stabilizing small extracellular vesicles outside of cold storage conditions.
Collapse
Affiliation(s)
- Alan B Dogan
- Virginia Tech Carilion School of Medicine, Roanoke, VA, 24016, USA
| | - Spencer R Marsh
- Fralin Biomedical Research Institute at Virginia Tech Carilion, Roanoke, VA, 24016, USA
- Center for Vascular and Heart Research, Virginia Tech, Roanoke, VA, 24016, USA
| | - Rachel J Tschetter
- Materials Science and Engineering, Virginia Tech, Blacksburg, VA, 24061, USA
| | - Claire E Beard
- Fralin Biomedical Research Institute at Virginia Tech Carilion, Roanoke, VA, 24016, USA
| | - Md R Amin
- Fralin Biomedical Research Institute at Virginia Tech Carilion, Roanoke, VA, 24016, USA
- Translational Biology, Medicine, and Health graduate program at Virginia Tech, Roanoke, VA, 24016, USA
| | - L Jane Jourdan
- Fralin Biomedical Research Institute at Virginia Tech Carilion, Roanoke, VA, 24016, USA
- Center for Vascular and Heart Research, Virginia Tech, Roanoke, VA, 24016, USA
| | - Robert G Gourdie
- Fralin Biomedical Research Institute at Virginia Tech Carilion, Roanoke, VA, 24016, USA.
- Center for Vascular and Heart Research, Virginia Tech, Roanoke, VA, 24016, USA.
- Department of Biomedical Engineering and Mechanics, Virginia Tech, Blacksburg, VA, 24061, USA.
- Department of Emergency Medicine, Virginia Tech Carilion School of Medicine, Virginia Tech, Roanoke, VA, 24016, USA.
- Faculty of Health Science, Virginia Tech, Blacksburg, VA, 24061, USA.
| |
Collapse
|
3
|
Estévez M, Cicuéndez M, Colilla M, Vallet-Regí M, González B, Izquierdo-Barba I. Magnetic colloidal nanoformulations to remotely trigger mechanotransduction for osteogenic differentiation. J Colloid Interface Sci 2024; 664:454-468. [PMID: 38484514 DOI: 10.1016/j.jcis.2024.03.043] [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: 12/11/2023] [Revised: 03/05/2024] [Accepted: 03/06/2024] [Indexed: 04/07/2024]
Abstract
Nowadays, diseases associated with an ageing population, such as osteoporosis, require the development of new biomedical approaches to bone regeneration. In this regard, mechanotransduction has emerged as a discipline within the field of bone tissue engineering. Herein, we have tested the efficacy of superparamagnetic iron oxide nanoparticles (SPIONs), obtained by the thermal decomposition method, with an average size of 13 nm, when exposed to the application of an external magnetic field for mechanotransduction in human bone marrow-derived mesenchymal stem cells (hBM-MSCs). The SPIONs were functionalized with an Arg-Gly-Asp (RGD) peptide as ligand to target integrin receptors on cell membrane and used in colloidal state. Then, a comprehensive and comparative bioanalytical characterization of non-targeted versus targeted SPIONs was performed in terms of biocompatibility, cell uptake pathways and mechanotransduction effect, demonstrating the osteogenic differentiation of hBM-MSCs. A key conclusion derived from this research is that when the magnetic stimulus is applied in the first 30 min of the in vitro assay, i.e., when the nanoparticles come into contact with the cell membrane surface to initiate endocytic pathways, a successful mechanotransduction effect is observed. Thus, under the application of a magnetic field, there was a significant increase in runt-related transcription factor 2 (Runx2) and alkaline phosphatase (ALP) gene expression as well as ALP activity, when cells were exposed to RGD-functionalized SPIONs, demonstrating osteogenic differentiation. These findings open new expectations for the use of remotely activated mechanotransduction using targeted magnetic colloidal nanoformulations for osteogenic differentiation by drug-free cell therapy using minimally invasive techniques in cases of bone loss.
Collapse
Affiliation(s)
- Manuel Estévez
- Departamento de Química en Ciencias Farmacéuticas, Facultad de Farmacia, Universidad Complutense de Madrid, Instituto de Investigación Sanitaria, Hospital 12 de Octubre i+12, Plaza Ramón y Cajal s/n, 28040 Madrid, Spain
| | - Mónica Cicuéndez
- Departamento de Química en Ciencias Farmacéuticas, Facultad de Farmacia, Universidad Complutense de Madrid, Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC), 28040 Madrid, Spain
| | - Montserrat Colilla
- Departamento de Química en Ciencias Farmacéuticas, Facultad de Farmacia, Universidad Complutense de Madrid, Instituto de Investigación Sanitaria, Hospital 12 de Octubre i+12, Plaza Ramón y Cajal s/n, 28040 Madrid, Spain; Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Spain
| | - María Vallet-Regí
- Departamento de Química en Ciencias Farmacéuticas, Facultad de Farmacia, Universidad Complutense de Madrid, Instituto de Investigación Sanitaria, Hospital 12 de Octubre i+12, Plaza Ramón y Cajal s/n, 28040 Madrid, Spain; Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Spain
| | - Blanca González
- Departamento de Química en Ciencias Farmacéuticas, Facultad de Farmacia, Universidad Complutense de Madrid, Instituto de Investigación Sanitaria, Hospital 12 de Octubre i+12, Plaza Ramón y Cajal s/n, 28040 Madrid, Spain; Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Spain.
| | - Isabel Izquierdo-Barba
- Departamento de Química en Ciencias Farmacéuticas, Facultad de Farmacia, Universidad Complutense de Madrid, Instituto de Investigación Sanitaria, Hospital 12 de Octubre i+12, Plaza Ramón y Cajal s/n, 28040 Madrid, Spain; Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Spain.
| |
Collapse
|
4
|
Latypova AA, Yaremenko AV, Pechnikova NA, Minin AS, Zubarev IV. Magnetogenetics as a promising tool for controlling cellular signaling pathways. J Nanobiotechnology 2024; 22:327. [PMID: 38858689 PMCID: PMC11163773 DOI: 10.1186/s12951-024-02616-z] [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: 03/28/2024] [Accepted: 06/04/2024] [Indexed: 06/12/2024] Open
Abstract
Magnetogenetics emerges as a transformative approach for modulating cellular signaling pathways through the strategic application of magnetic fields and nanoparticles. This technique leverages the unique properties of magnetic nanoparticles (MNPs) to induce mechanical or thermal stimuli within cells, facilitating the activation of mechano- and thermosensitive proteins without the need for traditional ligand-receptor interactions. Unlike traditional modalities that often require invasive interventions and lack precision in targeting specific cellular functions, magnetogenetics offers a non-invasive alternative with the capacity for deep tissue penetration and the potential for targeting a broad spectrum of cellular processes. This review underscores magnetogenetics' broad applicability, from steering stem cell differentiation to manipulating neuronal activity and immune responses, highlighting its potential in regenerative medicine, neuroscience, and cancer therapy. Furthermore, the review explores the challenges and future directions of magnetogenetics, including the development of genetically programmed magnetic nanoparticles and the integration of magnetic field-sensitive cells for in vivo applications. Magnetogenetics stands at the forefront of cellular manipulation technologies, offering novel insights into cellular signaling and opening new avenues for therapeutic interventions.
Collapse
Affiliation(s)
- Anastasiia A Latypova
- Institute of Future Biophysics, Dolgoprudny, 141701, Russia
- Moscow Center for Advanced Studies, Moscow, 123592, Russia
| | - Alexey V Yaremenko
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA.
- Aristotle University of Thessaloniki, Thessaloniki, 54124, Greece.
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, Moscow, 117997, Russia.
| | - Nadezhda A Pechnikova
- Aristotle University of Thessaloniki, Thessaloniki, 54124, Greece
- Saint Petersburg Pasteur Institute, Saint Petersburg, 197101, Russia
| | - Artem S Minin
- M.N. Mikheev Institute of Metal Physics of the Ural Branch of the Russian Academy of Sciences, Yekaterinburg, 620108, Russia
| | - Ilya V Zubarev
- Institute of Future Biophysics, Dolgoprudny, 141701, Russia.
| |
Collapse
|
5
|
Guan W, Gao H, Liu Y, Sun S, Li G. Application of magnetism in tissue regeneration: recent progress and future prospects. Regen Biomater 2024; 11:rbae048. [PMID: 38939044 PMCID: PMC11208728 DOI: 10.1093/rb/rbae048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Revised: 04/14/2024] [Accepted: 04/25/2024] [Indexed: 06/29/2024] Open
Abstract
Tissue regeneration is a hot topic in the field of biomedical research in this century. Material composition, surface topology, light, ultrasonic, electric field and magnetic fields (MFs) all have important effects on the regeneration process. Among them, MFs can provide nearly non-invasive signal transmission within biological tissues, and magnetic materials can convert MFs into a series of signals related to biological processes, such as mechanical force, magnetic heat, drug release, etc. By adjusting the MFs and magnetic materials, desired cellular or molecular-level responses can be achieved to promote better tissue regeneration. This review summarizes the definition, classification and latest progress of MFs and magnetic materials in tissue engineering. It also explores the differences and potential applications of MFs in different tissue cells, aiming to connect the applications of magnetism in various subfields of tissue engineering and provide new insights for the use of magnetism in tissue regeneration.
Collapse
Affiliation(s)
- Wenchao Guan
- Key Laboratory of Neuroregeneration, Co-innovation Center of Neuroregeneration, Nantong University, Nantong 226001, China
| | - Hongxia Gao
- Key Laboratory of Neuroregeneration, Co-innovation Center of Neuroregeneration, Nantong University, Nantong 226001, China
| | - Yaqiong Liu
- Key Laboratory of Neuroregeneration, Co-innovation Center of Neuroregeneration, Nantong University, Nantong 226001, China
| | - Shaolan Sun
- Key Laboratory of Neuroregeneration, Co-innovation Center of Neuroregeneration, Nantong University, Nantong 226001, China
| | - Guicai Li
- Key Laboratory of Neuroregeneration, Co-innovation Center of Neuroregeneration, Nantong University, Nantong 226001, China
- State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, China
| |
Collapse
|
6
|
Unnithan AR, Rotherham M, Markides H, El Haj AJ. Magnetic Ion Channel Activation (MICA)-Enabled Screening Assay: A Dynamic Platform for Remote Activation of Mechanosensitive Ion Channels. Int J Mol Sci 2023; 24:ijms24043364. [PMID: 36834776 PMCID: PMC9962865 DOI: 10.3390/ijms24043364] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Revised: 02/02/2023] [Accepted: 02/04/2023] [Indexed: 02/10/2023] Open
Abstract
This study reports results of a mechanical platform-based screening assay (MICA) to evaluate the remote activation of mechanosensitive ion channels. Here, we studied ERK pathway activation and the elevation in intracellular Ca2+ levels in response to the MICA application using the Luciferase assay and Fluo-8AM assay, respectively. Functionalised magnetic nanoparticles (MNPs) targeting membrane-bound integrins and mechanosensitive TREK1 ion channels were studied with HEK293 cell lines under MICA application. The study demonstrated that active targeting of mechanosensitive integrins via RGD (Arginylglycylaspartic acid) motifs or TREK1 (KCNK2, potassium channel subfamily K member 2) ion channels can stimulate the ERK pathway and intracellular calcium levels compared to non-MICA controls. This screening assay offers a powerful tool, which aligns with existing high-throughput drug screening platforms for use in the assessment of drugs that interact with ion channels and influence ion channel-modulated diseases.
Collapse
Affiliation(s)
- Afeesh Rajan Unnithan
- Healthcare Technology Institute, Institute of Translational Medicine, University of Birmingham, Birmingham B15 2TH, UK
- Centre for Pharmaceutical Engineering Science, School of Pharmacy and Medical Sciences, Faculty of Lifesciences, University of Bradford, Bradford BD7 1DP, UK
- Correspondence: (A.R.U.); (A.J.E.H.)
| | - Michael Rotherham
- Healthcare Technology Institute, Institute of Translational Medicine, University of Birmingham, Birmingham B15 2TH, UK
| | - Hareklea Markides
- Healthcare Technology Institute, Institute of Translational Medicine, University of Birmingham, Birmingham B15 2TH, UK
| | - Alicia J. El Haj
- Healthcare Technology Institute, Institute of Translational Medicine, University of Birmingham, Birmingham B15 2TH, UK
- Correspondence: (A.R.U.); (A.J.E.H.)
| |
Collapse
|
7
|
Yang J, Feng Y, Li Q, Zeng Y. Evidence of the static magnetic field effects on bone-related diseases and bone cells. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2023; 177:168-180. [PMID: 36462638 DOI: 10.1016/j.pbiomolbio.2022.11.006] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Revised: 11/28/2022] [Accepted: 11/30/2022] [Indexed: 12/03/2022]
Abstract
Static magnetic fields (SMFs), magnetic fields with constant intensity and orientation, have been extensively studied in the field of bone biology both fundamentally and clinically as a non-invasive physical factor. A large number of animal experiments and clinical studies have shown that SMFs have effective therapeutic effects on bone-related diseases such as non-healing fractures, bone non-union of bone implants, osteoporosis and osteoarthritis. The maintenance of bone health in adults depends on the basic functions of bone cells, such as bone formation by osteoblasts and bone resorption by osteoclasts. Numerous studies have revealed that SMFs can regulate the proliferation, differentiation, and function of bone tissue cells, including bone marrow mesenchymal stem cells (BMSCs), osteoblasts, bone marrow monocytes (BMMs), osteoclasts, and osteocytes. In this paper, the effects of SMFs on bone-related diseases and bone tissue cells are reviewed from both in vivo studies and in vitro studies, and the possible mechanisms are analyzed. In addition, some challenges that need to be further addressed in the research of SMF and bone are also discussed.
Collapse
Affiliation(s)
- Jiancheng Yang
- Department of Osteoporosis, Honghui Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Yan Feng
- Department of Osteoporosis, Honghui Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Qingmei Li
- Department of Osteoporosis, Honghui Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Yuhong Zeng
- Department of Osteoporosis, Honghui Hospital, Xi'an Jiaotong University, Xi'an, China.
| |
Collapse
|
8
|
Cabrera D, Eizadi Sharifabad M, Ranjbar JA, Telling ND, Harper AGS. Clot-targeted magnetic hyperthermia permeabilizes blood clots to make them more susceptible to thrombolysis. J Thromb Haemost 2022; 20:2556-2570. [PMID: 35950914 PMCID: PMC9826519 DOI: 10.1111/jth.15846] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 07/12/2022] [Accepted: 08/03/2022] [Indexed: 01/11/2023]
Abstract
BACKGROUND Thrombolysis is a frontline treatment for stroke, which involves the application of tissue plasminogen activator (tPA) to trigger endogenous clot-degradation pathways. However, it is only effective within 4.5 h of symptom onset because of clot contraction preventing tPA permeation into the clot. Magnetic hyperthermia (MH) mediated by tumor-targeted magnetic nanoparticles is used to treat cancer by using local heat generation to trigger apoptosis of cancer cells. OBJECTIVES To develop clot-targeting magnetic nanoparticles to deliver MH to the surface of human blood clots, and to assess whether this can improve the efficacy of thrombolysis of contracted blood clots. METHODS Clot-targeting magnetic nanoparticles were developed by functionalizing iron oxide nanoparticles with an antibody recognizing activated integrin αIIbβ3 (PAC-1). The magnetic properties of the PAC-1-tagged magnetic nanoparticles were characterized and optimized to deliver clot-targeted MH. RESULTS Clot-targeted MH increases the efficacy of tPA-mediated thrombolysis in contracted human blood clots, leading to a reduction in clot weight. MH increases the permeability of the clots to tPA, facilitating their breakdown. Scanning electron microscopy reveals that this effect is elicited through enhanced fibrin breakdown and triggering the disruption of red blood cells on the surface of the clot. Importantly, endothelial cells viability in a three-dimensional blood vessel model is unaffected by exposure to MH. CONCLUSIONS This study demonstrates that clot-targeted MH can enhance the thrombolysis of contracted human blood clots and can be safely applied to enhance the timeframe in which thrombolysis is effective.
Collapse
Affiliation(s)
- David Cabrera
- School of Pharmacy and BioengineeringGuy Hilton Research Centre, Keele UniversityStoke‐on‐TrentUK
| | - Maneea Eizadi Sharifabad
- School of Pharmacy and BioengineeringGuy Hilton Research Centre, Keele UniversityStoke‐on‐TrentUK
| | - Jacob A. Ranjbar
- School of Pharmacy and BioengineeringGuy Hilton Research Centre, Keele UniversityStoke‐on‐TrentUK
| | - Neil D. Telling
- School of Pharmacy and BioengineeringGuy Hilton Research Centre, Keele UniversityStoke‐on‐TrentUK
| | | |
Collapse
|
9
|
Labusca L, Danceanu C, Minuti AE, Herea DD, Ghemes A, Rotarescu C, Dragos-Pinzaru O, Tibu M, Marian G, Chiriac H, Lupu N. Magnetic nanowires substrate increases adipose-derived mesenchymal cells osteogenesis. Sci Rep 2022; 12:16698. [PMID: 36202902 PMCID: PMC9537172 DOI: 10.1038/s41598-022-21145-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Accepted: 09/22/2022] [Indexed: 11/08/2022] Open
Abstract
Magnetic nanomaterials are increasingly impacting the field of biology and medicine. Their versatility in terms of shape, structure, composition, coating, and magnetic responsivity make them attractive for drug delivery, cell targeting and imaging. Adipose derived-mesenchymal cells (ASCs) are intensely scrutinized for tissue engineering and regenerative medicine. However, differentiation into musculoskeletal lineages can be challenging. In this paper, we show that uncoated nickel nanowires (Ni NW) partially released from their alumina membrane offer a mechanically-responsive substrate with regular topography that can be used for the delivery of magneto-mechanical stimulation. We have used a tailored protocol for improving ASCs adherence to the substrate, and showed that cells retain their characteristic fibroblastic appearance, cytoskeletal fiber distribution and good viability. We report here for the first time significant increase in osteogenic but not adipogenic differentiation of ASCs on Ni NW exposed to 4 mT magnetic field compared to non-exposed. Moreover, magnetic actuation is shown to induce ASCs osteogenesis but not adipogenesis in the absence of external biochemical cues. While these findings need to be verified in vivo, the use of Ni NW substrate for inducing osteogenesis in the absence of specific differentiation factors is attractive for bone engineering. Implant coating with similar surfaces for orthopedic and dentistry could be as well envisaged as a modality to improve osteointegration.
Collapse
Affiliation(s)
- Luminita Labusca
- Department of Magnetic Devices and Materials, National Institute of Research and Development for Technical Physics, 700050, Iasi, Romania
- Orthopedics and Traumatology Clinic, County Emergency Hospital Saint Spiridon Iasi, 700111, Iasi, Romania
| | - Camelia Danceanu
- Department of Magnetic Devices and Materials, National Institute of Research and Development for Technical Physics, 700050, Iasi, Romania
| | - Anca Emanuela Minuti
- Department of Magnetic Devices and Materials, National Institute of Research and Development for Technical Physics, 700050, Iasi, Romania
- Alexandru Ioan Cuza University, Faculty of Physics, 700506, Iasi, Romania
| | - Dumitru-Daniel Herea
- Department of Magnetic Devices and Materials, National Institute of Research and Development for Technical Physics, 700050, Iasi, Romania.
| | - Adrian Ghemes
- Department of Magnetic Devices and Materials, National Institute of Research and Development for Technical Physics, 700050, Iasi, Romania
| | - Cristian Rotarescu
- Department of Magnetic Devices and Materials, National Institute of Research and Development for Technical Physics, 700050, Iasi, Romania
| | - Oana Dragos-Pinzaru
- Department of Magnetic Devices and Materials, National Institute of Research and Development for Technical Physics, 700050, Iasi, Romania
| | - Mihai Tibu
- Department of Magnetic Devices and Materials, National Institute of Research and Development for Technical Physics, 700050, Iasi, Romania
| | - Grigoras Marian
- Department of Magnetic Devices and Materials, National Institute of Research and Development for Technical Physics, 700050, Iasi, Romania
| | - Horia Chiriac
- Department of Magnetic Devices and Materials, National Institute of Research and Development for Technical Physics, 700050, Iasi, Romania
| | - Nicoleta Lupu
- Department of Magnetic Devices and Materials, National Institute of Research and Development for Technical Physics, 700050, Iasi, Romania
| |
Collapse
|
10
|
Hu B, Rotherham M, Farrow N, Roach P, Dobson J, El Haj AJ. Immobilization of Wnt Fragment Peptides on Magnetic Nanoparticles or Synthetic Surfaces Regulate Wnt Signaling Kinetics. Int J Mol Sci 2022; 23:10164. [PMID: 36077561 PMCID: PMC9456016 DOI: 10.3390/ijms231710164] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Revised: 08/29/2022] [Accepted: 08/30/2022] [Indexed: 11/18/2022] Open
Abstract
Wnt signaling plays an important role in embryogenesis and adult stem cell homeostasis. Its diminished activation is implicated in osteoporosis and degenerative neural diseases. However, systematic administration of Wnt-signaling agonists carries risk, as aberrantly activated Wnt/β-catenin signaling is linked to cancer. Therefore, technologies for local modulation and control of Wnt signaling targeted to specific sites of disease or degeneration have potential therapeutic value in the treatment of degenerative diseases. We reported a facile approach to locally activate the canonical Wnt signaling cascade using nanomagnetic actuation or ligand immobilized platforms. Using a human embryonic kidney (HEK293) Luc-TCF/LEF reporter cell line, we demonstrated that targeting the cell membrane Wnt receptor, Frizzled 2, with peptide-tagged magnetic nanoparticles (MNPs) triggered canonical Wnt signaling transduction when exposed to a high-gradient, time-varying magnetic field, and the induced TCF/LEF signal transduction was shown to be avidity-dependent. We also demonstrated that the peptide retained signaling activity after functionalization onto glass surfaces, providing a versatile platform for drug discovery or recreation of the cell niche. In conclusion, these results showed that peptide-mediated Wnt signaling kinetics depended not only on ligand concentration but also on the presentation method of the ligand, which may be further modulated by magnetic actuation. This has important implications when designing future therapeutic platforms involving Wnt mimetics.
Collapse
Affiliation(s)
- Bin Hu
- School of Pharmacy and Bioengineering, Guy Hilton Research Center, Keele University, Thornburrow Drive, Hartshill, Stoke-on-Trent, Staffordshire ST4 7QB, UK
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, China
| | - Michael Rotherham
- Healthcare Technologies Institute, School of Chemical Engineering, University of Birmingham, Heritage Building, Mindelsohn Way, Birmingham B15 2TH, UK
| | - Neil Farrow
- School of Pharmacy and Bioengineering, Guy Hilton Research Center, Keele University, Thornburrow Drive, Hartshill, Stoke-on-Trent, Staffordshire ST4 7QB, UK
| | - Paul Roach
- School of Pharmacy and Bioengineering, Guy Hilton Research Center, Keele University, Thornburrow Drive, Hartshill, Stoke-on-Trent, Staffordshire ST4 7QB, UK
- Department of Chemistry, Loughborough University, Leicestershire, Loughborough LE11 3TU, UK
| | - Jon Dobson
- J Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL 32611, USA
| | - Alicia J. El Haj
- School of Pharmacy and Bioengineering, Guy Hilton Research Center, Keele University, Thornburrow Drive, Hartshill, Stoke-on-Trent, Staffordshire ST4 7QB, UK
- Healthcare Technologies Institute, School of Chemical Engineering, University of Birmingham, Heritage Building, Mindelsohn Way, Birmingham B15 2TH, UK
| |
Collapse
|
11
|
Rotherham M, Nahar T, Broomhall TJ, Telling ND, El Haj AJ. Remote magnetic actuation of cell signalling for tissue engineering. CURRENT OPINION IN BIOMEDICAL ENGINEERING 2022. [DOI: 10.1016/j.cobme.2022.100410] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
|
12
|
Zhang J, Zhang M, Lin R, Du Y, Wang L, Yao Q, Zannettino A, Zhang H. Chondrogenic preconditioning of mesenchymal stem/stromal cells within a magnetic scaffold for osteochondral repair. Biofabrication 2022; 14. [PMID: 35226893 DOI: 10.1088/1758-5090/ac5935] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Accepted: 02/28/2022] [Indexed: 12/11/2022]
Abstract
Stem cell therapy using mesenchymal stromal/stem cells (MSCs) represents a novel approach to treating severe diseases, including osteoarthritis (OA). However, the therapeutic benefit of MSCs is highly dependent on their differentiation state, which can be regulated by many factors. Herein, three-dimensional (3D) magnetic scaffolds were successfully fabricated by incorporating magnetic nanoparticles (MNPs) into electrospun gelatin nanofibers. When positioned near a rotating magnet (f= 0.5 Hz), the magnetic scaffolds with the embedded MSCs were driven upward/downward in the culture container to induce mechanical stimulation to MSCs due to spatial confinement and fluid flow. The extracellular matrix-mimicking scaffold and the alternating magnetic field significantly enhanced chondrogenesis instead of osteogenesis. Furthermore, the fibre topography could be tuned with different compositions of the coating layer on MNPs, and the topography had a significant impact on MSC differentiation. Selective up-regulation of chondrogenesis-related genes (COL2A1andACAN) was found for the magnetic scaffolds with citric acid-coated MNPs (CAG). In contrast, osteogenesis-related genes (RUNX2andSPARC) were selectively and significantly up-regulated for the magnetic scaffolds with polyvinylpyrrolidone-coated MNPs (PVPG). Prior to implantation in vivo, chondrogenic preconditioning of MSCs within the CAG scaffolds under a dynamic magnetic field resulted in superior osteochondral repair. Hence, the magnetic scaffolds together with an in-house rotating magnet device could be a novel platform to initiate multiple stimuli on stem cell differentiation for effective repair of osteochondral defects.
Collapse
Affiliation(s)
- Jiabin Zhang
- Sun Yat-Sen University, Sun Yat-Sen University, Guangzhou, 510275, CHINA
| | - Ming Zhang
- Department of Orthopedics, Zhongda Hospital, School of Medicine, Southeast University, Department of Orthopedics, Zhongda Hospital, School of Medicine, Southeast University, Nanjing , Jiangsu Province, China, Nangjing, Jiangsu, 210009, CHINA
| | - Rongcai Lin
- Department of Orthopaedic Surgery, Institute of Digital Medicine, Nanjing First Hospital, Nanjing Medical University, Department of Orthopaedic Surgery, Institute of Digital Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing, China, Nanjing, 210006, CHINA
| | - Yuguang Du
- Institute of Process Engineering Chinese Academy of Sciences, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, China, Beijing, Beijing, 100190, CHINA
| | - Liming Wang
- Department of Orthopaedic Surgery Nanjing First Hospital, Nanjing Medical University, Department of Orthopaedic Surgery, Institute of Digital Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing, China, Nanjing, Jiangsu Province, 210006, CHINA
| | - Qingqiang Yao
- Department of Orthopaedic Surgery, Institute of Digital Medicine, Nanjing First Hospital, Nanjing Medical University, Department of Orthopaedic Surgery, Institute of Digital Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing, China, Nanjing, 210006, CHINA
| | - Andrew Zannettino
- Adelaide Medical School Research, The University of Adelaide, Adelaide, Australia, Adelaide, South Australia, 5005, AUSTRALIA
| | - Hu Zhang
- Amgen Bioprocessing Centre, Keck Graduate Institute, CA 91711, USA, 535 Watson Drive, Claremont, CA, USA, Claremont, California, 91711, UNITED STATES
| |
Collapse
|
13
|
Del Sol-Fernández S, Martínez-Vicente P, Gomollón-Zueco P, Castro-Hinojosa C, Gutiérrez L, Fratila RM, Moros M. Magnetogenetics: remote activation of cellular functions triggered by magnetic switches. NANOSCALE 2022; 14:2091-2118. [PMID: 35103278 PMCID: PMC8830762 DOI: 10.1039/d1nr06303k] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Accepted: 11/13/2021] [Indexed: 05/03/2023]
Abstract
During the last decade, the possibility to remotely control intracellular pathways using physical tools has opened the way to novel and exciting applications, both in basic research and clinical applications. Indeed, the use of physical and non-invasive stimuli such as light, electricity or magnetic fields offers the possibility of manipulating biological processes with spatial and temporal resolution in a remote fashion. The use of magnetic fields is especially appealing for in vivo applications because they can penetrate deep into tissues, as opposed to light. In combination with magnetic actuators they are emerging as a new instrument to precisely manipulate biological functions. This approach, coined as magnetogenetics, provides an exclusive tool to study how cells transform mechanical stimuli into biochemical signalling and offers the possibility of activating intracellular pathways connected to temperature-sensitive proteins. In this review we provide a critical overview of the recent developments in the field of magnetogenetics. We discuss general topics regarding the three main components for magnetic field-based actuation: the magnetic fields, the magnetic actuators and the cellular targets. We first introduce the main approaches in which the magnetic field can be used to manipulate the magnetic actuators, together with the most commonly used magnetic field configurations and the physicochemical parameters that can critically influence the magnetic properties of the actuators. Thereafter, we discuss relevant examples of magneto-mechanical and magneto-thermal stimulation, used to control stem cell fate, to activate neuronal functions, or to stimulate apoptotic pathways, among others. Finally, although magnetogenetics has raised high expectations from the research community, to date there are still many obstacles to be overcome in order for it to become a real alternative to optogenetics for instance. We discuss some controversial aspects related to the insufficient elucidation of the mechanisms of action of some magnetogenetics constructs and approaches, providing our opinion on important challenges in the field and possible directions for the upcoming years.
Collapse
Affiliation(s)
- Susel Del Sol-Fernández
- Instituto de Nanociencia y Materiales de Aragón (INMA), CSIC-Universidad de Zaragoza, Zaragoza 50009, Spain.
| | - Pablo Martínez-Vicente
- Instituto de Nanociencia y Materiales de Aragón (INMA), CSIC-Universidad de Zaragoza, Zaragoza 50009, Spain.
| | - Pilar Gomollón-Zueco
- Instituto de Nanociencia y Materiales de Aragón (INMA), CSIC-Universidad de Zaragoza, Zaragoza 50009, Spain.
| | - Christian Castro-Hinojosa
- Instituto de Nanociencia y Materiales de Aragón (INMA), CSIC-Universidad de Zaragoza, Zaragoza 50009, Spain.
| | - Lucía Gutiérrez
- Instituto de Nanociencia y Materiales de Aragón (INMA), CSIC-Universidad de Zaragoza, Zaragoza 50009, Spain.
- Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Spain
- Departamento de Química Analítica, Universidad de Zaragoza, Zaragoza 50009, Spain
| | - Raluca M Fratila
- Instituto de Nanociencia y Materiales de Aragón (INMA), CSIC-Universidad de Zaragoza, Zaragoza 50009, Spain.
- Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Spain
- Departamento de Química Orgánica, Universidad de Zaragoza, C/Pedro Cerbuna 12, Zaragoza 50009, Spain
| | - María Moros
- Instituto de Nanociencia y Materiales de Aragón (INMA), CSIC-Universidad de Zaragoza, Zaragoza 50009, Spain.
- Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Spain
| |
Collapse
|
14
|
Kush P, Kumar P, Singh R, Kaushik A. Aspects of high-performance and bio-acceptable magnetic nanoparticles for biomedical application. Asian J Pharm Sci 2021; 16:704-737. [PMID: 35027950 PMCID: PMC8737424 DOI: 10.1016/j.ajps.2021.05.005] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Revised: 05/01/2021] [Accepted: 05/22/2021] [Indexed: 12/11/2022] Open
Abstract
This review covers extensively the synthesis & surface modification, characterization, and application of magnetic nanoparticles. For biomedical applications, consideration should be given to factors such as design strategies, the synthesis process, coating, and surface passivation. The synthesis method regulates post-synthetic change and specific applications in vitro and in vivo imaging/diagnosis and pharmacotherapy/administration. Special insights have been provided on biodistribution, pharmacokinetics, and toxicity in a living system, which is imperative for their wider application in biology. These nanoparticles can be decorated with multiple contrast agents and thus can also be used as a probe for multi-mode imaging or double/triple imaging, for example, MRI-CT, MRI-PET. Similarly loading with different drug molecules/dye/fluorescent molecules and integration with other carriers have found application not only in locating these particles in vivo but simultaneously target drug delivery/hyperthermia inside the body. Studies are underway to collect the potential of these magnetically driven nanoparticles in various scientific fields such as particle interaction, heat conduction, imaging, and magnetism. Surely, this comprehensive data will help in the further development of advanced techniques for theranostics based on high-performance magnetic nanoparticles and will lead this research area in a new sustainable direction.
Collapse
Affiliation(s)
- Preeti Kush
- School of Pharmacy, Adarsh Vijendra Institute of Pharmaceutical Sciences, Shobhit University Gangoh, Saharanpur, Uttar Pradesh 247341, India
| | - Parveen Kumar
- Nanotechnology Division (H-1), CSIR-Central Scientific Instruments Organization, Chandigarh 160030, India
| | - Ranjit Singh
- School of Pharmacy, Adarsh Vijendra Institute of Pharmaceutical Sciences, Shobhit University Gangoh, Saharanpur, Uttar Pradesh 247341, India
| | - Ajeet Kaushik
- NanoBioTech Laboratory, Health System Engineering, Department of Natural Sciences, Florida Polytechnic University, Lakeland, FL 33805-8531, United States
| |
Collapse
|
15
|
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
|
16
|
Bonnet C, Brahmbhatt A, Deng SX, Zheng JJ. Wnt signaling activation: targets and therapeutic opportunities for stem cell therapy and regenerative medicine. RSC Chem Biol 2021; 2:1144-1157. [PMID: 34458828 PMCID: PMC8341040 DOI: 10.1039/d1cb00063b] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Accepted: 06/01/2021] [Indexed: 12/18/2022] Open
Abstract
Wnt proteins are secreted morphogens that play critical roles in embryonic development, stem cell proliferation, self-renewal, tissue regeneration and remodeling in adults. While aberrant Wnt signaling contributes to diseases such as cancer, activation of Wnt/β-catenin signaling is a target of interest in stem cell therapy and regenerative medicine. Recent high throughput screenings from chemical and biological libraries, combined with improved gene expression reporter assays of Wnt/β-catenin activation together with rational drug design, led to the development of a myriad of Wnt activators, with different mechanisms of actions. Among them, Wnt mimics, antibodies targeting Wnt inhibitors, glycogen-synthase-3β inhibitors, and indirubins and other natural product derivatives are emerging modalities to treat bone, neurodegenerative, eye, and metabolic disorders, as well as prevent ageing. Nevertheless, the creation of Wnt-based therapies has been hampered by challenges in developing potent and selective Wnt activators without off-target effects, such as oncogenesis. On the other hand, to avoid these risks, their use to promote ex vivo expansion during tissue engineering is a promising application.
Collapse
Affiliation(s)
- Clémence Bonnet
- Stein Eye Institute, University of California Los Angeles CA USA +1-3107947906 +1-3102062173
- INSERM, UMRS1138, Team 17, From Physiopathology of Ocular Diseases to Clinical Development, Paris University, Centre de Recherche des Cordeliers, and Cornea Departement, Cochin Hospital, AP-HP F-75014 Paris France
| | - Anvi Brahmbhatt
- Stein Eye Institute, University of California Los Angeles CA USA +1-3107947906 +1-3102062173
| | - Sophie X Deng
- Stein Eye Institute, University of California Los Angeles CA USA +1-3107947906 +1-3102062173
- Molecular Biology Institute, University of California Los Angeles CA USA
| | - Jie J Zheng
- Stein Eye Institute, University of California Los Angeles CA USA +1-3107947906 +1-3102062173
- Molecular Biology Institute, University of California Los Angeles CA USA
| |
Collapse
|
17
|
Markides H, Foster NC, McLaren JS, Hopkins T, Black C, Oreffo ROC, Scammell BE, Echevarria I, White LJ, El Haj AJ. Short-Term Evaluation of Cellular Fate in an Ovine Bone Formation Model. Cells 2021; 10:1776. [PMID: 34359945 PMCID: PMC8305225 DOI: 10.3390/cells10071776] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Revised: 07/08/2021] [Accepted: 07/09/2021] [Indexed: 02/07/2023] Open
Abstract
The ovine critical-sized defect model provides a robust preclinical model for testing tissue-engineered constructs for use in the treatment of non-union bone fractures and severe trauma. A critical question in cell-based therapies is understanding the optimal therapeutic cell dose. Key to defining the dose and ensuring successful outcomes is understanding the fate of implanted cells, e.g., viability, bio-distribution and exogenous infiltration post-implantation. This study evaluates such parameters in an ovine critical-sized defect model 2 and 7 days post-implantation. The fate of cell dose and behaviour post-implantation when combined with nanomedicine approaches for multi-model tracking and remote control using external magnetic fields is also addressed. Autologous STRO-4 selected mesenchymal stromal cells (MSCs) were labelled with a fluorescent lipophilic dye (CM-Dil), functionalised magnetic nanoparticles (MNPs) and delivered to the site within a naturally derived bone extracellular matrix (ECM) gel. Encapsulated cells were implanted within a critical-sized defect in an ovine medial femoral condyle and exposed to dynamic gradients of external magnetic fields for 1 h per day. Sheep were sacrificed at 2 and 7 days post-initial surgery where ECM was harvested. STRO-4-positive (STRO-4+) stromal cells expressed osteocalcin and survived within the harvested gels at day 2 and day 7 with a 50% loss at day 2 and a further 45% loss at 7 days. CD45-positive leucocytes were also observed in addition to endogenous stromal cells. No elevation in serum C-reactive protein (CRP) or non-haem iron levels was observed following implantation in groups containing MNPs with or without magnetic field gradients. The current study demonstrates how numbers of therapeutic cells reduce substantially after implantation in the repair site. Cell death is accompanied by enhanced leucocyte invasion, but not by inflammatory blood marker levels. Crucially, a proportion of implanted STRO-4+ stromal cells expressed osteocalcin, which is indicative of osteogenic differentiation. Furthermore, MNP labelling did not alter cell number or result in a further deleterious impact on stromal cells following implantation.
Collapse
Affiliation(s)
- Hareklea Markides
- Guy Hilton Research Centre, Institute of Science and Technology in Medicine, Keele University, Thornburrow Drive, Stoke-on-Trent ST4 7QB, UK; (H.M.); (T.H.); (I.E.)
- Healthcare Technologies Institute, Institute of Translational Medicine, School of Chemical Engineering, University of Birmingham, Birmingham B15 2TT, UK;
| | - Nicola C. Foster
- Healthcare Technologies Institute, Institute of Translational Medicine, School of Chemical Engineering, University of Birmingham, Birmingham B15 2TT, UK;
| | - Jane S. McLaren
- Centre for Biomolecular Sciences, University of Nottingham, Nottingham NG7 2RD, UK; (J.S.M.); (L.J.W.)
| | - Timothy Hopkins
- Guy Hilton Research Centre, Institute of Science and Technology in Medicine, Keele University, Thornburrow Drive, Stoke-on-Trent ST4 7QB, UK; (H.M.); (T.H.); (I.E.)
| | - Cameron Black
- Bone and Joint Research Group, Centre for Human Development, Stem Cells and Regeneration, Faculty of Medicine, University of Southampton, Southampton SO16 6YD, UK; (C.B.); (R.O.C.O.)
| | - Richard O. C. Oreffo
- Bone and Joint Research Group, Centre for Human Development, Stem Cells and Regeneration, Faculty of Medicine, University of Southampton, Southampton SO16 6YD, UK; (C.B.); (R.O.C.O.)
| | - Brigitte E. Scammell
- Academic Orthopaedics, Trauma and Sports Medicine, Queen’s Medical Centre, University of Nottingham, Nottingham NG7 2UH, UK;
| | - Iria Echevarria
- Guy Hilton Research Centre, Institute of Science and Technology in Medicine, Keele University, Thornburrow Drive, Stoke-on-Trent ST4 7QB, UK; (H.M.); (T.H.); (I.E.)
| | - Lisa J. White
- Centre for Biomolecular Sciences, University of Nottingham, Nottingham NG7 2RD, UK; (J.S.M.); (L.J.W.)
| | - Alicia J. El Haj
- Guy Hilton Research Centre, Institute of Science and Technology in Medicine, Keele University, Thornburrow Drive, Stoke-on-Trent ST4 7QB, UK; (H.M.); (T.H.); (I.E.)
- Healthcare Technologies Institute, Institute of Translational Medicine, School of Chemical Engineering, University of Birmingham, Birmingham B15 2TT, UK;
| |
Collapse
|
18
|
Almeida AF, Vinhas A, Gonçalves AI, Miranda MS, Rodrigues MT, Gomes ME. Magnetic triggers in biomedical applications - prospects for contact free cell sensing and guidance. J Mater Chem B 2021; 9:1259-1271. [PMID: 33410453 DOI: 10.1039/d0tb02474k] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
In recent years, the inputs from magnetically assisted strategies have been contributing to the development of more sensitive screening methods and precise means of diagnosis to overcome existing and emerging treatment challenges. The features of magnetic materials enabling in vivo traceability, specific targeting and space- and time-controlled delivery of nanomedicines have highlighted the resourcefulness of the magnetic toolbox for biomedical applications and theranostic strategies. The breakthroughs in magnetically assisted technologies for contact-free control of cell and tissue fate opens new perspectives to improve healing and instruct regeneration reaching a wide range of diseases and disorders. In this review, the contribution of magnetic nanoparticles (MNPs) will be explored as sophisticated and versatile nanotriggers, evidencing their unique cues to probe and control cell function. As cells detect and engage external magnetic features, these approaches will be overviewed considering molecular engineering and cell programming perspectives as well as cell and tissue targeting modalities. The therapeutic relevance of MNPs will be also emphasized as key components of nanostructured systems to control the release of nanomedicines and in the context of new therapy technologies.
Collapse
Affiliation(s)
- Ana F Almeida
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, 4805-017 Barco, Guimarães, Portugal. and ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Adriana Vinhas
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, 4805-017 Barco, Guimarães, Portugal. and ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Ana I Gonçalves
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, 4805-017 Barco, Guimarães, Portugal. and ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Margarida S Miranda
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, 4805-017 Barco, Guimarães, Portugal. and ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Márcia T Rodrigues
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, 4805-017 Barco, Guimarães, Portugal. and ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Manuela E Gomes
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, 4805-017 Barco, Guimarães, Portugal. and ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal
| |
Collapse
|
19
|
Fan D, Wang Q, Zhu T, Wang H, Liu B, Wang Y, Liu Z, Liu X, Fan D, Wang X. Recent Advances of Magnetic Nanomaterials in Bone Tissue Repair. Front Chem 2020; 8:745. [PMID: 33102429 PMCID: PMC7545026 DOI: 10.3389/fchem.2020.00745] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2020] [Accepted: 07/17/2020] [Indexed: 12/19/2022] Open
Abstract
The magnetic field has been proven to enhance bone tissue repair by affecting cell metabolic behavior. Magnetic nanoparticles are used as biomaterials due to their unique magnetic properties and good biocompatibility. Through endocytosis, entering the cell makes it easier to affect the physiological function of the cell. Once the magnetic particles are exposed to an external magnetic field, they will be rapidly magnetized. The magnetic particles and the magnetic field work together to enhance the effectiveness of their bone tissue repair treatment. This article reviews the common synthesis methods, the mechanism, and application of magnetic nanomaterials in the field of bone tissue repair.
Collapse
Affiliation(s)
- Daoyang Fan
- Department of Orthopedic, First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Qi Wang
- Department of Pediatrics, Peking University Third Hospital, Beijing, China
| | - Tengjiao Zhu
- Department of Orthopedic, Peking University Third Hospital, Beijing, China
| | - Hufei Wang
- Beijing National Laboratory for Molecular Sciences, State Key Laboratory of Polymer Physics & Chemistry, Institute of Chemistry, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Bingchuan Liu
- Department of Orthopedic, Peking University Third Hospital, Beijing, China
| | - Yifan Wang
- CED Education, North Carolina State University, Raleigh, NC, United States
| | - Zhongjun Liu
- Department of Orthopedic, Peking University Third Hospital, Beijing, China
| | - Xunyong Liu
- School of Chemistry and Materials Science, Ludong University, Yantai, China
| | - Dongwei Fan
- Department of Pediatrics, Peking University Third Hospital, Beijing, China
| | - Xing Wang
- Beijing National Laboratory for Molecular Sciences, State Key Laboratory of Polymer Physics & Chemistry, Institute of Chemistry, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
20
|
Matos AM, Gonçalves AI, Rodrigues MT, Miranda MS, Haj AJE, Reis RL, Gomes ME. Remote triggering of TGF-β/Smad2/3 signaling in human adipose stem cells laden on magnetic scaffolds synergistically promotes tenogenic commitment. Acta Biomater 2020; 113:488-500. [PMID: 32652226 DOI: 10.1016/j.actbio.2020.07.009] [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] [Received: 01/17/2020] [Revised: 07/01/2020] [Accepted: 07/02/2020] [Indexed: 12/29/2022]
Abstract
Injuries affecting load bearing tendon tissues are a significant clinical burden and efficient treatments are still unmet. Tackling tendon regeneration, tissue engineering strategies aim to develop functional substitutes that recreate native tendon milieu. Tendon mimetic scaffolds capable of remote magnetic responsiveness and functionalized magnetic nanoparticles (MNPs) targeting cellular mechanosensitive receptors are potential instructive tools to mediate mechanotransduction in guiding tenogenic responses. In this work, we combine magnetically responsive scaffolds and targeted Activin A type II receptor in human adipose stem cells (hASCs), under alternating magnetic field (AMF), to synergistically facilitate external control over signal transduction. The combination of remote triggering TGF-β/Smad2/3 using MNPs tagged hASCs, through magnetically actuated scaffolds, stimulates overall expression of tendon related genes and the deposition of tendon related proteins, in comparison to non-stimulated conditions. Moreover, the phosphorylation of Smad2/3 proteins and their nuclear co-localization was also more evident. Overall, biophysical stimuli resulting from magnetic scaffolds and magnetically triggered cells under AMF stimulation modulate the mechanosensing response of hASCs towards tenogenesis, holding therapeutic promise. STATEMENT OF SIGNIFICANCE: The concept of magnetically-assisted tissue engineering may assist the development of innovative solutions to treat tendon disorders upon remote control of biological processes as cell migration or differentiation. Herein, we originally combine a fibrous aligned superparamagnetic scaffold, based on a biodegradable polymeric blend of starch and poly-ɛ-caprolactone incorporating magnetic nanoparticles (MNPs), and human adipose stem cells (hASCs) labelled with MNPs functionalized with anti-activin receptor type IIA (ActRIIA). Constructs were stimulated using alternating magnetic field (AMF), to activate the ActRIIA and subsequent induction of TGF-β signaling, through Smad2/3 phosphorylation cascade, enhancing the expression of tendon-related markers. Altogether, these findings contribute with powerful bio-magnetic approaches to activate key tenogenic pathways, envisioning future translation of magnetic biomaterials into regenerative platforms for tendon repair.
Collapse
|
21
|
Rotherham M, Nahar T, Goodman T, Telling N, Gates M, El Haj A. Magnetic Mechanoactivation of Wnt Signaling Augments Dopaminergic Differentiation of Neuronal Cells. ACTA ACUST UNITED AC 2020; 3:e1900091. [PMID: 32648650 DOI: 10.1002/adbi.201900091] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2019] [Revised: 08/07/2019] [Indexed: 01/09/2023]
Abstract
Wnt signaling is a key developmental pathway that regulates dopaminergic progenitor cell proliferation and differentiation during neuronal development. This makes Wnt signaling an important therapeutic target for neurodegenerative conditions such as Parkinson's disease. Wnt signaling can be modulated using peptides such as UM206, which bind to the Wnt receptor Frizzled. Previous work has demonstrated remote activation of the Wnt pathway through Frizzled using peptide-functionalized magnetic nanoparticles (MNPs) with magnetic field stimulation. Using this technology, Wnt signaling is remotely activated in the neuronal cell line SH-SY5Y, and the phenotypic response to stimulation is assessed. Results indicate β-catenin translocalization and activation of TCF/LEF responsive transcription in response to MNP and magnetic fields, which result in dopaminergic marker expression when synergistically combined with differentiation factors retinoic acid and the phorbol ester phorbol 12-myristate 13-acetate. This approach is translated into ex vivo postnatal rat brain slices modeling the developing nigrostriatal pathway. Dopaminergic marker expression is maintained in MNP-labeled SH-SY5Y cells after injection and magnetic stimulation. These results demonstrate the translational value of remote control of signal transduction for controlling neuronal precursor cell behavior and highlight the potential applications for controlled cell differentiation as part of cell therapies for neurodegenerative disease.
Collapse
Affiliation(s)
- Michael Rotherham
- Institute for Science and Technology in Medicine, Keele University, Guy Hilton Research Centre, Thornburrow Drive, Stoke-on-Trent, ST4 7QB, UK
| | - Tasmin Nahar
- Institute for Science and Technology in Medicine, Keele University, Guy Hilton Research Centre, Thornburrow Drive, Stoke-on-Trent, ST4 7QB, UK
| | - Timothy Goodman
- Institute for Science and Technology in Medicine, Keele University, Guy Hilton Research Centre, Thornburrow Drive, Stoke-on-Trent, ST4 7QB, UK
| | - Neil Telling
- Institute for Science and Technology in Medicine, Keele University, Guy Hilton Research Centre, Thornburrow Drive, Stoke-on-Trent, ST4 7QB, UK
| | - Monte Gates
- Institute for Science and Technology in Medicine, Keele University, Guy Hilton Research Centre, Thornburrow Drive, Stoke-on-Trent, ST4 7QB, UK
| | - Alicia El Haj
- Institute for Science and Technology in Medicine, Keele University, Guy Hilton Research Centre, Thornburrow Drive, Stoke-on-Trent, ST4 7QB, UK.,Institute of Translational Medicine, University of Birmingham, Heritage Building, Mindelsohn Way, Edgbaston, Birmingham, B15 2TH, UK
| |
Collapse
|
22
|
Municoy S, Álvarez Echazú MI, Antezana PE, Galdopórpora JM, Olivetti C, Mebert AM, Foglia ML, Tuttolomondo MV, Alvarez GS, Hardy JG, Desimone MF. Stimuli-Responsive Materials for Tissue Engineering and Drug Delivery. Int J Mol Sci 2020; 21:E4724. [PMID: 32630690 PMCID: PMC7369929 DOI: 10.3390/ijms21134724] [Citation(s) in RCA: 97] [Impact Index Per Article: 19.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Revised: 06/28/2020] [Accepted: 06/30/2020] [Indexed: 02/07/2023] Open
Abstract
Smart or stimuli-responsive materials are an emerging class of materials used for tissue engineering and drug delivery. A variety of stimuli (including temperature, pH, redox-state, light, and magnet fields) are being investigated for their potential to change a material's properties, interactions, structure, and/or dimensions. The specificity of stimuli response, and ability to respond to endogenous cues inherently present in living systems provide possibilities to develop novel tissue engineering and drug delivery strategies (for example materials composed of stimuli responsive polymers that self-assemble or undergo phase transitions or morphology transformations). Herein, smart materials as controlled drug release vehicles for tissue engineering are described, highlighting their potential for the delivery of precise quantities of drugs at specific locations and times promoting the controlled repair or remodeling of tissues.
Collapse
Affiliation(s)
- Sofia Municoy
- Universidad de Buenos Aires, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Instituto de la Química y Metabolismo del Fármaco (IQUIMEFA), Facultad de Farmacia y Bioquímica Junín 956, Piso 3° (1113), Buenos Aires 1113, Argentina; (S.M.); (M.I.Á.E.); (P.E.A.); (J.M.G.); (C.O.); (A.M.M.); (M.L.F.); (M.V.T.); (G.S.A.)
| | - María I. Álvarez Echazú
- Universidad de Buenos Aires, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Instituto de la Química y Metabolismo del Fármaco (IQUIMEFA), Facultad de Farmacia y Bioquímica Junín 956, Piso 3° (1113), Buenos Aires 1113, Argentina; (S.M.); (M.I.Á.E.); (P.E.A.); (J.M.G.); (C.O.); (A.M.M.); (M.L.F.); (M.V.T.); (G.S.A.)
| | - Pablo E. Antezana
- Universidad de Buenos Aires, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Instituto de la Química y Metabolismo del Fármaco (IQUIMEFA), Facultad de Farmacia y Bioquímica Junín 956, Piso 3° (1113), Buenos Aires 1113, Argentina; (S.M.); (M.I.Á.E.); (P.E.A.); (J.M.G.); (C.O.); (A.M.M.); (M.L.F.); (M.V.T.); (G.S.A.)
| | - Juan M. Galdopórpora
- Universidad de Buenos Aires, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Instituto de la Química y Metabolismo del Fármaco (IQUIMEFA), Facultad de Farmacia y Bioquímica Junín 956, Piso 3° (1113), Buenos Aires 1113, Argentina; (S.M.); (M.I.Á.E.); (P.E.A.); (J.M.G.); (C.O.); (A.M.M.); (M.L.F.); (M.V.T.); (G.S.A.)
| | - Christian Olivetti
- Universidad de Buenos Aires, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Instituto de la Química y Metabolismo del Fármaco (IQUIMEFA), Facultad de Farmacia y Bioquímica Junín 956, Piso 3° (1113), Buenos Aires 1113, Argentina; (S.M.); (M.I.Á.E.); (P.E.A.); (J.M.G.); (C.O.); (A.M.M.); (M.L.F.); (M.V.T.); (G.S.A.)
| | - Andrea M. Mebert
- Universidad de Buenos Aires, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Instituto de la Química y Metabolismo del Fármaco (IQUIMEFA), Facultad de Farmacia y Bioquímica Junín 956, Piso 3° (1113), Buenos Aires 1113, Argentina; (S.M.); (M.I.Á.E.); (P.E.A.); (J.M.G.); (C.O.); (A.M.M.); (M.L.F.); (M.V.T.); (G.S.A.)
| | - María L. Foglia
- Universidad de Buenos Aires, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Instituto de la Química y Metabolismo del Fármaco (IQUIMEFA), Facultad de Farmacia y Bioquímica Junín 956, Piso 3° (1113), Buenos Aires 1113, Argentina; (S.M.); (M.I.Á.E.); (P.E.A.); (J.M.G.); (C.O.); (A.M.M.); (M.L.F.); (M.V.T.); (G.S.A.)
| | - María V. Tuttolomondo
- Universidad de Buenos Aires, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Instituto de la Química y Metabolismo del Fármaco (IQUIMEFA), Facultad de Farmacia y Bioquímica Junín 956, Piso 3° (1113), Buenos Aires 1113, Argentina; (S.M.); (M.I.Á.E.); (P.E.A.); (J.M.G.); (C.O.); (A.M.M.); (M.L.F.); (M.V.T.); (G.S.A.)
| | - Gisela S. Alvarez
- Universidad de Buenos Aires, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Instituto de la Química y Metabolismo del Fármaco (IQUIMEFA), Facultad de Farmacia y Bioquímica Junín 956, Piso 3° (1113), Buenos Aires 1113, Argentina; (S.M.); (M.I.Á.E.); (P.E.A.); (J.M.G.); (C.O.); (A.M.M.); (M.L.F.); (M.V.T.); (G.S.A.)
| | - John G. Hardy
- Department of Chemistry, Faraday Building, Lancaster University, Lancaster, Lancashire LA1 4YB, UK
- Materials Science Institute, Faraday Building, Lancaster University, Lancaster, Lancashire LA1 4YB, UK
| | - Martin F. Desimone
- Universidad de Buenos Aires, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Instituto de la Química y Metabolismo del Fármaco (IQUIMEFA), Facultad de Farmacia y Bioquímica Junín 956, Piso 3° (1113), Buenos Aires 1113, Argentina; (S.M.); (M.I.Á.E.); (P.E.A.); (J.M.G.); (C.O.); (A.M.M.); (M.L.F.); (M.V.T.); (G.S.A.)
| |
Collapse
|
23
|
Abdel Fattah AR, Ranga A. Nanoparticles as Versatile Tools for Mechanotransduction in Tissues and Organoids. Front Bioeng Biotechnol 2020; 8:240. [PMID: 32363177 PMCID: PMC7180186 DOI: 10.3389/fbioe.2020.00240] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Accepted: 03/09/2020] [Indexed: 12/28/2022] Open
Abstract
Organoids are 3D multicellular constructs that rely on self-organized cell differentiation, patterning and morphogenesis to recapitulate key features of the form and function of tissues and organs of interest. Dynamic changes in these systems are orchestrated by biochemical and mechanical microenvironments, which can be engineered and manipulated to probe their role in developmental and disease mechanisms. In particular, the in vitro investigation of mechanical cues has been the focus of recent research, where mechanical manipulations imparting local as well as large-scale mechanical stresses aim to mimic in vivo tissue deformations which occur through proliferation, folding, invagination, and elongation. However, current in vitro approaches largely impose homogeneous mechanical changes via a host matrix and lack the required positional and directional specificity to mimic the diversity of in vivo scenarios. Thus, while organoids exhibit limited aspects of in vivo morphogenetic events, how local forces are coordinated to enable large-scale changes in tissue architecture remains a difficult question to address using current techniques. Nanoparticles, through their efficient internalization by cells and dispersion through extracellular matrices, have the ability to provide local or global, as well as passive or active modulation of mechanical stresses on organoids and tissues. In this review, we explore how nanoparticles can be used to manipulate matrix and tissue mechanics, and highlight their potential as tools for fate regulation through mechanotransduction in multicellular model systems.
Collapse
Affiliation(s)
- Abdel Rahman Abdel Fattah
- Laboratory of Bioengineering and Morphogenesis, Department of Mechanical Engineering, KU Leuven, Leuven, Belgium
| | - Adrian Ranga
- Laboratory of Bioengineering and Morphogenesis, Department of Mechanical Engineering, KU Leuven, Leuven, Belgium
| |
Collapse
|
24
|
Zare EN, Jamaledin R, Naserzadeh P, Afjeh-Dana E, Ashtari B, Hosseinzadeh M, Vecchione R, Wu A, Tay FR, Borzacchiello A, Makvandi P. Metal-Based Nanostructures/PLGA Nanocomposites: Antimicrobial Activity, Cytotoxicity, and Their Biomedical Applications. ACS APPLIED MATERIALS & INTERFACES 2020; 12:3279-3300. [PMID: 31873003 DOI: 10.1021/acsami.9b19435] [Citation(s) in RCA: 87] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/20/2023]
Abstract
Among the different synthetic polymers developed for biomedical applications, poly(lactic-co-glycolic acid) (PLGA) has attracted considerable attention because of its excellent biocompatibility and biodegradability. Nanocomposites based on PLGA and metal-based nanostructures (MNSs) have been employed extensively as an efficient strategy to improve the structural and functional properties of PLGA polymer. The MNSs have been used to impart new properties to PLGA, such as antimicrobial properties and labeling. In the present review, the different strategies available for the fabrication of MNS/PLGA nanocomposites and their applications in the biomedical field will be discussed, beginning with a description of the preparation routes, antimicrobial activity, and cytotoxicity concerns of MNS/PLGA nanocomposites. The biomedical applications of these nanocomposites, such as carriers and scaffolds in tissue regeneration and other therapies are subsequently reviewed. In addition, the potential advantages of using MNS/PLGA nanocomposites in treatment illnesses are analyzed based on in vitro and in vivo studies, to support the potential of these nanocomposites in future research in the biomedical field.
Collapse
Affiliation(s)
| | - Rezvan Jamaledin
- Center for Advanced Biomaterials for Health Care , Istituto Italiano di Tecnologia , Naples 80125 , Italy
- Department of Chemical, Materials and Industrial Production Engineering , University of Naples Federico II , Naples 80125 , Italy
| | - Parvaneh Naserzadeh
- Shahdad Ronak Commercialization Company (SPE No 10320821698) , Pasdaran Street , Tehran 1947 , Iran
- Nanomedicine and Tissue Engineering Research Center , Shahid Beheshti University of Medical Sciences , Tehran 1985717443 , Iran
| | - Elham Afjeh-Dana
- Shahdad Ronak Commercialization Company (SPE No 10320821698) , Pasdaran Street , Tehran 1947 , Iran
- Radiation Biology Research Center , Iran University of Medical Sciences , Tehran 14496-14535 , Iran
| | - Behnaz Ashtari
- Radiation Biology Research Center , Iran University of Medical Sciences , Tehran 14496-14535 , Iran
- Department of Medical Nanotechnology, Faculty of Advanced Technologies in Medicine , Iran University of Medical Sciences , Tehran 14496-14535 , Iran
| | - Mehdi Hosseinzadeh
- Health Management and Economics Research Center , Iran University of Medical Sciences , Tehran 14496-14535 , Iran
- Computer Science , University of Human Development , Sulaymaniyah , Iraq
| | - Raffaele Vecchione
- Center for Advanced Biomaterials for Health Care , Istituto Italiano di Tecnologia , Naples 80125 , Italy
| | - Aimin Wu
- Department of Orthopedics, Bioprinting Research Group, Zhejiang Provincial Key Laboratory of Orthopedics , The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University , Wenzhou 325035 , China
| | - Franklin R Tay
- College of Graduate Studies , Augusta University , Augusta , Georgia 30912 , United States
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi Key Laboratory of Stomatology, Department of Prosthodontics, School of Stomatology , The Fourth Military Medical University , Xi'an , Shaanxi , China
| | - Assunta Borzacchiello
- Institute for Polymers, Composites, and Biomaterials (IPCB) , National Research Council (CNR) , Naples 80125 , Italy
| | - Pooyan Makvandi
- Department of Medical Nanotechnology, Faculty of Advanced Technologies in Medicine , Iran University of Medical Sciences , Tehran 14496-14535 , Iran
- Institute for Polymers, Composites, and Biomaterials (IPCB) , National Research Council (CNR) , Naples 80125 , Italy
| |
Collapse
|
25
|
Matos AM, Gonçalves AI, El Haj AJ, Gomes ME. Magnetic biomaterials and nano-instructive tools as mediators of tendon mechanotransduction. NANOSCALE ADVANCES 2020; 2:140-148. [PMID: 36133967 PMCID: PMC9417540 DOI: 10.1039/c9na00615j] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Accepted: 11/29/2019] [Indexed: 05/29/2023]
Abstract
Tendon tissues connect muscle to bone allowing the transmission of forces resulting in joint movement. Tendon injuries are prevalent in society and the impact on public health is of utmost concern. Thus, clinical options for tendon treatments are in demand, and tissue engineering aims to provide reliable and successful long-term regenerative solutions. Moreover, the possibility of regulating cell fate by triggering intracellular pathways is a current challenge in regenerative medicine. In the last decade, the use of magnetic nanoparticles as nano-instructive tools has led to great advances in diagnostics and therapeutics. Recent advances using magnetic nanomaterials for regenerative medicine applications include the incorporation of magnetic biomaterials within 3D scaffolds resulting in mechanoresponsive systems with unprecedented properties and the use of nanomagnetic actuators to control cell signaling. Mechano-responsive scaffolds and nanomagnetic systems can act as mechanostimulation platforms to apply forces directly to single cells and multicellular biological tissues. As transmitters of forces in a localized manner, the approaches enable the downstream activation of key tenogenic signaling pathways. In this minireview, we provide a brief outlook on the tenogenic signaling pathways which are most associated with the conversion of mechanical input into biochemical signals, the novel bio-magnetic approaches which can activate these pathways, and the efforts to translate magnetic biomaterials into regenerative platforms for tendon repair.
Collapse
Affiliation(s)
- Ana M Matos
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine Avepark - Zona Industrial da Gandra, 4805-017 Barco Guimarães Portugal
- ICVS/3B's - PT Government Associate Laboratory Braga/Guimarães Portugal
| | - Ana I Gonçalves
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine Avepark - Zona Industrial da Gandra, 4805-017 Barco Guimarães Portugal
- ICVS/3B's - PT Government Associate Laboratory Braga/Guimarães Portugal
| | - Alicia J El Haj
- Healthcare Technologies Institute, Birmingham University B15 2TT Birmingham UK
| | - Manuela E Gomes
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine Avepark - Zona Industrial da Gandra, 4805-017 Barco Guimarães Portugal
- ICVS/3B's - PT Government Associate Laboratory Braga/Guimarães Portugal
- The Discoveries Centre for Regenerative and Precision Medicine, Headquarters at the University of Minho Avepark, 4805-017 Barco Guimarães Portugal
| |
Collapse
|
26
|
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
|
27
|
Li J, Wang L, Tian J, Zhou Z, Li J, Yang H. Nongenetic engineering strategies for regulating receptor oligomerization in living cells. Chem Soc Rev 2020; 49:1545-1568. [DOI: 10.1039/c9cs00473d] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Nongenetic strategies for regulating receptor oligomerization in living cells based on DNA, protein, small molecules and physical stimuli.
Collapse
Affiliation(s)
- Jingying Li
- MOE Key Laboratory for Analytical Science of Food Safety and Biology
- Fujian Provincial Key Laboratory of Analysis and Detection Technology for Food Safety
- State Key Laboratory of Photocatalysis on Energy and Environment
- College of Chemistry
- Fuzhou University
| | - Liping Wang
- MOE Key Laboratory for Analytical Science of Food Safety and Biology
- Fujian Provincial Key Laboratory of Analysis and Detection Technology for Food Safety
- State Key Laboratory of Photocatalysis on Energy and Environment
- College of Chemistry
- Fuzhou University
| | - Jinmiao Tian
- Institute of Molecular Medicine
- Renji Hospital
- School of Medicine
- Shanghai Jiao Tong University
- Shanghai
| | - Zhilan Zhou
- Institute of Molecular Medicine
- Renji Hospital
- School of Medicine
- Shanghai Jiao Tong University
- Shanghai
| | - Juan Li
- MOE Key Laboratory for Analytical Science of Food Safety and Biology
- Fujian Provincial Key Laboratory of Analysis and Detection Technology for Food Safety
- State Key Laboratory of Photocatalysis on Energy and Environment
- College of Chemistry
- Fuzhou University
| | - Huanghao Yang
- MOE Key Laboratory for Analytical Science of Food Safety and Biology
- Fujian Provincial Key Laboratory of Analysis and Detection Technology for Food Safety
- State Key Laboratory of Photocatalysis on Energy and Environment
- College of Chemistry
- Fuzhou University
| |
Collapse
|
28
|
Marshall KM, Kanczler JM, Oreffo ROC. Evolving applications of the egg: chorioallantoic membrane assay and ex vivo organotypic culture of materials for bone tissue engineering. J Tissue Eng 2020; 11:2041731420942734. [PMID: 33194169 PMCID: PMC7594486 DOI: 10.1177/2041731420942734] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Accepted: 06/26/2020] [Indexed: 01/03/2023] Open
Abstract
The chick chorioallantoic membrane model has been around for over a century, applied in angiogenic, oncology, dental and xenograft research. Despite its often perceived archaic, redolent history, the chorioallantoic membrane assay offers new and exciting opportunities for material and growth factor evaluation in bone tissue engineering. Currently, superior/improved experimental methodology for the chorioallantoic membrane assay are difficult to identify, given an absence of scientific consensus in defining experimental approaches, including timing of inoculation with materials and the analysis of results. In addition, critically, regulatory and welfare issues impact upon experimental designs. Given such disparate points, this review details recent research using the ex vivo chorioallantoic membrane assay and the ex vivo organotypic culture to advance the field of bone tissue engineering, and highlights potential areas of improvement for their application based on recent developments within our group and the tissue engineering field.
Collapse
Affiliation(s)
- Karen M Marshall
- Bone and Joint Research Group, Centre for Human
Development, Stem Cells and Regeneration, Institute of Developmental Sciences,
University of Southampton, Southampton, UK
| | - Janos M Kanczler
- Bone and Joint Research Group, Centre for Human
Development, Stem Cells and Regeneration, Institute of Developmental Sciences,
University of Southampton, Southampton, UK
| | - Richard OC Oreffo
- Bone and Joint Research Group, Centre for Human
Development, Stem Cells and Regeneration, Institute of Developmental Sciences,
University of Southampton, Southampton, UK
| |
Collapse
|
29
|
Li J, Duan H, Pu K. Nanotransducers for Near-Infrared Photoregulation in Biomedicine. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2019; 31:e1901607. [PMID: 31199021 DOI: 10.1002/adma.201901607] [Citation(s) in RCA: 98] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/13/2019] [Revised: 04/05/2019] [Indexed: 06/09/2023]
Abstract
Photoregulation, which utilizes light to remotely control biological events, provides a precise way to decipher biology and innovate in medicine; however, its potential is limited by the shallow tissue penetration and/or phototoxicity of ultraviolet (UV)/visible light that are required to match the optical responses of endogenous photosensitive substances. Thereby, biologically friendly near-infrared (NIR) light with improved tissue penetration is desired for photoregulation. Since there are a few endogenous biomolecules absorbing or emitting light in the NIR region, the development of molecular transducers is essential to convert NIR light into the cues for regulation of biological events. In this regard, optical nanomaterials able to convert NIR light into UV/visible light, heat, or free radicals are suitable for this task. Here, the recent developments of optical nanotransducers for NIR-light-mediated photoregulation in medicine are summarized. The emerging applications, including photoregulation of neural activity, gene expression, and visual systems, as well as photochemical tissue bonding, are highlighted, along with the design principles of nanotransducers. Moreover, the current challenges and perspectives in this field are discussed.
Collapse
Affiliation(s)
- Jingchao Li
- School of Chemical and Biomedical Engineering, Nanyang Technological University, Singapore, 637457, Singapore
| | - Hongwei Duan
- School of Chemical and Biomedical Engineering, Nanyang Technological University, Singapore, 637457, Singapore
| | - Kanyi Pu
- School of Chemical and Biomedical Engineering, Nanyang Technological University, Singapore, 637457, Singapore
| |
Collapse
|
30
|
Stanley SA, Friedman JM. Electromagnetic Regulation of Cell Activity. Cold Spring Harb Perspect Med 2019; 9:cshperspect.a034322. [PMID: 30249601 DOI: 10.1101/cshperspect.a034322] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The ability to observe the effects of rapidly and reversibly regulating cell activity in targeted cell populations has provided numerous physiologic insights. Over the last decade, a wide range of technologies have emerged for regulating cellular activity using optical, chemical, and, more recently, electromagnetic modalities. Electromagnetic fields can freely penetrate cells and tissue and their energy can be absorbed by metal particles. When released, the absorbed energy can in turn gate endogenous or engineered receptors and ion channels to regulate cell activity. In this manner, electromagnetic fields acting on external nanoparticles have been used to exert mechanical forces on cell membranes and organelles to generate heat and interact with thermally activated proteins or to induce receptor aggregation and intracellular signaling. More recently, technologies using genetically encoded nanoparticles composed of the iron storage protein, ferritin, have been used for targeted, temporal control of cell activity in vitro and in vivo. These tools provide a means for noninvasively modulating gene expression, intracellular organelles, such as endosomes, and whole-cell activity both in vitro and in freely moving animals. The use of magnetic fields interacting with external or genetically encoded nanoparticles thus provides a rapid noninvasive means for regulating cell activity.
Collapse
Affiliation(s)
- Sarah A Stanley
- Diabetes, Obesity and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, New York 10029.,Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York 10029
| | - Jeffrey M Friedman
- Laboratory of Molecular Genetics, Rockefeller University, New York, New York 10065.,Howard Hughes Medical Institute, New York, New York 10065
| |
Collapse
|
31
|
Scialla S, Barca A, Palazzo B, D'Amora U, Russo T, Gloria A, De Santis R, Verri T, Sannino A, Ambrosio L, Gervaso F. Bioactive chitosan‐based scaffolds with improved properties induced by dextran‐grafted nano‐maghemite and
l
‐arginine amino acid. J Biomed Mater Res A 2019; 107:1244-1252. [DOI: 10.1002/jbm.a.36633] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Revised: 12/14/2018] [Accepted: 12/26/2018] [Indexed: 01/13/2023]
Affiliation(s)
- Stefania Scialla
- Department of Engineering for InnovationUniversity of Salento Lecce Italy
| | - Amilcare Barca
- General Physiology Laboratories, Department of Biological and Environmental Sciences and TechnologiesUniversity of Salento Lecce Italy
| | - Barbara Palazzo
- Department of Engineering for InnovationUniversity of Salento Lecce Italy
- Ghimas S.p.A., c/o Dhitech Scarl, Campus Ecotekne Lecce Italy
| | - Ugo D'Amora
- Institute of Polymers, Composites and BiomaterialsNational Research Council Naples Italy
| | - Teresa Russo
- Institute of Polymers, Composites and BiomaterialsNational Research Council Naples Italy
| | - Antonio Gloria
- Institute of Polymers, Composites and BiomaterialsNational Research Council Naples Italy
| | - Roberto De Santis
- Institute of Polymers, Composites and BiomaterialsNational Research Council Naples Italy
| | - Tiziano Verri
- General Physiology Laboratories, Department of Biological and Environmental Sciences and TechnologiesUniversity of Salento Lecce Italy
| | - Alessandro Sannino
- Department of Engineering for InnovationUniversity of Salento Lecce Italy
| | - Luigi Ambrosio
- Institute of Polymers, Composites and BiomaterialsNational Research Council Naples Italy
| | - Francesca Gervaso
- Department of Engineering for InnovationUniversity of Salento Lecce Italy
| |
Collapse
|
32
|
Moise S, Byrne JM, El Haj AJ, Telling ND. The potential of magnetic hyperthermia for triggering the differentiation of cancer cells. NANOSCALE 2018; 10:20519-20525. [PMID: 30397703 DOI: 10.1039/c8nr05946b] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/20/2023]
Abstract
Magnetic hyperthermia is a potential technique for cancer therapy that exploits heat generated by magnetic nanoparticles to kill cancerous cells. Many studies have shown that magnetic hyperthermia is effective at killing cancer cells both in vitro and in vivo, however little attention has been paid to the cellular functioning of the surviving cells. We report here new evidence demonstrating the onset of thermally triggered differentiation in osteosarcoma cancer cells that survive magnetic hyperthermia treatment. This raises the possibility that in addition to causing cell death, magnetic hyperthermia could induce surviving cancer cells to form more mature cell types and thereby inhibit their capacity to self-renew. Such processes could prove to be as important as cell death when considering magnetic hyperthermia for treating cancer.
Collapse
Affiliation(s)
- Sandhya Moise
- Department of Chemical Engineering, University of Bath, Bath BA2 7AY, UK.
| | | | | | | |
Collapse
|
33
|
Mechanoactivation of Wnt/β-catenin pathways in health and disease. Emerg Top Life Sci 2018; 2:701-712. [DOI: 10.1042/etls20180042] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Revised: 10/08/2018] [Accepted: 10/09/2018] [Indexed: 11/17/2022]
Abstract
Mechanical forces play an important role in regulating tissue development and homeostasis in multiple cell types including bone, joint, epithelial and vascular cells, and are also implicated in the development of diseases, e.g. osteoporosis, cardiovascular disease and osteoarthritis. Defining the mechanisms by which cells sense and respond to mechanical forces therefore has important implications for our understanding of tissue function in health and disease and may lead to the identification of targets for therapeutic intervention. Mechanoactivation of the Wnt signalling pathway was first identified in osteoblasts with a key role for β-catenin demonstrated in loading-induced osteogenesis. Since then, mechanoregulation of the Wnt pathway has also been observed in stem cells, epithelium, chondrocytes and vascular and lymphatic endothelium. Wnt can signal through both canonical and non-canonical pathways, and evidence suggests that both can mediate responses to mechanical strain, stretch and shear stress. This review will discuss our current understanding of the activation of the Wnt pathway in response to mechanical forces.
Collapse
|
34
|
Henstock JR, Rotherham M, El Haj AJ. Magnetic ion channel activation of TREK1 in human mesenchymal stem cells using nanoparticles promotes osteogenesis in surrounding cells. J Tissue Eng 2018; 9:2041731418808695. [PMID: 30397432 PMCID: PMC6207961 DOI: 10.1177/2041731418808695] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2018] [Accepted: 09/28/2018] [Indexed: 12/15/2022] Open
Abstract
Magnetic ion channel activation technology uses superparamagnetic nanoparticles conjugated with targeting antibodies to apply mechanical force directly to stretch-activated ion channels on the cell surface, stimulating mechanotransduction and downstream processes. This technique has been reported to promote differentiation towards musculoskeletal cell types and enhance mineralisation. Previous studies have shown how mesenchymal stem cells injected into a pre-mineralised environment such as a foetal chick epiphysis, results in large-scale osteogenesis at the target site. However, the relative contributions of stem cells and surrounding host tissue has not been resolved, that is, are the mesenchymal stem cells solely responsible for the observed mineralisation or do mechanically stimulated mesenchymal stem cells also promote a host-tissue mineralisation response? To address this, we established a novel two-dimensional co-culture assay, which indicated that magnetic ion channel activation stimulation of human mesenchymal stem cells does not significantly promote migration but does enhance collagen deposition and mineralisation in the surrounding cells. We conclude that one of the important functions of injected human mesenchymal stem cells is to release biological factors (e.g., cytokines and microvesicles) which guide the surrounding tissue response, and that remote control of this signalling process using magnetic ion channel activation technology may be a useful way to both drive and regulate tissue regeneration and healing.
Collapse
Affiliation(s)
- James R Henstock
- Institute of Ageing and Chronic Disease, University of Liverpool, Liverpool, UK
- James R Henstock, Institute of Ageing and Chronic Disease, University of Liverpool, William Henry Duncan Building, Liverpool L7 8TX, UK.
| | - Michael Rotherham
- Institute of Science and Technology in Medicine, Guy Hilton Research Centre, Keele University, Staffordshire, UK
| | - Alicia J El Haj
- Institute of Science and Technology in Medicine, Guy Hilton Research Centre, Keele University, Staffordshire, UK
| |
Collapse
|
35
|
Xia Y, Sun J, Zhao L, Zhang F, Liang XJ, Guo Y, Weir MD, Reynolds MA, Gu N, Xu HHK. Magnetic field and nano-scaffolds with stem cells to enhance bone regeneration. Biomaterials 2018; 183:151-170. [PMID: 30170257 DOI: 10.1016/j.biomaterials.2018.08.040] [Citation(s) in RCA: 167] [Impact Index Per Article: 23.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2018] [Revised: 08/10/2018] [Accepted: 08/20/2018] [Indexed: 12/20/2022]
Abstract
Novel strategies utilizing magnetic nanoparticles (MNPs) and magnetic fields are being developed to enhance bone tissue engineering efficacy. This article first reviewed cutting-edge research on the osteogenic enhancements via magnetic fields and MNPs. Then the current developments in magnetic strategies to improve the cells, scaffolds and growth factor deliveries were described. The magnetic-cell strategies included cell labeling, targeting, patterning, and gene modifications. MNPs were incorporated to fabricate magnetic composite scaffolds, as well as to construct delivery systems for growth factors, drugs and gene transfections. The novel methods using magnetic nanoparticles and scaffolds with magnetic fields and stem cells increased the osteogenic differentiation, angiogenesis and bone regeneration by 2-3 folds over those of the controls. The mechanisms of magnetic nanoparticles and scaffolds with magnetic fields and stem cells to enhance bone regeneration were identified as involving the activation of signaling pathways including MAPK, integrin, BMP and NF-κB. Potential clinical applications of magnetic nanoparticles and scaffolds with magnetic fields and stem cells include dental, craniofacial and orthopedic treatments with substantially increased bone repair and regeneration efficacy.
Collapse
Affiliation(s)
- Yang Xia
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, Jiangsu 210029, China; Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering, Southeast University, Nanjing, Jiangsu 210096, China; Department of Advanced Oral Sciences & Therapeutics, University of Maryland School of Dentistry, Baltimore, MD 21201, USA
| | - Jianfei Sun
- Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering, Southeast University, Nanjing, Jiangsu 210096, China
| | - Liang Zhao
- Department of Advanced Oral Sciences & Therapeutics, University of Maryland School of Dentistry, Baltimore, MD 21201, USA; Department of Orthopedic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Feimin Zhang
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, Jiangsu 210029, China; Collaborative Innovation Center of Suzhou Nano Science and Technology, Suzhou, Jiangsu 215123, China
| | - Xing-Jie Liang
- Chinese Academy of Sciences (CAS) Center for Excellence in Nanoscience, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology, Beijing 100190, China
| | - Yu Guo
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Michael D Weir
- Department of Advanced Oral Sciences & Therapeutics, University of Maryland School of Dentistry, Baltimore, MD 21201, USA
| | - Mark A Reynolds
- Department of Advanced Oral Sciences & Therapeutics, University of Maryland School of Dentistry, Baltimore, MD 21201, USA
| | - Ning Gu
- Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering, Southeast University, Nanjing, Jiangsu 210096, China; Collaborative Innovation Center of Suzhou Nano Science and Technology, Suzhou, Jiangsu 215123, China.
| | - Hockin H K Xu
- Department of Advanced Oral Sciences & Therapeutics, University of Maryland School of Dentistry, Baltimore, MD 21201, USA; Center for Stem Cell Biology and Regenerative Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, USA; University of Maryland Marlene and Stewart Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore, MD 21201, USA.
| |
Collapse
|
36
|
Gonçalves AI, Miranda MS, Rodrigues MT, Reis RL, Gomes ME. Magnetic responsive cell-based strategies for diagnostics and therapeutics. Biomed Mater 2018; 13:054001. [DOI: 10.1088/1748-605x/aac78b] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
|
37
|
Li X, Yang J, Bao M, Zeng K, Fu S, Wang C, Ye L. Wnt signaling in bone metastasis: mechanisms and therapeutic opportunities. Life Sci 2018; 208:33-45. [PMID: 29969609 DOI: 10.1016/j.lfs.2018.06.036] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2018] [Revised: 06/29/2018] [Accepted: 06/29/2018] [Indexed: 02/05/2023]
Abstract
Bone metastasis frequently occurs in advanced cancer patients, who will develop osteogenic/osteolytic bone lesions in the late stage of the disease. Wnt signaling pathway, which is mainly grouped into the β-catenin dependent pathway and β-catenin independent pathway, is a well-organized cascade that has been reported to play important roles in a variety of physiological and pathological conditions, including bone metastasis. Regulation of Wnt signaling in bone metastasis involves multiple stages, including dissemination of primary tumor cells to bone, dormancy and outgrowth of metastatic tumor cells, and tumor-induced osteogenic and osteolytic bone destruction, suggesting the importance of Wnt signaling in bone metastasis pathology. In this review, we will introduce the involvement of Wnt signaling components in specific bone metastasis stages and summarize the promising Wnt modulators that have shown potential as bone metastasis therapeutics, in the hope to maximize the therapeutic opportunities of Wnt signaling for bone metastasis.
Collapse
Affiliation(s)
- Xin Li
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Jing Yang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China; Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Minyue Bao
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Kan Zeng
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China; Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Shijin Fu
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China; Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Chenglin Wang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China; Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Ling Ye
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China; Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China.
| |
Collapse
|
38
|
Triggering the activation of Activin A type II receptor in human adipose stem cells towards tenogenic commitment using mechanomagnetic stimulation. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2018; 14:1149-1159. [PMID: 29471171 DOI: 10.1016/j.nano.2018.02.008] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/27/2017] [Revised: 01/16/2018] [Accepted: 02/10/2018] [Indexed: 12/21/2022]
Abstract
Stem cell therapies hold potential to stimulate tendon regeneration and homeostasis, which is maintained in response to the native mechanical environment. Activins are members of the mechano-responsive TGF-β superfamily that participates in the regulation of several downstream biological processes. Mechanosensitive membrane receptors such as activin can be activated in different types of stem cells via magnetic nanoparticles (MNPs) through remote magnetic actuation resulting in cell differentiation. In this work, we target the Activin receptor type IIA (ActRIIA) in human adipose stem cells (hASCs), using anti-ActRIIA functionalized MNPs, externally activated through a oscillating magnetic bioreactor. Upon activation, the phosphorylation of Smad2/3 is induced allowing translocation of the complex to the nucleus, regulating tenogenic transcriptional responses. Our study demonstrates the potential remote activation of MNPs tagged hASCs to trigger the Activin receptor leading to tenogenic differentiation. These results may provide insights toward tendon regeneration therapies.
Collapse
|