Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
Add more filters










Publication year range
1.
ACS Appl Mater Interfaces ; 16(26): 32930-32944, 2024 Jul 03.
Article in English | MEDLINE | ID: mdl-38888932

ABSTRACT

Protein materials are versatile tools in diverse biomedical fields. Among them, artificial secretory granules (SGs), mimicking those from the endocrine system, act as mechanically stable reservoirs for the sustained release of proteins as oligomeric functional nanoparticles. Only validated in oncology, the physicochemical properties of SGs, along with their combined drug-releasing and scaffolding abilities, make them suitable as smart topographies in regenerative medicine for the prolonged delivery of growth factors (GFs). Thus, considering the need for novel, safe, and cost-effective materials to present GFs, in this study, we aimed to biofabricate a protein platform combining both endocrine-like and extracellular matrix fibronectin-derived (ECM-FN) systems. This approach is based on the sustained delivery of a nanostructured histidine-tagged version of human fibroblast growth factor 2. The GF is presented onto polymeric surfaces, interacting with FN to spontaneously generate nanonetworks that absorb and present the GF in the solid state, to modulate mesenchymal stromal cell (MSC) behavior. The results show that SGs-based topographies trigger high rates of MSCs proliferation while preventing differentiation. While this could be useful in cell therapy manufacture demanding large numbers of unspecialized MSCs, it fully validates the hybrid platform as a convenient setup for the design of biologically active hybrid surfaces and in tissue engineering for the controlled manipulation of mammalian cell growth.


Subject(s)
Extracellular Matrix , Fibronectins , Mesenchymal Stem Cells , Humans , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/drug effects , Extracellular Matrix/chemistry , Extracellular Matrix/metabolism , Fibronectins/chemistry , Fibroblast Growth Factor 2/chemistry , Fibroblast Growth Factor 2/pharmacology , Cell Proliferation/drug effects , Cell Differentiation/drug effects , Nanostructures/chemistry
2.
Biomater Adv ; 158: 213766, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38232578

ABSTRACT

Medical implant-associated infections pose a significant challenge to modern medicine, with aseptic loosening and bacterial infiltration being the primary causes of implant failure. While nanostructured surfaces have demonstrated promising antibacterial properties, the translation of their efficacy from 2D to 3D substrates remains a challenge. Here, we used scalable alkaline etching to fabricate nanospike and nanonetwork topologies on 2D and laser powder-bed fusion printed 3D titanium. The fabricated surfaces were compared with regard to their antibacterial properties against Staphylococcus aureus, Escherichia coli, and Pseudomonas aeruginosa, and mesenchymal stromal cell responses with and without the presence of bacteria. Finite elemental analysis assessed the mechanical properties and permeability of the 3D substrate. Our findings suggest that 3D nanostructured surfaces have potential to both prevent implant infections and allow host cell integration. This work represents a significant step towards developing effective and scalable fabrication methods on 3D substrates with consistent and reproducible antibacterial activity, with important implications for the future of medical implant technology.


Subject(s)
Bacterial Adhesion , Titanium , Titanium/pharmacology , Coculture Techniques , Surface Properties , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Bacteria
3.
Sci Rep ; 9(1): 12944, 2019 09 10.
Article in English | MEDLINE | ID: mdl-31506561

ABSTRACT

In regenerative medicine, techniques which control stem cell lineage commitment are a rapidly expanding field of interest. Recently, nanoscale mechanical stimulation of mesenchymal stem cells (MSCs) has been shown to activate mechanotransduction pathways stimulating osteogenesis in 2D and 3D culture. This has the potential to revolutionise bone graft procedures by creating cellular graft material from autologous or allogeneic sources of MSCs without using chemical induction. With the increased interest in mechanical stimulation of cells and huge potential for clinical use, it is apparent that researchers and clinicians require a scalable bioreactor system that provides consistently reproducible results with a simple turnkey approach. A novel bioreactor system is presented that consists of: a bioreactor vibration plate, calibrated and optimised for nanometre vibrations at 1 kHz, a power supply unit, which supplies a 1 kHz sine wave signal necessary to generate approximately 30 nm of vibration amplitude, and custom 6-well cultureware with toroidal shaped magnets incorporated in the base of each well for conformal attachment to the bioreactor's magnetic vibration plate. The cultureware and vibration plate were designed using finite element analysis to determine the modal and harmonic responses, and validated by interferometric measurement. This helps ensure that the vibration plate and cultureware, and thus collagen and MSCs, all move as a rigid body, avoiding large deformations close to the resonant frequency of the vibration plate and vibration damping beyond the resonance. Assessment of osteogenic protein expression was performed to confirm differentiation of MSCs after initial biological experiments with the system, as well as atomic force microscopy of the 3D gel constructs during vibrational stimulation to verify that strain hardening of the gel did not occur. This shows that cell differentiation was the result of the nanovibrational stimulation provided by the bioreactor alone, and that other cell differentiating factors, such as stiffening of the collagen gel, did not contribute.


Subject(s)
Bioreactors , Cell Culture Techniques/methods , Cell Differentiation , Mechanotransduction, Cellular , Mesenchymal Stem Cells/cytology , Osteogenesis , Tissue Engineering/methods , Cells, Cultured , Equipment Design , Humans
4.
J Tissue Eng ; 9: 2041731418794007, 2018.
Article in English | MEDLINE | ID: mdl-30202512

ABSTRACT

The regenerative potential of skeletal stem cells provides an attractive prospect to generate bone tissue needed for musculoskeletal reparation. A central issue remains efficacious, controlled cell differentiation strategies to aid progression of cell therapies to the clinic. The nacre surface from Pinctada maxima shells is known to enhance bone formation. However, to date, there is a paucity of information on the role of the topography of P. maxima surfaces, nacre and prism. To investigate this, nacre and prism topographical features were replicated onto polycaprolactone and skeletal stem cell behaviour on the surfaces studied. Skeletal stem cells on nacre surfaces exhibited an increase in cell area, increase in expression of osteogenic markers ALP (p < 0.05) and OCN (p < 0.01) and increased metabolite intensity (p < 0.05), indicating a role of nacre surface to induce osteogenic differentiation, while on prism surfaces, skeletal stem cells did not show alterations in cell area or osteogenic marker expression and a decrease in metabolite intensity (p < 0.05), demonstrating a distinct role for the prism surface, with the potential to maintain the skeletal stem cell phenotype.

5.
J Tissue Eng ; 9: 2041731418790694, 2018.
Article in English | MEDLINE | ID: mdl-30116518

ABSTRACT

Titanium (Ti) plays a predominant role as the material of choice in orthopaedic and dental implants. Despite the majority of Ti implants having long-term success, premature failure due to unsuccessful osseointegration leading to aseptic loosening is still too common. Recently, surface topography modification and biological/non-biological coatings have been integrated into orthopaedic/dental implants in order to mimic the surrounding biological environment as well as reduce the inflammation/infection that may occur. In this review, we summarize the impact of various Ti coatings on cell behaviour both in vivo and in vitro. First, we focus on the Ti surface properties and their effects on osteogenesis and then on bacterial adhesion and viability. We conclude from the current literature that surface modification of Ti implants can be generated that offer both osteoinductive and antimicrobial properties.

6.
J Tissue Eng ; 9: 2041731418789838, 2018.
Article in English | MEDLINE | ID: mdl-30083308

ABSTRACT

The use of biomaterials in orthopaedics for joint replacement, fracture healing and bone regeneration is a rapidly expanding field. Infection of these biomaterials is a major healthcare burden, leading to significant morbidity and mortality. Furthermore, the cost to healthcare systems is increasing dramatically. With advances in implant design and production, research has predominately focussed on osseointegration; however, modification of implant material, surface topography and chemistry can also provide antibacterial activity. With the increasing burden of infection, it is vitally important that we consider the bacterial interaction with the biomaterial and the host when designing and manufacturing future implants. During this review, we will elucidate the interaction between patient, biomaterial surface and bacteria. We aim to review current and developing surface modifications with a view towards antibacterial orthopaedic implants for clinical applications.

7.
J Tissue Eng ; 9: 2041731418784098, 2018.
Article in English | MEDLINE | ID: mdl-30034770

ABSTRACT

The ability to influence stem cell differentiation is highly desirable as it would help us improve clinical outcomes for patients in various aspects. Many different techniques to achieve this have previously been investigated. This concise study, however, has focused on the topography on which cells grow. Current uncemented orthopaedic implants can fail if the implant fails to bind to the surrounding bone and, typically, forms a soft tissue interface which reduces direct bone contact. Here, we look at the effect of a previously reported nanotopography that utilises nanodisorder to influence mesenchymal stromal cell (as may be found in the bone marrow) differentiation towards bone and to also exert this effect on mature osteoblasts (as may be found in the bone). As topography is a physical technique, it can be envisaged for use in a range of materials such as polymers and metals used in the manufacture of orthopaedic implants.

8.
Oncogene ; 37(29): 3998-4012, 2018 07.
Article in English | MEDLINE | ID: mdl-29691476

ABSTRACT

Epstein-Barr virus (EBV)-associated Burkitt's lymphoma is characterised by the deregulation of c-Myc expression and a restricted viral gene expression pattern in which the EBV nuclear antigen-1 (EBNA1) is the only viral protein to be consistently expressed. EBNA1 is required for viral genome propagation and segregation during latency. However, it has been much debated whether the protein plays a role in viral-associated tumourigenesis. We show that the lymphomas which arise in EµEBNA1 transgenic mice are unequivocally linked to EBNA1 expression and that both C-Myc and Mdm2 deregulation are central to this process. Tumour cell survival is supported by IL-2 and there is a skew towards CD8-positive T cells in the tumour environment, while the immune check-point protein PD-L1 is upregulated in the tumours. Additionally, several isoforms of Mdm2 are upregulated in the EµEBNA1 tumours, with increased phosphorylation at ser166, an expression pattern not seen in Eµc-Myc transgenic tumours. Concomitantly, E2F1, Xiap, Mta1, C-Fos and Stat1 are upregulated in the tumours. Using four independent inhibitors of Mdm2 we demonstrate that the EµEBNA1 tumour cells are dependant upon Mdm2 for survival (as they are upon c-Myc) and that Mdm2 inhibition is not accompanied by upregulation of p53, instead cell death is linked to loss of E2F1 expression, providing new insight into the underlying tumourigenic mechanism. This opens a new path to combat EBV-associated disease.


Subject(s)
Epstein-Barr Virus Nuclear Antigens/metabolism , Lymphoma/virology , Proto-Oncogene Proteins c-mdm2/metabolism , Animals , B7-H1 Antigen/metabolism , CD8-Positive T-Lymphocytes/metabolism , Cell Death/physiology , Cell Line , E2F1 Transcription Factor/metabolism , Epstein-Barr Virus Infections/metabolism , Epstein-Barr Virus Infections/virology , Gene Expression Regulation, Viral/physiology , Herpesvirus 4, Human/metabolism , Humans , Interleukin-2/metabolism , Lymphoma/metabolism , Mice, Inbred C57BL , Mice, Transgenic , Proto-Oncogene Proteins c-myc/metabolism , Tumor Suppressor Protein p53/metabolism , Up-Regulation/physiology
9.
Sci Rep ; 7(1): 16363, 2017 11 27.
Article in English | MEDLINE | ID: mdl-29180787

ABSTRACT

Engineering the interface between biomaterials and tissues is important to increase implant lifetime and avoid failures and revision surgeries. Permanent devices should enhance attachment and differentiation of stem cells, responsible for injured tissue repair, and simultaneously discourage bacterial colonization; this represents a major challenge. To take first steps towards such a multifunctional surface we propose merging topographical and biochemical cues on the surface of a clinically relevant material such as titanium. In detail, our strategy combines antibacterial nanotopographical features with integrin selective synthetic ligands that can rescue the adhesive capacity of the surfaces and instruct mesenchymal stem cell (MSC) response. To this end, a smooth substrate and two different high aspect ratio topographies have been produced and coated either with an αvß3-selective peptidomimetic, an α5ß1-selective peptidomimetic, or an RGD/PHSRN peptidic molecule. Results showed that antibacterial effects of the substrates could be maintained when tested on pathogenic Pseudomonas aeruginosa. Further, functionalization increased MSC adhesion to the surfaces and the αvß3-selective peptidomimetic-coated nanotopographies promoted osteogenesis. Such a dual physicochemical approach to achieve multifunctional surfaces represents a first step in the design of novel cell-instructive biomaterial surfaces.


Subject(s)
Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/pharmacology , Biocompatible Materials/chemistry , Biocompatible Materials/pharmacology , Integrins/chemistry , Anti-Bacterial Agents/metabolism , Bacterial Adhesion , Biocompatible Materials/metabolism , Cell Differentiation , Cells, Cultured , Coated Materials, Biocompatible , Humans , Integrins/metabolism , Ligands , Mesenchymal Stem Cells/cytology , Microbial Viability , Osteogenesis/drug effects , Osteogenesis/genetics , Surface Properties , Titanium/chemistry
10.
Acta Biomater ; 60: 220-231, 2017 09 15.
Article in English | MEDLINE | ID: mdl-28754648

ABSTRACT

Despite microsurgical repair, recovery of function following peripheral nerve injury is slow and often incomplete. Outcomes could be improved by an increased understanding of the molecular biology of regeneration and by translation of experimental bioengineering strategies. Topographical cues have been shown to be powerful regulators of the rate and directionality of neurite regeneration, and in this study we investigated the downstream molecular effects of linear micropatterned structures in an organotypic explant model. Linear topographical cues enhanced neurite outgrowth and our results demonstrated that the mTOR pathway is important in regulating these responses. mTOR gene expression peaked between 48 and 72h, coincident with the onset of rapid neurite outgrowth and glial migration, and correlated with neurite length at 48h. mTOR protein was located to glia and in a punctate distribution along neurites. mTOR levels peaked at 72h and were significantly increased by patterned topography (p<0.05). Furthermore, the topographical cues could override pharmacological inhibition. Downstream phosphorylation assays and inhibition of mTORC1 using rapamycin highlighted mTORC2 as an important mediator, and more specific therapeutic target. Quantitative immunohistochemistry confirmed the presence of the mTORC2 component rictor at the regenerating front where it co-localised with F-actin and vinculin. Collectively, these results provide a deeper understanding of the mechanism of action of topography on neural regeneration, and support the incorporation of topographical patterning in combination with pharmacological mTORC2 potentiation within biomaterial constructs used to repair peripheral nerves. STATEMENT OF SIGNIFICANCE: Peripheral nerve injury is common and functionally devastating. Despite microsurgical repair, healing is slow and incomplete, with lasting functional deficit. There is a clear need to translate bioengineering approaches and increase our knowledge of the molecular processes controlling nerve regeneration to improve the rate and success of healing. Topographical cues are powerful determinants of neurite outgrowth and represent a highly translatable engineering strategy. Here we demonstrate, for the first time, that microtopography potentiates neurite outgrowth via the mTOR pathway, with the mTORC2 subtype being of particular importance. These results give further evidence for the incorporation of microtopographical cues into peripheral nerve regeneration conduits and indicate that mTORC2 may be a suitable therapeutic target to potentiate nerve regeneration.


Subject(s)
Gene Expression Regulation , Mechanistic Target of Rapamycin Complex 2/biosynthesis , Nerve Regeneration , Peripheral Nerve Injuries/metabolism , Peripheral Nerves/physiology , TOR Serine-Threonine Kinases/biosynthesis , Animals , Disease Models, Animal , Peripheral Nerve Injuries/pathology , Peripheral Nerves/pathology , Rats , Rats, Sprague-Dawley
11.
Nat Biomed Eng ; 1(9): 758-770, 2017 Sep.
Article in English | MEDLINE | ID: mdl-31015671

ABSTRACT

Bone grafts are one of the most commonly transplanted tissues. However, autologous grafts are in short supply, and can be associated with pain and donor-site morbidity. The creation of tissue-engineered bone grafts could help to fulfil clinical demand and provide a crucial resource for drug screening. Here, we show that vibrations of nanoscale amplitude provided by a newly developed bioreactor can differentiate a potential autologous cell source, mesenchymal stem cells (MSCs), into mineralized tissue in 3D. We demonstrate that nanoscale mechanotransduction can stimulate osteogenesis independently of other environmental factors, such as matrix rigidity. We show this by generating mineralized matrix from MSCs seeded in collagen gels with stiffness an order of magnitude below the stiffness of gels needed to induce bone formation in vitro. Our approach is scalable and can be compatible with 3D scaffolds.

12.
Nat Biomed Eng ; 1(12): 1004, 2017 12.
Article in English | MEDLINE | ID: mdl-31015702

ABSTRACT

In the version of this Article originally published, in Fig. 4f, the asterisk was missing; in Fig. 6a-c, the labels 'Wnt/ß-catenin signalling', 'Wnt/Ca+ pathway' and 'ERK' and their associated lines/arrows were missing; and in Fig. 6d and in the sentence beginning "In MSCs that were...", 'myosin' and 'nanostimulated', respectively, were spelt incorrectly. These errors have now been corrected in all versions of the Article.

13.
Sci Adv ; 2(8): e1600188, 2016 08.
Article in English | MEDLINE | ID: mdl-27574702

ABSTRACT

Growth factors (GFs) are powerful signaling molecules with the potential to drive regenerative strategies, including bone repair and vascularization. However, GFs are typically delivered in soluble format at supraphysiological doses because of rapid clearance and limited therapeutic impact. These high doses have serious side effects and are expensive. Although it is well established that GF interactions with extracellular matrix proteins such as fibronectin control GF presentation and activity, a translation-ready approach to unlocking GF potential has not been realized. We demonstrate a simple, robust, and controlled material-based approach to enhance the activity of GFs during tissue healing. The underlying mechanism is based on spontaneous fibrillar organization of fibronectin driven by adsorption onto the polymer poly(ethyl acrylate). Fibrillar fibronectin on this polymer, but not a globular conformation obtained on control polymers, promotes synergistic presentation of integrin-binding sites and bound bone morphogenetic protein 2 (BMP-2), which enhances mesenchymal stem cell osteogenesis in vitro and drives full regeneration of a nonhealing bone defect in vivo at low GF concentrations. This simple and translatable technology could unlock the full regenerative potential of GF therapies while improving safety and cost-effectiveness.


Subject(s)
Bone Morphogenetic Protein 2/genetics , Fibronectins/therapeutic use , Intercellular Signaling Peptides and Proteins/administration & dosage , Osteogenesis/drug effects , Regenerative Medicine , Acrylic Resins/chemistry , Acrylic Resins/therapeutic use , Binding Sites , Bone Morphogenetic Protein 2/chemistry , Bone Regeneration/drug effects , Cell Culture Techniques , Cell Differentiation/genetics , Fibronectins/chemistry , Fibronectins/genetics , Humans , Integrins/genetics , Integrins/metabolism , Mesenchymal Stem Cells/drug effects , Osteogenesis/genetics , Polymers/therapeutic use
14.
Adv Healthc Mater ; 5(8): 947-55, 2016 Apr 20.
Article in English | MEDLINE | ID: mdl-26890261

ABSTRACT

A focus of orthopedic research is to improve osteointegration and outcomes of joint replacement. Material surface topography has been shown to alter cell adhesion, proliferation, and growth. The use of nanotopographical features to promote cell adhesion and bone formation is hoped to improve osteointegration and clinical outcomes. Use of block-copolymer self-assembled nanopatterns allows nanopillars to form via templated anodization with control over height and order, which has been shown to be of cellular importance. This project assesses the outcome of a human bone marrow-derived co-culture of adherent osteoprogenitors and osteoclast progenitors on polished titania and titania patterned with 15 nm nanopillars, fabricated by a block-copolymer templated anodization technique. Substrate implantation in rabbit femurs is performed to confirm the in vivo bone/implant integration. Quantitative and qualitative results demonstrate increased osteogenesis on the nanopillar substrate with scanning electron microscopy, histochemical staining, and real-time quantitative reverse-transcription polymerase chain reaction analysis performed. Osteoblast/osteoclast co-culture analysis shows an increase in osteoblastogenesis-related gene expression and reduction in osteoclastogenesis. Supporting this in vitro finding, in vivo implantation of substrates in rabbit femora indicates increased implant/bone contact by ≈20%. These favorable osteogenic characteristics demonstrate the potential of 15 nm titania nanopillars fabricated by the block-copolymer templated anodization technique.


Subject(s)
Nanoparticles/chemistry , Nanotechnology/methods , Osteoblasts/cytology , Osteoclasts/cytology , Osteogenesis/drug effects , Titanium/pharmacology , Animals , Bone Marrow Cells/cytology , Bone Marrow Cells/drug effects , Cell Proliferation/drug effects , Coculture Techniques , Gene Expression Regulation/drug effects , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/drug effects , Humans , Imaging, Three-Dimensional , Implants, Experimental , Male , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/drug effects , Nanoparticles/ultrastructure , Osteoblasts/drug effects , Osteoclasts/drug effects , Rabbits , Surface Properties , Tartrate-Resistant Acid Phosphatase/metabolism
15.
J Funct Biomater ; 6(3): 598-622, 2015 Jul 16.
Article in English | MEDLINE | ID: mdl-26193326

ABSTRACT

Adult or mesenchymal stem cells (MSCs) have been found in different tissues in the body, residing in stem cell microenvironments called "stem cell niches". They play different roles but their main activity is to maintain tissue homeostasis and repair throughout the lifetime of an organism. Their ability to differentiate into different cell types makes them an ideal tool to study tissue development and to use them in cell-based therapies. This differentiation process is subject to both internal and external forces at the nanoscale level and this response of stem cells to nanostimuli is the focus of this review.

16.
Nanomedicine (Lond) ; 10(6): 949-57, 2015.
Article in English | MEDLINE | ID: mdl-25867859

ABSTRACT

BACKGROUND: Optimised nanotopography with controlled disorder (NSQ50) has been shown to stimulate osteogenesis and new bone formation in vitro. Following osteointegration the implant interface must undergo constant remodeling without inducing immune response. AIM: We aimed to assess the effect of nanotopography on bone remodelling using osteoclast and osteoblast cocultures. MATERIALS & METHODS: We developed a novel osteoblast/osteoclast coculture using solely human bone marrow derived mesenchymal and hematopeotic progenitor cells without extraneous supplementation. The coculture was been applied to NSQ50 or flat control polycarbonate substrates and assessed using immunohistochemical and immunofluorescent microscopy, scanning electron microscopy and quantitative reverse-transcription PCR methods. RESULTS: These confirm the presence of mature osteoclasts, osteoblasts and bone formation in coculture. Osteoblast differentiation increased on NSQ50, with no significant difference in osteoclast differentiation. CONCLUSION: Controlled disorder nanotopography appears to be selectively bioactive. We recommend this coculture method to be a better in vitro approximation of the osseous environment encountered by implants.


Subject(s)
Bone Marrow Cells/cytology , Cell Culture Techniques , Nanomedicine/methods , Osteoblasts/cytology , Osteoclasts/cytology , Polymers/chemistry , Bone Marrow/pathology , Cell Differentiation , Cells, Cultured , Coculture Techniques , Humans , Immunohistochemistry , Microscopy, Electron, Scanning , Microscopy, Fluorescence , Polycarboxylate Cement/chemistry , Polymerase Chain Reaction , Stromal Cells/cytology
17.
Expert Rev Med Devices ; 11(4): 417-23, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24801757

ABSTRACT

We consider three biophysical factors operating at the nanoscale which can affect gene expression, and thus, differentiation, in cultured mammalian cells. These factors are nanovibration, nanoforces and the local nanotopography. Work supporting these conclusions is reviewed. It is argued that stirring of the medium close to the cells cannot contribute to the effects. It is suggested that the three factors interact. Possible pathways by which this could occur are outlined.


Subject(s)
Epigenesis, Genetic , Nanoparticles/chemistry , Nanotechnology , Vibration , Animals , Humans , Stochastic Processes , Time Factors
18.
Nanomedicine (Lond) ; 9(2): 207-20, 2014 Feb.
Article in English | MEDLINE | ID: mdl-23631503

ABSTRACT

AIM: Bacterial inclusion bodies (IBs) are nanostructured (submicron), pseudospherical proteinaceous particles produced in recombinant bacteria resulting from ordered protein aggregation. Being mechanically stable, several physicochemical and biological properties of IBs can be tuned by appropriate selection of the producer strain and of culture conditions. It has been previously shown that IBs favor cell adhesion and surface colonization by mammalian cell lines upon decoration on materials surfaces, but how these biomaterials could influence the behavior of mesenchymal stem cells remains to be explored. MATERIALS & METHODS: Here, the authors vary topography, stiffness and wettability using the IBs to decorate polycaprolactone surfaces on which mesenchymal stem cells are cultured. RESULTS: The authors show that these topographies can be used to specifically target osteogenesis from mesenchymal stem cells, and through metabolomics, they show that the cells have increased energy demand during this bone-related differentiation. CONCLUSION: IBs as topographies can be used not only to direct cell proliferation but also to target differentiation of mesenchymal stem cells.


Subject(s)
Inclusion Bodies/chemistry , Mesenchymal Stem Cells/cytology , Osteogenesis , Polyesters/chemistry , Base Sequence , DNA Primers , Fluorescent Antibody Technique , Humans , Microscopy, Electron, Scanning , Real-Time Polymerase Chain Reaction
19.
J Cell Biochem ; 115(2): 380-90, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24123223

ABSTRACT

There is a rapidly growing body of literature on the effects of topography and critically, nanotopography on cell adhesion, apoptosis and differentiation. Understanding the effects of nanotopography on cell adhesion and morphology and the consequences of cell shape changes in the nucleus, and consequently, gene expression offers new approaches to the elucidation and potential control of stem cell differentiation. In the current study we have used molecular approaches in combination with immunohistology and transcript analysis to understand the role of nanotopography on mesenchymal stem cell morphology and phenotype. Results demonstrate large changes in cell adhesion, nucleus and lamin morphologies in response to the different nanotopographies. Furthermore, these changes relate to alterations in packing of chromosome territories within the interphase nucleus. This, in turn, leads to changes in transcription factor activity and functional (phenotypical) signalling including cell metabolism. Nanotopography provides a useful, non-invasive tool for studying cellular mechanotransduction, gene and protein expression patterns, through effects on cell morphology. The different nanotopographies examined, result in different morphological changes in the cyto- and nucleo-skeleton. We propose that both indirect (biochemical) and direct (mechanical) signalling are important in these early stages of regulating stem cell fate as a consequence of altered metabolic changes and altered phenotype. The current studies provide new insight on cell-surface interactions and enhance our understanding of the modulation of stem cell differentiation with significant potential application in regenerative medicine.


Subject(s)
Cell Differentiation/genetics , Cell Nucleus/ultrastructure , Cell Shape/genetics , Mesenchymal Stem Cells/ultrastructure , Apoptosis/genetics , Cell Adhesion/genetics , Cell Nucleus/genetics , Gene Expression Regulation , Humans , Mesenchymal Stem Cells/metabolism , Phenotype , Regenerative Medicine , Signal Transduction
20.
Nanomedicine (Lond) ; 8(10): 1587-99, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23394133

ABSTRACT

AIMS: Bacterial inclusion bodies (IBs) are protein-based, amyloidal nanomaterials that mechanically stimulate mammalian cell proliferation upon surface decoration. However, their biological performance as potentially functional scaffolds in mammalian cell culture still needs to be explored. MATERIALS & METHODS: Using fluorescent proteins, we demonstrate significant membrane penetration of surface-attached IBs and a corresponding intracellular bioavailability of the protein material. RESULTS: When IBs are formed by protein drugs, such as the intracellular acting human chaperone Hsp70 or the extracellular/intracellular acting human FGF-2, IB components intervene on top-growing cells, namely by rescuing them from chemically induced apoptosis or by stimulating cell division under serum starvation, respectively. Protein release from IBs seems to mechanistically mimic the sustained secretion of protein hormones from amyloid-like secretory granules in higher organisms. CONCLUSION: We propose bacterial IBs as biomimetic nanostructured scaffolds (bioscaffolds) suitable for tissue engineering that, while acting as adhesive materials, partially disintegrate for the slow release of their biologically active building blocks. The bottom-up delivery of protein drugs mediated by bioscaffolds offers a highly promising platform for emerging applications in regenerative medicine.


Subject(s)
Cell Membrane Permeability/drug effects , Fibroblast Growth Factor 2/administration & dosage , HSP70 Heat-Shock Proteins/administration & dosage , Inclusion Bodies , Nanostructures/administration & dosage , Animals , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Delivery Systems , Escherichia coli/chemistry , Fibroblast Growth Factor 2/chemistry , HSP70 Heat-Shock Proteins/chemistry , Humans , Nanostructures/chemistry , Tissue Engineering
SELECTION OF CITATIONS
SEARCH DETAIL
...