Recessive dystrophic epidermolysis bullosa (RDEB) is caused by loss-of-function mutations in the collagen type VII (C7) gene (COL7A1), and results in a potentially fatal and terribly debilitating disorder. Individuals with severe generalied RDEB develop blisters and erosions involving large portions of the body surface area, mutilating scarring, joint contractures, and strictures of the esophagus, with opportunistic infections and aggressive squamous cell carcinoma as principal causes of early death. We have shown that hematopoietic stem cell transplantation (HSCT) can increase C7 protein in skin and mucous membranes, and ameliorate many of the disease manifestations. Unfortunately not all mucocutaneous lesions heal, and the toxicities associated with HSCT are significant. Our overarching goal is to solve these problems by establishing a reliable method for delivering C7-expressing cells that home to the dermal-epidermal junction and secrete C7 protein. In order to accomplish this goal and to overcome the limitations of allogeneic HSCT, we will: 1) identify the optimal cellular vector and homing signal for targeting injured skin in RDEB, using human mesenchymal stromal/stem cells and induced pluripotent stem cells (iPSCs), 2) define the efficacy of systemic RDEB gene therapy by C7 gene augmentation, and 3) use nucleases for gene editing of individual mutations, as well as whole regions of the C7 gene. We propose to define the conditions conducive to wound healing in this severe blistering genodermatosis, but the impact of these studies may not be limited to RDEB. The function of C7 depends on extracellular polymerization, which makes RDEB an ideal model for understanding the mechanisms of cross- correction of structural protein deficiency in the extracellular matrix. In addition, wound healing integrates skin extracellular matrix and skin cells into a dynamic system, which in RDEB results in the depletion of skin stem cell niches and, in turn, provides a unique model of skin tissue repair with donor regenerative cells. By using powerful tools for studying and manipulating the information basis of biological systems-engineered restriction enzymes (nucleases), skin- targeting, and induced cell lineage conversion (iPSCs)-we will focus on developing personalized cell therapy for individuals with RDEB with the idea that our findings may have broad implications for understanding other connective tissue extracellular matrix diseases. Our proposal is equally motivated by wanting a better understanding of the biological mechanisms in injured skin and by needing to improve the lives of people with RDEB through maximizing the benefits and reducing the risks of potential novel therapies.

Public Health Relevance

More than 3,000 new cases of genetic inborn errors are found in U.S. newborns every year. The complex and life-long medical care for these patients represents a significant challenge to the health care system. Additionally, a deeper understanding of the basic biology of skin blistering disorders and the proof-of-concept for clinical translation in these relatively rare conditions is likely to have a profound impact on muc more common ones, such as chemical and thermal burns.

Agency
National Institute of Health (NIH)
Institute
National Institute of Arthritis and Musculoskeletal and Skin Diseases (NIAMS)
Type
Research Project (R01)
Project #
5R01AR063070-03
Application #
8836974
Study Section
Arthritis, Connective Tissue and Skin Study Section (ACTS)
Program Officer
Baker, Carl
Project Start
2013-04-01
Project End
2016-03-31
Budget Start
2015-04-01
Budget End
2016-03-31
Support Year
3
Fiscal Year
2015
Total Cost
Indirect Cost
Name
University of Minnesota Twin Cities
Department
Pediatrics
Type
Schools of Medicine
DUNS #
555917996
City
Minneapolis
State
MN
Country
United States
Zip Code
55455
Lee, Catherine A A; Seo, Hannah S; Armien, Anibal G et al. (2018) Modeling and rescue of defective blood-brain barrier function of induced brain microvascular endothelial cells from childhood cerebral adrenoleukodystrophy patients. Fluids Barriers CNS 15:9
Vanden Oever, Michael; Twaroski, Kirk; Osborn, Mark J et al. (2018) Inside out: regenerative medicine for recessive dystrophic epidermolysis bullosa. Pediatr Res 83:318-324
Tolar, Jakub; Bauer, Johann W; Kaplan, Daniel H et al. (2018) Montagna Symposium 2017-Precision Dermatology: Next Generation Prevention, Diagnosis, and Treatment. J Invest Dermatol 138:1243-1248
Osborn, Mark J; Lees, Christopher J; McElroy, Amber N et al. (2018) CRISPR/Cas9-Based Cellular Engineering for Targeted Gene Overexpression. Int J Mol Sci 19:
Zhang, Huanan; Lee, Catherine A A; Li, Zhuliu et al. (2018) A multitask clustering approach for single-cell RNA-seq analysis in Recessive Dystrophic Epidermolysis Bullosa. PLoS Comput Biol 14:e1006053
Zhu, Zhijie; Guo, Shuang-Zhuang; Hirdler, Tessa et al. (2018) 3D Printed Functional and Biological Materials on Moving Freeform Surfaces. Adv Mater 30:e1707495
Skvarova Kramarzova, Karolina; Osborn, Mark J; Webber, Beau R et al. (2017) CRISPR/Cas9-Mediated Correction of the FANCD1 Gene in Primary Patient Cells. Int J Mol Sci 18:
Mallhi, K; Orchard, P J; Miller, W P et al. (2017) Non-myeloablative conditioning for second hematopoietic cell transplantation for graft failure in patients with non-malignant disorders: a prospective study and review of the literature. Bone Marrow Transplant 52:726-732
Knipping, Friederike; Osborn, Mark J; Petri, Karl et al. (2017) Genome-wide Specificity of Highly Efficient TALENs and CRISPR/Cas9 for T Cell Receptor Modification. Mol Ther Methods Clin Dev 4:213-224
Boyle, William S; Senger, Kyle; Tolar, Jakub et al. (2017) Heparin Enhances Transfection in Concert with a Trehalose-Based Polycation with Challenging Cell Types. Biomacromolecules 18:56-67

Showing the most recent 10 out of 44 publications