1
|
Morren MA, Legius E, Giuliano F, Hadj-Rabia S, Hohl D, Bodemer C. Challenges in Treating Genodermatoses: New Therapies at the Horizon. Front Pharmacol 2022; 12:746664. [PMID: 35069188 PMCID: PMC8766835 DOI: 10.3389/fphar.2021.746664] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2021] [Accepted: 11/29/2021] [Indexed: 01/28/2023] Open
Abstract
Genodermatoses are rare inherited skin diseases that frequently affect other organs. They often have marked effects on wellbeing and may cause early death. Progress in molecular genetics and translational research has unravelled many underlying pathological mechanisms, and in several disorders with high unmet need, has opened the way for the introduction of innovative treatments. One approach is to intervene where cell-signaling pathways are dysregulated, in the case of overactive pathways by the use of selective inhibitors, or when the activity of an essential factor is decreased by augmenting a molecular component to correct disequilibrium in the pathway. Where inflammatory reactions have been induced by a genetically altered protein, another possible approach is to suppress the inflammation directly. Depending on the nature of the genodermatosis, the implicated protein or even on the particular mutation, to correct the consequences or the genetic defect, may require a highly personalised stratagem. Repurposed drugs, can be used to bring about a "read through" strategy especially where the genetic defect induces premature termination codons. Sometimes the defective protein can be replaced by a normal functioning one. Cell therapies with allogeneic normal keratinocytes or fibroblasts may restore the integrity of diseased skin and allogeneic bone marrow or mesenchymal cells may additionally rescue other affected organs. Genetic engineering is expanding rapidly. The insertion of a normal functioning gene into cells of the recipient is since long explored. More recently, genome editing, allows reframing, insertion or deletion of exons or disruption of aberrantly functioning genes. There are now several examples where these stratagems are being explored in the (pre)clinical phase of therapeutic trial programmes. Another stratagem, designed to reduce the severity of a given disease involves the use of RNAi to attenuate expression of a harmful protein by decreasing abundance of the cognate transcript. Most of these strategies are short-lasting and will thus require intermittent life-long administration. In contrast, insertion of healthy copies of the relevant gene or editing the disease locus in the genome to correct harmful mutations in stem cells is more likely to induce a permanent cure. Here we discuss the potential advantages and drawbacks of applying these technologies in patients with these genetic conditions. Given the severity of many genodermatoses, prevention of transmission to future generations remains an important goal including offering reproductive choices, such as preimplantation genetic testing, which can allow selection of an unaffected embryo for transfer to the uterus.
Collapse
Affiliation(s)
- Marie-Anne Morren
- Pediatric Dermatology Unit, Departments of Dermatology and Venereology and Pediatrics, University Hospital Lausanne, University of Lausanne, Lausanne, Switzerland
| | - Eric Legius
- Department for Human Genetics, University Hospitals Leuven, KU Leuven, ERN Genturis and ERN Skin, Leuven, Belgium
| | - Fabienne Giuliano
- Department of Medical Genetics, University Hospital Lausanne, Lausanne, Switzerland
| | - Smail Hadj-Rabia
- Department of Pediatric Dermatology and Dermatology, National Reference Centre for Genodermatosis and Rare Diseases of the Skin (MAGEC), Hôpital Necker-Enfants Malades, and Assistance Publique-Hôpitaux de Paris, Université Paris Descartes, ERN Skin, Paris, France
| | - Daniel Hohl
- Department of Dermatology and Venereology, University Hospital Lausanne, University of Lausanne, Lausanne, Switzerland
| | - Christine Bodemer
- Department of Pediatric Dermatology and Dermatology, National Reference Centre for Genodermatosis and Rare Diseases of the Skin (MAGEC), Hôpital Necker-Enfants Malades, and Assistance Publique-Hôpitaux de Paris, Université Paris Descartes, ERN Skin, Paris, France
| |
Collapse
|
2
|
Izmiryan A, Ganier C, Bovolenta M, Schmitt A, Mavilio F, Hovnanian A. Ex Vivo COL7A1 Correction for Recessive Dystrophic Epidermolysis Bullosa Using CRISPR/Cas9 and Homology-Directed Repair. MOLECULAR THERAPY-NUCLEIC ACIDS 2018; 12:554-567. [PMID: 30195791 PMCID: PMC6077132 DOI: 10.1016/j.omtn.2018.06.008] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/02/2018] [Revised: 06/04/2018] [Accepted: 06/22/2018] [Indexed: 02/06/2023]
Abstract
Recessive dystrophic epidermolysis bullosa is a rare and severe genetic skin disease resulting in blistering of the skin and mucosa. Recessive dystrophic epidermolysis bullosa (RDEB) is caused by a wide variety of mutations in COL7A1-encoding type VII collagen, which is essential for dermal-epidermal adhesion. Here we demonstrate the feasibility of ex vivo COL7A1 editing in primary RDEB cells and in grafted 3D skin equivalents through CRISPR/Cas9-mediated homology-directed repair. We designed five guide RNAs to correct a RDEB causative null mutation in exon 2 (c.189delG; p.Leu64Trpfs*40). Among the site-specific guide RNAs tested, one showed significant cleavage activity in primary RDEB keratinocytes and in fibroblasts when delivered as integration-deficient lentivirus. Genetic correction was detected in transduced keratinocytes and fibroblasts by allele-specific highly sensitive TaqMan-droplet digital PCR (ddPCR), resulting in 11% and 15.7% of corrected COL7A1 mRNA expression, respectively, without antibiotic selection. Grafting of genetically corrected 3D skin equivalents onto nude mice showed up to 26% re-expression and normal localization of type VII collagen as well as anchoring fibril formation at the dermal-epidermal junction. Our study provides evidence that precise genome editing in primary RDEB cells is a relevant strategy to genetically correct COL7A1 mutations for the development of future ex vivo clinical applications.
Collapse
Affiliation(s)
- Araksya Izmiryan
- Laboratory of Genetic Skin Diseases, INSERM UMR 1163, Imagine Institute, 24 bd du Montparnasse, Paris, France; University Paris Descartes-Sorbonne Cité, Paris, France
| | - Clarisse Ganier
- Laboratory of Genetic Skin Diseases, INSERM UMR 1163, Imagine Institute, 24 bd du Montparnasse, Paris, France; University Paris Descartes-Sorbonne Cité, Paris, France
| | | | - Alain Schmitt
- Electronic Microscopy Facility, INSERM UMR 1016, Cochin Institute, Paris, France
| | - Fulvio Mavilio
- University Paris Descartes-Sorbonne Cité, Paris, France; Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy; Imagine Institute, Paris, France
| | - Alain Hovnanian
- Laboratory of Genetic Skin Diseases, INSERM UMR 1163, Imagine Institute, 24 bd du Montparnasse, Paris, France; University Paris Descartes-Sorbonne Cité, Paris, France; Department of Genetics, Necker Hospital for Sick Children, APHP, Paris, France.
| |
Collapse
|
3
|
Webber BR, O’Connor KT, McElmurry RT, Durgin EN, Eide C, Lees CJ, Riddle MJ, Mathews W, Frank NY, Kluth MA, Ganss C, Moriarity BS, Frank MH, Osborn MJ, Tolar J. Rapid generation of Col7a1 -/- mouse model of recessive dystrophic epidermolysis bullosa and partial rescue via immunosuppressive dermal mesenchymal stem cells. J Transl Med 2017; 97:1218-1224. [PMID: 28892093 PMCID: PMC5623156 DOI: 10.1038/labinvest.2017.85] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2017] [Revised: 07/06/2017] [Accepted: 07/10/2017] [Indexed: 12/20/2022] Open
Abstract
Recessive dystrophic epidermolysis bullosa (RDEB) is a debilitating and ultimately lethal blistering disease caused by mutations to the Col7a1 gene. Development of novel cell therapies for the treatment of RDEB would be fostered by having immunodeficient mouse models able to accept human cell grafts; however, immunodeficient models of many genodermatoses such as RDEB are lacking. To overcome this limitation, we combined the clustered regularly interspaced short palindromic repeats and associated nuclease (CRISPR/Cas9) system with microinjection into NOD/SCID IL2rγcnull (NSG) embryos to rapidly develop an immunodeficient Col7a1-/- mouse model of RDEB. Through dose optimization, we achieve F0 biallelic knockout efficiencies exceeding 80%, allowing us to quickly generate large numbers of RDEB NSG mice for experimental use. Using this strategy, we clearly demonstrate important strain-specific differences in RDEB pathology that could underlie discordant results observed between independent studies and establish the utility of this system in proof-of-concept human cellular transplantation experiments. Importantly, we uncover the ability of a recently identified skin resident immunomodulatory dermal mesenchymal stem cell marked by ABCB5 to reduce RDEB pathology and markedly extend the lifespan of RDEB NSG mice via reduced skin infiltration of inflammatory myeloid derivatives.
Collapse
Affiliation(s)
- Beau R. Webber
- Department of Pediatrics, Division of Blood and Marrow Transplantation, University of Minnesota Medical School, Minneapolis, Minnesota, USA
| | - Kyle T. O’Connor
- Masonic Cancer Center at the University of Minnesota, Mouse Genetics Laboratory Shared Resource, University of Minnesota, Minneapolis, Minnesota, USA
| | - Ron T. McElmurry
- Department of Pediatrics, Division of Blood and Marrow Transplantation, University of Minnesota Medical School, Minneapolis, Minnesota, USA
| | - Elise N. Durgin
- Department of Pediatrics, Division of Blood and Marrow Transplantation, University of Minnesota Medical School, Minneapolis, Minnesota, USA
| | - Cindy Eide
- Department of Pediatrics, Division of Blood and Marrow Transplantation, University of Minnesota Medical School, Minneapolis, Minnesota, USA
| | - Christopher J. Lees
- Department of Pediatrics, Division of Blood and Marrow Transplantation, University of Minnesota Medical School, Minneapolis, Minnesota, USA
| | - Megan J. Riddle
- Department of Pediatrics, Division of Blood and Marrow Transplantation, University of Minnesota Medical School, Minneapolis, Minnesota, USA
| | - Wendy Mathews
- Department of Pediatrics, Division of Blood and Marrow Transplantation, University of Minnesota Medical School, Minneapolis, Minnesota, USA
| | - Natasha Y. Frank
- Department of Medicine, Boston VA Healthcare System, West Roxbury, Massachusetts, USA,Division of Genetics, Brigham and Women’s Hospital, Boston, Massachusetts, USA
| | - Mark A. Kluth
- Rheacell GmbH & Co. KG, Heidelberg, Germany,Ticeba GmbH, Heidelberg, Germany
| | - Christoph Ganss
- Rheacell GmbH & Co. KG, Heidelberg, Germany,Ticeba GmbH, Heidelberg, Germany
| | - Branden S. Moriarity
- Department of Pediatrics, Division of Blood and Marrow Transplantation, University of Minnesota Medical School, Minneapolis, Minnesota, USA,Center for Genome Engineering, University of Minnesota, Minneapolis, Minnesota, USA
| | - Markus H. Frank
- Transplant Research Program, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA,Harvard Skin Disease Research Center, Department of Dermatology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA,School of Medical Sciences, Edith Cowan University, Joondalup, Western Australia, Australia,Harvard Stem Cell Institute, Harvard University, Cambridge, Massachusetts, USA
| | - Mark J. Osborn
- Department of Pediatrics, Division of Blood and Marrow Transplantation, University of Minnesota Medical School, Minneapolis, Minnesota, USA,Center for Genome Engineering, University of Minnesota, Minneapolis, Minnesota, USA,Stem Cell Institute, University of Minnesota, Minneapolis, Minnesota, USA,Asan-Minnesota Institute for Innovating Transplantation, Seoul, Republic of Korea
| | - Jakub Tolar
- Department of Pediatrics, Division of Blood and Marrow Transplantation, University of Minnesota Medical School, Minneapolis, Minnesota, USA,Center for Genome Engineering, University of Minnesota, Minneapolis, Minnesota, USA,Stem Cell Institute, University of Minnesota, Minneapolis, Minnesota, USA,Asan-Minnesota Institute for Innovating Transplantation, Seoul, Republic of Korea,Correspondence to: Jakub Tolar, Pediatric BMT, 420 Delaware St SE, MMC 366, Minneapolis, MN 55455; 612-626-6723;
| |
Collapse
|
4
|
Xu L, Carrer A, Zonta F, Qu Z, Ma P, Li S, Ceriani F, Buratto D, Crispino G, Zorzi V, Ziraldo G, Bruno F, Nardin C, Peres C, Mazzarda F, Salvatore AM, Raspa M, Scavizzi F, Chu Y, Xie S, Yang X, Liao J, Liu X, Wang W, Wang S, Yang G, Lerner RA, Mammano F. Design and Characterization of a Human Monoclonal Antibody that Modulates Mutant Connexin 26 Hemichannels Implicated in Deafness and Skin Disorders. Front Mol Neurosci 2017; 10:298. [PMID: 29018324 PMCID: PMC5615210 DOI: 10.3389/fnmol.2017.00298] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2017] [Accepted: 09/05/2017] [Indexed: 12/21/2022] Open
Abstract
Background: Mutations leading to changes in properties, regulation, or expression of connexin-made channels have been implicated in 28 distinct human hereditary diseases. Eight of these result from variants of connexin 26 (Cx26), a protein critically involved in cell-cell signaling in the inner ear and skin. Lack of non-toxic drugs with defined mechanisms of action poses a serious obstacle to therapeutic interventions for diseases caused by mutant connexins. In particular, molecules that specifically modulate connexin hemichannel function without affecting gap junction channels are considered of primary importance for the study of connexin hemichannel role in physiological as well as pathological conditions. Monoclonal antibodies developed in the last three decades have become the most important class of therapeutic biologicals. Recombinant methods permit rapid selection and improvement of monoclonal antibodies from libraries with large diversity. Methods: By screening a combinatorial library of human single-chain fragment variable (scFv) antibodies expressed in phage, we identified a candidate that binds an extracellular epitope of Cx26. We characterized antibody action using a variety of biochemical and biophysical assays in HeLa cells, organotypic cultures of mouse cochlea and human keratinocyte-derived cells. Results: We determined that the antibody is a remarkably efficient, non-toxic, and completely reversible inhibitor of hemichannels formed by connexin 26 and does not affect direct cell-cell communication via gap junction channels. Importantly, we also demonstrate that the antibody efficiently inhibits hyperative mutant Cx26 hemichannels implicated in autosomal dominant non-syndromic hearing impairment accompanied by keratitis and hystrix-like ichthyosis-deafness (KID/HID) syndrome. We solved the crystal structure of the antibody, identified residues that are critical for binding and used molecular dynamics to uncover its mechanism of action. Conclusions: Although further studies will be necessary to validate the effect of the antibody in vivo, the methodology described here can be extended to select antibodies against hemichannels composed by other connexin isoforms and, consequently, to target other pathologies associated with hyperactive hemichannels. Our study highlights the potential of this approach and identifies connexins as therapeutic targets addressable by screening phage display libraries expressing human randomized antibodies.
Collapse
Affiliation(s)
- Liang Xu
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech UniversityShanghai, China.,Institute of Biochemistry and Cell Biology, Shanghai Institute for Biological Sciences, Chinese Academy of SciencesShanghai, China
| | - Andrea Carrer
- CNR Institute of Cell Biology and NeurobiologyMonterotondo, Italy.,Department of Physics and Astronomy "G. Galilei,", University of PadovaPadova, Italy
| | - Francesco Zonta
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech UniversityShanghai, China.,CNR Institute of Cell Biology and NeurobiologyMonterotondo, Italy
| | - Zhihu Qu
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech UniversityShanghai, China
| | - Peixiang Ma
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech UniversityShanghai, China
| | - Sheng Li
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech UniversityShanghai, China
| | - Federico Ceriani
- CNR Institute of Cell Biology and NeurobiologyMonterotondo, Italy.,Department of Physics and Astronomy "G. Galilei,", University of PadovaPadova, Italy
| | - Damiano Buratto
- CNR Institute of Cell Biology and NeurobiologyMonterotondo, Italy.,Department of Physics and Astronomy "G. Galilei,", University of PadovaPadova, Italy
| | - Giulia Crispino
- CNR Institute of Cell Biology and NeurobiologyMonterotondo, Italy.,Department of Physics and Astronomy "G. Galilei,", University of PadovaPadova, Italy.,Venetian Institute of Molecular MedicinePadova, Italy
| | - Veronica Zorzi
- CNR Institute of Cell Biology and NeurobiologyMonterotondo, Italy.,Institute of Otolaryngology, Catholic University School of MedicineRome, Italy
| | - Gaia Ziraldo
- CNR Institute of Cell Biology and NeurobiologyMonterotondo, Italy.,Department of Physics and Astronomy "G. Galilei,", University of PadovaPadova, Italy.,Institute of Otolaryngology, Catholic University School of MedicineRome, Italy
| | - Francesca Bruno
- Department of Physics and Astronomy "G. Galilei,", University of PadovaPadova, Italy.,Venetian Institute of Molecular MedicinePadova, Italy
| | - Chiara Nardin
- CNR Institute of Cell Biology and NeurobiologyMonterotondo, Italy.,Department of Science, Roma Tre UniversityRome, Italy
| | - Chiara Peres
- CNR Institute of Cell Biology and NeurobiologyMonterotondo, Italy
| | - Flavia Mazzarda
- CNR Institute of Cell Biology and NeurobiologyMonterotondo, Italy.,Department of Science, Roma Tre UniversityRome, Italy
| | - Anna M Salvatore
- CNR Institute of Cell Biology and NeurobiologyMonterotondo, Italy
| | - Marcello Raspa
- CNR Institute of Cell Biology and NeurobiologyMonterotondo, Italy
| | | | - Youjun Chu
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech UniversityShanghai, China
| | - Sichun Xie
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech UniversityShanghai, China
| | - Xuemei Yang
- School of Life Science and Technology, Shanghai Tech UniversityShanghai, China
| | - Jun Liao
- School of Life Science and Technology, Shanghai Tech UniversityShanghai, China
| | - Xiao Liu
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech UniversityShanghai, China.,Institute of Biochemistry and Cell Biology, Shanghai Institute for Biological Sciences, Chinese Academy of SciencesShanghai, China.,University of Chinese Academy of SciencesBeijing, China
| | - Wei Wang
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech UniversityShanghai, China
| | - Shanshan Wang
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech UniversityShanghai, China
| | - Guang Yang
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech UniversityShanghai, China
| | - Richard A Lerner
- Department of Cell and Molecular Biology, The Scripps Research InstituteLa Jolla, CA, United States
| | - Fabio Mammano
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech UniversityShanghai, China.,CNR Institute of Cell Biology and NeurobiologyMonterotondo, Italy.,Department of Physics and Astronomy "G. Galilei,", University of PadovaPadova, Italy.,Venetian Institute of Molecular MedicinePadova, Italy
| |
Collapse
|
5
|
Higgins CA, Roger MF, Hill RP, Ali-Khan AS, Garlick JA, Christiano AM, Jahoda CAB. Multifaceted role of hair follicle dermal cells in bioengineered skins. Br J Dermatol 2017; 176:1259-1269. [PMID: 27679975 DOI: 10.1111/bjd.15087] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/21/2016] [Indexed: 01/16/2023]
Abstract
BACKGROUND The method of generating bioengineered skin constructs was pioneered several decades ago; nowadays these constructs are used regularly for the treatment of severe burns and nonhealing wounds. Commonly, these constructs are comprised of skin fibroblasts within a collagen scaffold, forming the skin dermis, and stratified keratinocytes overlying this, forming the skin epidermis. In the past decade there has been a surge of interest in bioengineered skins, with researchers seeking alternative cell sources, or scaffolds, from which constructs can be established, and for more biomimetic equivalents with skin appendages. OBJECTIVES To evaluate whether human hair follicle dermal cells can act as an alternative cell source for engineering the dermal component of engineered skin constructs. METHODS We established in vitro skin constructs by incorporating into the collagenous dermal compartment: (i) primary interfollicular dermal fibroblasts, (ii) hair follicle dermal papilla cells or (iii) hair follicle dermal sheath cells. In vivo skins were established by mixing dermal cells and keratinocytes in chambers on top of immunologically compromised mice. RESULTS All fibroblast subtypes were capable of supporting growth of overlying epithelial cells, both in vitro and in vivo. However, we found hair follicle dermal sheath cells to be superior to fibroblasts in their capacity to influence the establishment of a basal lamina. CONCLUSIONS Human hair follicle dermal cells can be readily interchanged with interfollicular fibroblasts and used as an alternative cell source for establishing the dermal component of engineered skin both in vitro and in vivo.
Collapse
Affiliation(s)
- C A Higgins
- Department of Dermatology, Columbia University, New York, NY, U.S.A.,Department of Bioengineering, Imperial College London, London, U.K
| | - M F Roger
- School of Biological and Biomedical Sciences, Durham University, Durham, U.K
| | - R P Hill
- School of Biological and Biomedical Sciences, Durham University, Durham, U.K
| | - A S Ali-Khan
- Department of Plastic Surgery, University Hospital of Durham, Durham, U.K
| | - J A Garlick
- Sackler Graduate School of Biomedical Sciences, Tufts University, Boston, MA, U.S.A
| | - A M Christiano
- Department of Dermatology, Columbia University, New York, NY, U.S.A.,Department of Genetics and Development, Columbia University, New York, NY, U.S.A
| | - C A B Jahoda
- School of Biological and Biomedical Sciences, Durham University, Durham, U.K
| |
Collapse
|
6
|
Vahidnezhad H, Youssefian L, Zeinali S, Saeidian AH, Sotoudeh S, Mozafari N, Abiri M, Kajbafzadeh AM, Barzegar M, Ertel A, Fortina P, Uitto J. Dystrophic Epidermolysis Bullosa: COL7A1 Mutation Landscape in a Multi-Ethnic Cohort of 152 Extended Families with High Degree of Customary Consanguineous Marriages. J Invest Dermatol 2017; 137:660-669. [DOI: 10.1016/j.jid.2016.10.023] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2016] [Revised: 10/17/2016] [Accepted: 10/18/2016] [Indexed: 12/11/2022]
|
7
|
Storan ER, Irvine AD. Access to Genetic Diagnostics for Genodermatoses: Who Should Get Tested? Why? Who Pays? Pediatr Dermatol 2017; 34:105-108. [PMID: 27981618 DOI: 10.1111/pde.13041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Eoin R Storan
- Paediatric Dermatology, Our Lady's Children's Hospital Crumlin, Dublin, Ireland
| | - Alan D Irvine
- Paediatric Dermatology, Our Lady's Children's Hospital Crumlin, Dublin, Ireland.,Clinical Medicine, Trinity College, Dublin, Ireland.,National Children's Research Centre, Our Lady's Children's Hospital, Crumlin, Dublin, Ireland
| |
Collapse
|
8
|
Shagalov DR, Ferzli GM, Wildman T, Glick SA. Genetic Testing in Dermatology: A Survey Analyzing Obstacles to Appropriate Care. Pediatr Dermatol 2017; 34:33-38. [PMID: 27653748 DOI: 10.1111/pde.12981] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
BACKGROUND/OBJECTIVES The past several decades have witnessed unprecedented advances in genomic technology, bringing genetic testing to the forefront of medical practice and moving us towards the practice of personalized medicine. Genetic testing has become an important aspect in preempting and successfully treating diseases in dermatology, yet difficulty remains in regards to obtaining genetic testing for patients. We conducted a survey for pediatric dermatologists in order to try to gauge and understand where difficulties lie in obtaining genetic testing and to analyze how best these issues can be resolved. METHODS An 18-question survey was emailed to 480 dermatologists who have attended at least one of the last three annual Society for Pediatric Dermatology (SPD) meetings. RESULTS Virtually all providers encountered at least one situation in which they required genetic testing for a patient (97.3% [n = 108]) and 37.4% indicated needing genetic testing more than six times per year. Of the respondents who had attempted to obtain genetic testing, half were unsuccessful in obtaining coverage more than 75% of the time (45% [n = 32]) and only 7.0% (n = 5) achieved success 75% to 100% of the time. The most common reasons for obtaining genetic testing included the need to provide an accurate diagnosis, followed by the need to provide prognostic information and appropriate medical management. CONCLUSION The role of genetic testing in the practice of dermatology is expanding, yet obtaining coverage for genetic testing remains a challenge. We propose several solutions as to how this can be remedied.
Collapse
Affiliation(s)
- Devorah R Shagalov
- Dermatology, State University of New York Downstate Medical Center, Brooklyn, New York
| | - Georgina M Ferzli
- Dermatology, State University of New York Downstate Medical Center, Brooklyn, New York
| | | | - Sharon A Glick
- Dermatology, State University of New York Downstate Medical Center, Brooklyn, New York
| |
Collapse
|
9
|
Kim KH, Blasco-Morente G, Cuende N, Arias-Santiago S. Mesenchymal stromal cells: properties and role in management of cutaneous diseases. J Eur Acad Dermatol Venereol 2016; 31:414-423. [PMID: 27549663 DOI: 10.1111/jdv.13934] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2016] [Accepted: 07/28/2016] [Indexed: 12/18/2022]
Abstract
This review describes the current understanding and the potential use of mesenchymal stromal cells (MSCs) in cell-based therapies for clinical management of difficult wounds and other dermatoses. MSCs have been shown to possess many advantageous properties that make them a promising therapeutic modality in dermatology still under investigation. In fact, MSCs' ability to promote wound healing through its paracrine function and pro-angiogenic properties have generated increasing interest for treating acute and chronic wounds. There is also great interest in utilizing MSCs' immunological characteristics for therapeutic use especially for patients with debilitating systemic autoimmune and inflammatory skin conditions who have failed other therapies. Its role in aesthetics has also been explored with clinical data showing improvement of acne scars and wrinkles from photoaging. Clinical trials are underway investigating the safety and efficacy of MSCs in the treatment of different skin conditions such as acute burns, diabetic and venous stasis ulcers, epidermolysis bullosa and systemic sclerosis, among others. We anticipate that as our understanding of the characteristics and function of MSCs grow, so will its role in cell-based treatments of dermatological conditions.
Collapse
Affiliation(s)
- K H Kim
- Department of Dermatology, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - G Blasco-Morente
- Department of Dermatology, Virgen de las Nieves University Hospital, Granada, Spain
| | - N Cuende
- Andalusian Initiative for Advanced Therapies, Seville, Spain
| | - S Arias-Santiago
- Department of Dermatology, Virgen de las Nieves University Hospital, Granada, Spain.,Cell Production and Tissue Engineering Unit, Virgen de las Nieves University Hospital, Granada, Spain
| |
Collapse
|
10
|
Pourreyron C, Chen M, McGrath JA, Salas-Alanis JC, South AP, Leigh IM. High levels of type VII collagen expression in recessive dystrophic epidermolysis bullosa cutaneous squamous cell carcinoma keratinocytes increases PI3K and MAPK signalling, cell migration and invasion. Br J Dermatol 2016; 170:1256-65. [PMID: 24641191 DOI: 10.1111/bjd.12715] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/26/2013] [Indexed: 12/20/2022]
Abstract
BACKGROUND Epidermolysis bullosa is a group of inherited skin fragility diseases varying in severity from mild scarring to infant mortality. Great efforts are being undertaken to develop therapeutic strategies to treat the more pernicious forms of this disease, particularly those associated with recessive, loss-of-function mutations. In such cases significant effort is directed toward delivering recombinant protein at levels sufficient to demonstrate clinical benefit. Recessive dystrophic epidermolysis bullosa (RDEB) predisposes patients to a high incidence of life-threatening cutaneous squamous cell carcinoma (cSCC). Mutations in the gene encoding type VII collagen, COL7A1, are the sole cause of this disease and conflicting reports concerning type VII collagen and COL7A1 in carcinogenesis exist. OBJECTIVES To investigate potential oncogenic effects of expressing recombinant type VII collagen in patient cells. METHODS We used retroviral transduction to introduce type VII collagen into keratinocytes derived from patients with and without RDEB. RESULTS Retroviral expression of type VII collagen in cSCC keratinocytes established from patients with RDEB resulted in increased cell adhesion, migration and invasion coupled with a concurrent increase in PI3K and MAPK signalling. CONCLUSIONS Our data suggest caution when formulating strategies where delivery of type VII collagen is likely to exceed levels seen under normal physiological conditions in a patient group with a higher inherent risk of developing skin cancer.
Collapse
Affiliation(s)
- C Pourreyron
- Division of Cancer Research, Ninewells Hospital and Medical School, University of Dundee, Dundee, DD1 9SY, U.K
| | | | | | | | | | | |
Collapse
|
11
|
A living band-aid for epidermolysis bullosa. BLOOD TRANSFUSION = TRASFUSIONE DEL SANGUE 2015; 13:1-2. [PMID: 25636127 DOI: 10.2450/2014.0289-14] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
12
|
Healy E, Brown SJ, Langan SM, Nicholls SG, Shams K, Reynolds NJ. Identification of translational dermatology research priorities in the U.K.: results of an electronic Delphi exercise. Br J Dermatol 2015; 173:1191-1198. [PMID: 26149834 PMCID: PMC5038882 DOI: 10.1111/bjd.14022] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/02/2015] [Indexed: 01/29/2023]
Abstract
BACKGROUND Translational research is the direct application of basic and applied research to patient care. It is estimated that there are at least 2000 different skin diseases; thus, there are considerable challenges in seeking to undertake research on each of these disorders. OBJECTIVES This electronic Delphi (e-Delphi) exercise was conducted in order to generate a list of translational dermatology research questions that are regarded as a priority for further investigations. METHODS During the first phase of the e-Delphi exercise, 228 research questions were generated by an expert panel that included clinical academic dermatologists, clinical dermatologists, nonclinical scientists, dermatology trainees and representatives from patient support groups. RESULTS Following completion of the second and third phases, 40 questions on inflammatory skin disease, 20 questions on structural skin disorders/genodermatoses, 37 questions on skin cancer and eight miscellaneous questions were designated as priority translational dermatology research questions (PRQs). In addition to PRQs on a variety of disease areas (including multiple PRQs on psoriasis, eczema, squamous cell carcinoma and melanoma), there were a number of cross-cutting themes that identified a need to investigate mechanisms/pathogenesis of disease and the necessity to improve treatments for patients with skin disease. CONCLUSIONS It is predicted that this list of PRQs will help to provide a strategic direction for translational dermatology research in the U.K. and that addressing this list of questions will ultimately provide clinical benefit for substantial numbers of patients with skin disorders.
Collapse
Affiliation(s)
- E Healy
- Dermatopharmacology, Sir Henry Wellcome Laboratories, Faculty of Medicine, University of Southampton, Southampton, UK
- Southampton Dermatology Centre, Royal South Hants Hospital, University Hospitals Southampton NHS Foundation Trust, Southampton, UK
| | - S J Brown
- Department of Dermatology, Ninewells Hospital & Medical School, Dundee, UK
- Dermatology & Genetic Medicine, Medical Research Institute, University of Dundee, Dundee, UK
| | - S M Langan
- Faculty of Epidemiology and Population Health, London School of Tropical Medicine, London, UK
| | - S G Nicholls
- Department of Epidemiology and Community Medicine, Faculty of Medicine, University of Ottawa, Ottawa, Canada
| | - K Shams
- Chemokine Research Group, Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - N J Reynolds
- Dermatology, Institute of Cellular Medicine, Medical School, Newcastle University, Newcastle upon Tyne, UK
| |
Collapse
|
13
|
Cunningham L, Liu L, Menzies S, McGrath J, Lally A. Novel missense mutation in a patient with recessive pretibial epidermolysis bullosa and a mild phenotype. J Eur Acad Dermatol Venereol 2015; 30:e115-e116. [DOI: 10.1111/jdv.13385] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- L. Cunningham
- Department of Dermatology; St. Vincent's University Hospital; Dublin Ireland
| | - L. Liu
- Viapath; St. Thomas’ Hospital; London UK
| | - S. Menzies
- Department of Dermatology; St. Vincent's University Hospital; Dublin Ireland
| | - J.A. McGrath
- Viapath; St. Thomas’ Hospital; London UK
- St John's Institute of Dermatology; King's College London; London UK
| | - A. Lally
- Department of Dermatology; St. Vincent's University Hospital; Dublin Ireland
| |
Collapse
|
14
|
Uitto J, Wang G. Molecular Genetics of Heritable Skin Diseases: Pseudoxanthoma Elasticum as a Paradigm of China-US Collaboration. J Investig Dermatol Symp Proc 2015; 17:20-21. [PMID: 26067309 DOI: 10.1038/jidsymp.2015.17] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Affiliation(s)
- Jouni Uitto
- Department of Dermatology and Cutaneous Biology, Jefferson Medical College of Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Gang Wang
- Department of Dermatology, The Fourth Military Medical University, Xijing Hospital, Xi'an, Peoples Republic of China
| |
Collapse
|
15
|
From marrow to matrix: novel gene and cell therapies for epidermolysis bullosa. Mol Ther 2015; 23:987-992. [PMID: 25803200 DOI: 10.1038/mt.2015.47] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2014] [Accepted: 03/11/2015] [Indexed: 12/22/2022] Open
Abstract
Epidermolysis bullosa encompasses a group of inherited connective tissue disorders that range from mild to lethal. There is no cure, and current treatment is limited to palliative care that is largely ineffective in treating the systemic, life-threatening pathology associated with the most severe forms of the disease. Although allogeneic cell- and protein-based therapies have shown promise, both novel and combinatorial approaches will undoubtedly be required to totally alleviate the disorder. Progress in the development of next-generation therapies that synergize targeted gene-correction and induced pluripotent stem cell technologies offers exciting prospects for personalized, off-the-shelf treatment options that could avoid many of the limitations associated with current allogeneic cell-based therapies. Although no single therapeutic avenue has achieved complete success, each has substantially increased our collective understanding of the complex biology underlying the disease, both providing mechanistic insights and uncovering new hurdles that must be overcome.
Collapse
|
16
|
Epidermal cell junctions and their regulation by p63 in health and disease. Cell Tissue Res 2015; 360:513-28. [PMID: 25645146 DOI: 10.1007/s00441-014-2108-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2014] [Accepted: 12/17/2014] [Indexed: 12/17/2022]
Abstract
As the outermost tissue of the body, the epidermis is the first physical barrier for any pressure, stress or trauma. Several specialized cell-matrix and cell-cell adhesion structures, together with an intracellular network of dedicated intermediate filaments, are required to confer critical resilience to mechanical stress. The transcription factor p63 is a master regulator of gene expression in the epidermis and in other stratified epithelia. It has been extensively demonstrated that p63 positively controls a large number of tissue-specific genes, including those encoding a large fraction of tissue-restricted cell adhesion molecules. Consistent with p63 functions in cell adhesion and in epidermal differentiation, heterozygous mutations clustered mainly in the p63 C-terminus are causative of AEC syndrome, an autosomal dominant disorder characterized by cleft palate, ankyloblepharon and ectodermal dysplasia associated with severe skin erosions, bleeding and infections. The molecular basis of skin erosions in AEC patients is not fully understood, although defects in desmosomes and in other cell junctions are likely to be involved. Here, we provide an extensive review of the different epidermal cell junctions that cooperate to withstand mechanical stress and on the mechanisms by which p63 regulates gene expression of their components in healthy skin and in AEC syndrome. Collectively, advancement in understanding the molecular mechanisms by which epidermal cell junctions precisely exert their functions and how p63 orchestrates their coordinated expression, will ultimately lead to insight into developing future strategies for the treatment of AEC syndrome and more in generally for diseases that share an overlapping phenotype.
Collapse
|
17
|
Hegde V, Hickerson RP, Nainamalai S, Campbell PA, Smith FJD, McLean WHI, Pedrioli DML. In vivo gene silencing following non-invasive siRNA delivery into the skin using a novel topical formulation. J Control Release 2014; 196:355-62. [PMID: 25449884 PMCID: PMC4275573 DOI: 10.1016/j.jconrel.2014.10.022] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2014] [Revised: 10/20/2014] [Accepted: 10/23/2014] [Indexed: 12/17/2022]
Abstract
Therapeutics based on short interfering RNAs (siRNAs), which act by inhibiting the expression of target transcripts, represent a novel class of potent and highly specific next-generation treatments for human skin diseases. Unfortunately, the intrinsic barrier properties of the skin combined with the large size and negative charge of siRNAs make epidermal delivery of these macromolecules quite challenging. To help evaluate the in vivo activity of these therapeutics and refine delivery strategies we generated an innovative reporter mouse model that predominantly expresses firefly luciferase (luc2p) in the paw epidermis--the region of murine epidermis that most closely models the tissue architecture of human skin. Combining this animal model with state-of-the-art live animal imaging techniques, we have developed a real-time in vivo analysis work-flow that has allowed us to compare and contrast the efficacies of a wide range nucleic acid-based gene silencing reagents in the skin of live animals. While inhibition was achieved with all of the reagents tested, only the commercially available "self-delivery" modified Accell-siRNAs (Dharmacon) produced potent and sustained in vivo gene silencing. Together, these findings highlight just how informative reliable reporter mouse models can be when assessing novel therapeutics in vivo. Using this work-flow, we developed a novel clinically-relevant topical formulation that facilitates non-invasive epidermal delivery of unmodified and "self-delivery" siRNAs. Remarkably, a sustained >40% luc2p inhibition was observed after two 1-hour treatments with Accell-siRNAs in our topical formulation. Importantly, our ability to successfully deliver siRNA molecules topically brings these novel RNAi-based therapeutics one-step closer to clinical use.
Collapse
Affiliation(s)
- Vikas Hegde
- Centre for Dermatology and Genetic Medicine, Division of Molecular Medicine, University of Dundee, Dundee DD1 5EH, Scotland, UK
| | - Robyn P Hickerson
- Centre for Dermatology and Genetic Medicine, Division of Molecular Medicine, University of Dundee, Dundee DD1 5EH, Scotland, UK
| | - Sitheswaran Nainamalai
- Centre for Dermatology and Genetic Medicine, Division of Molecular Medicine, University of Dundee, Dundee DD1 5EH, Scotland, UK
| | - Paul A Campbell
- Centre for Dermatology and Genetic Medicine, Division of Molecular Medicine, University of Dundee, Dundee DD1 5EH, Scotland, UK; Carnegie Physics Laboratory, University of Dundee, Dundee DD1 4HN, Scotland, UK
| | - Frances J D Smith
- Centre for Dermatology and Genetic Medicine, Division of Molecular Medicine, University of Dundee, Dundee DD1 5EH, Scotland, UK
| | - W H Irwin McLean
- Centre for Dermatology and Genetic Medicine, Division of Molecular Medicine, University of Dundee, Dundee DD1 5EH, Scotland, UK.
| | - Deena M Leslie Pedrioli
- Centre for Dermatology and Genetic Medicine, Division of Molecular Medicine, University of Dundee, Dundee DD1 5EH, Scotland, UK.
| |
Collapse
|
18
|
Umegaki-Arao N, Pasmooij AMG, Itoh M, Cerise JE, Guo Z, Levy B, Gostyński A, Rothman LR, Jonkman MF, Christiano AM. Induced pluripotent stem cells from human revertant keratinocytes for the treatment of epidermolysis bullosa. Sci Transl Med 2014; 6:264ra164. [PMID: 25429057 DOI: 10.1126/scitranslmed.3009342] [Citation(s) in RCA: 88] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2023]
Abstract
Revertant mosaicism is a naturally occurring phenomenon involving spontaneous correction of a pathogenic gene mutation in a somatic cell. It has been observed in several genetic diseases, including epidermolysis bullosa (EB), a group of inherited skin disorders characterized by blistering and scarring. Induced pluripotent stem cells (iPSCs), generated from fibroblasts or keratinocytes, have been proposed as a treatment for EB. However, this requires genome editing to correct the mutations, and, in gene therapy, efficiency of targeted gene correction and deleterious genomic modifications are still limitations of translation. We demonstrate the generation of iPSCs from revertant keratinocytes of a junctional EB patient with compound heterozygous COL17A1 mutations. These revertant iPSCs were then differentiated into naturally genetically corrected keratinocytes that expressed type XVII collagen (Col17). Gene expression profiling showed a strong correlation between gene expression in revertant iPSC-derived keratinocytes and the original revertant keratinocytes, indicating the successful differentiation of iPSCs into the keratinocyte lineage. Revertant-iPSC keratinocytes were then used to create in vitro three-dimensional skin equivalents and reconstitute human skin in vivo in mice, both of which expressed Col17 in the basal layer. Therefore, revertant keratinocytes may be a viable source of spontaneously gene-corrected cells for developing iPSC-based therapeutic approaches in EB.
Collapse
Affiliation(s)
| | - Anna M G Pasmooij
- Department of Dermatology, University Medical Center Groningen, University of Groningen, Groningen 9700 RB, the Netherlands
| | - Munenari Itoh
- Department of Dermatology, Columbia University, New York, NY 10032, USA. Department of Dermatology, The Jikei University School of Medicine, Tokyo 105-8461, Japan
| | - Jane E Cerise
- Department of Dermatology, Columbia University, New York, NY 10032, USA
| | - Zongyou Guo
- Department of Dermatology, Columbia University, New York, NY 10032, USA
| | - Brynn Levy
- Department of Pathology and Cell Biology, Columbia University, New York, NY 10032, USA
| | - Antoni Gostyński
- Department of Dermatology, University Medical Center Groningen, University of Groningen, Groningen 9700 RB, the Netherlands
| | - Lisa R Rothman
- Department of Dermatology, Columbia University, New York, NY 10032, USA
| | - Marcel F Jonkman
- Department of Dermatology, University Medical Center Groningen, University of Groningen, Groningen 9700 RB, the Netherlands
| | - Angela M Christiano
- Department of Dermatology, Columbia University, New York, NY 10032, USA. Department of Genetics and Development, Columbia University, New York, NY 10032, USA.
| |
Collapse
|
19
|
Abstract
Mammalian skin research represents the convergence of three complementary disciplines: cell biology, mouse genetics, and dermatology. The skin provides a paradigm for current research in cell adhesion, inflammation, and tissue stem cells. Here, I discuss recent insights into the cell biology of skin. Single-cell analysis has revealed that human epidermal stem cells are heterogeneous and differentiate in response to multiple extrinsic signals. Live-cell imaging, optogenetics, and cell ablation experiments show skin cells to be remarkably dynamic. High-throughput, genome-wide approaches have yielded unprecedented insights into the circuitry that controls epidermal stem cell fate. Last, integrative biological analysis of human skin disorders has revealed unexpected functions for elements of the skin that were previously considered purely structural.
Collapse
Affiliation(s)
- Fiona M Watt
- King's College London Centre for Stem Cells and Regenerative Medicine, 28th Floor, Tower Wing, Guy's Hospital, Great Maze Pond, London SE1 9RT, UK.
| |
Collapse
|
20
|
Bone marrow stromal cells as immunomodulators. A primer for dermatologists. J Dermatol Sci 2014; 77:11-20. [PMID: 25476233 DOI: 10.1016/j.jdermsci.2014.10.004] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2014] [Revised: 09/29/2014] [Accepted: 10/15/2014] [Indexed: 02/07/2023]
Abstract
Bone marrow stromal cells (BMSCs, also known as mesenchymal stem cells or MSCs) represent a unique cell population in the bone marrow with a long-known function to support hematopoiesis and replace skeletal tissues. The recent discovery that BMSCs also possess potent immunoregulatory features attracted a great deal of attention from stem cell biologists, immunologists and clinicians of different specialties worldwide. Initial clinical experience along with several animal models suggested that intravenously delivered BMSCs are able to regulate a wide variety of host immune cells and act in a way that is beneficial for the recipient in a variety of diseases. The role of the present review is to give a short introduction to the biology of BMSCs and to summarize our current understanding of how BMSCs modulate the immune system with special emphasis on available clinical data. Considering the audience of this journal we will also attempt to guide dermatologists in choosing the right skin conditions where BMSCs might be considered as a therapeutic alternative.
Collapse
|
21
|
Lane SW, Williams DA, Watt FM. Modulating the stem cell niche for tissue regeneration. Nat Biotechnol 2014; 32:795-803. [PMID: 25093887 PMCID: PMC4422171 DOI: 10.1038/nbt.2978] [Citation(s) in RCA: 411] [Impact Index Per Article: 37.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2014] [Accepted: 07/06/2014] [Indexed: 02/06/2023]
Abstract
The field of regenerative medicine holds considerable promise for treating diseases that are currently intractable. Although many researchers are adopting the strategy of cell transplantation for tissue repair, an alternative approach to therapy is to manipulate the stem cell microenvironment, or niche, to facilitate repair by endogenous stem cells. The niche is highly dynamic, with multiple opportunities for intervention. These include administration of small molecules, biologics or biomaterials that target specific aspects of the niche, such as cell-cell and cell-extracellular matrix interactions, to stimulate expansion or differentiation of stem cells, or to cause reversion of differentiated cells to stem cells. Nevertheless, there are several challenges in targeting the niche therapeutically, not least that of achieving specificity of delivery and responses. We envisage that successful treatments in regenerative medicine will involve different combinations of factors to target stem cells and niche cells, applied at different times to effect recovery according to the dynamics of stem cell-niche interactions.
Collapse
Affiliation(s)
- Steven W Lane
- Division of Immunology, QIMR Berghofer Medical Research Institute, Royal Brisbane Hospital, Herston, Queensland, Australia
| | - David A Williams
- 1] Division of Hematology/Oncology, Boston Children's Hospital and Department of Pediatric Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA. [2] Harvard Stem Cell Institute, Boston, Massachusetts, USA
| | - Fiona M Watt
- Centre for Stem Cells and Regenerative Medicine, King's College London, Guy's Hospital, Great Maze Pond, London, UK
| |
Collapse
|
22
|
Abstract
Harnessing the regenerative capacity of keratinocytes and fibroblasts from human skin has created new opportunities to develop cell-based therapies for patients. Cultured cells and bioengineered skin products are being used to treat patients with inherited and acquired skin disorders associated with defective skin, and further clinical trials of new products are in progress. The capacity of extracutaneous sources of cells such as bone marrow is also being investigated for its plasticity in regenerating skin, and new strategies, such as the derivation of inducible pluripotent stem cells, also hold great promise for future cell therapies in dermatology. This article reviews some of the preclinical and clinical studies and future directions relating to cell therapy in dermatology, particularly for inherited skin diseases associated with fragile skin and poor wound healing.
Collapse
Affiliation(s)
- Gabriela Petrof
- St. John's Institute of Dermatology, King's College London (Guy's Campus), London SE1 1UL, United Kingdom
| | - Alya Abdul-Wahab
- St. John's Institute of Dermatology, King's College London (Guy's Campus), London SE1 1UL, United Kingdom
| | - John A McGrath
- St. John's Institute of Dermatology, King's College London (Guy's Campus), London SE1 1UL, United Kingdom
| |
Collapse
|
23
|
Vanden Oever MJ, Tolar J. Advances in understanding and treating dystrophic epidermolysis bullosa. F1000PRIME REPORTS 2014; 6:35. [PMID: 24860657 PMCID: PMC4017907 DOI: 10.12703/p6-35] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Epidermolysis bullosa is a group of inherited disorders that can be both systemic and life-threatening. Standard treatments for the most severe forms of this disorder, typically limited to palliative care, are ineffective in reducing the morbidity and mortality due to complications of the disease. Emerging therapies—such as the use of allogeneic cellular therapy, gene therapy, and protein therapy—have all shown promise, but it is likely that several approaches will need to be combined to realize a cure. For recessive dystrophic epidermolysis bullosa, each particular therapeutic approach has added to our understanding of type VII collagen (C7) function and the basic biology surrounding the disease. The efficacy of these therapies and the mechanisms by which they function also give us insight into developing future strategies for treating this and other extracellular matrix disorders.
Collapse
|
24
|
Abstract
Hair loss is a topic of enormous public interest and understanding the pathophysiology and treatment of various alopecias will likely make a large impact on patients' lives. The investigation of alopecias also provides important insight in the basic sciences; for instance, the abundance of stem cell populations and regenerative cycles that characterize a hair follicle render it an excellent model for the study of stem cell biology. This review seeks to provide a concise summary of the major alopecias with regard to presentation and management, and correlate these to recent advances in relevant research on pathogenesis.
Collapse
Affiliation(s)
- Ji Qi
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, Maryland 21287
| | | |
Collapse
|
25
|
Gostyński A, Pasmooij AMG, Jonkman MF. Successful therapeutic transplantation of revertant skin in epidermolysis bullosa. J Am Acad Dermatol 2014; 70:98-101. [PMID: 24176523 DOI: 10.1016/j.jaad.2013.08.052] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2013] [Revised: 08/16/2013] [Accepted: 08/23/2013] [Indexed: 11/27/2022]
Abstract
BACKGROUND Epidermolysis bullosa (EB) is a group of genetic blistering diseases. Despite many efforts, treatment for EB remains symptomatic. Revertant mosaicism, coexistence of cells carrying disease-causing mutations with cells in which the inherited mutation is genetically corrected by a spontaneous genetic event (revertant cells) in 1 individual, can be found in EB. The naturally corrected revertant keratinocytes provide an opportunity for autologous cell therapy. OBJECTIVE We sought to locally treat EB by transplantation of revertant skin. METHODS Persistent ulcers in a patient with non-Herlitz junctional EB caused by mutations in the LAMB3 gene were treated by transplantation of split-thickness biopsy specimens from one of his revertant patches. RESULTS All transplanted biopsy specimens were accepted and complete re-epithelialization occurred within 14 days. During 18 months of follow-up, the patient never experienced blisters or wounds in the grafted area, nor in the healed donor site. Immunofluorescence and DNA sequencing showed that acceptor sites healed with transplanted revertant keratinocytes. LIMITATIONS Punch grafting allows only limited expansion of revertant skin. CONCLUSIONS We demonstrate that phenotypical and genotypical correction of skin in patients with revertant mosaicism by expansion of revertant skin might be a promising therapeutic option for cutaneous manifestations of EB.
Collapse
Affiliation(s)
- Antoni Gostyński
- Center for Blistering Diseases and Department of Dermatology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands.
| | - Anna M G Pasmooij
- Center for Blistering Diseases and Department of Dermatology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Marcel F Jonkman
- Center for Blistering Diseases and Department of Dermatology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| |
Collapse
|
26
|
Phase I Study Protocol for Ex Vivo Lentiviral Gene Therapy for the Inherited Skin Disease, Netherton Syndrome. HUM GENE THER CL DEV 2013; 24:182-90. [DOI: 10.1089/humc.2013.195] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
|
27
|
Progress in Epidermolysis bullosa research: summary of DEBRA International Research Conference 2012. J Invest Dermatol 2013; 133:2121-6. [PMID: 23949764 DOI: 10.1038/jid.2013.127] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
28
|
Itoh M, Umegaki-Arao N, Guo Z, Liu L, Higgins CA, Christiano AM. Generation of 3D skin equivalents fully reconstituted from human induced pluripotent stem cells (iPSCs). PLoS One 2013; 8:e77673. [PMID: 24147053 PMCID: PMC3795682 DOI: 10.1371/journal.pone.0077673] [Citation(s) in RCA: 146] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2013] [Accepted: 09/05/2013] [Indexed: 01/01/2023] Open
Abstract
Recent generation of patient-specific induced pluripotent stem cells (PS-iPSCs) provides significant advantages for cell- and gene-based therapy. Establishment of iPSC-based therapy for skin diseases requires efficient methodology for differentiating iPSCs into both keratinocytes and fibroblasts, the major cellular components of the skin, as well as the reconstruction of skin structures using these iPSC-derived skin components. We previously reported generation of keratinocytes from human iPSCs for use in the treatment of recessive dystrophic epidermolysis bullosa (RDEB) caused by mutations in the COL7A1 gene. Here, we developed a protocol for differentiating iPSCs into dermal fibroblasts, which also produce type VII collagen and therefore also have the potential to treat RDEB. Moreover, we generated in vitro 3D skin equivalents composed exclusively human iPSC-derived keratinocytes and fibroblasts for disease models and regenerative therapies for skin diseases, first demonstrating that iPSCs can provide the basis for modeling a human organ derived entirely from two different types of iPSC-derived cells.
Collapse
Affiliation(s)
- Munenari Itoh
- Department of Dermatology, Columbia University, College of Physicians & Surgeons, New York, New York, United States of America
| | - Noriko Umegaki-Arao
- Department of Dermatology, Columbia University, College of Physicians & Surgeons, New York, New York, United States of America
| | - Zongyou Guo
- Department of Dermatology, Columbia University, College of Physicians & Surgeons, New York, New York, United States of America
| | - Liang Liu
- Department of Dermatology, Columbia University, College of Physicians & Surgeons, New York, New York, United States of America
| | - Claire A. Higgins
- Department of Dermatology, Columbia University, College of Physicians & Surgeons, New York, New York, United States of America
| | - Angela M. Christiano
- Department of Dermatology, Columbia University, College of Physicians & Surgeons, New York, New York, United States of America
- Department of Genetics & Development, Columbia University, College of Physicians & Surgeons, New York, New York, United States of America
- * E-mail:
| |
Collapse
|
29
|
Tolar J, Wagner JE. Allogeneic blood and bone marrow cells for the treatment of severe epidermolysis bullosa: repair of the extracellular matrix. Lancet 2013; 382:1214-23. [PMID: 24095195 PMCID: PMC3959900 DOI: 10.1016/s0140-6736(13)61897-8] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Contrary to the prevailing professional opinion of the past few decades, recent experimental and clinical data support the fact that protein replacement therapy by allogeneic blood and marrow transplantation is not limited to freely diffusible molecules such as enzymes, but also large structural proteins such as collagens. A prime example is the cross-correction of type VII collagen deficiency in generalised severe recessive dystrophic epidermolysis bullosa, in which blood and marrow transplantation can attenuate the mucocutaneous manifestations of the disease and improve patients' quality of life. Although allogeneic blood and marrow transplantation can improve the integrity of the skin and mucous membranes, today's accomplishments are only the first steps on the long pathway to cure. Future strategies will be built on the lessons learned from these first transplant studies.
Collapse
Affiliation(s)
- Jakub Tolar
- Division of Blood and Marrow Transplantation, Department of Pediatrics, University of Minnesota Medical School, Minneapolis, MN 55455, USA
| | | |
Collapse
|
30
|
Uitto J, Has C, Bruckner-Tuderman L. Cell-based therapies for epidermolysis bullosa - from bench to bedside. J Dtsch Dermatol Ges 2013; 10:803-7. [PMID: 23107326 DOI: 10.1111/j.1610-0387.2012.08035.x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Significant progress has been made over the past two decades in molecular genetics of epidermolysis bullosa (EB), a group of heritable blistering disorders, with diagnostic and prognostic implications. More recently, novel molecular approaches have been developed towards potential treatment of EB, with emphasis on gene-, protein-, and cell-based strategies. This overview highlights cell-based approaches that have recently been tested in pilot clinical trials, attesting to the potential of regenerative medicine for blistering skin diseases.
Collapse
Affiliation(s)
- Jouni Uitto
- Department of Dermatology and Cutaneous Biology, Jefferson Medical College, Philadelphia, Pennsylvania 19107, USA.
| | | | | |
Collapse
|
31
|
Highly rapid and efficient conversion of human fibroblasts to keratinocyte-like cells. J Invest Dermatol 2013; 134:335-344. [PMID: 23921950 PMCID: PMC3875612 DOI: 10.1038/jid.2013.327] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2013] [Revised: 06/07/2013] [Accepted: 06/27/2013] [Indexed: 02/04/2023]
Abstract
Cell fate commitment during development is achieved through the expression of lineage specific transcription factors. Recent studies have suggested that expression of combinations of these lineage specific transcription factors can convert adult somatic cells from one type to another. Here we report that the combination of p63, a master regulator of epidermal development and differentiation, and KLF4, a regulator of epidermal differentiation is sufficient to convert dermal fibroblasts to a keratinocyte phenotype. Induced keratinocytes expressed keratinocyte specific proteins and had a transcriptome similar to keratinocytes. Reprogramming to a keratinocyte phenotype was rapid and efficient with a vast majority of cells morphologically resembling and expressing keratinocyte specific genes within a week of p63 and KLF4 transduction. Furthermore, p63 and KLF4 are capable of inducing a keratinocyte phenotype even in a cancerous cell line highlighting their importance for epidermal specification. The robustness of the conversion process also allows the use of this as a model system to study the mechanisms of reprogramming.
Collapse
|
32
|
Disorders of the cutaneous basement membrane zone--the paradigm of epidermolysis bullosa. Matrix Biol 2013; 33:29-34. [PMID: 23917088 DOI: 10.1016/j.matbio.2013.07.007] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2013] [Revised: 07/01/2013] [Accepted: 07/01/2013] [Indexed: 12/19/2022]
Abstract
The cutaneous basement membrane zone (BMZ) is a highly specialized functional complex that provides the skin with structural adhesion and resistance to shearing forces. Its regulatory functions include control of epithelial-mesenchymal interactions under physiological and pathological conditions. Mutations in genes encoding components of the BMZ are associated with inherited skin disorders of the epidermolysis bullosa (EB) group, characterized by skin fragility, mechanically induced blisters and erosions of the skin and mucous membranes. Although most disease-associated genes are known, the genetic basis of new EB subtypes linked to mutations in genes for focal adhesion proteins was uncovered only recently. The molecular mechanisms leading to blistering, abnormal wound healing, predisposition to skin cancer, and other complications in EB have been elucidated using animal models and disease proteomics. The rapid progress in understanding the molecular basis of EB has enabled the development of strategies for biologically valid causal therapies.
Collapse
|
33
|
Abstract
During the past 20 years, tremendous progress has been made in our understanding of the molecular basis of many genetic skin conditions. The translation of these laboratory findings into effective therapies for affected individuals has been slow, however, in large part due to the risk of carcinogenesis from random viral genomic integration and the lack of efficacy of topically applied genetic material and most proteins. As intervention at the gene level still appears remote for most genetic disorders, increased knowledge about the cellular and biochemical pathogenesis of disease allows specific targeting of pathways with existing and/or novel drugs and molecules. In contrast to the requirement for personalization of most gene-based approaches, pathogenesis-based therapy is pathway specific, and in theory, it should have broader applicability. In this chapter, we provide an overview of the pathoetiology of the various types of ichthyoses and demonstrate how a pathogenesis-based approach can potentially lead to innovative treatments for these conditions. Notably, this strategy has been successfully validated for the treatment of the rare X-linked dominant condition, CHILD syndrome, in which topical applications of cholesterol and lovastatin together to affected skin resulted in marked improvement of the skin phenotype.
Collapse
Affiliation(s)
- Joey E Lai-Cheong
- St John's Institute of Dermatology, King's College London, London, United Kingdom
| | | | | |
Collapse
|
34
|
Mahto A, McGrath JA, Deroide F, Rustin MHA. Late-onset pretibial recessive dystrophic epidermolysis bullosa. Clin Exp Dermatol 2013; 38:630-2. [PMID: 23786535 DOI: 10.1111/ced.12072] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/30/2012] [Indexed: 12/30/2022]
Abstract
Pretibial epidermolysis bullosa (EB) is a rare form of localized dystrophic EB, characterized by recurrent blistering and scarring plaques occurring predominantly in the pretibial area. In most cases, nail dystrophy, especially of the toenails, is also present. Often there are no clinical abnormalities at birth, and the disorder may only appear after several years. We report a patient who developed symptoms in his fifth decade. Genetic testing identified compound heterozygosity for two pathogenic mutations in the COL7A1 gene. This case highlights a rare variant of mechanobullous disease, and stresses the importance of molecular screening in establishing a correct diagnosis. Precisely why the disorder specifically localizes to the shins or why it may only become apparent in later life is not known.
Collapse
Affiliation(s)
- A Mahto
- Department of Dermatology, Royal Free Hospital, London, UK.
| | | | | | | |
Collapse
|
35
|
Abdul-Wahab A, Petrof G, McGrath JA. Bone marrow transplantation in epidermolysis bullosa. Immunotherapy 2013; 4:1859-67. [PMID: 23240753 DOI: 10.2217/imt.12.120] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Epidermolysis bullosa (EB) is a heterogeneous group of inherited blistering skin diseases. Severe forms of EB are associated with increased morbidity and mortality, and there is currently no effective treatment. To combat severe complications of EB, such as chronic erosions, scarring and malignancy, effective therapy needs to be given systemically and at an early age. One recent therapeutic advancement has been a clinical trial of whole bone marrow (BM) transplantation in children with the dystrophic form of EB. This led to correction of the inherent skin basement membrane defect and better skin integrity in some individuals. The challenge now is to precisely identify which BM cells contribute to skin recovery and what mechanisms are involved in tissue regeneration. An improved understanding of the key aspects of BM skin repair is likely to lead to significant health improvements for patients with EB and other skin diseases.
Collapse
Affiliation(s)
- Alya Abdul-Wahab
- St John's Institute of Dermatology, King's College London (Guy's Campus), 9th Floor, Tower Wing, Guy's Hospital, Great Maze Pond, London, SE1 9RT, UK
| | | | | |
Collapse
|
36
|
|
37
|
New findings in genodermatoses. Dermatol Clin 2013; 31:303-15. [PMID: 23557657 DOI: 10.1016/j.det.2012.12.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
New technologies are accelerating the pace at which genetic defects leading to inherited skin disease are elucidated. Translation of these genetic discoveries into new therapies for patients with inherited skin diseases has not been as rapid but the pace is now accelerating. This article summarizes recent findings in genetic skin diseases, the scope of advances being made, the role of new DNA analysis technologies in these discoveries, as well as highlighting some examples of how an understanding of the genetic cause of inherited skin diseases can lead to therapeutic interventions for patients.
Collapse
|
38
|
|
39
|
Byron A, Humphries JD, Humphries MJ. Defining the extracellular matrix using proteomics. Int J Exp Pathol 2013; 94:75-92. [PMID: 23419153 DOI: 10.1111/iep.12011] [Citation(s) in RCA: 123] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2012] [Revised: 09/13/2012] [Accepted: 11/16/2012] [Indexed: 12/11/2022] Open
Abstract
The cell microenvironment has a profound influence on the behaviour, growth and survival of cells. The extracellular matrix (ECM) provides not only mechanical and structural support to cells and tissues but also binds soluble ligands and transmembrane receptors to provide spatial coordination of signalling processes. The ability of cells to sense the chemical, mechanical and topographical features of the ECM enables them to integrate complex, multiparametric information into a coherent response to the surrounding microenvironment. Consequently, dysregulation or mutation of ECM components results in a broad range of pathological conditions. Characterization of the composition of ECM derived from various cells has begun to reveal insights into ECM structure and function, and mechanisms of disease. Proteomic methodologies permit the global analysis of subcellular systems, but extracellular and transmembrane proteins present analytical difficulties to proteomic strategies owing to the particular biochemical properties of these molecules. Here, we review advances in proteomic approaches that have been applied to furthering our understanding of the ECM microenvironment. We survey recent studies that have addressed challenges in the analysis of ECM and discuss major outcomes in the context of health and disease. In addition, we summarize efforts to progress towards a systems-level understanding of ECM biology.
Collapse
Affiliation(s)
- Adam Byron
- Wellcome Trust Centre for Cell-Matrix Research, Faculty of Life Sciences, University of Manchester, Manchester, UK
| | | | | |
Collapse
|
40
|
Abstract
Laminin 332 is an essential component of the dermal-epidermal junction, a highly specialized basement membrane zone that attaches the epidermis to the dermis and thereby provides skin integrity and resistance to external mechanical forces. Mutations in the LAMA3, LAMB3 and LAMC2 genes that encode the three constituent polypeptide chains, α3, β3 and γ2, abrogate or perturb the functions of laminin 332. The phenotypic consequences are diminished dermal-epidermal adhesion and, as clinical symptoms, skin fragility and mechanically induced blistering. The disorder is designated as junctional epidermolysis bullosa (JEB). This article delineates the signs and symptoms of the different forms of JEB, the mutational spectrum, genotype-phenotype correlations as well as perspectives for future molecular therapies.
Collapse
Affiliation(s)
- Dimitra Kiritsi
- Department of Dermatology; University Freiburg Medical Center; Freiburg, Germany
| | - Cristina Has
- Department of Dermatology; University Freiburg Medical Center; Freiburg, Germany
| | | |
Collapse
|
41
|
Abstract
Groundbreaking advances on the molecular and cellular physiological and physiopathological skin processes, including the complete sequencing of the genome of several species and the increased availability of gene-modified organisms, paved the way to firmly establishing molecular approaches and methods in experimental, translational, and clinical dermatology. As a result, newly developed experimental ex vivo assays and animal models prove exquisite tools for addressing fundamental physiological cutaneous processes and pathogenic mechanisms of skin diseases. A plethora of new findings that were generated using these experimental tools serve as a strong basis for intense translational research efforts aiming at developing new, specific, and sensitive diagnostic tests and efficient "personalized" therapies with less side-effects. Consequently, a broad array of molecular diagnostic tests and therapies for a wide spectrum of skin diseases ranging from genodermatoses through skin neoplasms, allergy, inflammatory and autoimmune diseases, are already routinely used in the clinical dermatology practice. This article highlights several major developments in molecular experimental and clinical dermatology.
Collapse
Affiliation(s)
- Cristina Has
- Department of Molecular Dermatology, University of Freiburg, Freiburg, Germany.
| | | |
Collapse
|
42
|
Uitto J. Rare heritable skin diseases: targets for regenerative medicine. J Invest Dermatol 2012; 132:2485-8. [PMID: 23069901 DOI: 10.1038/jid.2012.334] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
43
|
[New developments in hereditary blistering skin diseases]. Hautarzt 2012; 64:7-11. [PMID: 23263713 DOI: 10.1007/s00105-012-2406-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Hereditary blistering skin diseases were described more than hundred years ago, but only the rapid scientific developments in molecular genetics in the last years have revealed the full spectrum of these diseases, delineated disease mechanisms and pointed to novel therapeutic strategies. Not only the classic forms of epidermolysis bullosa, but also new syndromic forms with multiorgan involvement, or skin fragility disorders that manifest with erosive, crusty lesions and pigment anomalies, instead of marked skin blistering belong to the group of hereditary blistering diseases. Understanding the biological functions of skin structures that provide intraepidermal and dermo-epidermal adhesion has furthered development of novel cell- and molecule-based therapies that are currently being tested in preclinical and clinical pilot trial settings.
Collapse
|
44
|
Kopecki Z, Ruzehaji N, Turner C, Iwata H, Ludwig RJ, Zillikens D, Murrell DF, Cowin AJ. Topically applied flightless I neutralizing antibodies improve healing of blistered skin in a murine model of epidermolysis bullosa acquisita. J Invest Dermatol 2012; 133:1008-16. [PMID: 23223144 DOI: 10.1038/jid.2012.457] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Epidermolysis bullosa (EB) is a chronic inheritable disease that leads to severe blistering and fibrosis. Previous studies have shown that the actin cytoskeletal protein flightless I (Flii) impairs wound healing associated with EB. Using a mouse model of EB acquisita (EBA), the effect of "mopping up" Flii using Flii-neutralizing antibodies (FnAbs) before, during, and after blister formation was determined. FnAbs, incorporated into a cream vehicle and applied topically to the skin, penetrated into the basal epidermis and upper papillary dermis but were not detected in serum or other organs and did not alter neutrophil or macrophage infiltration into the blistered skin. Histological assessment of blister severity showed that treatment of early-stage blisters with FnAb cream reduced their severity and improved their rate of healing. Treatment of established blisters with FnAb cream also improved healing and restored the skin's tensile strength toward that of normal skin. Repeated application of FnAbs to EBA skin before the onset of blistering reduced the severity of skin blistering. Independent of when the FnAbs were applied, skin barrier function and wound healing were improved and skin fragility was reduced, suggesting that FnAbs could potentially improve healing of patients with EB.
Collapse
Affiliation(s)
- Zlatko Kopecki
- Women's and Children's Health Research Institute, North Adelaide, South Australia, Australia.
| | | | | | | | | | | | | | | |
Collapse
|
45
|
Affiliation(s)
- Jouni Uitto
- Department of Dermatology and Cutaneous Biology, Jefferson Medical College, and Jefferson Institute of Molecular Medicine, Thomas Jefferson University, Philadelphia, Pennsylvania, USA.
| |
Collapse
|
46
|
|
47
|
Tolar J, Wagner JE. Management of severe epidermolysis bullosa by haematopoietic transplant: principles, perspectives and pitfalls. Exp Dermatol 2012; 21:896-900. [PMID: 23016552 DOI: 10.1111/exd.12014] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/15/2012] [Indexed: 12/17/2022]
Abstract
People with severe forms of epidermolysis bullosa (EB) develop widespread blistering and progressively debilitating multisystem complications that may result in a shortened lifespan. As some wounds in EB individuals are difficult or impossible to access with topical therapy, we examined the potential of systemic therapy with normal haematopoietic stem cells. In both animal models and children with EB, healthy donor cells from the haematopoietic graft migrated to the injured skin; simultaneously, there was an increase in the production of skin-specific structural proteins deficient in EB, increased skin integrity and reduced tendency to blister formation. Even though the majority of evaluable individuals have had a positive response in skin healing, frequently changing their quality of life, the improvement in lifestyle has been varied and the overall clinical response incomplete. To change the current amelioration of disease into a full cure, we propose to (i) increase safety as well as efficacy of haematopoietic cell transplant (HCT) using co-infusion of mesenchymal stromal/stem cells with haematopoietic stem cells and non-myeloablative conditioning for transplant; (ii) optimize homing of donor cells into the skin erosions in animal models of EB; and (iii) discover and test new drugs for EB therapy using patient-specific induced pluripotent stem cells. We conclude that although HCT has always been a risky treatment restricted to those with serious life-threatening or debilitating diseases, by most benchmarks, the results of HCT in EB have shown that HCT has the potential of being a durable, systemic therapy for people with severe forms of EB.
Collapse
Affiliation(s)
- Jakub Tolar
- Division of Blood and Marrow Transplantation, Department of Pediatrics, University of Minnesota, Minneapolis, MN 55455, USA.
| | | |
Collapse
|
48
|
Chamcheu JC, Wood GS, Siddiqui IA, Syed DN, Adhami VM, Teng JM, Mukhtar H. Progress towards genetic and pharmacological therapies for keratin genodermatoses: current perspective and future promise. Exp Dermatol 2012; 21:481-9. [PMID: 22716242 PMCID: PMC3556927 DOI: 10.1111/j.1600-0625.2012.01534.x] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Hereditary keratin disorders of the skin and its appendages comprise a large group of clinically heterogeneous disfiguring blistering and ichthyotic diseases, primarily characterized by the loss of tissue integrity, blistering and hyperkeratosis in severely affected tissues. Pathogenic mutations in keratins cause these afflictions. Typically, these mutations in concert with characteristic features have formed the basis for improved disease diagnosis, prognosis and most recently therapy development. Examples include epidermolysis bullosa simplex, keratinopathic ichthyosis, pachyonychia congenita and several other tissue-specific hereditary keratinopathies. Understanding the molecular and genetic events underlying skin dysfunction has initiated alternative treatment approaches that may provide novel therapeutic opportunities for affected patients. Animal and in vitro disease modelling studies have shed more light on molecular pathogenesis, further defining the role of keratins in disease processes and promoting the translational development of new gene and pharmacological therapeutic strategies. Given that the molecular basis for these monogenic disorders is well established, gene therapy and drug discovery targeting pharmacological compounds with the ability to reinforce the compromised cytoskeleton may lead to promising new therapeutic strategies for treating hereditary keratinopathies. In this review, we will summarize and discuss recent advances in the preclinical and clinical modelling and development of gene, natural product, pharmacological and protein-based therapies for these disorders, highlighting the feasibility of new approaches for translational clinical therapy.
Collapse
Affiliation(s)
- Jean Christopher Chamcheu
- Department of Dermatology, School of Medicine and Public Health, University of Wisconsin, Madison, WI, USA
| | | | | | | | | | | | | |
Collapse
|
49
|
|