Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
Wound Repair Regen ; 25(4): 632-640, 2017 08.
Article in English | MEDLINE | ID: mdl-28665034

ABSTRACT

Skin quality outcome after skin grafting is adversely affected by wound bed inflammation. Neomycin, gentamicin, and other aminoglycoside antibiotics are known to modulate inflammation, and topical application affords the use of higher doses than are possible to use systemically. Previous data suggest that clinically relevant doses of neomycin, but not gentamicin, may impair angiogenesis, which is critical to the durable survival of skin grafts. The role of gentamicin at ultrahigh doses compared with clinically relevant neomycin doses in regulating inflammatory expression and angiogenesis has been examined. In a porcine skin replacement excisional wound model, continuous exposure to gentamicin increased anti-angiogenic and inflammatory expression at 7 days postgrafting. In in vitro studies, gentamicin also impaired angiogenesis in a human umbilical vein endothelial cell (HUVEC) tube formation model, increased the expression of the anti-angiogenic gene C-X-C motif chemokine 10 (CXCL10) in HUVECs and macrophages, and increased pro-inflammatory cytokine expression of macrophages in a dose-dependent manner. Neomycin exerted similar effects in vitro at clinically relevant doses on HUVEC tube formation and macrophage pro-inflammatory expression. CXCL10 was upregulated in macrophages, but did not exhibit a change in HUVECs with neomycin treatment. Ultrahigh doses of gentamicin and clinically relevant doses of neomycin affect inflammation and angiogenesis in in vivo and in vitro models. These findings suggest that topical administration of aminoglycosides have the potential to adversely influence early skin graft survival.


Subject(s)
Anti-Bacterial Agents/pharmacology , Burns/pathology , Gentamicins/pharmacology , Inflammation/drug therapy , Neovascularization, Physiologic/drug effects , Wound Healing/drug effects , Wound Infection/drug therapy , Animals , Anti-Bacterial Agents/administration & dosage , Burns/immunology , Cell Proliferation/drug effects , Disease Models, Animal , Dose-Response Relationship, Drug , Female , Gentamicins/administration & dosage , Inflammation/metabolism , Swine , Wound Healing/immunology , Wound Infection/immunology
2.
FEBS Open Bio ; 7(7): 953-967, 2017 07.
Article in English | MEDLINE | ID: mdl-28680809

ABSTRACT

The healing of burn wounds is often hampered by bacterial infection and the formation of biofilms. Antimicrobial peptides (AMPs) are effective in promoting wound healing, but are susceptible to degradation. We have tested the ability of ceragenins (CSAs), mimics of antimicrobial peptides, to mitigate preformed biofilms and stimulate wound healing in vitro. Potent CSAs (MICs < 10 µg·mL-1) were tested against biofilms formed from a mixture of Pseudomonas aeruginosa and Staphylococcus aureus grown for 22 h and subjected to 20 h treatment. Many CSAs showed more potent anti-biofilm activity than the endogenous AMP LL-37, and CSA-13 and CSA-90 decreased the amount of biofilm matrix substances detected by SYPRO Ruby stain. Effects on mammalian cells were measured by viability, migration, and tube formation assays in vitro. Although CSAs were toxic to immortalized human keratinocytes (HaCaTs) at higher concentrations (>10 µg·mL-1), lower concentrations of CSA-13 and CSA-192 stimulated cell migration. CSA-13, CSA-90, and CSA-142 also stimulated tube formation in an in vitro angiogenesis model. An inhibitor of vascular endothelial growth factor receptor 2 (VEGFR2) blocked tube formation stimulated by CSA-13, suggesting that CSA-13 signals through this receptor. Ceragenins display anti-biofilm activity and stimulate migration and tube formation in vitro. This work suggests that ceragenins have the potential to be both topical antimicrobials and wound-healing adjunct therapeutics.

3.
Technology (Singap World Sci) ; 5(2): 81-86, 2017 06.
Article in English | MEDLINE | ID: mdl-29552603

ABSTRACT

The transition of macrophages from the pro-inflammatory M1 to the anti-inflammatory M2 phenotype is crucial for the progression of normal wound healing. Persistent M1 macrophages within the injury site may lead to an uncontrolled macrophage-mediated inflammatory response and ultimately a failure of the wound healing cascade, leading to chronic wounds. Mesenchymal stromal cells (MSCs) have been widely reported to promote M1 to M2 macrophage transition; however, it is unclear whether MSCs can drive this transition in the hypoxic environment typically observed in chronic wounds. Here we report on the effect of hypoxia (1% O2) on MSCs' ability to transition macrophages from the M1 to the M2 phenotype. While hypoxia had no effect on MSC secretion, it inhibited MSC-induced M1 to M2 macrophage transition, and suppressed macrophage expression and production of the anti-inflammatory mediator interleukin-10 (IL-10). These results suggest that hypoxic environments may impede the therapeutic effects of MSCs.

4.
Wound Repair Regen ; 23(5): 711-23, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26110250

ABSTRACT

Chronic skin wounds are a common complication of diabetes. When standard wound care fails to heal such wounds, a promising approach consists of using decellularized matrices and other porous scaffold materials to promote the restoration of skin. Proper revascularization is critical for the efficacy of such materials in regenerative medicine. Stromal cell-derived factor-1 (SDF-1) is a chemokine known to play a key role for angiogenesis in ischemic tissues. Herein we developed nanosized SDF-1 liposomes, which were then incorporated into decellularized dermis scaffolds used for skin wound healing applications. SDF-1 peptide associated with liposomes with an efficiency of 80%, and liposomes were easily dispersed throughout the acellular dermis. Acellular dermis spiked with SDF-1 liposomes exhibited more persistent cell proliferation in the dermis, especially in CD31(+) areas, compared to acellular dermis spiked with free SDF-1, which resulted in increased improved wound closure at day 21, and increased granulation tissue thickness at day 28. SDF-1 liposomes may increase the performance of a variety of decellularized matrices used in tissue engineering.


Subject(s)
Acellular Dermis , Chemokine CXCL12/administration & dosage , Diabetes Mellitus, Experimental/complications , Skin/pathology , Tissue Scaffolds , Wound Healing/physiology , Wounds and Injuries/therapy , Animals , Cell Proliferation , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/pathology , Humans , Liposomes , Mice , Skin/metabolism , Tissue Engineering , Wounds and Injuries/etiology , Wounds and Injuries/metabolism
5.
Biochem Biophys Res Commun ; 458(1): 8-13, 2015 Feb 27.
Article in English | MEDLINE | ID: mdl-25625213

ABSTRACT

During wound healing, fibroblasts deposit extracellular matrix that guides angiogenesis and supports the migration and proliferation of cells that eventually form the scar. They also promote wound closure via differentiation into α-smooth muscle actin (SMA)-expressing myofibroblasts, which cause wound contraction. Low oxygen tension typical of chronic nonhealing wounds inhibits fibroblast collagen production and differentiation. It has been suggested that hypoxic mesenchymal stromal cells (MSCs) secrete factors that promote wound healing in animal models; however, it is unclear whether these factors are equally effective on the target cells in a hypoxic wound environment. Here we investigated the impact of MSC-derived soluble factors on the function of fibroblasts cultured in hypoxic fibroblast-populated collagen lattices (FPCLs). Hypoxia alone significantly decreased FPCL contraction and α-SMA expression. MSC-conditioned medium restored hypoxic FPCL contraction and α-SMA expression to levels similar to normoxic FPCLs. SB431542, an inhibitor of transforming growth factor-ß1 (TGF-ß1)-mediated signaling, blocked most of the MSC effect on FPCL contraction, while exogenous TGF-ß1 at levels similar to that secreted by MSCs reproduced the MSC effect. These results suggest that TGF-ß1 is a major paracrine signal secreted by MSCs that can restore fibroblast functions relevant to the wound healing process and that are impaired in hypoxia.


Subject(s)
Actins/metabolism , Fibroblasts/metabolism , Mesenchymal Stem Cells/metabolism , Alginates , Cell Differentiation , Cell Hypoxia/physiology , Cells, Cultured , Cells, Immobilized , Collagen/metabolism , Culture Media, Conditioned/pharmacology , Fibroblasts/cytology , Fibroblasts/drug effects , Glucuronic Acid , Hexuronic Acids , Humans , Mesenchymal Stem Cells/cytology , Muscle, Smooth/metabolism , Myofibroblasts/metabolism , Skin/cytology , Skin/metabolism , Transforming Growth Factor beta1/metabolism , Wound Healing
SELECTION OF CITATIONS
SEARCH DETAIL