Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Theriogenology ; 210: 143-153, 2023 Oct 15.
Article in English | MEDLINE | ID: mdl-37499372

ABSTRACT

Gonocytes play an important role in early development of spermatogonial stem cells and fertility preservation to acquire more high quality gonocytes in vitro for further germ cell-related research and applications, it is necessarily needed to enrich and in vitro propagate gonocytes from cryopreserved bovine testicular tissues. This study aimed to investigate the isolation, enrichment, and colony formation of gonocytes in vitro for germ cell expansion from cryopreserved neonatal bovine testicular tissues. The effects of several different in vitro culture conditions, including seeding density, temperature, serum replacement and extracellular matrices were investigated for the maintenance, proliferation and formation of gonocyte colonies in vitro. Frozen/thawed two-week-old neonatal bovine testicular tissues were digested and gonocytes were enriched using a Percoll density gradient. Cell viability was accessed by trypan blue staining and cell apoptosis was evaluated by TUNEL assays. Gonocytes were identified and confirmed by immunofluorescence with the PGP9.5 germ cell marker and the OCT4 pluripotency marker while Sertoli cells were stained with vimentin. We found that neonatal bovine gonocytes were efficiently enriched by a 30%-40% Percoll density gradient (p < 0.05). No significant differences were detected between neonatal bovine testicular cells cultured at 34 °C or 37 °C. The formation of gonocyte colonies was observed in culture medium supplemented with knockout serum replacement (KSR), but not fetal bovine serum (FBS), at a seeding density higher than 5.0 × 104 cells/well. A greater number of gonocyte colonies were observed in culture plates coated with laminin (38.00 ± 6.24/well) and Matrigel (38.67 ± 3.78/well) when compared to plates coated with collagen IV and fibronectin (p < 0.05). In conclusion, bovine neonatal gonocytes were able to be efficiently isolated, enriched and maintained in gonocyte colonies in vitro; the development of this protocol provides vital information for the clinical translation of this technology and the future restoration of human fertility.


Subject(s)
Sertoli Cells , Testis , Male , Animals , Humans , Germ Cells , Cell Proliferation , Spermatogonia
2.
Polymers (Basel) ; 15(5)2023 Feb 24.
Article in English | MEDLINE | ID: mdl-36904392

ABSTRACT

In the search for a novel and scalable skin scaffold for wound healing and tissue regeneration, we fabricated a class of fibrin/polyvinyl alcohol (PVA) scaffolds using an emulsion templating method. The fibrin/PVA scaffolds were formed by enzymatic coagulation of fibrinogen with thrombin in the presence of PVA as a bulking agent and an emulsion phase as the porogen, with glutaraldehyde as the cross-linking agent. After freeze drying, the scaffolds were characterized and evaluated for biocompatibility and efficacy of dermal reconstruction. SEM analysis showed that the formed scaffolds had interconnected porous structures (average pore size e was around 330 µm) and preserved the nano-scale fibrous architecture of the fibrin. Mechanical testing showed that the scaffolds' ultimate tensile strength was around 0.12 MPa with an elongation of around 50%. The proteolytic degradation of scaffolds could be controlled over a wide range by varying the type or degree of cross-linking and by fibrin/PVA composition. Assessment of cytocompatibility by human mesenchymal stem cell (MSC) proliferation assays shows that MSC can attach, penetrate, and proliferate into the fibrin/PVA scaffolds with an elongated and stretched morphology. The efficacy of scaffolds for tissue reconstruction was evaluated in a murine full-thickness skin excision defect model. The scaffolds were integrated and resorbed without inflammatory infiltration and, compared to control wounds, promoted deeper neodermal formation, greater collagen fiber deposition, facilitated angiogenesis, and significantly accelerated wound healing and epithelial closure. The experimental data showed that the fabricated fibrin/PVA scaffolds are promising for skin repair and skin tissue engineering.

3.
Adv Wound Care (New Rochelle) ; 12(10): 574-600, 2023 10.
Article in English | MEDLINE | ID: mdl-36680749

ABSTRACT

Significance: Tissue-engineered artificial skin for clinical reconstruction can be regarded as an established practice. Bi-layered skin equivalents are available as established allogenic or autologous therapy, and various acellular skin replacements can support tissue repair. Moreover, there is considerable commonality between the skin and other soft tissue reconstruction products. This article presents an attempt to create a comprehensive global landscape review of advanced replacement materials and associated strategies for skin and soft tissue reconstruction. Recent Advances: There has been rapid growth in the number of commercial and pre-commercial products over the past decade. In this survey, 263 base products for advanced skin therapy have been identified, across 8 therapeutic categories, giving over 350 products in total. The largest market is in the United States, followed by the E.U. zone. However, despite these advances, and the investment of resources in each product development, there are key issues concerning the clinical efficacy, cost-benefit of products, and clinical impact. Each therapeutic strategy has relative merits and limitations. Critical Issues: A critical consideration in developing and evaluating products is the therapeutic modality, associated regulatory processes, and the potential for clinical adoption geographically, determined by regulatory territory, intellectual property, and commercial distribution factors. The survey identifies an opportunity for developments that improve basic efficacy or cost-benefit. Future Directions: The economic pressures on health care systems, compounded by the demands of our increasingly ageing population, and the imperative to distribute effective health care, create an urgent global need for effective and affordable products.


Subject(s)
Skin, Artificial , Tissue Engineering , Skin , Wound Healing , Skin Transplantation
4.
J Mater Sci Mater Med ; 29(6): 79, 2018 Jun 05.
Article in English | MEDLINE | ID: mdl-29872930

ABSTRACT

There are significant challenges for using emulsion templating as a method of manufacturing macro-porous protein scaffolds. Issues include protein denaturation by adsorption at hydrophobic interfaces, emulsion instability, oil droplet and surfactant removal after protein gelation, and compatible cross-linking methods. We investigated an oil-in-water macro-emulsion stabilised with a surfactant blend, as a template for manufacturing protein-based nano-structured bio-intelligent scaffolds (EmDerm) with tuneable micro-scale porosity for tissue regeneration. Prototype EmDerm scaffolds were made using either collagen, through thermal gelation, fibrin, through enzymatic coagulation or collagen-fibrin composite. Pore size was controlled via surfactant-to-oil phase ratio. Scaffolds were crosslink-stabilised with EDC/NHS for varying durations. Scaffold micro-architecture and porosity were characterised with SEM, and mechanical properties by tensiometry. Hydrolytic and proteolytic degradation profiles were quantified by mass decrease over time. Human dermal fibroblasts, endothelial cells and bone marrow derived mesenchymal stem cells were used to investigate cytotoxicity and cell proliferation within each scaffold. EmDerm scaffolds showed nano-scale based hierarchical structures, with mean pore diameters ranging from 40-100 microns. The Young's modulus range was 1.1-2.9 MPa, and ultimate tensile strength was 4-16 MPa. Degradation rate was related to cross-linking duration. Each EmDerm scaffold supported excellent cell ingress and proliferation compared to the reference materials Integra™ and Matriderm™. Emulsion templating is a novel rapid method of fabricating nano-structured fibrous protein scaffolds with micro-scale pore dimensions. These scaffolds hold promising clinical potential for regeneration of the dermis and other soft tissues, e.g., for burns or chronic wound therapies.


Subject(s)
Biocompatible Materials/chemistry , Collagen/chemistry , Tissue Engineering/methods , Tissue Scaffolds/chemistry , Bone Marrow Cells/cytology , Bone Regeneration , Cell Proliferation , Elastic Modulus , Elastin/chemistry , Emulsions , Fibrin/metabolism , Fibroblasts/cytology , Humans , Mesenchymal Stem Cells/cytology , Osteoblasts , Osteogenesis , Porosity , Stress, Mechanical , Tensile Strength , Wound Healing
5.
Biomed Res Int ; 2015: 108571, 2015.
Article in English | MEDLINE | ID: mdl-26504774

ABSTRACT

Mesenchymal stem cells (MSCs) have been shown to improve tissue regeneration in several preclinical and clinical trials. These cells have been used in combination with three-dimensional scaffolds as a promising approach in the field of regenerative medicine. We compare the behavior of human adipose-derived MSCs (AdMSCs) on four different biomaterials that are awaiting or have already received FDA approval to determine a suitable regenerative scaffold for delivering these cells to dermal wounds and increasing healing potential. AdMSCs were isolated, characterized, and seeded onto scaffolds based on chitosan, fibrin, bovine collagen, and decellularized porcine dermis. In vitro results demonstrated that the scaffolds strongly influence key parameters, such as seeding efficiency, cellular distribution, attachment, survival, metabolic activity, and paracrine release. Chick chorioallantoic membrane assays revealed that the scaffold composition similarly influences the angiogenic potential of AdMSCs in vivo. The wound healing potential of scaffolds increases by means of a synergistic relationship between AdMSCs and biomaterial resulting in the release of proangiogenic and cytokine factors, which is currently lacking when a scaffold alone is utilized. Furthermore, the methods used herein can be utilized to test other scaffold materials to increase their wound healing potential with AdMSCs.


Subject(s)
Mesenchymal Stem Cell Transplantation/instrumentation , Mesenchymal Stem Cells/cytology , Skin, Artificial , Tissue Scaffolds , Wounds and Injuries/pathology , Wounds and Injuries/therapy , Cell Adhesion/physiology , Cell Proliferation/physiology , Cell Survival/physiology , Cells, Cultured , Equipment Design , Equipment Failure Analysis , Humans , Materials Testing , Mesenchymal Stem Cells/physiology
6.
Biochim Open ; 1: 6-10, 2015.
Article in English | MEDLINE | ID: mdl-29632825

ABSTRACT

Commercially available two component human fibrin sealants are commonly used to manufacture human fibrin-based biomaterials. However, this method is costly and allows little room for further tuning of the biomaterial. Human fibrinogen solutions offer a more cost-effective and versatile alternative to manufacture human fibrin-based biomaterials. Yet, human fibrinogen is highly unstable and contains certain impurities like human albumin. Within the context of biomaterials and tissue engineering we offer a simple yet novel solution based on classical biochemical techniques to significantly reduce albumin in human fibrinogen solutions. This method can be used for various tissue engineering and biomedical applications as an initial step in the manufacturing of human fibrin-based biomaterials to optimise their regenerative application.

7.
Biochim Open ; 1: 40-50, 2015.
Article in English | MEDLINE | ID: mdl-29632828

ABSTRACT

Composite biomaterials made from synthetic and protein-based polymers are extensively researched in tissue engineering. To successfully fabricate a protein-polymer composite, it is critical to understand how strongly the protein binds to the synthetic polymer, which occurs through protein adsorption. Currently, there is no cost-effective and simple method for characterizing this interfacial binding. To characterize this interfacial binding, we introduce a simple three-step method that involves: 1) synthetic polymer surface characterisation, 2) a quick, inexpensive and robust novel immuno-based assay that uses protein extraction compounds to characterize protein binding strength followed by 3) an in vitro 2D model of cell culture to confirm the results of the immuno-based assay. Fibrinogen, precursor of fibrin, was adsorbed (test protein) on three different polymeric surfaces: silicone, poly(acrylic acid)-coated silicone and poly(allylamine)-coated silicone. Polystyrene surface was used as a reference. Characterisation of the different surfaces revealed different chemistry and roughness. The novel immuno-based assay showed significantly stronger binding of fibrinogen to both poly(acrylic acid) and poly(allylamine) coated silicone. Finally, cell studies showed that the strength of the interaction between the protein and the polymer had an effect on cell growth. This novel immuno-based assay is a valuable tool in developing composite biomaterials of synthetic and protein-based polymers with the potential to be applied in other fields of research where protein adsorption onto surfaces plays an important role.

8.
Anal Biochem ; 470: 22-4, 2015 Feb 01.
Article in English | MEDLINE | ID: mdl-25447464

ABSTRACT

Recent studies show that apoptosis affects surrounding tissue, playing a role in diseases such as fibrosis, a significant global disease burden. Elucidating the mechanisms by which the different apoptotic cells present during fibrotic wound healing affect their environment would enable development of new therapies. We describe here a simple, rapid, and cost-effective method for inducing apoptosis of primary normal human dermal fibroblasts without affecting the overall cell viability of the population. Such population could be used for in vitro models of fibrotic wound healing in co-culture with other cells involved in this process to study events such as apoptosis-induced proliferation.


Subject(s)
Apoptosis , Cytological Techniques/methods , Fibroblasts/cytology , Skin/cytology , Cell Proliferation , Cost-Benefit Analysis , Cytological Techniques/economics , Fibroblasts/pathology , Fibrosis , Humans , Time Factors
9.
Biomed Mater Eng ; 24(2): 1457-68, 2014.
Article in English | MEDLINE | ID: mdl-24642973

ABSTRACT

PURPOSE: The yield of a critical number of basal epithelial cells with high mitotic rates from native tissue is a challenge in the field of tissue engineering. There are many protocols that use enzymatic methods for isolation of epithelial cells with unsatisfactory results for tissue engineering. This study aimed to develop a protocol for isolating a sufficient number of epithelial cells with a high Proliferating Index from ovine esophagus for tissue engineering applications. METHODS: Esophageal mucosa was pretreated with dispase-collagenase solution and plated on collagen-coated culture dishes. Distinction of the various types of epithelial cells and developmental stages was done with specific primary antibodies to Cytokeratins and to Proliferating Cell Nuclear Antigen (PCNA). RESULTS: Up to approximately 8100 epithelial cells/mm2 of mucosa tissue were found after one week of migration. Cytokeratin 14 (CK 14) was positive identified in cells even after 83 days. At the same time the Proliferating Index was 71%. CONCLUSION: Our protocol for isolation of basal epithelial cells was successful to yield sufficient numbers of cells predominantly with proliferative character and without noteworthy negative enzymatic affection. The results at this study offer the possibility of generation critical cell numbers for tissue engineering applications.


Subject(s)
Cell Separation/methods , Epithelial Cells/cytology , Esophagus/cytology , Animals , Cell Count , Cell Proliferation , Cells, Cultured , Keratin-14/analysis , Sheep , Tissue Engineering
10.
Biores Open Access ; 2(6): 412-20, 2013 Dec 01.
Article in English | MEDLINE | ID: mdl-24380051

ABSTRACT

The clinical demand for effective dermal substitutes continues as current commercially available products present limitations. However, there are no definitive in vitro methods to predict in vivo outcomes such as integration, cellularization and contraction, which may help the development of new dermal scaffolds. This study aimed to develop a multiparameter in vitro model of three-dimensional (3D) cell ingress into dermal scaffolds to predict in vivo outcomes of new dermal scaffolds under development. A new dermal scaffold, Smart Matrix, was compared to the scar-forming contractile collagen gel model and to the clinically well-established Integra(®) and Matriderm(®). Parameters studied were cell viability and proliferation, apoptosis, matrix contraction, cell morphology, α-smooth muscle actin, and growth factor expression. Combinatorial evaluation of the results in a scoring matrix showed that Smart Matrix could offer an advantage over existing products. This method would be useful as an international golden scoring matrix to develop new dermal scaffolds that effectively improve the existing products, thus enabling better treatments for burns or chronic wounds.

11.
Biomaterials ; 32(29): 7096-105, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21741704

ABSTRACT

Severe skin loss constitutes a major unsolved clinical problem worldwide. For this reason, in the last decades there has been a major push towards the development of novel therapeutic approaches to enhance skin wound healing. Neo-vessel formation through angiogenesis is a critical step during the wound healing process. Besides the contribution of pre-existing endothelial cells (EC), endothelial progenitor cells (EPCs) have also been implicated in wound healing acting either by differentiating into EC that incorporate the neo-vessels, or via the production of paracrine factors that improve angiogenesis. Here we tested the importance of different extracellular matrices (ECM) in regulating the angiogenic and wound healing potential of cord blood-derived EPC (CB-EPC). We compared the properties of several ECM and particularly of fibrin fragment E (FbnE) in regulating EPC adhesion, proliferation, differentiation and healing-promotion in vitro and in vivo. Our results show that CB-EPCs have increased adhesion and endothelial differentiation when plated on FbnE compared to collagens, fibronectin or fibrin. Using integrin neutralizing antibodies, we show that CB-EPC adhesion to FbnE is mediated by integrin α5ß1. Gene expression analysis of CB-EPCs plated on different substrates revealed that CB-EPC grown on FbnE shows increased expression of paracrine factors such as VEGF-A, TGF-ß1, SDF-1, IL-8 and MIP-1α. Accordingly, conditioned media from CB-EPC grown on FbnE induced EC tube formation and monocyte migration in vitro. To test the wound healing effects of FbnE in vivo we used an FbnE enriched scaffold in a cutaneous wound healing mouse model. In accordance with our in vitro data, co-administration of the FbnE enriched scaffold with CB-EPC significantly accelerated wound closure and wound vascularization, compared FbnE enriched scaffold alone or to using collagen-based scaffolds. Our results show that FbnE modulates several CB-EPC properties in vivo and in vitro, and as such promotes wound healing. We suggest the use of FbnE-based scaffolds represents a promising approach to resolve wound healing complications arising from different pathologies.


Subject(s)
Cell Adhesion/physiology , Cell Differentiation/physiology , Endothelial Cells/physiology , Fibrin/metabolism , Stem Cells/physiology , Wound Healing/physiology , Animals , Biomarkers/metabolism , Cell Proliferation , Cells, Cultured , Endothelial Cells/cytology , Extracellular Matrix/metabolism , Female , Humans , Mice , Mice, Inbred BALB C , Mice, SCID , Neovascularization, Physiologic/physiology , Protein Isoforms/metabolism , Stem Cells/cytology , Vascular Endothelial Growth Factor A/metabolism
12.
J Biomed Mater Res A ; 96(3): 552-65, 2011 Mar 01.
Article in English | MEDLINE | ID: mdl-21254387

ABSTRACT

Calcium ions are utilized in biomolecular biomaterial design for osteomimetic scaffolds and as divalent cross-linking agents, typically for gelation of alginates, stabilisation of protein structure (e.g., fibrinogen) and enzyme activation (e.g., thrombin). Biological interactions with defined calcium phosphates (e.g., hydroxyapatite) are exploited for osteogenesis, although crystalline calcium phosphates (e.g., calcium pyrophosphate) stimulate inflammation. We found that the calcium concentration used in the manufacture of prototype dermal scaffolds made from fibrin/alginate composite was related to the inflammatory infiltration during in vivo integration. In investigating a cause for this inflammatory response, we have identified and characterized a cytolytic inflammatory effect of amorphous calcium phosphate (CaP) formed in physiological solutions, relevant to biomaterial biocompatibility. Isolated human neutrophils (Nφ) were incubated in phosphate-buffered saline with CaCl(2) ranging 2.5-20 mM total calcium. Nφ activation was assessed by morphology and integrin-ß2 (CD18a) expression. Mediator release (Nφ-elastase, IL-8, and TNFα) was measured from both Nφ and whole blood cultures plus CaCl(2). CaP exposure increased CD18a expression over 1 h (maximal at 10 mM calcium/ phosphate) with concurrent phagocytosis, cytolysis, and Nφ-elastase release. CaCl(2) induced expression of IL-8 and TNFα in whole blood cultures. These results suggest that CaP formed from the resorption of calcium-containing biomaterials could induce inflammation and accelerate biomaterial degradation, driving further CaP release. This demonstrates a novel mechanism for biomaterial-induced inflammation. The in vitro system described could aid preclinical evaluation of novel biomaterial inflammatory potential.


Subject(s)
Biocompatible Materials/adverse effects , Calcium Phosphates/pharmacology , Cytotoxicity, Immunologic/drug effects , Inflammation/pathology , Models, Biological , Neutrophil Activation/drug effects , Alginates/pharmacology , Buffers , CD18 Antigens/metabolism , Calcium/pharmacology , Dermis/drug effects , Fibrin/pharmacology , Glucuronic Acid/pharmacology , Hexuronic Acids/pharmacology , Humans , Inflammation Mediators/metabolism , Leukocyte Elastase/metabolism , Magnesium/pharmacology , Neutrophils/cytology , Neutrophils/drug effects , Neutrophils/immunology , Neutrophils/ultrastructure , Phagocytosis/drug effects , Phosphates/pharmacology , Spectrometry, X-Ray Emission , Time Factors , Tissue Scaffolds/chemistry
13.
Biomaterials ; 30(30): 5918-26, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19651436

ABSTRACT

Clinical success in tissue regeneration requires improvements in vascularization capacity of scaffolds. Several efforts have been made in this field including cellular and acellular technologies. In this work we combined the use of stem cells derived from pancreas or submandibular glands expressing green fluorescent protein (GFP(+)) with a commercially available scaffold for dermal regeneration. Cells were isolated, characterized and seeded in a scaffold for dermal regeneration. Scaffolds containing cells were used to induce dermal regeneration in a full skin defect model. After 3 weeks of in vivo regeneration, tissues were harvested and vascularization was analyzed. Results showed that gland-derived stem cells displayed stem cell features and presented multipotential differentiation capacity because they were able to differentiate in cell types representing the 3 different germ layers. After seeding, cells were homogeneously distributed and formed focal adhesions with the scaffold. Metabolic assays showed that cells can be cultured for at least 3 weeks in the scaffold. In vivo, the presence of pancreatic or submandibular stem cells significantly enhanced the vascularization compared to empty scaffolds. Presence of gland-derived stem cells in the regenerating tissue was confirmed by the detection of GFP expression in the wound area. In order to explore the possible mechanisms behind the improvement in vascular regeneration, in vitro experiments were performed, showing that gland-derived stem cells could contribute by angiogenic and vasculogenic mechanisms to this process. Our results suggest that the combined use of stem cells derived from glands and scaffold for dermal regeneration could be a rational alternative to improve vascularization in scaffold-mediated dermal regeneration.


Subject(s)
Biocompatible Materials/chemistry , Regeneration , Skin/pathology , Stem Cells/cytology , Tissue Scaffolds , Animals , Blood Vessels/pathology , Cell Differentiation , Cell Survival , Collagen/chemistry , Dermis/pathology , Drug Combinations , Green Fluorescent Proteins/chemistry , Laminin/chemistry , Mice , Mice, Inbred C57BL , Proteoglycans/chemistry , Skin/metabolism , Tissue Engineering/methods
14.
Ann Plast Surg ; 62(1): 92-6, 2009 Jan.
Article in English | MEDLINE | ID: mdl-19131729

ABSTRACT

Topical negative pressure is an effective technique to promote wound healing and the integration of skin graft and synthetic dermal equivalents. We describe an in vitro model to investigate the effect of negative pressure on angiogenesis, a pivotal step. Dermal fibroblasts or human microvascular endothelial cells were cultured on Integra and subjected to intermittent or continuous negative pressure. At fixed intervals of over 120 hours, the Integra was fixed and assessed for cell migration (microscopy), cell viability (MTS assay), and cell proliferation (Ki67 immunostaining). Under control conditions, endothelial cells formed a monolayer and failed to ingress, whereas fibroblasts migrated throughout the Integra within 24 hours. Negative pressure switches endothelial cell to a migratory and proliferative phenotype. Ingress is greatest with intermittent rather than continuous negative pressure. It has no effect on dermal fibroblast function. This study identifies an important, potential pro-angiogenic mechanism by which topical negative pressure promotes wound healing.


Subject(s)
Cell Movement , Cell Proliferation , Chondroitin Sulfates , Collagen , Endothelial Cells/physiology , Negative-Pressure Wound Therapy , Wound Healing , Cells, Cultured , Endothelium, Vascular/cytology , Fibroblasts/physiology , Humans
15.
Crit Care Med ; 36(5): 1594-602, 2008 May.
Article in English | MEDLINE | ID: mdl-18434887

ABSTRACT

OBJECTIVE: Polydeoxyribonucleotide contains a mixture of nucleotides and interacts with adenosine receptors, stimulating vascular endothelial growth factor expression and wound healing. The purpose of this study was to investigate the effect of polydeoxyribonucleotide on experimental burn wounds. DESIGN: Randomized experiment. SETTING: Research laboratory at a university hospital. SUBJECTS: Thermal injury in mice. INTERVENTIONS: Mice were immersed in 80 degrees C water for 10 secs to achieve a deep-dermal second-degree burn. Animals were randomized to receive either polydeoxyribonucleotide (8 mg/kg/day intraperitoneally for 14 days) or its vehicle alone (0.9% NaCl solution at 100 microL/day intraperitoneally). On days 7 and 14 the animals were killed. Blood was collected for tumor necrosis factor-alpha measurement; burn areas were used for histologic and immunohistochemical examination, for the evaluation of vascular endothelial growth factor and nitric oxide synthases by Western blot, and for the determination of wound nitric oxide products. MEASUREMENTS AND MAIN RESULTS: Polydeoxyribonucleotide increased burn wound re-epithelialization and reduced the time to final wound closure. Polydeoxyribonucleotide improved healing of burn wound through increased epithelial proliferation and maturation of the extracellular matrix as confirmed by fibronectin and laminin immunostaining. Polydeoxyribonucleotide also improved neoangiogenesis as suggested by the marked increase in microvessel density and by the robust expression of platelet-endothelial cell adhesion molecule-1. Furthermore, polydeoxyribonucleotide blunted serum tumor necrosis factor-alpha and enhanced inducible nitric oxide synthase and vascular endothelial growth factor expression and the wound content of nitric oxide products. CONCLUSIONS: Our study suggests that polydeoxyribonucleotide may be an effective therapeutic approach to improve clinical outcomes after thermal injury.


Subject(s)
Burns/drug therapy , Polydeoxyribonucleotides/therapeutic use , Wound Healing/drug effects , Animals , Male , Mice , Mice, Inbred C57BL , Neovascularization, Physiologic/drug effects
16.
Burns ; 34(2): 164-74, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18242874

ABSTRACT

BACKGROUND: The use of synthetic dermal replacements (SDRs) in the treatment of large wounds, which have associated morbidity and mortality, has attracted great interest. However, because of poor outcome, SDRs have limited use. The addition of topical negative pressure (TNP) has increased their success, but little research has focused on the underlying mechanisms. This paper studies the in vitro effects of TNP on commonly used SDRs to identify the most effective TNP regimen and optimum SDR for encouraging endothelial cell ingress. METHODS: Endothelial cells were co-cultured in vitro on four SDRs with or without TNP. Negative pressure (125mmHg) was applied intermittently, continuously, for 4h per day, or not at all. Endothelial ingress was measured for each condition. RESULTS: In the collagen controls, cell migration was minimal. Integratrade mark gave the greatest endothelial cell migration (p<0.05, n=3). TNP increased endothelial cell migration, intermittent application being the optimum regimen. CONCLUSIONS: Integratrade mark has an open sponge structure which may account for greater angiogenicity than Allodermtrade mark, Permacoltrade mark and Xenodermtrade mark. In vitro intermittent TNP stimulates the greatest angiogenic response. The majority of clinical studies investigating SDR success with TNP have used continuous regimens; this study suggests a change in clinical practice to intermittent application.


Subject(s)
Burns/therapy , Dermis/blood supply , Negative-Pressure Wound Therapy/methods , Neovascularization, Physiologic/physiology , Skin, Artificial , Humans , Skin Diseases, Bacterial/prevention & control , Stress, Mechanical
17.
Plast Reconstr Surg ; 117(6): 1876-85, 2006 May.
Article in English | MEDLINE | ID: mdl-16651960

ABSTRACT

BACKGROUND: Acute and chronic wounds are costly and invariably expose significant structures. Surgical reconstruction causes donor-site morbidity, scarring, and the need for intensive care. Reconstruction using an artificial dermis avoids donor sites, but available collagen-based solutions are susceptible to poor take. Using an in vitro angiogenic assay, the authors investigated dermal matrices for potential inclusion in a second-generation proangiogenic synthetic dermal replacement. METHODS: Human placental endothelial cells were cocultured on Cytodex beads (Pharmacia Biotech) and plated in eight different extracellular matrix gels (collagen, fibrin, four glycosaminoglycans, vitronectin, and fibronectin), with or without stimulation from two soluble angiogenic factors. Three different cell lines were used, with 30 beads per condition. Cellular invasion into gels was calculated using Sigma Scan computer software, and statistical comparisons were made. RESULTS: The authors found that fibrin provided greatest stimulus for endothelial invasion, with invasion in fibrin inhibited by collagen in a concentration-dependent fashion. Invasion by alternative extracellular matrix components and soluble angiogenic factors was far less than that in fibrin. CONCLUSIONS: The authors identified that extracellular matrices can provide greater angiogenic potential than soluble angiogenic factors. Fibrin provided a better proangiogenic scaffold than collagen. This could well be used to encourage blood vessel ingress and eventual take of a second-generation proangiogenic synthetic dermal replacement.


Subject(s)
Endothelial Cells/cytology , Endothelium, Vascular/cytology , Extracellular Matrix/physiology , Neovascularization, Physiologic , Skin, Artificial , Angiogenesis Inducing Agents/pharmacology , Cell Culture Techniques/instrumentation , Cell Culture Techniques/methods , Cell Movement/drug effects , Chondroitin Sulfates , Collagen , Culture Media , Dermatan Sulfate , Endothelial Cells/drug effects , Female , Fibrin , Fibronectins , Gels , Glycosaminoglycans , Humans , Microspheres , Neovascularization, Physiologic/drug effects , Placenta/blood supply , Pregnancy , Vascular Endothelial Growth Factor A/pharmacology , Vitronectin
SELECTION OF CITATIONS
SEARCH DETAIL
...