39
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Lentiviral Engineered Fibroblasts Expressing Codon-Optimized COL7A1 Restore Anchoring Fibrils in RDEB

      research-article

      Read this article at

      Bookmark
          There is no author summary for this article yet. Authors can add summaries to their articles on ScienceOpen to make them more accessible to a non-specialist audience.

          Abstract

          Cells therapies, engineered to secrete replacement proteins, are being developed to ameliorate otherwise debilitating diseases. Recessive dystrophic epidermolysis bullosa (RDEB) is caused by defects of type VII collagen, a protein essential for anchoring fibril formation at the dermal-epidermal junction. Whereas allogeneic fibroblasts injected directly into the dermis can mediate transient disease modulation, autologous gene-modified fibroblasts should evade immunological rejection and support sustained delivery of type VII collagen at the dermal-epidermal junction. We demonstrate the feasibility of such an approach using a therapeutic grade, self-inactivating-lentiviral vector, encoding codon-optimized COL7A1, to transduce RDEB fibroblasts under conditions suitable for clinical application. Expression and secretion of type VII collagen was confirmed with transduced cells exhibiting supranormal levels of protein expression, and ex vivo migration of fibroblasts was restored in functional assays. Gene-modified RDEB fibroblasts also deposited type VII collagen at the dermal-epidermal junction of human RDEB skin xenografts placed on NOD- scid IL2Rgamma null recipients, with reconstruction of human epidermal structure and regeneration of anchoring fibrils at the dermal-epidermal junction. Fibroblast-mediated restoration of protein and structural defects in this RDEB model strongly supports proposed therapeutic applications in man.

          Related collections

          Most cited references40

          • Record: found
          • Abstract: found
          • Article: not found

          Inherited epidermolysis bullosa: updated recommendations on diagnosis and classification.

          Several new targeted genes and clinical subtypes have been identified since publication in 2008 of the report of the last international consensus meeting on diagnosis and classification of epidermolysis bullosa (EB). As a correlate, new clinical manifestations have been seen in several subtypes previously described. We sought to arrive at an updated consensus on the classification of EB subtypes, based on newer data, both clinical and molecular. In this latest consensus report, we introduce a new approach to classification ("onion skinning") that takes into account sequentially the major EB type present (based on identification of the level of skin cleavage), phenotypic characteristics (distribution and severity of disease activity; specific extracutaneous features; other), mode of inheritance, targeted protein and its relative expression in skin, gene involved and type(s) of mutation present, and--when possible--specific mutation(s) and their location(s). This classification scheme critically takes into account all published data through June 2013. Further modifications are likely in the future, as more is learned about this group of diseases. The proposed classification scheme should be of value both to clinicians and researchers, emphasizing both clinical and molecular features of each EB subtype, and has sufficient flexibility incorporated in its structure to permit further modifications in the future. Copyright © 2014 American Academy of Dermatology, Inc. Published by Mosby, Inc. All rights reserved.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found
            Is Open Access

            TALEN-based gene correction for epidermolysis bullosa.

            Recessive dystrophic epidermolysis bullosa (RDEB) is characterized by a functional deficit of type VII collagen protein due to gene defects in the type VII collagen gene (COL7A1). Gene augmentation therapies are promising, but run the risk of insertional mutagenesis. To abrogate this risk, we explored the possibility of using engineered transcription activator-like effector nucleases (TALEN) for precise genome editing. We report the ability of TALEN to induce site-specific double-stranded DNA breaks (DSBs) leading to homology-directed repair (HDR) from an exogenous donor template. This process resulted in COL7A1 gene mutation correction in primary fibroblasts that were subsequently reprogrammed into inducible pluripotent stem cells and showed normal protein expression and deposition in a teratoma-based skin model in vivo. Deep sequencing-based genome-wide screening established a safety profile showing on-target activity and three off-target (OT) loci that, importantly, were at least 10 kb from a coding sequence. This study provides proof-of-concept for TALEN-mediated in situ correction of an endogenous patient-specific gene mutation and used an unbiased screen for comprehensive TALEN target mapping that will cooperatively facilitate translational application.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Bone marrow transplantation for recessive dystrophic epidermolysis bullosa.

              Recessive dystrophic epidermolysis bullosa is an incurable, often fatal mucocutaneous blistering disease caused by mutations in COL7A1, the gene encoding type VII collagen (C7). On the basis of preclinical data showing biochemical correction and prolonged survival in col7 −/− mice, we hypothesized that allogeneic marrow contains stem cells capable of ameliorating the manifestations of recessive dystrophic epidermolysis bullosa in humans. Between October 2007 and August 2009, we treated seven children who had recessive dystrophic epidermolysis bullosa with immunomyeloablative chemotherapy and allogeneic stem-cell transplantation. We assessed C7 expression by means of immunofluorescence staining and used transmission electron microscopy to visualize anchoring fibrils. We measured chimerism by means of competitive polymerase-chain-reaction assay, and documented blister formation and wound healing with the use of digital photography. One patient died of cardiomyopathy before transplantation. Of the remaining six patients, one had severe regimen-related cutaneous toxicity, with all having improved wound healing and a reduction in blister formation between 30 and 130 days after transplantation. We observed increased C7 deposition at the dermal-epidermal junction in five of the six recipients, albeit without normalization of anchoring fibrils. Five recipients were alive 130 to 799 days after transplantation; one died at 183 days as a consequence of graft rejection and infection. The six recipients had substantial proportions of donor cells in the skin, and none had detectable anti-C7 antibodies. Increased C7 deposition and a sustained presence of donor cells were found in the skin of children with recessive dystrophic epidermolysis bullosa after allogeneic bone marrow transplantation. Further studies are needed to assess the long-term risks and benefits of such therapy in patients with this disorder. (Funded by the National Institutes of Health; ClinicalTrials.gov number, NCT00478244.)
                Bookmark

                Author and article information

                Contributors
                Journal
                J Invest Dermatol
                J. Invest. Dermatol
                The Journal of Investigative Dermatology
                Elsevier
                0022-202X
                1523-1747
                1 January 2016
                January 2016
                : 136
                : 1
                : 284-292
                Affiliations
                [1 ]UCL Institute of Child Health, Molecular and Cellular Immunology Section & Great Ormond Street Hospital NHS Foundation Trust, London, United Kingdom
                [2 ]St John’s Institute of Dermatology, King’s College London (Guy’s campus), London, United Kingdom
                [3 ]Centre for Ultrastructural Imaging, King’s College London, London, United Kingdom
                [4 ]Department of Haematological Medicine, King’s College London, The Rayne Institute, London, United Kingdom
                [5 ]The Robin Eady National Diagnostic Epidermolysis Bullosa Laboratory, Viapath LLP, St Thomas’ Hospital, London, United Kingdom
                [6 ]Department of Dermatology, University of Southern California, Los Angeles, California, USA
                Author notes
                []Correspondence: Waseem Qasim, Reader in Cell & Gene Therapy, UCL Institute of Child Health & Great Ormond Street Hospital NHS Foundation Trust, 30 Guilford Street, London WC1N 1EH, United Kingdom.Reader in Cell & Gene TherapyUCL Institute of Child Health & Great Ormond Street Hospital NHS Foundation Trust30 Guilford StreetLondon WC1N 1EHUnited Kingdom W.Qasim@ 123456ucl.ac.uk
                [7]

                These authors have contributed equally to this work.

                [8]

                These authors have contributed equally to this work.

                Article
                S0022-202X(15)00027-5
                10.1038/JID.2015.364
                4759620
                26763448
                fc3180cb-870b-46a3-98a7-efddec0411e1
                © 2015 The Authors

                This is an open access article under the CC BY license (http://creativecommons.org/licenses/by/4.0/).

                History
                : 7 May 2015
                : 27 July 2015
                : 3 August 2015
                Categories
                Original Article
                Wound Healing

                Dermatology
                af, anchoring fibril,dej, dermalepidermal junction,lv, lentiviral,rdeb, recessive dystrophic epidermolysis bullosa,c7, type vii collagen

                Comments

                Comment on this article