Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 111
Filter
Add more filters

Country/Region as subject
Affiliation country
Publication year range
1.
J Cutan Pathol ; 51(6): 430-433, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38525523

ABSTRACT

The histopathologic diagnosis of poorly differentiated cutaneous angiosarcoma can be challenging. We report a case of cutaneous epithelioid angiosarcoma with numerous multinucleated giant cells (MGCs) developing pulmonary metastasis. A 79-year-old man presented with a red-purple plaque on the scalp. A skin biopsy revealed epithelioid cell proliferation, admixed with numerous MGCs, and background hemorrhage. Vascular spaces were focally present and lined by atypical endothelial cells, including MGCs. Immunohistochemically, tumor cells, including MGCs, were positive for CD31, D2-40, and ERG. The patient received radiation therapy and chemotherapy, after which a follow-up CT scan revealed symptomless pneumothorax and pulmonary metastases. The patient received palliative partial lung resection, and the specimen revealed histopathological and immunohistochemical features similar to the primary cutaneous lesion. Our report expands the morphologic spectrum of cutaneous epithelioid angiosarcoma. Cutaneous angiosarcoma is an aggressive neoplasm; thus, awareness of this rare manifestation is important.


Subject(s)
Giant Cells , Hemangiosarcoma , Lung Neoplasms , Skin Neoplasms , Humans , Male , Aged , Skin Neoplasms/pathology , Skin Neoplasms/diagnosis , Skin Neoplasms/secondary , Lung Neoplasms/pathology , Lung Neoplasms/diagnosis , Lung Neoplasms/secondary , Giant Cells/pathology , Hemangiosarcoma/pathology , Hemangiosarcoma/diagnosis , Scalp/pathology , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , Epithelioid Cells/pathology
2.
Clin Exp Dermatol ; 48(3): 199-210, 2023 Mar 01.
Article in English | MEDLINE | ID: mdl-36656063

ABSTRACT

BACKGROUND: Epidermolytic ichthyosis (EI) is a major form of nonsyndromic inherited ichthyosis, characterized by erythroderma, marked hyperkeratosis and scale, bulla and erosion at birth, associated with KRT1/KRT10 mutations. The cytokine and chemokine profiles in EI are poorly understood, and specific treatment options have not been established. AIM: To explore novel biomarkers and therapeutic targets in patients with EI. METHODS: We analysed cytokine levels in serum and skin samples from 10 patients with inherited ichthyosis, including seven patients with EI. Wild-type and mutant KRT1 constructs were established and transfected into HaCaT cells, an immortalized keratinocyte cell line, for in vitro immunoblotting and immunocytochemistry analyses. RESULTS: Multiplex cytokine/chemokine analysis revealed that 10 cytokines/chemokines [interleukin (IL)-1ß, IL-4, IL-17A, IL-16, IL-18, IL-1 receptor-α, macrophage colony-stimulating factor, interferon-α2, basic fibroblast growth factor and monocyte chemotactic protein-3] were significantly increased in patients with EI. Furthermore, IL-18 levels were significantly higher in patients with EI [n = 7; 2714.1 (1438.0) pg mL-1] than in healthy controls [n = 11; 218.4 (28.4) pg mL-1, P < 0.01]. Immunohistochemical analyses showed that IL-18 expression was elevated in skin samples from patients with EI. Serum IL-18 levels correlated with the severity of ichthyosis, as measured by the Ichthyosis Scoring System. Immunoblotting analysis revealed that mature IL-18 levels were increased in the supernatant of mutant KRT1 expressing HaCaT cells. Additionally, these cells showed NLRP3 aggregation in the cytoplasm and ASC clustered around mutant keratin aggregations. These findings suggest that mutant keratin might promote the activation of the NLRP3 inflammasome and its downstream caspase-1-mediated IL-18 release in keratinocytes from patients with EI. CONCLUSIONS: Our results suggest that serum IL-18 is a severity marker released from the skin of patients with EI. Blockade of IL-18 may be a useful novel therapeutic option for patients with EI.


Subject(s)
Hyperkeratosis, Epidermolytic , Ichthyosis, Lamellar , Humans , Infant, Newborn , Cytokines , Hyperkeratosis, Epidermolytic/genetics , Interleukin-18 , Keratins , NLR Family, Pyrin Domain-Containing 3 Protein
3.
Hum Mutat ; 43(7): 877-881, 2022 07.
Article in English | MEDLINE | ID: mdl-35446444

ABSTRACT

An autosomal recessive disease is caused by biallelic loss-of-function mutations. However, when more than two disease-causing variants are found in a patient's gene, it is challenging to determine which two of the variants are responsible for the disease phenotype. Here, to decipher the pathogenic variants by precise haplotyping, we applied nanopore Cas9-targeted sequencing (nCATS) to three truncation COL7A1 variants detected in a patient with recessive dystrophic epidermolysis bullosa (EB). The distance between the most 5' and 3' variants was approximately 19 kb at the level of genomic DNA. nCATS successfully demonstrated that the most 5' and 3' variants were located in one allele while the variant in between was located in the other allele. Interestingly, the proband's mother, who was phenotypically intact, was heterozygous for the allele that harbored the two truncation variants, which could otherwise be misinterpreted as those of typical recessive dystrophic EB. Our study highlights the usefulness of nCATS as a tool to determine haplotypes of complicated genetic cases. Haplotyping of multiple variants in a gene can determine which variant should be therapeutically targeted when nucleotide-specific gene therapy is applied.


Subject(s)
Collagen Type VII , Epidermolysis Bullosa Dystrophica , CRISPR-Cas Systems , Collagen Type VII/genetics , Epidermolysis Bullosa Dystrophica/genetics , Epidermolysis Bullosa Dystrophica/pathology , Epidermolysis Bullosa Dystrophica/therapy , Genes, Recessive , Haplotypes , Humans , Mutation
4.
Proc Natl Acad Sci U S A ; 116(52): 26846-26852, 2019 Dec 26.
Article in English | MEDLINE | ID: mdl-31818947

ABSTRACT

Recessive dystrophic epidermolysis bullosa (RDEB) is a severe inherited skin disorder caused by mutations in the COL7A1 gene encoding type VII collagen (C7). The spectrum of severity depends on the type of mutation in the COL7A1 gene. C7 is the major constituent of anchoring fibrils (AFs) at the basement membrane zone (BMZ). Patients with RDEB lack functional C7 and have severely impaired dermal-epidermal stability, resulting in extensive blistering and open wounds on the skin that greatly affect the patient's quality of life. There are currently no therapies approved for the treatment of RDEB. Here, we demonstrated the correction of mutations in exon 19 (c.2470insG) and exon 32 (c.3948insT) in the COL7A1 gene through homology-directed repair (HDR). We used the clustered regulatory interspaced short palindromic repeats (CRISPR) Cas9-gRNAs system to modify induced pluripotent stem cells (iPSCs) derived from patients with RDEB in both the heterozygous and homozygous states. Three-dimensional human skin equivalents (HSEs) were generated from gene-corrected iPSCs, differentiated into keratinocytes (KCs) and fibroblasts (FBs), and grafted onto immunodeficient mice, which showed normal expression of C7 at the BMZ as well as restored AFs 2 mo postgrafting. Safety assessment for potential off-target Cas9 cleavage activity did not reveal any unintended nuclease activity. Our findings represent a crucial advance for clinical applications of innovative autologous stem cell-based therapies for RDEB.

6.
Acta Derm Venereol ; 99(12): 1166-1169, 2019 Nov 01.
Article in English | MEDLINE | ID: mdl-31453627

ABSTRACT

Some bone lesions are reported to mimic bone metastasis on imaging tests. Herein, we report a case of a 55-year-old Japanese man who presented with a skin tumor on the left lower extremity. He also had a history of recurrent generalized cutaneous blister and erosion formation since childhood. His skin lesions were diagnosed as cutaneous squamous cell carcinoma complicated by recessive dystrophic epidermolysis bullosa. Magnetic resonance imaging of the left lower extremity detected multiple focal bone lesions mimicking bone metastases in the left femur and tibia. However, bone biopsy revealed that the bone lesions were osteonecrosis without tumor cells. We suggest that cancer-induced osteonecrosis should be included in the differential diagnosis of bone lesions suspected of being metastases on magnetic resonance imaging.


Subject(s)
Bone Neoplasms/diagnostic imaging , Carcinoma, Squamous Cell/diagnostic imaging , Epidermolysis Bullosa Dystrophica/diagnostic imaging , Femoral Neoplasms/diagnostic imaging , Magnetic Resonance Imaging , Osteonecrosis/diagnostic imaging , Skin Neoplasms/diagnostic imaging , Tibia/diagnostic imaging , Biopsy , Bone Neoplasms/secondary , Carcinoma, Squamous Cell/secondary , Diagnosis, Differential , Epidermolysis Bullosa Dystrophica/pathology , Femoral Neoplasms/secondary , Humans , Male , Middle Aged , Osteonecrosis/pathology , Predictive Value of Tests , Skin Neoplasms/pathology , Tibia/pathology
7.
Acta Derm Venereol ; 99(12): 1110-1115, 2019 Nov 01.
Article in English | MEDLINE | ID: mdl-31513274

ABSTRACT

Nail-patella syndrome is an autosomal dominant disorder characterized by nail dysplasia and skeletal anomaly. Some patients have been shown to have ultrastructural abnormalities of the glomerular basement membrane that result in nephrosis. However, little has been reported on the epidermal basement membrane in this condition. This paper reports 2 families with nail-patella syndrome. Direct sequencing analysis of LMX1B revealed that family 1 and family 2 were heterozygous for the mutations c.140-1G>C and c.326+1G>C, respectively. To evaluate the epidermal basement membrane zone, ultrastructural and immunohistochemical analyses were performed using skin specimens obtained from the dorsal thumb. Electron microscopy showed intact hemidesmosomes, lamina lucida, lamina densa, and anchoring fibrils. Immunofluorescence studies with antibodies against components of the epidermal basement membrane zone revealed a normal expression pattern among the components, including type IV collagen. These data suggest that nail dysplasia in patients with nail-patella syndrome is not caused by structural abnormalities of the epidermal basement membrane.


Subject(s)
Basement Membrane/chemistry , Basement Membrane/ultrastructure , Collagen Type IV/analysis , Epidermis/chemistry , Epidermis/ultrastructure , Fluorescent Antibody Technique , Microscopy, Electron, Transmission , Nail-Patella Syndrome/diagnosis , Biomarkers/analysis , Child , Female , Genetic Predisposition to Disease , Heterozygote , Humans , Infant , LIM-Homeodomain Proteins/genetics , Male , Mutation , Nail-Patella Syndrome/genetics , Nail-Patella Syndrome/metabolism , Nail-Patella Syndrome/pathology , Phenotype , Predictive Value of Tests , Transcription Factors/genetics
8.
Proc Natl Acad Sci U S A ; 113(20): 5676-81, 2016 May 17.
Article in English | MEDLINE | ID: mdl-27143720

ABSTRACT

Genome editing with engineered site-specific endonucleases involves nonhomologous end-joining, leading to reading frame disruption. The approach is applicable to dominant negative disorders, which can be treated simply by knocking out the mutant allele, while leaving the normal allele intact. We applied this strategy to dominant dystrophic epidermolysis bullosa (DDEB), which is caused by a dominant negative mutation in the COL7A1 gene encoding type VII collagen (COL7). We performed genome editing with TALENs and CRISPR/Cas9 targeting the mutation, c.8068_8084delinsGA. We then cotransfected Cas9 and guide RNA expression vectors expressed with GFP and DsRed, respectively, into induced pluripotent stem cells (iPSCs) generated from DDEB fibroblasts. After sorting, 90% of the iPSCs were edited, and we selected four gene-edited iPSC lines for further study. These iPSCs were differentiated into keratinocytes and fibroblasts secreting COL7. RT-PCR and Western blot analyses revealed gene-edited COL7 with frameshift mutations degraded at the protein level. In addition, we confirmed that the gene-edited truncated COL7 could neither associate with normal COL7 nor undergo triple helix formation. Our data establish the feasibility of mutation site-specific genome editing in dominant negative disorders.


Subject(s)
Epidermolysis Bullosa Dystrophica/genetics , Induced Pluripotent Stem Cells/physiology , Adult , Animals , Base Sequence , Cell Differentiation , Cell Transformation, Neoplastic , Cells, Cultured , Collagen Type VII/genetics , Collagen Type VII/metabolism , DNA Mutational Analysis , Fibroblasts/metabolism , Gene Editing , Gene Expression , Humans , Induced Pluripotent Stem Cells/transplantation , Male , Mice, Nude , Teratoma/pathology
9.
Hum Mol Genet ; 25(2): 328-39, 2016 Jan 15.
Article in English | MEDLINE | ID: mdl-26604146

ABSTRACT

In skin, basal keratinocytes in the epidermis are tightly attached to the underlying dermis by the basement membrane (BM). The correct expression of hemidesmosomal and extracellular matrix (ECM) proteins is essential for BM formation, and the null-expression of one molecule may induce blistering diseases associated with immature BM formation in humans. However, little is known about the significance of post-translational processing of hemidesmosomal or ECM proteins in BM formation. Here we show that the C-terminal cleavage of hemidesmosomal transmembrane collagen XVII (COL17) is essential for correct BM formation. The homozygous p.R1303Q mutation in COL17 induces BM duplication and blistering in humans. Although laminin 332, a major ECM protein, interacts with COL17 around p.R1303, the mutation leaves the binding of both molecules unchanged. Instead, the mutation hampers the physiological C-terminal cleavage of COL17 in the ECM. Consequently, non-cleaved COL17 ectodomain remnants induce the aberrant deposition of laminin 332 in the ECM, which is thought to be the major pathogenesis of the BM duplication that results from this mutation. As an example of impaired cleavage of COL17, this study shows that regulated processing of hemidesmosomal proteins is essential for correct BM organization in skin.


Subject(s)
Autoantigens/genetics , Autoantigens/metabolism , Basement Membrane/metabolism , Blister/metabolism , Cell Adhesion Molecules/metabolism , Non-Fibrillar Collagens/genetics , Non-Fibrillar Collagens/metabolism , Protein Processing, Post-Translational , Adult , Blister/genetics , Child , Epidermis/metabolism , Female , Humans , Japan , Keratinocytes/metabolism , Middle Aged , Mutation , Pedigree , Kalinin , Collagen Type XVII
10.
J Am Acad Dermatol ; 79(6): 1039-1046, 2018 Dec.
Article in English | MEDLINE | ID: mdl-29928911

ABSTRACT

BACKGROUND: Mycosis fungoides (MF) is the most common cutaneous T-cell lymphoma. Early-stage MF patches or plaques often resemble inflammatory skin disorders (ISDs), including psoriasis and atopic dermatitis. Cell adhesion molecule 1 gene (CADM1), which was initially identified as a tumor suppressor gene in human non-small cell lung cancer, has been reported as a diagnostic marker for adult T-cell leukemia/lymphoma. OBJECTIVE: We investigated CADM1 expression in MF neoplastic cells, especially during early stages, and evaluated its usefulness as a diagnostic marker for MF. METHODS: We conducted a retrospective study by using immunohistochemical staining and confirmed the expression of CADM1 in MF. In addition, we compared CADM1 messenger RNA expression in microdissected MF samples and ISD samples. RESULTS: In the overall study period, 55 of 58 MF samples (94.8 %) stained positive for CADM1. None of the 50 ISD samples showed positive reactivity (P < .0001). We found CADM1 messenger RNA expression in the intradermal lymphocytes of patients with MF but not in those of patients with an ISD. LIMITATIONS: We did not conduct a validation study for MF cases in other institutions. CONCLUSIONS: CADM1-positive cells can be identified in early stages with fewer infiltrating cells and may be useful as a diagnostic marker for early-stage MF.


Subject(s)
Biomarkers, Tumor/analysis , Cell Adhesion Molecule-1/analysis , Mycosis Fungoides/chemistry , Neoplasm Proteins/analysis , Adult , Aged , Aged, 80 and over , Cell Adhesion Molecule-1/biosynthesis , Cell Adhesion Molecule-1/genetics , Dermatitis/metabolism , Early Diagnosis , Female , Gene Expression Regulation, Neoplastic , Humans , Japan/epidemiology , Lymphocytes/metabolism , Lymphocytes/pathology , Middle Aged , Mycosis Fungoides/diagnosis , Mycosis Fungoides/genetics , Mycosis Fungoides/pathology , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/genetics , Neoplasm Staging , RNA, Messenger/biosynthesis , RNA, Neoplasm/biosynthesis , Retrospective Studies
12.
Hum Mutat ; 38(12): 1666-1670, 2017 12.
Article in English | MEDLINE | ID: mdl-28941359

ABSTRACT

Plectin is a linker protein that interacts with intermediate filaments and ß4 integrin in hemidesmosomes of the epidermal basement membrane zone (BMZ). Type XVII collagen (COL17) has been suggested as another candidate plectin binding partner in hemidesmosomes. Here, we demonstrate that plectin-COL17 binding helps to maintain epidermal BMZ organization. We identified an epidermolysis bullosa (EB) simplex patient as having markedly diminished expression of plectin and COL17 in skin. The patient is compound heterozygous for sequence variants in the plectin gene (PLEC); one is a truncation and the other is a small in-frame deletion sequence variant. The in-frame deletion is located in the putative COL17-binding domain of plectin and abolishes the plectin-COL17 interaction in vitro. These results imply that disrupted interaction between plectin and COL17 is involved in the development of EB. Our study suggests that protein-protein binding defects may underlie EB in patients with unidentified disease-causing sequence variants.


Subject(s)
Autoantigens/metabolism , Epidermolysis Bullosa Simplex/genetics , Non-Fibrillar Collagens/metabolism , Plectin/genetics , Autoantigens/genetics , Basement Membrane/metabolism , Epidermolysis Bullosa Simplex/diagnosis , Epidermolysis Bullosa Simplex/pathology , Genetic Variation , Hemidesmosomes/metabolism , Humans , Infant, Newborn , Keratinocytes/metabolism , Male , Non-Fibrillar Collagens/genetics , Plectin/metabolism , Protein Binding , Protein Domains , Sequence Deletion , Skin/pathology , Collagen Type XVII
13.
Exp Dermatol ; 26(9): 811-819, 2017 09.
Article in English | MEDLINE | ID: mdl-28111846

ABSTRACT

In this study, we generated a new set of monoclonal antibodies (mAbs) to bovine and human type VII collagen (COL7) by immunizing mice with bovine cornea-derived basement membrane zone (BMZ) fraction. The four mAbs, tentatively named as COL7-like mAbs, showed speckled subepidermal staining in addition to linear BMZ staining of normal human skin and bovine cornea, a characteristic immunofluorescence feature of COL7, but showed no reactivity with COL7 by in vitro biochemical analyses. Taking advantage of the phenomenon that COL7-like mAbs did not react with mouse BMZ, we compared immunofluorescence reactivity between wild-type and COL7-rescued humanized mice and found that COL7-like mAbs reacted with BMZ of COL7-rescued humanized mice. In ELISAs, COL7-like mAbs reacted with intact triple-helical mammalian recombinant protein (RP) of COL7 but not with bacterial RP. Furthermore, COL7-like mAbs did not react with COL7 within either cultured DJM-1 cells or basal cells of skin of a bullous dermolysis of the newborn patient. These results confirmed that COL7-like mAbs reacted with human and bovine COL7. The epitopes for COL7-like mAbs were considered to be present only on mature COL7 after secretion from keratinocytes and deposition to BMZ and to be easily destroyed during immunoblotting procedure. Additional studies indicated association of the speckled subepidermal staining with both type IV collagen and elastin. These unique anti-COL7 mAbs should be useful in studies of both normal and diseased conditions, particularly dystrophic epidermolysis bullosa, which produces only immature COL7.


Subject(s)
Basement Membrane/metabolism , Collagen Type VII/immunology , Collagen Type VII/metabolism , Animals , Antibodies, Monoclonal/biosynthesis , Cattle , HEK293 Cells , Humans , Mice
14.
Lab Invest ; 96(12): 1301-1310, 2016 12.
Article in English | MEDLINE | ID: mdl-27775687

ABSTRACT

Macropinocytosis is an endocytic pathway that is involved in the nonselective fluid uptake of extracellular fluid. Bullous pemphigoid (BP) is an autoimmune subepidermal blistering disease associated with autoantibodies to type XVII collagen (COL17), which is a component of hemidesmosome. When keratinocytes are treated with BP-IgG, COL17 internalizes into cells by way of the macropinocytosis. We investigated the mechanism of COL17 macropinocytosis using DJM-1 cells, a cutaneous squamous cell carcinoma cell line. First, non-hemidesmosomal COL17 was preferentially depleted by stimulation with the BP-IgG in the DJM-1 cells. To investigate the signaling involved in COL17-macropinocytosis, the inhibition of small GTPase family members Rac1 and Cdc42 was found to strongly repress COL17 internalization; in addition, the Rho inhibitor also partially blocked that internalization, suggesting these small GTPases are involved in signaling to mediate COL17-macropinocytosis. Western blotting using Phostag-SDS-PAGE demonstrated high levels of COL17 phosphorylation in DJM-1 cells under steady-state condition. Treatment with BP-IgG increased the intracellular calcium level within a minute, and induced the overabundant phosphorylation of COL17. The overabundant phosphorylation of COL17 was suppressed by a protein kinase C (PKC) inhibitor. In addition, PKC inhibitor repressed COL17 endocytosis using cell culture and organ culture systems. Finally, the depletion of COL17 was not observed in the HEK293 cells transfected COL17 without intracellular domain. These results suggest that COL17 internalization induced by BP-IgG may be mediated by a PKC pathway. In summary, BP-IgG initially binds to COL17 distributed on the plasma membrane, and COL17 may be internalized by means of a macropinocytic pathway related to the phosphorylation of the intracellular domain by PKC.


Subject(s)
Autoantibodies/pharmacology , Autoantigens/metabolism , Immunoglobulin G/pharmacology , Keratinocytes/drug effects , Non-Fibrillar Collagens/metabolism , Pemphigoid, Bullous/immunology , Pinocytosis/drug effects , Protein Kinase C/metabolism , Animals , Antibodies, Monoclonal/pharmacology , Autoantigens/chemistry , Autoantigens/genetics , Calcium Signaling/drug effects , Cell Line, Tumor , HEK293 Cells , Humans , Keratinocytes/immunology , Keratinocytes/metabolism , Keratinocytes/pathology , Mice , Non-Fibrillar Collagens/chemistry , Non-Fibrillar Collagens/genetics , Pemphigoid, Bullous/metabolism , Pemphigoid, Bullous/pathology , Peptide Fragments , Phosphorylation/drug effects , Protein Interaction Domains and Motifs , Protein Kinase C/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Protein Processing, Post-Translational/drug effects , Recombinant Proteins , Tissue Culture Techniques , Up-Regulation/drug effects , Collagen Type XVII
15.
J Immunol ; 193(9): 4415-28, 2014 Nov 01.
Article in English | MEDLINE | ID: mdl-25261486

ABSTRACT

Complement activation and subsequent recruitment of inflammatory cells at the dermal/epidermal junction are thought to be essential for blister formation in bullous pemphigoid (BP), an autoimmune blistering disease induced by autoantibodies against type XVII collagen (COL17); however, this theory does not fully explain the pathological features of BP. Recently, the involvement of complement-independent pathways has been proposed. To directly address the question of the necessity of the complement activation in blister formation, we generated C3-deficient COL17-humanized mice. First, we show that passive transfer of autoantibodies from BP patients induced blister formation in neonatal C3-deficient COL17-humanized mice without complement activation. By using newly generated human and murine mAbs against the pathogenic noncollagenous 16A domain of COL17 with high (human IgG1, murine IgG2), low (murine IgG1), or no (human IgG4) complement activation abilities, we demonstrate that the deposition of Abs, and not complements, is relevant to the induction of blister formation in neonatal and adult mice. Notably, passive transfer of BP autoantibodies reduced the amount of COL17 in lesional mice skin, as observed in cultured normal human keratinocytes treated with the same Abs. Moreover, the COL17 depletion was associated with a ubiquitin/proteasome pathway. In conclusion, the COL17 depletion induced by BP autoantibodies, and not complement activation, is essential for the blister formation under our experimental system.


Subject(s)
Autoantibodies/immunology , Blister/immunology , Complement Activation/immunology , Complement System Proteins/immunology , Pemphigoid, Bullous/immunology , Pemphigoid, Bullous/pathology , Animals , Animals, Newborn , Antigen-Antibody Complex/immunology , Autoantigens/immunology , Autoantigens/metabolism , Cell Line , Complement System Proteins/deficiency , Epitopes/immunology , Humans , Immunization, Passive , Immunoglobulin G/immunology , Mice , Mice, Transgenic , Non-Fibrillar Collagens/immunology , Non-Fibrillar Collagens/metabolism , Pemphigoid, Bullous/genetics , Pemphigoid, Bullous/metabolism , Proteasome Endopeptidase Complex/metabolism , Protein Binding/immunology , Proteolysis , Skin/immunology , Skin/metabolism , Skin/pathology , Ubiquitination , Collagen Type XVII
18.
J Immunol ; 188(11): 5792-9, 2012 Jun 01.
Article in English | MEDLINE | ID: mdl-22523387

ABSTRACT

In bullous pemphigoid (BP), the most prevalent autoimmune blistering disease, type XVII collagen (COL17) is targeted by circulating autoantibodies. BP is thought to be an autoantibody-mediated complement-fixing blistering disease, and a juxtamembranous noncollagenous 16A (NC16A) domain spanning Glu(490) to Arg(566) was proved to be the main pathogenic region on COL17, although precise pathogenic epitopes within NC16A have not been elucidated. In this study, we showed that injection of rabbit IgG Abs targeting Asp(522) to Gln(545) induced skin fragility associated with in vivo deposition of IgG and complement in neonatal COL17-humanized mice. Notably, immunoadsorption of rabbit anti-NC16A IgG Ab with this epitope (Asp(522) to Gln(545)) or the anti-NC16A IgG administered together with the peptides of this epitope as a decoy ameliorated skin fragility in the injected neonatal COL17-humanized mice compared with the anti-NC16A IgG alone even though all of the mice showed both IgG and complement deposition. These results led us to investigate an additional, complement-independent mechanism of skin fragility in the mice injected with anti-COL17 Abs. The rabbit anti-NC16A IgG depleted the expression of COL17 in cultured normal human keratinocytes, whereas immunoadsorption of the same IgG with this epitope significantly suppressed the depletion effect. Moreover, passive transfer of F(ab')(2) fragments of the human BP or rabbit IgG Abs against COL17 demonstrated skin fragility in neonatal COL17-humanized mice. In summary, this study reveals the importance of Abs directed against distinct epitopes on COL17, which induce skin fragility in complement-dependent as well as complement-independent ways.


Subject(s)
Autoantibodies/administration & dosage , Autoantigens/immunology , Complement System Proteins/physiology , Epitopes/immunology , Non-Fibrillar Collagens/immunology , Pemphigoid, Bullous/immunology , Animals , Animals, Newborn , Autoantibodies/adverse effects , Autoantigens/adverse effects , Cells, Cultured , Epitopes/adverse effects , Humans , Immunization, Passive , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Non-Fibrillar Collagens/adverse effects , Pemphigoid, Bullous/pathology , Rabbits , Collagen Type XVII
19.
J Dermatol ; 51(1): 56-61, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37804078

ABSTRACT

Nevus lipomatosus cutaneous superficialis is a rare, benign hamartoma characterized by mature adipocyte proliferation in the dermis. It is frequently difficult to distinguish clinically from soft tissue tumors, including lipoma, neurofibroma, venous malformation, and angiolipoma. Notably, the classical form, which shows multiple and sometimes enlarged nodules, is difficult to differentiate from liposarcoma based on clinical examination, computed tomography, and magnetic resonance imaging findings. Therefore, to ascertain the utility of ultrasonography in diagnosing nevus lipomatosus cutaneous superficialis, sonographic examinations were performed on eight patients with nevus lipomatosus cutaneous superficialis. All patients had ill-defined hyperechoic masses in the dermis or from the dermis to the subcutis, and the posterior echoes were attenuated in seven patients. Color Doppler sonography revealed no blood flow to the lesions. Ultrasound images were created using the reflections of ultrasound waves at interfaces with different acoustic impedances. Therefore, it is assumed that, in nevus lipomatosus cutaneous superficialis, the ultrasound beam is scattered by ectopic mature adipocytes intermingled with collagen bundles, which are shown as hyperechoic masses. Furthermore, the scattering of the ultrasound beam is thought to reduce tissue penetration, which may attenuate the posterior echo.


Subject(s)
Hamartoma , Lipomatosis , Nevus , Skin Neoplasms , Soft Tissue Neoplasms , Humans , Lipomatosis/diagnostic imaging , Lipomatosis/pathology , Skin Neoplasms/diagnostic imaging , Skin Neoplasms/pathology , Hamartoma/diagnostic imaging , Hamartoma/pathology , Nevus/diagnostic imaging , Nevus/pathology
20.
J Dermatol ; 51(1): 125-129, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37789597

ABSTRACT

Infancy associated eosinophilic pustular folliculitis (I-EPF) is a clinical variant of EPF that develops in childhood. Previous studies have suggested that I-EPF exhibits clinical and histological differences distinct from other variants, including classic EPF. Herein, we report two patients with I-EPF treated with topical indomethacin. These two cases exhibited less perifollicular and more perivascular eosinophilic infiltration, which is different in distribution from that of classic EPF. Immunohistochemical study demonstrated that the infiltrating mononuclear cells were CD4-dominant T cells in classic EPF and I-EPF, whereas the number of CD68-positive cells was significantly higher in classic EPF than in I-EPF. Immunohistochemical staining was also performed for eosinophilic pustular folliculitis (HPGDS), which has been reported to induce eosinophils and is a therapeutic target of indomethacin in classic EPF. HPGDS-positive cells were also observed in I-EPF, which may explain the effectiveness of topical indomethacin. Although clinical and histopathological features of I-EPF are different from other variants, the arachidonic acid pathway could be involved in eosinophil infiltration, not only in classic EPF but also in I-EPF.


Subject(s)
Eosinophilia , Folliculitis , Skin Diseases, Vesiculobullous , Humans , Indomethacin/therapeutic use , Eosinophilia/drug therapy , Eosinophilia/pathology , Folliculitis/drug therapy , Folliculitis/pathology , Skin Diseases, Vesiculobullous/drug therapy , Skin Diseases, Vesiculobullous/pathology
SELECTION OF CITATIONS
SEARCH DETAIL