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










Publication year range
1.
Ann Dermatol ; 27(5): 578-92, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26512172

ABSTRACT

BACKGROUND: Since the treatment guidelines for atopic dermatitis (AD) were issued by the Korean Atopic Dermatitis Association (KADA) work group in 2006, there have been further advances in the systemic treatment of AD. OBJECTIVE: We aimed to establish updated evidence- and experience-based systemic treatment guidelines for Korean AD. METHODS: We compiled a database of references from relevant systematic reviews and guidelines regarding the systemic management of AD, including antihistamines, antimicrobials, systemic immunomodulators, allergen-specific immunotherapy, phototherapy, adjunctive treatment, and complementary and alternative medicines. Evidence for each statement was graded and classified based on the strength of the recommendation. Thirty-nine council members of KADA participated in the three rounds of votes and expert consensus recommendations were established. RESULTS: The use of antihistamines is recommended to relieve pruritus and to prevent exacerbation due to scratching in AD patients. Infection should be controlled as needed and long-term medication should be avoided. For moderate to severe AD patients, concomitant active treatments with systemic immunomodulators are indicated. Cyclosporine is the first choice among systemic immunomodulators and others should be considered as second-line alternatives. Allergen-specific immunotherapy could be effective in AD patients with aeroallergen hypersensitivity. Phototherapy can be useful for moderate to severe AD patients and narrow-band ultraviolet B is the most effective option. Complementary and alternative medicines cannot be recommended for treating AD. CONCLUSION: We expect these recommendations to be a reference guide for physicians and AD patients in choosing the appropriate treatment to improve quality of life and decrease unnecessary social medical costs.

2.
Ann Dermatol ; 27(5): 563-77, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26512171

ABSTRACT

BACKGROUND: Since the treatment guidelines for atopic dermatitis (AD) were released by the Korean Atopic Dermatitis Association (KADA) work group in 2006, there have been several advances in AD management. OBJECTIVE: We aimed to establish updated evidence- and experience-based treatment guidelines for Korean AD. METHODS: We collected a database of references from relevant systematic AD reviews and guidelines regarding general AD management such as bathing and skin care, avoidance of exacerbating factors, education and psychosocial support, and the use of moisturizers and topical anti-inflammatory and antipruritic drugs. Evidence for each statement was graded and the strength of the recommendation for each statement classified. Thirty-nine KADA council members participated in three rounds of voting to establish an expert consensus of recommendations. RESULTS: Basic AD treatment includes proper bathing and skin care, avoidance of exacerbating factors, proper education and psychosocial support, and use of moisturizers. The regular use of moisturizer has a steroid-sparing effect and reduces relapse episodes. The short- and long-term use of topical corticosteroids and calcineurin inhibitors improves AD symptoms and should be encouraged to use in an active and proactive treatment. Wet-wrap therapy can be used for rapid recovery of acute exacerbation. Topical antipruritic drugs cannot be recommended for the treatment of AD. CONCLUSION: This report provides up-to-date evidence- and experience-based treatment guidelines for AD regarding general management and topical treatment. In addition, the average agreement scores obtained by a panel of experts based on the Korean healthcare system and patient adherence are presented.

3.
Exp Dermatol ; 16(4): 311-7, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17359337

ABSTRACT

In a search for the wound healing accelerators, we found that tetraacetyl-phytosphingosine (TAPS), a sphingolipid metabolite produced by phytosphingosine acetylation, has significant inhibitory potential on healing of rabbit ear wound. As angiogenesis is fundamental to proper wound healing, we examined the effect of TAPS on angiogenesis using human umbilical vein endothelial cells cultured in vitro. TAPS markedly decreased vascular endothelial growth factor (VEGF)-induced chemotactic migration and capillary-like tube formation. Recognizing its inhibitory potential on angiogenesis, we further investigated the action mechanism of TAPS. TAPS significantly inhibited VEGF-induced proteolytic enzyme production, including matrix metalloproteinase-2, urokinase-type plasminogen activator and plasminogen activator inhibitor-1. TAPS also suppressed VEGF-induced phosphorylation of p42/44 extracellular signal-regulated kinase and c-Jun N-terminal kinase. In addition, TAPS abolished VEGF-induced intracellular calcium increase, measured using laser scanning confocal microscopy. Together, these results suggest that TAPS exerts its inhibitory action on angiogenesis through the inhibition of mitogen-activated protein kinase activation and intracellular calcium increase, thereby affecting the process of wound healing negatively.


Subject(s)
Endothelial Cells/drug effects , Neovascularization, Physiologic/drug effects , Sphingosine/analogs & derivatives , Wound Healing/drug effects , Acetylation , Animals , Calcium/metabolism , Cell Movement , Cells, Cultured , Endothelial Cells/cytology , Endothelial Cells/enzymology , Extracellular Signal-Regulated MAP Kinases/metabolism , Humans , JNK Mitogen-Activated Protein Kinases/metabolism , Matrix Metalloproteinase 2/metabolism , Phosphorylation/drug effects , Plasminogen Activator Inhibitor 1/metabolism , Rabbits , Signal Transduction/drug effects , Sphingosine/chemistry , Sphingosine/pharmacology , Umbilical Veins/cytology , Urokinase-Type Plasminogen Activator/metabolism , Vascular Endothelial Growth Factor A/pharmacology
4.
J Dermatol Sci ; 46(2): 91-9, 2007 May.
Article in English | MEDLINE | ID: mdl-17321112

ABSTRACT

BACKGROUND: Sphingosylphosphorylcholine (SPC) has been reported as a novel lipid mediator that exerts various actions on wound healing process. OBJECTIVE: The aim of this study is to evaluate the involvement of interleukin-6 (IL-6) in SPC-induced wound healing acceleration. METHODS: We performed immunohistochemical analysis to demonstrate the IL-6 induction by SPC. To analyze the signaling events, skin fibroblasts were treated with SPC, and then RT-PCR, ELISA and Western blot analyses were carried out. RESULTS: SPC markedly induced interleukin-6 (IL-6) expression in rabbit ear wound. SPC also induced IL-6 expression at both the mRNA and protein levels in human dermal fibroblasts cultured in vitro. SPC rapidly phosphorylated p42/44 extracellular signal-regulated kinase (ERK). Pretreatment with PD 98059, a specific MAPK kinase 1/2 inhibitor, markedly suppressed SPC-induced IL-6 expression in a dose-dependent manner. Protein kinase C (PKC) activation by phorbol myristate acetate (PMA) potentiated IL-6 mRNA expression, whereas PKC inhibition by bisindolylmaleimide blocked SPC-induced p42/44 ERK phosphorylation and IL-6 expression. Over-expression of PKCalpha markedly induced the IL-6 expression and p42/44 ERK activation. CONCLUSION: These results suggest that SPC-induced IL-6 production is mediated by PKC-dependent p42/44 ERK activation in human dermal fibroblasts cultured in vitro.


Subject(s)
Dermis/metabolism , Fibroblasts/metabolism , Interleukin-6/biosynthesis , MAP Kinase Signaling System , Phosphorylcholine/analogs & derivatives , Protein Kinase C/metabolism , Sphingosine/analogs & derivatives , Wound Healing , Animals , Cells, Cultured , Dermis/cytology , Dermis/drug effects , Dose-Response Relationship, Drug , Enzyme Activation , Epidermis/metabolism , Epidermis/surgery , Fibroblasts/drug effects , Flavonoids/pharmacology , Humans , Indoles/pharmacology , Interleukin-6/genetics , MAP Kinase Signaling System/drug effects , Maleimides/pharmacology , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Phosphorylation , Phosphorylcholine/metabolism , Phosphorylcholine/pharmacology , Protein Kinase C/genetics , Protein Kinase C-alpha/metabolism , Protein Kinase Inhibitors/pharmacology , RNA, Messenger/biosynthesis , Rabbits , Sphingosine/metabolism , Sphingosine/pharmacology , Tetradecanoylphorbol Acetate/pharmacology , Time Factors , Transfection , Wound Healing/drug effects
5.
J Biomed Opt ; 11(4): 041124, 2006.
Article in English | MEDLINE | ID: mdl-16965152

ABSTRACT

We use polarization-sensitive optical coherence tomography (PS-OCT) to monitor the wound healing process in vitro and in vivo, which are affected by various drugs. Five rabbit subjects are used for in vitro studies and another five are used for in vivo studies. The in vitro studies are conducted to compare the PS-OCT images with histopathology. For each subject, three biopsy lesions are created on each ear: one site is not treated (control); the second site is treated with sphingosylphosphorylcholine, which is expected to promote healing; and the last is administered with tetraacetylphytosphingosine, which negatively affects the healing process. Each site is examined with a PS-OCT system at 1, 4, 7, 10, and 14- days after wound generation. The variations of phase retardation values caused by the collagen morphology changes on wound sites are quantified for all cases. Our results suggest that PS-OCT may be a useful tool for visualization of collagen fiber regeneration and for quantification of various drug effects during the wound healing process.


Subject(s)
Image Enhancement/methods , Image Interpretation, Computer-Assisted/methods , Microscopy, Polarization/methods , Tomography, Optical Coherence/methods , Wound Healing/physiology , Wounds, Penetrating/pathology , Animals , Phosphorylcholine/administration & dosage , Phosphorylcholine/analogs & derivatives , Rabbits , Reproducibility of Results , Sensitivity and Specificity , Sphingosine/administration & dosage , Sphingosine/analogs & derivatives , Treatment Outcome , Wound Healing/drug effects , Wounds, Penetrating/drug therapy , Wounds, Penetrating/physiopathology
6.
Exp Dermatol ; 14(7): 509-14, 2005 Jul.
Article in English | MEDLINE | ID: mdl-15946238

ABSTRACT

Sphingosylphosphorylcholine (SPC) is a bioactive sphingolipid metabolite that can enhance wound healing. In an effort to find downstream effectors of SPC, we performed microarray analysis and found that the expression of the gene for connective tissue growth factor (CTGF) was significantly affected in human skin fibroblasts cultured in vitro. Northern blot analysis showed that SPC markedly induced CTGF mRNA expression in a dose- and time-dependent manner. Consistent with this result, Western blot analysis also showed that SPC significantly induced the CTGF production. Pretreatment with cycloheximide did not prevent the CTGF induction by SPC, indicating that SPC stimulates CTGF mRNA expression without the increased synthesis of a regulatory protein. Inhibition by pretreatment with Y27632, but not by PD98059 (a mitogen-activated protein kinase 1/2 inhibitor) and LY294002 (a phosphatidylinositol 3-kinase inhibitor), indicated that rho-kinase pathway was involved in SPC-induced CTGF expression. Together, these results reveal the potential importance of CTGF induction as a downstream event in SPC-induced cellular responses.


Subject(s)
Immediate-Early Proteins/genetics , Intercellular Signaling Peptides and Proteins/genetics , Phosphorylcholine/analogs & derivatives , Skin/drug effects , Skin/metabolism , Sphingosine/analogs & derivatives , Base Sequence , Cells, Cultured , Connective Tissue Growth Factor , DNA, Complementary/genetics , Fibroblasts/drug effects , Fibroblasts/metabolism , Gene Expression/drug effects , Gene Expression Profiling , Humans , Oligonucleotide Array Sequence Analysis , Phosphorylcholine/metabolism , Phosphorylcholine/pharmacology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Signal Transduction , Sphingosine/metabolism , Sphingosine/pharmacology
7.
Exp Dermatol ; 14(5): 356-62, 2005 May.
Article in English | MEDLINE | ID: mdl-15854129

ABSTRACT

Sphingosylphosphorylcholine (SPC) has been shown to accelerate wound healing. As angiogenesis is fundamental to proper wound healing, we examined the effect of SPC on angiogenesis using a well-established rat aortic ring assay. SPC significantly stimulated the sprouting of endothelial cells from rat aortic ring. Recognizing its potential effect on angiogenesis, we further investigated the action of SPC using human umbilical vein endothelial cells (HUVECs) cultured in vitro. SPC significantly accelerated the closure of in vitro wound. In addition, SPC markedly enhanced the chemotactic migration and capillary-like tube formation. Subsequently, we examined whether SPC affected the production of urokinase-type plasminogen activator (uPA), an important regulator of angiogenesis, and found that SPC stimulated the expression of uPA at both the transcriptional and translational levels. Consistent with these results, SPC increased the activity of cell-surface-associated plasminogen activator. Pretreatment with antiuPA antibody significantly diminished both the chemotactic migration and capillary-like tube formation, indicating the potential importance of uPA in SPC-induced angiogenesis. Together, these results suggest that SPC may affect angiogenesis in the wound-healing process via regulation of uPA production.


Subject(s)
Endothelium, Vascular/enzymology , Neovascularization, Physiologic/drug effects , Neovascularization, Physiologic/physiology , Phosphorylcholine/analogs & derivatives , Phosphorylcholine/pharmacology , Sphingosine/analogs & derivatives , Sphingosine/pharmacology , Urokinase-Type Plasminogen Activator/metabolism , Animals , Aorta/drug effects , Aorta/enzymology , Capillaries/cytology , Capillaries/drug effects , Cell Movement/drug effects , Cells, Cultured , Endothelium, Vascular/cytology , Endothelium, Vascular/drug effects , Female , Humans , In Vitro Techniques , Rats , Rats, Inbred F344 , Umbilical Veins/cytology , Wound Healing/drug effects
8.
J Invest Dermatol ; 122(6): 1365-71, 2004 Jun.
Article in English | MEDLINE | ID: mdl-15175025

ABSTRACT

Sphingosylphosphorylcholine (SPC) is a bioactive sphingolipid metabolite that can enhance wound healing. In a search for effectors downstream of SPC in the wound-healing process, we found that the expression of the gene for plasminogen activator inhibitor-1 (PAI-1) was significantly affected. ELISA and western blot analyses showed that SPC markedly induced PAI-1 production in human dermal fibroblasts cultured in vitro. Inhibition by pre-treatment with pertussis toxin (PTx), but not by tyrphostin A47 (a receptor tyrosine kinase inhibitor), indicated that PTx-sensitive G proteins were involved in SPC-induced PAI-1 expression. SPC elicited a rapid and transient increase in intracellular calcium levels ([Ca2+]i), measured using laser scanning confocal microscopy, which was partly mediated through PTx-sensitive G proteins. Pre-treatment with thapsigargin, but not with EGTA, abolished SPC-induced PAI-1 expression, indicating the importance of Ca2+ release from internal stores. Phorbol-12-myristate-13-acetate (PMA) induced the expression of PAI-1, and pre-treatment with Ro 31-8220 (a PKC inhibitor) markedly suppressed SPC-induced PAI-1 expression. SPC-induced PAI-1 expression was also significantly suppressed by PD98059 (a specific MAPK kinase 1/2 inhibitor). Consistent with this result, SPC stimulated the phosphorylation of p42/44 extracellular signal-regulated kinase (ERK). Together, these results suggest that SPC induces PAI-1 production through a G protein-coupled calcium increase and downstream kinase signaling events in cultured human dermal fibroblasts.


Subject(s)
Dermis/cytology , Fibroblasts/drug effects , Fibroblasts/physiology , Phosphorylcholine/analogs & derivatives , Phosphorylcholine/pharmacology , Plasminogen Activator Inhibitor 1/genetics , Sphingosine/analogs & derivatives , Sphingosine/pharmacology , Animals , Calcium/metabolism , Cells, Cultured , Fibroblasts/cytology , GTP-Binding Proteins/metabolism , Gene Expression/drug effects , Humans , MAP Kinase Signaling System/drug effects , Plasminogen Activator Inhibitor 1/metabolism , Protein Kinase C/metabolism , Rabbits , Wound Healing/drug effects , Wound Healing/physiology
9.
Biochemistry ; 42(42): 12181-91, 2003 Oct 28.
Article in English | MEDLINE | ID: mdl-14567679

ABSTRACT

The lysophospholipids, lysophosphatidic acid, sphingosine-1-phosphate, and sphingosylphosphorylcholine (SPC), are bioactive lipid molecules that regulate diverse biological processes. Although the specific G protein-coupled receptors for lysophosphatidic acid and sphingosine-1-phosphate have been well-characterized, much less is known of the SPC receptors. It has been reported that ovarian cancer G protein-coupled receptor 1 (OGR1) is a high affinity receptor for SPC, and its closely related homologue GPR4 is a high affinity receptor for SPC with low affinity for lysophosphatidylcholine (LPC). However, in a functional assay to examine the specificity of ligand binding, we found that neither SPC nor LPC, or other related lysophospholipids, induced internalization of GPR4 from the plasma membrane. In agreement, these lysolipids also did not induce translocation of beta-arrestin2-GFP from the cytosol to the plasma membrane in GPR4 expressing cells. However, when these cells were cotransfected with G protein-coupled receptor kinase 2, in the absence of added ligands, beta-arrestin2-GFP accumulated in cytoplasmic vesicles, reminiscent of vesicular labeling usually observed after agonist stimulation of GPCRs. In addition, neither SPC nor LPC stimulated the binding of GTPgammaS to membranes prepared from GPR4 expressing cells and did not activate ERK1/2. Surprisingly, enforced expression of GPR4 inhibited activation of ERK1/2 induced by several stimuli, including SPC, sphingosine-1-phosphate, and even EGF. Collectively, our results suggest that SPC and LPC are not the ligands for GPR4 and that this receptor may constitutively inhibit ERK1/2 activation.


Subject(s)
Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinases/metabolism , Receptors, G-Protein-Coupled/metabolism , Animals , Base Sequence , CHO Cells , Cell Line , Cricetinae , DNA Primers , Endocytosis , Enzyme Activation , ErbB Receptors/metabolism , Humans , Ligands , Mitogen-Activated Protein Kinase 3 , Signal Transduction
10.
Arch Dermatol Res ; 294(10-11): 433-7, 2003 Jan.
Article in English | MEDLINE | ID: mdl-12563540

ABSTRACT

Sphingosylphosphorylcholine (SPC) has been reported to stimulate wound healing by its potent mitogenic effect. Fibronectin (FN) is a cell-adhesion protein that plays an important role in cell migration and collagen deposition during wound healing. In order to elucidate further the mechanism involved in the accelerated wound healing stimulated by SPC, we studied the role of SPC in FN production in cultured human dermal fibroblasts. We demonstrated that SPC dose- and time-dependently enhanced the expression of FN in human dermal fibroblast at the protein and mRNA levels. IL-6 is known to stimulate the production of FN in fibroblasts. SPC also markedly induced IL-6 production in cultured human dermal fibroblasts in a dose- and time-dependent manner. We also demonstrated that FN mRNA expression in human dermal fibroblasts was upregulated 4 h after IL-6 treatment. Moreover, pretreatment with neutralizing anti-IL-6 antibodies partially blocked the upregulation of FN mRNA expression induced by SPC in human dermal fibroblasts. These results indicate that SPC may stimulate FN synthesis through IL-6 production in cultured human dermal fibroblasts.


Subject(s)
Dermis/metabolism , Fibroblasts/metabolism , Fibronectins/metabolism , Interleukin-6/metabolism , Phosphorylcholine/analogs & derivatives , Phosphorylcholine/pharmacology , Sphingosine/analogs & derivatives , Sphingosine/pharmacology , Up-Regulation , Antibodies/pharmacology , Cells, Cultured , Dermis/cytology , Fibronectins/genetics , Humans , Interleukin-6/immunology , Protein Biosynthesis , RNA, Messenger/antagonists & inhibitors , RNA, Messenger/metabolism , Transcription, Genetic
12.
Arch Dermatol Res ; 294(9): 411-8, 2002 Dec.
Article in English | MEDLINE | ID: mdl-12522579

ABSTRACT

Suppression subtractive hybridization, a PCR-based method for cDNA subtraction, was used to identify differentially expressed genes in keratinocytes. Differentiation was induced by elevating the calcium level in the cell culture medium. Using HaCaT immortalized keratinocytes cultured in the presence of a high calcium concentration, we isolated 60 clones representing 48 different genes. By reverse Northern analysis, 13 genes were scored as overexpressed in these HaCaT cells. Northern blot analysis was used to confirm differential gene expression. Six genes, keratin 1, plasminogen activator inhibitor type 2 (PAI-2), ferritin H, peroxiredoxin 5 (PRDX5), insulin-like growth factor binding protein-3 (IGFBP-3), and one EST gene, were differentially expressed in HaCaT cells cultured in the presence of a high calcium concentration. Two of these genes, keratin 1 and PAI-2, are differentially expressed during keratinocyte terminal differentiation. IGFBP-3, which has reduced expression during epidermal differentiation, was increased after culture in a high-calcium medium for 2 or 5 days. Overexpression of the ferritin H and PRDX5 genes due to elevated calcium has not been reported in keratinocytes. We demonstrated the expression of IGFBP-3, ferritin H, PRDX5, and one gene of a matching sequence from the EST database during differentiation in primary cultured normal human keratinocytes. The EST gene expressed two transcripts of 1.8 kb and 2.5 kb in HaCaT cells, and the transcripts were confirmed to increase in keratinocytes cultured in a high-calcium medium.


Subject(s)
Calcium/administration & dosage , Gene Expression/drug effects , Keratinocytes/physiology , Cell Differentiation/physiology , Cell Line, Transformed , Culture Media/chemistry , Dose-Response Relationship, Drug , Ferritins/genetics , Humans , Insulin-Like Growth Factor Binding Protein 3/genetics , Keratinocytes/cytology , Keratins/genetics , Nucleic Acid Hybridization , Osmolar Concentration , Plasminogen Activator Inhibitor 2/genetics , Polymerase Chain Reaction
SELECTION OF CITATIONS
SEARCH DETAIL
...