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










Publication year range
1.
Methods Mol Biol ; 2225: 93-105, 2021.
Article in English | MEDLINE | ID: mdl-33108659

ABSTRACT

In vivo wound healing models are predictive preclinical tests for therapeutics that enhance skin repair or limit scarring. Large animals, such as swine, heal in a manner similar to humans, but testing is impractical and expensive. Experiments in mice are more economic, but may be less translatable as this species heals primarily through contraction, not by the processes of epithelialization and granulation tissue formation as seen in human wounds. Here, we describe a murine model of thermal burn injury that closely mimics human healing, resulting in a large, hypertrophic-like scar. This practical, reproducible model is ideal for testing promising wound-healing therapies, such as virus-derived growth factors and immune-modulatory proteins.


Subject(s)
Burns/pathology , Cicatrix/prevention & control , Disease Models, Animal , Re-Epithelialization/genetics , Animals , Burns/genetics , Burns/therapy , Cicatrix/genetics , Cicatrix/pathology , Female , Gene Expression , Hot Temperature , Humans , Immunologic Factors/biosynthesis , Immunologic Factors/genetics , Immunologic Factors/pharmacology , Intercellular Signaling Peptides and Proteins/biosynthesis , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/pharmacology , Mice , Mice, Inbred C57BL , Re-Epithelialization/drug effects , Skin/drug effects , Skin/injuries , Transgenes , Viruses/genetics
2.
Front Vet Sci ; 7: 577835, 2020.
Article in English | MEDLINE | ID: mdl-33195583

ABSTRACT

Fibroproliferative disorders occur in both humans and horses following skin injury. In horses, wound healing on the limb is often complicated by the formation of fibroproliferative exuberant granulation tissue, characterized by persistent expression of pro-fibrotic transforming growth factor-beta1 (TGF-ß1) and deficient expression of anti-inflammatory interleukin-10 (IL-10). IL-10 has been shown to directly modulate fibrotic gene expression in human fibroblasts, so we hypothesized that equine IL-10 (eIL-10) may exert similar anti-fibrotic effects on equine dermal fibroblasts. Cell-lines were created from the limb skin of six individual horses. Recombinant eIL-10 was produced and purified, and its effects on the cells investigated in the presence and absence of equine TGF-ß1 (eTGF-ß1). Myofibroblast differentiation and collagen production were examined using immunofluorescent cytometry, cell contractility in a collagen gel assay, and fibrotic gene expression using quantitative PCR. In response to eTGF-ß1, fibroblasts increased in contractility and expression of alpha-smooth muscle actin, collagen types 1 and 3, and matrix metalloproteinase 1, 2, and 9. Equine IL-10 limited cell contractility and production of alpha-smooth muscle actin and type 3 collagen, and decreased mRNA levels of eCol3a1 and eMMP9, while increasing that of eMMP1. Opposing effects on eTGF-ßR3 and eIL-10R1 gene expression were also observed, with mRNA levels decreasing following eTGF-ß1 treatment, and increasing with eIL-10 treatment. These findings indicate that eIL-10 limits the pro-fibrotic effects of eTGF-ß1, potentially through the modulation of fibrotic and receptor gene expression. Further investigations are warranted to assess the therapeutic utility of eIL-10 in the treatment of exuberant granulation tissue.

3.
J Clin Med ; 9(4)2020 Apr 11.
Article in English | MEDLINE | ID: mdl-32290480

ABSTRACT

Orf virus (OV) is a zoonotic parapoxvirus that causes highly proliferative skin lesions which resolve with minimal inflammation and scarring. OV encodes two immunomodulators, vascular endothelial growth factor (VEGF)-E and interleukin-10 (ovIL-10), which individually modulate skin repair and inflammation. This study examined the effects of the VEGF-E and ovIL-10 combination on healing processes in a murine wound model. Treatments with viral proteins, individually and in combination, were compared to a mammalian VEGF-A and IL-10 combination. Wound biopsies were harvested to measure re-epithelialisation and scarring (histology), inflammation, fibrosis and angiogenesis (immunofluorescence), and gene expression (quantitative polymerase chain reaction). VEGF-E and ovIL-10 showed additive effects on wound closure and re-epithelialisation, and suppressed M1 macrophage and myofibroblast infiltration, while allowing M2 macrophage recruitment. The viral combination also increased endothelial cell density and pericyte coverage, and improved collagen deposition while reducing the scar area. The mammalian combination showed equivalent effects on wound closure, re-epithelialisation and fibrosis, but did not promote blood vessel stabilisation or collagen remodeling. The combination treatments also differentially altered the expression of transforming growth factor beta isoforms, Tgfß1 and Tgfß3. These findings show that the OV proteins synergistically enhance skin repair, and act in a complimentary fashion to improve scar quality.

4.
Adv Wound Care (New Rochelle) ; 7(8): 283-297, 2018 Aug 01.
Article in English | MEDLINE | ID: mdl-30087804

ABSTRACT

Objective: Vascular endothelial growth factor (VEGF) family members are critical regulators of tissue repair and depending on their distinct pattern of receptor specificity can also promote inflammation and scarring. This study utilized a receptor-selective VEGF to examine the role of VEGF receptor (VEGFR)-2 in scar tissue (ST) formation. Approach: Cutaneous skin wounds were created in mice using a 4 mm biopsy punch and then treated until closure with purified VEGF-E derived from orf virus stain NZ-2. Tissue samples were harvested to measure gene expression using quantitative PCR and to observe ST formation through histological examination and changes in cell populations by immunofluorescence. Results: VEGFR-2-activation with VEGF-E increased expression of anti-inflammatory cytokine interleukin (IL)-10 and reduced macrophage infiltration and myofibroblast differentiation in wounded skin compared with controls. VEGF-E treatment also increased microvascular density and improved pericyte coverage of blood vessels in the healing wounds. The ST that formed following treatment with VEGF-E was reduced in size and showed improved collagen structure. Innovation: The role of VEGFR-2 activation in wound epithelialization and angiogenesis is well established; but its contribution to ST formation is unclear. This study tests the effect of a selective VEGFR-2 activation on ST formation following cutaneous wounding in a murine model. Conclusion: VEGFR-2 stimulation can enhance the quality of skin repair, at least, in part, through the induction of IL-10 expression and dampening of wound inflammation and fibrosis. Therapies that selectively activate VEGFR-2 may therefore be beneficial to treat impaired healing or to prevent excess scarring.

5.
PLoS One ; 13(5): e0197223, 2018.
Article in English | MEDLINE | ID: mdl-29763436

ABSTRACT

Bandaging of limb wounds in horses leads to formation of exuberant granulation tissue (EGT) that retards healing due to protracted inflammation, aberrant vascularisation and delayed epithelialisation. EGT is not observed if wounds are left undressed or when wounds are on the body. A previous study showed that short-term administration of proteins derived from orf virus dampened inflammation and promoted epithelialisation of open wounds in horses. Here, we investigated the impact of orf virus interleukin-10 and vascular endothelial growth factor-E on the development and resolution of EGT. Excisional wounds were created on the forelimb of four horses, and bandages were maintained until full healing to induce EGT formation. Matching body wounds were created to ensure EGT was limited to the limb, and to differentiate the effects of the viral proteins on normal healing and on EGT formation. Viral proteins or the hydrogel vehicle control were administered topically to site-matched wounds at day 1, with repeat administration at day 8. Wound healing and EGT formation were monitored macroscopically. Wound margin samples were harvested at 2, 7 and 14 days, and at full healing, with histology used to observe epithelialisation, immunofluorescence used to detect inflammatory cells, angiogenesis and cell death, and qPCR to measure expression of genes regulating inflammation and angiogenesis. Limb wounds developed EGT, and exhibited slower healing than body wounds. Viral protein treatment did not accelerate healing at either location nor limit EGT formation in limb wounds. Treatment of limb wounds did however increase epithelialisation and angiogenesis, without dampening inflammatory cell infiltration or gene expression. The healed wounds also had less occlusion and death of blood vessels and fewer epidermal rete ridges following viral protein treatment. These findings indicate that the viral protein treatment does not suppress wound inflammation or EGT formation, but does promote vascular and epidermal repair and EGT resolution.


Subject(s)
Hindlimb , Horses , Hydrogels/pharmacology , Interleukin-10/pharmacology , Viral Proteins/pharmacology , Wound Healing/drug effects , Wounds and Injuries , Animals , Hindlimb/metabolism , Hindlimb/pathology , Wounds and Injuries/drug therapy , Wounds and Injuries/metabolism , Wounds and Injuries/pathology
6.
PLoS One ; 11(12): e0168007, 2016.
Article in English | MEDLINE | ID: mdl-27936239

ABSTRACT

Bovine papular stomatitis virus (BPSV) is a Parapoxvirus that induces acute pustular skin lesions in cattle and is transmissible to humans. Previous studies have shown that BPSV encodes a distinctive chemokine-binding protein (CBP). Chemokines are critically involved in the trafficking of immune cells to sites of inflammation and infected tissue, suggesting that the CBP plays a role in immune evasion by preventing immune cells reaching sites of infection. We hypothesised that the BPSV-CBP binds a wide range of inflammatory chemokines particularly those involved in BPSV skin infection, and inhibits the recruitment of immune cells from the blood into inflamed skin. Molecular analysis of the purified protein revealed that the BPSV-CBP is a homodimeric polypeptide with a MW of 82.4 kDa whilst a comprehensive screen of inflammatory chemokines by surface plasmon resonance showed high-affinity binding to a range of chemokines within the CXC, CC and XC subfamilies. Structural analysis of BPSV-CBP, based on the crystal structure of orf virus CBP, provided a probable explanation for these chemokine specificities at a molecular level. Functional analysis of the BPSV-CBP using transwell migration assays demonstrated that it potently inhibited chemotaxis of murine neutrophils and monocytes in response to CXCL1, CXCL2 as well as CCL2, CCL3 and CCL5 chemokines. In order to examine the effects of CBP in vivo, we used murine skin models to determine its impact on inflammatory cell recruitment such as that observed during BPSV infection. Intradermal injection of BPSV-CBP blocked the influx of neutrophils and monocytes in murine skin in which inflammation was induced with lipopolysaccharide. Furthermore, intradermal injection of BPSV-CBP into injured skin, which more closely mimics BPSV lesions, delayed the influx of neutrophils and reduced the recruitment of MHC-II+ immune cells to the wound bed. Our findings suggest that the CBP could be important in pathogenesis of BPSV infections.


Subject(s)
Chemokines/metabolism , Chemotaxis, Leukocyte/physiology , Disease Models, Animal , Inflammation/pathology , Monocytes/pathology , Neutrophils/pathology , Parapoxvirus/metabolism , Viral Proteins/physiology , Wounds and Injuries/pathology , Amino Acid Sequence , Animals , Dimerization , Mice , Protein Conformation , Sequence Homology, Amino Acid , Surface Plasmon Resonance , Viral Proteins/chemistry
7.
Wound Repair Regen ; 24(6): 966-980, 2016 11.
Article in English | MEDLINE | ID: mdl-27681311

ABSTRACT

Healing is delayed in limb wounds relative to body wounds of horses, partly because of sustained inflammation and inefficient angiogenesis. In laboratory animals, proteins derived from orf virus modulate these processes and enhance healing. We aimed to compare immune cell trafficking and the inflammatory, vascular, and epidermal responses in body and limb wounds of horses and then to investigate the impact of orf virus interleukin-10 and vascular endothelial growth factor-E on these processes. Standardized excisional wounds were created on the body and forelimb of horses and their progression monitored macroscopically until healed. Tissue samples were harvested to measure the expression of genes regulating inflammation and repair (quantitative polymerase chain reaction) and to observe epithelialization (histology), innate immune cell infiltration, and angiogenesis (immunofluorescence). Delayed healing of limb wounds was characterized by intensified and extended pro-inflammatory signaling and exacerbated innate immune response, concomitant with the absence of anti-inflammatory eIL-10. Blood vessels were initially more permeable and then matured belatedly, concomitant with retarded production of angiogenic factors. Epithelial coverage was achieved belatedly in limb wounds. Viral proteins were administered to wounds of one body and one limb site/horse at days 1-3, while wounds at matching sites served as controls. Treatment dampened pro-inflammatory gene expression and the innate immune response in all wounds. It also improved angiogenic gene expression, but primarily in body wounds, where it altered blood vessel density and myofibroblast persistence. Moreover, the viral proteins increased epithelialization of all wounds. The short-term viral protein therapy did not, however, improve the healing rate of wounds in either location, likely due to suboptimal dosing. In conclusion, we have further detailed the processes contributing to protracted healing in limb wounds of horses and shown that short-term administration of viral proteins exerts several promising though transient effects that, if optimized, may positively influence healing.


Subject(s)
Inflammation/genetics , Inflammation/therapy , Interleukin-10/genetics , Orf virus/genetics , Viral Proteins/genetics , Wound Healing , Wounds and Injuries/therapy , Animals , Cells, Cultured , Extremities/injuries , Extremities/pathology , Extremities/virology , Gene Expression Regulation , Horses , Humans , Inflammation/pathology , Inflammation/virology , Interleukin-10/metabolism , Male , Neovascularization, Physiologic , Viral Proteins/metabolism , Wounds and Injuries/genetics
8.
Wound Repair Regen ; 22(3): 356-67, 2014.
Article in English | MEDLINE | ID: mdl-24844335

ABSTRACT

Interleukin (IL)-10 plays a critical role in controlling wound inflammation and scar formation. Orf virus, a zoonotic parapoxvirus, induces proliferative skin lesions that resolve with minimal scarring. Orf virus encodes a range of factors that subvert the host's response to infection, including a homolog of IL-10. This study investigated, using a murine full-thickness wound model, whether purified orf virus IL-10 (ovIL-10) can regulate skin repair and scarring. Repeat injections of ovIL-10 into wounded skin accelerated wound closure. Histological analyses of wound sections revealed that treatment with ovIL-10 accelerated wound reepithelialization, granulation tissue coverage of the wound bed, and improved wound revascularization. In addition, wounds treated with ovIL-10 showed a reduction in macrophage infiltration, myofibroblast differentiation, and wound contraction. Treatment of wounds with ovIL-10 also resulted in a reduction in visible scarring that was consistent with the extent of scar tissue formed. Quantitative polymerase chain reaction analysis confirmed that ovIL-10 reduced the expression of key mediators of inflammation and granulation tissue formation. These findings show that ovIL-10, like mammalian IL-10, limits inflammation and scar tissue formation and reveal a new role for both mammalian and viral IL-10 in mediating tissue repair.


Subject(s)
Cicatrix/pathology , Inflammation/pathology , Interleukin-10/pharmacology , Orf virus/pathogenicity , Skin/pathology , Wound Healing , Wounds and Injuries/pathology , Animals , Cells, Cultured , Cytokines/metabolism , Female , Inflammation/immunology , Mice , Orf virus/immunology , Wound Healing/immunology , Wounds and Injuries/immunology
9.
Cell Microbiol ; 14(9): 1376-90, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22507661

ABSTRACT

Vascular endothelial growth factor (VEGF)-A, a key regulator of cutaneous blood vessel formation, appears to have an additional role during wound healing, enhancing re-epithelialization. Orf virus, a zoonotic parapoxvirus, induces proliferative skin lesions that initiate in wounds and are characterized by extensive blood vessel formation, epidermal hyperplasia and rete ridge formation. The vascular changes beneath the lesion are largely due to viral-expressed VEGF-E. This study investigated using mouse skin models whether VEGF-E can induce epidermal changes such as that seen in the viral lesion. Injection of VEGF-E into normal skin increased the number of endothelial cells and blood vessels within the dermis and increased epidermal thickening and keratinocyte number. Injection of VEGF-E into wounded skin, which more closely mimics orf virus lesions, increased neo-epidermal thickness and area, promoted rete ridge formation, and enhanced wound re-epithelialization. Quantitative RT-PCR analysis showed that VEGF-E did not induce expression of epidermal-specific growth factors within the wound, but did increase matrix metalloproteinase (MMP)-2 and MMP-9 expression. In cell-based assays, VEGF-E induced keratinocyte migration and proliferation, responses that were inhibited by a neutralizing antibody against VEGF receptor (VEGFR)-2. These findings demonstrate that VEGF-E, both directly and indirectly, regulates keratinocyte function, thereby promoting epidermal regeneration.


Subject(s)
Epidermis/pathology , Keratinocytes/drug effects , Orf virus/pathogenicity , Viral Proteins/metabolism , Virulence Factors/metabolism , Animals , Cell Movement , Cell Proliferation , Gene Expression Profiling , Matrix Metalloproteinase 2/biosynthesis , Matrix Metalloproteinase 9/biosynthesis , Mice , Real-Time Polymerase Chain Reaction , Regeneration
10.
Environ Microbiol ; 9(12): 2923-32, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17991023

ABSTRACT

The ability to utilize the siderophore ferrichrome as an iron source was found to be a variable trait in a field population of mesorhizobia. To investigate the genetic basis of this variation, genes required for ferrichrome utilization (fhu genes) were characterized in Mesorhizobium strain R88B, an Fhu(+) member of the population. Functional fhu genes were present at three loci. Two genes of the ferrichrome ABC transporter, fhuBD, were identified at an fhu1 locus downstream of the symbiosis island that was integrated at the phe-tRNA gene. The fhuA gene encoding the ferrichrome outer membrane receptor was located in the fhu2 locus together with non-functional fhuDB genes, while the fhuC gene encoding the ATPase required for ferrichrome transport was part of the fhu3 locus that included genes required to form a functional TonB complex. None of the fhu genes were present in the sequenced Mesorhizobium loti strain MAFF303099. Comparisons with MAFF303099 suggested that the fhu2 and fhu3 loci evolved through small-scale (< 5 kb) acquisitions and deletions. Despite their independent origins, the three fhu loci were coordinately regulated in response to iron availability. Within the mesorhizobial population, the ability to utilize ferrichrome was most strongly correlated with the presence of the fhuA gene. We hypothesize that the ferrichrome transport system evolved through cycles of gene acquisition and deletion, with the positive selection pressure of an iron-poor or siderophore-rich environment being offset by the negative pressure of the outer membrane receptor being a target for phage.


Subject(s)
Alphaproteobacteria/metabolism , Bacterial Proteins/genetics , Evolution, Molecular , Ferrichrome/metabolism , Gene Expression Regulation, Bacterial , ATP-Binding Cassette Transporters/genetics , ATP-Binding Cassette Transporters/metabolism , Alphaproteobacteria/classification , Alphaproteobacteria/genetics , Alphaproteobacteria/growth & development , Bacterial Outer Membrane Proteins/genetics , Bacterial Outer Membrane Proteins/metabolism , Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Base Sequence , DNA, Bacterial/analysis , Genetic Variation , Iron/metabolism , Molecular Sequence Data , Selection, Genetic , Sequence Analysis, DNA
11.
Mol Microbiol ; 62(3): 723-34, 2006 Nov.
Article in English | MEDLINE | ID: mdl-17076666

ABSTRACT

The Mesorhizobium loti strain R7A symbiosis island is an Integrative Conjugative Element (ICE), herein termed ICEMlSymR7A, which integrates into a phetRNA gene. Integration reconstructs the phetRNA gene at one junction with the core chromosome, and a direct repeat of the 3-prime 17 bp of the gene is formed at the other junction. We show that the ICEMlSymR7AintS gene, which encodes an integrase of the phage P4 family, is required for integration and excision of the island. Excision also depended on a novel recombination directionality factor encoded by msi109 (rdfS). Constitutive expression of rdfS resulted in curing of ICEMlSymR7A. The rdfS gene is part of an operon with genes required for conjugative transfer, allowing co-ordinate regulation of ICEMlSymR7A excision and transfer. The excised form of ICEMlSymR7A was detectable during exponential growth but occurred at higher frequency during stationary phase. ICEMlSymR7A encodes homologues of the traR and traI genes of Agrobacterium tumefaciens that regulate Ti plasmid transfer via quorum sensing. The presence of a plasmid with cloned island traR traI2 genes resulted in excision of ICEMlSymR7A in all cells regardless of culture density, indicating that excision may be similarly regulated. Maintenance of ICEMlSymR7A in these cells depended on msi106 (rlxS) that encodes a putative relaxase. Transfer of the island to non-symbiotic mesorhizobia required intS, rlxS and rdfS. The rdfS and rlxS genes are conserved across a diverse range of alpha-, beta- and gamma-proteobacteria and identify a large family of genomic islands with a common transfer mechanism.


Subject(s)
Bacterial Proteins/metabolism , DNA Nucleotidyltransferases/metabolism , Genomic Islands/physiology , Integrases/metabolism , Rhizobiaceae/genetics , Amino Acid Sequence , Attachment Sites, Microbiological , Bacterial Proteins/genetics , Conserved Sequence , DNA Nucleotidyltransferases/genetics , Gene Expression Regulation, Bacterial , Integrases/genetics , Molecular Sequence Data , Phylogeny , Recombination, Genetic , Rhizobiaceae/physiology , Sequence Homology, Amino Acid , Symbiosis/physiology
12.
J Bacteriol ; 184(11): 3086-95, 2002 Jun.
Article in English | MEDLINE | ID: mdl-12003951

ABSTRACT

The Mesorhizobium loti strain R7A symbiosis island is a 502-kb chromosomally integrated element which transfers to nonsymbiotic mesorhizobia in the environment, converting them to Lotus symbionts. It integrates into a phenylalanine tRNA gene in a process mediated by a P4-type integrase encoded at the left end of the element. We have determined the nucleotide sequence of the island and compared its deduced genetic complement with that reported for the 611-kb putative symbiosis island of M. loti strain MAFF303099. The two islands share 248 kb of DNA, with multiple deletions and insertions of up to 168 kb interrupting highly conserved colinear DNA regions in the two strains. The shared DNA regions contain all the genes likely to be required for Nod factor synthesis, nitrogen fixation, and island transfer. Transfer genes include a trb operon and a cluster of potential tra genes which are also present on the strain MAFF303099 plasmid pMLb. The island lacks plasmid replication genes, suggesting that it is a site-specific conjugative transposon. The R7A island encodes a type IV secretion system with strong similarity to the vir pilus from Agrobacterium tumefaciens that is deleted from MAFF303099, which in turn encodes a type III secretion system not found on the R7A island. The 414 genes on the R7A island also include putative regulatory genes, transport genes, and an array of metabolic genes. Most of the unique hypothetical genes on the R7A island are strain-specific and clustered, suggesting that they may represent other acquired genetic elements rather than symbiotically relevant DNA.


Subject(s)
Genes, Bacterial , Rhizobiaceae/genetics , Symbiosis , Amino Acids/metabolism , Carbon/metabolism , Gene Transfer, Horizontal/genetics , Genes, Regulator , Lotus/microbiology , Microtubule Proteins/biosynthesis , Microtubule Proteins/genetics , Molecular Sequence Data , Multigene Family , Nitrogen Fixation/genetics , Phosphates/metabolism , Rhizobiaceae/metabolism , Species Specificity
SELECTION OF CITATIONS
SEARCH DETAIL
...