Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
1.
J Invest Dermatol ; 143(12): 2356-2365.e5, 2023 12.
Article in English | MEDLINE | ID: mdl-37263487

ABSTRACT

TRPV4 is a calcium ion channel that is widely expressed in various cells. It is also involved in physiological and pathological processes. However, the role of TRPV4 in psoriasis remains unknown. We aimed to investigate the role of TRPV4 in psoriasis using human psoriasis skin samples and an imiquimod-induced psoriasis-like mouse model. Keratinocytes in human psoriasis skin had high TRPV4 expression. Trpv4-knockout mice had less severe dermatitis than wild-type mice in the imiquimod-induced mouse model. Knockout mice had significantly reduced epidermal thickness and a low number of infiltrated CD3+ T cells and CD68+ macrophages on the basis of histopathological studies and decreased mRNA expression of Il17a, Il17f, and Il23, as detected through qPCR. Furthermore, knockout mice had a significantly low expression of neuropeptides and the neuron marker PGP9.5. Adenosine triphosphate release was significantly suppressed by TRPV4 knockdown in both human and mouse keratinocytes in vitro. Finally, treatment with TRPV4 antagonist was significantly effective in preventing the progression of psoriasis-like dermatitis. In conclusion, TRPV4 mediates the expression of keratinocyte-derived adenosine triphosphate and increases the secretion of neuropeptides, resulting in the activation and amplification of IL-23/Th17 responses. Hence, TRPV4 can serve as a novel therapeutic target in psoriasis.


Subject(s)
Dermatitis , Neuropeptides , Psoriasis , Humans , Animals , Mice , Imiquimod/pharmacology , TRPV Cation Channels/genetics , TRPV Cation Channels/metabolism , Adenosine Triphosphate/metabolism , Mice, Knockout , Keratinocytes/metabolism , Psoriasis/chemically induced , Psoriasis/genetics , Psoriasis/drug therapy , Skin/metabolism , Dermatitis/pathology , Neuropeptides/metabolism , Disease Models, Animal , Mice, Inbred BALB C
5.
World Allergy Organ J ; 14(2): 100511, 2021 Feb.
Article in English | MEDLINE | ID: mdl-33643518

ABSTRACT

BACKGROUND: Hereditary angioedema (HAE) is a rare but life-threatening condition. HAE types I and II (HAE-1/2) result from C1-inhibitor (C1-INH) deficiency. However, recent genetic analysis has established a new type of HAE with normal C1-INH (HAEnC1-INH). The mutations of factor XII, plasminogen, angiopoietin 1, and kininogen 1 genes may be the cause of HAEnC1-INH. Nevertheless, other causative molecules (HAE-unknown) may be involved. The Japanese therapeutic environment for HAE has been improving owing to the self-subcutaneous injection of icatibant, which was approved for the treatment of acute attack and enables early therapy. Erythema marginatum (EM) is a visible prodromal symptom which occasionally occurs prior to an angioedema attack; hence, recognizing the risk of an acute attack is important for early treatment. However, the detailed characteristics of EM remain unclear. In this study, we first investigated the clinical manifestations of EM in Japanese patients with HAE. METHODS: A 20-point survey was developed and distributed to 40 physicians to gather clinical data on EM from patients with HAE. RESULTS: Data on 68 patients with HAE (58 patients with HAE-1/2 and 10 patients with HAE-unknown) were collected. Of the patients with HAE-1/2, 53.4% experienced EM, whereas 43.1% did not. The forearm was the most frequent area of EM (64.5%), followed by the abdomen (29.0%) and upper arm and precordium (19.3%). Of the HAE-1/2 patients with EM, 41.9% always had angioedema following EM, while 29.0% always had colocalization of EM with angioedema. Moreover, 3.2% showed a correlation between the awareness of EM and severity of an angioedema attack. In 60.9% of HAE-1/2 patients with EM, the interval between the awareness of EM and appearance of angioedema was <3 h. Of the patients with HAE-unknown, 30.0% also experienced EM. CONCLUSION: We confirmed that more than one-half of Japanese patients with HAE-1/2 and one-third of those with HAE-unknown develop EM as the prodromal symptom of an angioedema attack. Physicians should communicate the significance of EM to patients with HAE to prepare them for possible imminent attacks.

6.
J Dermatol Sci ; 101(1): 58-68, 2021 Jan.
Article in English | MEDLINE | ID: mdl-33176965

ABSTRACT

BACKGROUND: Psoriasis is a multifactorial disease arises from a complex interaction of genetics, immune system, and environmental aspects. IL-23/Th17 immune axis has been considered as a primary modulator in psoriasis. In addition, several findings imply that nervous system may take a part in the pathogenesis of psoriasis, suggesting that nervous system, through its neuropeptide, may interact with immune system and lead to the formation of psoriasis. OBJECTIVE: We aimed to ascertain the role of neuropeptides secreted from neurons in the pathogenesis of psoriasis in vivo. METHODS: The release of neuropeptide was inhibited by injecting Botulinum toxin B (BTX-B) on Imiquimod (IMQ)-induced psoriasis-like dermatitis mice model. Quantification of skin dermatitis, infiltrating inflammatory cells, and the production of cytokines at the lesional skin area were performed by PSI score, immunostaining, and real-time PCR. We also tested the effect of selective CGRP antagonist (CGRP8-37) on psoriasis-like dermatitis in IMQ-treated mice. RESULTS: BTX-B injection significantly suppressed PSI score and reduced the number of CD4+ T cells, CD11c+ dendritic cells, and the production of IL-17A/F in the lesional skin. The expressions of PGP9.5+ nerve fibers and neuropeptides (SP, CGRP) were also significantly reduced following BTX-B injection. Additionally, CGRP antagonist also suppressed the development of IMQ-induced psoriasis-like dermatitis in mice. CONCLUSION: The suppression of neuropeptide secretion in the skin by BTX injection might inhibit nerve elongation, the infiltration of immune cells, as well as IL-17 production, resulting in the improvement of psoriasis. Neuropeptide inhibitor could also be applied to the treatment of psoriasis.


Subject(s)
Botulinum Toxins, Type A/administration & dosage , Calcitonin Gene-Related Peptide/metabolism , Neuroimmunomodulation/drug effects , Psoriasis/drug therapy , Substance P/metabolism , Animals , Calcitonin Gene-Related Peptide/immunology , Disease Models, Animal , Female , Humans , Imiquimod/administration & dosage , Imiquimod/immunology , Injections, Subcutaneous , Mice , Mice, Inbred BALB C , Psoriasis/chemically induced , Psoriasis/diagnosis , Psoriasis/immunology , Severity of Illness Index , Skin/drug effects , Skin/immunology , Skin/innervation , Skin/pathology , Substance P/immunology
7.
Wound Repair Regen ; 28(5): 600-608, 2020 09.
Article in English | MEDLINE | ID: mdl-32356363

ABSTRACT

Ischemia-reperfusion (I/R) is associated with various pathogenic conditions, and there has been increasing evidence that cutaneous I/R injury is associated with the pathogenesis of pressure ulcers (PUs), especially at the early stage presenting as non-blanchable erythema. Several studies demonstrated that oxidative stress is a key player in I/R injury, and the inhibition of oxidative stress may be capable of protecting tissue damage after I/R injury in various organs including skin. Dimethyl fumarate (DMF) approved by the Food and Drug Administration is Nrf2 activator, and recent studies revealed the antioxidative and anti-inflammatory effects of DMF on I/R injury in animal models. Our objective was to assess the effects of oral administration of DMF on the development of PUs after cutaneous I/R injury in mice. We found that DMF administration significantly decreased the size of PUs after cutaneous I/R. Cutaneous I/R-induced oxidative stress was also significantly inhibited by DMF in OKD48 mice, in which oxidative stress can be visually assessed. In addition, DMF treatment decreased hypoxic area, the numbers of apoptotic cells, and vascular loss in I/R area. DMF treatment suppressed the infiltration of MPO+ neutrophils and the production of proinflammatory cytokines in I/R site after cutaneous I/R injury. in vitro experiments, DMF treatment suppressed the production of reactive oxygen species in pericyte-like cells. These results suggest that DMF treatment might prevent the formation of PUs induced by cutaneous I/R injury via suppressing oxidative stress and subsequent inflammation. DMF treatment during the early phase of decubitus ulcers might protect against further progression.


Subject(s)
Dimethyl Fumarate/pharmacology , Pressure Ulcer/etiology , Pressure Ulcer/prevention & control , Reperfusion Injury/complications , Administration, Oral , Animals , Dimethyl Fumarate/administration & dosage , Disease Models, Animal , Mice , Mice, Inbred C57BL , Oxidative Stress
8.
Sci Rep ; 10(1): 1349, 2020 Jan 28.
Article in English | MEDLINE | ID: mdl-31992828

ABSTRACT

Several studies have demonstrated potential roles for apelin/APJ signaling in the regulation of oxidative stress associated with ischemia-reperfusion (I/R) injury in several organs. Objective was to assess the role of apelin/APJ signaling in the development of pressure ulcers (PUs) formation after cutaneous I/R injury in mice. We identified that cutaneous I/R injury increased the expression of apelin in the skin at I/R site. Administration of apelin significantly inhibited the formation of PUs. The reductions of blood vessels, hypoxic area and apoptosis in I/R site were inhibited by apelin injection. Oxidative stress signals in OKD48 mice and the expressions of oxidative stress related genes in the skin were suppressed by apelin injection. H2O2-induced intracellular ROS and apoptosis in endothelial cells and fibroblasts were suppressed by apelin in vitro. Furthermore, MM07, biased agonist of APJ, also significantly suppressed the development of PUs after cutaneous I/R, and the inhibitory effect of MM07 on PUs formation was higher than that in apelin. We conclude that apelin/APJ signaling may inhibit cutaneous I/R injury-induced PUs formation by protecting the reduction of vascularity and tissue damage via suppression of oxidative stress. Exogenous application of apelin or MM07 might have therapeutic potentials against the development of PUs.


Subject(s)
Apelin Receptors/metabolism , Apelin/metabolism , Reperfusion Injury/metabolism , Signal Transduction , Skin/blood supply , Skin/metabolism , Ulcer/etiology , Ulcer/metabolism , Animals , Apelin/genetics , Apelin Receptors/genetics , Apoptosis/genetics , Cytokines/genetics , Cytokines/metabolism , Disease Models, Animal , Fluorescent Antibody Technique , Gene Expression , Mice , Oxidative Stress , Reactive Oxygen Species/metabolism , Reperfusion Injury/etiology , Reperfusion Injury/pathology , Ulcer/pathology
9.
J Dermatol Sci ; 96(1): 18-25, 2019 Oct.
Article in English | MEDLINE | ID: mdl-31447183

ABSTRACT

BACKGROUND: Previous studies have indicated that MFG-E8 enhances tumor cell survival, invasion and angiogenesis. However, the role of MFG-E8 in angiosarcoma (AS) has not been clarified. OBJECTIVE: Objective was to elucidate the mechanism of the regulation by MFG-E8 in AS and the association between MFG-E8 and clinicopathological features of AS. METHODS: The effects of the depletion of MFG-E8 by siRNA on tube formation, migration and proliferation in murine AS cells were examined. The effect of administration of anti-MFG-E8 antibody (Ab) on tumor growth of AS in mice was examined. The associations of MFG-E8 expression and clinicopathological features of human AS were assessed. RESULTS: The expressions of MFG-E8 in murine and human AS cells were significantly higher than those in melanoma cells, macrophages and endothelial cells. Depletion of MFG-E8 in murine AS cells by siRNA significantly inhibited the formation of capillary-like structures and migration, but not proliferation. Administration of anti-MFG-E8 Ab significantly inhibited tumor growth and decreased the number of tumor-associated macrophages (TAMs) in AS tumors. Tumor size and the number of TAMs in human AS with high expression of MFG-E8 were significantly increased compared to those of AS with low expression of MFG-E8. Progression-free survival and overall survival time of the patients of AS with high expression of MFG-E8 were significantly shorter than those of AS with low expression of MFG-E8. CONCLUSIONS: AS-derived MFG-E8 might enhance tumor growth via angiogenesis and the induction of TAMs in autocrine/paracrine manner, and administration of anti-MFG-E8 Ab could be a therapeutic potential for AS.


Subject(s)
Antigens, Surface/metabolism , Hemangiosarcoma/pathology , Milk Proteins/metabolism , Neovascularization, Pathologic/pathology , Skin Neoplasms/pathology , Aged , Animals , Antibodies/administration & dosage , Antigens, Surface/genetics , Biopsy , Cell Line, Tumor/transplantation , Disease Models, Animal , Female , Gene Knockdown Techniques , Hemangiosarcoma/blood supply , Hemangiosarcoma/drug therapy , Human Umbilical Vein Endothelial Cells , Humans , Male , Mice , Milk Proteins/antagonists & inhibitors , Milk Proteins/genetics , Neovascularization, Pathologic/drug therapy , Pericytes , RAW 264.7 Cells , RNA, Small Interfering/metabolism , Skin/blood supply , Skin/pathology , Skin Neoplasms/blood supply , Skin Neoplasms/drug therapy
10.
J Dermatol Sci ; 96(1): 8-17, 2019 Oct.
Article in English | MEDLINE | ID: mdl-31447184

ABSTRACT

BACKGROUND: There is growing evidence that vasculopathy-induced hypoxia and oxidative stress enhance the process of fibrosis in systemic sclerosis (SSc). Kaempferol is a natural flavonoid widely found in various vegetables and fruits, and has been reported to have excellent antioxidant activity. OBJECTIVE: Objective was to elucidate the effect of kaempferol on skin fibrosis and the mechanism of the inhibitory regulation of fibrosis by kaempferol. METHODS: We assessed the effect of intraperitoneally administered kaempferol on bleomycin-induced dermal fibrosis in mice. The effect of kaempferol on oxidative stress in bleomycin-treated mice and SSc fibroblasts was assessed in vivo and in vitro. RESULTS: We identified that kaempferol injection significantly inhibited bleomycin-induced dermal fibrosis in mice. The number of αSMA+ myofibroblasts, CD3+ T-cells, and CD68+ macrophages in lesional skin was significantly decreased by kaempferol injections. Kaempferol administration also significantly suppressed the bleomycin-induced oxidative stress signal in OKD48 mice. Additionally, mRNA levels of oxidative stress-associated factors, such as HO-1 and NOX2, as well as inflammatory and pro-fibrotic cytokines, including IL-6, TGF-ß and TNFα in sclerotic skin were significantly decreased by kaempferol. Kaempferol also reduced bleomycin-induced TUNEL+ apoptotic cells in the lesional skin of bleomycin-treated mice. Furthermore, the oxidant-induced intracellular accumulation of reactive oxygen species (ROS) in SSc fibroblasts was inhibited by kaempferol treatment. In addition, the oxidant-induced apoptosis of SSc fibroblasts was decreased by kaempferol in vitro. CONCLUSION: Kaempferol might improve bleomycin-induced fibrosis by reducing oxidative stress, inflammation, and oxidative cellular damage. Administration of kaempferol might be an alternative treatment for skin fibrosis in SSc.


Subject(s)
Antioxidants/administration & dosage , Fibroblasts/drug effects , Kaempferols/administration & dosage , Scleroderma, Systemic/drug therapy , Skin/pathology , Animals , Apoptosis/drug effects , Biopsy , Bleomycin/toxicity , Disease Models, Animal , Female , Fibroblasts/pathology , Fibrosis , Humans , Injections, Intraperitoneal , Mice , Oxidative Stress/drug effects , Primary Cell Culture , Reactive Oxygen Species/metabolism , Scleroderma, Systemic/pathology , Skin/drug effects
12.
J Dermatol ; 46(3): 213-218, 2019 Mar.
Article in English | MEDLINE | ID: mdl-30614031

ABSTRACT

18 F-Fluorodeoxyglucose (FDG) positron emission tomography (PET)/computed tomography (CT) is usually used to screen malignancy in patients with dermatomyositis (DM). Additionally, it is well known that FDG-PET/CT provides valuable information for evaluating the activity of several inflammatory diseases, such as sarcoidosis, atherosclerosis, inflammatory bowel disease and rheumatoid arthritis. Therefore, the objective of this study was to evaluate the clinical usefulness of FDG-PET/CT for the detection of inflammatory lesions and disease activity of both myopathy and interstitial lung disease (ILD) in DM patients. We measured the maximum standardized uptake value (SUVmax) in the muscles and lungs in 22 DM patients, and compared with magnetic resonance imaging (MRI) and high-resolution computed tomography (HRCT) findings in the same muscle and lung regions as well as with clinical findings. We found that the location of increased FDG uptake was nearly consistent with the region of ILD and myositis detected by HRCT or MRI, respectively. There was a significant positive correlation between lung HRCT score and SUVmax in each lung. Serum Krebs von den Lungen-6 levels also revealed significant positive correlation with total SUVmax of right and left lungs. Regarding FDG-PET/CT and myopathy, total SUVmax in the muscles was significantly correlated with serum cytokeratin levels. Our results suggest that FDG uptake (SUVmax) might be useful for not only the detection of malignant tumors, but also the evaluation of the location and activity of ILD and myositis in DM patients.


Subject(s)
Dermatomyositis/complications , Fluorodeoxyglucose F18/administration & dosage , Lung Diseases, Interstitial/diagnostic imaging , Positron Emission Tomography Computed Tomography/methods , Radiopharmaceuticals/administration & dosage , Adult , Aged , Dermatomyositis/diagnostic imaging , Female , Humans , Lung , Lung Diseases, Interstitial/blood , Lung Diseases, Interstitial/etiology , Magnetic Resonance Imaging , Male , Middle Aged , Mucin-1/blood , Muscle, Skeletal/diagnostic imaging , Retrospective Studies
13.
Arthritis Rheumatol ; 71(2): 302-314, 2019 02.
Article in English | MEDLINE | ID: mdl-30175895

ABSTRACT

OBJECTIVE: Several studies have demonstrated that the secreted glycoprotein and integrin ligand milk fat globule-associated protein with epidermal growth factor- and factor VIII-like domains (MFG-E8) negatively regulates fibrosis in the liver, lungs, and respiratory tract. However, the mechanisms and roles of MFG-E8 in skin fibrosis in systemic sclerosis (SSc) have not been characterized. We undertook this study to elucidate the role of MFG-E8 in skin fibrosis in SSc. METHODS: We assessed expression of MFG-E8 in the skin and serum in SSc patients. We examined the effect of recombinant MFG-E8 (rMFG-E8) on latent transforming growth factor ß (TGFß)-induced gene/protein expression in SSc fibroblasts. We examined the effects of deficiency or administration of MFG-E8 on fibrosis mouse models. RESULTS: We demonstrated that MFG-E8 expression around dermal blood vessels and the serum MFG-E8 level in SSc patients (n = 7 and n = 44, respectively) were lower than those in healthy individuals (n = 6 and n = 28, respectively). Treatment with rMFG-E8 significantly inhibited latent TGFß-induced expression of type I collagen, α-smooth muscle actin, and CCN2 in SSc fibroblasts (n = 3-8), which suggested that MFG-E8 inhibited activation of latent TGFß as well as TGFß signaling via binding to αv integrin. In a mouse model of bleomycin-induced fibrosis (n = 5-8) and in a TSK mouse model (a genetic model of SSc) (n = 5-10), deficient expression of MFG-E8 significantly enhanced both pulmonary and skin fibrosis, and administration of rMFG-E8 significantly inhibited bleomycin-induced dermal fibrosis. CONCLUSION: These results suggest that vasculopathy-induced dysfunction of pericytes and endothelial cells, the main cells secreting MFG-E8, may be associated with the decreased expression of MFG-E8 in SSc and that the deficient inhibitory regulation of latent TGFß-induced skin fibrosis by MFG-E8 may be involved in the pathogenesis of SSc and may be a therapeutic target for fibrosis in SSc patients.


Subject(s)
Antigens, Surface/metabolism , Fibroblasts/metabolism , Integrin alphaV/metabolism , Milk Proteins/metabolism , Scleroderma, Systemic/metabolism , Skin/metabolism , Transforming Growth Factor beta/metabolism , Animals , Antibiotics, Antineoplastic/toxicity , Antigens, Surface/genetics , Antigens, Surface/pharmacology , Bleomycin/toxicity , Collagen Type I/drug effects , Collagen Type I/metabolism , Female , Fibroblasts/drug effects , Fibrosis , Humans , Lung/drug effects , Lung/pathology , Male , Mice , Mice, Knockout , Middle Aged , Milk Proteins/genetics , Milk Proteins/pharmacology , Scleroderma, Systemic/chemically induced , Scleroderma, Systemic/pathology , Skin/drug effects , Skin/pathology
15.
J Invest Dermatol ; 139(4): 890-899, 2019 04.
Article in English | MEDLINE | ID: mdl-30404019

ABSTRACT

Tissue injury/hypoxia and oxidative stress induced-extracellular adenosine triphosphate (ATP) can act as damage-associated molecular pattern molecules, which initiate inflammatory response. Our objective was to elucidate the role of extracellular ATP in skin fibrosis in systemic sclerosis (SSc). We identified that hypoxia enhanced ATP release and that extracellular ATP enhanced IL-6 production more significantly in SSc fibroblasts than in normal fibroblasts. There were no significant differences of P2X and P2Y receptor expression levels between normal and SSc fibroblasts. Nonselective P2 receptor antagonist and selective P2Y2 receptor antagonists, kaempferol and AR-C118925XX, significantly inhibited ATP-induced IL-6 production and phosphorylation of p38 in SSc fibroblasts. ATP-induced IL-6 production was significantly inhibited by p38 inhibitors, SB203580, and doramapimod. Collagen type I production in SSc fibroblasts by ATP-induced IL-6/IL-6 receptor trans-signaling was inhibited by kaempferol and SB203580. The amount of ATP in bleomycin-treated skin was increased, and administration of AR-C118925XX significantly inhibited bleomycin-induced dermal fibrosis in mice. These results suggest that vasculopathy-induced hypoxia and oxidative stress might enhance ATP release in the dermis in SSc and that extracellular ATP-induced phosphorylation of p38 via P2Y2 receptor might enhance IL-6 and collagen type I production in SSc fibroblasts. P2Y2 receptor antagonist therapy could be a treatment for skin sclerosis in patients with SSc.


Subject(s)
Receptors, Purinergic P2Y2/biosynthesis , Scleroderma, Systemic/complications , Skin/pathology , Adenosine Triphosphate/metabolism , Animals , Cells, Cultured , Endothelial Cells/metabolism , Endothelial Cells/pathology , Fibroblasts/metabolism , Fibroblasts/pathology , Fibrosis/etiology , Fibrosis/metabolism , Fibrosis/pathology , Humans , Immunoblotting , Interleukin-6/biosynthesis , Mice , Mice, Inbred C57BL , Scleroderma, Systemic/metabolism , Scleroderma, Systemic/pathology , Signal Transduction , Skin/metabolism
19.
Arthritis Rheumatol ; 70(10): 1661-1672, 2018 10.
Article in English | MEDLINE | ID: mdl-29676521

ABSTRACT

OBJECTIVE: Apelin/APJ signaling has been determined to regulate cardiac and arterial fibrosis and to be involved in the pathogenesis of pulmonary arterial hypertension. Our objective was to elucidate the role of apelin in skin fibrosis in systemic sclerosis (SSc). METHODS: Expression of apelin/APJ in normal and SSc fibroblasts was compared. Effects of small interfering RNA depletion and the addition of apelin in fibroblasts were analyzed. The effect of apelin injections on bleomycin-induced dermal fibrosis in mice was investigated. We analyzed the effects of the biased agonist of APJ, MM07, on skin fibrosis in vitro and in vivo. RESULTS: The expression of apelin in SSc fibroblasts was significantly lower than that in normal fibroblasts. Serum apelin levels were negatively correlated with the modified Rodnan skin thickness score in SSc patients. Stimulation with transforming growth factor ß1 (TGFß1) inhibited apelin expression in fibroblasts, suggesting that activation of TGFß1 signaling in SSc might be responsible for reduced apelin expression in SSc fibroblasts. Small interfering RNA depletion of apelin from fibroblasts significantly enhanced fibrosis-related gene expression, and treatment with apelin protein significantly inhibited TGFß1 signaling in fibroblasts. Administration of apelin significantly inhibited bleomycin-induced dermal fibrosis in mice. We demonstrated that MM07 had greater potential than apelin to inhibit fibrosis in vivo and in vitro. CONCLUSION: Collectively, TGFß1 signaling and apelin signaling may counteract each other in the fibrotic process of SSc. Inhibitory regulation of TGFß1-induced skin fibrosis by apelin/APJ signaling may be involved in the pathogenesis of SSc and could be a therapeutic target for fibrosis in SSc patients.


Subject(s)
Apelin Receptors/metabolism , Apelin/metabolism , Scleroderma, Systemic/pathology , Skin Diseases/pathology , Skin/pathology , Adult , Animals , Bleomycin , Cells, Cultured , Disease Models, Animal , Female , Fibroblasts/metabolism , Fibrosis , Humans , Male , Mice , Signal Transduction , Skin/cytology , Skin Diseases/chemically induced
SELECTION OF CITATIONS
SEARCH DETAIL
...