Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Int J Mol Sci ; 24(4)2023 Feb 08.
Article in English | MEDLINE | ID: mdl-36834776

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.


Subject(s)
Integrins , Ion Channels , Humans , HEK293 Cells , Magnetics , Magnetic Phenomena
2.
Cells ; 10(7)2021 07 14.
Article in English | MEDLINE | ID: mdl-34359945

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.


Subject(s)
Bone Marrow Cells/cytology , Cell Differentiation/physiology , Mesenchymal Stem Cells/metabolism , Osteogenesis/physiology , Animals , Bone and Bones/cytology , Sheep , Stromal Cells/cytology
3.
Sci Rep ; 10(1): 11171, 2020 Jul 02.
Article in English | MEDLINE | ID: mdl-32612189

ABSTRACT

An amendment to this paper has been published and can be accessed via a link at the top of the paper.

4.
Sci Rep ; 10(1): 8451, 2020 05 21.
Article in English | MEDLINE | ID: mdl-32439838

ABSTRACT

Multipotent Mesenchymal Stem/Stromal Cells (MSCs) are widely used in cellular therapy for joint repair. However, the use of MSC therapies is complicated by a lack of understanding of the behaviour of cells and repair within the joint. Current methods of MSC tracking include labelling the cells with Super Paramagnetic Iron Oxide nanoparticles (SPIOs). However, standard acquisition sequences (T2 and T2*) give poor anatomical definition in the presence of SPIOs. To avoid anatomical compromise in the presence of SPIOs, we have investigated the use of Ultra-short Echo Time (UTE) MRI, using a 3D cones acquisition trajectory. This method was used to track SPIO labelled MSC injected into joints containing osteochondral defects in experimental sheep. This study demonstrates that multiple echo times from UTE with 3 T MRI can provide excellent anatomical detail of osteochondral defects and demonstrate similar features to histology. This work also monitors the location of SPIO-labelled cells for regenerative medicine of the knee with MRI, histology, and Prussian blue staining. With these methods, we show that the SPIOs do not hone to the site of defect but instead aggregate in the location of injection, which suggests that any repair mechanism with this disease model must trigger a secondary process.


Subject(s)
Iron/chemistry , Knee Injuries/veterinary , Magnetic Resonance Imaging/methods , Mesenchymal Stem Cell Transplantation/methods , Mesenchymal Stem Cells/cytology , Nanoparticles/chemistry , Sheep Diseases/therapy , Animals , Disease Models, Animal , Female , Knee Injuries/pathology , Sheep
5.
Stem Cell Res Ther ; 10(1): 25, 2019 01 11.
Article in English | MEDLINE | ID: mdl-30635066

ABSTRACT

BACKGROUND: Osteochondral injuries represent a significant clinical problem requiring novel cell-based therapies to restore function of the damaged joint with the use of mesenchymal stromal cells (MSCs) leading research efforts. Pre-clinical studies are fundamental in translating such therapies; however, technologies to minimally invasively assess in vivo cell fate are currently limited. We investigate the potential of a MRI- (magnetic resonance imaging) and superparamagnetic iron oxide nanoparticle (SPION)-based technique to monitor cellular bio-distribution in an ovine osteochondral model of acute and chronic injuries. METHODS: MSCs were isolated, expanded and labelled with Nanomag, a 250-nm SPION, and using a novel cell-penetrating technique, glycosaminoglycan-binding enhanced transduction (GET). MRI visibility thresholds, cellular toxicity and differentiation potential post-labelling were assessed in vitro. A single osteochondral defect was created in the medial femoral condyle in the left knee joint of each sheep with the contralateral joint serving as the control. Cells, either GET-Nanomag labelled or unlabelled, were delivered 1 week or 4.5 weeks later. Sheep were sacrificed 7 days post implantation and immediately MR imaged using a 0.2-T MRI scanner and validated on a 3-T MRI scanner prior to histological evaluation. RESULTS: MRI data demonstrated a significant increase in MRI contrast as a result of GET-Nanomag labelling whilst cell viability, proliferation and differentiation capabilities were not affected. MRI results revealed evidence of implanted cells within the synovial joint of the injured leg of the chronic model only with no signs of cell localisation to the defect site in either model. This was validated histologically determining the location of implanted cells in the synovium. Evidence of engulfment of Nanomag-labelled cells by leukocytes is observed in the injured legs of the chronic model only. Finally, serum c-reactive protein (CRP) levels were measured by ELISA with no obvious increase in CRP levels observed as a result of P21-8R:Nanomag delivery. CONCLUSION: This study has the potential to be a powerful translational tool with great implications in the clinical translation of stem cell-based therapies. Further, we have demonstrated the ability to obtain information linked to key biological events occurring post implantation, essential in designing therapies and selecting pre-clinical models.


Subject(s)
Cell Tracking , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/cytology , Animals , Bone Marrow Cells/cytology , Bone Marrow Cells/drug effects , Cell Differentiation/drug effects , Cell Survival/drug effects , Ferric Compounds/pharmacology , Knee Joint/diagnostic imaging , Knee Joint/physiopathology , Magnetic Resonance Imaging , Mesenchymal Stem Cells/drug effects , Sheep , Synovial Membrane/cytology , Synovial Membrane/transplantation
6.
NPJ Regen Med ; 3: 9, 2018.
Article in English | MEDLINE | ID: mdl-29675269

ABSTRACT

The role of biomechanical stimuli, or mechanotransduction, in normal bone homeostasis and repair is understood to facilitate effective osteogenesis of mesenchymal stem cells (MSCs) in vitro. Mechanotransduction has been integrated into a multitude of in vitro bone tissue engineering strategies and provides an effective means of controlling cell behaviour towards therapeutic outcomes. However, the delivery of mechanical stimuli to exogenous MSC populations, post implantation, poses a significant translational hurdle. Here, we describe an innovative bio-magnetic strategy, MICA, where magnetic nanoparticles (MNPs) are used to remotely deliver mechanical stimuli to the mechano-receptor, TREK-1, resulting in activation and downstream signalling via an external magnetic array. In these studies, we have translated MICA to a pre-clinical ovine model of bone injury to evaluate functional bone repair. We describe the development of a magnetic array capable of in vivo MNP manipulation and subsequent osteogenesis at equivalent field strengths in vitro. We further demonstrate that the viability of MICA-activated MSCs in vivo is unaffected 48 h post implantation. We present evidence to support early accelerated repair and preliminary enhanced bone growth in MICA-activated defects within individuals compared to internal controls. The variability in donor responses to MICA-activation was evaluated in vitro revealing that donors with poor osteogenic potential were most improved by MICA-activation. Our results demonstrate a clear relationship between responders to MICA in vitro and in vivo. These unique experiments offer exciting clinical applications for cell-based therapies as a practical in vivo source of dynamic loading, in real-time, in the absence of pharmacological agents.

7.
Stem Cells Int ; 2017: 2905104, 2017.
Article in English | MEDLINE | ID: mdl-29434641

ABSTRACT

BACKGROUND: Mesenchymal stem cells (MSCs) have a therapeutic potential for the treatment of osteoarthritic (OA) joint pathology and pain. The aims of this study were to determine the influence of a passage number on the effects of MSCs on pain behaviour and cartilage and bone features in a rodent model of OA. METHODS: Rats underwent either medial meniscal transection (MNX) or sham surgery under anaesthesia. Rats received intra-articular injection of either 1.5 × 106 late passage MSCs labelled with 10 µg/ml SiMAG, 1.5 × 106 late passage mesenchymal stem cells, the steroid Kenalog (200 µg/20 µL), 1.5 × 106 early passage MSCs, or serum-free media (SFM). Sham-operated rats received intra-articular injection of SFM. Pain behaviour was quantified until day 42 postmodel induction. Magnetic resonance imaging (MRI) was used to localise the labelled cells within the knee joint. RESULTS: Late passage MSCs and Kenalog attenuated established pain behaviour in MNX rats, but did not alter MNX-induced joint pathology at the end of the study period. Early passage MSCs exacerbated MNX-induced pain behaviour for up to one week postinjection and did not alter joint pathology. CONCLUSION: Our data demonstrate for the first time the role of a passage number in influencing the therapeutic effects of MSCs in a model of OA pain.

8.
J Tissue Eng Regen Med ; 11(8): 2333-2348, 2017 08.
Article in English | MEDLINE | ID: mdl-27151571

ABSTRACT

Mesenchymal stem cells (MSCs) represent a valuable resource for regenerative medicine treatments for orthopaedic repair and beyond. Following developments in isolation, expansion and differentiation protocols, efforts to promote clinical translation of emerging cellular strategies now seek to improve cell delivery and targeting. This study shows efficient live MSC labelling using silica-coated magnetic particles (MPs), which enables 3D tracking and guidance of stem cells. A procedure developed for the efficient and unassisted particle uptake was shown to support MSC viability and integrity, while surface marker expression and MSC differentiation capability were also maintained. In vitro, MSCs showed a progressive decrease in labelling over increasing culture time, which appeared to be linked to the dilution effect of cell division, rather than to particle release, and did not lead to detectable secondary particle uptake. Labelled MSC populations demonstrated magnetic responsiveness in vitro through directed migration in culture and, when seeded onto a scaffold, supporting MP-based approaches to cell targeting. The potential of these silica-coated MPs for MRI cell tracking of MSC populations was validated in 2D and in a cartilage repair model following cell delivery. These results highlight silica-coated magnetic particles as a simple, safe and effective resource to enhance MSC targeting for therapeutic applications and improve patient outcomes. © 2016 The Authors Journal of Tissue Engineering and Regenerative Medicine Published by John Wiley & Sons Ltd.


Subject(s)
Cell Tracking/methods , Cell- and Tissue-Based Therapy , Coated Materials, Biocompatible , Magnetic Fields , Mesenchymal Stem Cells , Silicon Dioxide , Staining and Labeling/methods , Antigens, Differentiation/biosynthesis , Cell Differentiation/drug effects , Coated Materials, Biocompatible/chemistry , Coated Materials, Biocompatible/pharmacology , Gene Expression Regulation/drug effects , Humans , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Silicon Dioxide/chemistry , Silicon Dioxide/pharmacology
9.
Proc Natl Acad Sci U S A ; 113(3): E291-9, 2016 Jan 19.
Article in English | MEDLINE | ID: mdl-26733682

ABSTRACT

Protein transduction domains (PTDs) are powerful nongenetic tools that allow intracellular delivery of conjugated cargoes to modify cell behavior. Their use in biomedicine has been hampered by inefficient delivery to nuclear and cytoplasmic targets. Here we overcame this deficiency by developing a series of novel fusion proteins that couple a membrane-docking peptide to heparan sulfate glycosaminoglycans (GAGs) with a PTD. We showed that this GET (GAG-binding enhanced transduction) system could deliver enzymes (Cre, neomycin phosphotransferase), transcription factors (NANOG, MYOD), antibodies, native proteins (cytochrome C), magnetic nanoparticles (MNPs), and nucleic acids [plasmid (p)DNA, modified (mod)RNA, and small inhibitory RNA] at efficiencies of up to two orders of magnitude higher than previously reported in cell types considered hard to transduce, such as mouse embryonic stem cells (mESCs), human ESCs (hESCs), and induced pluripotent stem cells (hiPSCs). This technology represents an efficient strategy for controlling cell labeling and directing cell fate or behavior that has broad applicability for basic research, disease modeling, and clinical application.


Subject(s)
Cell-Penetrating Peptides/metabolism , Drug Delivery Systems , Glycosaminoglycans/metabolism , Amino Acid Motifs , Animals , Cell Differentiation/drug effects , Cell Membrane/drug effects , Cell Membrane/metabolism , Cell Proliferation/drug effects , Cell-Penetrating Peptides/chemistry , Detergents/pharmacology , Endocytosis/drug effects , Genome , Homeodomain Proteins/metabolism , Human Embryonic Stem Cells/cytology , Human Embryonic Stem Cells/drug effects , Human Embryonic Stem Cells/metabolism , Humans , Induced Pluripotent Stem Cells/drug effects , Induced Pluripotent Stem Cells/metabolism , Integrases/metabolism , Mice , Mouse Embryonic Stem Cells/cytology , Mouse Embryonic Stem Cells/drug effects , Mouse Embryonic Stem Cells/metabolism , Muscle Development/drug effects , MyoD Protein/metabolism , NIH 3T3 Cells , Nanog Homeobox Protein , Nanoparticles , Nucleic Acids/metabolism , Protein Structure, Tertiary , Solubility , Trypsin/metabolism
10.
Int J Biochem Cell Biol ; 69: 162-72, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26482595

ABSTRACT

Despite major medical advances, non-union bone fractures and skeletal defects continue to place significant burden on the patient, the clinicians and the healthcare system as a whole. Current bone substitute approaches are still limited in effectiveness and to date no adequate bone substitute material has been developed for routine clinical application. Tissue engineering presents a novel approach to tackling this clinical burden and developing an acceptable solution for the treatment of skeletal defects. Over the past three decades the field has evolved to appreciate the key biological, material and physical parameters influencing the development of a cell-based tissue engineered therapy and to create associated technologies to exploit such parameters. In recent years a number of therapies have started progressing along the pre-clinical pipeline to build a case for regulatory approval and ultimately clinical adoption. However, little emphasis has been given to the translational challenges faced when moving from "bench-to-bedside". One particular challenge lies in the delivery of functional mechanical stimuli to implanted cell populations to activate and promote osteogenic activities. This review introduces novel bio-magnetic approaches to overcoming this challenge.


Subject(s)
Bone and Bones/physiology , Fractures, Bone/therapy , Animals , Biomechanical Phenomena , Bone Regeneration , Bone Substitutes/therapeutic use , Fracture Healing , Humans , Tissue Engineering , Translational Research, Biomedical
11.
Stem Cell Res Ther ; 4(5): 126, 2013 Oct 17.
Article in English | MEDLINE | ID: mdl-24406201

ABSTRACT

INTRODUCTION: The application of mesenchymal stem cells (MSCs) in treating rheumatoid arthritis (RA) has been made possible by the immunosuppressive and differentiation abilities of these cells. A non-invasive means of assessing cell integration and bio-distribution is fundamental in evaluating the risks and success of this therapy, thereby enabling clinical translation. This paper defines the use of superparamagnetic iron oxide nanoparticles (SPIONs) in conjunction with magnetic resonance imaging (MRI) to image and track MSCs in vivo within a murine model of RA. METHODS: Murine MSCs (mMSCs) were isolated, expanded and labelled with SiMAG, a commercially available particle. In vitro MRI visibility thresholds were investigated by labelling mMSCs with SiMAG with concentrations ranging from 0 to 10 µg/ml and resuspending varying cell doses (10930 to 5 × 10950 cells) in 2 mg/ml collagen prior to MR-imaging. Similarly, in vivo detection thresholds were identified by implanting 3 × 10950 mMSCs labelled with 0 to 10 µg/ml SiMAG within the synovial cavity of a mouse and MR-imaging. Upon RA induction, 300,000 mMSCs labelled with SiMAG (10 µg/ml) were implanted via intra-articular injection and joint swelling monitored as an indication of RA development over seven days. Furthermore, the effect of SiMAG on cell viability, proliferation and differentiation was investigated. RESULTS: A minimum particle concentration of 1 µg/ml (300,000 cells) and cell dose of 100,000 cells (5 and 10 µg/ml) were identified as the in vitro MRI detection threshold. Cell viability, proliferation and differentiation capabilities were not affected, with labelled populations undergoing successful differentiation down osteogenic and adipogenic lineages. A significant decrease (P < 0.01) in joint swelling was measured in groups containing SiMAG-labelled and unlabelled mMSCs implying that the presence of SPIONs does not affect the immunomodulating properties of the cells. In vivo MRI scans demonstrated good contrast and the identification of SiMAG-labelled populations within the synovial joint up to 7 days post implantation. This was further confirmed using histological analysis. CONCLUSIONS: We have been able to monitor and track the migration of stem cell populations within the rheumatic joint in a non-invasive manner. This manuscript goes further to highlight the key characteristics (biocompatible and the ability to create significant contrast at realistic doses within a clinical relevant system) demonstrated by SiMAG that should be incorporated into the design of a new clinically approved tracking agent.


Subject(s)
Arthritis, Rheumatoid/diagnostic imaging , Dextrans/chemistry , Magnetite Nanoparticles/chemistry , Mesenchymal Stem Cells/chemistry , Mesenchymal Stem Cells/cytology , Whole Body Imaging , Animals , Arthritis, Rheumatoid/pathology , Arthritis, Rheumatoid/surgery , Bone Marrow Cells/cytology , Cell Differentiation , Cell Proliferation , Cell Survival , Cells, Cultured , Disease Models, Animal , Fluorescent Dyes/chemistry , Magnetic Resonance Imaging , Mesenchymal Stem Cell Transplantation , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Radiography
12.
Stem Cell Res Ther ; 3(2): 13, 2012 Apr 19.
Article in English | MEDLINE | ID: mdl-22520594

ABSTRACT

Stem cells have tremendous applications in the field of regenerative medicine and tissue engineering. These are pioneering fields that aim to create new treatments for disease that currently have limited therapies or cures. A particularly popular avenue of research has been the regeneration of bone and cartilage to combat various orthopaedic diseases. Magnetic nanoparticles (MNPs) have been applied to aid the development and translation of these therapies from research to the clinic. This review highlights contemporary research for the applications of iron-oxide-based MNPs for the therapeutic implementation of stem cells in orthopaedics. These MNPs comprise of an iron oxide core, coated with a choice of biological polymers that can facilitate the uptake of MNPs by cells through improving endocytic activity. The combined use of these oxides and the biological polymer coatings meet biological requirements, effectively encouraging the use of MNPs in regenerative medicine. The association of MNPs with stem cells can be achieved via the process of endocytosis resulting in the internalisation of these particles or the attachment to cell surface receptors. This allows for the investigation of migratory patterns through various tracking studies, the targeting of particle-labelled cells to desired locations via the application of an external magnetic field and, finally, for activation stem cells to initiate various cellular responses to induce the differentiation. Characterisation of cell localisation and associated tissue regeneration can therefore be enhanced, particularly for in vivo applications. MNPs have been shown to have the potential to stimulate differentiation of stem cells for orthopaedic applications, without limiting proliferation. However, careful consideration of the use of active agents associated with the MNP is suggested, for differentiation towards specific lineages. This review aims to broaden the knowledge of current applications, paving the way to translate the in vitro and in vivo work into further orthopaedic clinical studies.


Subject(s)
Bone Diseases/therapy , Cell Differentiation , Magnetite Nanoparticles/therapeutic use , Stem Cells/cytology , Translational Research, Biomedical , Cartilage/growth & development , Cartilage/transplantation , Cell Lineage , Cell Proliferation , Endocytosis , Ferric Compounds/chemistry , Humans , Magnetite Nanoparticles/chemistry , Regenerative Medicine/methods
SELECTION OF CITATIONS
SEARCH DETAIL
...