Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters










Publication year range
1.
Burns ; 42(6): 1277-86, 2016 Sep.
Article in English | MEDLINE | ID: mdl-27222384

ABSTRACT

Dermal wound healing, in which transforming growth factor beta 1 (TGFß1) plays an important role, is a complex process. Previous studies suggest that vitamin D has a potential regulatory role in TGFß1 induced activation in bone formation, and there is cross-talk between their signaling pathways, but research on their effects in other types of wound healing is limited. The authors therefore wanted to explore the role of vitamin D and its interaction with low concentration of TGFß1 in dermal fibroblast-mediated wound healing through an in vitro study. Human dermal fibroblasts were treated with vitamin D, TGFß1, both, or vehicle, and then the wound healing functions of dermal fibroblasts were measured. To further explore possible mechanisms explaining the synergistic effect of vitamin D and TGFß1, targeted gene silencing of the vitamin D receptor was performed. Compared to either factor alone, treatment of fibroblasts with both vitamin D and low concentration of TGFß1 increased gene expression of TGFß1, connective tissue growth factor, and fibronectin 1, and enhanced fibroblast migration, myofibroblast formation, and collagen production. Vitamin D receptor gene silencing blocked this synergistic effect of vitamin D and TGFß1 on both collagen production and myofibroblast differentiation. Thus a synergistic effect of vitamin D and low TGFß1 concentration was found in dermal fibroblast-mediated wound healing in vitro. This study suggests that supplementation of vitamin D may be an important step to improve wound healing and regeneration in patients with a vitamin D deficiency.


Subject(s)
Calcitriol/pharmacology , Dermis/drug effects , Fibroblasts/drug effects , Transforming Growth Factor beta1/pharmacology , Vitamins/pharmacology , Wound Healing/drug effects , Adult , Cell Differentiation/drug effects , Cell Movement/drug effects , Cells, Cultured , Chromatography, Liquid , Connective Tissue Growth Factor/drug effects , Connective Tissue Growth Factor/genetics , Dermis/metabolism , Drug Synergism , Female , Fibroblasts/metabolism , Fibronectins/drug effects , Fibronectins/genetics , Humans , Hydroxyproline/metabolism , In Vitro Techniques , Mass Spectrometry , Myofibroblasts/drug effects , Real-Time Polymerase Chain Reaction , Receptors, Calcitriol/drug effects , Receptors, Calcitriol/genetics , Reverse Transcriptase Polymerase Chain Reaction , Smad2 Protein/drug effects , Smad2 Protein/genetics , Smad3 Protein/drug effects , Smad3 Protein/genetics , Smad7 Protein/drug effects , Smad7 Protein/genetics , Transforming Growth Factor beta1/drug effects , Transforming Growth Factor beta1/genetics
2.
Burns ; 41(5): 998-1007, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25683215

ABSTRACT

Hypertrophic scars (HTS) are generally believed to result from proliferation and activation of resident connective tissue fibroblasts after burns. To demonstrate a potential role of blood-borne cells, the peripheral blood mononuclear cells (PBMCs) and the effect of PBMCs on dermal fibroblast behavior was investigated. Flow cytometry was used to analyze the surface and intracellular protein expression of PBMCs and fibroblasts. Transwell migration assay, enzyme-linked immunosorbent assay and real-time reverse transcription polymerase chain reaction was performed to assess fibroblast functions. We identified a novel subpopulation of PBMCs in burn patients in vivo that appears at an early stage following major thermal injuries, which primarily express procollagen 1, leukocyte specific protein 1, CD204, toll-like receptor 4 and stromal cell-derived factor 1 (SDF-1) receptor CXCR4. In vitro, the conditioned media from burn patient PBMCs up-regulated the expression of fibrotic growth factors and extracellular matrix molecules, down-regulated antifibrotic factor decorin, enhanced cell chemotaxis and promoted cell differentiation into contractile myofibroblasts in dermal fibroblasts. After thermal injury, this novel subpopulation of PBMCs is systemically triggered and attracted to the wounds under SDF-1/CXCR4 signaling where they appear to modulate the functions of resident connective tissue cells and thus contribute to the development of HTS.


Subject(s)
Burns/blood , Chemotaxis , Cicatrix, Hypertrophic/blood , Leukocytes, Mononuclear/cytology , Macrophages/cytology , Adult , Body Surface Area , Burns/pathology , Cell Differentiation , Cell Migration Assays , Cells, Cultured , Connective Tissue Cells/cytology , Connective Tissue Cells/metabolism , Decorin/metabolism , Down-Regulation , Enzyme-Linked Immunosorbent Assay , Extracellular Matrix Proteins/metabolism , Female , Fibroblasts/cytology , Humans , In Vitro Techniques , Leukocytes, Mononuclear/metabolism , Macrophages/metabolism , Male , Microfilament Proteins/metabolism , Middle Aged , Myofibroblasts/cytology , Procollagen/metabolism , Receptors, CXCR4/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Scavenger Receptors, Class A/metabolism , Signal Transduction , Skin/cytology , Toll-Like Receptor 4/metabolism , Trauma Severity Indices , Young Adult
3.
Wound Repair Regen ; 22(5): 622-30, 2014.
Article in English | MEDLINE | ID: mdl-25139227

ABSTRACT

Effective prevention and treatment of hypertrophic scars (HTSs), a dermal form of fibrosis that frequently occurs following thermal injury to deep dermis, are unsolved significant clinical problems. Previously, we have found that stromal cell-derived factor 1/CXCR4 signaling is up-regulated during wound healing in burn patients and HTS tissue after thermal injury. We hypothesize that blood-borne mononuclear cells are recruited into wound sites after burn injury through the chemokine pathway of stromal cell-derived factor 1 and its receptor CXCR4. Deep dermal injuries to the skin are often accompanied by prolonged inflammation, which leads to chemotaxis of mononuclear cells into the wounds by chemokine signaling where fibroblast activation occurs and ultimately HTS are formed. Blocking mononuclear cell recruitment and fibroblast activation, CXCR4 antagonism is expected to reduce or minimize scar formation. In this study, the inhibitory effect of CXCR4 antagonist CTCE-9908 on dermal fibrosis was determined in vivo using a human HTS-like nude mouse model, in which split-thickness human skin is transplanted into full-thickness dorsal excisional wounds in athymic mice, where these wounds subsequently develop fibrotic scars that resemble human HTS as previously described. CTCE-9908 significantly attenuated scar formation and contraction, reduced the accumulation of macrophages and myofibroblasts, enhanced the remodeling of collagen fibers, and down-regulated the gene and protein expression of fibrotic growth factors in the human skin tissues. These findings support the potential therapeutic value of CXCR4 antagonist in dermal fibrosis and possibly other fibroproliferative disorders.


Subject(s)
Cicatrix, Hypertrophic/prevention & control , Dermis/drug effects , Peptides/pharmacology , Receptors, CXCR4/antagonists & inhibitors , Wound Healing/drug effects , Adult , Animals , Cicatrix, Hypertrophic/pathology , Dermis/pathology , Disease Models, Animal , Female , Fibrosis , Humans , Male , Mice, Nude , Middle Aged , Skin Transplantation
4.
Wound Repair Regen ; 21(3): 448-55, 2013.
Article in English | MEDLINE | ID: mdl-23627585

ABSTRACT

Hypertrophic scars are a significant fibroproliferative disorder complicating deep injuries to the skin. We hypothesize that activated deep dermal fibroblasts are subject to regulation by bone marrow-derived mesenchymal stem cells (BM-MSCs), which leads to the development of excessive fibrosis following deep dermal injury. We found that the expression of fibrotic factors was higher in deep burn wounds compared with superficial burn wounds collected from burn patients with varying depth of skin injury. We characterized deep and superficial dermal fibroblasts, which were cultured from the deep and superficial dermal layers of normal uninjured skin obtained from abdominoplasty patients, and examined the paracrine effects of BM-MSCs on the fibrotic activities of the cells. In vitro, deep dermal fibroblasts were found higher in the messenger RNA (mRNA) levels of type 1 collagen, alpha smooth muscle actin, transforming growth factor beta, stromal cell-derived factor 1, and tissue inhibitor of metalloproteinase 1, an inhibitor of collagenase (matrix metalloproteinase 1). As well, deep dermal fibroblasts had low matrix metalloproteinase 1 mRNA, produced more collagen, and contracted collagen lattices significantly greater than superficial fibroblasts. By co-culturing layered fibroblasts with BM-MSCs in a transwell insert system, BM-MSCs enhanced the fibrotic behavior of deep dermal fibroblasts, which suggests a possible involvement of BM-MSCs in the pathogenesis of hypertrophic scarring.


Subject(s)
Burns/pathology , Cicatrix, Hypertrophic/prevention & control , Skin/pathology , Stem Cell Transplantation/methods , Wound Healing/physiology , Adult , Burns/metabolism , Burns/surgery , Cell Proliferation , Cells, Cultured , Cicatrix, Hypertrophic/metabolism , Cicatrix, Hypertrophic/pathology , Collagen/biosynthesis , Collagen/genetics , Female , Fibroblasts/metabolism , Fibroblasts/pathology , Gene Expression Regulation , Humans , Laser-Doppler Flowmetry , Male , Mesenchymal Stem Cells , Middle Aged , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Skin/metabolism
5.
J Cell Commun Signal ; 7(4): 239-52, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23504443

ABSTRACT

Hypertrophic scar (HTS) is a dermal form of fibroproliferative disorder which often develops after thermal or traumatic injury to the deep regions of the skin and is characterized by excessive deposition and alterations in morphology of collagen and other extracellular matrix (ECM) proteins. HTS are cosmetically disfiguring and can cause functional problems that often recur despite surgical attempts to remove or improve the scars. In this review, the roles of various fibrotic and anti-fibrotic molecules are discussed in order to improve our understanding of the molecular mechanism of the pathogenesis of HTS. These molecules include growth factors, cytokines, ECM molecules, and proteolytic enzymes. By exploring the mechanisms of this form of dermal fibrosis, we seek to provide some insight into this form of dermal fibrosis that may allow clinicians to improve treatment and prevention in the future.

6.
J Burn Care Res ; 33(6): 741-6, 2012.
Article in English | MEDLINE | ID: mdl-23147213

ABSTRACT

Burn-specific guidelines for optimal catheter rotation, catheter type, insertion methods, and catheter site care do not exist, and practices vary widely from one burn unit to another. The purpose of this study was to define current practices and identify areas of practice variation for future clinical investigation. An online survey was sent to the directors of 123 U.S. burn centers. The survey consisted of 23 questions related to specific practices in placement and maintenance of central venous catheters (CVCs), arterial catheters, and peripherally inserted central catheters (PICCs). The overall response rate was 36%; response rate from verified centers was 52%. Geographic representation was wide. CVC and arterial catheter replacement varied from every 3 days (24% of sites) to only for overt infection (24% of sites); 23% of sites did not use the femoral position for CVC placement. Nearly 60% of units used some kind of antiseptic catheter. Physicians inserted the majority of catheters, and 22% of sites used nonphysicians for at least some insertions. Ultrasound was routinely used by less than 50% of units. A wide variety of post-insertion dressing protocols were followed. PICCs were used in some critically injured patients in 37% of units; the majority of these users did not rotate PICCs. Thus, it can be surmised that wide practice variation exists among burn centers with regard to insertion and maintenance of invasive catheters. Areas with particular variability that would be appropriate targets of clinical investigation are line rotation protocols, catheter site care protocols, and use of PICCs in acute burns.


Subject(s)
Burn Units , Catheterization/statistics & numerical data , Practice Patterns, Physicians'/statistics & numerical data , Catheterization/standards , Humans , Surveys and Questionnaires , United States
7.
J Burn Care Res ; 33(6): 764-71, 2012.
Article in English | MEDLINE | ID: mdl-22955162

ABSTRACT

Hypertrophic scarring (HTS) is a fibroproliferative disorder that commonly develops after severe burn injuries. Overexpression of transforming growth factor-ß (TGF-ß) by an increased number of fibrocytes has been associated with increased extracellular matrix molecule expression leading to HTS. The most widely accepted adjuvant to clinical assessment of burn depth is laser Doppler imaging (LDI) and may predict injury to the dermis that corresponds to cellular and molecular changes associated with HTS. A prospective, blinded, control trial was performed comparing LDI and clinical assessment for the decision to operate. Immunohistochemistry and real-time reverse transcription polymerase chain reaction was performed to determine whether there is a correlation between histological assessment of burn depth and LDI, and the presence of fibrocytes was detected using confocal microscopy. The positive predictive value for a burn requiring a graft was calculated to be >90%. Immunohistochemistry on biopsy samples revealed an increased expression of TGF-ß, connective tissue growth factor, heat shock protein 47, and collagen type I in deep burn wounds compared to superficial burns. Using the fibrocyte-specific markers procollagen type I and lymphocyte-specific protein-1, there was an increased number of fibrocytes in deep burn areas compared to superficial burn. In deep burn injuries, increased infiltration of fibrocytes occurs leading to an overexpression of TGF-ß1 and connective tissue growth factor. More importantly, LDI was >90% accurate at predicting the need for excision and grafting. The accuracy of the decision to debride deep dermal burns to avoid HTS using both clinical parameters and LDI was supported by histological and biochemical measurements.


Subject(s)
Burns/pathology , Cicatrix, Hypertrophic/pathology , Laser-Doppler Flowmetry/methods , Adolescent , Adult , Aged , Biopsy , Burns/metabolism , Child , Child, Preschool , Cicatrix, Hypertrophic/metabolism , Collagen Type I/metabolism , Connective Tissue Growth Factor/metabolism , Female , HSP47 Heat-Shock Proteins/metabolism , Humans , Immunohistochemistry , Infant , Male , Microfilament Proteins/metabolism , Microscopy, Confocal , Middle Aged , Predictive Value of Tests , Prospective Studies , Real-Time Polymerase Chain Reaction , Transforming Growth Factor beta/metabolism
8.
J Burn Care Res ; 33(5): 668-77, 2012.
Article in English | MEDLINE | ID: mdl-22210076

ABSTRACT

Hypertrophic scar (HTS) represents the dermal equivalent of fibroproliferative disorders. Fibroblasts from the deep dermis are implicated in the development of HTS after injuries that involve deeper areas of the skin. However, fibroblasts that reside in the superficial layer of the skin show antifibrotic properties, and injuries limited to this area heal with little or no scarring. Previously, cellular and molecular characteristics of superficial fibroblasts and deep dermal fibroblasts that may influence HTS formation were analyzed. In this study, differences in cellular behavior between superficial fibroblasts and deep dermal fibroblasts that may also affect the development of HTS or tissue fibrosis were further characterized. Immunostaining and migration, adhesion, apoptosis, and cell viability assays were performed in fibroblasts from the superficial and deep dermis. Reverse-transcription polymerase chain reaction was used to examine the gene expression of molecules involved in cell death after treatment of fibroblasts with decorin. When compared with superficial fibroblasts, deep dermal fibroblasts showed lower migration rates. Although all the fibroblasts tested showed no difference in adhesion to fibronectin, superficial fibroblasts demonstrated increased apoptotic and dead cells when treated with decorin. Decorin resulted in a significant increase in the expression of apoptosis markers, histone-1, caspase-1, caspase-8, and p53 in superficial fibroblasts when compared with deep dermal fibroblasts. Taken together, the findings suggest that reduced migration, lack of decorin, and resistance of deep dermal fibroblasts to decorin-induced apoptosis may result in hypercellularity in injuries involving the deep dermis, leading to deposition of excess extracellular matrix and HTS formation.


Subject(s)
Apoptosis , Cicatrix, Hypertrophic/pathology , Decorin , Fibroblasts , Caspase 8 , Cell Adhesion , Cell Migration Assays , Extracellular Matrix , Extracellular Matrix Proteins , Humans , Proteoglycans , RNA, Messenger , Signal Transduction , Wound Healing
9.
J Burn Care Res ; 33(2): 218-27, 2012.
Article in English | MEDLINE | ID: mdl-22079916

ABSTRACT

Hypertrophic scar (HTS) occurs after injuries involving the deep dermis, while superficial wounds (SWs) to the skin heal with minimal or no scarring. The levels of transforming growth factor (TGF)-ß1 and small leucine-rich proteoglycans (SLRPs) with fibroblast subtype and function may influence the development of HTS. The aim of this study was to characterize the expression and localization of factors that regulate wound healing including SLRPs, TGF-ß1, and TGF-ß3 in an experimental human SW and deep wound (DW) scar model including fibroblasts from superficial and deep layers of normal dermis. A 6-cm horizontal dermal scratch experimental wound was created, which consisted of progressively deeper wounds that were superficial at one end (0-0.75 mm deep) and deep (0.75-3 mm deep) at the other end, located on the anterior thigh of an adult male. Immunofluorescence staining, immunoblotting, reverse transcription polymerase chain reaction, and flow cytometry were performed to analyze the cellular and molecular differences between the SW scar and DW scar as well as fibroblasts isolated from superficial layer (L1) and deep layer (L5) of normal dermis. Comparing SWs and L1 fibroblasts, the expression of decorin, fibromodulin, and TGF-ß3 was considerably lower than in DWs and L5 fibroblasts; however, TGF-ß1 was higher in the deeper dermal wounds. When compared with L1 fibroblasts, L5 fibroblasts had lower Thy-1 immunoreactivity and significantly higher expression of TGF-ß receptor type II. Decreased antifibrotic molecules in matrix of deep dermis of the skin and the unique features of the associated fibroblasts including an increased sensitivity to TGF-ß1 stimulation contribute to the development of HTS after injuries involving the deep dermis.


Subject(s)
Burns/metabolism , Cicatrix, Hypertrophic/metabolism , Decorin/metabolism , Extracellular Matrix Proteins/metabolism , Proteoglycans/metabolism , Transforming Growth Factor beta3/metabolism , Analysis of Variance , Blotting, Western , Burns/complications , Burns/therapy , Cicatrix, Hypertrophic/etiology , Down-Regulation , Fibroblasts/metabolism , Fibromodulin , Flow Cytometry , Fluorescent Antibody Technique , Humans , Male , Microscopy, Confocal , Occlusive Dressings , Polymerase Chain Reaction , Thigh
10.
Wound Repair Regen ; 19(5): 568-78, 2011.
Article in English | MEDLINE | ID: mdl-22092795

ABSTRACT

Recent data support the involvement of stromal cell-derived factor 1 (SDF-1) in the homing of bone marrow-derived stem cells to wound sites during skeletal, myocardial, vascular, lung, and skin wound repair as well as some fibrotic disorders via its receptor CXCR4. In this study, the role of SDF-1/CXCR4 signaling in the formation of hypertrophic scar (HTS) following burn injury and after treatment with systemic interferon α2b (IFNα2b) is investigated. Studies show SDF-1/CXCR4 signaling was up-regulated in burn patients, including SDF-1 level in HTS tissue and serum as well as CD14+ CXCR4+ cells in the peripheral blood mononuclear cells. In vitro, dermal fibroblasts constitutively expressed SDF-1 and deep dermal fibroblasts expressed more SDF-1 than superficial fibroblasts. Lipopolysaccharide increased SDF-1 gene expression in fibroblasts. Also, recombinant SDF-1 and lipopolysaccharide stimulated fibroblast-conditioned medium up-regulated peripheral blood mononuclear cell mobility. In the burn patients with HTS who received subcutaneous IFNα2b treatment, increased SDF-1/CXCR4 signaling was found prior to treatment which was down-regulated after IFNα2b administration, coincident with enhanced remodeling of their HTS. Our results suggest that SDF-1/CXCR4 signaling is involved in the development of HTS by promoting migration of activated CD14+ CXCR4+ cells from the bloodstream to wound sites, where they may differentiate into fibrocyte and myofibroblasts and contribute to the development of HTS.


Subject(s)
Burns/metabolism , Chemokine CXCL12/metabolism , Cicatrix, Hypertrophic/metabolism , Receptors, CXCR4/metabolism , Adult , Burns/complications , Burns/pathology , Cell Migration Assays , Cicatrix, Hypertrophic/etiology , Dermis/pathology , Down-Regulation , Enzyme-Linked Immunosorbent Assay , Female , Fibroblasts/metabolism , Flow Cytometry , Humans , Interferon alpha-2 , Interferon-alpha/pharmacology , Male , Middle Aged , Real-Time Polymerase Chain Reaction , Recombinant Proteins/pharmacology , Signal Transduction , Skin/metabolism , Skin/pathology , Young Adult
11.
Wound Repair Regen ; 19(3): 368-78, 2011.
Article in English | MEDLINE | ID: mdl-21518082

ABSTRACT

Small leucine-rich proteoglycans (SLRPs) are extracellular matrix molecules that regulate collagen fibrillogenesis and inhibit transforming growth factor-ß activity; thus, they may play a critical role in wound healing and scar formation. Hypertrophic scarring is a dermal form of fibroproliferative disorders, which occurs in over 70% of burn patients and leads to disfigurement and limitations in function. By understanding the cellular and molecular mechanisms that lead to scarring after injury, new clinical therapeutic approaches can by developed to minimize abnormal scar formation in hypertrophic scarring and other fibroproliferative disorders. To study the expression and localization of SLRPs with connective tissue cells in tissue immunohistochemistry, immunofluorescence staining, immunoblotting, and reverse-transcription polymerase chain reaction were used in normal skin and hypertrophic scar (HTS). In normal skin, there was more decorin and fibromodulin accumulation in the superficial layers than in the deeper dermal layers. The levels of decorin and fibromodulin were significantly lower in HTS, whereas biglycan was increased when compared with normal skin. There was an increased expression of biglycan, fibromodulin, and lumican in the basement membrane and around basal epithelial cells. In contrast, these proteoglycans were absent or weakly expressed in HTS. The findings suggest that down-regulation of SLRPs after wound healing in deep injuries to the skin plays an important role in the development of fibrosis and HTS.


Subject(s)
Cicatrix, Hypertrophic/metabolism , Decorin/metabolism , Extracellular Matrix Proteins/metabolism , Proteoglycans/metabolism , Wound Healing/physiology , Adult , Biglycan/metabolism , Blotting, Western , Burns/complications , Burns/metabolism , Child , Cicatrix, Hypertrophic/etiology , Down-Regulation/physiology , Female , Fibroblasts/physiology , Fibromodulin , Fluorescent Antibody Technique , Humans , Male , Microscopy, Confocal , Middle Aged , Reverse Transcriptase Polymerase Chain Reaction
12.
Wound Repair Regen ; 19(2): 274-85, 2011.
Article in English | MEDLINE | ID: mdl-21362096

ABSTRACT

Hypertrophic scar (HTS) following thermal injury and other forms of trauma is a dermal fibroproliferative disorder that leads to considerable morbidity. Because of the lack of an ideal animal model, research is difficult. We have established an HTS model that involves transplanting human split-thickness skin graft (STSG) or full-thickness skin graft (FTSG) onto the backs of nude mice. The animals developed raised, firm, and reddish scars 2 months following transplantation. Histology and micromeasurement indicate raised, thickened engrafted skin with STSG and FTSG. In contrast, thickening was not observed with full-thickness rat skin grafts used as controls. Masson's trichrome staining demonstrates increased accumulations of collagen fibrils in the dermis in both scars grafted with STSG and FTSG. Staining cells with toludine blue and an antibody for F4/80 showed an increase in the infiltration of mast cells and macrophages. Quantification of fibrocytes reveals increased fibrocytes. Moreover, STSG grafted skin had significantly more macrophages, mast cells, and fibrocytes than FTSG. Real-time polymerase chain reaction analysis showed significantly elevated mRNA levels for type I collagen, transforming growth factor-ß, connective tissue growth factor and heat shock protein 47 in both types of engrafted skin. These data demonstrate that human skin grafted onto nude mice develops red raised and thickened scars having intrinsic properties that closely resemble HTS formation as seen in humans. Interestingly, STSG developed more scar than FTSG. Furthermore, inflammatory cells and bone marrow-derived fibrocytes may play a critical role in HTS development in this animal model.


Subject(s)
Cicatrix, Hypertrophic/metabolism , Cicatrix, Hypertrophic/pathology , Models, Animal , Animals , Collagen Type I/metabolism , Connective Tissue Growth Factor/metabolism , HSP47 Heat-Shock Proteins/metabolism , Humans , Immunohistochemistry , Macrophages/pathology , Mast Cells/pathology , Mice , Mice, Inbred BALB C , Mice, Nude , Myofibroblasts/pathology , Skin/metabolism , Skin Transplantation , Transforming Growth Factor beta1/metabolism , Transplantation, Heterologous
13.
Lab Invest ; 88(12): 1278-90, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18955978

ABSTRACT

Hypertrophic scar (HTS) following thermal injury is a dermal fibroproliferative disorder that leads to considerable morbidity. The development of HTS involves numerous cell types and cytokines with dermal fibroblasts being a key cell. We have previously reported that the phenotype of fibroblasts isolated from HTS was altered compared to fibroblasts from normal skin. In this study, normal skin was horizontally sectioned into five layers using a dermatome from which fibroblasts were isolated and cultured. Cells from the deeper layers were observed to proliferate at a slow rate, but were morphologically larger. In ELISA and FACS assays, cells from the deeper layers produced more TGF-beta1 and TGF-beta1 producing cells were higher. In quantitative RT-PCR, the cells from the deeper layers had higher CTGF and HSP47 mRNA levels compared to those from superficial layers. In western blot, FACS and collagen gel assays, fibroblasts from the deeper layers produced more alpha-smooth muscle actin (alpha-SMA), had higher alpha-SMA positive cells and contracted collagen gels more. Fibroblasts from the deeper layers were also found to produce more collagen, but less collagenase by mass spectrometry and collagenase assay. Interestingly, cells from the deeper layers also produced more of the proteoglycan, versican, but less decorin. Taken together, these data strongly demonstrate that fibroblasts from the deeper layers of the dermis resemble HTS fibroblasts, suggesting that the deeper layer fibroblasts may be critical in the formation of HTS.


Subject(s)
Cicatrix, Hypertrophic/pathology , Skin/pathology , Adolescent , Adult , Base Sequence , Case-Control Studies , Cell Proliferation , Cell Separation , Cell Size , Child, Preschool , Cicatrix, Hypertrophic/metabolism , Collagenases/metabolism , DNA Primers , Enzyme-Linked Immunosorbent Assay , Female , Fibroblasts/cytology , Fibroblasts/metabolism , Flow Cytometry , Humans , Male , Middle Aged , Reverse Transcriptase Polymerase Chain Reaction , Skin/metabolism , Transforming Growth Factor beta1/biosynthesis
14.
J Interferon Cytokine Res ; 28(7): 423-34, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18597619

ABSTRACT

Hypertrophic scar (HTS) after thermal injury is a dermal fibroproliferative disorder, which leads to considerable morbidity. Previous clinical studies from our laboratory have suggested that interferon-alpha2b (IFN-alpha2b) improves scar quality as a result of the suppression of fibroblast function. More recently, our work has demonstrated that the improvement of scar in patients with HTS after IFN-alpha2b treatment may be associated with a decreased number of fibrocytes and/or altered fibrocyte function. In this study, we report that the improvement of HTS after IFNalpha-2b treatment may be associated with a decrease in angiogenesis. Using immunohistochemistry, we demonstrate an increase in angiogenesis in HTS compared to normal skin, and also show an increase in the expression of vascular endothelial cell growth factor (VEGF) in HTS. Subsequently, we demonstrate a significant reduction in angiogenesis in HTS tissue from patients after treatment with systemic IFN-alpha2b. By using a [3H] thymidine incorporation assay, we demonstrate that IFN-alpha2b suppresses the proliferation of human umbilical vein endothelial cells (HUVECs) in a dose-dependent manner. In addition, IFN-alpha2b inhibits VEGF-induced proliferation and tube formation by using HUVECs. All these effects may be a result of the blocking of VEGF receptor expression on endothelial cells by IFN-alpha2b. Taken together with previous results, the present study suggests that the improvement of scar quality in HTS patients after IFN-alpha2b treatment may also be associated with decreased angiogenesis in HTS. The current in vitro results may provide some insights into the scar improvement that is seen with systemic IFN-alpha2b treatment.


Subject(s)
Cicatrix, Hypertrophic/drug therapy , Interferon-alpha/administration & dosage , Neovascularization, Pathologic/drug therapy , Adolescent , Adult , Burns/pathology , Cell Proliferation/drug effects , Cells, Cultured , Child , Cicatrix, Hypertrophic/metabolism , Cicatrix, Hypertrophic/pathology , Cicatrix, Hypertrophic/physiopathology , Dose-Response Relationship, Drug , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Endothelial Cells/pathology , Female , Humans , Interferon alpha-2 , Male , Middle Aged , Neovascularization, Pathologic/pathology , Recombinant Proteins , Skin/blood supply , Skin/drug effects , Skin/metabolism , Skin/pathology , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Vascular Endothelial Growth Factor A/biosynthesis , Vascular Endothelial Growth Factor Receptor-2/antagonists & inhibitors , Vascular Endothelial Growth Factor Receptor-2/biosynthesis
15.
J Invest Dermatol ; 128(12): 2767-76, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18580965

ABSTRACT

Fibrosis is a complex process resulting from persistent inflammation following tissue damage. It involves the interaction of numerous cell types, soluble mediators, and extracellular matrix. Recently, a newly identified cell type, the fibrocyte, has been reported to contribute to wound healing and to fibrotic conditions such as hypertrophic scarring. Previously, we established leukocyte-specific protein 1 (LSP1) as a new marker for fibrocytes. In the present study, we examined the biological role of LSP1 in the development of skin fibrosis using bleomycin in an Lsp1(-/-) mice. These animals showed a significant increase in fibrosis, with increased thickness of the skin and collagen content. The skin in Lsp1(-/-) mice injected with bleomycin had higher densities of neutrophils, macrophages, and fibrocytes. In accordance with the increased leukocyte infiltration, the expression levels of macrophage-derived chemokines, transforming growth factor-beta1, and connective tissue growth factor were all upregulated in Lsp1(-/-) mice. These results demonstrate that the absence of LSP1 promotes fibrosis in the skin. The most likely mechanism for this effect seems to be through an increase in leukocyte infiltration, leading to locally elevated synthesis and the release of chemokines and growth factors.


Subject(s)
Bleomycin/pharmacology , Blood Proteins/deficiency , Fibrosis/chemically induced , Gene Expression Regulation , Skin/drug effects , Animals , Antibiotics, Antineoplastic/pharmacology , Blood Proteins/genetics , Blood Proteins/physiology , Connective Tissue Growth Factor/biosynthesis , Fibroblasts/metabolism , Immunohistochemistry/methods , Inflammation , Intracellular Signaling Peptides and Proteins , Leukocytes/cytology , Membrane Proteins , Mice , Mice, Transgenic , Skin/pathology , Transforming Growth Factor beta1/metabolism
16.
J Interferon Cytokine Res ; 27(11): 921-30, 2007 Nov.
Article in English | MEDLINE | ID: mdl-18052725

ABSTRACT

Hypertrophic scar (HTS) following thermal injury is a dermal fibroproliferative disorder that leads to considerable morbidity. Previous clinical studies from our laboratory have suggested that interferon-alpha2b (IFN-alpha2b) improves scar quality as a result of suppression of fibroblast functions. Fibrocytes, which constitute a unique cell population, have recently been reported to contribute to wound healing and to a variety of fibrotic conditions, including HTS. Therefore, we hypothesize that improvement of scar in HTS patients after IFN-alpha2b treatment may be associated with a decreased number of fibrocytes or altered fibrocyte function. Using flow cytometry, immunofluorescent staining, and confocal microscopy, we demonstrate here that the marker protein leukocyte-specific protein 1 (LSP1) is stably expressed by fibrocytes for at least 2 months, whereas other potential fibrocyte markers, such as CD34 and CD45, gradually disappear. Using dual staining immunohistochemistry for LSP1 and procollagen, we demonstrated a significant reduction in numbers of fibrocytes in HTS tissue from patients after treatment with systemic IFN-alpha2b. IFN-alpha2b was shown to abolish fibrocyte differentiation from peripheral blood mononuclear cells (PBMCs) in vitro in a dose-dependent fashion. In addition, IFN-alpha2b inhibits proliferation of fibrocytes and T lymphocytes and reduces transforming growth factor-beta (TGF-beta)-mediated alpha-smooth muscle actin (alpha-SMA) expression in fibrocytes. Taken together with our previous study in which we showed that fibrocytes could indirectly regulate dermal fibroblasts in burn patients, the present study suggests that the improvement of scar quality in HTS patients after IFN-alpha2b treatment is associated with decreased numbers and activities of fibrocytes.


Subject(s)
Burns/drug therapy , Cicatrix, Hypertrophic/drug therapy , Cicatrix, Hypertrophic/pathology , Interferon-alpha/therapeutic use , Leukocytes/physiology , Actins/metabolism , Adolescent , Adult , Burns/complications , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Child , Cicatrix, Hypertrophic/metabolism , Female , Humans , Interferon alpha-2 , Interferon-alpha/metabolism , Leukocytes/cytology , Male , Microfilament Proteins/metabolism , Middle Aged , Procollagen/metabolism , Recombinant Proteins , Transforming Growth Factor beta/metabolism , Wound Healing/drug effects
17.
J Leukoc Biol ; 82(6): 1554-63, 2007 Dec.
Article in English | MEDLINE | ID: mdl-18040084

ABSTRACT

Wound healing is a complex process involving the integrated actions of numerous cell types, soluble mediators, and ECM. Recently, a newly identified cell type, the fibrocyte, has been reported to contribute to wound healing and fibrotic conditions such as hypertrophic scarring. We previously established leukocyte-specific protein 1 (LSP1) as a marker for fibrocytes. LSP1 is an F-actin binding protein and substrate of p38 mitogen-activated protein kinase and protein kinase C, and has been reported to be important in leukocyte chemotaxis. We examine the biological roles of LSP1 in skin wound healing using Lsp1(-/-) null mice. These animals showed accelerated healing of full-thickness skin wounds, with increased re-epithelialization rates, collagen synthesis, and angiogenesis. Healing wounds in Lsp1(-/-) mice had higher densities of neutrophiles, macrophages, and fibrocytes. Along with increased leukocyte infiltration, levels of macrophage-derived chemokine expression, TGF-beta1, and VEGF were all up-regulated. These results demonstrate that the absence of LSP1 promotes healing of skin wounds. The primary mechanism seems to be an increase in leukocyte infiltration, leading to locally elevated synthesis and release of chemokines and growth factors. Further analysis of Lsp1(-/-) mice may suggest ways to improve wound healing and/or treat fibrotic conditions of skin and other tissue.


Subject(s)
Calcium-Binding Proteins/deficiency , Leukocytes/cytology , Leukocytes/immunology , Wound Healing/physiology , Animals , Cell Count , Cell Proliferation , Chemokines/genetics , Chemokines/metabolism , Collagen/biosynthesis , Epithelium/pathology , Gene Expression Regulation , Keratinocytes/cytology , Keratinocytes/immunology , Macrophages/cytology , Macrophages/immunology , Mice , Microfilament Proteins , Neovascularization, Pathologic , Neutrophil Infiltration , Skin/immunology , Skin/pathology , Transforming Growth Factor beta/metabolism
18.
J Trauma ; 63(1): 164-71, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17622885

ABSTRACT

BACKGROUND: Nosocomially-acquired Pseudomonas aeruginosa remains a serious cause of infection and septic mortality in burn patients. This study was conducted to quantify the impact of nosocomially-transmitted resistant P. aeruginosa in a burn population. METHODS: Using a TRACS burn database, 48 patients with P. aeruginosa resistant to gentamicin were identified (Pseudomonas group). Thirty-nine were case-matched to controls without resistant P. aeruginosa cultures (control group) for age, total body surface area, admission year, and presence of inhalation injury. Mortality and various morbidity endpoints were examined, as well as antibiotic costs. RESULTS: There was a significantly higher mortality rate in the Pseudomonas group (33% vs. 8%, p < 0.001) compared with in the control group. Length of stay was increased in the Pseudomonas group (73.4 +/- 11.6 vs. 58.3 +/- 8.3 days). Ventilatory days (23.9 +/- 5.4 vs. 10.8 +/- 2.4, p < 0.05), number of surgical procedures (5.2 +/- 0.6 vs. 3.4 +/- 0.4, p < 0.05), and amount of blood products used (packed cells 51.1 +/- 8.0 vs. 21.1 +/- 3.4, p < 0.01; platelets 11.9 +/- 3.0 vs. 1.4 +/- 0.7, p < 0.01) were all significantly higher in the Pseudomonas group. Cost of antibiotics was also significantly higher ($2,658.52 +/- $647.93 vs. $829.22 +/- $152.82, p < 0.01). CONCLUSIONS: Nosocomial colonization or infection, or both, of burn patients with aminoglycoside-resistant P. aeruginosa is associated with significantly higher morbidity, mortality, and cost of care. Increased resource consumption did not prevent significantly higher mortality rates when compared with that of control patients. Thus, prevention, identification, and eradication of nosocomial Pseudomonas contamination are critical for cost-effective, successful burn care.


Subject(s)
Burns/microbiology , Pseudomonas Infections , Adult , Alberta , Anti-Bacterial Agents/economics , Anti-Bacterial Agents/pharmacology , Burns/economics , Burns/mortality , Case-Control Studies , Cross Infection , Crown-Rump Length , Female , Gentamicins/pharmacology , Humans , Male , Pseudomonas Infections/economics , Pseudomonas Infections/mortality , Pseudomonas aeruginosa/drug effects , Respiration, Artificial
19.
Wound Repair Regen ; 15(4): 530-9, 2007.
Article in English | MEDLINE | ID: mdl-17650097

ABSTRACT

The development of hypertrophic scar involves a complex interplay between cells and cytokines. Although the mechanism underlying its pathogenesis is not well understood, a polarized T-helper type 2 immune response has been reported, indicating a role for CD4+ T lymphocytes in hypertrophic scarring. Here, we report an increased frequency of CD4+/transforming growth factor-beta (TGF-beta)-producing T cells in the peripheral blood and hypertrophic scar tissue of burn patients. These cells may play an indirect regulatory role in hypertrophic scar by affecting the functions of dermal fibroblasts. Our results show an increase in cell proliferation and collagen synthesis by dermal fibroblasts treated with medium derived from burn patient CD4+ T lymphocytes but not from the CD4+ T cells of normal subjects. Using confocal microscopy and immunoblotting, we found the level of alpha-smooth muscle actin to be elevated in these treated dermal fibroblasts, which also showed an enhanced ability to contract collagen lattices. TGF-beta levels in medium conditioned by the culture of CD4+ T lymphocytes from burn patients were significantly higher than in the conditioned medium from CD4+ T lymphocytes of normal subjects. In addition, the application of a TGF-beta-neutralizing antibody significantly reduced the effect of burn patient CD4+ T lymphocyte medium on dermal fibroblast proliferation and collagen lattice contraction. Our study suggests that CD4+/TGF-beta-producing T lymphocytes may play an important role in postburn hypertrophic scarring.


Subject(s)
Burns/metabolism , CD4-Positive T-Lymphocytes/metabolism , Cicatrix, Hypertrophic/metabolism , Fibroblasts/physiology , Transforming Growth Factor beta/metabolism , Adolescent , Adult , Burns/blood , Cell Proliferation , Child, Preschool , Cicatrix, Hypertrophic/blood , Collagen/metabolism , Culture Media, Conditioned , Female , Flow Cytometry , Humans , Male , Microscopy, Confocal , Skin/cytology , Up-Regulation/physiology
20.
Wound Repair Regen ; 15(1): 113-21, 2007.
Article in English | MEDLINE | ID: mdl-17244327

ABSTRACT

Wound healing requires an elaborate interplay between numerous cell types that orchestrate a series of regulated and overlapping events. Fibrocytes are a unique leukocyte subpopulation implicated in this process. One role proposed for these cells in wound healing is to synthesize extracellular matrix. Interestingly, using mass spectrometry to quantify hydroxyproline, we discovered that the capacity of fibrocytes from normal subjects or from burn patients to produce collagen is much less than that of dermal fibroblasts. Therefore, we investigated whether fibrocytes could play an indirect, regulatory, role in the healing of burn wounds by affecting the functions of dermal fibroblasts. Dermal fibroblasts treated with medium conditioned by burn patient fibrocytes, but not by those derived from normal subjects, showed an increase in cell proliferation and migration. Using confocal microscopy, flow cytometry, and immunoblotting, we found the level of alpha-smooth muscle actin (alpha-SMA) expression to be increased in these treated dermal fibroblasts, which also showed an enhanced ability to contract collagen lattices. To determine whether these effects could be attributed to transforming growth factor beta (TGF-beta1) or to connective tissue growth factor (CTGF), we measured total TGF-beta1 levels in the conditioned medium by an enzyme-linked immunosorbtion assay and assessed levels of CTGF mRNA and protein in fibroblasts and fibrocytes by reverse transcription-polymerase chain reaction and Western blotting. The results showed significantly higher levels of TGF-beta1 and CTGF produced by burn patient fibrocytes. In addition, the application of a TGF-beta1 neutralizing antibody significantly reduced the effect of burn patient fibrocyte medium on dermal fibroblast proliferation, migration, and collagen lattice contraction. Our results suggest that in healing burn wounds, fibrocytes could regulate the activities of local fibroblasts.


Subject(s)
Burns/pathology , Burns/physiopathology , Cell Communication/physiology , Fibroblasts/physiology , Leukocytes/physiology , Wound Healing/physiology , Adult , Burns/metabolism , Case-Control Studies , Cell Culture Techniques , Cell Movement/physiology , Cell Proliferation , Connective Tissue Growth Factor , Culture Media, Conditioned , Female , Humans , Hydroxyproline/metabolism , Immediate-Early Proteins/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Male , Transforming Growth Factor beta1/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...