Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 19 de 19
1.
Eur J Pediatr ; 183(1): 305-311, 2024 Jan.
Article En | MEDLINE | ID: mdl-37875632

Patients with familial hypokalemic periodic paralysis (HOKPP) experience episodes of reversible immobility and are at an increased risk of limited sunlight exposure, potentially leading to vitamin D deficiency. However, there is a lack of data on vitamin D levels in this population. We investigated serum vitamin D levels and their associated factors in children with HOKPP. This study included 170 genetically-confirmed children with HOKPP, aged 3-18 years, and 170 age-, sex-, and body mass index (BMI)-matched healthy controls from the Korean Channelopathy Study, a prospective controlled investigation. Anthropometric and clinical characteristics were recorded, and serum levels of calcium, ionized calcium, phosphorus, alkaline phosphatase, 25-hydroxyvitamin D, and intact parathyroid hormone (PTH) were analyzed. Vitamin D deficiency (< 20 ng/mL) was observed in 87.0% of the patients compared to 45.5% of the controls (P < 0.05) during the summer-fall season. During the winter-spring season, 91.7% of the patients and 73.4% of the controls were deficient (P < 0.05). A strong positive correlation was found between onset age of the first paralytic attack and vitamin D levels (r = 0.78, P < 0.01). Conversely, the frequency and duration of paralytic attacks were negatively correlated with vitamin D levels (r = -0.82 and r = -0.65, P < 0.01, respectively). Age, BMI, age at onset, frequency and duration of attacks, and PTH levels were independently associated with vitamin D levels (ß = -0.10, -0.12, 0.19, -0.27, -0.21, and -0.13, P < 0.05, respectively). CONCLUSIONS: Vitamin D deficiency was highly prevalent in children with HOKPP, and vitamin D levels correlated with various disease characteristics. We recommend routine screening for vitamin D levels in these patients to address this prevalent deficiency. Considering the high prevalence of vitamin D deficiency observed, further research on other diseases characterized by reversible immobility is warranted. WHAT IS KNOWN: • A correlation between immobility and low serum vitamin D levels has been established. However, the vitamin D status of patients with familial hypokalemic periodic paralysis (HOKPP) who experience periods of reversible immobility remains unknown. WHAT IS NEW: • Vitamin D deficiency was highly prevalent in children with HOKPP, and vitamin D levels correlated with various disease characteristics.


Hypokalemic Periodic Paralysis , Vitamin D Deficiency , Child , Humans , Adolescent , Calcium , Hypokalemic Periodic Paralysis/etiology , Hypokalemic Periodic Paralysis/complications , Prospective Studies , Prevalence , Vitamin D , Vitamin D Deficiency/complications , Vitamin D Deficiency/epidemiology , Risk Factors , Vitamins , Parathyroid Hormone , Seasons
2.
Int J Mol Sci ; 24(21)2023 Nov 06.
Article En | MEDLINE | ID: mdl-37958976

Skin microbiome dysbiosis has deleterious effects, and the factors influencing burn scar formation, which affects the scar microbiome composition, are unknown. Therefore, we investigated the effects of various factors influencing scar formation on the scar microbiome composition in patients with burns. We collected samples from the burn scar center and margin of 40 patients with burns, subgrouped by factors influencing scar formation. Scar microbiome composition-influencing factors were analyzed using univariate and multivariate analyses. Skin graft, hospitalization period, intensive care unit (ICU) admission, burn degree, sex, age, total body surface area burned (TBSA), time post-injury, transepidermal water loss, the erythrocyte sedimentation rate, and C-reactive protein levels were identified as factors influencing burn scar microbiome composition. Only TBSA and ICU admission were associated with significant differences in alpha diversity. Alpha diversity significantly decreased with an increase in TBSA and was significantly lower in patients admitted to the ICU than in those not admitted to the ICU. Furthermore, we identified microorganisms associated with various explanatory variables. Our cross-sectional systems biology study confirmed that various variables influence the scar microbiome composition in patients with burns, each of which is associated with various microorganisms. Therefore, these factors should be considered during the application of skin microbiota for burn scar management.


Burns , Cicatrix , Humans , Cicatrix/pathology , Cross-Sectional Studies , Retrospective Studies , Hospitalization
3.
Ann Dermatol ; 35(4): 293-302, 2023 Aug.
Article En | MEDLINE | ID: mdl-37550230

BACKGROUND: Cytoplasmic polyadenylation element binding (CPEB) proteins are sequence-specific RNA-binding proteins that control translation via cytoplasmic polyadenylation. We previously reported that CPEB1 or CPEB4 knockdown suppresses TAK1 and SMAD signaling in an in vitro study. OBJECTIVE: This study aimed to investigate whether suppression of CPEB1 or CPEB4 expression inhibits scar formation in a mice model of acute dermal wound healing. METHODS: CPEB1 and CPEB4 expression levels were suppressed by siRNA treatment. Skin wounds were created by pressure-induced ulcers in mice. Images of the wound healing were obtained using a digital camera and contraction was measured by ImageJ. mRNA and protein expression was analyzed using quantitative real time polymerase chain reaction and western blotting, respectively. RESULTS: Wound contraction was significantly decreased by pre-treatment with CPEB1 or CPEB4 siRNA compared to the control. Suppression of CPEB1 or CPEB4 expression decreased TAK1 signaling by reducing the levels of TLR4 and TNF-α, phosphorylated TAK1, p38, ERK, JNK, and NF-κB-p65. Decreased levels of phosphorylated SMAD2 and SMAD3 indicated a reduction in SMAD signaling as well. Consequently, the expression of α-SMA, fibronectin, and type I collagen decreased. CONCLUSION: CPEB1 siRNA or CPEB4 siRNA inhibit scar formation by modulating the TAK1 and SMAD signaling pathways. Our study highlights CPEB1 and CPEB4 as potential therapeutic targets for the treatment of scar formation.

4.
Wound Repair Regen ; 31(4): 547-558, 2023.
Article En | MEDLINE | ID: mdl-37129034

Sex differences are observed in various spectrums of skin diseases, and there are differences in wound healing rate. Herein, sex differences were identified for the newly healed skin microbiome of burn patients. Fifty-two skin samples (26 normal skin, 26 burn scars) were collected from 26 burn patients (12 male, 14 female) and microbiota analysis was performed. The correlation between skin microbiota and biomechanical properties of burn scars was also investigated. There were no significant differences in clinical characteristics between male and female patients. Considering the biomechanical properties of burn scars and normal skin around it performed before sample collection, the mean erythema level of men's normal skin was significantly higher than that of women, whereas the mean levels of melanin, transepidermal water loss and skin hydration showed no significant sex differences. The erythrocyte sedimentation rate was significantly higher in females than that in males. Alpha diversity showed no significant differences between normal skin and burn scars in the male group. However, the scar was significantly higher than that of normal skin in the female group. Microbial network analysis revealed that the male group had more complex microbial network than the female group. Additionally, in the male group, the edge density and clustering coefficient were higher in burn scars when compared to normal skin, than the female group. There were sex differences in the results of microbiome of normal skin and burn scars. Some of the altered microbiota have been correlated with the biomechanical properties of burn scars. In conclusion, sex difference in the burn scar microbiome was confirmed. These results suggest that burn treatment strategies should vary with sex.


Burns , Cicatrix, Hypertrophic , Microbiota , Female , Humans , Male , Cicatrix/pathology , Sex Characteristics , Wound Healing , Skin/pathology , Burns/pathology , Cicatrix, Hypertrophic/pathology
5.
Int J Mol Sci ; 24(7)2023 Mar 24.
Article En | MEDLINE | ID: mdl-37047109

Epidermal keratinocytes are highly activated, hyper-proliferated, and abnormally differentiated in the post-burn hypertrophic scar (HTS); however, the effects of scar fibroblasts (SFs) on keratinocytes through cell-cell interaction in HTS remain unknown. Here, we investigated the effects of HTSF-derived exosomes on the proliferation and differentiation of normal human keratinocytes (NHKs) compared with normal fibroblasts (NFs) and their possible mechanism to provide a reference for clinical intervention of HTS. Fibroblasts were isolated and cultured from HTS and normal skin. Both HTSF-exosomes and NF-exosomes were extracted via a column-based method from the cell culture supernatant. NHKs were treated for 24 or 48 h with 100 µg/mL of cell-derived exosomes. The expression of proliferation markers (Ki-67 and keratin 14), activation markers (keratins 6, 16, and 17), differentiation markers (keratins 1 and 10), apoptosis factors (Bax, Bcl2, caspase 14, and ASK1), proliferation/differentiation regulators (p21 and p27), and epithelial-mesenchymal transition (EMT) markers (E-cadherin, N-cadherin, and vimentin) was investigated. Compared with NF-exosomes, HTSF-exosomes altered the molecular pattern of proliferation, activation, differentiation, and apoptosis, proliferation/differentiation regulators of NHKs, and EMT markers differently. In conclusion, our findings indicate that HTSF-derived exosomes may play a role in the epidermal pathological development of HTS.


Cicatrix, Hypertrophic , Exosomes , Humans , Cicatrix, Hypertrophic/metabolism , Exosomes/metabolism , Keratinocytes/metabolism , Fibroblasts/metabolism , Keratins/metabolism , Cell Proliferation , Cells, Cultured
6.
Arch Biochem Biophys ; 722: 109215, 2022 06 15.
Article En | MEDLINE | ID: mdl-35430216

Post-burn hypertrophic scars are characterized by excessive accumulation of extracellular matrix secreted by fibroblasts. Exosomes are membrane lipid extracellular vesicles that play a pivotal role in cellular communication. Previous studies revealed the role of stem cell-derived exosomes in repairing damaged tissues, and also showed that cancer cell-derived exosomes could affect the disease pathogenesis. However, the functional properties of exosomes derived from hypertrophic scar fibroblasts (HTSFs) have not yet been studied extensively. In this study, we aimed to investigate whether HTSFs-derived exosomes can change the fibrosis-related signaling pathways in human normal fibroblasts (HNFs). HTSFs and HNFs were isolated from human hypertrophic scar tissues. HTSFs-derived exosomes were extracted and treated to HNFs. Reverse transcription-quantitative polymerase chain reaction and western blotting were used to detect mRNA and protein expression, respectively, and cell proliferation and mobility were also assessed. Exosome treatment markedly increased cell proliferation and migration, and induced small mother against decapentaplegic (SMAD) signaling by increasing the levels of phosphorylated SMAD2 and SMAD1/5/8. The levels of TAK1 signaling components were also increased after exosome treatment to HNFs, including phosphorylated TAK1, p38, ERK, and JNK. HTSFs-derived exosomes further induced the epithelial-mesenchymal transition by decreasing the expression level of E-cadherin and increasing the expression levels of N-cadherin and vimentin. Consequently, the expression levels of fibronectin, type Ⅰ collagen, and type Ⅲ collagen were increased. Our results demonstrate the fibrotic property of HTSFs-derived exosomes, which suggests a potential functional role in hypertrophic scar development and a new therapeutic target.


Cicatrix, Hypertrophic , Exosomes , Cells, Cultured , Cicatrix, Hypertrophic/metabolism , Exosomes/metabolism , Fibroblasts/metabolism , Fibrosis , Humans , Signal Transduction
7.
Sci Rep ; 11(1): 17296, 2021 08 27.
Article En | MEDLINE | ID: mdl-34453089

Hypertrophic scars represent a common complication in burn patients. In addition to cosmetic defects, they may cause serious sensory abnormalities such as pain and itching, severe dysfunction depending on the site, and emotional disorders such as anxiety and depression. The present study aimed to identify the molecular mechanisms underlying the use of extracorporeal shock wave therapy in keratinocytes. Keratinocytes derived from hypertrophic scar tissue were cultured and expression of proliferation markers (keratin 5 and 14), activation markers (keratin 6 and 17), differentiation markers (keratin 1, 10, and involucrin), apoptosis factors (Bax, Bcl2, and Caspase 14), and proliferation/differentiation regulators (p21 and p27) was investigated to compared with that of those in keratinocytes derived from normal skin tissue. Scar-derived keratinocytes were treated with extracorporeal shock waves under 1000 impulses at 0.1, 0.2, and 0.3 mJ/mm2. Shock waves altered the molecular pattern of proliferation, activation, differentiation, and apoptosis, as well as proliferation/ differentiation regulators, including Bax, Bcl2, ASK1, p21, p27, and Notch1. In summary, we show that extracorporeal shock wave therapy regulates the proliferation and differentiation of keratinocytes derived from hypertrophic scar to maintain normal epidermal integrity.


Cicatrix, Hypertrophic/therapy , Extracorporeal Shockwave Therapy/methods , Keratinocytes/cytology , Biomarkers/metabolism , Caspase 14/metabolism , Cell Differentiation , Humans , Keratin-14/metabolism , Keratin-5/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Skin , Treatment Outcome , bcl-2-Associated X Protein/metabolism
8.
Int J Mol Sci ; 22(11)2021 May 28.
Article En | MEDLINE | ID: mdl-34071277

Hypertrophic scars, the most common complication of burn injuries, are characterized by excessive deposition of fibroblast-derived extracellular matrix proteins. Calpain, a calcium-dependent protease, is involved in the fibroblast proliferation and extracellular matrix production observed in certain fibrotic diseases. However, its role in the formation of post-burn hypertrophic skin scars remains largely unknown. Here, calpain expression and activity were assessed in skin fibroblasts obtained directly from patients with third-degree burns, who consequently developed post-burn hypertrophic scars. Furthermore, the antifibrotic effect of calpastatin, an endogenous calpain inhibitor, was evaluated in human fibroblasts and a murine burn model. The activity, mRNA levels, and protein levels of calpain were markedly higher in fibroblasts from the burn wounds of patients than in normal cells. Selective calpain inhibition by calpastatin markedly reduced not only the proliferation of burn-wound fibroblasts but also the mRNA and protein expression of calpain, transforming growth factor-beta 1, α-smooth muscle actin, type I and type III collagens, fibronectin, and vimentin in burn-wound fibroblasts. The anti-scarring effects of calpastatin were validated using a murine burn model by molecular, histological, and visual analyses. This study demonstrates the pathological role of calpain and the antifibrotic effect of calpastatin via calpain inhibition in post-burn hypertrophic scar formation.


Burns/metabolism , Calcium-Binding Proteins/metabolism , Calpain/metabolism , Adult , Animals , Burns/complications , Calcium-Binding Proteins/pharmacology , Calpain/antagonists & inhibitors , Cell Proliferation , Cicatrix, Hypertrophic/metabolism , Collagen Type III , Extracellular Matrix/metabolism , Extracellular Matrix Proteins/metabolism , Female , Fibroblasts/metabolism , Fibronectins/metabolism , Humans , Hypertrophy , Male , Mice , Middle Aged , RNA, Messenger/metabolism , Skin/metabolism , Skin/pathology , Transforming Growth Factor beta1/metabolism , Young Adult
9.
Transl Res ; 236: 133-146, 2021 10.
Article En | MEDLINE | ID: mdl-33905948

Hypertrophic scars are the most common post-burn complications characterized by fibroblast proliferation and excessive extracellular matrix deposition. The intermediate-conductance Ca2+-activated K+ channel (KCa3.1) mediates fibroblast activation, resulting in several fibrotic diseases; however, this channel's role in the formation of post-burn hypertrophic skin scars remains unknown. Herein, we investigated the role of KCa3.1 and the therapeutic potential of TRAM-34, a selective inhibitor of KCa3.1, in hypertrophic skin scar formation following burn injury. Cytosolic Ca2+ levels, the expression of KCa3.1 and hypertrophic markers, and the proliferation of skin fibroblasts obtained directly from patients with third-degree burns who consequently developed post-burn hypertrophic scars were assessed. The anti-fibrotic effect of KCa3.1 inhibition by TRAM-34 was evaluated in vitro (fibroblasts) and in vivo (mouse burn models). Fibroblasts from burn wounds exhibited remarkably higher levels of cytosolic Ca2+ than normal cells. KCa3.1 expression was markedly higher in the membrane fraction but lower in the cytosolic fraction of burn wound fibroblasts than in normal cells. Selective inhibition of KCa3.1 by TRAM-34 markedly reduced not only the proliferation of burn wound fibroblasts but also the expression of hypertrophic markers in these cells. Anti-scarring molecular, histological, and visual effects of TRAM-34 were confirmed in murine burn models. Altered subcellular expression of KCa3.1 is a novel mechanism underlying the cellular response to burn injury. Our results suggest that selective inhibition of KCa3.1 by TRAM-34 has therapeutic potential against post-burn hypertrophic scar formation.


Burns/drug therapy , Burns/metabolism , Cicatrix, Hypertrophic/drug therapy , Cicatrix, Hypertrophic/etiology , Intermediate-Conductance Calcium-Activated Potassium Channels/metabolism , Pyrazoles/therapeutic use , Adolescent , Adult , Aged , Animals , Biomarkers/metabolism , Burns/genetics , Burns/pathology , Calcium/metabolism , Cell Membrane/drug effects , Cell Membrane/metabolism , Cell Proliferation/drug effects , Cicatrix, Hypertrophic/genetics , Cytosol/drug effects , Cytosol/metabolism , Female , Fibroblasts/drug effects , Fibroblasts/metabolism , Fibroblasts/pathology , Humans , Intermediate-Conductance Calcium-Activated Potassium Channels/genetics , Male , Mice , Middle Aged , Pyrazoles/pharmacology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Young Adult
10.
Int J Mol Sci ; 21(10)2020 May 23.
Article En | MEDLINE | ID: mdl-32456187

Low-temperature plasma (LTP; 3 min/day), negative pressure wound therapy (NPWT; 4 h/day), and bone marrow mesenchymal stem cells (MSCs; 1×106 cells/day) were used as mono- and combination therapy in an acute excisional skin wound-healing ICR mouse model. These therapies have been beneficial in treating wounds. We investigated the effectiveness of monotherapy with LTP, NPWT, and MSC and combination therapy with LTP + MSC, LTP + NPWT, NPWT + MSC, and LTP + NPWT + MSC on skin wounds in mice for seven consecutive days. Gene expression, protein expression, and epithelial thickness were analyzed using real time polymerase chain reaction (RT-qPCR), western blotting, and hematoxylin and eosin staining (H&E), respectively. Wound closure was also evaluated. Wound closure was significantly accelerated in monotherapy groups, whereas more accelerated in combination therapy groups. Tumor necrosis factor-α (TNF-α) expression was increased in the LTP monotherapy group but decreased in the NPWT, MSC, and combination therapy groups. Expressions of vascular endothelial growth factor (VEGF), α-smooth muscle actin (α-SMA), and type I collagen were increased in the combination therapy groups. Re-epithelialization was also considerably accelerated in combination therapy groups. Our findings suggest that combination therapy with LPT, NPWT, and MSC exert a synergistic effect on wound healing, representing a promising strategy for the treatment of acute wounds.


Mesenchymal Stem Cell Transplantation/methods , Plasma Gases/therapeutic use , Pressure , Re-Epithelialization , Skin/injuries , Actins/genetics , Actins/metabolism , Animals , Bone Marrow Cells/cytology , Cells, Cultured , Cold Temperature , Collagen Type I/genetics , Collagen Type I/metabolism , Female , Mice , Mice, Inbred ICR , Skin/metabolism , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism
11.
Arch Biochem Biophys ; 683: 108322, 2020 04 15.
Article En | MEDLINE | ID: mdl-32113875

Post-burn hypertrophic scar (HTS) is a form of excessive dermal fibrosis characterized by cutaneous scarring, which is common in patients following burn injury. Moreover, at least 50% of HTS are accompanied by inflammation. Cytoplasmic polyadenylation element binding (CPEB) proteins are key mRNA-binding proteins that control the translation of several mRNAs. However, their potential roles in treating dermal fibrosis and scarring remain unknown. Therefore, in this study, we aimed to investigate the effects of small interfering RNA (siRNA)-mediated knockdown of CPEB1 or CPEB4 in human THP-1 macrophages and dermal fibroblasts treated with LPS and TGF-ß1. We found significantly increased CPEB1 and CPEB4 mRNA and protein levels in LPS-treated THP-1 cells and TGF-ß1-treated fibroblasts. CPEB1 and CPEB4 knockdowns suppressed LPS-activated TAK1 signaling cascades by reducing the levels of TNF-α and phosphorylated TAK1, p38, ERK, JNK, and NF-κB-p65 in THP-1 cells. CPEB1 and CPEB4 knockdowns also attenuated TGF-ß1-activated Smad-dependent and -independent signaling cascades by reducing the levels of TAK1, p38, ERK, JNK, and phosphorylated Smad 2 and Smad 1/5/8 in fibroblasts. Furthermore, CPEB1 or CPEB4 knockdown markedly decreased the levels of fibrosis markers, including α-SMA, type I collagen, and fibronectin in fibroblasts. Our findings indicate that CPEB1 and CPEB4 are involved in the regulation of the TAK1 and Smad signalings in human macrophages and dermal fibroblasts. These activities may play a role in cutaneous scarring responses.


MAP Kinase Kinase Kinases/metabolism , RNA-Binding Proteins/genetics , Signal Transduction , Smad Proteins/metabolism , Transcription Factors/genetics , mRNA Cleavage and Polyadenylation Factors/genetics , Animals , Dermis/cytology , Fibroblasts/cytology , Gene Expression Regulation , Humans , Inflammation , Macrophages/cytology , Mice , Phosphorylation , RAW 264.7 Cells , RNA, Small Interfering/metabolism , THP-1 Cells , Transforming Growth Factor beta1/pharmacology , Up-Regulation
12.
Tissue Eng Regen Med ; 16(6): 585-593, 2019 12.
Article En | MEDLINE | ID: mdl-31824821

BACKGROUND: Low temperature plasma (LTP) was recently shown to be potentially useful for biomedical applications such as bleeding cessation, cancer treatment, and wound healing, among others. Keratinocytes are a major cell type that migrates directionally into the wound bed, and their proliferation leads to complete wound closure during the cutaneous repair/regeneration process. However, the beneficial effects of LTP on human keratinocytes have not been well studied. Therefore, we investigated migration, growth factor production, and cytokine secretion in primary human keratinocytes after LTP treatment. METHODS: Primary cultured keratinocytes were obtained from human skin biopsies. Cell viability was measured with the EZ-Cytox cell viability assay, cell migration was evaluated by an in vitro wound healing assay, gene expression was analyzed by quantitative real-time polymerase chain reaction, and protein expression was measured by enzyme-linked immunosorbent assays and western blotting after LTP treatment. RESULTS: Cell migration, the secretion of several cytokines, and gene and protein levels of angiogenic growth factors increased in LTP-treated human keratinocytes without associated cell toxicity. LTP treatment also significantly induced the expression of hypoxia inducible factor-1α (HIF-1α), an upstream regulator of angiogenesis. Further, the inhibition of HIF-1α expression blocked the production of angiogenic growth factors induced by LTP in human keratinocytes. CONCLUSION: Our results suggest that LTP treatment is an effective approach to modulate wound healing-related molecules in epidermal keratinocytes and might promote angiogenesis, leading to improved wound healing.


Cell Movement/drug effects , Plasma Gases/pharmacology , Wound Healing/drug effects , Angiotensin I/genetics , Angiotensin I/metabolism , Cell Survival/drug effects , Cells, Cultured , Cytokines/metabolism , Fibroblast Growth Factor 2/genetics , Fibroblast Growth Factor 2/metabolism , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Keratinocytes/cytology , Keratinocytes/metabolism , Plasma Gases/chemistry , Temperature , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism
13.
Int J Mol Sci ; 19(1)2018 Jan 02.
Article En | MEDLINE | ID: mdl-29301325

Extracorporeal shock wave therapy (ESWT) considerably improves the appearance and symptoms of post-burn hypertrophic scars (HTS). However, the mechanism underlying the observed beneficial effects is not well understood. The objective of this study was to elucidate the mechanism underlying changes in cellular and molecular biology that is induced by ESWT of fibroblasts derived from scar tissue (HTSFs). We cultured primary dermal fibroblasts derived from human HTS and exposed these cells to 1000 impulses of 0.03, 0.1, and 0.3 mJ/mm². At 24 h and 72 h after treatment, real-time PCR and western blotting were used to detect mRNA and protein expression, respectively, and cell viability and mobility were assessed. While HTSF viability was not affected, migration was decreased by ESWT. Transforming growth factor beta 1 (TGF-ß1) expression was reduced and alpha smooth muscle actin (α-SMA), collagen-I, fibronectin, and twist-1 were reduced significantly after ESWT. Expression of E-cadherin was increased, while that of N-cadherin was reduced. Expression of inhibitor of DNA binding 1 and 2 was increased. In conclusion, suppressed epithelial-mesenchymal transition might be responsible for the anti-scarring effect of ESWT, and has potential as a therapeutic target in the management of post-burn scars.


Cicatrix, Hypertrophic/genetics , Cicatrix, Hypertrophic/pathology , Extracorporeal Shockwave Therapy , Fibroblasts/metabolism , Fibroblasts/pathology , Gene Expression Regulation , Actins/genetics , Actins/metabolism , Cadherins/metabolism , Cell Movement/genetics , Cell Survival/genetics , Extracellular Matrix Proteins/metabolism , Fibrosis , Humans , Transcription Factors/metabolism , Transforming Growth Factor beta1/genetics , Transforming Growth Factor beta1/metabolism , Vimentin/genetics , Vimentin/metabolism
14.
Arch Biochem Biophys ; 630: 9-17, 2017 09 15.
Article En | MEDLINE | ID: mdl-28750820

Numerous studies on the application of low temperature plasma (LTP) have produced impressive results, including antimicrobial, antitumor, and wound healing effects. Although LTP research has branched out to include medical applications, the detailed effects and working mechanisms of LTP on wound healing have not been fully investigated. Here, we investigated the potential effect of inducing growth factor after exposure to LTP and demonstrated the increased expression of angiogenic growth factor mediated by LTP-induced HIF1α expression in primary cultured human dermal fibroblasts. In cell viability assays, fibroblast viability was reduced 6 h and 24 h after LTP treatment for only 5 min, and pre-treating with NAC, a ROS scavenger, prevented cell loss. Fibroblast migration significantly increased at 6 h and 24 h in scratch wound healing assays, the expression of cytokines significantly changed, and regulatory growth factors were induced at 6 h and 24 h after exposure to LTP in RT-PCR or ELISAs. Specifically, LTP treatment significantly induced the expression of HIF1α, an upstream regulator of angiogenesis. Pre-treatment with the inhibitor CAY10585 abolished HIF1α expression and prevented LTP-induced angiogenic growth factor production according to immunoblotting, immunocytochemistry, and ELISA results. Taken together, our results provide information on the molecular mechanism by which LTP application may promote angiogenesis and will aid in developing methods to improve wound healing.


Angiogenesis Inducing Agents/metabolism , Dermis/metabolism , Fibroblasts/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/biosynthesis , Neovascularization, Physiologic/drug effects , Plasma Gases/pharmacology , Up-Regulation/drug effects , Dermis/cytology , Fibroblasts/cytology , Humans
15.
Ann Dermatol ; 26(5): 570-5, 2014 Oct.
Article En | MEDLINE | ID: mdl-25324648

BACKGROUND: Atopic dermatitis (AD) is characterized by itching and eczema-like skin lesions, and its symptoms alleviate with age. Recently, the prevalence of AD has increased among adolescents and adults. The increasing prevalence of AD seems to be related to westernized lifestyles and dietary patterns. OBJECTIVE: The aim of this study was to evaluate the dietary patterns and nutrient intake of patients with AD. METHODS: The study population consisted of 50 children with AD who visited the Department of Dermatology at Kangnam Sacred Heart Hospital, Seoul, Korea from May 2008 to May 2009. Physical condition and calorie intake were evaluated using the Eczema Area and Severity Index score and Food Record Questionnaire completed by the subjects, and the data were analyzed using the Nutritional Assessment Program Can-pro 3.0 (The Korean Nutrition Society, 2005) program to determine the gap between the actual ingestion and average requirements of 3 major nutrients (i.e. carbohydrates, proteins, and lipids), vitamins (i.e. A, B, C, and E), niacin, folic acid, calcium, iron, phosphorus, and zinc in all subjects. RESULTS: The intake rate of proteins was 18.02% (recommended dietary allowance [RDA], 7%~20%), of carbohydrates was 67.7% (RDA, 55%~70%), and of lipids was 14.24% (RDA, 15%~30%). Thirty-one subjects (62%) showed deficiency of folic acid, and 21 subjects (42%), of iron supplements. CONCLUSION: Essential nutrient intake tends to be lower in AD patients than in healthy subjects, and this low intake is closely related to the severity of AD.

16.
Neurochem Res ; 38(11): 2276-86, 2013 Nov.
Article En | MEDLINE | ID: mdl-23990224

Although propofol has been reported to offer neuroprotection against cerebral ischemia injury, its impact on cerebral edema following ischemia is not clear. The objective of this investigation is to evaluate the effects of propofol post-treatment on blood-brain barrier (BBB) integrity and cerebral edema after transient cerebral ischemia and its mechanism of action, focusing on modulation of aquaporins (AQPs), matrix metalloproteinases (MMPs), and hypoxia inducible factor (HIF)-1α. Cerebral ischemia was induced in male Sprague-Dawley rats (n = 78) by occlusion of the right middle cerebral artery for 1 h. For post-treatment with propofol, 1 mg kg(-1) min(-1) of propofol was administered for 1 h from the start of reperfusion. Nineteen rats undergoing sham surgery were also included in the investigation. Edema and BBB integrity were assessed by quantification of cerebral water content and extravasation of Evans blue, respectively, following 24 h of reperfusion. In addition, the expression of AQP-1, AQP-4, MMP-2, and MMP-9 was determined 24 h after reperfusion and the expression of HIF-1α was determined 8 h after reperfusion. Propofol post-treatment significantly reduced cerebral edema (P < 0.05) and BBB disruption (P < 0.05) compared with the saline-treated control. The expression of AQP-1, AQP-4, MMP-2, and MMP-9 at 24 h and of HIF-1α at 8 h following ischemia/reperfusion was significantly suppressed in the propofol post-treatment group (P < 0.05). Propofol post-treatment attenuated cerebral edema after transient cerebral ischemia, in association with reduced expression of AQP-1, AQP-4, MMP-2, and MMP-9. The decreased expression of AQPs and MMPs after propofol post-treatment might result from suppression of HIF-1α expression.


Blood-Brain Barrier/drug effects , Brain Edema/prevention & control , Ischemic Attack, Transient/physiopathology , Propofol/pharmacology , Animals , Aquaporin 1/biosynthesis , Aquaporin 4/biosynthesis , Hypoxia-Inducible Factor 1, alpha Subunit/biosynthesis , Infarction, Middle Cerebral Artery/physiopathology , Male , Matrix Metalloproteinase 2/biosynthesis , Matrix Metalloproteinase 9/biosynthesis , Rats , Rats, Sprague-Dawley
17.
J Pharm Pharmacol ; 59(4): 521-8, 2007 Apr.
Article En | MEDLINE | ID: mdl-17430635

The neuroprotective effect of 9-hydroxypinoresinol was examined in mice challenged with kainic acid (KA), a potent central nervous system excitotoxin. For this purpose, mice were administered intraperitoneally with 9-hydroxypinoresinol before KA injection. A remarkable neuroprotective effect was observed with a single dose of 9-hydroxypinoresinol (30 mg kg(-1)) 24 h before KA challenge. Furthermore, 9-hydroxypinoresinol (20 mg kg(-1)) administered for 3 days before KA challenge reduced the mortality (60%) induced by KA to zero, and alleviated behavioural signs of KA neurotoxicity. Additionally, pretreatment with 9-hydroxypinoresinol (20 mg kg(-1)) prevented the decrease in the levels of total glutathione (GSH) and thiobarbituric acid reactive substances (P < 0.05). GSH peroxidase activity in brain tissue was restored to control levels, although GSH reductase activity and GSH S-transferase activity were not affected. Such a protective action was also observed even with a lower dose (10 mg kg(-1)) of 9-hydroxypinoresinol administered for 3 days, albeit to a lesser extent. From the results, it is proposed that 9-hydroxypinoresinol exerts a potent neuroprotective effect mainly by preventing oxidative stress in brain tissue of mice challenged with KA.


Antioxidants/pharmacology , Behavior, Animal/drug effects , Brain/drug effects , Kainic Acid/toxicity , Lignans/pharmacology , Neurotoxins/toxicity , Oxidative Stress/drug effects , Animals , Antioxidants/administration & dosage , Body Weight/drug effects , Brain/pathology , Glutathione/metabolism , Glutathione Peroxidase/metabolism , Glutathione Reductase/metabolism , Glutathione Transferase/metabolism , Lignans/administration & dosage , Lipid Peroxidation/drug effects , Male , Mice , Mice, Inbred ICR , Organ Size , Thiobarbituric Acid Reactive Substances/metabolism
18.
J Agric Food Chem ; 53(22): 8526-32, 2005 Nov 02.
Article En | MEDLINE | ID: mdl-16248548

The neuroprotective effect of petaslignolide A (PA), a furfuran lignan isolated from butanol fraction of Petasites japonicus (Sieb. et Zucc.) Maxim. (Compositae) leaves, on the oxidative damage in the brain of mice challenged with kainic acid was examined using behavioral signs and biochemical parameters of oxidative stress. PA (40 mg/kg) was administered to ICR male mice through a gavage for 4 days consecutively, and on the final day, kainic acid (50 mg/kg) was administered intraperitoneally. During the 4-day treatment with PA, the body weight gain was not significantly different from that of vehicle-treated control animals. PA (40 mg/kg) alleviated the behavioral signs of kainic acid neurotoxicity and reduced the mortality (50%) by kainic acid to 12.5%. Moreover, the administration of PA restored the levels of glutathione and thiobarbituric acid-reactive substances as well as GSH-peroxidase activity in the brains of mice administered kainic acid to control levels (P < 0.05). In comparison, PA (40 mg/kg) was approximately comparable to the butanol fraction (200 mg/kg) of P. japonicus extract in reducing kainic acid neurotoxicity. On the basis of these results, PA is suggested to be a major neuroprotective agent primarily responsible for the protective action of the butanol fraction of P. japonicus extract against kainic acid-induced neurotoxicity in the brains of mice.


Brain Diseases/prevention & control , Furans/administration & dosage , Kainic Acid , Lignans/administration & dosage , Neuroprotective Agents/administration & dosage , Oxidative Stress/drug effects , Petasites/chemistry , Animals , Blood-Brain Barrier , Brain Diseases/chemically induced , Butanols , Enzyme Induction/drug effects , Glutathione/metabolism , Male , Mice , Mice, Inbred ICR , Plant Extracts/chemistry , Plant Leaves/chemistry
19.
Arch Pharm Res ; 28(9): 1023-6, 2005 Sep.
Article En | MEDLINE | ID: mdl-16212231

A new furofuran lignan (1) was isolated from the n-butanol fraction of the methanolic extract of the leaves of Petasites japonicus (Sieb. et Zucc.) Maxim. (Compositae). The structure of compound 1 was determined to be 2alpha-(4'-hydroxy-3'-methoxyphenyl)-6alpha-(4"-hydroxy-3"-methoxyphenyl)-8alpha-hydroxy-3,7-dioxabicyclo[3.3.0octane 4'-O-(beta-D-glucopyranoside) by spectroscopic methods including 2D-NMR. In further studies, it was found that the compound 1 expressed an antioxidant activity in DPPH radical scavenging assay, and moreover, ameliorated the seizure in kainic acid-treated mice.


Anticonvulsants/isolation & purification , Antioxidants/isolation & purification , Lignans/isolation & purification , Petasites/chemistry , Plant Leaves/chemistry , Animals , Anticonvulsants/chemistry , Anticonvulsants/pharmacology , Antioxidants/chemistry , Antioxidants/pharmacology , Lignans/chemistry , Lignans/pharmacology , Mice
...