1
|
Steinbeck BJ, Gao XD, McElroy AN, Pandey S, Doman JL, Riddle MJ, Xia L, Chen W, Eide CR, Lengert AH, Han SW, Blazar BR, Wandall HH, Dabelsteen S, Liu DR, Tolar J, Osborn MJ. Twin Prime Editing Mediated Exon Skipping/Reinsertion for Restored Collagen VII Expression in Recessive Dystrophic Epidermolysis Bullosa. J Invest Dermatol 2024; 144:2764-2777.e9. [PMID: 38763174 PMCID: PMC12050016 DOI: 10.1016/j.jid.2024.04.013] [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: 08/28/2023] [Revised: 04/12/2024] [Accepted: 04/26/2024] [Indexed: 05/21/2024]
Abstract
Gene editing nucleases, base editors, and prime editors are potential locus-specific genetic treatment strategies for recessive dystrophic epidermolysis bullosa; however, many recessive dystrophic epidermolysis bullosa COL7A1 pathogenic nucleotide variations (PNVs) are unique, making the development of personalized editing reagents challenging. A total of 270 of the ∼320 COL7A1 epidermolysis bullosa PNVs reside in exons that can be skipped, and antisense oligonucleotides and gene editing nucleases have been used to create in-frame deletions. Antisense oligonucleotides are transient, and nucleases generate deleterious double-stranded DNA breaks and uncontrolled mixtures of allele products. We developed a twin prime editing strategy using the PEmax and recently evolved PE6 prime editors and dual prime editing guide RNAs flanking COL7A1 exon 5. Prime editing-mediated deletion of exon 5 with a homozygous premature stop codon was achieved in recessive dystrophic epidermolysis bullosa fibroblasts, keratinocytes, and induced pluripotent stem cells with minimal double-stranded DNA breaks, and collagen type VII protein was restored. Twin prime editing can replace the target exon with recombinase attachment sequences, and we exploited this to reinsert a normal copy of exon 5 using the Bxb1 recombinase. These findings demonstrate that twin prime editing can facilitate locus-specific, predictable, in-frame deletions and sequence replacement with few double-stranded DNA breaks as a strategy that may enable a single therapeutic agent to treat multiple recessive dystrophic epidermolysis bullosa patient cohorts.
Collapse
Affiliation(s)
- Benjamin J Steinbeck
- Division of Pediatric Blood and Marrow Transplantation & Cellular Therapy, Department of Pediatrics, University of Minnesota, Minneapolis, Minnesota, USA
| | - Xin D Gao
- Merkin Institute of Transformative Technologies in Healthcare, The Broad Institute of Harvard and MIT, Cambridge, Massachusetts, USA; Department of Chemistry and Chemical Biology, Harvard University, Cambridge, Massachusetts, USA; Howard Hughes Medical Institute, Harvard University, Cambridge, Massachusetts, USA
| | - Amber N McElroy
- Division of Pediatric Blood and Marrow Transplantation & Cellular Therapy, Department of Pediatrics, University of Minnesota, Minneapolis, Minnesota, USA
| | - Smriti Pandey
- Merkin Institute of Transformative Technologies in Healthcare, The Broad Institute of Harvard and MIT, Cambridge, Massachusetts, USA; Department of Chemistry and Chemical Biology, Harvard University, Cambridge, Massachusetts, USA; Howard Hughes Medical Institute, Harvard University, Cambridge, Massachusetts, USA
| | - Jordan L Doman
- Merkin Institute of Transformative Technologies in Healthcare, The Broad Institute of Harvard and MIT, Cambridge, Massachusetts, USA; Department of Chemistry and Chemical Biology, Harvard University, Cambridge, Massachusetts, USA; Howard Hughes Medical Institute, Harvard University, Cambridge, Massachusetts, USA
| | - Megan J Riddle
- Division of Pediatric Blood and Marrow Transplantation & Cellular Therapy, Department of Pediatrics, University of Minnesota, Minneapolis, Minnesota, USA
| | - Lily Xia
- Division of Pediatric Blood and Marrow Transplantation & Cellular Therapy, Department of Pediatrics, University of Minnesota, Minneapolis, Minnesota, USA
| | - Weili Chen
- Division of Pediatric Blood and Marrow Transplantation & Cellular Therapy, Department of Pediatrics, University of Minnesota, Minneapolis, Minnesota, USA
| | - Cindy R Eide
- Division of Pediatric Blood and Marrow Transplantation & Cellular Therapy, Department of Pediatrics, University of Minnesota, Minneapolis, Minnesota, USA
| | - Andre H Lengert
- Escola Paulista de Medicina, Universidade Federal de Sao Paulo, Sao Paulo, Brazil
| | - Sang Won Han
- Escola Paulista de Medicina, Universidade Federal de Sao Paulo, Sao Paulo, Brazil
| | - Bruce R Blazar
- Division of Pediatric Blood and Marrow Transplantation & Cellular Therapy, Department of Pediatrics, University of Minnesota, Minneapolis, Minnesota, USA
| | - Hans H Wandall
- Copenhagen Center for Glycomics, Department of Cellular and Molecular Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Sally Dabelsteen
- Copenhagen Center for Glycomics, Department of Cellular and Molecular Medicine, University of Copenhagen, Copenhagen, Denmark
| | - David R Liu
- Merkin Institute of Transformative Technologies in Healthcare, The Broad Institute of Harvard and MIT, Cambridge, Massachusetts, USA; Department of Chemistry and Chemical Biology, Harvard University, Cambridge, Massachusetts, USA; Howard Hughes Medical Institute, Harvard University, Cambridge, Massachusetts, USA
| | - Jakub Tolar
- Division of Pediatric Blood and Marrow Transplantation & Cellular Therapy, Department of Pediatrics, University of Minnesota, Minneapolis, Minnesota, USA
| | - Mark J Osborn
- Division of Pediatric Blood and Marrow Transplantation & Cellular Therapy, Department of Pediatrics, University of Minnesota, Minneapolis, Minnesota, USA.
| |
Collapse
|
2
|
Gariballa N, Mohamed F, Badawi S, Ali BR. The double whammy of ER-retention and dominant-negative effects in numerous autosomal dominant diseases: significance in disease mechanisms and therapy. J Biomed Sci 2024; 31:64. [PMID: 38937821 PMCID: PMC11210014 DOI: 10.1186/s12929-024-01054-1] [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: 03/24/2024] [Accepted: 06/20/2024] [Indexed: 06/29/2024] Open
Abstract
The endoplasmic reticulum (ER) employs stringent quality control mechanisms to ensure the integrity of protein folding, allowing only properly folded, processed and assembled proteins to exit the ER and reach their functional destinations. Mutant proteins unable to attain their correct tertiary conformation or form complexes with their partners are retained in the ER and subsequently degraded through ER-associated protein degradation (ERAD) and associated mechanisms. ER retention contributes to a spectrum of monogenic diseases with diverse modes of inheritance and molecular mechanisms. In autosomal dominant diseases, when mutant proteins get retained in the ER, they can interact with their wild-type counterparts. This interaction may lead to the formation of mixed dimers or aberrant complexes, disrupting their normal trafficking and function in a dominant-negative manner. The combination of ER retention and dominant-negative effects has been frequently documented to cause a significant loss of functional proteins, thereby exacerbating disease severity. This review aims to examine existing literature and provide insights into the impact of dominant-negative effects exerted by mutant proteins retained in the ER in a range of autosomal dominant diseases including skeletal and connective tissue disorders, vascular disorders, neurological disorders, eye disorders and serpinopathies. Most crucially, we aim to emphasize the importance of this area of research, offering substantial potential for understanding the factors influencing phenotypic variability associated with genetic variants. Furthermore, we highlight current and prospective therapeutic approaches targeted at ameliorating the effects of mutations exhibiting dominant-negative effects. These approaches encompass experimental studies exploring treatments and their translation into clinical practice.
Collapse
Affiliation(s)
- Nesrin Gariballa
- Department of Genetics and Genomics, College of Medicine and Health Sciences, United Arab Emirates University, P.O. Box: 15551, Al-Ain, United Arab Emirates
| | - Feda Mohamed
- Department of Genetics and Genomics, College of Medicine and Health Sciences, United Arab Emirates University, P.O. Box: 15551, Al-Ain, United Arab Emirates
- ASPIRE Precision Medicine Research Institute Abu Dhabi, United Arab Emirates University, Abu Dhabi, United Arab Emirates
| | - Sally Badawi
- Department of Genetics and Genomics, College of Medicine and Health Sciences, United Arab Emirates University, P.O. Box: 15551, Al-Ain, United Arab Emirates
| | - Bassam R Ali
- Department of Genetics and Genomics, College of Medicine and Health Sciences, United Arab Emirates University, P.O. Box: 15551, Al-Ain, United Arab Emirates.
- ASPIRE Precision Medicine Research Institute Abu Dhabi, United Arab Emirates University, Abu Dhabi, United Arab Emirates.
| |
Collapse
|
3
|
Ramesh A, Hongal A, Srinivasa M, Desai S, Mala R, Jayashankar CK, Abhigna R, Jyothi V, Asha K, Meenakshi B, Ravi H, Gurudatta BV. Clinical and molecular studies in two patients with dystrophic epidermolysis bullosa. Indian J Dermatol Venereol Leprol 2023; 89:880-883. [PMID: 37067131 DOI: 10.25259/ijdvl_225_2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Accepted: 09/01/2022] [Indexed: 03/31/2023]
Affiliation(s)
- Asha Ramesh
- Department of Dermatology, Bangalore Medical College and Research Institute, Bengaluru, Karnataka, India
| | - Amrita Hongal
- Department of Dermatology, Bangalore Medical College and Research Institute, Bengaluru, Karnataka, India
| | | | - Sheetal Desai
- Centre for Human Genetics, Bengaluru, Karnataka, India
| | - R Mala
- Centre for Human Genetics, Bengaluru, Karnataka, India
| | | | - Rai Abhigna
- Department of Dermatology, Bangalore Medical College and Research Institute, Bengaluru, Karnataka, India
| | - Vishwanth Jyothi
- Department of Dermatology, Bangalore Medical College and Research Institute, Bengaluru, Karnataka, India
| | - Kubba Asha
- Department of Dermatology, Bangalore Medical College and Research Institute, Bengaluru, Karnataka, India
| | | | | | | |
Collapse
|
4
|
Guide SV, Gonzalez ME, Bağcı IS, Agostini B, Chen H, Feeney G, Steimer M, Kapadia B, Sridhar K, Quesada Sanchez L, Gonzalez F, Van Ligten M, Parry TJ, Chitra S, Kammerman LA, Krishnan S, Marinkovich MP. Trial of Beremagene Geperpavec (B-VEC) for Dystrophic Epidermolysis Bullosa. N Engl J Med 2022; 387:2211-2219. [PMID: 36516090 DOI: 10.1056/nejmoa2206663] [Citation(s) in RCA: 85] [Impact Index Per Article: 28.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
BACKGROUND Dystrophic epidermolysis bullosa is a rare genetic blistering skin disease caused by mutations in COL7A1, which encodes type VII collagen (C7). Beremagene geperpavec (B-VEC) is a topical investigational herpes simplex virus type 1 (HSV-1)-based gene therapy designed to restore C7 protein by delivering COL7A1. METHODS We conducted a phase 3, double-blind, intrapatient randomized, placebo-controlled trial involving patients 6 months of age or older with genetically confirmed dystrophic epidermolysis bullosa. For each patient, a primary wound pair was selected, with the wounds matched according to size, region, and appearance. The wounds within each pair were randomly assigned in a 1:1 ratio to receive weekly application of either B-VEC or placebo for 26 weeks. The primary end point was complete wound healing of treated as compared with untreated wounds at 6 months. Secondary end points included complete wound healing at 3 months and the change from baseline to weeks 22, 24, and 26 in pain severity during changes in wound dressing, assessed with the use of a visual analogue scale (scores range from 0 to 10, with higher scores indicating greater pain). RESULTS Primary wound pairs were exposed to B-VEC and placebo in 31 patients. At 6 months, complete wound healing occurred in 67% of the wounds exposed to B-VEC as compared with 22% of those exposed to placebo (difference, 46 percentage points; 95% confidence interval [CI], 24 to 68; P = 0.002). Complete wound healing at 3 months occurred in 71% of the wounds exposed to B-VEC as compared with 20% of those exposed to placebo (difference, 51 percentage points; 95% CI, 29 to 73; P<0.001). The mean change from baseline to week 22 in pain severity during wound-dressing changes was -0.88 with B-VEC and -0.71 with placebo (adjusted least-squares mean difference, -0.61; 95% CI, -1.10 to -0.13); similar mean changes were observed at weeks 24 and 26. Adverse events with B-VEC and placebo included pruritus and chills. CONCLUSIONS Complete wound healing at 3 and 6 months in patients with dystrophic epidermolysis bullosa was more likely with topical administration of B-VEC than with placebo. Pruritus and mild systemic side effects were observed in patients treated with B-VEC. Longer and larger trials are warranted to determine the durability and side effects of B-VEC for this disease. (Funded by Krystal Biotech; GEM-3 ClinicalTrials.gov number, NCT04491604.).
Collapse
Affiliation(s)
- Shireen V Guide
- From the Mission Dermatology Center, Department of Dermatology, Children's Hospital of Orange County, University of California Irvine, Rancho Santa Margarita (S.V.G., M.V.L.), the Department of Dermatology, Stanford University School of Medicine, Stanford (I.S.B., K.S., M.P.M.), and the Veterans Affairs Medical Center, Palo Alto (M.P.M.) - all in California; Pediatric Skin Research, Coral Gables, FL (M.E.G., L.Q.S., F.G.); Krystal Biotech, Pittsburgh (B.A., H.C., G.F., M.S., B.K., T.J.P., S.K.); Savio Group Analytics, Hockessin, DE (S.C.); and Kammerman Consulting, Chevy Chase, MD (L.A.K.)
| | - Mercedes E Gonzalez
- From the Mission Dermatology Center, Department of Dermatology, Children's Hospital of Orange County, University of California Irvine, Rancho Santa Margarita (S.V.G., M.V.L.), the Department of Dermatology, Stanford University School of Medicine, Stanford (I.S.B., K.S., M.P.M.), and the Veterans Affairs Medical Center, Palo Alto (M.P.M.) - all in California; Pediatric Skin Research, Coral Gables, FL (M.E.G., L.Q.S., F.G.); Krystal Biotech, Pittsburgh (B.A., H.C., G.F., M.S., B.K., T.J.P., S.K.); Savio Group Analytics, Hockessin, DE (S.C.); and Kammerman Consulting, Chevy Chase, MD (L.A.K.)
| | - I Sinem Bağcı
- From the Mission Dermatology Center, Department of Dermatology, Children's Hospital of Orange County, University of California Irvine, Rancho Santa Margarita (S.V.G., M.V.L.), the Department of Dermatology, Stanford University School of Medicine, Stanford (I.S.B., K.S., M.P.M.), and the Veterans Affairs Medical Center, Palo Alto (M.P.M.) - all in California; Pediatric Skin Research, Coral Gables, FL (M.E.G., L.Q.S., F.G.); Krystal Biotech, Pittsburgh (B.A., H.C., G.F., M.S., B.K., T.J.P., S.K.); Savio Group Analytics, Hockessin, DE (S.C.); and Kammerman Consulting, Chevy Chase, MD (L.A.K.)
| | - Brittani Agostini
- From the Mission Dermatology Center, Department of Dermatology, Children's Hospital of Orange County, University of California Irvine, Rancho Santa Margarita (S.V.G., M.V.L.), the Department of Dermatology, Stanford University School of Medicine, Stanford (I.S.B., K.S., M.P.M.), and the Veterans Affairs Medical Center, Palo Alto (M.P.M.) - all in California; Pediatric Skin Research, Coral Gables, FL (M.E.G., L.Q.S., F.G.); Krystal Biotech, Pittsburgh (B.A., H.C., G.F., M.S., B.K., T.J.P., S.K.); Savio Group Analytics, Hockessin, DE (S.C.); and Kammerman Consulting, Chevy Chase, MD (L.A.K.)
| | - Hubert Chen
- From the Mission Dermatology Center, Department of Dermatology, Children's Hospital of Orange County, University of California Irvine, Rancho Santa Margarita (S.V.G., M.V.L.), the Department of Dermatology, Stanford University School of Medicine, Stanford (I.S.B., K.S., M.P.M.), and the Veterans Affairs Medical Center, Palo Alto (M.P.M.) - all in California; Pediatric Skin Research, Coral Gables, FL (M.E.G., L.Q.S., F.G.); Krystal Biotech, Pittsburgh (B.A., H.C., G.F., M.S., B.K., T.J.P., S.K.); Savio Group Analytics, Hockessin, DE (S.C.); and Kammerman Consulting, Chevy Chase, MD (L.A.K.)
| | - Gloria Feeney
- From the Mission Dermatology Center, Department of Dermatology, Children's Hospital of Orange County, University of California Irvine, Rancho Santa Margarita (S.V.G., M.V.L.), the Department of Dermatology, Stanford University School of Medicine, Stanford (I.S.B., K.S., M.P.M.), and the Veterans Affairs Medical Center, Palo Alto (M.P.M.) - all in California; Pediatric Skin Research, Coral Gables, FL (M.E.G., L.Q.S., F.G.); Krystal Biotech, Pittsburgh (B.A., H.C., G.F., M.S., B.K., T.J.P., S.K.); Savio Group Analytics, Hockessin, DE (S.C.); and Kammerman Consulting, Chevy Chase, MD (L.A.K.)
| | - Molly Steimer
- From the Mission Dermatology Center, Department of Dermatology, Children's Hospital of Orange County, University of California Irvine, Rancho Santa Margarita (S.V.G., M.V.L.), the Department of Dermatology, Stanford University School of Medicine, Stanford (I.S.B., K.S., M.P.M.), and the Veterans Affairs Medical Center, Palo Alto (M.P.M.) - all in California; Pediatric Skin Research, Coral Gables, FL (M.E.G., L.Q.S., F.G.); Krystal Biotech, Pittsburgh (B.A., H.C., G.F., M.S., B.K., T.J.P., S.K.); Savio Group Analytics, Hockessin, DE (S.C.); and Kammerman Consulting, Chevy Chase, MD (L.A.K.)
| | - Binoy Kapadia
- From the Mission Dermatology Center, Department of Dermatology, Children's Hospital of Orange County, University of California Irvine, Rancho Santa Margarita (S.V.G., M.V.L.), the Department of Dermatology, Stanford University School of Medicine, Stanford (I.S.B., K.S., M.P.M.), and the Veterans Affairs Medical Center, Palo Alto (M.P.M.) - all in California; Pediatric Skin Research, Coral Gables, FL (M.E.G., L.Q.S., F.G.); Krystal Biotech, Pittsburgh (B.A., H.C., G.F., M.S., B.K., T.J.P., S.K.); Savio Group Analytics, Hockessin, DE (S.C.); and Kammerman Consulting, Chevy Chase, MD (L.A.K.)
| | - Kunju Sridhar
- From the Mission Dermatology Center, Department of Dermatology, Children's Hospital of Orange County, University of California Irvine, Rancho Santa Margarita (S.V.G., M.V.L.), the Department of Dermatology, Stanford University School of Medicine, Stanford (I.S.B., K.S., M.P.M.), and the Veterans Affairs Medical Center, Palo Alto (M.P.M.) - all in California; Pediatric Skin Research, Coral Gables, FL (M.E.G., L.Q.S., F.G.); Krystal Biotech, Pittsburgh (B.A., H.C., G.F., M.S., B.K., T.J.P., S.K.); Savio Group Analytics, Hockessin, DE (S.C.); and Kammerman Consulting, Chevy Chase, MD (L.A.K.)
| | - Lori Quesada Sanchez
- From the Mission Dermatology Center, Department of Dermatology, Children's Hospital of Orange County, University of California Irvine, Rancho Santa Margarita (S.V.G., M.V.L.), the Department of Dermatology, Stanford University School of Medicine, Stanford (I.S.B., K.S., M.P.M.), and the Veterans Affairs Medical Center, Palo Alto (M.P.M.) - all in California; Pediatric Skin Research, Coral Gables, FL (M.E.G., L.Q.S., F.G.); Krystal Biotech, Pittsburgh (B.A., H.C., G.F., M.S., B.K., T.J.P., S.K.); Savio Group Analytics, Hockessin, DE (S.C.); and Kammerman Consulting, Chevy Chase, MD (L.A.K.)
| | - Franshesca Gonzalez
- From the Mission Dermatology Center, Department of Dermatology, Children's Hospital of Orange County, University of California Irvine, Rancho Santa Margarita (S.V.G., M.V.L.), the Department of Dermatology, Stanford University School of Medicine, Stanford (I.S.B., K.S., M.P.M.), and the Veterans Affairs Medical Center, Palo Alto (M.P.M.) - all in California; Pediatric Skin Research, Coral Gables, FL (M.E.G., L.Q.S., F.G.); Krystal Biotech, Pittsburgh (B.A., H.C., G.F., M.S., B.K., T.J.P., S.K.); Savio Group Analytics, Hockessin, DE (S.C.); and Kammerman Consulting, Chevy Chase, MD (L.A.K.)
| | - Matthew Van Ligten
- From the Mission Dermatology Center, Department of Dermatology, Children's Hospital of Orange County, University of California Irvine, Rancho Santa Margarita (S.V.G., M.V.L.), the Department of Dermatology, Stanford University School of Medicine, Stanford (I.S.B., K.S., M.P.M.), and the Veterans Affairs Medical Center, Palo Alto (M.P.M.) - all in California; Pediatric Skin Research, Coral Gables, FL (M.E.G., L.Q.S., F.G.); Krystal Biotech, Pittsburgh (B.A., H.C., G.F., M.S., B.K., T.J.P., S.K.); Savio Group Analytics, Hockessin, DE (S.C.); and Kammerman Consulting, Chevy Chase, MD (L.A.K.)
| | - Trevor J Parry
- From the Mission Dermatology Center, Department of Dermatology, Children's Hospital of Orange County, University of California Irvine, Rancho Santa Margarita (S.V.G., M.V.L.), the Department of Dermatology, Stanford University School of Medicine, Stanford (I.S.B., K.S., M.P.M.), and the Veterans Affairs Medical Center, Palo Alto (M.P.M.) - all in California; Pediatric Skin Research, Coral Gables, FL (M.E.G., L.Q.S., F.G.); Krystal Biotech, Pittsburgh (B.A., H.C., G.F., M.S., B.K., T.J.P., S.K.); Savio Group Analytics, Hockessin, DE (S.C.); and Kammerman Consulting, Chevy Chase, MD (L.A.K.)
| | - Surya Chitra
- From the Mission Dermatology Center, Department of Dermatology, Children's Hospital of Orange County, University of California Irvine, Rancho Santa Margarita (S.V.G., M.V.L.), the Department of Dermatology, Stanford University School of Medicine, Stanford (I.S.B., K.S., M.P.M.), and the Veterans Affairs Medical Center, Palo Alto (M.P.M.) - all in California; Pediatric Skin Research, Coral Gables, FL (M.E.G., L.Q.S., F.G.); Krystal Biotech, Pittsburgh (B.A., H.C., G.F., M.S., B.K., T.J.P., S.K.); Savio Group Analytics, Hockessin, DE (S.C.); and Kammerman Consulting, Chevy Chase, MD (L.A.K.)
| | - Lisa A Kammerman
- From the Mission Dermatology Center, Department of Dermatology, Children's Hospital of Orange County, University of California Irvine, Rancho Santa Margarita (S.V.G., M.V.L.), the Department of Dermatology, Stanford University School of Medicine, Stanford (I.S.B., K.S., M.P.M.), and the Veterans Affairs Medical Center, Palo Alto (M.P.M.) - all in California; Pediatric Skin Research, Coral Gables, FL (M.E.G., L.Q.S., F.G.); Krystal Biotech, Pittsburgh (B.A., H.C., G.F., M.S., B.K., T.J.P., S.K.); Savio Group Analytics, Hockessin, DE (S.C.); and Kammerman Consulting, Chevy Chase, MD (L.A.K.)
| | - Suma Krishnan
- From the Mission Dermatology Center, Department of Dermatology, Children's Hospital of Orange County, University of California Irvine, Rancho Santa Margarita (S.V.G., M.V.L.), the Department of Dermatology, Stanford University School of Medicine, Stanford (I.S.B., K.S., M.P.M.), and the Veterans Affairs Medical Center, Palo Alto (M.P.M.) - all in California; Pediatric Skin Research, Coral Gables, FL (M.E.G., L.Q.S., F.G.); Krystal Biotech, Pittsburgh (B.A., H.C., G.F., M.S., B.K., T.J.P., S.K.); Savio Group Analytics, Hockessin, DE (S.C.); and Kammerman Consulting, Chevy Chase, MD (L.A.K.)
| | - M Peter Marinkovich
- From the Mission Dermatology Center, Department of Dermatology, Children's Hospital of Orange County, University of California Irvine, Rancho Santa Margarita (S.V.G., M.V.L.), the Department of Dermatology, Stanford University School of Medicine, Stanford (I.S.B., K.S., M.P.M.), and the Veterans Affairs Medical Center, Palo Alto (M.P.M.) - all in California; Pediatric Skin Research, Coral Gables, FL (M.E.G., L.Q.S., F.G.); Krystal Biotech, Pittsburgh (B.A., H.C., G.F., M.S., B.K., T.J.P., S.K.); Savio Group Analytics, Hockessin, DE (S.C.); and Kammerman Consulting, Chevy Chase, MD (L.A.K.)
| |
Collapse
|
5
|
Morren MA, Legius E, Giuliano F, Hadj-Rabia S, Hohl D, Bodemer C. Challenges in Treating Genodermatoses: New Therapies at the Horizon. Front Pharmacol 2022; 12:746664. [PMID: 35069188 PMCID: PMC8766835 DOI: 10.3389/fphar.2021.746664] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2021] [Accepted: 11/29/2021] [Indexed: 01/28/2023] Open
Abstract
Genodermatoses are rare inherited skin diseases that frequently affect other organs. They often have marked effects on wellbeing and may cause early death. Progress in molecular genetics and translational research has unravelled many underlying pathological mechanisms, and in several disorders with high unmet need, has opened the way for the introduction of innovative treatments. One approach is to intervene where cell-signaling pathways are dysregulated, in the case of overactive pathways by the use of selective inhibitors, or when the activity of an essential factor is decreased by augmenting a molecular component to correct disequilibrium in the pathway. Where inflammatory reactions have been induced by a genetically altered protein, another possible approach is to suppress the inflammation directly. Depending on the nature of the genodermatosis, the implicated protein or even on the particular mutation, to correct the consequences or the genetic defect, may require a highly personalised stratagem. Repurposed drugs, can be used to bring about a "read through" strategy especially where the genetic defect induces premature termination codons. Sometimes the defective protein can be replaced by a normal functioning one. Cell therapies with allogeneic normal keratinocytes or fibroblasts may restore the integrity of diseased skin and allogeneic bone marrow or mesenchymal cells may additionally rescue other affected organs. Genetic engineering is expanding rapidly. The insertion of a normal functioning gene into cells of the recipient is since long explored. More recently, genome editing, allows reframing, insertion or deletion of exons or disruption of aberrantly functioning genes. There are now several examples where these stratagems are being explored in the (pre)clinical phase of therapeutic trial programmes. Another stratagem, designed to reduce the severity of a given disease involves the use of RNAi to attenuate expression of a harmful protein by decreasing abundance of the cognate transcript. Most of these strategies are short-lasting and will thus require intermittent life-long administration. In contrast, insertion of healthy copies of the relevant gene or editing the disease locus in the genome to correct harmful mutations in stem cells is more likely to induce a permanent cure. Here we discuss the potential advantages and drawbacks of applying these technologies in patients with these genetic conditions. Given the severity of many genodermatoses, prevention of transmission to future generations remains an important goal including offering reproductive choices, such as preimplantation genetic testing, which can allow selection of an unaffected embryo for transfer to the uterus.
Collapse
Affiliation(s)
- Marie-Anne Morren
- Pediatric Dermatology Unit, Departments of Dermatology and Venereology and Pediatrics, University Hospital Lausanne, University of Lausanne, Lausanne, Switzerland
| | - Eric Legius
- Department for Human Genetics, University Hospitals Leuven, KU Leuven, ERN Genturis and ERN Skin, Leuven, Belgium
| | - Fabienne Giuliano
- Department of Medical Genetics, University Hospital Lausanne, Lausanne, Switzerland
| | - Smail Hadj-Rabia
- Department of Pediatric Dermatology and Dermatology, National Reference Centre for Genodermatosis and Rare Diseases of the Skin (MAGEC), Hôpital Necker-Enfants Malades, and Assistance Publique-Hôpitaux de Paris, Université Paris Descartes, ERN Skin, Paris, France
| | - Daniel Hohl
- Department of Dermatology and Venereology, University Hospital Lausanne, University of Lausanne, Lausanne, Switzerland
| | - Christine Bodemer
- Department of Pediatric Dermatology and Dermatology, National Reference Centre for Genodermatosis and Rare Diseases of the Skin (MAGEC), Hôpital Necker-Enfants Malades, and Assistance Publique-Hôpitaux de Paris, Université Paris Descartes, ERN Skin, Paris, France
| |
Collapse
|
6
|
Welponer T, Prodinger C, Pinon-Hofbauer J, Hintersteininger A, Breitenbach-Koller H, Bauer JW, Laimer M. Clinical Perspectives of Gene-Targeted Therapies for Epidermolysis Bullosa. Dermatol Ther (Heidelb) 2021; 11:1175-1197. [PMID: 34110606 PMCID: PMC8322229 DOI: 10.1007/s13555-021-00561-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Indexed: 02/06/2023] Open
Abstract
New insights into molecular genetics and pathomechanisms in epidermolysis bullosa (EB), methodological and technological advances in molecular biology as well as designated funding initiatives and facilitated approval procedures for orphan drugs have boosted translational research perspectives for this devastating disease. This is echoed by the increasing number of clinical trials assessing innovative molecular therapies in the field of EB. Despite remarkable progress, gene-corrective modalities, aimed at sustained or permanent restoration of functional protein expression, still await broad clinical availability. This also reflects the methodological and technological shortcomings of current strategies, including the translatability of certain methodologies beyond preclinical models as well as the safe, specific, efficient, feasible, sustained and cost-effective delivery of therapeutic/corrective information to target cells. This review gives an updated overview on status, prospects, challenges and limitations of current gene-targeted therapies.
Collapse
Affiliation(s)
- Tobias Welponer
- Department of Dermatology and Allergology and EB House Austria, University Hospital of the Paracelsus Medical University, Salzburg, Austria
| | - Christine Prodinger
- Department of Dermatology and Allergology and EB House Austria, University Hospital of the Paracelsus Medical University, Salzburg, Austria
| | - Josefina Pinon-Hofbauer
- Department of Dermatology and Allergology and EB House Austria, University Hospital of the Paracelsus Medical University, Salzburg, Austria
| | - Arno Hintersteininger
- Department of Dermatology and Allergology, University Hospital of the Paracelsus Medical University, Salzburg, Austria
| | | | - Johann W Bauer
- Department of Dermatology and Allergology and EB House Austria, University Hospital of the Paracelsus Medical University, Salzburg, Austria
- Department of Biosciences, Paris Lodron University of Salzburg, Salzburg, Austria
| | - Martin Laimer
- Department of Dermatology and Allergology and EB House Austria, University Hospital of the Paracelsus Medical University, Salzburg, Austria.
| |
Collapse
|
7
|
Mutant collagen COL11A1 enhances cancerous invasion. Oncogene 2021; 40:6299-6307. [PMID: 34584216 PMCID: PMC8566234 DOI: 10.1038/s41388-021-02013-y] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 08/17/2021] [Accepted: 09/07/2021] [Indexed: 02/08/2023]
Abstract
Collagens are the most abundant proteins in the body and comprise the basement membranes and stroma through which cancerous invasion occurs; however, a pro-neoplastic function for mutant collagens is undefined. Here we identify COL11A1 mutations in 66 of 100 cutaneous squamous cell carcinomas (cSCCs), the second most common U.S. cancer, concentrated in a triple helical region known to produce trans-dominant collagens. Analysis of COL11A1 and other collagen genes found that they are mutated across common epithelial malignancies. Knockout of mutant COL11A1 impairs cSCC tumorigenesis in vivo. Compared to otherwise genetically identical COL11A1 wild-type tissue, gene-edited mutant COL11A1 skin is characterized by induction of β1 integrin targets and accelerated neoplastic invasion. In mosaic tissue, mutant COL11A1 cells enhanced invasion by neighboring wild-type cells. These results suggest that specific collagens are commonly mutated in cancer and that mutant collagens may accelerate this process.
Collapse
|
8
|
Bornert O, Hogervorst M, Nauroy P, Bischof J, Swildens J, Athanasiou I, Tufa SF, Keene DR, Kiritsi D, Hainzl S, Murauer EM, Marinkovich MP, Platenburg G, Hausser I, Wally V, Ritsema T, Koller U, Haisma EM, Nyström A. QR-313, an Antisense Oligonucleotide, Shows Therapeutic Efficacy for Treatment of Dominant and Recessive Dystrophic Epidermolysis Bullosa: A Preclinical Study. J Invest Dermatol 2020; 141:883-893.e6. [PMID: 32946877 DOI: 10.1016/j.jid.2020.08.018] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Revised: 07/21/2020] [Accepted: 08/12/2020] [Indexed: 02/06/2023]
Abstract
Dystrophic epidermolysis bullosa (DEB) is a blistering skin disease caused by mutations in the gene COL7A1 encoding collagen VII. DEB can be inherited as recessive DEB (RDEB) or dominant DEB (DDEB) and is associated with a high wound burden. Perpetual cycles of wounding and healing drive fibrosis in DDEB and RDEB, as well as the formation of a tumor-permissive microenvironment. Prolonging wound-free episodes by improving the quality of wound healing would therefore confer substantial benefit for individuals with DEB. The collagenous domain of collagen VII is encoded by 82 in-frame exons, which makes splice-modulation therapies attractive for DEB. Indeed, antisense oligonucleotide-based exon skipping has shown promise for RDEB. However, the suitability of antisense oligonucleotides for treatment of DDEB remains unexplored. Here, we developed QR-313, a clinically applicable, potent antisense oligonucleotide specifically targeting exon 73. We show the feasibility of topical delivery of QR-313 in a carbomer-composed gel for treatment of wounds to restore collagen VII abundance in human RDEB skin. Our data reveal that QR-313 also shows direct benefit for DDEB caused by exon 73 mutations. Thus, the same topically applied therapeutic could be used to improve the wound healing quality in RDEB and DDEB.
Collapse
Affiliation(s)
- Olivier Bornert
- Department of Dermatology, Medical Faculty, Medical Center - University of Freiburg, Freiburg, Germany
| | | | - Pauline Nauroy
- Department of Dermatology, Medical Faculty, Medical Center - University of Freiburg, Freiburg, Germany
| | - Johannes Bischof
- EB House Austria, Research Program for Molecular Therapy of Genodermatoses, Department of Dermatology and Allergology, University Hospital of the Paracelsus Medical University Salzburg, Salzburg, Austria
| | - Jim Swildens
- ProQR Therapeutics N.V., Leiden, The Netherlands
| | - Ioannis Athanasiou
- Department of Dermatology, Medical Faculty, Medical Center - University of Freiburg, Freiburg, Germany
| | - Sara F Tufa
- Micro-Imaging Center, Shriners Hospital for Children, Portland, Oregon, USA
| | - Douglas R Keene
- Micro-Imaging Center, Shriners Hospital for Children, Portland, Oregon, USA
| | - Dimitra Kiritsi
- Department of Dermatology, Medical Faculty, Medical Center - University of Freiburg, Freiburg, Germany
| | - Stefan Hainzl
- EB House Austria, Research Program for Molecular Therapy of Genodermatoses, Department of Dermatology and Allergology, University Hospital of the Paracelsus Medical University Salzburg, Salzburg, Austria
| | - Eva M Murauer
- EB House Austria, Research Program for Molecular Therapy of Genodermatoses, Department of Dermatology and Allergology, University Hospital of the Paracelsus Medical University Salzburg, Salzburg, Austria
| | - M Peter Marinkovich
- Department of Dermatology, Stanford University School of Medicine, Stanford, California, USA; Dermatology, Veteran's Affairs Medical Center, Palo Alto, California, USA
| | | | - Ingrid Hausser
- Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany
| | - Verena Wally
- EB House Austria, Research Program for Molecular Therapy of Genodermatoses, Department of Dermatology and Allergology, University Hospital of the Paracelsus Medical University Salzburg, Salzburg, Austria
| | - Tita Ritsema
- ProQR Therapeutics N.V., Leiden, The Netherlands
| | - Ulrich Koller
- EB House Austria, Research Program for Molecular Therapy of Genodermatoses, Department of Dermatology and Allergology, University Hospital of the Paracelsus Medical University Salzburg, Salzburg, Austria
| | | | - Alexander Nyström
- Department of Dermatology, Medical Faculty, Medical Center - University of Freiburg, Freiburg, Germany.
| |
Collapse
|
9
|
Bremer J, van der Heijden EH, Eichhorn DS, Meijer R, Lemmink HH, Scheffer H, Sinke RJ, Jonkman MF, Pasmooij AMG, Van den Akker PC. Natural Exon Skipping Sets the Stage for Exon Skipping as Therapy for Dystrophic Epidermolysis Bullosa. MOLECULAR THERAPY. NUCLEIC ACIDS 2019; 18:465-475. [PMID: 31670143 PMCID: PMC6831832 DOI: 10.1016/j.omtn.2019.09.009] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Revised: 09/10/2019] [Accepted: 09/10/2019] [Indexed: 02/04/2023]
Abstract
Dystrophic epidermolysis bullosa (DEB) is a devastating blistering disease affecting skin and mucous membranes. It is caused by pathogenic variants in the COL7A1 gene encoding type VII collagen, and can be inherited dominantly or recessively. Recently, promising proof-of-principle has been shown for antisense oligonucleotide (AON)-mediated exon skipping as a therapeutic approach for DEB. However, the precise phenotypic effect to be anticipated from exon skipping, and which patient groups could benefit, is not yet clear. To answer these questions, we studied new clinical and molecular data on seven patients from the Dutch EB registry and reviewed the literature on COL7A1 exon skipping variants. We found that phenotypes associated with dominant exon skipping cannot be distinguished from phenotypes caused by other dominant DEB variants. Recessive exon skipping phenotypes are generally relatively mild in the spectrum of recessive DEB. Therefore, for dominant DEB, AON-mediated exon skipping is unlikely to ameliorate the phenotype. In contrast, the overall severity of phenotypes associated with recessive natural exon skipping pivots toward the milder end of the spectrum. Consequently, we anticipate AON-mediated exon skipping for recessive DEB caused by bi-allelic null variants should lead to a clinically relevant improvement of this devastating phenotype.
Collapse
Affiliation(s)
- Jeroen Bremer
- Department of Genetics, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands; Department of Dermatology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands.
| | - Elisabeth H van der Heijden
- Department of Genetics, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Daryll S Eichhorn
- Department of Dermatology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Rowdy Meijer
- Department of Human Genetics, Radboud University Nijmegen Medical Centre, Nijmegen, the Netherlands
| | - Henny H Lemmink
- Department of Genetics, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Hans Scheffer
- Department of Human Genetics, Radboud University Nijmegen Medical Centre, Nijmegen, the Netherlands
| | - Richard J Sinke
- Department of Genetics, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Marcel F Jonkman
- Department of Dermatology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Anna M G Pasmooij
- Department of Dermatology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Peter C Van den Akker
- Department of Genetics, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands; Department of Dermatology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| |
Collapse
|
10
|
Abstract
The size and relatively high GC content of cDNAs are challenges for efficient targeted engineering of large collagens. There are both basic biological and therapeutic interests in the ability to modify collagens, as this would allow for studies precisely describing interactions of collagens with specific interaction partners, addressing consequences of individual disease-causing mutations, and assessing therapeutic applicability of precision medicine approaches. Using collagen VII as an example, we will here describe a strategy for rapid and simple modification of cDNAs encoding large collagens. The method is flexible and can be used for the creation of point mutations, small or large deletions, and insertion of DNA.
Collapse
Affiliation(s)
- Olivier Bornert
- Department of Dermatology, Faculty of Medicine, Medical Center, University of Freiburg, Freiburg, Germany
| | - Alexander Nyström
- Department of Dermatology, Faculty of Medicine, Medical Center, University of Freiburg, Freiburg, Germany.
| |
Collapse
|
11
|
Basement membranes in the cornea and other organs that commonly develop fibrosis. Cell Tissue Res 2018; 374:439-453. [PMID: 30284084 DOI: 10.1007/s00441-018-2934-7] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2018] [Accepted: 09/20/2018] [Indexed: 12/19/2022]
Abstract
Basement membranes are thin connective tissue structures composed of organ-specific assemblages of collagens, laminins, proteoglycan-like perlecan, nidogens, and other components. Traditionally, basement membranes are thought of as structures which primarily function to anchor epithelial, endothelial, or parenchymal cells to underlying connective tissues. While this role is important, other functions such as the modulation of growth factors and cytokines that regulate cell proliferation, migration, differentiation, and fibrosis are equally important. An example of this is the critical role of both the epithelial basement membrane and Descemet's basement membrane in the cornea in modulating myofibroblast development and fibrosis, as well as myofibroblast apoptosis and the resolution of fibrosis. This article compares the ultrastructure and functions of key basement membranes in several organs to illustrate the variability and importance of these structures in organs that commonly develop fibrosis.
Collapse
|
12
|
Bornert O, Peking P, Bremer J, Koller U, van den Akker PC, Aartsma-Rus A, Pasmooij AMG, Murauer EM, Nyström A. RNA-based therapies for genodermatoses. Exp Dermatol 2017; 26:3-10. [PMID: 27376675 PMCID: PMC5593095 DOI: 10.1111/exd.13141] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/29/2016] [Indexed: 12/14/2022]
Abstract
Genetic disorders affecting the skin, genodermatoses, constitute a large and heterogeneous group of diseases, for which treatment is generally limited to management of symptoms. RNA-based therapies are emerging as a powerful tool to treat genodermatoses. In this review, we discuss in detail RNA splicing modulation by antisense oligonucleotides and RNA trans-splicing, transcript replacement and genome editing by in vitro-transcribed mRNAs, and gene knockdown by small interfering RNA and antisense oligonucleotides. We present the current state of these therapeutic approaches and critically discuss their opportunities, limitations and the challenges that remain to be solved. The aim of this review was to set the stage for the development of new and better therapies to improve the lives of patients and families affected by a genodermatosis.
Collapse
Affiliation(s)
- Olivier Bornert
- Department of Dermatology, Medical Center – University of
Freiburg, Freiburg, Germany
| | - Patricia Peking
- EB House Austria, Research Program for Molecular Therapy of
Genodermatoses, Department of Dermatology, University Hospital of the Paracelsus
Medical University, Salzburg, Austria
| | - Jeroen Bremer
- Department of Dermatology, University Medical Center Groningen,
University of Groningen, Groningen, The Netherlands
| | - Ulrich Koller
- EB House Austria, Research Program for Molecular Therapy of
Genodermatoses, Department of Dermatology, University Hospital of the Paracelsus
Medical University, Salzburg, Austria
| | - Peter C. van den Akker
- Department of Dermatology, University Medical Center Groningen,
University of Groningen, Groningen, The Netherlands
- Department of Genetics, University Medical Center Groningen,
University of Groningen, Groningen, The Netherlands
| | - Annemieke Aartsma-Rus
- Department of Human Genetics, Leiden University Medical Center,
Leiden, The Netherlands
| | - Anna M. G. Pasmooij
- Department of Dermatology, University Medical Center Groningen,
University of Groningen, Groningen, The Netherlands
| | - Eva M. Murauer
- EB House Austria, Research Program for Molecular Therapy of
Genodermatoses, Department of Dermatology, University Hospital of the Paracelsus
Medical University, Salzburg, Austria
| | - Alexander Nyström
- Department of Dermatology, Medical Center – University of
Freiburg, Freiburg, Germany
| |
Collapse
|
13
|
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: 4.1] [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]
|
14
|
Bornert O, Kühl T, Bremer J, van den Akker PC, Pasmooij AM, Nyström A. Analysis of the functional consequences of targeted exon deletion in COL7A1 reveals prospects for dystrophic epidermolysis bullosa therapy. Mol Ther 2016; 24:1302-11. [PMID: 27157667 DOI: 10.1038/mt.2016.92] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2016] [Accepted: 05/03/2016] [Indexed: 12/18/2022] Open
Abstract
Genetically evoked deficiency of collagen VII causes dystrophic epidermolysis bullosa (DEB)-a debilitating disease characterized by chronic skin fragility and progressive fibrosis. Removal of exons carrying frame-disrupting mutations can reinstate protein expression in genetic diseases. The therapeutic potential of this approach is critically dependent on gene, protein, and disease intrinsic factors. Naturally occurring exon skipping in COL7A1, translating collagen VII, suggests that skipping of exons containing disease-causing mutations may be feasible for the treatment of DEB. However, despite a primarily in-frame arrangement of exons in the COL7A1 gene, no general conclusion of the aptitude of exon skipping for DEB can be drawn, since regulation of collagen VII functionality is complex involving folding, intra- and intermolecular interactions. To directly address this, we deleted two conceptually important exons located at both ends of COL7A1, exon 13, containing recurrent mutations, and exon 105, predicted to impact folding. The resulting recombinantly expressed proteins showed conserved functionality in biochemical and in vitro assays. Injected into DEB mice, the proteins promoted skin stability. By demonstrating functionality of internally deleted collagen VII variants, our study provides support of targeted exon deletion or skipping as a potential therapy to treat a large number of individuals with DEB.
Collapse
Affiliation(s)
- Olivier Bornert
- Department of Dermatology, Medical Center - University of Freiburg, Freiburg, Germany
| | - Tobias Kühl
- Department of Dermatology, Medical Center - University of Freiburg, Freiburg, Germany
| | - Jeroen Bremer
- Department of Dermatology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Peter C van den Akker
- Department of Dermatology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands.,Department of Genetics, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Anna Mg Pasmooij
- Department of Dermatology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Alexander Nyström
- Department of Dermatology, Medical Center - University of Freiburg, Freiburg, Germany
| |
Collapse
|
15
|
Kovács G, Kalmár T, Endreffy E, Ondrik Z, Iványi B, Rikker C, Haszon I, Túri S, Sinkó M, Bereczki C, Maróti Z. Efficient Targeted Next Generation Sequencing-Based Workflow for Differential Diagnosis of Alport-Related Disorders. PLoS One 2016; 11:e0149241. [PMID: 26934356 PMCID: PMC4775026 DOI: 10.1371/journal.pone.0149241] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2015] [Accepted: 01/28/2016] [Indexed: 01/02/2023] Open
Abstract
Alport syndrome (AS) is an inherited type IV collagen nephropathies characterized by microscopic hematuria during early childhood, the development of proteinuria and progression to end-stage renal disease. Since choosing the right therapy, even before the onset of proteinuria, can delay the onset of end-stage renal failure and improve life expectancy, the earliest possible differential diagnosis is desired. Practically, this means the identification of mutation(s) in COL4A3-A4-A5 genes. We used an efficient, next generation sequencing based workflow for simultaneous analysis of all three COL4A genes in three individuals and fourteen families involved by AS or showing different level of Alport-related symptoms. We successfully identified mutations in all investigated cases, including 14 unpublished mutations in our Hungarian cohort. We present an easy to use unified clinical/diagnostic terminology and workflow not only for X-linked but for autosomal AS, but also for Alport-related diseases. In families where a diagnosis has been established by molecular genetic analysis, the renal biopsy may be rendered unnecessary.
Collapse
Affiliation(s)
- Gábor Kovács
- University of Szeged, Faculty of Medicine, Department of Pediatrics and Pediatric Health Center, Szeged, Hungary
| | - Tibor Kalmár
- University of Szeged, Faculty of Medicine, Department of Pediatrics and Pediatric Health Center, Szeged, Hungary
| | - Emőke Endreffy
- University of Szeged, Faculty of Medicine, Department of Pediatrics and Pediatric Health Center, Szeged, Hungary
| | - Zoltán Ondrik
- University of Szeged, Faculty of Medicine, First Department of Internal Medicine, Szeged, Hungary
| | - Béla Iványi
- University of Szeged, Faculty of Medicine, Department of Pathology, Szeged, Hungary
| | - Csaba Rikker
- Péterfy Sándor Hospital Department of Internal Medicine 1, Budapest, Hungary
| | - Ibolya Haszon
- University of Szeged, Faculty of Medicine, Department of Pediatrics and Pediatric Health Center, Szeged, Hungary
| | - Sándor Túri
- University of Szeged, Faculty of Medicine, Department of Pediatrics and Pediatric Health Center, Szeged, Hungary
| | - Mária Sinkó
- University of Szeged, Faculty of Medicine, Department of Pediatrics and Pediatric Health Center, Szeged, Hungary
| | - Csaba Bereczki
- University of Szeged, Faculty of Medicine, Department of Pediatrics and Pediatric Health Center, Szeged, Hungary
| | - Zoltán Maróti
- University of Szeged, Faculty of Medicine, Department of Pediatrics and Pediatric Health Center, Szeged, Hungary
| |
Collapse
|
16
|
Shen J, Zhang J, Wang Z, Ni C, Li H, Cheng R, Liang J, Li M, Yao Z. Gene diagnosis and prenatal genetic diagnosis of a case of dystrophic epidermolysis bullosa family caused by gonadosomatic mosaicism for the COL7A1 mutation p.Gly2043Arg in the pregnant mother. J Eur Acad Dermatol Venereol 2015; 30:1627-9. [PMID: 26289024 DOI: 10.1111/jdv.13229] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- J Shen
- Department of Dermatology, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - J Zhang
- Department of Dermatology, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Z Wang
- Department of Dermatology, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - C Ni
- Department of Dermatology, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - H Li
- Department of Dermatology, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - R Cheng
- Department of Dermatology, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - J Liang
- Department of Dermatology, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - M Li
- Department of Dermatology, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Z Yao
- Department of Dermatology, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| |
Collapse
|
17
|
van den Akker P, Pasmooij A, Meijer R, Scheffer H, Jonkman M. Somatic mosaicism for theCOL7A1mutation p.Gly2034Arg in the unaffected mother of a patient with dystrophic epidermolysis bullosa pruriginosa. Br J Dermatol 2015; 172:778-81. [DOI: 10.1111/bjd.13336] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/03/2014] [Indexed: 12/14/2022]
Affiliation(s)
- P.C. van den Akker
- Department of Dermatology; University of Groningen; University Medical Center Groningen; The Netherlands
- Department of Genetics; University of Groningen; University Medical Center Groningen; The Netherlands
| | - A.M.G. Pasmooij
- Department of Dermatology; University of Groningen; University Medical Center Groningen; The Netherlands
| | - R. Meijer
- Department of Human Genetics; Radboud University Medical Center; Nijmegen The Netherlands
| | - H. Scheffer
- Department of Human Genetics; Radboud University Medical Center; Nijmegen The Netherlands
| | - M.F. Jonkman
- Department of Dermatology; University of Groningen; University Medical Center Groningen; The Netherlands
| |
Collapse
|
18
|
Koller U, Hainzl S, Kocher T, Hüttner C, Klausegger A, Gruber C, Mayr E, Wally V, Bauer JW, Murauer EM. Trans-splicing improvement by the combined application of antisense strategies. Int J Mol Sci 2015; 16:1179-91. [PMID: 25569093 PMCID: PMC4307297 DOI: 10.3390/ijms16011179] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2014] [Accepted: 12/25/2014] [Indexed: 11/16/2022] Open
Abstract
Spliceosome-mediated RNA trans-splicing has become an emergent tool for the repair of mutated pre-mRNAs in the treatment of genetic diseases. RNA trans-splicing molecules (RTMs) are designed to induce a specific trans-splicing reaction via a binding domain for a respective target pre-mRNA region. A previously established reporter-based screening system allows us to analyze the impact of various factors on the RTM trans-splicing efficiency in vitro. Using this system, we are further able to investigate the potential of antisense RNAs (AS RNAs), presuming to improve the trans-splicing efficiency of a selected RTM, specific for intron 102 of COL7A1. Mutations in the COL7A1 gene underlie the dystrophic subtype of the skin blistering disease epidermolysis bullosa (DEB). We have shown that co-transfections of the RTM and a selected AS RNA, interfering with competitive splicing elements on a COL7A1-minigene (COL7A1-MG), lead to a significant increase of the RNA trans-splicing efficiency. Thereby, accurate trans-splicing between the RTM and the COL7A1-MG is represented by the restoration of full-length green fluorescent protein GFP on mRNA and protein level. This mechanism can be crucial for the improvement of an RTM-mediated correction, especially in cases where a high trans-splicing efficiency is required.
Collapse
Affiliation(s)
- Ulrich Koller
- Department of Dermatology and EB House Austria, Paracelsus Medical University, Salzburg 5020, Austria.
| | - Stefan Hainzl
- Department of Dermatology and EB House Austria, Paracelsus Medical University, Salzburg 5020, Austria.
| | - Thomas Kocher
- Department of Dermatology and EB House Austria, Paracelsus Medical University, Salzburg 5020, Austria.
| | - Clemens Hüttner
- Department of Dermatology and EB House Austria, Paracelsus Medical University, Salzburg 5020, Austria.
| | - Alfred Klausegger
- Department of Dermatology and EB House Austria, Paracelsus Medical University, Salzburg 5020, Austria.
| | - Christina Gruber
- Department of Dermatology and EB House Austria, Paracelsus Medical University, Salzburg 5020, Austria.
| | - Elisabeth Mayr
- Department of Dermatology and EB House Austria, Paracelsus Medical University, Salzburg 5020, Austria.
| | - Verena Wally
- Department of Dermatology and EB House Austria, Paracelsus Medical University, Salzburg 5020, Austria.
| | - Johann W Bauer
- Department of Dermatology and EB House Austria, Paracelsus Medical University, Salzburg 5020, Austria.
| | - Eva M Murauer
- Department of Dermatology and EB House Austria, Paracelsus Medical University, Salzburg 5020, Austria.
| |
Collapse
|
19
|
Chiaverini C, Charlesworth A, Fernandez A, Barbarot S, Bessis D, Bodemer C, Bursztejn AC, Cobo AM, Del Rio M, D'Incan M, Labrèze C, Langlet C, Mazereeuw J, Miquel J, Vabres P, Meneguzzi G, Lacour JP. Aplasia cutis congenita with dystrophic epidermolysis bullosa: clinical and mutational study. Br J Dermatol 2014; 170:901-6. [DOI: 10.1111/bjd.12741] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/14/2013] [Indexed: 12/11/2022]
Affiliation(s)
- C. Chiaverini
- French Reference Centre for Inherited Epidermolysis Bullosa; Archet Hospital; BP 3079 06202 NICE Cedex3 France
- INSERM U1081, CNRS UMR7284; Institute for Research on Cancer and Aging, Nice (IRCAN); University of Nice Sophia Antipolis, Faculty of Medicine; 28 Avenue Valombrose F-06107 Nice France
| | - A. Charlesworth
- French Reference Centre for Inherited Epidermolysis Bullosa; Archet Hospital; BP 3079 06202 NICE Cedex3 France
- INSERM U1081, CNRS UMR7284; Institute for Research on Cancer and Aging, Nice (IRCAN); University of Nice Sophia Antipolis, Faculty of Medicine; 28 Avenue Valombrose F-06107 Nice France
| | - A. Fernandez
- French Reference Centre for Inherited Epidermolysis Bullosa; Archet Hospital; BP 3079 06202 NICE Cedex3 France
| | - S. Barbarot
- Competence Centre for Rare Skin Diseases; Department of Dermatology; CHU Nantes; Nantes France
| | - D. Bessis
- Department of Dermatology; Saint Eloi Hospital; Montpellier France
| | - C. Bodemer
- French Reference Centre for Genetic Skin Disorders (MAGEC); CHU Necker for Sick Children; University-Paris Descartes; Institute Imagine; APHP; Paris France
| | | | - A.-M. Cobo
- Department of Genetics; Donostia Hospital; San Sebastian Spain
| | - M. Del Rio
- Regenerative Medicine Unit; Department of Bioengineering; CIEMAT and CIBER on Rare Diseases; Universidad Carlos III; Madrid Spain
| | - M. D'Incan
- Department of Dermatology; Estaing Hospital; Clermont-Ferrand France
| | - C. Labrèze
- National Centre for Rare Skin Disorders and Diseases; Pellegrin Enfants Hospital; Bordeaux France
| | - C. Langlet
- Department of Pediatrics 2; Hautepierre Hospital; Strasbourg France
| | - J. Mazereeuw
- Reference Centre for Rare Skin Diseases; Department of Dermatology; Larrey Hospital; Toulouse France
| | - J. Miquel
- Department of Dermatology; Pontchaillou Hospital; Rennes France
| | - P. Vabres
- Department of Dermatology; Bocage Hospital; Dijon France
| | - G. Meneguzzi
- INSERM U1081, CNRS UMR7284; Institute for Research on Cancer and Aging, Nice (IRCAN); University of Nice Sophia Antipolis, Faculty of Medicine; 28 Avenue Valombrose F-06107 Nice France
| | - J.-P. Lacour
- French Reference Centre for Inherited Epidermolysis Bullosa; Archet Hospital; BP 3079 06202 NICE Cedex3 France
- INSERM U1081, CNRS UMR7284; Institute for Research on Cancer and Aging, Nice (IRCAN); University of Nice Sophia Antipolis, Faculty of Medicine; 28 Avenue Valombrose F-06107 Nice France
| |
Collapse
|
20
|
Iqbal MA, Siddiqui FA, Chaman N, Gupta V, Kumar B, Gopinath P, Bamezai RNK. Missense mutations in pyruvate kinase M2 promote cancer metabolism, oxidative endurance, anchorage independence, and tumor growth in a dominant negative manner. J Biol Chem 2014; 289:8098-105. [PMID: 24492614 DOI: 10.1074/jbc.m113.515742] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The present study was designed to examine the functional relevance of two heterozygous mutations (H391Y and K422R), observed earlier by us in the Bloom syndrome condition. Cells stably expressing exogenous wild-type or mutant PKM2 (K422R or H391Y) or co-expressing both wild type and mutant (PKM2-K422R or PKM2-H391Y) were assessed for cancer metabolism and tumorigenic potential. Interestingly, cells co-expressing PKM2 and mutant (K422R or H391Y) showed significantly aggressive cancer metabolism as compared with cells expressing either wild-type or mutant PKM2 independently. A similar trend was observed for oxidative endurance, tumorigenic potential, cellular proliferation, and tumor growth. These observations signify the dominant negative nature of mutations. Remarkably, PKM2-H391Y co-expressed cells showed a maximal effect on all the studied parameters. Such a dominant negative impaired function of PKM2 in tumor development is not known; this study demonstrates for the first time the possible predisposition of Bloom syndrome patients with impaired PKM2 activity to cancer and the importance of studying genetic variations in PKM2 in the future to understand their relevance in cancer in general.
Collapse
Affiliation(s)
- Mohd Askandar Iqbal
- From the National Centre of Applied Human Genetics, School of Life Sciences, Jawaharlal Nehru University, New Mehrauli Road, New Delhi, 110067 India
| | | | | | | | | | | | | |
Collapse
|
21
|
Amato J, Stellato MI, Pizzo E, Petraccone L, Oliviero G, Borbone N, Piccialli G, Orecchia A, Bellei B, Castiglia D, Giancola C. PNA as a potential modulator of COL7A1 gene expression in dominant dystrophic epidermolysis bullosa: a physico-chemical study. MOLECULAR BIOSYSTEMS 2013; 9:3166-74. [PMID: 24121392 DOI: 10.1039/c3mb70283a] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Dominant diseases are single gene disorders occurring in the heterozygous state. The mutated allele exerts a dominant effect because it produces an abnormal polypeptide that interferes with the function of the normal allele product. Peptide Nucleic Acids (PNAs) offer a route for a potential therapy for dominant diseases by selectively silencing the allele carrying the dominant mutation. Here, we have synthesized and studied the properties of a 15-mer PNA fully complementary to the site of the c.5272-38T>A sequence variation, which identifies a recurrent mutant COL7A1 allele causing dominant dystrophic epidermolysis bullosa (DDEB), a mendelian disease characterized by skin blistering. The PNA was conjugated with four lysine residues at the C-terminus and a fluorescent probe at the N-terminus. Physico-chemical results proved the formation of a stable, selective PNA/mutant-DNA heteroduplex in vitro. Intriguingly, when transfected into normal human fibroblasts, the PNA correctly localized in the cell nucleus. Our results open new therapeutic possibilities for patients with DDEB.
Collapse
Affiliation(s)
- Jussara Amato
- Department of Pharmacy, University of Naples "Federico II", via D. Montesano 49, 80131 Naples, Italy.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Nyström A, Velati D, Mittapalli VR, Fritsch A, Kern JS, Bruckner-Tuderman L. Collagen VII plays a dual role in wound healing. J Clin Invest 2013; 123:3498-509. [PMID: 23867500 PMCID: PMC3726167 DOI: 10.1172/jci68127] [Citation(s) in RCA: 159] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2012] [Accepted: 05/09/2013] [Indexed: 01/01/2023] Open
Abstract
Although a host of intracellular signals is known to contribute to wound healing, the role of the cell microenvironment in tissue repair remains elusive. Here we employed 2 different mouse models of genetic skin fragility to assess the role of the basement membrane protein collagen VII (COL7A1) in wound healing. COL7A1 secures the attachment of the epidermis to the dermis, and its mutations cause a human skin fragility disorder coined recessive dystrophic epidermolysis bullosa (RDEB) that is associated with a constant wound burden. We show that COL7A1 is instrumental for skin wound closure by 2 interconnected mechanisms. First, COL7A1 was required for re-epithelialization through organization of laminin-332 at the dermal-epidermal junction. Its loss perturbs laminin-332 organization during wound healing, which in turn abrogates strictly polarized expression of integrin α6β4 in basal keratinocytes and negatively impacts the laminin-332/integrin α6β4 signaling axis guiding keratinocyte migration. Second, COL7A1 supported dermal fibroblast migration and regulates their cytokine production in the granulation tissue. These findings, which were validated in human wounds, identify COL7A1 as a critical player in physiological wound healing in humans and mice and may facilitate development of therapeutic strategies not only for RDEB, but also for other chronic wounds.
Collapse
Affiliation(s)
- Alexander Nyström
- Department of Dermatology, University Medical Center, Freiburg, Germany.
Freiburg Institute for Advanced Studies, School of Life Sciences — LifeNet, Freiburg, Germany
| | - Daniela Velati
- Department of Dermatology, University Medical Center, Freiburg, Germany.
Freiburg Institute for Advanced Studies, School of Life Sciences — LifeNet, Freiburg, Germany
| | - Venugopal R. Mittapalli
- Department of Dermatology, University Medical Center, Freiburg, Germany.
Freiburg Institute for Advanced Studies, School of Life Sciences — LifeNet, Freiburg, Germany
| | - Anja Fritsch
- Department of Dermatology, University Medical Center, Freiburg, Germany.
Freiburg Institute for Advanced Studies, School of Life Sciences — LifeNet, Freiburg, Germany
| | - Johannes S. Kern
- Department of Dermatology, University Medical Center, Freiburg, Germany.
Freiburg Institute for Advanced Studies, School of Life Sciences — LifeNet, Freiburg, Germany
| | - Leena Bruckner-Tuderman
- Department of Dermatology, University Medical Center, Freiburg, Germany.
Freiburg Institute for Advanced Studies, School of Life Sciences — LifeNet, Freiburg, Germany
| |
Collapse
|
23
|
Nyström A, Bruckner-Tuderman L, Kern JS. Cell- and protein-based therapy approaches for epidermolysis bullosa. Methods Mol Biol 2013; 961:425-40. [PMID: 23325662 DOI: 10.1007/978-1-62703-227-8_29] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/20/2023]
Abstract
Dystrophic epidermolysis bullosa (DEB) is a clinically heterogeneous heritable skin fragility disorder characterized by mechanically induced mucocutaneous blistering. On the molecular level DEB is caused by mutations leading to deficiency in collagen VII (CVII), a large extracellular protein building anchoring fibrils that attach the epidermis to the dermis. Severely affected patients suffer from wounds, which heal with excessive scarring causing mutilating deformities of hands and feet. The patients are also predisposed to development of aggressive squamous cell carcinomas at sites of chronic wounds. Currently no available therapies exist for this extremely disabling and stigmatizing disorder. We are developing and evaluating cell- and protein-based therapies for the management of DEB. Dermal fibroblasts are easy to propagate in vitro, they produce CVII, and they have immunomodulating capacities, which makes it possible to use allogeneic fibroblasts for therapy without risking major adverse effects from the host's immune system. Hence, fibroblasts, and fibroblast-like cells such as mesenchymal stromal cells, are prime candidates for cell-based DEB therapies. An alternative for management of disorders caused by defects in proteins with relatively low turnover rate is to introduce the protein de novo to the tissue by direct application of the protein. CVII is long-lived and expressed in moderate amounts in the skin; this makes injection of collagen VII protein a realistic approach for the treatment of DEB. Here we present methods and protocols that we are using for fibroblast- and recombinant CVII-based therapies of DEB in our model of this disease, the CVII hypomorphic mouse. These protocols are directed towards management of DEB but they can be easily adapted for the treatment of other skin fragility disorders.
Collapse
Affiliation(s)
- Alexander Nyström
- Department of Dermatology, University Freiburg Medical Center, Freiburg, Germany
| | | | | |
Collapse
|
24
|
Morgan CP, Allen DSI, Millington-Ward S, O'Dwyer GE, Palfi A, Jane Farrar G. A mutation-independent therapeutic strategy for dominant dystrophic epidermolysis bullosa. J Invest Dermatol 2013; 133:2793-2796. [PMID: 23743647 DOI: 10.1038/jid.2013.241] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- Clare P Morgan
- Department of Genetics, Trinity College Dublin, Dublin, Ireland.
| | - Danny S I Allen
- Department of Genetics, Trinity College Dublin, Dublin, Ireland
| | | | | | - Arpad Palfi
- Department of Genetics, Trinity College Dublin, Dublin, Ireland
| | - G Jane Farrar
- Department of Genetics, Trinity College Dublin, Dublin, Ireland
| |
Collapse
|
25
|
Nyström A, Buttgereit J, Bader M, Shmidt T, Özcelik C, Hausser I, Bruckner-Tuderman L, Kern JS. Rat model for dominant dystrophic epidermolysis bullosa: glycine substitution reduces collagen VII stability and shows gene-dosage effect. PLoS One 2013; 8:e64243. [PMID: 23717576 PMCID: PMC3662756 DOI: 10.1371/journal.pone.0064243] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2013] [Accepted: 04/10/2013] [Indexed: 02/07/2023] Open
Abstract
Dystrophic epidermolysis bullosa, a severely disabling hereditary skin fragility disorder, is caused by mutations in the gene coding for collagen VII, a specialized adhesion component of the dermal-epidermal junction zone. Both recessive and dominant forms are known; the latter account for about 40% of cases. Patients with dominant dystrophic epidermolysis bullosa exhibit a spectrum of symptoms ranging from mild localized to generalized skin manifestations. Individuals with the same mutation can display substantial phenotypic variance, emphasizing the role of modifying genes in this disorder. The etiology of dystrophic epidermolysis bullosa has been known for around two decades; however, important pathogenetic questions such as involvement of modifier genes remain unanswered and a causative therapy has yet to be developed. Much of the failure to make progress in these areas is due to the lack of suitable animal models that capture all aspects of this complex monogenetic disorder. Here, we report the first rat model of dominant dystrophic epidermolysis bullosa. Affected rats carry a spontaneous glycine to aspartic acid substitution, p.G1867D, within the main structural domain of collagen VII. This confers dominant-negative interference of protein folding and decreases the stability of mutant collagen VII molecules and their polymers, the anchoring fibrils. The phenotype comprises fragile and blister-prone skin, scarring and nail dystrophy. The model recapitulates all signs of the human disease with complete penetrance. Homozygous carriers of the mutation are more severely affected than heterozygous ones, demonstrating for the first time a gene-dosage effect of mutated alleles in dystrophic epidermolysis bullosa. This novel viable and workable animal model for dominant dystrophic epidermolysis bullosa will be valuable for addressing molecular disease mechanisms, effects of modifying genes, and development of novel molecular therapies for patients with dominantly transmitted skin disease.
Collapse
Affiliation(s)
- Alexander Nyström
- Department of Dermatology, University Freiburg Medical Center, Freiburg, Germany
| | - Jens Buttgereit
- Max Delbück Center for Molecular Medicine (MDC), Campus Berlin-Buch, Berlin, Germany
- Experimental and Clinical Research Center (ECRC), Berlin, Germany
| | - Michael Bader
- Max Delbück Center for Molecular Medicine (MDC), Campus Berlin-Buch, Berlin, Germany
| | - Tatiana Shmidt
- Max Delbück Center for Molecular Medicine (MDC), Campus Berlin-Buch, Berlin, Germany
- Experimental and Clinical Research Center (ECRC), Berlin, Germany
| | - Cemil Özcelik
- Max Delbück Center for Molecular Medicine (MDC), Campus Berlin-Buch, Berlin, Germany
- Experimental and Clinical Research Center (ECRC), Berlin, Germany
| | - Ingrid Hausser
- Department of Dermatology, University of Heidelberg, Heidelberg, Germany
| | - Leena Bruckner-Tuderman
- Department of Dermatology, University Freiburg Medical Center, Freiburg, Germany
- Freiburg Institute for Advanced Studies, School of Life Sciences, LifeNet, University of Freiburg, Freiburg, Germany
- * E-mail:
| | - Johannes S. Kern
- Department of Dermatology, University Freiburg Medical Center, Freiburg, Germany
| |
Collapse
|
26
|
Breitkreutz D, Koxholt I, Thiemann K, Nischt R. Skin basement membrane: the foundation of epidermal integrity--BM functions and diverse roles of bridging molecules nidogen and perlecan. BIOMED RESEARCH INTERNATIONAL 2013; 2013:179784. [PMID: 23586018 PMCID: PMC3618921 DOI: 10.1155/2013/179784] [Citation(s) in RCA: 124] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/10/2012] [Revised: 01/18/2013] [Accepted: 01/28/2013] [Indexed: 02/06/2023]
Abstract
The epidermis functions in skin as first defense line or barrier against environmental impacts, resting on extracellular matrix (ECM) of the dermis underneath. Both compartments are connected by the basement membrane (BM), composed of a set of distinct glycoproteins and proteoglycans. Herein we are reviewing molecular aspects of BM structure, composition, and function regarding not only (i) the dermoepidermal interface but also (ii) the resident microvasculature, primarily focusing on the per se nonscaffold forming components perlecan and nidogen-1 and nidogen-2. Depletion or functional deficiencies of any BM component are lethal at some stage of development or around birth, though BM defects vary between organs and tissues. Lethality problems were overcome by developmental stage- and skin-specific gene targeting or by cell grafting and organotypic (3D) cocultures of normal or defective cells, which allows recapitulating BM formation de novo. Thus, evidence is accumulating that BM assembly and turnover rely on mechanical properties and composition of the adjacent ECM and the dynamics of molecular assembly, including further "minor" local components, nidogens largely functioning as catalysts or molecular adaptors and perlecan as bridging stabilizer. Collectively, orchestration of BM assembly, remodeling, and the role of individual players herein are determined by the developmental, tissue-specific, or functional context.
Collapse
Affiliation(s)
- Dirk Breitkreutz
- Department of Dermatology, University of Cologne, Kerpener Strasse 62, 50937 Cologne, Germany.
| | | | | | | |
Collapse
|
27
|
siRNA-Mediated Allele-Specific Inhibition of Mutant Type VII Collagen in Dominant Dystrophic Epidermolysis Bullosa. J Invest Dermatol 2012; 132:1741-3. [DOI: 10.1038/jid.2012.11] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
28
|
Abstract
Spliceosome-mediated RNA trans-splicing (SMaRT) is an RNA-based technology to reprogram genes for diagnostic and therapeutic purposes. For the correction of genetic diseases, SMaRT offers several advantages over traditional gene-replacement strategies. SMaRT protocols have recently been used for in vitro phenotypic correction of a variety of genetic disorders, ranging from epidermolysis bullosa to neurodegenerative diseases. In vivo studies are currently bringing trans-splicing RNA therapy toward clinical application. In this review, we summarize the progress made toward the medical use of SMaRT and provide an outlook on its upcoming applications.
Collapse
|
29
|
Licarete E, Ganz S, Recknagel MJ, Di Zenzo G, Hashimoto T, Hertl M, Zambruno G, Hundorfean G, Mudter J, Neurath MF, Bruckner-Tuderman L, Sitaru C. Prevalence of collagen VII-specific autoantibodies in patients with autoimmune and inflammatory diseases. BMC Immunol 2012; 13:16. [PMID: 22471736 PMCID: PMC3368718 DOI: 10.1186/1471-2172-13-16] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2011] [Accepted: 04/04/2012] [Indexed: 11/20/2022] Open
Abstract
Background Autoimmunity to collagen VII is typically associated with the skin blistering disease epidermolysis bullosa acquisita (EBA), but also occurs occasionally in patients with systemic lupus erythematosus or inflammatory bowel disease. The aim of our present study was to develop an accurate immunoassay for assessing the presence of autoantibodies against collagen VII in large cohorts of patients and healthy donors. Methods Based on in silico antigenic analysis and previous wetlab epitope mapping data, we designed a chimeric collagen VII construct containing all collagen VII epitopes with higher antigenicity. ELISA was performed with sera from patients with EBA (n = 50), Crohn's disease (CD, n = 50), ulcerative colitis (UC, n = 50), bullous pemphigoid (BP, n = 76), and pemphigus vulgaris (PV, n = 42) and healthy donors (n = 245). Results By ELISA, the receiver operating characteristics analysis yielded an area under the curve of 0.98 (95% CI: 0.9638-1.005), allowing to set the cut-off at 0.32 OD at a calculated specificity of 98% and a sensitivity of 94%. Running the optimized test showed that serum IgG autoantibodies from 47 EBA (94%; 95% CI: 87.41%-100%), 2 CD (4%; 95% CI: 0%-9.43%), 8 UC (16%; 95% CI: 5.8%-26%), 2 BP (2.63%; 95% CI: 0%-6.23%), and 4 PV (9.52%; 95% CI: 0%-18.4%) patients as well as from 4 (1.63%; 95% CI: 0%-3.21%) healthy donors reacted with the chimeric protein. Further analysis revealed that in 34%, 37%, 16% and 100% of sera autoantibodies of IgG1, IgG2, IgG3, and IgG4 isotype, respectively, recognized the recombinant autoantigen. Conclusions Using a chimeric protein, we developed a new sensitive and specific ELISA to detect collagen specific antibodies. Our results show a low prevalence of collagen VII-specific autoantibodies in inflammatory bowel disease, pemphigus and bullous pemphigoid. Furthermore, we show that the autoimmune response against collagen VII is dominated by IgG4 autoantibodies. The new immunoassay should prove a useful tool for clinical and translational research and should improve the routine diagnosis and disease monitoring in diseases associated with collagen VII-specific autoimmunity.
Collapse
Affiliation(s)
- Emilia Licarete
- Department of Dermatology, University of Freiburg, Hauptstr, 7, Freiburg 79104, Germany
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Steplewski A, Kasinskas A, Fertala A. Remodeling of the dermal-epidermal junction in bilayered skin constructs after silencing the expression of the p.R2622Q and p.G2623C collagen VII mutants. Connect Tissue Res 2012; 53:379-89. [PMID: 22352907 PMCID: PMC4246506 DOI: 10.3109/03008207.2012.668252] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
The integrity of skin depends on a complex system of extracellular matrix molecules that form a biological scaffold. One of its elements is the dermal basement membrane that provides a link between the epidermis and the dermis. Mutations in collagen VII, a key component of the dermal membrane zone, are associated with dystrophic epidermolysis bullosa. Although it has been proposed that silencing the mutated COL7A1 allele is a promising approach to restore the dermal basement membrane zone formed in the presence of collagen VII mutants, limitations exist to testing this proposal. Here, we employed a model that utilized skin-like constructs in which engineered collagen VII mutant chains harboring the R2622Q or G2623C substitution were expressed conditionally, but the wild-type chains were expressed unconditionally. We demonstrated that switching off the production of the mutant collagen VII chains in skin constructs restores the organization of collagen VII and laminin 332 deposits in the dermal-epidermal junction to the level of control. We also demonstrated that remodeling of collagen IV deposits was not fully effective after silencing the expression of collagen VII mutants. Thus, our study suggests that while silencing mutant alleles of COL7A1 may repair critical elements of the affected dermal basement membrane, it may not be sufficient to fully remodel its entire architecture initially formed in the presence of the mutant collagen VII chains.
Collapse
Affiliation(s)
- Andrzej Steplewski
- Department of Dermatology and Cutaneous Biology, Jefferson Medical College, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Anthony Kasinskas
- Department of Dermatology and Cutaneous Biology, Jefferson Medical College, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Andrzej Fertala
- Department of Dermatology and Cutaneous Biology, Jefferson Medical College, Thomas Jefferson University, Philadelphia, Pennsylvania,Correspondence to: Andrzej Fertala, Department of Orthopaedic Surgery, Jefferson Medical College, Thomas Jefferson University, Curtis Building, Room 501, 1015 Walnut Street, Philadelphia, PA 19107., Tel: 215-503-0113,
| |
Collapse
|
31
|
A cellular model for the investigation of Fuchs' endothelial corneal dystrophy. Exp Eye Res 2011; 93:880-8. [PMID: 22020132 DOI: 10.1016/j.exer.2011.10.001] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2011] [Revised: 08/28/2011] [Accepted: 10/03/2011] [Indexed: 11/22/2022]
Abstract
Fuchs' endothelial corneal dystrophy is the most common corneal endotheliopathy, and a leading indication for corneal transplantation in the US. Relatively little is known about its underlying pathology. We created a cellular model of the disease focusing on collagen VIII alpha 2 (COL8A2), a collagen which is normally present in the cornea, but which is found in abnormal amounts and distribution in both early and late-onset forms of the disease. We performed cellular transfections using COL8A2 cDNAs including both wild-type and mutant alleles which are known to result in early-onset FECD. We used this cell model to explore the cellular production of wild-type and mutant monomeric and trimeric collagen VIII and measured production levels and patterns using Western blotting and immunofluorescence. We studied the thermal stability of the mutated collagen VIII helices using computer modeling, and further investigated these differences using collagen mimetic peptides. The Western blots demonstrated that similar amounts of wild-type and mutant collagen VIII monomers were produced in the cells. However, the levels of trimeric collagen peptide in the mutant-transfected cells were elevated. Intracellular accumulation of trimeric collagen VIII was confirmed on immunofluorescence studies. Both the computer model and the collagen mimetic peptides demonstrated that the L450W mutant was less thermally stable than either the Q455K or wild-type collagen VIII. Thus, although both mutant collagen VIII peptides were retained intracellularly, the biochemical reasons for the retention varied between genotypes. Collagen VIII mutations, which clinically result in Fuchs' dystrophy, are associated with abnormal cellular accumulation of collagen VIII. Different collagen VIII mutations may act via distinct biochemical mechanisms to produce the FECD phenotype.
Collapse
|
32
|
Atkinson SD, McGilligan VE, Liao H, Szeverenyi I, Smith FJ, Tara Moore C, Irwin McLean W. Development of Allele-Specific Therapeutic siRNA for Keratin 5 Mutations in Epidermolysis Bullosa Simplex. J Invest Dermatol 2011; 131:2079-86. [DOI: 10.1038/jid.2011.169] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
33
|
Leverkus M, Ambach A, Hoefeld-Fegeler M, Kohlhase J, Schmidt E, Schumann H, Has C, Gollnick H. Late-onset inversa recessive dystrophic epidermolysis bullosa caused by glycine substitutions in collagen type VII. Br J Dermatol 2011; 164:1104-6. [PMID: 21275939 DOI: 10.1111/j.1365-2133.2011.10230.x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Dystrophic epidermolysis bullosa (DEB) is a rare hereditary skin disorder caused by mutations in COL7A1, encoding collagen type VII.1 Clinical manifestations of COL7A1 mutations range from generalized skin blistering to mild localized blistering or nail dystrophy.2 The investigation of the molecular basis of DEB has revealed more than 540 different mutations that cannot entirely explain phenotypic variations (HGMD Professional 2010.3, https://portal.biobase-international. com/hgmd/). Inversa recessive DEB (RDEB-I) is a subtype characterized by generalized blistering in the neonatal period. The condition improves with age, and in adults blistering is restricted to intertriginous areas, and severe lesions of the oral and genital mucosa and nail changes occur in the majority of described patients.2 Recent data suggested that amino-acid substitutions affecting arginines or glycines at borders of collagenic subdomains might cause this phenotype.3 We report a German patient with an unusually mild RDEB-I harbouring compound heterozygous mutations in COL7A1.
Collapse
Affiliation(s)
- M Leverkus
- Department of Dermatology and Venereology, Otto-von-Guericke-University, Magdeburg, Germany.
| | | | | | | | | | | | | | | |
Collapse
|
34
|
Jensen DA, Steplewski A, Gawron K, Fertala A. Persistence of intracellular and extracellular changes after incompletely suppressing expression of the R789C (p.R989C) and R992C (p.R1192C) collagen II mutants. Hum Mutat 2011; 32:794-805. [PMID: 21472893 DOI: 10.1002/humu.21506] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2011] [Accepted: 03/23/2011] [Indexed: 11/06/2022]
Abstract
Mutations in COL2A1 produce a spectrum of disorders whose hallmark feature is alterations in skeletal development. Attempts to counteract the effects of collagen mutations at the molecular level have been relatively ineffective due to the inability to selectively suppress a mutant allele, and failure to deliver a sufficient number of cells expressing wild-type collagen. Moreover, these approaches are hampered because the minimal therapeutic conditions that would allow extracellular matrix remodeling and recovery of cells from stress are not known. Here, we employed a tetracycline-inducible system for expressing the R789C or R992C collagen II mutants, allowing us to decrease the production of mutant proteins by 25, 50, 75, or 100% with respect to their initial production. Through analysis of intracellular and extracellular parameters we have shown that affected cell/matrix systems are able to recover from mutation-induced aberrations only when 100% expression of mutant collagens is shut off, but not if the expression of small amounts of mutant molecules persists in the system. Our data suggest that efficient remodeling of tissues affected by the presence of thermolabile collagen mutants may depend on their complete elimination rather than on partial reduction.
Collapse
Affiliation(s)
- Deborah A Jensen
- Department of Dermatology and Cutaneous Biology, Jefferson Medical College, Thomas Jefferson University, 233 S. 10th Street, Philadelphia, PA 19107, USA
| | | | | | | |
Collapse
|
35
|
Chiaverini C, Charlesworth AV, Youssef M, Cuny JF, Rabia SH, Lacour JP, Meneguzzi G. Inversa Dystrophic Epidermolysis Bullosa Is Caused by Missense Mutations at Specific Positions of the Collagenic Domain of Collagen Type VII. J Invest Dermatol 2010; 130:2508-11. [DOI: 10.1038/jid.2010.159] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
36
|
Wally V, Brunner M, Lettner T, Wagner M, Koller U, Trost A, Murauer EM, Hainzl S, Hintner H, Bauer JW. K14 mRNA reprogramming for dominant epidermolysis bullosa simplex. Hum Mol Genet 2010; 19:4715-25. [DOI: 10.1093/hmg/ddq405] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
|
37
|
Abstract
PURPOSE OF REVIEW This review highlights key findings, both clinical and basic, that have been published in the field of inherited epidermolysis bullosa within the past few years. RECENT FINDINGS New epidermolysis bullosa phenotypes, genotypes and modes of transmission have been identified, resulting in a revised classification system. Detailed evidence-based data are now available on the risk of extracutaneous complications in each of the major epidermolysis bullosa subtypes. Studies are now underway to try to better explain the biological aggressiveness of squamous cell carcinomas arising in epidermolysis bullosa skin. Cell and animal models have been refined and used to ascertain the feasibility of gene replacement therapy, stem cell transplantation, and treatment with injected allogeneic fibroblasts or recombinant type VII collagen. As a result, clinical trials are now being pursued to test each of these in humans. SUMMARY Epidermolysis bullosa is caused by mutations in at least 14 genes, leading to a broad spectrum of entities, each of which has its own relative risk for the development of specific extracutaneous complications and/or premature death. Intensive research, both basic and clinical, is bringing us closer to more effective treatments and possibly even a cure.
Collapse
|