Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 168
Filter
1.
J Tissue Viability ; 32(4): 508-515, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37442720

ABSTRACT

Pressure ulcers (PUs) remain a chronic health problem with severe impacts on healthcare systems. Early detection is crucial to providing effective interventions. However, detecting PUs currently relies on subjective tissue evaluations, such as visual skin assessment, precluding interventions prior to the development of visible tissue damage. There is an unmet need for solutions that can detect early tissue damage before visual and tactile signs occur. Assessments based on sub-epidermal moisture (SEM) measurements represent an opportunity for robust and objective early detection of PUs, preventing broken skin PUs in more high-risk patients at high-risk anatomical locations. While SEM assessment technology has been validated in computational, bench and tissue phantom models, validation in soft tissue was absent. In this study, we successfully validated the ability of a commercially available SEM assessment device to measure and detect sub-epidermal moisture changes in a novel ex vivo porcine soft tissue model of localised oedema. When controlled and incremental fluid volumes (Phosphate Buffer Solution) were injected into porcine soft tissues, statistically significant differences were found in SEM values between fluid-injected sites, representing an inflammatory oedematous condition, and healthy tissue control sites, as measured by the SEM device. The device provided reproducible readings by detecting localised oedema changes in soft tissues, reflecting the build-up of fluid as small as 1 ml into the underlying tissue. Spatial characterization experiments described the ability of the device technology to differentiate between healthy and oedematous tissue. Our findings validate the use of SEM assessment technology to measure and quantify localized oedema.


Subject(s)
Pressure Ulcer , Humans , Swine , Animals , Pressure Ulcer/diagnosis , Pressure Ulcer/prevention & control , Epidermis , Skin , Edema/diagnosis , Suppuration
2.
Eur Cell Mater ; 45: 158-172, 2023 Jun 29.
Article in English | MEDLINE | ID: mdl-37382477

ABSTRACT

Extracellular matrix (ECM) biomaterials have shown promise for treating small artucular-joint defetcs. However, ECM-based biomaterials generally lack appropriate mechanical properties to support physiological loads and are prone to delamination in larger cartilage defects. To overcome these common mechanical limitations, a collagen hyaluronic-acid (CHyA) matrix, with proven regenerative potential, was reinforced with a bioabsorbable 3D-printed framework to support physiological loads. Polycaprolactone (PCL) was 3D-printed in two configurations, rectilinear and gyroid designs, that were extensively mechanically characterised. Both scaffold designs increased the compressive modulus of the CHyA matrices by three orders of magnitude, mimicking the physiological range (0.5-2.0 MPa) of healthy cartilage. The gyroid scaffold proved to be more flexible compared to the rectilinear scaffold, thus better contouring to the curvature of a femoral condyle. Additionally, PCL reinforcement of the CHyA matrix increased the tensile modulus and allowed for suture fixation of the scaffold to the subchondral bone, thus addressing the major challenge of biomaterial fixation to articular joint surfaces in shallow defects. In vitro evaluation confirmed successful infiltration of human mesenchymal stromal cells (MSCs) within the PCL-CHyA scaffolds, which resulted in increased production of sulphated glycosaminoglycans (sGAG/DNA; p = 0.0308) compared to non-reinforced CHyA matrices. Histological staining using alcian blue confirmed these results, while also indicating greater spatial distribution of sGAG throughout the PCL-CHyA scaffold. These findings have a great clinical importance as they provide evidence that reinforced PCL-CHyA scaffolds, with their increased chondroinductive potential and compatibility with joint fixation techniques, could be used to repair large-area chondral defects that currently lack effective treatment options.


Subject(s)
Absorbable Implants , Cartilage , Humans , Glycosaminoglycans , Hyaluronic Acid , Biocompatible Materials/pharmacology , Printing, Three-Dimensional
3.
Int Urogynecol J ; 33(8): 2177-2184, 2022 08.
Article in English | MEDLINE | ID: mdl-35312806

ABSTRACT

INTRODUCTION AND HYPOTHESIS: The use of polypropylene (PP) mesh for stress urinary incontinence (SUI) surgery has declined because of safety concerns. The aim of this study is to evaluate a biodegradable polycaprolactone (PCL) mesh and a PCL composite mesh tissue engineered with human uterine fibroblasts (HUFs) for SUI surgery by comparing mechanical properties and in vitro biocompatibility to commercially available PP and porcine dermis (PD). METHODS: The mechanical properties of four scaffold materials were evaluated: PCL, PCL-collagen-hyaluronic acid composite, acellular porcine dermal collagen (PD) (Pelvicol™) and polypropylene (Gynecare TVT™ Exact®). HUFs were seeded on separate scaffolds. After 7 and 14 days scaffolds were assessed for metabolic activity and cell proliferation using Alamar Blue, Live/Dead and PicoGreen assays. Soluble collagen production was evaluated using a Sircol assay. RESULTS: PCL and the composite scaffold reached ultimate tensile strength (UTS) values closest to healthy pelvic floor tissue (PCL = 1.19 MPa; composite = 1.13 MPa; pelvic floor = 0.79 MPa; Lei et al. Int Urogynecol J Pelvic Floor Dysfunct. 18(6):603-7, 2007). Cells on PCL showed significantly greater cell viability than PP at day 7 (p < 0.0001). At D14 the composite scaffold showed significantly greater cell viability than PP (p = 0.0006). PCL was the best performing scaffold for soluble collagen production at day 14 (106.1 µg versus 13.04 µg for PP, p = 0.0173). CONCLUSIONS: We have designed a biodegradable PCL mesh and a composite mesh which demonstrate better biocompatibility than PP and mechanical properties closer to that of healthy pelvic floor tissue. This in vitro study provides promising evidence that these two implants should be evaluated in animal and human trials.


Subject(s)
Urinary Incontinence, Stress , Animals , Collagen , Humans , Polyesters , Polypropylenes , Surgical Mesh/adverse effects , Swine , Tissue Engineering , Tissue Scaffolds , Urinary Incontinence, Stress/surgery
4.
Biomaterials ; 279: 121187, 2021 12.
Article in English | MEDLINE | ID: mdl-34678648

ABSTRACT

In recent decades, an increasing number of tissue engineered bone grafts have been developed. However, expensive and laborious screenings in vivo are necessary to assess the safety and efficacy of their formulations. Rodents are the first choice for initial in vivo screens but their size limits the dimensions and number of the bone grafts that can be tested in orthotopic locations. Here, we report the development of a refined murine subcutaneous model for semi-orthotopic bone formation that allows the testing of up to four grafts per mouse one order of magnitude greater in volume than currently possible in mice. Crucially, these defects are also "critical size" and unable to heal within the timeframe of the study without intervention. The model is based on four bovine bone implants, ring-shaped, where the bone healing potential of distinct grafts can be evaluated in vivo. In this study we demonstrate that promotion and prevention of ossification can be assessed in our model. For this, we used a semi-automatic algorithm for longitudinal micro-CT image registration followed by histological analyses. Taken together, our data supports that this model is suitable as a platform for the real-time screening of bone formation, and provides the possibility to study bone resorption, osseointegration and vascularisation.


Subject(s)
Bone Regeneration , Regenerative Medicine , Animals , Biocompatible Materials , Cattle , Mice , Osteogenesis , Tissue Engineering , Tissue Scaffolds
5.
Eur J Obstet Gynecol Reprod Biol ; 259: 153-160, 2021 Apr.
Article in English | MEDLINE | ID: mdl-33676124

ABSTRACT

Stress urinary incontinence (SUI) was managed with techniques such as colposuspension, autologous fascia sling and urethral bulking agents. The introduction of the mid-urethral polypropylene (PP) sling in the 1990s led to a significant and rapid global change in SUI surgery. The synthetic non-degradable PP sling had superior results to traditional SUI procedures but its use has now declined due to significant complications such as pain and mesh erosion. These complications are attributed to its poor biocompatibility and integration into vaginal tissues. The efficacy of PP was extrapolated from studies on abdominal wall repair and it is now clear that integration of implanted materials in the pelvic floor differs from the abdominal wall. With PP prohibited in some jurisdictions, female patients with SUI have few management options. In the present review we summarise recent advances in SUI surgery and evaluate potential alternatives to PP slings with a particular focus on degradable materials. Allograft and xenograft materials demonstrate good biocompatibility but have yielded suboptimal cure rates. Tissue engineered synthetic degradable materials outperform unmodified synthetic degradable materials in terms of biomechanics and cell support. Synthetic tissue engineered degradable materials show promising results from in vitro studies and future research should focus on animal and human trials in this field.


Subject(s)
Suburethral Slings , Urinary Incontinence, Stress , Female , Humans , Male , Polypropylenes , Suburethral Slings/adverse effects , Surgical Mesh/adverse effects , Urethra , Urinary Incontinence, Stress/surgery , Urologic Surgical Procedures
6.
J Pers Med ; 11(1)2020 Dec 22.
Article in English | MEDLINE | ID: mdl-33375065

ABSTRACT

Skin wounds can lead to serious morbidity complications in diabetic patients due to the reduced healing potential of autologous stem cells. One reason for the low functional potency of stem cells from diabetic patients (diabetic stem cells) is attributed to their senescent-like nature. Here, we investigated if an anti-ageing protein, ß-klotho, could be used to rejuvenate diabetic stem cells and to promote pro-angiogenic gene-activated scaffold (GAS)-induced functional response for wound healing applications. Human stem cells derived from the adipose tissue (adipose-derived stem cells (ADSCs)) of normal and diabetic (type 2) donors were used for the study. We report that the ß-klotho priming facilitated inflammatory signal pruning by reducing interleukin-8 release by more than half while concurrently doubling the release of monocyte chemoattractant protein-1. Additionally, ß-klotho priming enhanced the pro-angiogenic response of diabetic ADSCs on GAS by dampening the release of anti-angiogenic factors (i.e., pigment epithelium-derived factor, tissue inhibitor of metalloproteinase-1 and thrombospondin-1) while simultaneously supporting the expression of pro-angiogenic factors (i.e., Vascular Endothelial Growth Factor (VEGF), angiopoietin-2 and angiogenin). Finally, we show that ß-klotho pre-treatment expedites the cellular expression of matrix proteins such as collagen IV and collagen VI, which are implicated in tissue maturation. Taken together, our study provides evidence that the synergistic effect of the pro-angiogenic GAS and ß-klotho activation effectively accelerates the functional development of diabetic ADSCs for wound healing applications.

7.
J Control Release ; 301: 13-27, 2019 05 10.
Article in English | MEDLINE | ID: mdl-30853527

ABSTRACT

The regeneration of complex tissues and organs remains a major clinical challenge. With a view towards bioprinting such tissues, we developed a new class of pore-forming bioink to spatially and temporally control the presentation of therapeutic genes within bioprinted tissues. By blending sacrificial and stable hydrogels, we were able to produce bioinks whose porosity increased with time following printing. When combined with amphipathic peptide-based plasmid DNA delivery, these bioinks supported enhanced non-viral gene transfer to stem cells in vitro. By modulating the porosity of these bioinks, it was possible to direct either rapid and transient (pore-forming bioinks), or slower and more sustained (solid bioinks) transfection of host or transplanted cells in vivo. To demonstrate the utility of these bioinks for the bioprinting of spatially complex tissues, they were next used to zonally position stem cells and plasmids encoding for either osteogenic (BMP2) or chondrogenic (combination of TGF-ß3, BMP2 and SOX9) genes within networks of 3D printed thermoplastic fibers to produce mechanically reinforced, gene activated constructs. In vivo, these bioprinted tissues supported the development of a vascularised, bony tissue overlaid by a layer of stable cartilage. When combined with multiple-tool biofabrication strategies, these gene activated bioinks can enable the bioprinting of a wide range of spatially complex tissues.


Subject(s)
Bioprinting , Gene Transfer Techniques , Ink , Tissue Engineering , Alginates , Animals , Bone Morphogenetic Protein 2/genetics , DNA/administration & dosage , Hydrogels , Mesenchymal Stem Cells , Methylcellulose , Plasmids , Porosity , Printing, Three-Dimensional , SOX9 Transcription Factor/genetics , Swine , Transforming Growth Factor beta3/genetics
8.
Mater Today Bio ; 3: 100009, 2019 Jun.
Article in English | MEDLINE | ID: mdl-32159148

ABSTRACT

There is an urgent, clinical need for an alternative to the use of autologous grafts for the ever increasing number of bone grafting procedures performed annually. Herein, we describe a developmentally inspired approach to bone tissue engineering, which focuses on leveraging biomaterials as platforms for recapitulating the process of endochondral ossification. To begin, we describe the traditional biomaterial-based approaches to tissue engineering that have been investigated as methods to promote in vivo bone regeneration, including the use of three-dimensional biomimetic scaffolds, the delivery of growth factors and recombinant proteins, and the in vitro engineering of mineralized bone-like tissue. Thereafter, we suggest that some of the hurdles encountered by these traditional tissue engineering approaches may be circumvented by modulating the endochondral route to bone repair and, to that end, we assess various biomaterials that can be used in combination with cells and signaling factors to engineer hypertrophic cartilaginous grafts capable of promoting endochondral bone formation. Finally, we examine the emerging trends in biomaterial-based approaches to endochondral bone regeneration, such as the engineering of anatomically shaped templates for bone and osteochondral tissue engineering, the fabrication of mechanically reinforced constructs using emerging three-dimensional bioprinting techniques, and the generation of gene-activated scaffolds, which may accelerate the field towards its ultimate goal of clinically successful bone organ regeneration.

9.
Acta Biomater ; 70: 84-97, 2018 04 01.
Article in English | MEDLINE | ID: mdl-29447961

ABSTRACT

3D scaffold-based in vitro cell culturing is a recent technological advancement in cancer research bridging the gap between conventional 2D culture and in vivo tumours. The main challenge in treating neuroblastoma, a paediatric cancer of the sympathetic nervous system, is to combat tumour metastasis and resistance to multiple chemotherapeutic drugs. The aim of this study was to establish a physiologically relevant 3D neuroblastoma tissue-engineered system and explore its therapeutic relevance. Two neuroblastoma cell lines, chemotherapeutic sensitive Kelly and chemotherapeutic resistant KellyCis83 were cultured in a 3D in vitro model on two collagen-based scaffolds containing either glycosaminoglycan (Coll-GAG) or nanohydroxyapatite (Coll-nHA) and compared to 2D cell culture and an orthotopic murine model. Both neuroblastoma cell lines actively infiltrated the scaffolds and proliferated displaying >100-fold increased resistance to cisplatin treatment when compared to 2D cultures, exhibiting chemosensitivity similar to orthotopic xenograft in vivo models. This model demonstrated its applicability to validate miRNA-based gene delivery. The efficacy of liposomes bearing miRNA mimics uptake and gene knockdown was similar in both 2D and 3D in vitro culturing models highlighting the proof-of-principle for the applicability of 3D collagen-based scaffolds cell system for validation of miRNA function. Collectively, this data shows the successful development and characterisation of a physiologically relevant, scaffold-based 3D tissue-engineered neuroblastoma cell model, strongly supporting its value in the evaluation of chemotherapeutics, targeted therapies and investigation of neuroblastoma pathogenesis. While neuroblastoma is the specific disease being focused upon, the platform may have multi-functionality beyond this tumour type. STATEMENT OF SIGNIFICANCE: Traditional 2D cell cultures do not completely capture the 3D architecture of cells and extracellular matrix contributing to a gap in our understanding of mammalian biology at the tissue level and may explain some of the discrepancies between in vitro and in vivo results. Here, we demonstrated the successful development and characterisation of a physiologically relevant, scaffold-based 3D tissue-engineered neuroblastoma cell model, strongly supporting its value in the evaluation of chemotherapeutics, targeted therapies and investigation of neuroblastoma pathogenesis. The ability to test drugs in this reproducible and controllable tissue-engineered model system will help reduce the attrition rate of the drug development process and lead to more effective and tailored therapies. Importantly, such 3D cell models help to reduce and replace animals for pre-clinical research addressing the principles of the 3Rs.


Subject(s)
Collagen/chemistry , Gene Transfer Techniques , Neuroblastoma , Tissue Scaffolds/chemistry , Xenograft Model Antitumor Assays , Animals , Cell Line, Tumor , Female , Heterografts , Humans , Mice , Mice, Nude , Neuroblastoma/genetics , Neuroblastoma/metabolism , Neuroblastoma/pathology , Neuroblastoma/therapy
10.
Surgeon ; 16(1): 55-65, 2018 Feb.
Article in English | MEDLINE | ID: mdl-28811169

ABSTRACT

Autologous gastrointestinal tissue has remained the gold-standard reconstructive biomaterial in urology for >100 years. Mucus-secreting epithelium is associated with lifelong metabolic and neuromechanical complications when implanted into the urinary tract. Therefore, the availability of biocompatible tissue-engineered biomaterials such as extracellular matrix (ECM) scaffolds may provide an attractive alternative for urologists. ECMs are decellularised, biodegradable membranes that have shown promise for repairing defective urinary tract segments in vitro and in vivo by inducing a host-derived tissue remodelling response after implantation. In urology, porcine small intestinal submucosa (SIS) and porcine urinary bladder matrix (UBM) are commonly selected as ECMs for tissue regeneration. Both ECMs support ingrowth of native tissue and differentiation of multi-layered urothelial and smooth muscle cells layers while providing mechanical support in vivo. In their native acellular state, ECM scaffolds can repair small urinary tract defects. Larger urinary tract segments can be repaired when ECMs are manipulated by seeding them with various cell types prior to in vivo implantation. In the present review, we evaluate and summarise the clinical potential of tissue engineered ECMs in reconstructive urology with emphasis on their long-term outcomes in urological clinical trials.


Subject(s)
Extracellular Matrix , Tissue Engineering/methods , Tissue Scaffolds , Urinary Tract/surgery , Humans , Tissue Engineering/trends , Tissue Scaffolds/trends
11.
Ir J Med Sci ; 187(1): 33-37, 2018 Feb.
Article in English | MEDLINE | ID: mdl-28478609

ABSTRACT

INTRODUCTION: The aim of this study was to compare the performance of preoperative risk nomograms or detecting lymph node invasion in a cohort of men undergoing radical prostatectomy (RP). METHODS: A retrospective analysis was performed on all men (n = 145) who underwent RP between 2012 and 2015. Preoperative data was inputted to the Memorial Sloan-Kettering Cancer Centre (MSKCC), Partin 2011 and Briganti nomograms and the University of California San Francisco- Centre of the Prostate Risk Assessment tool (UCSF-CAPRA). The risk of lymph node involvement (LNI) was calculated and compared to final histology. RESULTS: One hundred three (71%) men underwent a lymph node dissection at RP. Ten (9.7%) demonstrated LNI. The median nodal yield was 15 nodes, with no difference between those with LNI and those without (19.5 vs 14.5, p = 0.22).No patient classified as low risk on the UCSF-CAPRA score had evidence of LNI. In patients with LNI, no patient breached the 2% threshold for lymph node dissection (LND) on the MSKCC nomogram; four patients breached the 5% threshold on the Partin tables while three patients breached the 2.5% threshold for the Briganti nomogram. CONCLUSION: Nomograms produce useful information regarding risk of disease; however, they often have not been validated on different populations. Risk predictions need to be considered carefully and treatment decisions were made on a patient specific basis.


Subject(s)
Lymph Node Excision/methods , Lymphatic Metastasis/diagnosis , Nomograms , Prostatectomy/methods , Prostatic Neoplasms/diagnosis , Humans , Male , Middle Aged , Retrospective Studies , Risk Assessment
12.
Gene Ther ; 24(11): 681-691, 2017 11.
Article in English | MEDLINE | ID: mdl-28905887

ABSTRACT

Recent developments within the field of tissue engineering (TE) have shown that biomaterial scaffold systems can be augmented via the incorporation of gene therapeutics. The objective of this study was to assess the potential of the activated polyamidoamine dendrimer (dPAMAM) transfection reagent (SuperfectTM) as a gene delivery system to mesenchymal stem cells (MSCs) in both monolayer and 3D culture on collagen based scaffolds. dPAMAM-pDNA polyplexes at a mass ratio (M:R) 10:1 (dPAMAM : pDNA) (1 ug pDNA) were capable of facilitating prolonged reporter gene expression in monolayer MSCs which was superior to that facilitated using polyethylenimine (PEI)-pDNA polyplexes (2 ug pDNA). When dPAMAM-pDNA polyplexes (1 ug pDNA) were soak loaded onto a collagen-chondroitin sulphate (CS) scaffold prolonged transgene expression was facilitated which was higher than that obtained for a PEI-pDNA polyplex (2 ug pDNA) loaded scaffold. Transgene expression was dependent on the composite nature of the collagen scaffold with varying expression profiles obtained from a suite of collagen constructs including a collagen alone, collagen-CS, collagen-hydroxyapatite, collagen-nanohydroxyapatite and collagen-hyaluronic acid scaffold. Therefore, the dPAMAM vector described herein represents a biocompatible, effective gene delivery vector for TE applications which, via matching with a particular composite scaffold type, can be tailored for regeneration of various tissue defects.


Subject(s)
Dendrimers/metabolism , Tissue Engineering/methods , Transfection/methods , Animals , Biocompatible Materials , Collagen/metabolism , Dendrimers/chemistry , Dendrites/physiology , Durapatite/metabolism , Gene Transfer Techniques , Genetic Therapy/methods , Mesenchymal Stem Cells/metabolism , Plasmids , Polyethyleneimine/metabolism , Rats , Rats, Sprague-Dawley , Tissue Scaffolds
13.
Acta Biomater ; 55: 226-238, 2017 06.
Article in English | MEDLINE | ID: mdl-28363788

ABSTRACT

Controlling the phenotype of mesenchymal stem cells (MSCs) through the delivery of regulatory genes is a promising strategy in tissue engineering (TE). Essential to effective gene delivery is the choice of gene carrier. Non-viral delivery vectors have been extensively used in TE, however their intrinsic effects on MSC differentiation remain poorly understood. The objective of this study was to investigate the influence of three different classes of non-viral gene delivery vectors: (1) cationic polymers (polyethylenimine, PEI), (2) inorganic nanoparticles (nanohydroxyapatite, nHA) and (3) amphipathic peptides (RALA peptide) on modulating stem cell fate after reporter and therapeutic gene delivery. Despite facilitating similar reporter gene transfection efficiencies, these nanoparticle-based vectors had dramatically different effects on MSC viability, cytoskeletal morphology and differentiation. After reporter gene delivery (pGFP or pLUC), the nHA and RALA vectors supported an elongated MSC morphology, actin stress fibre formation and the development of mature focal adhesions, while cells appeared rounded and less tense following PEI transfection. These changes in MSC morphology correlated with enhanced osteogenesis following nHA and RALA transfection and adipogenesis following PEI transfection. When therapeutic genes encoding for transforming growth factor beta 3 (TGF-ß3) and/or bone morphogenic protein 2 (BMP2) were delivered to MSCs, nHA promoted osteogenesis in 2D culture and the development of an endochondral phenotype in 3D culture, while RALA was less osteogenic and appeared to promote a more stable hyaline cartilage-like phenotype. In contrast, PEI failed to induce robust osteogenesis or chondrogenesis of MSCs, despite effective therapeutic protein production. Taken together, these results demonstrate that the differentiation of MSCs through the application of non-viral gene delivery strategies depends not only on the gene delivered, but also on the gene carrier itself. STATEMENT OF SIGNIFICANCE: Nanoparticle-based non-viral gene delivery vectors have been extensively used in regenerative medicine, however their intrinsic effects on mesenchymal stem cell (MSC) differentiation remain poorly understood. This paper demonstrates that different classes of commonly used non-viral vectors are not inert and they have a strong effect on cell morphology, stress fiber formation and gene transcription in MSCs, which in turn modulates their capacity to differentiate towards osteogenic, adipogenic and chondrogenic lineages. These results also point to the need for careful and tissue-specific selection of nanoparticle-based delivery vectors to prevent undesired phenotypic changes and off-target effects when delivering therapeutic genes to damaged or diseased tissues.


Subject(s)
Gene Transfer Techniques , Materials Testing , Mesenchymal Stem Cells/metabolism , Nanoparticles/metabolism , Animals , Durapatite/chemistry , Durapatite/pharmacology , Mesenchymal Stem Cells/cytology , Peptides/chemistry , Peptides/pharmacology , Polyethyleneimine/chemistry , Polyethyleneimine/pharmacology , Swine
14.
Eur Cell Mater ; 33: 130-142, 2017 02 14.
Article in English | MEDLINE | ID: mdl-28197989

ABSTRACT

Limitations associated with demineralised bone matrix and other grafting materials have motivated the development of alternative strategies to enhance the repair of large bone defects. The growth plate (GP) of developing limbs contain a plethora of growth factors and matrix cues which contribute to long bone growth, suggesting that biomaterials derived from its extracellular matrix (ECM) may be uniquely suited to promoting bone regeneration. The goal of this study was to generate porous scaffolds from decellularised GP ECM and to evaluate their ability to enhance host mediated bone regeneration following their implantation into critically-sized rat cranial defects. The scaffolds were first assessed by culturing with primary human macrophages, which demonstrated that decellularisation resulted in reduced IL-1ß and IL-8 production. In vitro, GP derived scaffolds were found capable of supporting osteogenesis of mesenchymal stem cells via either an intramembranous or an endochondral pathway, demonstrating the intrinsic osteoinductivity of the biomaterial. Furthermore, upon implantation into cranial defects, GP derived scaffolds were observed to accelerate vessel in-growth, mineralisation and de novo bone formation. These results support the use of decellularised GP ECM as a scaffold for large bone defect regeneration.


Subject(s)
Bone Regeneration , Bone and Bones/pathology , Extracellular Matrix/metabolism , Growth Plate/metabolism , Tissue Scaffolds/chemistry , Wound Healing , Animals , Bone and Bones/diagnostic imaging , Chondrogenesis , Cytokines/biosynthesis , Glycosaminoglycans/metabolism , Growth Plate/ultrastructure , Humans , Macrophages/cytology , Male , Osteogenesis , Phenotype , Porosity , Rats, Inbred F344 , Skull/diagnostic imaging , Skull/pathology , Sus scrofa , X-Ray Microtomography
15.
Case Rep Oncol Med ; 2016: 5471862, 2016.
Article in English | MEDLINE | ID: mdl-27830100

ABSTRACT

A 37-year-old male presented with a traumatic injury to the scrotal region necessitating emergency surgery. Evacuation of a haematoma and bilateral orchidectomy were performed. A left sided nonseminomatous germ cell tumour (NSGCT), predominantly yolk sac, was identified. Microscopic margins were positive for tumour. Initial tumour markers revealed an AFP of 22,854 ng/mL, HCG of <1 mIU/mL, and LDH of 463 IU/L. Eight weeks after surgery, AFP levels remained elevated at 11,646 ng/mL. Computed tomography (CT) scanning demonstrated left inguinal adenopathy, 1.5 cm in max dimension. On review, extensive evidence of scrotal involvement was evident. His tumour was staged as stage IIIC, poor risk NSGCT. He was treated with 4 cycles of bleomycin, etoposide, and cisplatin over a 12-week period. His tumour markers normalised after 3 cycles. There was a marked improvement noted clinically. Follow-up CT scans demonstrated complete resolution of his tumour. He later underwent further surgery to remove a small amount of remaining spermatic cord. Histology revealed no malignant tissue. The patient suffered many complications including testosterone deficiency, osteopenia, infertility, and psychological distress. Discussion. A small proportion of testicular cancer may present in an atypical manner. The scrotum and testicle have markedly different embryonic origins and therefore a distinct anatomic separation. As a result the scrotum is not a typical site of spread of testicular cancer. Case reports have been described that were managed in a similar manner with good outcomes. Therefore, even with significant scrotal involvement, if timely and appropriate treatment is administered, complete resolution of the tumour may be achieved.

16.
J Antimicrob Chemother ; 71(12): 3372-3375, 2016 12.
Article in English | MEDLINE | ID: mdl-27566311

ABSTRACT

BACKGROUND: In Western Australia (WA), clonal complex 5, ST835, community-associated (CA) MRSA is isolated almost exclusively from aged care facilities. In WA four different staphylococcal cassette chromosome (SCC) mec (SCCmec) elements have been identified in this ST, indicating high genetic activity in the SCCmec region. OBJECTIVES: To investigate the SCC region of ST835 CA-MRSA WA MRSA-40 and determine the distribution of an SCCsorbitol element found within the region. RESULTS: The SCC region contained a composite island, SCCmecWA MRSA-40-CI, that was composed of three elements, ΨSCCpls, SCCsorbitol and SCCmecVT (5C2&5). This is the first time that a sorbitol operon has been reported in an SCC element. CONCLUSIONS: Generation of SCCmecWA MRSA-40-CI has involved multiple genetic events and recombination with CoNS has occurred during evolution of the SCC elements. While Staphylococcus aureus is renowned for its ability to utilize mobile genetic elements to disseminate antimicrobial resistance, the SCC region of WA MRSA-40 shows that this clone has also utilized SCC elements to acquire extra virulence and possibly adapt to a niche environment.


Subject(s)
Chromosomes, Bacterial , Community-Acquired Infections/microbiology , Genomic Islands , Homes for the Aged , Methicillin-Resistant Staphylococcus aureus/genetics , Nursing Homes , Staphylococcal Infections/microbiology , Aged , Evolution, Molecular , Genotype , Humans , Methicillin-Resistant Staphylococcus aureus/isolation & purification , Recombination, Genetic , Western Australia
17.
Acta Biomater ; 36: 55-62, 2016 05.
Article in English | MEDLINE | ID: mdl-26961807

ABSTRACT

UNLABELLED: Freshly isolated stromal cells can potentially be used as an alternative to in vitro expanded cells in regenerative medicine. Their use requires the development of bioactive hydrogels or scaffolds which provide an environment to enhance their proliferation and tissue-specific differentiation in vivo. The goal of the current study was to develop an injectable fibrin hydrogel functionalized with cartilage ECM microparticles and transforming growth factor (TGF)-ß3 as a putative therapeutic for articular cartilage regeneration. ECM microparticles were produced by cryomilling and freeze-drying porcine articular cartilage. Up to 2% (w/v) ECM could be incorporated into fibrin without detrimentally affecting its capacity to form stable hydrogels. To access the chondroinductivity of cartilage ECM, we compared chondrogenesis of infrapatellar fat pad-derived stem cells in fibrin hydrogels functionalized with either particulated ECM or control gelatin microspheres. Cartilage ECM particles could be used to control the delivery of TGF-ß3 to IFP-derived stem cells within fibrin hydrogels in vitro, and furthermore, led to higher levels of sulphated glycosaminoglycan (sGAG) and collagen accumulation compared to control constructs loaded with gelatin microspheres. In vivo, freshly isolated stromal cells generated a more cartilage-like tissue within fibrin hydrogels functionalized with cartilage ECM particles compared to the control gelatin loaded constructs. These tissues stained strongly for type II collagen and contained higher levels of sGAGs. These results support the use of fibrin hydrogels functionalized with cartilage ECM components in single-stage, cell-based therapies for joint regeneration. STATEMENT OF SIGNIFICANCE: An alternative to the use of in vitro expanded cells in regenerative medicine is the use of freshly isolated stromal cells, where a bioactive scaffold or hydrogel is used to provide an environment that enhances their proliferation and tissue-specific differentiation in vivo. The objective of this study was to develop an injectable fibrin hydrogel functionalized with cartilage ECM micro-particles and the growth factor TGF-ß3 as a therapeutic for articular cartilage regeneration. This study demonstrates that freshly isolated stromal cells generate cartilage tissue in vivo when incorporated into such a fibrin hydrogels functionalized with cartilage ECM particles. These findings open up new possibilities for in-theatre, single-stage, cell-based therapies for joint regeneration.


Subject(s)
Cartilage/physiology , Chondrogenesis , Extracellular Matrix/chemistry , Fibrin/chemistry , Hydrogels/chemistry , Regeneration , Animals , Cartilage/cytology , Female , Humans , Male , Stromal Cells/cytology , Stromal Cells/metabolism , Swine
18.
J Hand Surg Eur Vol ; 41(8): 809-14, 2016 Oct.
Article in English | MEDLINE | ID: mdl-26936746

ABSTRACT

UNLABELLED: We divided 21 flexor digitorum profundus tendons in the index, middle and ring fingers in seven cadaver hands into three groups. The tendons were cut in zone 2 and repaired using a 4-strand cruciate core suture repair with one of the following three materials in each group: (1) a knotless repair with a 2-0 bidirectional-barbed suture, which has similar tensile strength as a 4-0 non-barbed suture used in the other two groups; (2) a knotted locking repair with a non-barbed 4-0 conventional suture; and (3) a non-locking repair with a non-barbed 4-0 knotless suture. The repaired fingers were cyclically loaded through a simulated active range of motion to a 5 N load. We monitored and recorded the gap sizes at regular intervals during the test. The 2-0 bidirectional-barbed suture group and non-barbed suture groups developed gaps of 2.2 mm after 10 cycles and 2.4 mm after 20 cycles, respectively. Over 1000 cycles, the mean gaps were 3.2 mm in the 4-0 conventional suture group and 9.1 mm in the 2-0 bidirectional-barbed group. The tendons in the 2-0 bidirectional-barbed group gapped earlier, with statistically significant differences compared with those in the locking repair with a non-barbed 4-0 knotless suture group. The repair strength of the barbed suture technique was inferior to the cruciate repairs using a conventional 4-0 non-barbed suture tested in this cyclic-loading model. LEVEL OF EVIDENCE: Level V.


Subject(s)
Finger Injuries/surgery , Suture Techniques , Sutures , Tendon Injuries/surgery , Tensile Strength , Cadaver , Humans , Range of Motion, Articular
19.
Eur Cell Mater ; 30: 271-81, 2015 Nov 27.
Article in English | MEDLINE | ID: mdl-26614482

ABSTRACT

Osteoporosis is one of the most prevalent bone diseases worldwide and is characterised by high levels of bone turnover, a marked loss in bone mass and accumulation of microdamage, which leads to an increased fracture incidence that places a huge burden on global health care systems. Bisphosphonates have been used to treat osteoporosis and have shown great success in conserving bone mass and reducing fracture incidence. In spite of the existing knowledge of the in vivo responses of bone to bisphosphonates, the cellular responses to these drugs have yet to be fully elucidated. In vitro model systems that allow the decoupling of complex highly integrated events, such as bone remodelling, provide a tool whereby these biological processes may be studied in a more simplified context. This study firstly utilised an in vitro model system of bone remodelling and comprising all three major cell types of the bone (osteocytes, osteoclasts and osteoblasts), which was representative of the bone's capacity to sense microdamage and subsequently initiate a basic multicellular unit response. Secondly, this system was used to study the effect of two commonly utilised aminobisphosphonate treatments for osteoporosis, alendronate and zoledronate. We demonstrated that microinjury to osteocyte networks being treated with bisphosphonates modulates receptor activator of nuclear factor kappa-B ligand and osteoprotegerin activity, and subsequently osteoclastogenesis. Furthermore, bisphosphonates increased the osteogenic potential following microinjury. Thus, we have shown for the first time that bisphosphonates act at all three stages of bone remodelling, from microinjury to osteoclastogenesis and ultimately osteogenesis.


Subject(s)
Bone Density Conservation Agents/pharmacology , Bone Remodeling/drug effects , Bone and Bones/injuries , Diphosphonates/pharmacology , Imidazoles/pharmacology , Osteoblasts/drug effects , Osteoclasts/drug effects , Animals , Bone and Bones/cytology , Mice , Osteoblasts/cytology , Osteoclasts/cytology , Osteocytes/cytology , Osteocytes/drug effects , Osteogenesis/physiology , Osteoporosis/drug therapy , Zoledronic Acid
20.
Microbes Infect ; 17(6): 395-401, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25749709

ABSTRACT

Staphylococcus epidermidis is the leading etiologic agent of orthopaedic implant infection. Contamination of the implanted device during insertion allows bacteria gain entry into the sterile bone environment leading to condition known as osteomyelitis. Osteomyelitis is characterised by weakened bones associated with progressive bone loss. The mechanism through which S. epidermidis interacts with bone cells to cause osteomyelitis is poorly understood. We demonstrate here that S. epidermidis can bind to osteoblasts in the absence of matrix proteins. S. epidermidis strains lacking the cell wall protein SdrG had a significantly reduced ability to bind to osteoblasts. Consistent with this, expression of SdrG in Lactococcus lactis resulted in significantly increased binding to the osteoblasts. Protein analysis identified that SdrG contains a potential integrin recognition motif. αVß3 is a major integrin expressed on osteoblasts and typically recognises RGD motifs in its ligands. Our results demonstrate that S. epidermidis binds to recombinant purified αVß3, and that a mutant lacking SdrG failed to bind. Blocking αVß3 on osteoblasts significantly reduced binding to S. epidermidis. These studies are the first to identify a mechanism through which S. epidermidis binds to osteoblasts and potentially offers a mechanism through which implant infection caused by S. epidermidis leads to osteomyelitis.


Subject(s)
Bacterial Proteins/metabolism , Carrier Proteins/metabolism , Integrin alphaVbeta3/metabolism , Osteoblasts/metabolism , Staphylococcus epidermidis/growth & development , Carrier Proteins/immunology , Humans , Osteomyelitis/etiology , Osteomyelitis/immunology , Osteomyelitis/therapy , Protein Binding/immunology , Serine/antagonists & inhibitors , Serine/immunology , Staphylococcus epidermidis/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...