1
|
Levian B, Hou Y, Tang X, Bainvoll L, Zheng K, Badarinarayana V, Aghamohammadzadeh S, Chen M. Novel readthrough agent suppresses nonsense mutations and restores functional type VII collagen and laminin 332 in epidermolysis bullosa. MOLECULAR THERAPY. NUCLEIC ACIDS 2024; 35:102334. [PMID: 39391765 PMCID: PMC11465179 DOI: 10.1016/j.omtn.2024.102334] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Accepted: 09/05/2024] [Indexed: 10/12/2024]
Abstract
Recessive dystrophic epidermolysis bullosa (RDEB) and junctional epidermolysis bullosa (JEB) are lethal blistering skin disorders resulting from mutations in genes coding for type VII collagen (COL7A1) and laminin 332 (LAMA3, LAMB3, or LAMC2), respectively. In RDEB, 25% of patients harbor nonsense mutations causing premature termination codons (PTCs). In JEB, a majority of mutations in LAMB3 are nonsense mutations (80%). ELX-02, an aminoglycoside analog, has demonstrated superior PTC readthrough activity and lower toxicity compared to gentamicin in various genetic disorders. This study investigated the ability of ELX-02 to suppress PTCs and promote the expression of C7 and laminin 332 in primary RDEB keratinocytes/fibroblasts and primary JEB keratinocytes harboring nonsense mutations. ELX-02 induced a dose-dependent production of C7 or laminin β3 that surpassed the results achieved with gentamicin. ELX-02 reversed RDEB and JEB cellular hypermotility and improved poor cell-substratum adhesion in JEB cells. Importantly, ELX-02-induced C7 and laminin 332 localized to the dermal-epidermal junction. This is the first study demonstrating that ELX-02 can induce PTC readthrough and restore functional C7 and laminin 332 in RDEB and JEB caused by nonsense mutations. Therefore, ELX-02 may offer a novel and safe therapy for RDEB, JEB, and other inherited skin diseases caused by nonsense mutations.
Collapse
Affiliation(s)
- Brandon Levian
- Department of Dermatology, The Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Yingping Hou
- Department of Dermatology, The Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Xin Tang
- Department of Dermatology, The Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Liat Bainvoll
- Department of Dermatology, The Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Kate Zheng
- Department of Dermatology, The Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | | | | | - Mei Chen
- Department of Dermatology, The Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| |
Collapse
|
2
|
Zorina A, Zorin V, Isaev A, Kudlay D, Manturova N, Ustugov A, Kopnin P. Current Status of Biomedical Products for Gene and Cell Therapy of Recessive Dystrophic Epidermolysis Bullosa. Int J Mol Sci 2024; 25:10270. [PMID: 39408598 PMCID: PMC11476579 DOI: 10.3390/ijms251910270] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 08/22/2024] [Accepted: 08/27/2024] [Indexed: 10/20/2024] Open
Abstract
This detailed review describes innovative strategies and current products for gene and cell therapy at different stages of research and development to treat recessive dystrophic epidermolysis bullosa (RDEB) which is associated with the functional deficiency of collagen type VII alpha 1 (C7) caused by defects in the COL7A1 gene. The use of allogenic mesenchymal stem/stromal cells, which can be injected intradermally and intravenously, appears to be the most promising approach in the field of RDEB cell therapy. Injections of genetically modified autologous dermal fibroblasts are also worth mentioning under this framework. The most common methods of RDEB gene therapy are gene replacement using viral vectors and gene editing using programmable nucleases. Ex vivo epidermal transplants (ETs) based on autologous keratinocytes (Ks) have been developed using gene therapy methods; one such ET successively passed phase III clinical trials. Products based on the use of two-layer transplants have also been developed with both types of skin cells producing C7. Gene products have also been developed for local use. To date, significant progress has been achieved in the development of efficient biomedical products to treat RDEB, one of the most severe hereditary diseases.
Collapse
Affiliation(s)
- Alla Zorina
- Artgen Biotech, Moscow 119333, Russia; (A.Z.)
- Skincell LLC, Moscow 119333, Russia
| | - Vadim Zorin
- Artgen Biotech, Moscow 119333, Russia; (A.Z.)
- Skincell LLC, Moscow 119333, Russia
| | - Artur Isaev
- Artgen Biotech, Moscow 119333, Russia; (A.Z.)
| | - Dmitry Kudlay
- Department of Pharmacology, The I. M. Sechenov First Moscow State Medical University (The Sechenov University), Moscow 119991, Russia
- Department of Pharmacognosy and Industrial Pharmacy, Lomonosov Moscow State University, Moscow 119992, Russia
| | - Natalia Manturova
- Department of Plastic and Reconstructive surgery, Cosmetology and Cell Technologies, Pirogov Russian National Research Medical University, Moscow 117997, Russia
- JSC Plastic Surgery and Cosmetology Institute, Moscow 125047, Russia
| | - Andrei Ustugov
- Department of Plastic and Reconstructive surgery, Cosmetology and Cell Technologies, Pirogov Russian National Research Medical University, Moscow 117997, Russia
- JSC Plastic Surgery and Cosmetology Institute, Moscow 125047, Russia
| | - Pavel Kopnin
- Scientific Research Institute of Carcinogenesis, N. N. Blokhin National Medical Research Center of Oncology, Moscow 115522, Russia
| |
Collapse
|
3
|
Chai AC, Siegwart DJ, Wang RC. Nucleic Acid Therapy for the Skin. J Invest Dermatol 2024:S0022-202X(24)02062-1. [PMID: 39269387 DOI: 10.1016/j.jid.2024.07.029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Accepted: 07/16/2024] [Indexed: 09/15/2024]
Abstract
Advances in sequencing technologies have facilitated the identification of the genes and mechanisms for many inherited skin diseases. Although targeted nucleic acid therapeutics for diseases in other organs have begun to be deployed in patients, the goal of precise therapeutics for skin diseases has not yet been realized. First, we review the current and emerging nucleic acid-based gene-editing and delivery modalities. Next, current and emerging viral and nanoparticle vehicles for the delivery of gene therapies are reviewed. Finally, specific skin diseases that could benefit optimally from nucleic acid therapies are highlighted. By adopting the latest technologies and addressing specific barriers related to skin biology, nucleic acid therapeutics have the potential to revolutionize treatments for patients with skin disease.
Collapse
Affiliation(s)
- Andreas C Chai
- Department of Dermatology, The University of Texas Southwestern Medical Center, Dallas, Texas, USA; Medical Scientist Training Program, The University of Texas Southwestern Medical Center, Dallas, Texas, USA; Harmon Center for Regenerative Science and Medicine, The University of Texas Southwestern Medical Center, Dallas, Texas, USA.
| | - Daniel J Siegwart
- Department of Biomedical Engineering, The University of Texas Southwestern Medical Center, Dallas, Texas, USA; Department of Biochemistry, The University of Texas Southwestern Medical Center, Dallas, Texas, USA; Program in Genetic Drug Engineering, The University of Texas Southwestern Medical Center, Dallas, Texas, USA; Harold C. Simmons Comprehensive Cancer Center, The University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Richard C Wang
- Department of Dermatology, The University of Texas Southwestern Medical Center, Dallas, Texas, USA; Harold C. Simmons Comprehensive Cancer Center, The University of Texas Southwestern Medical Center, Dallas, Texas, USA
| |
Collapse
|
4
|
Woodley DT, Hao M, Kwong A, Levian B, Cogan J, Hou Y, Mosallaei D, Kleinman E, Zheng K, Chung C, Kim G, Peng D, Chen M. Intravenous gentamicin therapy induces functional type VII collagen in patients with recessive dystrophic epidermolysis bullosa: an open-label clinical trial. Br J Dermatol 2024; 191:267-274. [PMID: 38366625 PMCID: PMC11250489 DOI: 10.1093/bjd/ljae063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Revised: 02/02/2024] [Accepted: 02/05/2024] [Indexed: 02/18/2024]
Abstract
BACKGROUND Recessive dystrophic epidermolysis bullosa (RDEB) is an incurable widespread blistering skin disorder caused by mutations in the gene encoding for type VII collagen (C7), the major component of anchoring fibrils. OBJECTIVES To evaluate the efficacy and safety of intravenous (IV) gentamicin readthrough therapy in patients with RDEB harbouring nonsense mutations. The primary outcomes were increased expression of C7 in patients' skin and safety assessments (ototoxicity, nephrotoxicity, autoimmune response); secondary outcomes included measuring wound healing in target wounds and assessment by a validated Epidermolysis Bullosa Disease Activity and Scarring Index (EBDASI) scoring system. METHODS An open-label pilot trial to assess two different IV gentamicin regimens between August 2018 and March 2020 with follow-up through to 180 days post-treatment was carried out. Three patients with RDEB with confirmed nonsense mutations in COL7A1 in either one or two alleles and decreased baseline expression of C7 at the dermal-epidermal junction (DEJ) of their skin participated in the study. Three patients received gentamicin 7.5 mg kg-1 daily for 14 days and two of the three patients further received 7.5 mg kg-1 IV gentamicin twice weekly for 12 weeks. Patients who had pre-existing auditory or renal impairment, were currently using ototoxic or nephrotoxic medications, or had allergies to aminoglycosides or sulfate compounds were excluded. RESULTS After gentamicin treatment, skin biopsies from all three patients (age range 18-28 years) exhibited increased C7 in their DEJ. With both regimens, the new C7 persisted for at least 6 months post-treatment. At 1 and 3 months post-treatment, 100% of the monitored wounds exhibited > 85% closure. Both IV gentamicin infusion regimens decreased EBDASI total activity scores. Of the patients assessed with the EBDASI, all exhibited decreased total activity scores 3 months post-treatment. All three patients completed the study; no adverse effects or anti-C7 antibodies were detected. CONCLUSIONS IV gentamicin induced the readthrough of nonsense mutations in patients with RDEB and restored functional C7 in their skin, enhanced wound healing and improved clinical parameters. IV gentamicin may be a safe, efficacious, low-cost and readily available treatment for this population of patients with RDEB.
Collapse
Affiliation(s)
- David T Woodley
- Department of Dermatology, The Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Michelle Hao
- Department of Dermatology, The Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Andrew Kwong
- Department of Dermatology, The Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Brandon Levian
- Department of Dermatology, The Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Jon Cogan
- Department of Dermatology, The Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Yingping Hou
- Department of Dermatology, The Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Daniel Mosallaei
- Department of Dermatology, The Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Elana Kleinman
- Department of Dermatology, The Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Kate Zheng
- Department of Dermatology, The Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Claire Chung
- Department of Dermatology, The Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Gene Kim
- Department of Dermatology, The Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - David Peng
- Department of Dermatology, The Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Mei Chen
- Department of Dermatology, The Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| |
Collapse
|
5
|
du Rand A, Hunt J, Samson C, Loef E, Malhi C, Meidinger S, Chen CJ, Nutsford A, Taylor J, Dunbar R, Purvis D, Feisst V, Sheppard H. Highly efficient CRISPR/Cas9-mediated exon skipping for recessive dystrophic epidermolysis bullosa. Bioeng Transl Med 2024; 9:e10640. [PMID: 39036091 PMCID: PMC11256143 DOI: 10.1002/btm2.10640] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Revised: 12/05/2023] [Accepted: 12/13/2023] [Indexed: 07/23/2024] Open
Abstract
Gene therapy based on the CRISPR/Cas9 system has emerged as a promising strategy for treating the monogenic fragile skin disorder recessive dystrophic epidermolysis bullosa (RDEB). With this approach problematic wounds could be grafted with gene edited, patient-specific skin equivalents. Precise gene editing using homology-directed repair (HDR) is the ultimate goal, however low efficiencies have hindered progress. Reframing strategies based on highly efficient non-homologous end joining (NHEJ) repair aimed at excising dispensable, mutation-harboring exons offer a promising alternative approach for restoring the COL7A1 open reading frame. To this end, we employed an exon skipping strategy using dual single guide RNA (sgRNA)/Cas9 ribonucleoproteins (RNPs) targeted at three novel COL7A1 exons (31, 68, and 109) containing pathogenic heterozygous mutations, and achieved exon deletion rates of up to 95%. Deletion of exon 31 in both primary human RDEB keratinocytes and fibroblasts resulted in the restoration of type VII collagen (C7), leading to increased cellular adhesion in vitro and accurate C7 deposition at the dermal-epidermal junction in a 3D skin model. Taken together, we extend the list of COL7A1 exons amenable to therapeutic deletion. As an incidental finding, we find that long-read Nanopore sequencing detected large on-target structural variants comprised of deletions up to >5 kb at a frequency of ~10%. Although this frequency may be acceptable given the high rates of intended editing outcomes, our data demonstrate that standard short-read sequencing may underestimate the full range of unexpected Cas9-mediated editing events.
Collapse
Affiliation(s)
- Alex du Rand
- School of Biological SciencesThe University of AucklandAucklandNew Zealand
| | - John Hunt
- School of Biological SciencesThe University of AucklandAucklandNew Zealand
| | - Christopher Samson
- School of Biological SciencesThe University of AucklandAucklandNew Zealand
| | - Evert Loef
- School of Biological SciencesThe University of AucklandAucklandNew Zealand
| | - Chloe Malhi
- School of Biological SciencesThe University of AucklandAucklandNew Zealand
| | - Sarah Meidinger
- School of Biological SciencesThe University of AucklandAucklandNew Zealand
| | | | - Ashley Nutsford
- School of Biological SciencesThe University of AucklandAucklandNew Zealand
| | - John Taylor
- School of Biological SciencesThe University of AucklandAucklandNew Zealand
| | - Rod Dunbar
- School of Biological SciencesThe University of AucklandAucklandNew Zealand
| | - Diana Purvis
- Te Whatu Ora Health New ZealandTe Toka TumaiAucklandNew Zealand
| | - Vaughan Feisst
- School of Biological SciencesThe University of AucklandAucklandNew Zealand
| | - Hilary Sheppard
- School of Biological SciencesThe University of AucklandAucklandNew Zealand
| |
Collapse
|
6
|
Villavisanis DF, Perrault DP, Kiani SN, Cholok D, Fox PM. Current Treatment Landscape for Dystrophic Epidermolysis Bullosa: From Surgical Management to Emerging Gene Therapies and Novel Skin Grafts. J Hand Surg Am 2024; 49:472-480. [PMID: 38085193 DOI: 10.1016/j.jhsa.2023.10.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2023] [Revised: 08/27/2023] [Accepted: 10/16/2023] [Indexed: 05/05/2024]
Abstract
Epidermolysis bullosa is a genetic skin disorder characterized by blister formation from mechanical trauma. Dystrophic epidermolysis bullosa (DEB) is caused by mutations in the COL7A1 gene presenting as generalized blisters from birth, which can result in extensive scarring, alopecia, esophageal stenosis, corneal erosions, and nail dystrophy. This disease also often leads to pseudosyndactyly of the digits from the closure of webspaces, progressing to a "mitten hand" deformity. Although traditional and current treatment for DEB is largely supportive with wound care and iterative surgical pseudosyndactyly release, emerging gene therapies and novel skin grafts may offer promising treatment. Studies published in the early 2020s have used HSV-1 vectors expressing missing COL7A1 genes to restore collagen function. One of these treatments, B-VEC, is an HSV-1-based topical gene therapy designed to restore collagen 7 by delivering the COL7A1 gene, leveraging a differentiated HSV-1 vector platform that evades the patient's immune system response. Other work has been performed to retrovirally modify autologous keratinocytes, but limitations of this process include increased labor in harvesting and engineering autologous cells. This article provides an overview of DEB treatment with an emphasis on emerging gene therapies and novel skin grafts, especially as they pertain to pseudosyndactyly treatment.
Collapse
Affiliation(s)
- Dillan F Villavisanis
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Palo Alto, CA; Robert A Chase Hand and Upper Limb Center, Stanford University School of Medicine, Palo Alto, CA; Division of Plastic Surgery, Department of Surgery, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA
| | - David P Perrault
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Palo Alto, CA
| | - Sara N Kiani
- Department of Orthopedic Surgery, University of California San Francisco School of Medicine, San Francisco, CA
| | - David Cholok
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Palo Alto, CA
| | - Paige M Fox
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Palo Alto, CA; Robert A Chase Hand and Upper Limb Center, Stanford University School of Medicine, Palo Alto, CA.
| |
Collapse
|
7
|
AlMarshad FA, AlZahrani AM, Mahabbat NA, AlShammari EM, AlObaida SA, AlMalaq AA. Skin Allograft after Bone Marrow Transplantation of Patient with Recessive Dystrophic Epidermolysis Bullosa. PLASTIC AND RECONSTRUCTIVE SURGERY-GLOBAL OPEN 2023; 11:e5389. [PMID: 37954213 PMCID: PMC10635616 DOI: 10.1097/gox.0000000000005389] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Accepted: 09/15/2023] [Indexed: 11/14/2023]
Abstract
In this study, we present a 26-year-old woman with case presentation of recessive dystrophic epidermolysis bullosa who had developed squamous cell carcinoma. The patient underwent bone marrow transplant and skin grafting with the same bone marrow donor. After excision of squamous cell carcinoma and skin grafting, no tumor was observed; thus, chemotherapy and radiation were no longer needed.
Collapse
Affiliation(s)
- Felwa A. AlMarshad
- From the Plastic and Reconstructive Surgery Section, Department of Surgery, King Faisal Specialist Hospital & Research Centre, Riyadh, Saudi Arabia
| | - Abdullah M. AlZahrani
- From the Plastic and Reconstructive Surgery Section, Department of Surgery, King Faisal Specialist Hospital & Research Centre, Riyadh, Saudi Arabia
| | - Nehal A. Mahabbat
- From the Plastic and Reconstructive Surgery Section, Department of Surgery, King Faisal Specialist Hospital & Research Centre, Riyadh, Saudi Arabia
| | - Eman M. AlShammari
- From the Plastic and Reconstructive Surgery Section, Department of Surgery, King Faisal Specialist Hospital & Research Centre, Riyadh, Saudi Arabia
| | - Saud A. AlObaida
- Department of Dermatology, King Faisal Specialist Hospital & Research Centre, Riyadh, Saudi Arabia
| | - Ali A. AlMalaq
- From the Plastic and Reconstructive Surgery Section, Department of Surgery, King Faisal Specialist Hospital & Research Centre, Riyadh, Saudi Arabia
| |
Collapse
|
8
|
Hou PC, del Agua N, Lwin SM, Hsu CK, McGrath JA. Innovations in the Treatment of Dystrophic Epidermolysis Bullosa (DEB): Current Landscape and Prospects. Ther Clin Risk Manag 2023; 19:455-473. [PMID: 37337559 PMCID: PMC10277004 DOI: 10.2147/tcrm.s386923] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Accepted: 06/02/2023] [Indexed: 06/21/2023] Open
Abstract
Dystrophic epidermolysis bullosa (DEB) is one of the major types of EB, a rare hereditary group of trauma-induced blistering skin disorders. DEB is caused by inherited pathogenic variants in the COL7A1 gene, which encodes type VII collagen, the major component of anchoring fibrils which maintain adhesion between the outer epidermis and underlying dermis. DEB can be subclassified into dominant (DDEB) and recessive (RDEB) forms. Generally, DDEB has a milder phenotype, while RDEB patients often have more extensive blistering, chronic inflammation, skin fibrosis, and a propensity for squamous cell carcinoma development, collectively impacting on daily activities and life expectancy. At present, best practice treatments are mostly supportive, and thus there is a considerable burden of disease with unmet therapeutic need. Over the last 20 years, considerable translational research efforts have focused on either trying to cure DEB by direct correction of the COL7A1 gene pathology, or by modifying secondary inflammation to lessen phenotypic severity and improve patient symptoms such as poor wound healing, itch, and pain. In this review, we provide an overview and update on various therapeutic innovations for DEB, including gene therapy, cell-based therapy, protein therapy, and disease-modifying and symptomatic control agents. We outline the progress and challenges for each treatment modality and identify likely prospects for future clinical impact.
Collapse
Affiliation(s)
- Ping-Chen Hou
- Department of Dermatology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Nathalie del Agua
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan
- International Center for Wound Repair and Regeneration (iWRR), National Cheng Kung University, Tainan, Taiwan
| | - Su M Lwin
- St John’s Institute of Dermatology, School of Basic and Medical Biosciences, King’s College London, London, UK
| | - Chao-Kai Hsu
- Department of Dermatology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan
- International Center for Wound Repair and Regeneration (iWRR), National Cheng Kung University, Tainan, Taiwan
| | - John A McGrath
- Department of Dermatology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
- International Center for Wound Repair and Regeneration (iWRR), National Cheng Kung University, Tainan, Taiwan
- St John’s Institute of Dermatology, School of Basic and Medical Biosciences, King’s College London, London, UK
| |
Collapse
|
9
|
De Gregorio C, Catalán E, Garrido G, Morandé P, Bennett JC, Muñoz C, Cofré G, Huang YL, Cuadra B, Murgas P, Calvo M, Altermatt F, Yubero MJ, Palisson F, South AP, Ezquer M, Fuentes I. Maintenance of chronicity signatures in fibroblasts isolated from recessive dystrophic epidermolysis bullosa chronic wound dressings under culture conditions. Biol Res 2023; 56:23. [PMID: 37161592 PMCID: PMC10170710 DOI: 10.1186/s40659-023-00437-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Accepted: 04/27/2023] [Indexed: 05/11/2023] Open
Abstract
BACKGROUND Recessive Dystrophic Epidermolysis Bullosa (RDEB) is a rare inherited skin disease caused by variants in the COL7A1 gene, coding for type VII collagen (C7), an important component of anchoring fibrils in the basement membrane of the epidermis. RDEB patients suffer from skin fragility starting with blister formation and evolving into chronic wounds, inflammation and skin fibrosis, with a high risk of developing aggressive skin carcinomas. Restricted therapeutic options are limited by the lack of in vitro models of defective wound healing in RDEB patients. RESULTS In order to explore a more efficient, non-invasive in vitro model for RDEB studies, we obtained patient fibroblasts derived from discarded dressings) and examined their phenotypic features compared with fibroblasts derived from non-injured skin of RDEB and healthy-donor skin biopsies. Our results demonstrate that fibroblasts derived from RDEB chronic wounds (RDEB-CW) displayed characteristics of senescent cells, increased myofibroblast differentiation, and augmented levels of TGF-β1 signaling components compared to fibroblasts derived from RDEB acute wounds and unaffected RDEB skin as well as skin from healthy-donors. Furthermore, RDEB-CW fibroblasts exhibited an increased pattern of inflammatory cytokine secretion (IL-1β and IL-6) when compared with RDEB and control fibroblasts. Interestingly, these aberrant patterns were found specifically in RDEB-CW fibroblasts independent of the culturing method, since fibroblasts obtained from dressing of acute wounds displayed a phenotype more similar to fibroblasts obtained from RDEB normal skin biopsies. CONCLUSIONS Our results show that in vitro cultured RDEB-CW fibroblasts maintain distinctive cellular and molecular characteristics resembling the inflammatory and fibrotic microenvironment observed in RDEB patients' chronic wounds. This work describes a novel, non-invasive and painless strategy to obtain human fibroblasts chronically subjected to an inflammatory and fibrotic environment, supporting their use as an accessible model for in vitro studies of RDEB wound healing pathogenesis. As such, this approach is well suited to testing new therapeutic strategies under controlled laboratory conditions.
Collapse
Affiliation(s)
- Cristian De Gregorio
- Centro de Medicina Regenerativa, Facultad de Medicina Clínica Alemana, Universidad del Desarrollo, Santiago, 7610658, Chile
| | - Evelyng Catalán
- DEBRA Chile, Francisco de Villagra 392, Ñuñoa, Santiago, Chile
| | - Gabriel Garrido
- DEBRA Chile, Francisco de Villagra 392, Ñuñoa, Santiago, Chile
| | - Pilar Morandé
- DEBRA Chile, Francisco de Villagra 392, Ñuñoa, Santiago, Chile
| | | | - Catalina Muñoz
- DEBRA Chile, Francisco de Villagra 392, Ñuñoa, Santiago, Chile
| | - Glenda Cofré
- DEBRA Chile, Francisco de Villagra 392, Ñuñoa, Santiago, Chile
| | - Ya-Lin Huang
- Centro de Medicina Regenerativa, Facultad de Medicina Clínica Alemana, Universidad del Desarrollo, Santiago, 7610658, Chile
| | - Bárbara Cuadra
- Centro de Medicina Regenerativa, Facultad de Medicina Clínica Alemana, Universidad del Desarrollo, Santiago, 7610658, Chile
| | - Paola Murgas
- Instituto de Bioquímica y Microbiología, Facultad de Ciencias, Universidad Austral de Chile, Valdivia, Chile
| | - Margarita Calvo
- Facultad de Ciencias Biológicas y División de Anestesiología, Escuela de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
- Núcleo milenio para el estudio del dolor MINUSPAIN, Santiago, Chile
| | - Fernando Altermatt
- División de Anestesiología, Escuela de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - María Joao Yubero
- DEBRA Chile, Francisco de Villagra 392, Ñuñoa, Santiago, Chile
- Pediatrics and Pediatric Infectious Diseases of Clínica Alemana, Facultad de Medicina Alemana, Universidad del Desarrollo, Santiago, Chile
| | - Francis Palisson
- DEBRA Chile, Francisco de Villagra 392, Ñuñoa, Santiago, Chile
- Servicio de Dermatología, Facultad de Medicina Clínica Alemana, Universidad del Desarrollo, Santiago, Chile
| | - Andrew P South
- Department of Dermatology & Cutaneous Biology, Thomas Jefferson University, Philadelphia, USA
| | - Marcelo Ezquer
- Centro de Medicina Regenerativa, Facultad de Medicina Clínica Alemana, Universidad del Desarrollo, Santiago, 7610658, Chile.
| | - Ignacia Fuentes
- DEBRA Chile, Francisco de Villagra 392, Ñuñoa, Santiago, Chile.
- Centro de Genética y Genómica, Facultad de Medicina Clínica Alemana, Universidad del Desarrollo, Santiago, 7610658, Chile.
- Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile.
| |
Collapse
|
10
|
Sheriff A, Guri I, Zebrowska P, Llopis-Hernandez V, Brooks IR, Tekkela S, Subramaniam K, Gebrezgabher R, Naso G, Petrova A, Balon K, Onoufriadis A, Kujawa D, Kotulska M, Newby G, Łaczmański Ł, Liu DR, McGrath JA, Jacków J. ABE8e adenine base editor precisely and efficiently corrects a recurrent COL7A1 nonsense mutation. Sci Rep 2022; 12:19643. [PMID: 36385635 PMCID: PMC9666996 DOI: 10.1038/s41598-022-24184-8] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Accepted: 11/11/2022] [Indexed: 11/17/2022] Open
Abstract
Base editing introduces precise single-nucleotide edits in genomic DNA and has the potential to treat genetic diseases such as the blistering skin disease recessive dystrophic epidermolysis bullosa (RDEB), which is characterized by mutations in the COL7A1 gene and type VII collagen (C7) deficiency. Adenine base editors (ABEs) convert A-T base pairs to G-C base pairs without requiring double-stranded DNA breaks or donor DNA templates. Here, we use ABE8e, a recently evolved ABE, to correct primary RDEB patient fibroblasts harboring the recurrent RDEB nonsense mutation c.5047 C > T (p.Arg1683Ter) in exon 54 of COL7A1 and use a next generation sequencing workflow to interrogate post-treatment outcomes. Electroporation of ABE8e mRNA into a bulk population of RDEB patient fibroblasts resulted in remarkably efficient (94.6%) correction of the pathogenic allele, restoring COL7A1 mRNA and expression of C7 protein in western blots and in 3D skin constructs. Off-target DNA analysis did not detect off-target editing in treated patient-derived fibroblasts and there was no detectable increase in A-to-I changes in the RNA. Taken together, we have established a highly efficient pipeline for gene correction in primary fibroblasts with a favorable safety profile. This work lays a foundation for developing therapies for RDEB patients using ex vivo or in vivo base editing strategies.
Collapse
Affiliation(s)
- Adam Sheriff
- St John's Institute of Dermatology, Faculty of Life Sciences and Medicine, King's College London, 9th Floor Tower Wing, Guy's Hospital, Great Maze Pond Road, London, SE1 9RT, UK
| | - Ina Guri
- St John's Institute of Dermatology, Faculty of Life Sciences and Medicine, King's College London, 9th Floor Tower Wing, Guy's Hospital, Great Maze Pond Road, London, SE1 9RT, UK
| | - Paulina Zebrowska
- Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wroclaw, Poland
| | - Virginia Llopis-Hernandez
- St John's Institute of Dermatology, Faculty of Life Sciences and Medicine, King's College London, 9th Floor Tower Wing, Guy's Hospital, Great Maze Pond Road, London, SE1 9RT, UK
| | - Imogen R Brooks
- St John's Institute of Dermatology, Faculty of Life Sciences and Medicine, King's College London, 9th Floor Tower Wing, Guy's Hospital, Great Maze Pond Road, London, SE1 9RT, UK
| | - Stavroula Tekkela
- St John's Institute of Dermatology, Faculty of Life Sciences and Medicine, King's College London, 9th Floor Tower Wing, Guy's Hospital, Great Maze Pond Road, London, SE1 9RT, UK
| | - Kavita Subramaniam
- St John's Institute of Dermatology, Faculty of Life Sciences and Medicine, King's College London, 9th Floor Tower Wing, Guy's Hospital, Great Maze Pond Road, London, SE1 9RT, UK
| | - Ruta Gebrezgabher
- St John's Institute of Dermatology, Faculty of Life Sciences and Medicine, King's College London, 9th Floor Tower Wing, Guy's Hospital, Great Maze Pond Road, London, SE1 9RT, UK
| | - Gaetano Naso
- Molecular and Cellular Immunology Unit, UCL GOS Institute of Child Health, London, UK
| | - Anastasia Petrova
- Molecular and Cellular Immunology Unit, UCL GOS Institute of Child Health, London, UK
| | - Katarzyna Balon
- Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wroclaw, Poland
| | - Alexandros Onoufriadis
- St John's Institute of Dermatology, Faculty of Life Sciences and Medicine, King's College London, 9th Floor Tower Wing, Guy's Hospital, Great Maze Pond Road, London, SE1 9RT, UK
| | - Dorota Kujawa
- Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wroclaw, Poland
| | - Martyna Kotulska
- Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wroclaw, Poland
| | - Gregory Newby
- Merkin Institute of Transformative Technologies in Healthcare, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA, USA
- Howard Hughes Medical Institute, Harvard University, Cambridge, MA, USA
| | - Łukasz Łaczmański
- Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wroclaw, Poland
| | - David R Liu
- Merkin Institute of Transformative Technologies in Healthcare, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA, USA
- Howard Hughes Medical Institute, Harvard University, Cambridge, MA, USA
| | - John A McGrath
- St John's Institute of Dermatology, Faculty of Life Sciences and Medicine, King's College London, 9th Floor Tower Wing, Guy's Hospital, Great Maze Pond Road, London, SE1 9RT, UK
| | - Joanna Jacków
- St John's Institute of Dermatology, Faculty of Life Sciences and Medicine, King's College London, 9th Floor Tower Wing, Guy's Hospital, Great Maze Pond Road, London, SE1 9RT, UK.
| |
Collapse
|
11
|
Hong SA, Kim SE, Lee AY, Hwang GH, Kim JH, Iwata H, Kim SC, Bae S, Lee SE. Therapeutic base editing and prime editing of COL7A1 mutations in recessive dystrophic epidermolysis bullosa. Mol Ther 2022; 30:2664-2679. [PMID: 35690907 PMCID: PMC9372317 DOI: 10.1016/j.ymthe.2022.06.005] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2021] [Revised: 05/09/2022] [Accepted: 06/06/2022] [Indexed: 12/17/2022] Open
Abstract
Recessive dystrophic epidermolysis bullosa (RDEB) is a severe skin fragility disorder caused by loss-of-function mutations in the COL7A1 gene, which encodes type VII collagen (C7), a protein that functions in skin adherence. From 36 Korean RDEB patients, we identified a total of 69 pathogenic mutations (40 variants without recurrence), including point mutations (72.5%) and insertion/deletion mutations (27.5%). For fibroblasts from two patients (Pat1 and Pat2), we applied adenine base editors (ABEs) to correct the pathogenic mutation of COL7A1 or to bypass a premature stop codon in Pat1-derived primary fibroblasts. To expand the targeting scope, we also utilized prime editors (PEs) to correct the COL7A1 mutations in Pat1- and Pat2-derived fibroblasts. Ultimately, we found that transfer of edited patient-derived skin equivalents (i.e., RDEB keratinocytes and PE-corrected RDEB fibroblasts from the RDEB patient) into the skin of immunodeficient mice led to C7 deposition and anchoring fibril formation within the dermal-epidermal junction, suggesting that base editing and prime editing could be feasible strategies for ex vivo gene editing to treat RDEB.
Collapse
Affiliation(s)
- Sung-Ah Hong
- Genomic Medicine Institute, Medical Research Center, Seoul National University College of Medicine, Seoul 03080, South Korea
| | - Song-Ee Kim
- Department of Dermatology, Gangnam Severance Hospital, Cutaneous Biology Research Institute, Yonsei University College of Medicine, Seoul 06273, South Korea
| | - A-Young Lee
- Department of Dermatology, Gangnam Severance Hospital, Cutaneous Biology Research Institute, Yonsei University College of Medicine, Seoul 06273, South Korea
| | - Gue-Ho Hwang
- Department of Chemistry, Hanyang University, Seoul 04763, South Korea
| | - Jong Hoon Kim
- Department of Dermatology, Gangnam Severance Hospital, Cutaneous Biology Research Institute, Yonsei University College of Medicine, Seoul 06273, South Korea
| | - Hiroaki Iwata
- Department of Dermatology, Hokkaido University Graduate School of Medicine, Sapporo 060-8638, Japan
| | - Soo-Chan Kim
- Department of Dermatology, Yongin Severance Hospital, Yonsei University College of Medicine, Yongin 16995, South Korea
| | - Sangsu Bae
- Genomic Medicine Institute, Medical Research Center, Seoul National University College of Medicine, Seoul 03080, South Korea; Department of Biomedical Sciences, Department of Biochemistry and Molecular Biology, Seoul National University College of Medicine, Seoul 03080, South Korea.
| | - Sang Eun Lee
- Department of Dermatology, Gangnam Severance Hospital, Cutaneous Biology Research Institute, Yonsei University College of Medicine, Seoul 06273, South Korea.
| |
Collapse
|
12
|
Natsuga K, Shinkuma S, Hsu CK, Fujita Y, Ishiko A, Tamai K, McGrath JA. Current topics in Epidermolysis bullosa: Pathophysiology and therapeutic challenges. J Dermatol Sci 2021; 104:164-176. [PMID: 34916041 DOI: 10.1016/j.jdermsci.2021.11.004] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Accepted: 11/06/2021] [Indexed: 12/14/2022]
Abstract
Epidermolysis bullosa (EB) is a group of inherited skin and mucosal fragility disorders resulting from mutations in genes encoding basement membrane zone (BMZ) components or proteins that maintain the integrity of BMZ and adjacent keratinocytes. More than 30 years have passed since the first causative gene for EB was identified, and over 40 genes are now known to be responsible for the protean collection of mechanobullous diseases included under the umbrella term of EB. Through the elucidation of disease mechanisms using human skin samples, animal models, and cultured cells, we have now reached the stage of developing more effective therapeutics for EB. This review will initially focus on what is known about blister wound healing in EB, since recent and emerging basic science data are very relevant to clinical translation and therapeutic strategies for patients. We then place these studies in the context of the latest information on gene therapy, read-through therapy, and cell therapy that provide optimism for improved clinical management of people living with EB.
Collapse
Affiliation(s)
- Ken Natsuga
- Department of Dermatology, Hokkaido University Faculty of Medicine and Graduate School of Medicine, Hokkaido, Japan.
| | - Satoru Shinkuma
- Department of Dermatology, Nara Medical University School of Medicine, Kashihara, Japan
| | - Chao-Kai Hsu
- Department of Dermatology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan; Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan; International Center for Wound Repair and Regeneration (iWRR), National Cheng Kung University, Tainan, Taiwan
| | - Yasuyuki Fujita
- Department of Dermatology, Hokkaido University Faculty of Medicine and Graduate School of Medicine, Hokkaido, Japan; Department of Dermatology, Sapporo City General Hospital, Sapporo, Japan
| | - Akira Ishiko
- Department of Dermatology, Toho University School of Medicine, Tokyo, Japan
| | - Katsuto Tamai
- Department of Stem Cell Therapy Science, Graduate School of Medicine, Osaka University, Suita, Japan
| | - John A McGrath
- St. John's Institute of Dermatology, School of Basic and Medical Biosciences, King's College London, London, United Kingdom
| |
Collapse
|
13
|
Subramaniam KS, Antoniou MN, McGrath JA, Lwin SM. The potential of gene therapy for recessive dystrophic epidermolysis bullosa. Br J Dermatol 2021; 186:609-619. [PMID: 34862606 DOI: 10.1111/bjd.20910] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Revised: 11/12/2021] [Accepted: 11/28/2021] [Indexed: 11/30/2022]
Abstract
Epidermolysis bullosa (EB) encompasses a heterogeneous group of inherited skin fragility disorders with mutations in genes encoding the basement membrane zone (BMZ) proteins that normally ensure dermal-epidermal integrity. Of the four main EB types, recessive dystrophic EB (RDEB), especially the severe variant, represents one of the most debilitating clinical entities with recurrent mucocutaneous blistering and ulceration leading to chronic wounds, infections, inflammation, scarring and ultimately cutaneous squamous cell carcinoma, which leads to premature death. Improved understanding of the molecular genetics of EB over the past three decades and advances in biotechnology has led to rapid progress in developing gene and cell-based regenerative therapies for EB. In particular, RDEB is at the vanguard of advances in human clinical trials of advanced therapeutics. Furthermore, the past decade has witnessed the emergence of a real collective, global effort involving academia and industry, supported by international EB patient organisations such as the Dystrophic Epidermolysis Bullosa Research Association (DEBRA), amongst others, to develop clinically relevant and marketable targeted therapeutics for EB. Thus, there is an increasing need for the practising dermatologist to become familiar with the concept of gene therapy, fundamental differences between various approaches and their human applications. This review explains the principles of different approaches of gene therapy; summarises its journey and discusses its current and future impact in RDEB.
Collapse
Affiliation(s)
- K S Subramaniam
- Genetic Skin Diseases Group, St John's Institute of Dermatology, King's College London, Guy's Hospital, London, UK
| | - M N Antoniou
- Gene Expression and Therapy Group, Department of Medical & Molecular Genetics, King's College London, Guy's Hospital, London, UK
| | - J A McGrath
- Genetic Skin Diseases Group, St John's Institute of Dermatology, King's College London, Guy's Hospital, London, UK
| | - S M Lwin
- Genetic Skin Diseases Group, St John's Institute of Dermatology, King's College London, Guy's Hospital, London, UK
| |
Collapse
|
14
|
Woodley DT, Cogan J, Mosallaei D, Yim K, Chen M. Characterization of mutant type VII collagens underlying the inversa subtype of recessive dystrophic epidermolysis bullosa. J Dermatol Sci 2021; 104:104-111. [PMID: 34674926 DOI: 10.1016/j.jdermsci.2021.09.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 08/26/2021] [Accepted: 09/20/2021] [Indexed: 10/20/2022]
Abstract
BACKGROUND Patients with recessive dystrophic epidermolysis bullosa (RDEB) lack functional type VII collagen (C7) leading to skin fragility, bullae, and erosive wounds. RDEB-Inversa (RDEB-I), a subset of RDEB, is characterized by lesions localized to body areas with higher skin temperatures such as flexures and skin folds. OBJECTIVE We aimed to determine if C7 derived from RDEB-I mutations had structural and functional aberrancies that were temperature sensitive and could be reversed by lowering the temperature. METHODS In this study, we generated 12 substitution mutations associated with RDEB-I via site-directed mutagenesis and purified recombinant C7 protein. These C7 mutants were evaluated for structural parameters (trimer formation and protease sensitivity) and the ability to promote keratinocyte migration at 37 °C (the temperature of skin folds) and 30 °C (the maximum skin temperature of arms and legs). Fibroblasts derived from RDEB-I patients were evaluated for C7 secretion and cellular migration at both temperatures. RESULTS C7s from RDEB-I mutations exhibited decreased thermal stability, increased sensitivity to protease digestion, diminished formation of collagen trimers, and reduced ability to promote keratinocyte migration compared with wild-type C7. In addition, fibroblasts derived from RDEB-I patients demonstrated intracellular accumulation of C7 and abnormal cell migration at 37 °C. All of these aberrancies were corrected by reducing the temperature to 30 °C. C7s generated from severe-RDEB mutations (non-Inversa) did not display temperature-dependent perturbations. CONCLUSION These data demonstrate that RDEB-I mutations generate C7 aberrancies that are temperature dependent. This may explain why RDEB-I patients develop clinical lesions in areas where their skin is considerably warmer.
Collapse
Affiliation(s)
- David T Woodley
- Department of Dermatology, The Keck School of Medicine, University of Southern California, Los Angeles, USA
| | - Jon Cogan
- Department of Dermatology, The Keck School of Medicine, University of Southern California, Los Angeles, USA
| | - Daniel Mosallaei
- Department of Dermatology, The Keck School of Medicine, University of Southern California, Los Angeles, USA
| | - Kaitlyn Yim
- Department of Dermatology, The Keck School of Medicine, University of Southern California, Los Angeles, USA
| | - Mei Chen
- Department of Dermatology, The Keck School of Medicine, University of Southern California, Los Angeles, USA.
| |
Collapse
|
15
|
Li J, Chen Y, Tiwari M, Bansal V, Sen GL. Regulation of integrin and extracellular matrix genes by HNRNPL is necessary for epidermal renewal. PLoS Biol 2021; 19:e3001378. [PMID: 34543262 PMCID: PMC8452081 DOI: 10.1371/journal.pbio.3001378] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Accepted: 07/30/2021] [Indexed: 01/05/2023] Open
Abstract
Stratified epithelia such as the epidermis require coordinated regulation of stem and progenitor cell proliferation, survival, and differentiation to maintain homeostasis. Integrin-mediated anchorage of the basal layer stem cells of the epidermis to the underlying dermis through extracellular matrix (ECM) proteins is crucial for this process. It is currently unknown how the expression of these integrins and ECM genes are regulated. Here, we show that the RNA-binding protein (RBP) heterogeneous nuclear ribonucleoprotein L (HNRNPL) binds to these genes on chromatin to promote their expression. HNRNPL recruits RNA polymerase II (Pol II) to integrin/ECM genes and is required for stabilizing Pol II transcription through those genes. In the absence of HNRNPL, the basal layer of the epidermis where the stem cells reside prematurely differentiates and detaches from the underlying dermis due to diminished integrin/ECM expression. Our results demonstrate a critical role for RBPs on chromatin to maintain stem and progenitor cell fate by dictating the expression of specific classes of genes.
Collapse
Affiliation(s)
- Jingting Li
- Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Yifang Chen
- Department of Dermatology, Department of Cellular and Molecular Medicine, UCSD Stem Cell Program, University of California San Diego, La Jolla, California, United States of America
| | - Manisha Tiwari
- Department of Dermatology, Department of Cellular and Molecular Medicine, UCSD Stem Cell Program, University of California San Diego, La Jolla, California, United States of America
| | - Varun Bansal
- Department of Dermatology, Department of Cellular and Molecular Medicine, UCSD Stem Cell Program, University of California San Diego, La Jolla, California, United States of America
| | - George L. Sen
- Department of Dermatology, Department of Cellular and Molecular Medicine, UCSD Stem Cell Program, University of California San Diego, La Jolla, California, United States of America
| |
Collapse
|
16
|
Review of the Latest Methods of Epidermolysis Bullosa and Other Chronic Wounds Treatment Including BIOOPA Dressing. Dermatol Ther (Heidelb) 2021; 11:1469-1480. [PMID: 34269966 PMCID: PMC8484405 DOI: 10.1007/s13555-021-00578-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Indexed: 11/20/2022] Open
Abstract
Epidermolysis bullosa (EB) is a hereditary genetic skin disorder, classified as a type of genodermatosis, which causes severe, chronic skin blisters associated with painful and potentially life-threatening complications. Currently, there is no effective therapy or cure for EB. However, over the past decade, there have been several important advances in treatment methods, which are now approaching clinical application, including gene therapy, protein replacement therapy, cell therapy (allogeneic fibroblasts, mesenchymal stromal cells), bone marrow stem cell transplant, culture/vaccination of revertant mosaic keratinocytes, gene editing/engineering, and the clinical application of inducible pluripotent stem cells. Tissue engineering scientists are developing materials that mimic the structure and natural healing process to promote skin reconstruction in the event of an incurable injury. Although a cure for EB remains elusive, recent data from animal models and preliminary human clinical trials have raised the expectations of patients, clinicians, and researchers, where modifying the disease and improving patients’ quality of life are now considered attainable goals. In addition, the lessons learned from the treatment of EB may improve the treatment of other genetic diseases.
Collapse
|
17
|
Fujimura Y, Watanabe M, Ohno K, Kobayashi Y, Takashima S, Nakamura H, Kosumi H, Wang Y, Mai Y, Lauria A, Proserpio V, Ujiie H, Iwata H, Nishie W, Nagayama M, Oliviero S, Donati G, Shimizu H, Natsuga K. Hair follicle stem cell progeny heal blisters while pausing skin development. EMBO Rep 2021; 22:e50882. [PMID: 34085753 DOI: 10.15252/embr.202050882] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Revised: 04/30/2021] [Accepted: 05/05/2021] [Indexed: 12/13/2022] Open
Abstract
Injury in adult tissue generally reactivates developmental programs to foster regeneration, but it is not known whether this paradigm applies to growing tissue. Here, by employing blisters, we show that epidermal wounds heal at the expense of skin development. The regenerated epidermis suppresses the expression of tissue morphogenesis genes accompanied by delayed hair follicle (HF) growth. Lineage tracing experiments, cell proliferation dynamics, and mathematical modeling reveal that the progeny of HF junctional zone stem cells, which undergo a morphological transformation, repair the blisters while not promoting HF development. In contrast, the contribution of interfollicular stem cell progeny to blister healing is small. These findings demonstrate that HF development can be sacrificed for the sake of epidermal wound regeneration. Our study elucidates the key cellular mechanism of wound healing in skin blistering diseases.
Collapse
Affiliation(s)
- Yu Fujimura
- Department of Dermatology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Mika Watanabe
- Department of Dermatology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan.,Department of Life Sciences and Systems Biology, Molecular Biotechnology Centre, University of Turin, Turin, Italy
| | - Kota Ohno
- Research Institute for Electronic Science, Hokkaido University, Sapporo, Japan
| | - Yasuaki Kobayashi
- Research Institute for Electronic Science, Hokkaido University, Sapporo, Japan
| | - Shota Takashima
- Department of Dermatology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Hideki Nakamura
- Department of Dermatology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Hideyuki Kosumi
- Department of Dermatology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Yunan Wang
- Department of Dermatology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Yosuke Mai
- Department of Dermatology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Andrea Lauria
- Department of Life Sciences and Systems Biology, Molecular Biotechnology Centre, University of Turin, Turin, Italy.,Italian Institute for Genomic Medicine, Candiolo, Italy
| | - Valentina Proserpio
- Italian Institute for Genomic Medicine, Candiolo, Italy.,Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Italy
| | - Hideyuki Ujiie
- Department of Dermatology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Hiroaki Iwata
- Department of Dermatology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Wataru Nishie
- Department of Dermatology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Masaharu Nagayama
- Research Institute for Electronic Science, Hokkaido University, Sapporo, Japan
| | - Salvatore Oliviero
- Department of Life Sciences and Systems Biology, Molecular Biotechnology Centre, University of Turin, Turin, Italy.,Italian Institute for Genomic Medicine, Candiolo, Italy
| | - Giacomo Donati
- Department of Life Sciences and Systems Biology, Molecular Biotechnology Centre, University of Turin, Turin, Italy
| | - Hiroshi Shimizu
- Department of Dermatology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Ken Natsuga
- Department of Dermatology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| |
Collapse
|
18
|
Naso G, Petrova A. Cellular therapy options for genetic skin disorders with a focus on recessive dystrophic epidermolysis bullosa. Br Med Bull 2020; 136:30-45. [PMID: 32888294 DOI: 10.1093/bmb/ldaa029] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Revised: 07/20/2020] [Accepted: 08/04/2020] [Indexed: 12/21/2022]
Abstract
INTRODUCTION Combinatorial cell and gene therapies for life-threatening inherited skin disorders have shown tremendous potential for preclinical and clinical implementation with significant progress made for recessive dystrophic epidermolysis bullosa (RDEB). To date, various cell lineages including resident skin cells and adult stem cells have been investigated for gene and cell therapy for RDEB reaching the clinical trial stage. SOURCES OF DATA Sources of data are key recent literature, ClinicalTrials.gov, Clinicaltrialsregister.eu and pharma press releases. AREAS OF AGREEMENT Cell-based gene transfer using autologous patients' cells has demonstrated positive outcomes in preclinical and clinical trials and highlighted the importance of targeting resident skin stem cells to achieve a meaningful long-term effect. Additionally, adult stem cells, such as mesenchymal stromal cells, have the potential to ameliorate systemic manifestations of the disease. AREAS OF CONTROVERSY While proven safe, the clinical trials of localized treatment have reported only modest and transient improvements. On the other hand, the risks associated with systemic therapies remain high and should be carefully weighed against the potential benefits. It is unclear to what extent adult stem cells can contribute to skin regeneration/wound healing. GROWING POINTS Further research is warranted in order to fulfil the potential of cellular therapies for RDEB. The development of combinatorial gene and cell-based approaches is required to achieve long-term clinical benefits. AREAS TIMELY FOR DEVELOPING RESEARCH Induced pluripotent stem cells can potentially provide a valuable source of autologous patient material for cellular therapies. In addition, recent advances in the field of gene editing can overcome hurdles associated with conventional gene addition approaches. DATA AVAILABILITY STATEMENT No new data were generated or analysed in support of this review.
Collapse
Affiliation(s)
- Gaetano Naso
- Molecular and Cellular Immunology Section, UCL Great Ormond Street Institute of Child Health, 30 Guilford street, London WC1N 1EH, UK
| | - Anastasia Petrova
- Molecular and Cellular Immunology Section, UCL Great Ormond Street Institute of Child Health, 30 Guilford street, London WC1N 1EH, UK
| |
Collapse
|
19
|
Titeux M, Bonnet des Claustres M, Izmiryan A, Ragot H, Hovnanian A. Emerging drugs for the treatment of epidermolysis bullosa. Expert Opin Emerg Drugs 2020; 25:467-489. [DOI: 10.1080/14728214.2020.1839049] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Affiliation(s)
- Matthias Titeux
- Imagine Institute, Laboratory of Genetic Skin Diseases, INSERM UMR 1163, Université de Paris, Paris, France
| | | | - Araksya Izmiryan
- Imagine Institute, Laboratory of Genetic Skin Diseases, INSERM UMR 1163, Université de Paris, Paris, France
| | - Helene Ragot
- Imagine Institute, Laboratory of Genetic Skin Diseases, INSERM UMR 1163, Université de Paris, Paris, France
| | - Alain Hovnanian
- Imagine Institute, Laboratory of Genetic Skin Diseases, INSERM UMR 1163, Université de Paris, Paris, France
- Départment de Génétique, AP-HP, Hôpital Necker-Enfants Malades, Paris, France
| |
Collapse
|
20
|
Lincoln V, Cogan J, Hou Y, Hirsch M, Hao M, Alexeev V, De Luca M, De Rosa L, Bauer JW, Woodley DT, Chen M. Gentamicin induces LAMB3 nonsense mutation readthrough and restores functional laminin 332 in junctional epidermolysis bullosa. Proc Natl Acad Sci U S A 2018; 115:E6536-E6545. [PMID: 29946029 PMCID: PMC6048497 DOI: 10.1073/pnas.1803154115] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Herlitz junctional epidermolysis bullosa (H-JEB) is an incurable, devastating, and mostly fatal inherited skin disease for which there is only supportive care. H-JEB is caused by loss-of-function mutations in LAMA3, LAMB3, or LAMC2, leading to complete loss of laminin 332, the major component of anchoring filaments, which mediate epidermal-dermal adherence. LAMB3 (laminin β3) mutations account for 80% of patients with H-JEB, and ∼95% of H-JEB-associated LAMB3 mutations are nonsense mutations leading to premature termination codons (PTCs). In this study, we evaluated the ability of gentamicin to induce PTC readthrough in H-JEB laminin β3-null keratinocytes transfected with expression vectors encoding eight different LAMB3 nonsense mutations. We found that gentamicin induced PTC readthrough in all eight nonsense mutations tested. We next used lentiviral vectors to generate stably transduced H-JEB cells with the R635X and C290X nonsense mutations. Incubation of these cell lines with various concentrations of gentamicin resulted in the synthesis and secretion of full-length laminin β3 in a dose-dependent and sustained manner. Importantly, the gentamicin-induced laminin β3 led to the restoration of laminin 332 assembly, secretion, and deposition within the dermal/epidermal junction, as well as proper polarization of α6β4 integrin in basal keratinocytes, as assessed by immunoblot analysis, immunofluorescent microscopy, and an in vitro 3D skin equivalent model. Finally, newly restored laminin 332 corrected the abnormal cellular phenotype of H-JEB cells by reversing abnormal cell morphology, poor growth potential, poor cell-substratum adhesion, and hypermotility. Therefore, gentamicin may offer a therapy for H-JEB and other inherited skin diseases caused by PTC mutations.
Collapse
Affiliation(s)
- Vadim Lincoln
- Department of Dermatology, The Keck School of Medicine of University of Southern California, Los Angeles, CA 90033
| | - Jon Cogan
- Department of Dermatology, The Keck School of Medicine of University of Southern California, Los Angeles, CA 90033
| | - Yingping Hou
- Department of Dermatology, The Keck School of Medicine of University of Southern California, Los Angeles, CA 90033
| | - Michaela Hirsch
- Department of Dermatology, The Keck School of Medicine of University of Southern California, Los Angeles, CA 90033
| | - Michelle Hao
- Department of Dermatology, The Keck School of Medicine of University of Southern California, Los Angeles, CA 90033
| | - Vitali Alexeev
- Department of Dermatology and Cutaneous Biology, Sidney Kimmel Medical College, Jefferson Institute of Molecular Medicine, Thomas Jefferson University, Philadelphia, PA 19107
| | - Michele De Luca
- Center for Regenerative Medicine "Stefano Ferrari," Department of Life Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy
| | - Laura De Rosa
- Center for Regenerative Medicine "Stefano Ferrari," Department of Life Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy
| | - Johann W Bauer
- EB House Austria and Department of Dermatology, University Hospital of the Paracelsus Medical University, 5020 Salzburg, Austria
| | - David T Woodley
- Department of Dermatology, The Keck School of Medicine of University of Southern California, Los Angeles, CA 90033
| | - Mei Chen
- Department of Dermatology, The Keck School of Medicine of University of Southern California, Los Angeles, CA 90033;
| |
Collapse
|
21
|
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: 7.7] [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
|
22
|
Peking P, Koller U, Murauer EM. Functional therapies for cutaneous wound repair in epidermolysis bullosa. Adv Drug Deliv Rev 2018; 129:330-343. [PMID: 29248480 DOI: 10.1016/j.addr.2017.12.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2017] [Revised: 11/07/2017] [Accepted: 12/09/2017] [Indexed: 12/20/2022]
Abstract
Chronic wounding as a result of recurrent skin blistering in the painful genetic skin disease epidermolysis bullosa, may lead to life-threatening infections, increased risk of tumor formation, and other serious medical complications. Therefore, epidermolysis bullosa patients have an urgent need for optimal wound care and tissue regeneration. Therapeutic strategies using gene-, protein-, and cell-therapies are being developed to improve clinical symptoms, and some of them have already been investigated in early clinical trials. The most favorable options of functional therapies include gene replacement, gene editing, RNA targeting, and harnessing natural gene therapy. This review describes the current progress of the different approaches targeting autologous skin cells, and will discuss the benefits and challenges of their application.
Collapse
|
23
|
Skin-Derived Stem Cells for Wound Treatment Using Cultured Epidermal Autografts: Clinical Applications and Challenges. Stem Cells Int 2018; 2018:4623615. [PMID: 29765411 PMCID: PMC5889868 DOI: 10.1155/2018/4623615] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2017] [Accepted: 01/03/2018] [Indexed: 12/15/2022] Open
Abstract
The human skin fulfills important barrier, sensory, and immune functions-all of which contribute significantly to health and organism integrity. Widespread skin damage requires immediate treatment and coverage because massive skin loss fosters the invasion of pathogens, causes critical fluid loss, and may ultimately lead to death. Since the skin is a highly immunocompetent organ, autologous transplants are the only viable approach to permanently close a widespread skin wound. Despite the development of tissue-saving autologous transplantation techniques such as mesh and Meek grafts, treatment options for extensive skin damage remain severely limited. Yet, the skin is also a rich source of stem and progenitor cells. These cells promote wound healing under physiological conditions and are potential sources for tissue engineering approaches aiming to augment transplantable tissue by generating cultured epidermal autografts (CEAs). Here, we review autologous tissue engineering strategies as well as transplantation products based on skin-derived stem cells. We further provide an overview of clinical trial activities in the field and discuss relevant translational and clinical challenges associated with the use of these products.
Collapse
|
24
|
Osborn MJ, Lees CJ, McElroy AN, Merkel SC, Eide CR, Mathews W, Feser CJ, Tschann M, McElmury RT, Webber BR, Kim CJ, Blazar BR, Tolar J. CRISPR/Cas9-Based Cellular Engineering for Targeted Gene Overexpression. Int J Mol Sci 2018; 19:E946. [PMID: 29565806 PMCID: PMC5979553 DOI: 10.3390/ijms19040946] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2018] [Revised: 03/15/2018] [Accepted: 03/18/2018] [Indexed: 12/27/2022] Open
Abstract
Gene and cellular therapies hold tremendous promise as agents for treating genetic disorders. However, the effective delivery of genes, particularly large ones, and expression at therapeutic levels can be challenging in cells of clinical relevance. To address this engineering hurdle, we sought to employ the clustered regularly interspaced short palindromic repeats (CRISPR)/Cas9 system to insert powerful regulatory elements upstream of an endogenous gene. We achieved robust activation of the COL7A1 gene in primary human umbilical cord blood CD34⁺ hematopoietic stem cells and peripheral blood T-cells. CD34⁺ cells retained their colony forming potential and, in a second engineering step, we disrupted the T-cell receptor complex in T-cells. These cellular populations are of high translational impact due to their engraftment potential, broad circulatory properties, and favorable immune profile that supports delivery to multiple recipients. This study demonstrates the feasibility of targeted knock in of a ubiquitous chromatin opening element, promoter, and marker gene that doubles as a suicide gene for precision gene activation. This system merges the specificity of gene editing with the high level, sustained gene expression achieved with gene therapy vectors. We predict that this design concept will be highly transferrable to most genes in multiple model systems representing a facile cellular engineering platform for promoting gene expression.
Collapse
Affiliation(s)
- Mark J Osborn
- Department of Pediatrics, Division of Blood and Marrow Transplantation, Medical School, University of Minnesota, Minneapolis, MN 55455, USA.
- Stem Cell Institute, University of Minnesota, Minneapolis, MN 55455, USA.
- Center for Genome Engineering, University of Minnesota, Minneapolis, MN 55455, USA.
- Institute for Engineering in Medicine, University of Minnesota, Minneapolis, MN 55455, USA.
- Asan-Minnesota Institute for Innovating Transplantation, University of Minnesota, Minneapolis, MN 55455, USA.
| | - Christopher J Lees
- Department of Pediatrics, Division of Blood and Marrow Transplantation, Medical School, University of Minnesota, Minneapolis, MN 55455, USA.
| | - Amber N McElroy
- Department of Pediatrics, Division of Blood and Marrow Transplantation, Medical School, University of Minnesota, Minneapolis, MN 55455, USA.
| | - Sarah C Merkel
- Department of Pediatrics, Division of Blood and Marrow Transplantation, Medical School, University of Minnesota, Minneapolis, MN 55455, USA.
| | - Cindy R Eide
- Department of Pediatrics, Division of Blood and Marrow Transplantation, Medical School, University of Minnesota, Minneapolis, MN 55455, USA.
| | - Wendy Mathews
- Department of Pediatrics, Division of Blood and Marrow Transplantation, Medical School, University of Minnesota, Minneapolis, MN 55455, USA.
| | - Colby J Feser
- Department of Pediatrics, Division of Blood and Marrow Transplantation, Medical School, University of Minnesota, Minneapolis, MN 55455, USA.
| | - Madison Tschann
- Stem Cell Institute, University of Minnesota, Minneapolis, MN 55455, USA.
| | - Ron T McElmury
- Department of Pediatrics, Division of Blood and Marrow Transplantation, Medical School, University of Minnesota, Minneapolis, MN 55455, USA.
| | - Beau R Webber
- Stem Cell Institute, University of Minnesota, Minneapolis, MN 55455, USA.
- Department of Pediatrics, Division of Hematology, Oncology, and Transplantation, Medical School, University of Minnesota, Minneapolis, MN 55455, USA.
| | - Chong Jai Kim
- Asan-Minnesota Institute for Innovating Transplantation, University of Minnesota, Minneapolis, MN 55455, USA.
- Asan Institute for Life Sciences, Asan Medical Center, Seoul 138-736, Korea.
| | - Bruce R Blazar
- Department of Pediatrics, Division of Blood and Marrow Transplantation, Medical School, University of Minnesota, Minneapolis, MN 55455, USA.
- Stem Cell Institute, University of Minnesota, Minneapolis, MN 55455, USA.
- Center for Genome Engineering, University of Minnesota, Minneapolis, MN 55455, USA.
| | - Jakub Tolar
- Department of Pediatrics, Division of Blood and Marrow Transplantation, Medical School, University of Minnesota, Minneapolis, MN 55455, USA.
- Stem Cell Institute, University of Minnesota, Minneapolis, MN 55455, USA.
- Center for Genome Engineering, University of Minnesota, Minneapolis, MN 55455, USA.
- Asan-Minnesota Institute for Innovating Transplantation, University of Minnesota, Minneapolis, MN 55455, USA.
| |
Collapse
|
25
|
Watanabe M, Natsuga K, Shinkuma S, Shimizu H. Epidermal aspects of type VII collagen: Implications for dystrophic epidermolysis bullosa and epidermolysis bullosa acquisita. J Dermatol 2018; 45:515-521. [PMID: 29352483 DOI: 10.1111/1346-8138.14222] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2017] [Accepted: 12/13/2017] [Indexed: 02/02/2023]
Abstract
Type VII collagen (COL7), a major component of anchoring fibrils in the epidermal basement membrane zone, has been characterized as a defective protein in dystrophic epidermolysis bullosa and as an autoantigen in epidermolysis bullosa acquisita. Although COL7 is produced and secreted by both epidermal keratinocytes and dermal fibroblasts, the role of COL7 with regard to the epidermis is rarely discussed. This review focuses on COL7 physiology and pathology as it pertains to epidermal keratinocytes. We summarize the current knowledge of COL7 production and trafficking, its involvement in keratinocyte dynamics, and epidermal carcinogenesis in COL7 deficiency and propose possible solutions to unsolved issues in this field.
Collapse
Affiliation(s)
- Mika Watanabe
- Department of Dermatology, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Ken Natsuga
- Department of Dermatology, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Satoru Shinkuma
- Division of Dermatology, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Hiroshi Shimizu
- Department of Dermatology, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| |
Collapse
|
26
|
Watt SM, Pleat JM. Stem cells, niches and scaffolds: Applications to burns and wound care. Adv Drug Deliv Rev 2018; 123:82-106. [PMID: 29106911 DOI: 10.1016/j.addr.2017.10.012] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2017] [Revised: 10/19/2017] [Accepted: 10/22/2017] [Indexed: 12/11/2022]
Abstract
The importance of skin to survival, and the devastating physical and psychological consequences of scarring following reparative healing of extensive or difficult to heal human wounds, cannot be disputed. We discuss the significant challenges faced by patients and healthcare providers alike in treating these wounds. New state of the art technologies have provided remarkable insights into the role of skin stem and progenitor cells and their niches in maintaining skin homeostasis and in reparative wound healing. Based on this knowledge, we examine different approaches to repair extensive burn injury and chronic wounds, including full and split thickness skin grafts, temporising matrices and scaffolds, and composite cultured skin products. Notable developments include next generation skin substitutes to replace split thickness skin autografts and next generation gene editing coupled with cell therapies to treat genodermatoses. Further refinements are predicted with the advent of bioprinting technologies, and newly defined biomaterials and autologous cell sources that can be engineered to more accurately replicate human skin architecture, function and cosmesis. These advances will undoubtedly improve quality of life for patients with extensive burns and difficult to heal wounds.
Collapse
Affiliation(s)
- Suzanne M Watt
- Stem Cell Research, Nuffield Division of Clinical Laboratory Medicine, Radcliffe Department of Medicine, University of Oxford, John Radcliffe Hospital, Oxford OX3 9BQ, UK.
| | - Jonathan M Pleat
- Department of Plastic and Reconstructive Surgery, North Bristol NHS Trust and University of Bristol, Westbury on Trym, Bristol BS9 3TZ, UK.
| |
Collapse
|
27
|
Inside out: regenerative medicine for recessive dystrophic epidermolysis bullosa. Pediatr Res 2018; 83:318-324. [PMID: 29593249 DOI: 10.1038/pr.2017.244] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2017] [Accepted: 09/14/2017] [Indexed: 02/07/2023]
Abstract
Epidermolysis bullosa is classified as a genodermatosis, an inherited genetic skin disorder that results in severe, chronic skin blistering with painful and life-threatening complications. Although there is currently no cure for epidermolysis bullosa, concurrent advances in gene and stem cell therapies are converging toward combinatorial therapies that hold the promise of clinically meaningful and lifelong improvement. Recent studies using hematopoietic stem cells and mesenchymal stromal/stem cells to treat epidermolysis bullosa have demonstrated the potential for sustained, effective management of the most severe cases. Furthermore, advances in the use of gene therapy and gene-editing techniques, coupled with the development of induced pluripotent stem cells from patients with epidermolysis bullosa, allow for autologous therapies derived from a renewable population of cells that are patient-specific. Here we describe emerging treatments for epidermolysis bullosa and other genodermatoses, along with a discussion of their benefits and limitations as effective therapies.
Collapse
|
28
|
Woodley DT, Cogan J, Hou Y, Lyu C, Marinkovich MP, Keene D, Chen M. Gentamicin induces functional type VII collagen in recessive dystrophic epidermolysis bullosa patients. J Clin Invest 2017; 127:3028-3038. [PMID: 28691931 DOI: 10.1172/jci92707] [Citation(s) in RCA: 77] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2017] [Accepted: 05/16/2017] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND Recessive dystrophic epidermolysis bullosa (RDEB) is an incurable disease caused by mutations in the gene encoding type VII collagen, the major component of anchoring fibrils (AF). We previously demonstrated that gentamicin produced functional type VII collagen in RDEB cells harboring nonsense mutations. Herein, we determined whether topical or intradermal gentamicin administration induces type VII collagen and AFs in RDEB patients. METHODS A double-blind, placebo-controlled pilot trial assessed safety and efficacy of topical and intradermal gentamicin in 5 RDEB patients with nonsense mutations. The topical arm tested 0.1% gentamicin ointment or placebo application 3 times daily at 2 open erosion sites for 2 weeks. The intradermal arm tested daily intradermal injection of gentamicin solution (8 mg) or placebo into 2 intact skin sites for 2 days in 4 of 5 patients. Primary outcomes were induction of type VII collagen and AFs at the test sites and safety assessment. A secondary outcome assessed wound closure of topically treated erosions. RESULTS Both topical and intradermal gentamicin administration induced type VII collagen and AFs at the dermal-epidermal junction of treatment sites. Newly created type VII collagen varied from 20% to 165% of that expressed in normal human skin and persisted for 3 months. Topical gentamicin corrected dermal-epidermal separation, improved wound closure, and reduced blister formation. There were no untoward side effects from gentamicin treatments. Type VII collagen induction did not generate anti-type VII collagen autoantibodies in patients' blood or skin. CONCLUSION Topical and intradermal gentamicin suppresses nonsense mutations and induces type VII collagen and AFs in RDEB patients. Gentamicin therapy may provide a readily available treatment for RDEB patients with nonsense mutations. TRIAL REGISTRATION ClinicalTrials.gov NCT02698735. FUNDING Epidermolysis Bullosa Research Partnership, Epidermolysis Bullosa Medical Research Foundation, NIH, and VA Merit Award.
Collapse
Affiliation(s)
- David T Woodley
- Department of Dermatology, The Keck School of Medicine, University of Southern California (USC), Los Angeles, California, USA
| | - Jon Cogan
- Department of Dermatology, The Keck School of Medicine, University of Southern California (USC), Los Angeles, California, USA
| | - Yingping Hou
- Department of Dermatology, The Keck School of Medicine, University of Southern California (USC), Los Angeles, California, USA
| | - Chao Lyu
- Department of Dermatology, The Keck School of Medicine, University of Southern California (USC), Los Angeles, California, USA
| | - M Peter Marinkovich
- Department of Dermatology, Stanford University School of Medicine, Stanford, California, USA.,Dermatology, Veteran's Affairs Medical Center, Palo Alto, California, USA
| | - Douglas Keene
- Shriners Hospital for Children, Portland, Oregon, USA
| | - Mei Chen
- Department of Dermatology, The Keck School of Medicine, University of Southern California (USC), Los Angeles, California, USA
| |
Collapse
|
29
|
Atanasova VS, Jiang Q, Prisco M, Gruber C, Piñón Hofbauer J, Chen M, Has C, Bruckner-Tuderman L, McGrath JA, Uitto J, South AP. Amlexanox Enhances Premature Termination Codon Read-Through in COL7A1 and Expression of Full Length Type VII Collagen: Potential Therapy for Recessive Dystrophic Epidermolysis Bullosa. J Invest Dermatol 2017; 137:1842-1849. [PMID: 28549954 DOI: 10.1016/j.jid.2017.05.011] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2016] [Revised: 04/20/2017] [Accepted: 05/06/2017] [Indexed: 01/08/2023]
Abstract
Recessive dystrophic epidermolysis bullosa (RDEB) is a rare monogenic blistering disorder caused by the lack of functional type VII collagen, leading to skin fragility and subsequent trauma-induced separation of the epidermis from the underlying dermis. A total of 46% of patients with RDEB harbor at least one premature termination codon (PTC) mutation in COL7A1, and previous studies have shown that aminoglycosides are able to overcome RDEB PTC mutations by inducing "read-through" and incorporation of an amino acid at the PTC site. However, aminoglycoside toxicity will likely prevent widespread clinical application. Here the FDA-approved drug amlexanox was tested for its ability to read-through PTC mutations in cells derived from patients with RDEB. Eight of 12 different PTC alleles responded to treatment and produced full length protein, in some cases more than 50% relative to normal controls. Read-through type VII collagen was readily detectable in cell culture media and also localized to the dermal-epidermal junction in organotypic skin culture. Amlexanox increased COL7A1 transcript and the phosphorylation of UPF-1, an RNA helicase associated with nonsense-mediated mRNA decay, suggesting that amlexanox inhibits nonsense-mediated mRNA decay in cells from patients with RDEB that respond to read-through treatment. This preclinical study demonstrates the potential of repurposing amlexanox for the treatment of patients with RDEB harboring PTC mutation in COL7A1.
Collapse
Affiliation(s)
- Velina S Atanasova
- Department of Dermatology and Cutaneous Biology, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Qiujie Jiang
- Department of Dermatology and Cutaneous Biology, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Marco Prisco
- Department of Dermatology and Cutaneous Biology, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Christina Gruber
- Department of Dermatology and EB House Austria, Paracelsus Medical University, Salzburg, Austria
| | - Josefina Piñón Hofbauer
- Department of Dermatology and EB House Austria, Paracelsus Medical University, Salzburg, Austria
| | - Mei Chen
- Department of Dermatology, University of Southern California, Los Angeles, California, USA
| | - Cristina Has
- Department of Dermatology, Medical Center - University of Freiburg, Freiburg, Germany
| | | | - John A McGrath
- St. John's Institute of Dermatology, King's College London (Guy's Campus), UK
| | - Jouni Uitto
- Department of Dermatology and Cutaneous Biology, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Andrew P South
- Department of Dermatology and Cutaneous Biology, Thomas Jefferson University, Philadelphia, Pennsylvania, USA.
| |
Collapse
|
30
|
Abstract
Human skin wounds heal largely by reparative wound healing rather than regenerative wound healing. Human skin wounds heal with scarring and without pilosebaceous units or other appendages. Dermal fibroblasts come from 2 distinct lineages of cells that have distinct cell markers and, more importantly, distinct functional abilities. Human skin wound healing largely involves the dermal fibroblast lineage from the reticular dermis and not the papillary dermis. If scientists could find a way to stimulate the dermal fibroblast lineages from the papillary dermis in early wound healing, perhaps human skin wounds could heal without scarring and with skin appendages.
Collapse
|
31
|
Rashidghamat E, McGrath JA. Novel and emerging therapies in the treatment of recessive dystrophic epidermolysis bullosa. Intractable Rare Dis Res 2017; 6:6-20. [PMID: 28357176 PMCID: PMC5359356 DOI: 10.5582/irdr.2017.01005] [Citation(s) in RCA: 71] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Epidermolysis bullosa (EB) is a clinically and genetically heterogeneous group of inherited blistering diseases that affects ∼ 500,000 people worldwide. Clinically, individuals with EB have fragile skin and are susceptible to blistering following minimal trauma, with mucous membrane and other organ involvement in some subtypes. Within the spectrum of EB, ∼ 5% of affected individuals have the clinically more severe recessive dystrophic (RDEB) variant with a prevalence of 8 per one million of the population. RDEB is caused by loss-of-function mutations in the type VII collagen gene, COL7A1, which leads to reduced or absent type VII collagen (C7) and a paucity of structurally effective anchoring fibrils at the dermal-epidermal junction (DEJ). Currently, there is no cure for RDEB, although considerable progress has been made in testing novel treatments including gene therapy (lentiviral and gamma retroviral vectors for COL7A1 supplementation in keratinocytes and fibroblasts), as well as cell therapy (use of allogeneic fibroblasts, mesenchymal stromal cells (MSCs), and bone marrow transplantation (BMT)). Here, we review current treatment modalities available as well as novel and emerging therapies in the treatment of RDEB. Clinical trials of new translational therapies in RDEB offer hope for improved clinical management of patients as well as generating broader lessons for regenerative medicine that could be applicable to other inherited or acquired abnormalities of wound healing or scarring.
Collapse
Affiliation(s)
- Ellie Rashidghamat
- St. John's Institute of Dermatology, King's College London, London, United Kingdom
| | - John A. McGrath
- St. John's Institute of Dermatology, King's College London, London, United Kingdom
- Address correspondence to: Dr. John A. McGrath, Dermatology Research Laboratories, Floor 9 Tower Wing, Guy's Hospital, Great Maze Pond, London SE1 9RT, United Kingdom. E-mail:
| |
Collapse
|
32
|
Efficient in vivo gene editing using ribonucleoproteins in skin stem cells of recessive dystrophic epidermolysis bullosa mouse model. Proc Natl Acad Sci U S A 2017; 114:1660-1665. [PMID: 28137859 DOI: 10.1073/pnas.1614775114] [Citation(s) in RCA: 81] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
The prokaryotic CRISPR/Cas9 system has recently emerged as a powerful tool for genome editing in mammalian cells with the potential to bring curative therapies to patients with genetic diseases. However, efficient in vivo delivery of this genome editing machinery and indeed the very feasibility of using these techniques in vivo remain challenging for most tissue types. Here, we show that nonreplicable Cas9/sgRNA ribonucleoproteins can be used to correct genetic defects in skin stem cells of postnatal recessive dystrophic epidermolysis bullosa (RDEB) mice. We developed a method to locally deliver Cas9/sgRNA ribonucleoproteins into the skin of postnatal mice. This method results in rapid gene editing in epidermal stem cells. Using this method, we show that Cas9/sgRNA ribonucleoproteins efficiently excise exon80, which covers the point mutation in our RDEB mouse model, and thus restores the correct localization of the collagen VII protein in vivo. The skin blistering phenotype is also significantly ameliorated after treatment. This study provides an in vivo gene correction strategy using ribonucleoproteins as curative treatment for genetic diseases in skin and potentially in other somatic tissues.
Collapse
|
33
|
Cubo N, Garcia M, del Cañizo JF, Velasco D, Jorcano JL. 3D bioprinting of functional human skin: production and
in vivo
analysis. Biofabrication 2016; 9:015006. [DOI: 10.1088/1758-5090/9/1/015006] [Citation(s) in RCA: 254] [Impact Index Per Article: 31.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
34
|
Jacków J, Titeux M, Portier S, Charbonnier S, Ganier C, Gaucher S, Hovnanian A. Gene-Corrected Fibroblast Therapy for Recessive Dystrophic Epidermolysis Bullosa using a Self-Inactivating COL7A1 Retroviral Vector. J Invest Dermatol 2016; 136:1346-1354. [DOI: 10.1016/j.jid.2016.02.811] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2015] [Revised: 02/12/2016] [Accepted: 02/26/2016] [Indexed: 12/16/2022]
|
35
|
Georgiadis C, Syed F, Petrova A, Abdul-Wahab A, Lwin SM, Farzaneh F, Chan L, Ghani S, Fleck RA, Glover L, McMillan JR, Chen M, Thrasher AJ, McGrath JA, Di WL, Qasim W. Lentiviral Engineered Fibroblasts Expressing Codon-Optimized COL7A1 Restore Anchoring Fibrils in RDEB. J Invest Dermatol 2016; 136:284-92. [PMID: 26763448 PMCID: PMC4759620 DOI: 10.1038/jid.2015.364] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2015] [Revised: 07/27/2015] [Accepted: 08/03/2015] [Indexed: 01/06/2023]
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.
Collapse
Affiliation(s)
- Christos Georgiadis
- UCL Institute of Child Health, Molecular and Cellular Immunology Section & Great Ormond Street Hospital NHS Foundation Trust, London, United Kingdom
| | - Farhatullah Syed
- UCL Institute of Child Health, Molecular and Cellular Immunology Section & Great Ormond Street Hospital NHS Foundation Trust, London, United Kingdom
| | - Anastasia Petrova
- UCL Institute of Child Health, Molecular and Cellular Immunology Section & Great Ormond Street Hospital NHS Foundation Trust, London, United Kingdom
| | - Alya Abdul-Wahab
- St John's Institute of Dermatology, King's College London (Guy's campus), London, United Kingdom
| | - Su M Lwin
- St John's Institute of Dermatology, King's College London (Guy's campus), London, United Kingdom
| | - Farzin Farzaneh
- Department of Haematological Medicine, King's College London, The Rayne Institute, London, United Kingdom
| | - Lucas Chan
- Department of Haematological Medicine, King's College London, The Rayne Institute, London, United Kingdom
| | - Sumera Ghani
- UCL Institute of Child Health, Molecular and Cellular Immunology Section & Great Ormond Street Hospital NHS Foundation Trust, London, United Kingdom
| | - Roland A Fleck
- Centre for Ultrastructural Imaging, King's College London, London, United Kingdom
| | - Leanne Glover
- Centre for Ultrastructural Imaging, King's College London, London, United Kingdom
| | - James R McMillan
- The Robin Eady National Diagnostic Epidermolysis Bullosa Laboratory, Viapath LLP, St Thomas' Hospital, London, United Kingdom
| | - Mei Chen
- Department of Dermatology, University of Southern California, Los Angeles, California, USA
| | - Adrian J Thrasher
- UCL Institute of Child Health, Molecular and Cellular Immunology Section & Great Ormond Street Hospital NHS Foundation Trust, London, United Kingdom
| | - John A McGrath
- St John's Institute of Dermatology, King's College London (Guy's campus), London, United Kingdom
| | - Wei-Li Di
- UCL Institute of Child Health, Molecular and Cellular Immunology Section & Great Ormond Street Hospital NHS Foundation Trust, London, United Kingdom
| | - Waseem Qasim
- UCL Institute of Child Health, Molecular and Cellular Immunology Section & Great Ormond Street Hospital NHS Foundation Trust, London, United Kingdom.
| |
Collapse
|
36
|
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: 2.5] [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
|
37
|
Perdoni C, Osborn MJ, Tolar J. Gene editing toward the use of autologous therapies in recessive dystrophic epidermolysis bullosa. Transl Res 2016; 168:50-58. [PMID: 26073463 PMCID: PMC4662628 DOI: 10.1016/j.trsl.2015.05.008] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2015] [Accepted: 05/19/2015] [Indexed: 01/22/2023]
Abstract
Recessive dystrophic epidermolysis bullosa (RDEB) is a disease caused by mutations in the COL7A1 gene that result in absent or dysfunctional type VII collagen protein production. Clinically, RDEB manifests as early and severe chronic cutaneous blistering, damage to internal epithelium, an increased risk for squamous cell carcinoma, and an overall reduced life expectancy. Recent localized and systemic treatments have shown promise for lessening the disease severity in RDEB, but the concept of ex vivo therapy would allow a patient's own cells to be engineered to express functional type VII collagen. Here, we review gene delivery and editing platforms and their application toward the development of next-generation treatments designed to correct the causative genetic defects of RDEB.
Collapse
Affiliation(s)
- Christopher Perdoni
- Stem Cell Institute, University of Minnesota, Minneapolis, Minn; Division of Blood and Marrow Transplantation, Department of Pediatrics, University of Minnesota, Minneapolis, Minn
| | - Mark J Osborn
- Stem Cell Institute, University of Minnesota, Minneapolis, Minn; Division of Blood and Marrow Transplantation, Department of Pediatrics, University of Minnesota, Minneapolis, Minn
| | - Jakub Tolar
- Stem Cell Institute, University of Minnesota, Minneapolis, Minn; Division of Blood and Marrow Transplantation, Department of Pediatrics, University of Minnesota, Minneapolis, Minn.
| |
Collapse
|
38
|
Geyer MB, Radhakrishnan K, Giller R, Umegaki N, Harel S, Kiuru M, Morel KD, LeBoeuf N, Kandel J, Bruckner A, Fabricatore S, Chen M, Woodley D, McGrath J, Baxter-Lowe L, Uitto J, Christiano AM, Cairo MS. Reduced Toxicity Conditioning and Allogeneic Hematopoietic Progenitor Cell Transplantation for Recessive Dystrophic Epidermolysis Bullosa. J Pediatr 2015; 167:765-9.e1. [PMID: 26148662 DOI: 10.1016/j.jpeds.2015.05.051] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2014] [Revised: 04/03/2015] [Accepted: 05/26/2015] [Indexed: 10/23/2022]
Abstract
Recessive dystrophic epidermolysis bullosa is a severe, incurable, inherited blistering disease caused by COL7A1 mutations. Emerging evidence suggests hematopoietic progenitor cells (HPCs) can be reprogrammed into skin; HPC-derived cells can restore COL7 expression in COL7-deficient mice. We report two children with recessive dystrophic epidermolysis bullosa treated with reduced-toxicity conditioning and HLA-matched HPC transplantation.
Collapse
Affiliation(s)
- Mark B Geyer
- Department of Medicine (Hematology and Medical Oncology), Memorial Sloan-Kettering Cancer Center, New York, NY
| | - Kavita Radhakrishnan
- Department of Pediatrics, University of California, San Francisco, San Francisco, CA
| | - Roger Giller
- Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO
| | - Noriko Umegaki
- Department of Dermatology, Columbia University, New York, NY
| | - Sivan Harel
- Department of Dermatology, Columbia University, New York, NY
| | - Maija Kiuru
- Department of Medicine (Dermatology Service), Memorial Sloan-Kettering Cancer Center, New York, NY; Department of Dermatology, Weill Cornell Medical College, New York, NY
| | - Kimberly D Morel
- Department of Dermatology, Columbia University, New York, NY; Department of Pediatrics, Columbia University, New York, NY
| | - Nicole LeBoeuf
- Department of Dermatology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Jessica Kandel
- Department of Surgery, The University of Chicago Medicine Comer Children's Hospital, Chicago, IL
| | - Anna Bruckner
- Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO; Department of Dermatology, University of Colorado School of Medicine, Aurora, CO
| | | | - Mei Chen
- Department of Dermatology, University of Southern California, Los Angeles, CA
| | - David Woodley
- Department of Dermatology, University of Southern California, Los Angeles, CA
| | - John McGrath
- Department of Genetics and Molecular Medicine, King's College, London, United Kingdom
| | - LeeAnn Baxter-Lowe
- Department of Pathology and Laboratory Medicine, Children's Hospital of Los Angeles, Los Angeles, CA
| | - Jouni Uitto
- Department of Dermatology and Cutaneous Biology, The Thomas Jefferson University, Philadelphia, PA
| | - Angela M Christiano
- Department of Dermatology, Columbia University, New York, NY; Department of Genetics, Columbia University, New York, NY
| | - Mitchell S Cairo
- Department of Pediatrics, New York Medical College, Valhalla, NY; Department of Medicine, New York Medical College, Valhalla, NY; Department of Pathology, New York Medical College, Valhalla, NY; Department of Microbiology and Immunology, New York Medical College, Valhalla, NY; Department of Cell Biology and Anatomy, New York Medical College, Valhalla, NY.
| |
Collapse
|
39
|
Hou Y, Guey LT, Wu T, Gao R, Cogan J, Wang X, Hong E, Vivian Ning W, Keene D, Liu N, Huang Y, Kaftan C, Tangarone B, Quinones-Garcia I, Uitto J, Francone OL, Woodley DT, Chen M. Intravenously Administered Recombinant Human Type VII Collagen Derived from Chinese Hamster Ovary Cells Reverses the Disease Phenotype in Recessive Dystrophic Epidermolysis Bullosa Mice. J Invest Dermatol 2015. [PMID: 26203639 DOI: 10.1038/jid.2015.291] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Recessive dystrophic epidermolysis bullosa (RDEB) is an inherited disorder characterized by skin fragility, blistering, and multiple skin wounds with no currently approved or consistently effective treatment. It is due to mutations in the gene encoding type VII collagen (C7). Using recombinant human C7 (rhC7) purified from human dermal fibroblasts (FB-rhC7), we showed previously that intravenously injected rhC7 distributed to engrafted RDEB skin, incorporated into its dermal-epidermal junction (DEJ), and reversed the RDEB disease phenotype. Human dermal fibroblasts, however, are not used for commercial production of therapeutic proteins. Therefore, we generated rhC7 from Chinese hamster ovary (CHO) cells. The CHO-derived recombinant type VII collagen (CHO-rhC7), similar to FB-rhC7, was secreted as a correctly folded, disulfide-bonded, helical trimer resistant to protease degradation. CHO-rhC7 bound to fibronectin and promoted human keratinocyte migration in vitro. A single dose of CHO-rhC7, administered intravenously into new-born C7-null RDEB mice, incorporated into the DEJ of multiple skin sites, tongue and esophagus, restored anchoring fibrils, improved dermal-epidermal adherence, and increased the animals' life span. Furthermore, no circulating or tissue-bound anti-C7 antibodies were observed in the mice. These data demonstrate the efficacy of CHO-rhC7 in a preclinical murine model of RDEB.
Collapse
Affiliation(s)
- Yingping Hou
- Department of Dermatology, University of Southern California, Los Angeles, California, USA
| | | | - Timothy Wu
- Department of Dermatology, University of Southern California, Los Angeles, California, USA
| | - Robert Gao
- Department of Dermatology, University of Southern California, Los Angeles, California, USA
| | - Jon Cogan
- Department of Dermatology, University of Southern California, Los Angeles, California, USA
| | - Xinyi Wang
- Department of Dermatology, University of Southern California, Los Angeles, California, USA
| | - Elizabeth Hong
- Department of Dermatology, University of Southern California, Los Angeles, California, USA
| | - Weihuang Vivian Ning
- Department of Dermatology, University of Southern California, Los Angeles, California, USA
| | - Douglas Keene
- Shriners Hospital for Children, Portland, Oregon, USA
| | - Nan Liu
- Shire, Lexington, Massachussetts, USA
| | - Yan Huang
- Shire, Lexington, Massachussetts, USA
| | | | | | | | - Jouni Uitto
- Department of Dermatology and Cutaneous Biology, Jefferson Medical College, Philadelphia, PA, USA
| | | | - David T Woodley
- Department of Dermatology, University of Southern California, Los Angeles, California, USA
| | - Mei Chen
- Department of Dermatology, University of Southern California, Los Angeles, California, USA.
| |
Collapse
|
40
|
Sebastiano V, Zhen HH, Haddad B, Derafshi BH, Bashkirova E, Melo SP, Wang P, Leung TL, Siprashvili Z, Tichy A, Li J, Ameen M, Hawkins J, Lee S, Li L, Schwertschkow A, Bauer G, Lisowski L, Kay MA, Kim SK, Lane AT, Wernig M, Oro AE. Human COL7A1-corrected induced pluripotent stem cells for the treatment of recessive dystrophic epidermolysis bullosa. Sci Transl Med 2015; 6:264ra163. [PMID: 25429056 DOI: 10.1126/scitranslmed.3009540] [Citation(s) in RCA: 157] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Patients with recessive dystrophic epidermolysis bullosa (RDEB) lack functional type VII collagen owing to mutations in the gene COL7A1 and suffer severe blistering and chronic wounds that ultimately lead to infection and development of lethal squamous cell carcinoma. The discovery of induced pluripotent stem cells (iPSCs) and the ability to edit the genome bring the possibility to provide definitive genetic therapy through corrected autologous tissues. We generated patient-derived COL7A1-corrected epithelial keratinocyte sheets for autologous grafting. We demonstrate the utility of sequential reprogramming and adenovirus-associated viral genome editing to generate corrected iPSC banks. iPSC-derived keratinocytes were produced with minimal heterogeneity, and these cells secreted wild-type type VII collagen, resulting in stratified epidermis in vitro in organotypic cultures and in vivo in mice. Sequencing of corrected cell lines before tissue formation revealed heterogeneity of cancer-predisposing mutations, allowing us to select COL7A1-corrected banks with minimal mutational burden for downstream epidermis production. Our results provide a clinical platform to use iPSCs in the treatment of debilitating genodermatoses, such as RDEB.
Collapse
Affiliation(s)
- Vittorio Sebastiano
- Institute for Stem Cell Biology and Regenerative Medicine, and Department of Pathology, Stanford University, Stanford, CA 94305, USA. Department of Obstetrics and Gynecology, Stanford University, Stanford, CA 94305, USA
| | - Hanson Hui Zhen
- Program in Epithelial Biology, Department of Dermatology, Stanford University, Stanford, CA 94305, USA
| | | | - Bahareh Haddad Derafshi
- Institute for Stem Cell Biology and Regenerative Medicine, and Department of Pathology, Stanford University, Stanford, CA 94305, USA
| | - Elizaveta Bashkirova
- Institute for Stem Cell Biology and Regenerative Medicine, and Department of Pathology, Stanford University, Stanford, CA 94305, USA
| | - Sandra P Melo
- Program in Epithelial Biology, Department of Dermatology, Stanford University, Stanford, CA 94305, USA
| | - Pei Wang
- Department of Developmental Biology, Stanford University, Stanford, CA 94305, USA
| | - Thomas L Leung
- Department of Developmental Biology, Stanford University, Stanford, CA 94305, USA
| | - Zurab Siprashvili
- Program in Epithelial Biology, Department of Dermatology, Stanford University, Stanford, CA 94305, USA
| | - Andrea Tichy
- Program in Epithelial Biology, Department of Dermatology, Stanford University, Stanford, CA 94305, USA
| | - Jiang Li
- Program in Epithelial Biology, Department of Dermatology, Stanford University, Stanford, CA 94305, USA
| | - Mohammed Ameen
- Program in Epithelial Biology, Department of Dermatology, Stanford University, Stanford, CA 94305, USA
| | - John Hawkins
- Institute for Stem Cell Biology and Regenerative Medicine, and Department of Pathology, Stanford University, Stanford, CA 94305, USA
| | - Susie Lee
- Program in Epithelial Biology, Department of Dermatology, Stanford University, Stanford, CA 94305, USA
| | - Lingjie Li
- Program in Epithelial Biology, Department of Dermatology, Stanford University, Stanford, CA 94305, USA
| | - Aaron Schwertschkow
- Institute for Regenerative Cures, School of Medicine, University of California, Davis, Sacramento, CA 95817, USA
| | - Gerhard Bauer
- Institute for Regenerative Cures, School of Medicine, University of California, Davis, Sacramento, CA 95817, USA
| | - Leszek Lisowski
- Departments of Pediatrics and Genetics, Stanford University, Stanford, CA 94305, USA
| | - Mark A Kay
- Departments of Pediatrics and Genetics, Stanford University, Stanford, CA 94305, USA
| | - Seung K Kim
- Department of Developmental Biology, Stanford University, Stanford, CA 94305, USA
| | - Alfred T Lane
- Program in Epithelial Biology, Department of Dermatology, Stanford University, Stanford, CA 94305, USA
| | - Marius Wernig
- Institute for Stem Cell Biology and Regenerative Medicine, and Department of Pathology, Stanford University, Stanford, CA 94305, USA.
| | - Anthony E Oro
- Program in Epithelial Biology, Department of Dermatology, Stanford University, Stanford, CA 94305, USA.
| |
Collapse
|
41
|
Recessive Dystrophic Epidermolysis Bullosa: Advances in the Laboratory Leading to New Therapies. J Invest Dermatol 2015; 135:1705-1707. [DOI: 10.1038/jid.2015.149] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
|
42
|
Abstract
In the past few years, substantial preclinical and experimental advances have been made in the treatment of the severe monogenic skin blistering disease epidermolysis bullosa (EB). Promising approaches have been developed in the fields of protein and cell therapies, including allogeneic stem cell transplantation; in addition, the application of gene therapy approaches has become reality. The first ex vivo gene therapy for a junctional EB (JEB) patient was performed in Italy more than 8 years ago and was shown to be effective. We have now continued this approach for an Austrian JEB patient. Further, clinical trials for a gene therapy treatment of recessive dystrophic EB are currently under way in the United States and in Europe. In this review, we aim to point out that sustainable correction of autologous keratinocytes by stable genomic integration of a therapeutic gene represents a realistic option for patients with EB.
Collapse
|
43
|
Abstract
Dystrophic epidermolysis bullosa is a rare inherited blistering disorder caused by mutations in the COL7A1 gene encoding type VII collagen. The deficiency and/or dysfunction of type VII collagen leads to subepidermal blistering immediately below the lamina densa, resulting in mucocutaneous fragility and disease complications such as intractable ulcers, extensive scarring, malnutrition, and malignancy. The disease is usually diagnosed by immunofluorescence mapping and/or transmission electron microscopy and subsequently subclassified into one of 14 subtypes. This review provides practical knowledge on the disease, including new therapeutic strategies.
Collapse
Affiliation(s)
- Satoru Shinkuma
- Department of Dermatology, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| |
Collapse
|
44
|
Cutlar L, Greiser U, Wang W. Gene therapy: pursuing restoration of dermal adhesion in recessive dystrophic epidermolysis bullosa. Exp Dermatol 2014; 23:1-6. [PMID: 24107073 DOI: 10.1111/exd.12246] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/17/2013] [Indexed: 12/13/2022]
Abstract
The replacement of a defective gene with a fully functional copy is the goal of the most basic gene therapy. Recessive dystrophic epidermolysis bullosa (RDEB) is characterised by a lack of adhesion of the epidermis to the dermis. It is an ideal target for gene therapy as all variants of hereditary RDEB are caused by mutations in a single gene, COL7A1, coding for type VII collagen, a key component of anchoring fibrils that secure attachment of the epidermis to the dermis. RDEB is one of the most severe variants in the epidermolysis bullosa (EB) group of heritable skin diseases. Epidermolysis bullosa is defined by chronic fragility and blistering of the skin and mucous membranes due to mutations in the genes responsible for production of the basement membrane proteins. This condition has a high personal, medical and socio-economic impact. People with RDEB require a broad spectrum of medications and specialised care. Due to this being a systemic condition, most research focus is in the area of gene therapy. Recently, preclinical works have begun to show promise. They focus on the virally mediated ex vivo correction of autologous epithelium. These corrected cells are then to be expanded and grafted onto the patient following the lead of the first successful gene therapy in dermatology being a grafting of corrected tissue for junctional EB treatment. Current progress, outstanding challenges and future directions in translating these approaches in clinics are reviewed in this article.
Collapse
Affiliation(s)
- Lara Cutlar
- Network of Excellence for Functional Biomaterials, National University of Ireland, Galway, Ireland
| | | | | |
Collapse
|
45
|
Aminoglycosides restore full-length type VII collagen by overcoming premature termination codons: therapeutic implications for dystrophic epidermolysis bullosa. Mol Ther 2014; 22:1741-52. [PMID: 25155989 DOI: 10.1038/mt.2014.140] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2014] [Accepted: 07/16/2014] [Indexed: 11/08/2022] Open
Abstract
Patients with recessive dystrophic epidermolysis bullosa (RDEB) have severe, incurable skin fragility, blistering, and multiple skin wounds due to mutations in the gene encoding type VII collagen (C7), the major component of anchoring fibrils mediating epidermal-dermal adherence. Nearly 10-25% of RDEB patients carry nonsense mutations leading to premature stop codons (PTCs) that result in truncated C7. In this study, we evaluated the feasibility of using aminoglycosides to suppress PTCs and induce C7 expression in two RDEB keratinocyte cell lines (Q251X/Q251X and R578X/R906) and two primary RDEB fibroblasts (R578X/R578X and R163X/R1683X). Incubation of these cells with aminoglycosides (geneticin, gentamicin, and paromomycin) resulted in the synthesis and secretion of a full-length C7 in a dose-dependent and sustained manner. Importantly, aminoglycoside-induced C7 reversed the abnormal RDEB cell phenotype and incorporated into the dermal-epidermal junction of skin equivalents. We further demonstrated the general utility of aminoglycoside-mediated readthrough in 293 cells transiently transfected with expression vectors encoding 22 different RDEB nonsense mutations. This is the first study demonstrating that aminoglycosides can induce PTC readthrough and restore functional C7 in RDEB caused by nonsense mutations. Therefore, aminoglycosides may have therapeutic potential for RDEB patients and other inherited skin diseases caused by nonsense mutations.
Collapse
|
46
|
Nogueira C, Erlmann P, Villeneuve J, Santos AJ, Martínez-Alonso E, Martínez-Menárguez JÁ, Malhotra V. SLY1 and Syntaxin 18 specify a distinct pathway for procollagen VII export from the endoplasmic reticulum. eLife 2014. [PMID: 24842878 DOI: 10.7554/elife.02784.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
TANGO1 binds and exports Procollagen VII from the endoplasmic reticulum (ER). In this study, we report a connection between the cytoplasmic domain of TANGO1 and SLY1, a protein that is required for membrane fusion. Knockdown of SLY1 by siRNA arrested Procollagen VII in the ER without affecting the recruitment of COPII components, general protein secretion, and retrograde transport of the KDEL-containing protein BIP, and ERGIC53. SLY1 is known to interact with the ER-specific SNARE proteins Syntaxin 17 and 18, however only Syntaxin 18 was required for Procollagen VII export. Neither SLY1 nor Syntaxin 18 was required for the export of the equally bulky Procollagen I from the ER. Altogether, these findings reveal the sorting of bulky collagen family members by TANGO1 at the ER and highlight the existence of different export pathways for secretory cargoes one of which is mediated by the specific SNARE complex containing SLY1 and Syntaxin 18.DOI: http://dx.doi.org/10.7554/eLife.02784.001.
Collapse
Affiliation(s)
- Cristina Nogueira
- Cell and Developmental Biology Program, Center for Genomic Regulation (CRG), Barcelona, Spain Universitat Pompeu Fabra (UPF), Barcelona, Spain
| | - Patrik Erlmann
- Cell and Developmental Biology Program, Center for Genomic Regulation (CRG), Barcelona, Spain Universitat Pompeu Fabra (UPF), Barcelona, Spain
| | - Julien Villeneuve
- Cell and Developmental Biology Program, Center for Genomic Regulation (CRG), Barcelona, Spain Universitat Pompeu Fabra (UPF), Barcelona, Spain
| | - António Jm Santos
- Cell and Developmental Biology Program, Center for Genomic Regulation (CRG), Barcelona, Spain Universitat Pompeu Fabra (UPF), Barcelona, Spain
| | - Emma Martínez-Alonso
- Department of Cellular Biology and Histology, Faculty of Medicine, University of Murcia, Murcia, Spain
| | | | - Vivek Malhotra
- Cell and Developmental Biology Program, Center for Genomic Regulation (CRG), Barcelona, Spain Universitat Pompeu Fabra (UPF), Barcelona, Spain Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain
| |
Collapse
|
47
|
Nogueira C, Erlmann P, Villeneuve J, Santos AJ, Martínez-Alonso E, Martínez-Menárguez JÁ, Malhotra V. SLY1 and Syntaxin 18 specify a distinct pathway for procollagen VII export from the endoplasmic reticulum. eLife 2014; 3:e02784. [PMID: 24842878 PMCID: PMC4054776 DOI: 10.7554/elife.02784] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
TANGO1 binds and exports Procollagen VII from the endoplasmic reticulum (ER). In this study, we report a connection between the cytoplasmic domain of TANGO1 and SLY1, a protein that is required for membrane fusion. Knockdown of SLY1 by siRNA arrested Procollagen VII in the ER without affecting the recruitment of COPII components, general protein secretion, and retrograde transport of the KDEL-containing protein BIP, and ERGIC53. SLY1 is known to interact with the ER-specific SNARE proteins Syntaxin 17 and 18, however only Syntaxin 18 was required for Procollagen VII export. Neither SLY1 nor Syntaxin 18 was required for the export of the equally bulky Procollagen I from the ER. Altogether, these findings reveal the sorting of bulky collagen family members by TANGO1 at the ER and highlight the existence of different export pathways for secretory cargoes one of which is mediated by the specific SNARE complex containing SLY1 and Syntaxin 18.DOI: http://dx.doi.org/10.7554/eLife.02784.001.
Collapse
Affiliation(s)
- Cristina Nogueira
- Cell and Developmental Biology Program, Center for Genomic Regulation (CRG), Barcelona, Spain Universitat Pompeu Fabra (UPF), Barcelona, Spain
| | - Patrik Erlmann
- Cell and Developmental Biology Program, Center for Genomic Regulation (CRG), Barcelona, Spain Universitat Pompeu Fabra (UPF), Barcelona, Spain
| | - Julien Villeneuve
- Cell and Developmental Biology Program, Center for Genomic Regulation (CRG), Barcelona, Spain Universitat Pompeu Fabra (UPF), Barcelona, Spain
| | - António Jm Santos
- Cell and Developmental Biology Program, Center for Genomic Regulation (CRG), Barcelona, Spain Universitat Pompeu Fabra (UPF), Barcelona, Spain
| | - Emma Martínez-Alonso
- Department of Cellular Biology and Histology, Faculty of Medicine, University of Murcia, Murcia, Spain
| | | | - Vivek Malhotra
- Cell and Developmental Biology Program, Center for Genomic Regulation (CRG), Barcelona, Spain Universitat Pompeu Fabra (UPF), Barcelona, Spain Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain
| |
Collapse
|
48
|
Gorell E, Nguyen N, Lane A, Siprashvili Z. Gene therapy for skin diseases. Cold Spring Harb Perspect Med 2014; 4:a015149. [PMID: 24692191 DOI: 10.1101/cshperspect.a015149] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
The skin possesses qualities that make it desirable for gene therapy, and studies have focused on gene therapy for multiple cutaneous diseases. Gene therapy uses a vector to introduce genetic material into cells to alter gene expression, negating a pathological process. This can be accomplished with a variety of viral vectors or nonviral administrations. Although results are promising, there are several potential pitfalls that must be addressed to improve the safety profile to make gene therapy widely available clinically.
Collapse
Affiliation(s)
- Emily Gorell
- Department of Dermatology, Stanford School of Medicine, Palo Alto, California 94305
| | | | | | | |
Collapse
|
49
|
Kern JS, Has C. Update on diagnosis and therapy of inherited epidermolysis bullosa. ACTA ACUST UNITED AC 2014. [DOI: 10.1586/17469872.3.6.721] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
50
|
Woodley DT, Cogan J, Wang X, Hou Y, Haghighian C, Kudo G, Keene DR, Chen M. De novo anti-type VII collagen antibodies in patients with recessive dystrophic epidermolysis bullosa. J Invest Dermatol 2013; 134:1138-1140. [PMID: 24213372 PMCID: PMC3961494 DOI: 10.1038/jid.2013.475] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- David T Woodley
- Department of Dermatology, Keck School of Medicine, University of Southern California, Los Angeles, California, USA.
| | - Jon Cogan
- Department of Dermatology, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Xinyi Wang
- Department of Dermatology, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Yingping Hou
- Department of Dermatology, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Cyrus Haghighian
- Department of Dermatology, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Gail Kudo
- Department of Dermatology, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | | | - Mei Chen
- Department of Dermatology, Keck School of Medicine, University of Southern California, Los Angeles, California, USA.
| |
Collapse
|