Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 190
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Bioessays ; 46(5): e2300223, 2024 May.
Article in English | MEDLINE | ID: mdl-38522027

ABSTRACT

Ageing causes progressive decline in metabolic, behavioural, and physiological functions, leading to a reduced health span. The extracellular matrix (ECM) is the three-dimensional network of macromolecules that provides our tissues with structure and biomechanical resilience. Imbalance between damage and repair/regeneration causes the ECM to undergo structural deterioration with age, contributing to age-associated pathology. The ECM 'Ageing Across the Life Course' interdisciplinary research network (ECMage) was established to bring together researchers in the United Kingdom, and internationally, working on the emerging field of ECM ageing. Here we report on a consultation at a joint meeting of ECMage and the Medical Research Council / Versus Arthritis Centre for Integrated Research into Musculoskeletal Ageing, held in January 2023, in which delegates analysed the key questions and research opportunities in the field of ECM ageing. We examine fundamental biological questions, enabling technologies, systems of study and emerging in vitro and in silico models, alongside consideration of the broader challenges facing the field.


Subject(s)
Aging , Extracellular Matrix , Animals , Humans , Extracellular Matrix/metabolism , United Kingdom
2.
Cell Mol Life Sci ; 79(7): 386, 2022 Jun 28.
Article in English | MEDLINE | ID: mdl-35760917

ABSTRACT

The gut microbiota plays a central role in regulating host metabolism. While substantial progress has been made in discerning how the microbiota influences host functions post birth and beyond, little is known about how key members of the maternal gut microbiota can influence feto-placental growth. Notably, in pregnant women, Bifidobacterium represents a key beneficial microbiota genus, with levels observed to increase across pregnancy. Here, using germ-free and specific-pathogen-free mice, we demonstrate that the bacterium Bifidobacterium breve UCC2003 modulates maternal body adaptations, placental structure and nutrient transporter capacity, with implications for fetal metabolism and growth. Maternal and placental metabolome were affected by maternal gut microbiota (i.e. acetate, formate and carnitine). Histological analysis of the placenta confirmed that Bifidobacterium modifies placental structure via changes in Igf2P0, Dlk1, Mapk1 and Mapk14 expression. Additionally, B. breve UCC2003, acting through Slc2a1 and Fatp1-4 transporters, was shown to restore fetal glycaemia and fetal growth in association with changes in the fetal hepatic transcriptome. Our work emphasizes the importance of the maternal gut microbiota on feto-placental development and sets a foundation for future research towards the use of probiotics during pregnancy.


Subject(s)
Gastrointestinal Microbiome , Placenta , Animals , Bifidobacterium , Female , Fetal Development , Humans , Mice , Nutrients , Placenta/metabolism , Pregnancy
3.
FASEB J ; 34(5): 7075-7088, 2020 05.
Article in English | MEDLINE | ID: mdl-32253791

ABSTRACT

The early life gut microbiota plays a crucial role in regulating and maintaining the intestinal barrier, with disturbances in these communities linked to dysregulated renewal and replenishment of intestinal epithelial cells. Here we sought to determine pathological cell shedding outcomes throughout the postnatal developmental period, and which host and microbial factors mediate these responses. Surprisingly, neonatal mice (Day 14 and 21) were highly refractory to induction of cell shedding after intraperitoneal administration of liposaccharide (LPS), with Day 29 mice showing strong pathological responses, more similar to those observed in adult mice. These differential responses were not linked to defects in the cellular mechanisms and pathways known to regulate cell shedding responses. When we profiled microbiota and metabolites, we observed significant alterations. Neonatal mice had high relative abundances of Streptococcus, Escherichia, and Enterococcus and increased primary bile acids. In contrast, older mice were dominated by Candidatus Arthromitus, Alistipes, and Lachnoclostridium, and had increased concentrations of SCFAs and methyamines. Antibiotic treatment of neonates restored LPS-induced small intestinal cell shedding, whereas adult fecal microbiota transplant alone had no effect. Our findings further support the importance of the early life window for microbiota-epithelial interactions in the presence of inflammatory stimuli and highlights areas for further investigation.


Subject(s)
Animals, Newborn/microbiology , Gastrointestinal Microbiome/physiology , Intestinal Mucosa/microbiology , Intestinal Mucosa/pathology , Animals , Animals, Newborn/metabolism , Anti-Bacterial Agents/administration & dosage , Bile Acids and Salts/metabolism , Epithelial Cells/metabolism , Epithelial Cells/pathology , Fecal Microbiota Transplantation , Female , Gastrointestinal Microbiome/drug effects , Gastrointestinal Microbiome/genetics , Host Microbial Interactions/drug effects , Host Microbial Interactions/physiology , Inflammation/metabolism , Inflammation/microbiology , Inflammation/pathology , Intestinal Mucosa/metabolism , Intestine, Small/metabolism , Intestine, Small/microbiology , Intestine, Small/pathology , Lipopolysaccharides/administration & dosage , Mice , Mice, Inbred C57BL , Signal Transduction
4.
Cell Biol Toxicol ; 37(6): 915-933, 2021 12.
Article in English | MEDLINE | ID: mdl-33420657

ABSTRACT

Mesenchymal stem cells represent an important resource, for bone regenerative medicine and therapeutic applications. This review focuses on new advancements and biophysical tools which exploit different physical and chemical markers of mesenchymal stem cell populations, to finely characterize phenotype changes along their osteogenic differentiation process. Special attention is paid to recently developed label-free methods, which allow monitoring cell populations with minimal invasiveness. Among them, quantitative phase imaging, suitable for single-cell morphometric analysis, and nanoindentation, functional to cellular biomechanics investigation. Moreover, the pool of ion channels expressed in cells during differentiation is discussed, with particular interest for calcium homoeostasis.Altogether, a biophysical perspective of osteogenesis is proposed, offering a valuable tool for the assessment of the cell stage, but also suggesting potential physiological links between apparently independent phenomena.


Subject(s)
Mesenchymal Stem Cells , Osteogenesis , Biomarkers , Cell Differentiation , Cells, Cultured
5.
Biochem J ; 477(17): 3349-3366, 2020 09 18.
Article in English | MEDLINE | ID: mdl-32941644

ABSTRACT

Twenty-five years have passed since the first clinical trial utilising mesenchymal stomal/stem cells (MSCs) in 1995. In this time academic research has grown our understanding of MSC biochemistry and our ability to manipulate these cells in vitro using chemical, biomaterial, and mechanical methods. Research has been emboldened by the promise that MSCs can treat illness and repair damaged tissues through their capacity for immunomodulation and differentiation. Since 1995, 31 therapeutic products containing MSCs and/or progenitors have reached the market with the level of in vitro manipulation varying significantly. In this review, we summarise existing therapeutic products containing MSCs or mesenchymal progenitor cells and examine the challenges faced when developing new therapeutic products. Successful progression to clinical trial, and ultimately market, requires a thorough understanding of these hurdles at the earliest stages of in vitro pre-clinical development. It is beneficial to understand the health economic benefit for a new product and the reimbursement potential within various healthcare systems. Pre-clinical studies should be selected to demonstrate efficacy and safety for the specific clinical indication in humans, to avoid duplication of effort and minimise animal usage. Early consideration should also be given to manufacturing: how cell manipulation methods will integrate into highly controlled workflows and how they will be scaled up to produce clinically relevant quantities of cells. Finally, we summarise the main regulatory pathways for these clinical products, which can help shape early therapeutic design and testing.


Subject(s)
Cell Differentiation , Cell Proliferation , Immunologic Factors , Immunomodulation , Mesenchymal Stem Cells/metabolism , Animals , Humans , Immunologic Factors/metabolism , Immunologic Factors/therapeutic use
6.
Philos Trans A Math Phys Eng Sci ; 376(2120)2018 May 28.
Article in English | MEDLINE | ID: mdl-29661978

ABSTRACT

Mechanical signals are ubiquitous in our everyday life and the process of converting these mechanical signals into a biological signalling response is known as mechanotransduction. Our understanding of mechanotransduction, and its contribution to vital cellular responses, is a rapidly expanding field of research involving complex processes that are still not clearly understood. The use of mechanical vibration as a stimulus of mechanotransduction, including variation of frequency and amplitude, allows an alternative method to control specific cell behaviour without chemical stimulation (e.g. growth factors). Chemical-independent control of cell behaviour could be highly advantageous for fields including drug discovery and clinical tissue engineering. In this review, a novel technique is described based on nanoscale sinusoidal vibration. Using finite-element analysis in conjunction with laser interferometry, techniques that are used within the field of gravitational wave detection, optimization of apparatus design and calibration of vibration application have been performed. We further discuss the application of nanovibrational stimulation, or 'nanokicking', to eukaryotic and prokaryotic cells including the differentiation of mesenchymal stem cells towards an osteoblast cell lineage. Mechanotransductive mechanisms are discussed including mediation through the Rho-A kinase signalling pathway. Optimization of this technique was first performed in two-dimensional culture using a simple vibration platform with an optimal frequency and amplitude of 1 kHz and 22 nm. A novel bioreactor was developed to scale up cell production, with recent research demonstrating that mesenchymal stem cell differentiation can be efficiently triggered in soft gel constructs. This important step provides first evidence that clinically relevant (three-dimensional) volumes of osteoblasts can be produced for the purpose of bone grafting, without complex scaffolds and/or chemical induction. Initial findings have shown that nanovibrational stimulation can also reduce biofilm formation in a number of clinically relevant bacteria. This demonstrates additional utility of the bioreactor to investigate mechanotransduction in other fields of research.This article is part of a discussion meeting issue 'The promises of gravitational-wave astronomy'.

7.
Nanomedicine ; 14(7): 2455-2464, 2018 10.
Article in English | MEDLINE | ID: mdl-28552647

ABSTRACT

Regulation of cell behavior in response to nanoscale features has been the focus of much research in recent years and the successful generation of nanoscale features capable of mimicking the natural nanoscale interface has been of great interest in the field of biomaterials research. In this review, we discuss relevant nanofabrication techniques and how they are combined with bioengineering applications to mimic the natural extracellular matrix (ECM) and create valuable nanoscale interfaces.


Subject(s)
Biocompatible Materials/chemistry , Biomimetics , Cell Adhesion , Extracellular Matrix/chemistry , Nanostructures/chemistry , Animals , Humans
8.
Langmuir ; 32(3): 800-9, 2016 Jan 26.
Article in English | MEDLINE | ID: mdl-26715432

ABSTRACT

Cells, by interacting with surfaces indirectly through a layer of extracellular matrix proteins, can respond to a variety of physical properties, such as topography or stiffness. Polymer surface mobility is another physical property that is less well understood but has been indicated to hold the potential to modulate cell behavior. Polymer mobility is related to the glass-transition temperature (Tg) of the system, the point at which a polymer transitions from an amorphous solid to a more liquid-like state. This work shows that changes in polymer mobility translate to interfacial mobility of extracellular matrix proteins adsorbed on the material surface. This study has utilized a family of polyalkyl acrylates with similar chemistry but different degrees of mobility, obtained through increasing length of the side chain. These materials are used, in conjunction with fluorescent fibronectin, to determine the mobility of this interfacial layer of protein that constitutes the initial cell-material interface. Furthermore, the extent of fibronectin domain availability (III9, III10, - the integrin binding site), cell-mediated reorganization, and cell differentiation was also determined. A nonmonotonic dependence of fibronectin mobility on polymer surface mobility was observed, with a similar trend noted in cell-mediated reorganization of the protein layer by L929 fibroblasts. The availability of the integrin-binding site was higher on the more mobile surfaces, where a similar organization of the protein into networks at the material interface was observed. Finally, differentiation of C2C12 myoblasts was seen to be highly sensitive to surface mobility upon inhibition of cell contractility. Altogether, these findings show that polymer mobility is a subtle influence that translates to the cell/material interface through the protein layer to alter the biological activity of the surface.


Subject(s)
Acrylates/chemistry , Extracellular Matrix Proteins/chemistry , Fibronectins/chemistry , Integrins/chemistry , Animals , Cell Adhesion , Cell Line , Fibroblasts/chemistry , Fibroblasts/cytology , Fluorescein-5-isothiocyanate/chemistry , Fluorescent Dyes/chemistry , Mice , Myoblasts/chemistry , Myoblasts/cytology , Phase Transition , Protein Binding , Protein Transport , Surface Properties , Transition Temperature
9.
Nat Mater ; 13(6): 558-69, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24845995

ABSTRACT

Stem cells respond to nanoscale surface features, with changes in cell growth and differentiation mediated by alterations in cell adhesion. The interaction of nanotopographical features with integrin receptors in the cells' focal adhesions alters how the cells adhere to materials surfaces, and defines cell fate through changes in both cell biochemistry and cell morphology. In this Review, we discuss how cell adhesions interact with nanotopography, and we provide insight as to how materials scientists can exploit these interactions to direct stem cell fate and to understand how the behaviour of stem cells in their niche can be controlled. We expect knowledge gained from the study of cell-nanotopography interactions to accelerate the development of next-generation stem cell culture materials and implant interfaces, and to fuel discovery of stem cell therapeutics to support regenerative therapies.


Subject(s)
Extracellular Matrix/chemistry , Focal Adhesions , Integrins/chemistry , Stem Cells/metabolism , Cell Adhesion , Extracellular Matrix/metabolism , Humans , Integrins/metabolism , Stem Cells/cytology
10.
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
11.
Nano Lett ; 13(2): 570-6, 2013 Feb 13.
Article in English | MEDLINE | ID: mdl-23252684

ABSTRACT

The function and fate of cells is influenced by many different factors, one of which is surface topography of the support culture substrate. Systematic studies of nanotopography and cell response have typically been limited to single cell types and a small set of topographical variations. Here, we show a radical expansion of experimental throughput using automated detection, measurement, and classification of co-cultured cells on a nanopillar array where feature height changes continuously from planar to 250 nm over 9 mm. Individual cells are identified and characterized by more than 200 descriptors, which are used to construct a set of rules for label-free segmentation into individual cell types. Using this approach we can achieve label-free segmentation with 84% confidence across large image data sets and suggest optimized surface parameters for nanostructuring of implant devices such as vascular stents.


Subject(s)
Cell Tracking/methods , Endothelial Cells/cytology , Fibroblasts/cytology , Nanostructures/chemistry , Nanotechnology/methods , Cell Tracking/instrumentation , Coculture Techniques , Fluorescence , Humans , Nanotechnology/instrumentation
12.
Exp Hematol ; 135: 104232, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38729553

ABSTRACT

The bone marrow (BM) niche is a complex microenvironment that provides the signals required for regulation of hematopoietic stem cells (HSCs) and the process of hematopoiesis they are responsible for. Bioengineered models of the BM niche incorporate various elements of the in vivo BM microenvironment, including cellular components, soluble factors, a three-dimensional environment, mechanical stimulation of included cells, and perfusion. Recent advances in the bioengineering field have resulted in a spate of new models that shed light on BM function and are approaching precise imitation of the BM niche. These models promise to improve our understanding of the in vivo microenvironment in health and disease. They also aim to serve as platforms for HSC manipulation or as preclinical models for screening novel therapies for BM-associated disorders and diseases.


Subject(s)
Bone Marrow , Hematopoiesis , Hematopoietic Stem Cells , Stem Cell Niche , Humans , Animals , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Bone Marrow/metabolism , Models, Biological , Bone Marrow Cells/metabolism , Bone Marrow Cells/cytology
13.
Biomater Adv ; 160: 213861, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38663159

ABSTRACT

Novel strategies employing mechano-transducing materials eliciting biological outcomes have recently emerged for controlling cellular behaviour. Targeted cellular responses are achieved by manipulating physical, chemical, or biochemical modification of material properties. Advances in techniques such as nanopatterning, chemical modification, biochemical molecule embedding, force-tuneable materials, and artificial extracellular matrices are helping understand cellular mechanotransduction. Collectively, these strategies manipulate cellular sensing and regulate signalling cascades including focal adhesions, YAP-TAZ transcription factors, and multiple osteogenic pathways. In this minireview, we are providing a summary of the influence that these materials, particularly titanium-based orthopaedic materials, have on cells. We also highlight recent complementary methodological developments including, but not limited to, the use of metabolomics for identification of active biomolecules that drive cellular differentiation.


Subject(s)
Mechanotransduction, Cellular , Osteogenesis , Osteogenesis/physiology , Humans , Titanium/chemistry , Animals , Cell Differentiation , Surface Properties , Biocompatible Materials/chemistry , Extracellular Matrix/metabolism , Extracellular Matrix/physiology , Extracellular Matrix/chemistry
14.
Acta Biomater ; 180: 154-170, 2024 05.
Article in English | MEDLINE | ID: mdl-38621600

ABSTRACT

Bacterial infection remains a significant problem associated with orthopaedic surgeries leading to surgical site infection (SSI). This unmet medical need can become an even greater complication when surgery is due to malignant bone tumor. In the present study, we evaluated in vitro titanium (Ti) implants subjected to gallium (Ga) and silver (Ag)-doped thermochemical treatment as strategy to prevent SSI and improve osteointegration in bone defects caused by diseases such as osteoporosis, bone tumor, or bone metastasis. Firstly, as Ga has been reported to be an osteoinductive and anti-resorptive agent, its performance in the mixture was proved by studying human mesenchymal stem cells (hMSC) and pre-osteoclasts (RAW264.7) behaviour. Then, the antibacterial potential provided by Ag was assessed by resembling "The Race for the Surface" between hMSC and Pseudomonas aeruginosa in two co-culture methods. Moreover, the presence of quorum sensing molecules in the co-culture was evaluated. The results highlighted the suitability of the mixture to induce osteodifferentiation and reduce osteoclastogenesis in vitro. Furthermore, the GaAg surface promoted strong survival rate and retained osteoinduction potential of hMSCs even after bacterial inoculation. Therefore, GaAg-modified titanium may be an ideal candidate to repair bone defects caused by excessive bone resorption, in addition to preventing SSI. STATEMENT OF SIGNIFICANCE: This article provides important insights into titanium for fractures caused by osteoporosis or bone metastases with high incidence in surgical site infection (SSI) because in this situation bacterial infection can become a major disaster. In order to solve this unmet medical need, we propose a titanium implant modified with gallium and silver to improve osteointegration, reduce bone resorption and avoid bacterial infection. For that aim, we study osteoblast and osteoclast behavior with the main novelty focused on the antibacterial evaluation. In this work, we recreate "the race for the surface" in long-term experiments and study bacterial virulence factors (quorum sensing). Therefore, we believe that our article could be of great interest, providing a great impact on future orthopedic applications.


Subject(s)
Coculture Techniques , Gallium , Mesenchymal Stem Cells , Osteogenesis , Pseudomonas aeruginosa , Silver , Titanium , Titanium/chemistry , Titanium/pharmacology , Silver/pharmacology , Silver/chemistry , Humans , Gallium/pharmacology , Gallium/chemistry , Mice , Mesenchymal Stem Cells/drug effects , Animals , Osteogenesis/drug effects , Pseudomonas aeruginosa/drug effects , Bone Resorption/pathology , Surface Properties , RAW 264.7 Cells , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Bacterial Infections/prevention & control
15.
Adv Mater ; 36(23): e2310789, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38253339

ABSTRACT

Conventional osteogenic platforms utilize active growth factors to repair bone defects that are extensive in size, but they can adversely affect patient health. Here, an unconventional osteogenic platform is reported that functions by promoting capture of inactive osteogenic growth factor molecules to the site of cell growth for subsequent integrin-mediated activation, using a recombinant fragment of latent transforming growth factor beta-binding protein-1 (rLTBP1). It is shown that rLTBP1 binds to the growth-factor- and integrin-binding domains of fibronectin on poly(ethyl acrylate) surfaces, which immobilizes rLTBP1 and promotes the binding of latency associated peptide (LAP), within which inactive transforming growth factor beta 1 (TGF-ß1) is bound. rLTBP1 facilitates the interaction of LAP with integrin ß1 and the subsequent mechanically driven release of TGF-ß1 to stimulate canonical TGF-ß1 signaling, activating osteogenic marker expression in vitro and complete regeneration of a critical-sized bone defect in vivo.


Subject(s)
Osteogenesis , Transforming Growth Factor beta1 , Animals , Humans , Transforming Growth Factor beta1/metabolism , Fibronectins/metabolism , Fibronectins/chemistry , Latent TGF-beta Binding Proteins/metabolism , Latent TGF-beta Binding Proteins/chemistry , Bone Regeneration , Surface Properties , Integrins/metabolism , Protein Binding , Integrin beta1/metabolism , Signal Transduction
16.
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
17.
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
18.
ACS Nano ; 18(27): 17630-17641, 2024 Jul 09.
Article in English | MEDLINE | ID: mdl-38924391

ABSTRACT

Osteoporosis disrupts the fine-tuned balance between bone formation and resorption, leading to reductions in bone quantity and quality and ultimately increasing fracture risk. Prevention and treatment of osteoporotic fractures is essential for reductions in mortality, morbidity, and the economic burden, particularly considering the aging global population. Extreme bone loss that mimics time-accelerated osteoporosis develops in the paralyzed limbs following complete spinal cord injury (SCI). In vitro nanoscale vibration (1 kHz, 30 or 90 nm amplitude) has been shown to drive differentiation of mesenchymal stem cells toward osteoblast-like phenotypes, enhancing osteogenesis and inhibiting osteoclastogenesis simultaneously. Here, we develop and characterize a wearable device designed to deliver and monitor continuous nanoamplitude vibration to the hindlimb long bones of rats with complete SCI. We investigate whether a clinically feasible dose of nanovibration (two 2 h/day, 5 days/week for 6 weeks) is effective at reversing the established SCI-induced osteoporosis. Laser interferometry and finite element analysis confirmed transmission of nanovibration into the bone, and microcomputed tomography and serum bone formation and resorption markers assessed effectiveness. The intervention did not reverse SCI-induced osteoporosis. However, serum analysis indicated an elevated concentration of the bone formation marker procollagen type 1 N-terminal propeptide (P1NP) in rats receiving 40 nm amplitude nanovibration, suggesting increased synthesis of type 1 collagen, the major organic component of bone. Therefore, enhanced doses of nanovibrational stimulus may yet prove beneficial in attenuating/reversing osteoporosis, particularly in less severe forms of osteoporosis.


Subject(s)
Osteoporosis , Spinal Cord Injuries , Vibration , Animals , Rats , Osteoporosis/pathology , Osteoporosis/prevention & control , Rats, Sprague-Dawley , X-Ray Microtomography , Osteogenesis/drug effects , Female , Wearable Electronic Devices , Nanotechnology
19.
Nat Commun ; 15(1): 5791, 2024 Jul 10.
Article in English | MEDLINE | ID: mdl-38987295

ABSTRACT

Long-term reconstituting haematopoietic stem cells (LT-HSCs) are used to treat blood disorders via stem cell transplantation. The very low abundance of LT-HSCs and their rapid differentiation during in vitro culture hinders their clinical utility. Previous developments using stromal feeder layers, defined media cocktails, and bioengineering have enabled HSC expansion in culture, but of mostly short-term HSCs and progenitor populations at the expense of naive LT-HSCs. Here, we report the creation of a bioengineered LT-HSC maintenance niche that recreates physiological extracellular matrix organisation, using soft collagen type-I hydrogels to drive nestin expression in perivascular stromal cells (PerSCs). We demonstrate that nestin, which is expressed by HSC-supportive bone marrow stromal cells, is cytoprotective and, via regulation of metabolism, is important for HIF-1α expression in PerSCs. When CD34+ve HSCs were added to the bioengineered niches comprising nestin/HIF-1α expressing PerSCs, LT-HSC numbers were maintained with normal clonal and in vivo reconstitution potential, without media supplementation. We provide proof-of-concept that our bioengineered niches can support the survival of CRISPR edited HSCs. Successful editing of LT-HSCs ex vivo can have potential impact on the treatment of blood disorders.


Subject(s)
Extracellular Matrix , Hematopoietic Stem Cells , Hypoxia-Inducible Factor 1, alpha Subunit , Nestin , Hematopoietic Stem Cells/metabolism , Hematopoietic Stem Cells/cytology , Animals , Nestin/metabolism , Nestin/genetics , Extracellular Matrix/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Mice , Stem Cell Niche , Hydrogels/chemistry , Bioengineering/methods , Humans , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/cytology , Hematopoietic Stem Cell Transplantation , Antigens, CD34/metabolism , Collagen Type I/metabolism , Cell Differentiation , Mice, Inbred C57BL
20.
Small ; 9(12): 2140-51, 2013 Jun 24.
Article in English | MEDLINE | ID: mdl-23362187

ABSTRACT

The production of bone-forming osteogenic cells for research purposes or transplantation therapies remains a significant challenge. Using planar polycarbonate substrates lacking in topographical cues and substrates displaying a nanotopographical pattern, mesenchymal differentiation of human embryonic stem cells is directed in the absence of chemical factors and without induction of differentiation by embryoid body formation. Cells incubated on nanotopographical substrates show enhanced expression of mesenchymal or stromal markers and expression of early osteogenic progenitors at levels above those detected in cells on planar substrates in the same basal media. Evidence of epithelial-to-mesenchymal transition during substrate differentiation and DNA methylation changes akin to chemical induction are also observed. These studies provide a suitable approach to overcome regenerative medical challenges and describe a defined, reproducible platform for human embryonic stem cell differentiation.


Subject(s)
Embryonic Stem Cells/cytology , Cell Differentiation/drug effects , DNA Methylation , Epithelial-Mesenchymal Transition , Humans , Mesenchymal Stem Cells/cytology , Nanotechnology , Osteogenesis/physiology
SELECTION OF CITATIONS
SEARCH DETAIL