Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
Int J Mol Sci ; 22(4)2021 Feb 11.
Article in English | MEDLINE | ID: mdl-33670258

ABSTRACT

The recessive form of dystrophic epidermolysis bullosa (RDEB) is a debilitating disease caused by impairments in the junctions of the dermis and the basement membrane of the epidermis. Mutations in the COL7A1 gene induce multiple abnormalities, including chronic inflammation and profibrotic changes in the skin. However, the correlations between the specific mutations in COL7A1 and their phenotypic output remain largely unexplored. The mutations in the COL7A1 gene, described here, were found in the DEB register. Among them, two homozygous mutations and two cases of compound heterozygous mutations were identified. We created the panel of primary patient-specific RDEB fibroblast lines (FEB) and compared it with control fibroblasts from healthy donors (FHC). The set of morphological features and the contraction capacity of the cells distinguished FEB from FHC. We also report the relationships between the mutations and several phenotypic traits of the FEB. Based on the analysis of the available RNA-seq data of RDEB fibroblasts, we performed an RT-qPCR gene expression analysis of our cell lines, confirming the differential status of multiple genes while uncovering the new ones. We anticipate that our panels of cell lines will be useful not only for studying RDEB signatures but also for investigating the overall mechanisms involved in disease progression.


Subject(s)
Collagen Type VII , Dermis , Epidermolysis Bullosa Dystrophica , Fibroblasts , Gene Expression Regulation , Homozygote , Mutation , Adolescent , Adult , Child , Collagen Type VII/biosynthesis , Collagen Type VII/genetics , Dermis/metabolism , Dermis/pathology , Epidermolysis Bullosa Dystrophica/genetics , Epidermolysis Bullosa Dystrophica/metabolism , Epidermolysis Bullosa Dystrophica/pathology , Female , Fibroblasts/metabolism , Fibroblasts/pathology , Humans , Male , Middle Aged
2.
ACS Appl Mater Interfaces ; 11(34): 30661-30672, 2019 Aug 28.
Article in English | MEDLINE | ID: mdl-31390173

ABSTRACT

Recessive dystrophic epidermolysis bullosa (RDEB) is a severe congenital skin fragility disease caused by COL7A1 mutations that result in type VII collagen (C7) deficiency. Herein, we report a synergistic polyplex system that can efficiently restore C7 expression in RDEB keratinocytes. A highly branched multifunctional poly(ß-amino ester) (HPAE), termed as HC32-122, was optimized systematically as the high-performance gene delivery vector for keratinocytes, achieving much higher transfection capability than polyethylenimine, SuperFect, and Lipofectamine 2000 without inducing obvious cytotoxicity. Concurrently, a 12 kb length minicircle DNA encoding ∼9 kb full-length COL7A1 (MCC7) devoid of bacterial sequence was biosynthesized as the therapeutic gene. Combining the highly potent polymer and the miniaturized gene structure, HC32-122/MCC7 polyplexes achieve 96.4% cellular uptake efficiency, 4019-fold COL7A1 mRNA enhancement, and robust recombinant C7 expression. Structure-property investigations reveal that HC32-122 can effectively condense MCC7 to form small, uniform, compact, and positively charged spherical nanoparticles with high DNA release flexibility. Moreover, formulation study shows that sucrose is conductive to lyophilized HC32-122/DNA polyplexes for maintaining the transfection capability. Direct frozen polyplexes can maintain full gene transfection capability after one-year storage. High efficiency, biocompatibility, facile manipulation, and long-term stability make the HC32-122/MCC7 system a promising bench-to-bed candidate for treating the debilitating RDEB.


Subject(s)
Collagen Type VII , Epidermolysis Bullosa , Gene Transfer Techniques , Genetic Therapy , Keratinocytes , Nanoparticles/chemistry , Animals , Cell Line , Collagen Type VII/biosynthesis , Collagen Type VII/genetics , Epidermolysis Bullosa/genetics , Epidermolysis Bullosa/metabolism , Epidermolysis Bullosa/pathology , Epidermolysis Bullosa/therapy , Humans , Keratinocytes/metabolism , Keratinocytes/pathology , Polymers/chemistry , Polymers/pharmacology
3.
Cell Transplant ; 28(9-10): 1242-1256, 2019.
Article in English | MEDLINE | ID: mdl-31271052

ABSTRACT

The blistering disease recessive dystrophic epidermolysis bullosa (RDEB) is caused by mutations in the gene encoding collagen VII (COL7), which forms anchoring fibrils that attach the epidermis to the dermis. Cutaneous gene therapy to restore COL7 expression in RDEB patient cells has been proposed, and cultured epithelial autograft containing COL7-modified keratinocytes was previously tested in clinical trials. Because COL7 in normal skin is expressed in both fibroblasts and keratinocytes, cutaneous gene therapy using a bilayer skin substitute may enable faster restoration of anchoring fibrils. Hypothetically, COL7 expression in either dermal fibroblasts or epidermal keratinocytes might be sufficient for functional anchoring fibril formation in a bilayer skin substitute. To test this, engineered skin substitutes (ESS) were prepared using four combinations of normal + RDEB cells: (1) RDEB fibroblasts + RDEB keratinocytes; (2) RDEB fibroblasts + normal keratinocytes; (3) normal fibroblasts + RDEB keratinocytes; and (4) normal fibroblasts + normal keratinocytes. ESS were incubated in vitro for 2 weeks prior to grafting to full-thickness wounds in immunodeficient mice. Biopsies were analyzed in vitro and at 1, 2, or 3 weeks after grafting. COL7 was undetectable in ESS prepared using all RDEB cells (group 1), and macroscopic blistering was observed by 2 weeks after grafting in ESS containing RDEB cells. COL7 was expressed, in vitro and in vivo, in ESS prepared using combinations of normal + RDEB cells (groups 2 and 3) or all normal cells (group 4). However, transmission electron microscopy revealed structurally normal anchoring fibrils, in vitro and by week 2 in vivo, only in ESS prepared using all normal cells (group 4). The results suggest that although COL7 protein is produced in engineered skin when cells in only one layer express the COL7 gene, formation of structurally normal anchoring fibrils appears to require expression of COL7 in both dermal fibroblasts and epidermal keratinocytes.


Subject(s)
Collagen Type VII/biosynthesis , Fibroblasts , Gene Expression Regulation , Keratinocytes , Skin, Artificial , Tissue Engineering , Adult , Animals , Collagen Type VII/genetics , Epidermolysis Bullosa Dystrophica/genetics , Epidermolysis Bullosa Dystrophica/metabolism , Fibroblasts/metabolism , Fibroblasts/pathology , Fibroblasts/transplantation , Heterografts , Humans , Keratinocytes/metabolism , Keratinocytes/pathology , Keratinocytes/transplantation , Male , Mice , Mutation , Wound Healing , Wounds and Injuries/genetics , Wounds and Injuries/metabolism , Wounds and Injuries/pathology
4.
Exp Dermatol ; 28(10): 1153-1155, 2019 10.
Article in English | MEDLINE | ID: mdl-30019435

ABSTRACT

Human skin graft mouse models are widely used to investigate and develop therapeutic strategies for the severe generalized form of recessive dystrophic epidermolysis bullosa (RDEB), which is caused by biallelic null mutations in COL7A1 and the complete absence of type VII collagen (C7). Most therapeutic approaches are focused on reintroducing C7. Therefore, C7 and anchoring fibrils are widely used as readouts in therapeutic research with skin graft models. In this study, we investigated the expression pattern of human and murine C7 in a grafting model, in which human skin is reconstituted out of in vitro cultured keratinocytes and fibroblasts. The model revealed that murine C7 was deposited in both human healthy control and RDEB skin grafts. Moreover, we found that murine C7 is able to form anchoring fibrils in human grafts. Therefore, we advocate the use of human-specific antibodies when assessing the reintroduction of C7 using RDEB skin graft mouse models.


Subject(s)
Collagen Type VII/biosynthesis , Collagen Type VII/metabolism , Epidermolysis Bullosa Dystrophica/pathology , Fibroblasts/metabolism , Keratinocytes/metabolism , Skin Transplantation , Animals , Antibodies, Heterophile/immunology , Basement Membrane/metabolism , Cells, Cultured , Collagen Type VII/deficiency , Collagen Type VII/genetics , Collagen Type VII/immunology , Dermis/pathology , Epidermolysis Bullosa Dystrophica/immunology , Female , Fibroblasts/transplantation , Gene Expression , Heterografts , Humans , Keratinocytes/transplantation , Male , Mice , Mice, SCID , Models, Animal , Skin Window Technique
5.
PLoS One ; 10(11): e0141989, 2015.
Article in English | MEDLINE | ID: mdl-26528823

ABSTRACT

All previous reports concerning the effect of stretch on cultured skin cells dealt with experiments on epidermal keratinocytes or dermal fibroblasts alone. The aim of the present study was to develop a system that allows application of stretch stimuli to human skin equivalents (HSEs), prepared by coculturing of these two types of cells. In addition, this study aimed to analyze the effect of a stretch on keratinization of the epidermis and on the basement membrane. HSEs were prepared in a gutter-like structure created with a porous silicone sheet in a silicone chamber. After 5-day stimulation with stretching, HSEs were analyzed histologically and immunohistologically. Stretch-stimulated HSEs had a thicker epidermal layer and expressed significantly greater levels of laminin 5 and collagen IV/VII in the basal layer compared with HSEs not subjected to stretch stimulation. Transmission electron microscopy revealed that the structure of the basement membrane was more developed in HSEs subjected to stretching. Our model may be relevant for extrapolating the effect of a stretch on the skin in a state similar to an in vivo system. This experimental system may be useful for analysis of the effects of stretch stimuli on skin properties and wound healing and is also expected to be applicable to an in vitro model of a hypertrophic scar in the future.


Subject(s)
Basement Membrane/metabolism , Cicatrix, Hypertrophic/metabolism , Epidermis/metabolism , Fibroblasts/metabolism , Stress, Mechanical , Wound Healing , Basement Membrane/pathology , Cell Adhesion Molecules/biosynthesis , Cells, Cultured , Cicatrix, Hypertrophic/pathology , Collagen Type IV/biosynthesis , Collagen Type VII/biosynthesis , Epidermis/pathology , Fibroblasts/pathology , Humans , Kalinin
6.
EMBO Mol Med ; 7(4): 380-93, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25724200

ABSTRACT

There is a widespread agreement from patient and professional organisations alike that the safety of stem cell therapeutics is of paramount importance, particularly for ex vivo autologous gene therapy. Yet current technology makes it difficult to thoroughly evaluate the behaviour of genetically corrected stem cells before they are transplanted. To address this, we have developed a strategy that permits transplantation of a clonal population of genetically corrected autologous stem cells that meet stringent selection criteria and the principle of precaution. As a proof of concept, we have stably transduced epidermal stem cells (holoclones) obtained from a patient suffering from recessive dystrophic epidermolysis bullosa. Holoclones were infected with self-inactivating retroviruses bearing a COL7A1 cDNA and cloned before the progeny of individual stem cells were characterised using a number of criteria. Clonal analysis revealed a great deal of heterogeneity among transduced stem cells in their capacity to produce functional type VII collagen (COLVII). Selected transduced stem cells transplanted onto immunodeficient mice regenerated a non-blistering epidermis for months and produced a functional COLVII. Safety was assessed by determining the sites of proviral integration, rearrangements and hit genes and by whole-genome sequencing. The progeny of the selected stem cells also had a diploid karyotype, was not tumorigenic and did not disseminate after long-term transplantation onto immunodeficient mice. In conclusion, a clonal strategy is a powerful and efficient means of by-passing the heterogeneity of a transduced stem cell population. It guarantees a safe and homogenous medicinal product, fulfilling the principle of precaution and the requirements of regulatory affairs. Furthermore, a clonal strategy makes it possible to envision exciting gene-editing technologies like zinc finger nucleases, TALENs and homologous recombination for next-generation gene therapy.


Subject(s)
Collagen Type VII , Epidermolysis Bullosa Dystrophica/therapy , Genetic Therapy/methods , Stem Cells/metabolism , Transduction, Genetic , Adult , Animals , Cells, Cultured , Collagen Type VII/biosynthesis , Collagen Type VII/genetics , Epidermis , Epidermolysis Bullosa Dystrophica/genetics , Epidermolysis Bullosa Dystrophica/metabolism , Epidermolysis Bullosa Dystrophica/pathology , Female , Heterografts , Humans , Infant, Newborn , Male , Mice , Mice, SCID , Retroviridae/genetics , Stem Cell Transplantation , Stem Cells/pathology
7.
J Vis Exp ; (85)2014 Mar 18.
Article in English | MEDLINE | ID: mdl-24686341

ABSTRACT

3D organotypic cultures of epithelial cells on a matrix embedded with mesenchymal cells are widely used to study epithelial cell differentiation and invasion. Rat tail type I collagen and/or matrix derived from Engelbreth-Holm-Swarm mouse sarcoma cells have been traditionally employed as the substrates to model the matrix or stromal microenvironment into which mesenchymal cells (usually fibroblasts) are populated. Although experiments using such matrices are very informative, it can be argued that due to an overriding presence of a single protein (such as in type I Collagen) or a high content of basement membrane components and growth factors (such as in matrix derived from mouse sarcoma cells), these substrates do not best reflect the contribution to matrix composition made by the stromal cells themselves. To study native matrices produced by primary dermal fibroblasts isolated from patients with a tumor prone, genetic blistering disorder (recessive dystrophic epidermolysis bullosa), we have adapted an existing native matrix protocol to study tumor cell invasion. Fibroblasts are induced to produce their own matrix over a prolonged period in culture. This native matrix is then detached from the culture dish and epithelial cells are seeded onto it before the entire coculture is raised to the air-liquid interface. Cellular differentiation and/or invasion can then be assessed over time. This technique provides the ability to assess epithelial-mesenchymal cell interactions in a 3D setting without the need for a synthetic or foreign matrix with the only disadvantage being the prolonged period of time required to produce the native matrix. Here we describe the application of this technique to assess the ability of a single molecule expressed by fibroblasts, type VII collagen, to inhibit tumor cell invasion.


Subject(s)
Carcinoma, Squamous Cell/pathology , Skin Neoplasms/pathology , Stromal Cells/pathology , Tissue Engineering/methods , Tumor Microenvironment/physiology , Animals , Cell Differentiation/physiology , Cell Growth Processes/physiology , Collagen Type VII/biosynthesis , Collagen Type VII/genetics , Fibroblasts/metabolism , Fibroblasts/pathology , Fibroblasts/physiology , Humans , Keratinocytes/pathology , Mice , Neoplasm Invasiveness , Rats , Sarcoma, Experimental/metabolism , Sarcoma, Experimental/pathology
8.
Hum Pathol ; 45(2): 382-93, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24439225

ABSTRACT

Acute tubular necrosis (ATN), elicited by ischemia and/or toxicity, is a potentially life-threatening condition. Histologically, ATN corresponds to necrosis and detachment of renal tubular epithelial cells. However, the tubules possess a considerable regenerative capacity and may be restored. We have previously identified a scattered population of progenitor-like cells within the proximal tubules, sharing marker expression with the parietal epithelial cells of Bowman's capsule as well as with renal tubules regenerating after ATN. In the present analysis, we use transmission electron microscopy, immunoelectron microscopy and immunofluorescence of human kidney cortex to further explore these cells. We demonstrate that the cells are smaller and have drastically fewer mitochondria than the surrounding proximal tubule cells. They also display strong expression of several structural proteins such as vimentin, collagen-7A1 and the tight junction protein claudin-1. To functionally assess these cells, we also developed a novel human kidney explant model of ATN demonstrating that the cells are more resilient to injury than the surrounding proximal tubular cells. Taken together the results suggest a novel robust cell type with a contrasting biological role to that of the bulk of proximal tubular epithelium.


Subject(s)
Kidney Tubules, Proximal/cytology , Stem Cells/metabolism , Bowman Capsule/cytology , Claudin-1/biosynthesis , Collagen Type VII/biosynthesis , Epithelium/metabolism , Humans , Kidney/cytology , Kidney Tubular Necrosis, Acute/pathology , Kidney Tubules, Proximal/ultrastructure , Vimentin/biosynthesis
9.
J Am Acad Dermatol ; 68(1): 103-12, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23110966

ABSTRACT

BACKGROUND: Laser is one of the main tools for skin resurfacing. Erbium:yttrium-aluminum-garnet (Er:YAG) was the second ablative laser, after carbon dioxide, emitting wavelength of 2940 nm. Fractional laser resurfacing has been developed to overcome the drawbacks of ablative lasers. OBJECTIVE: We aimed to objectively evaluate the histopathological and immunohistochemical effects of Er:YAG 2940-nm laser for facial rejuvenation (multiple sessions of fractional vs single session of ablative Er:YAG laser). METHODS: Facial resurfacing with single-session ablative Er:YAG laser was performed on 6 volunteers. Another 6 were resurfaced using fractional Er:YAG laser (4 sessions). Histopathological (hematoxylin-eosin, orcein, Masson trichrome, and picrosirius red stains) and immunohistochemical assessment for skin biopsy specimens were done before laser resurfacing and after 1 and 6 months. Histometry for epidermal thickness and quantitative assessment for neocollagen formation; collagen I, III, and VII; elastin; and tropoelastin were done for all skin biopsy specimens. RESULTS: Both lasers resulted in increased epidermal thickness. Dermal collagen showed increased neocollagen formation with increased concentration of collagen types I, III, and VII. Dermal elastic tissue studies revealed decreased elastin whereas tropoelastin concentration increased after laser resurfacing. Neither laser showed significant difference between their effects clinically and on dermal collagen. Changes in epidermal thickness, elastin, and tropoelastin were significantly more marked after ablative laser. LIMITATIONS: The small number of patients is a limitation, yet the results show significant improvement. CONCLUSION: Multiple sessions of fractional laser have comparable effects to a single session of ablative Er:YAG laser on dermal collagen but ablative laser has more effect on elastic tissue and epidermis.


Subject(s)
Laser Therapy/methods , Lasers, Solid-State/therapeutic use , Rejuvenation , Skin/anatomy & histology , Skin/metabolism , Adult , Aged , Collagen Type I/biosynthesis , Collagen Type III/biosynthesis , Collagen Type VII/biosynthesis , Dermatologic Surgical Procedures , Elastin/biosynthesis , Erythema/etiology , Female , Humans , Immunohistochemistry , Laser Therapy/adverse effects , Lasers, Solid-State/adverse effects , Male , Middle Aged , Tropoelastin/biosynthesis
10.
Cancer Res ; 72(14): 3522-34, 2012 Jul 15.
Article in English | MEDLINE | ID: mdl-22564523

ABSTRACT

Patients with the genetic skin blistering disease recessive dystrophic epidermolysis bullosa (RDEB) develop aggressive cutaneous squamous cell carcinoma (cSCC). Metastasis leading to mortality is greater in RDEB than in other patient groups with cSCC. Here we investigate the dermal component in RDEB using mRNA expression profiling to compare cultured fibroblasts isolated from individuals without cSCC and directly from tumor matrix in RDEB and non-RDEB samples. Although gene expression of RDEB normal skin fibroblasts resembled that of cancer-associated fibroblasts, RDEB cancer-associated fibroblasts exhibited a distinct and divergent gene expression profile, with a large proportion of the differentially expressed genes involved in matrix and cell adhesion. RDEB cancer-associated fibroblasts conferred increased adhesion and invasion to tumor and nontumor keratinocytes. Reduction of COL7A1, the defective gene in RDEB, in normal dermal fibroblasts led to increased type XII collagen, thrombospondin-1, and Wnt-5A, while reexpression of wild type COL7A1 in RDEB fibroblasts decreased type XII collagen, thrombospondin-1, and Wnt-5A expression, reduced tumor cell invasion in organotypic culture, and restricted tumor growth in vivo. Overall, our findings show that matrix composition in patients with RDEB is a permissive environment for tumor development, and type VII collagen directly regulates the composition of matrix proteins secreted by dermal and cancer-associated fibroblasts.


Subject(s)
Carcinoma, Squamous Cell/genetics , Collagen Type VII/genetics , Epidermolysis Bullosa Dystrophica/genetics , Cell Proliferation , Cells, Cultured , Collagen Type VII/biosynthesis , Epidermolysis Bullosa Dystrophica/pathology , Fibroblasts/metabolism , Gene Expression Profiling , Humans , Neoplasm Invasiveness , Proto-Oncogene Proteins/biosynthesis , RNA, Small Interfering , Skin/cytology , Skin/metabolism , Thrombospondin 1/biosynthesis , Wnt Proteins/biosynthesis , Wnt-5a Protein
12.
J Pathol ; 225(3): 401-13, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21984127

ABSTRACT

Epidermolysis bullosa (EB) is a severe genetic skin fragility syndrome characterized by blister formation. The molecular basis of EB is still largely unknown and wound healing in patients suffering from EB remains a major challenge to their survival. Our previous studies have identified the actin remodelling protein Flightless I (Flii) as an important mediator of wound repair. Here we identify Flii as a novel target involved in skin blistering. Flii expression was significantly elevated in 30 patients with EB, most prominently in patients with recessive dystrophic EB (RDEB) who have defects in production of type VII collagen (ColVII). Using an autoimmune ColVII murine model of EB acquisita (EBA) and an immunocompetent-ColVII-hypomorphic genetic mouse model of RDEB together with murine Flii alleles, we investigated the contribution of Flii to EB. Overexpression of Flii produced severe blistering post-induction of EBA, while decreased Flii reduced blister severity, elevated integrin expression, and improved ColVII production. Flii(+/-) blistered skin showed reduced α-SMA, TGF-ß1, and Smad 2/3 expression, suggesting that decreasing Flii may affect fibrosis. In support of this, Flii-deficient fibroblasts from EBA mice were less able to contract collagen gels in vitro; however, addition of TGF-ß1 restored collagen contraction, suggesting an interplay between Flii and TGF-ß1. Elevated Flii gene and protein expression was further observed in the blisters of ColVII hypomorphic mice, a murine model of RDEB, suggesting that reducing Flii in blistered skin could be a potential new approach for treating patients with EB.


Subject(s)
Autoimmune Diseases/metabolism , Cytoskeletal Proteins/biosynthesis , Epidermolysis Bullosa Acquisita/metabolism , Animals , Autoimmune Diseases/genetics , Autoimmune Diseases/pathology , Carrier Proteins , Cell Adhesion/physiology , Cell Differentiation/physiology , Cell Proliferation , Cells, Cultured , Collagen Type VII/biosynthesis , Cytoskeletal Proteins/genetics , Disease Models, Animal , Epidermolysis Bullosa Acquisita/genetics , Epidermolysis Bullosa Acquisita/pathology , Fibroblasts/pathology , Fibroblasts/physiology , Gene Expression Regulation , Humans , Integrins/metabolism , Mice , Mice, Inbred BALB C , Mice, Transgenic , Microfilament Proteins/biosynthesis , Microfilament Proteins/genetics , Receptors, Cytoplasmic and Nuclear/biosynthesis , Receptors, Cytoplasmic and Nuclear/genetics , Signal Transduction/physiology , Skin/metabolism , Smad Proteins/physiology , Trans-Activators , Transforming Growth Factor beta1/physiology , Wound Healing/physiology
13.
J Pathol ; 224(2): 234-44, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21381035

ABSTRACT

Type VII collagen (COL7) is a major constituent of the cutaneous basement membrane. Loss of tolerance to COL7 leads to the blistering skin disease epidermolysis bullosa acquisita (EBA). Antibodies to COL7 have also been detected in patients with inflammatory bowel disease (IBD), yet reports on the expression of COL7 in the gut are controversial and a pathogenic relevance of anti-COL7 autoantibodies in IBD has not been demonstrated. We therefore characterized the expression patterns of COL7 in murine gastrointestinal organs and investigated if anti-COL7 antibodies induce an inflammatory response in the gut. COL7 expression was analysed on the mRNA and protein levels. Mice were injected with rabbit anti-murine COL7 IgG (passive EBA) or immunized with a fragment of murine COL7 (active EBA). COL7 was found to be expressed in buccal mucosa, oesophagus, stomach, small intestine, and colon. In addition to skin blistering, in both passive and active EBA, autoantibodies bound to the gastrointestinal basement membrane, fixed complement, and led to recruitment of leukocytes. Furthermore, blister formation was observed in the oesophagus (40%/38% of mice in passive/active model), stomach (40%/63%), small intestine (20%/13%), and colon (20%/13%). Compared to control animals, we found a significantly reduced body weight in diseased mice, suggesting that autoantibody-induced gastrointestinal inflammation is clinically relevant. Those observations may help us to understand the co-incidence of IBD with EBA, and vice versa: The inflammatory response in IBD might expose novel antigens (COL7), which leads to the formation of anti-COL7 antibodies. On the contrary, anti-COL7 antibody-induced gastrointestinal inflammation might pave the way for IBD pathogenesis. In summary, our results provide strong evidence that COL7 is expressed in different portions of the gut and that anti-COL7 antibodies induce distinct gastrointestinal tissue damage.


Subject(s)
Autoantibodies/immunology , Colitis/immunology , Epidermolysis Bullosa Acquisita/immunology , Weight Loss/immunology , Animals , Blister/immunology , Blister/pathology , Colitis/etiology , Colitis/pathology , Collagen Type VII/biosynthesis , Collagen Type VII/genetics , Collagen Type VII/immunology , Colon/immunology , Colon/ultrastructure , Complement C3/metabolism , Disease Models, Animal , Epidermolysis Bullosa Acquisita/complications , Esophagus/immunology , Esophagus/ultrastructure , Gene Expression , Immunoglobulin G/metabolism , Leukocytes/immunology , Macrophages/immunology , Mice , Mice, Inbred BALB C , RNA, Messenger/genetics
14.
Biofactors ; 35(5): 435-41, 2009.
Article in English | MEDLINE | ID: mdl-19753652

ABSTRACT

Coenzyme Q10 (CoQ10), which has both energizing and anti-oxidative effects, is also reported to have antiaging action, e.g., reducing the area of facial wrinkles. However, the mechanism of its anti-aging activity is not fully established. Here, we examined the effect of CoQ10 on human dermal and epidermal cells. CoQ10 promoted proliferation of fibroblasts but not keratinocytes. It also accelerated production of basement membrane components, i.e., laminin 332 and type IV and VII collagens, in keratinocytes and fibroblasts, respectively; however, it had no effect on type I collagen production in fibroblasts. CoQ10 also showed protective effects against cell death induced by several reactive oxygen species in keratinocytes, but only when its cellular absorption was enhanced by pretreatment of the cells with highly CoQ10-loaded serum. These results suggest that protection of epidermis against oxidative stress and enhancement of production of epidermal basement membrane components may be involved in the antiaging properties of CoQ10 in skin.


Subject(s)
Basement Membrane/physiology , Cell Death/drug effects , Dermis/metabolism , Epidermis/metabolism , Oxidative Stress/drug effects , Ubiquinone/analogs & derivatives , Amidines/pharmacology , Cell Adhesion Molecules/biosynthesis , Cell Proliferation/drug effects , Collagen Type IV/biosynthesis , Collagen Type VII/biosynthesis , Dermis/cytology , Epidermal Cells , Fibroblasts/cytology , Fibroblasts/drug effects , Humans , Hydrogen Peroxide/pharmacology , Keratinocytes/cytology , Keratinocytes/drug effects , Male , Ubiquinone/physiology , Kalinin
15.
Exp Dermatol ; 16(2): 151-5, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17222230

ABSTRACT

Type VII collagen is the major component of anchoring fibrils in the epidermal basement membrane. Its expression has been analyzed by immunostaining or Northern blotting, but rarely at the protein level. In this study, we have quantitatively examined the effects of ascorbic acid and various cytokines/growth factors on the protein synthesis and secretion of type VII collagen by human dermal fibroblasts in culture, using a developed, highly sensitive sandwich enzyme-linked immunoassay with two kinds of specific monoclonal antibodies against the non-collagenous domain-1. Ascorbic acid and its derivative induced a twofold increase in type VII collagen synthesis, and markedly increased the secretion of type VII collagen into the medium when compared with the control culture. This effect was not influenced by the presence of transforming growth factor-beta1 (TGF-beta1). The synthesis of type VII collagen was elevated by TGF-beta1, platelet-derived growth factor, tumor necrosis factor-alpha, and interleukin-1beta, but not by TGF-alpha. Thus, our data indicate that the synthesis and secretion of type VII collagen in human dermal fibroblasts are regulated by ascorbate and the enhancement of type VII collagen gene expression by cytokines/growth factors is accompanied with elevated production of type VII collagen at the protein level.


Subject(s)
Collagen Type VII/biosynthesis , Enzyme-Linked Immunosorbent Assay/methods , Fibroblasts/metabolism , Skin/metabolism , Adolescent , Adult , Age Factors , Aged , Aged, 80 and over , Ascorbic Acid/physiology , Cells, Cultured , Child , Child, Preschool , Collagen Type VII/analysis , Collagen Type VII/metabolism , Cytokines/physiology , Humans , Infant , Infant, Newborn , Middle Aged , Transforming Growth Factor beta1/physiology , Tumor Necrosis Factor-alpha/physiology
17.
J Invest Dermatol ; 121(5): 1021-8, 2003 Nov.
Article in English | MEDLINE | ID: mdl-14708601

ABSTRACT

Type VII collagen is synthesized and secreted by both human keratinocytes and fibroblasts. Although both cell types can secrete type VII collagen, it is thought that keratinocytes account for type VII collagen at the dermal-epidermal junction (DEJ). In this study, we examined if type VII collagen secreted solely by dermal fibroblasts could be transported to the DEJ. We established organotypic, skin-equivalent cultures composed of keratinocytes from patients with recessive dystrophic epidermolysis bullosa (RDEB) and normal dermal fibroblasts. Immuno-labeling of skin equivalent sections with the anti-type VII collagen antibody revealed tight linear staining at the DEJ. RDEB fibroblasts, were gene-corrected to make type VII collagen and used to regenerate human skin on immune-deficient mice. The human skin generated by gene-corrected RDEB fibroblasts or normal human fibroblasts combined with RDEB keratinocytes restored type VII collagen expression at the DEJ in vivo. Further, intradermal injection of normal human or gene-corrected RDEB fibroblasts into mouse skin resulted in the stable expression of human type VII collagen at the mouse DEJ. These data demonstrate that human dermal fibroblasts alone are capable of producing type VII collagen at the DEJ, and it is possible to restore type VII collagen gene expression in RDEB skin in vivo by direct intradermal injection of fibroblasts.


Subject(s)
Collagen Type VII/biosynthesis , Collagen Type VII/genetics , Epidermolysis Bullosa Dystrophica/genetics , Gene Transfer, Horizontal , Skin/metabolism , Animals , Basement Membrane/metabolism , Cell Line , Epidermolysis Bullosa Dystrophica/metabolism , Epidermolysis Bullosa Dystrophica/therapy , Fibroblasts/metabolism , Genetic Therapy , Humans , Mice , Skin/cytology
18.
Nat Genet ; 32(4): 670-5, 2002 Dec.
Article in English | MEDLINE | ID: mdl-12426566

ABSTRACT

Dystrophic epidermolysis bullosa (DEB) is a family of inherited mechano-bullous disorders caused by mutations in the human type VII collagen gene (COL7A1). Individuals with DEB lack type VII collagen and anchoring fibrils, structures that attach epidermis and dermis. The current lack of treatment for DEB is an impetus to develop gene therapy strategies that efficiently transfer and stably express genes delivered to skin cells in vivo. In this study, we delivered and expressed full-length type VII collagen using a self-inactivating minimal lentivirus-based vector. Transduction of lentiviral vectors containing the COL7A1 transgene into recessive DEB (RDEB) keratinocytes and fibroblasts (in which type VII collagen was absent) resulted in persistent synthesis and secretion of type VII collagen. Unlike RDEB parent cells, the gene-corrected cells had normal morphology, proliferative potential, matrix attachment and motility. We used these gene-corrected cells to regenerate human skin on immune-deficient mice. Human skin regenerated by gene-corrected RDEB cells had restored expression of type VII collagen and formation of anchoring fibrils at the dermal-epidermal junction in vivo. These studies demonstrate that it is possible to restore type VII collagen gene expression in RDEB skin in vivo.


Subject(s)
Collagen Type VII/genetics , Collagen Type VII/physiology , Epidermolysis Bullosa Dystrophica/metabolism , Cell Adhesion , Cell Division , Cell Line , Cell Movement , Cell Transformation, Viral , Cells, Cultured , Collagen Type VII/biosynthesis , DNA, Complementary , Epidermal Cells , Epidermolysis Bullosa Dystrophica/genetics , Epidermolysis Bullosa Dystrophica/therapy , Fibroblasts/metabolism , Fibroblasts/pathology , Fibroblasts/ultrastructure , Gene Transfer Techniques , Genes, Recessive , Genetic Therapy , Genetic Vectors , Humans , Keratinocytes/metabolism , Keratinocytes/pathology , Keratinocytes/ultrastructure , Laminin/metabolism , Lentivirus/genetics , Mutation , Transfection , Transgenes
19.
Hum Gene Ther ; 13(13): 1655-62, 2002 Sep 01.
Article in English | MEDLINE | ID: mdl-12228020

ABSTRACT

Dystrophic epidermolysis bullosa (DEB) comprises a family of inherited blistering skin disorders for which no corrective therapy currently exists. In the most severe form, the Hallopeau-Siemens subtype (RDEB-HS), the epidermal adhesion protein collagen VII is absent from the skin as a consequence of null mutations in the COL7A1 gene. In order to develop an ex vivo gene therapy approach for DEB, we aimed to restore expression of intact procollagen VII in RDEB-HS keratinocytes. The entire human COL7A1 locus in a P1-derived artificial chromosome (PAC) was transferred to RDEB-HS keratinocytes by microinjection, after which sustained biosynthesis and secretion of procollagen VII was detected for 1 year in vitro. Protein chemical analysis demonstrated that the chain composition, domain structure, N-glycosylation and protein folding of the newly produced procollagen VII were similar, if not identical, to its authentic counterpart, indicating that transgenic procollagen VII was structurally normal. These data demonstrate a "proof of principle" for genomic DNA vectors as a means of restoring collagen VII production in RDEB-HS skin and help develop future gene therapy protocols.


Subject(s)
Chromosomes, Artificial, P1 Bacteriophage/genetics , Collagen Type VII/genetics , Epidermolysis Bullosa Dystrophica/therapy , Genetic Vectors , Procollagen/genetics , Collagen Type VII/biosynthesis , Epidermolysis Bullosa Dystrophica/genetics , Epidermolysis Bullosa Dystrophica/metabolism , Genetic Vectors/administration & dosage , Humans , Keratinocytes/metabolism , Microinjections , Procollagen/biosynthesis
SELECTION OF CITATIONS
SEARCH DETAIL