1
|
Yudaeva A, Kostyusheva A, Kachanov A, Brezgin S, Ponomareva N, Parodi A, Pokrovsky VS, Lukashev A, Chulanov V, Kostyushev D. Clinical and Translational Landscape of Viral Gene Therapies. Cells 2024; 13:1916. [PMID: 39594663 PMCID: PMC11592828 DOI: 10.3390/cells13221916] [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: 09/23/2024] [Revised: 10/29/2024] [Accepted: 11/14/2024] [Indexed: 11/28/2024] Open
Abstract
Gene therapies hold significant promise for treating previously incurable diseases. A number of gene therapies have already been approved for clinical use. Currently, gene therapies are mostly limited to the use of adeno-associated viruses and the herpes virus. Viral vectors, particularly those derived from human viruses, play a critical role in this therapeutic approach due to their ability to efficiently deliver genetic material to target cells. Despite their advantages, such as stable gene expression and efficient transduction, viral vectors face numerous limitations that hinder their broad application. These limitations include small cloning capacities, immune and inflammatory responses, and risks of insertional mutagenesis. This review explores the current landscape of viral vectors used in gene therapy, discussing the different types of DNA- and RNA-based viral vectors, their characteristics, limitations, and current medical and potential clinical applications. The review also highlights strategies to overcome existing challenges, including optimizing vector design, improving safety profiles, and enhancing transgene expression both using molecular techniques and nanotechnologies, as well as by approved drug formulations.
Collapse
Affiliation(s)
- Alexandra Yudaeva
- Laboratory of Genetic Technologies, Martsinovsky Institute of Medical Parasitology, Tropical and Vector-Borne Diseases, First Moscow State Medical University (Sechenov University), 119991 Moscow, Russia; (A.Y.); (A.K.); (A.K.); (S.B.); (N.P.); (A.L.)
| | - Anastasiya Kostyusheva
- Laboratory of Genetic Technologies, Martsinovsky Institute of Medical Parasitology, Tropical and Vector-Borne Diseases, First Moscow State Medical University (Sechenov University), 119991 Moscow, Russia; (A.Y.); (A.K.); (A.K.); (S.B.); (N.P.); (A.L.)
| | - Artyom Kachanov
- Laboratory of Genetic Technologies, Martsinovsky Institute of Medical Parasitology, Tropical and Vector-Borne Diseases, First Moscow State Medical University (Sechenov University), 119991 Moscow, Russia; (A.Y.); (A.K.); (A.K.); (S.B.); (N.P.); (A.L.)
| | - Sergey Brezgin
- Laboratory of Genetic Technologies, Martsinovsky Institute of Medical Parasitology, Tropical and Vector-Borne Diseases, First Moscow State Medical University (Sechenov University), 119991 Moscow, Russia; (A.Y.); (A.K.); (A.K.); (S.B.); (N.P.); (A.L.)
- Division of Biotechnology, Sirius University of Science and Technology, 354340 Sochi, Russia; (A.P.); (V.S.P.)
| | - Natalia Ponomareva
- Laboratory of Genetic Technologies, Martsinovsky Institute of Medical Parasitology, Tropical and Vector-Borne Diseases, First Moscow State Medical University (Sechenov University), 119991 Moscow, Russia; (A.Y.); (A.K.); (A.K.); (S.B.); (N.P.); (A.L.)
- Division of Biotechnology, Sirius University of Science and Technology, 354340 Sochi, Russia; (A.P.); (V.S.P.)
- Department of Pharmaceutical and Toxicological Chemistry, Sechenov First Moscow State Medical University, 119146 Moscow, Russia
| | - Alessandro Parodi
- Division of Biotechnology, Sirius University of Science and Technology, 354340 Sochi, Russia; (A.P.); (V.S.P.)
| | - Vadim S. Pokrovsky
- Division of Biotechnology, Sirius University of Science and Technology, 354340 Sochi, Russia; (A.P.); (V.S.P.)
- Blokhin National Medical Research Center of Oncology, 115478 Moscow, Russia
- Department of Biochemistry, People’s Friendship University, 117198 Moscow, Russia
| | - Alexander Lukashev
- Laboratory of Genetic Technologies, Martsinovsky Institute of Medical Parasitology, Tropical and Vector-Borne Diseases, First Moscow State Medical University (Sechenov University), 119991 Moscow, Russia; (A.Y.); (A.K.); (A.K.); (S.B.); (N.P.); (A.L.)
- Research Institute for Systems Biology and Medicine, 117246 Moscow, Russia
| | - Vladimir Chulanov
- Department of Infectious Diseases, First Moscow State Medical University (Sechenov University), 119991 Moscow, Russia;
| | - Dmitry Kostyushev
- Laboratory of Genetic Technologies, Martsinovsky Institute of Medical Parasitology, Tropical and Vector-Borne Diseases, First Moscow State Medical University (Sechenov University), 119991 Moscow, Russia; (A.Y.); (A.K.); (A.K.); (S.B.); (N.P.); (A.L.)
- Division of Biotechnology, Sirius University of Science and Technology, 354340 Sochi, Russia; (A.P.); (V.S.P.)
- Faculty of Bioengineering and Bioinformatics, Lomonosov Moscow State University, 119234 Moscow, Russia
| |
Collapse
|
2
|
Boulinguiez A, Dhiab J, Crisol B, Muraine L, Gaut L, Rouxel C, Flaire J, Mouigni H, Lemaitre M, Giroux B, Audoux L, SaintPierre B, Ferry A, Mouly V, Butler‐Browne G, Negroni E, Malerba A, Trollet C. Different outcomes of endurance and resistance exercise in skeletal muscles of Oculopharyngeal muscular dystrophy. J Cachexia Sarcopenia Muscle 2024; 15:1976-1988. [PMID: 39113268 PMCID: PMC11446690 DOI: 10.1002/jcsm.13546] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Revised: 05/07/2024] [Accepted: 06/25/2024] [Indexed: 10/04/2024] Open
Abstract
BACKGROUND Exercise is widely considered to have beneficial impact on skeletal muscle aging. In addition, there are also several studies demonstrating a positive effect of exercise on muscular dystrophies. Oculopharyngeal muscular dystrophy (OPMD) is a late-onset autosomal dominant inherited neuromuscular disorder caused by mutations in the PAPBN1 gene. These mutations consist in short (1-8) and meiotically stable GCN trinucleotide repeat expansions in its coding region responsible for the formation of PAPBN1 intranuclear aggregates. This study aims to characterize the effects of two types of chronic exercise, resistance and endurance, on the OPMD skeletal muscle phenotype using a relevant murine model of OPMD. METHODS In this study, we tested two protocols of exercise. In the first, based on endurance exercise, FvB (wild-type) and A17 (OPMD) mice underwent a 6-week-long motorized treadmill protocol consisting in three sessions per week of running 20 cm/s for 20 min. In the second protocol, based on resistance exercise generated by chronic mechanical overload (OVL), surgical removal of gastrocnemius and soleus muscles was performed, inducing hypertrophy of the plantaris muscle. In both types of exercise, muscles of A17 and FvB mice were compared with those of respective sedentary mice. For all the groups, force measurement, muscle histology, and molecular analyses were conducted. RESULTS Following the endurance exercise protocol, we did not observe any major changes in the muscle physiological parameters, but an increase in the number of PABPN1 intranuclear aggregates in both tibialis anterior (+24%, **P = 0.0026) and gastrocnemius (+18%, ****P < 0.0001) as well as enhanced collagen deposition (+20%, **P = 0.0064 in the tibialis anterior; +35%, **P = 0.0042 in the gastrocnemius) in the exercised A17 OPMD mice. In the supraphysiological resistance overload protocol, we also observed an increased collagen deposition (×2, ****P < 0.0001) in the plantaris muscle of A17 OPMD mice which was associated with larger muscle mass (×2, ****P < 0.0001) and fibre cross sectional area (×2, ***P = 0.0007) and increased absolute maximal force (×2, ****P < 0.0001) as well as a reduction in PABPN1 aggregate number (-16%, ****P < 0.0001). CONCLUSIONS Running exercise and mechanical overload led to very different outcome in skeletal muscles of A17 mice. Both types of exercise enhanced collagen deposition but while the running protocol increased aggregates, the OVL reduced them. More importantly OVL reversed muscle atrophy and maximal force in the A17 mice. Our study performed in a relevant model gives an indication of the effect of different types of exercise on OPMD muscle which should be further evaluated in humans for future recommendations as a part of the lifestyle of individuals with OPMD.
Collapse
Affiliation(s)
- Alexis Boulinguiez
- Department of Biological Sciences, School of Life Sciences and the EnvironmentRoyal Holloway University of LondonLondonUK
| | - Jamila Dhiab
- Centre de Recherche en MyologieSorbonne Université, INSERM, Institut de MyologieParisFrance
| | - Barbara Crisol
- Centre de Recherche en MyologieSorbonne Université, INSERM, Institut de MyologieParisFrance
| | - Laura Muraine
- Centre de Recherche en MyologieSorbonne Université, INSERM, Institut de MyologieParisFrance
| | - Ludovic Gaut
- Centre de Recherche en MyologieSorbonne Université, INSERM, Institut de MyologieParisFrance
| | - Corentin Rouxel
- Centre de Recherche en MyologieSorbonne Université, INSERM, Institut de MyologieParisFrance
| | - Justine Flaire
- Centre de Recherche en MyologieSorbonne Université, INSERM, Institut de MyologieParisFrance
| | - Hadidja‐Rose Mouigni
- Centre de Recherche en MyologieSorbonne Université, INSERM, Institut de MyologieParisFrance
| | - Mégane Lemaitre
- Sorbonne Université, INSERM, UMS28 – Phénotypage du petit animalParisFrance
| | - Benoit Giroux
- Sorbonne Université, INSERM, UMS28 – Phénotypage du petit animalParisFrance
| | - Lucie Audoux
- Université Paris Cité, CNRS, INSERM, Institut CochinParisFrance
| | | | - Arnaud Ferry
- Centre de Recherche en MyologieSorbonne Université, INSERM, Institut de MyologieParisFrance
| | - Vincent Mouly
- Centre de Recherche en MyologieSorbonne Université, INSERM, Institut de MyologieParisFrance
| | - Gillian Butler‐Browne
- Centre de Recherche en MyologieSorbonne Université, INSERM, Institut de MyologieParisFrance
| | - Elisa Negroni
- Centre de Recherche en MyologieSorbonne Université, INSERM, Institut de MyologieParisFrance
| | - Alberto Malerba
- Department of Biological Sciences, School of Life Sciences and the EnvironmentRoyal Holloway University of LondonLondonUK
| | - Capucine Trollet
- Centre de Recherche en MyologieSorbonne Université, INSERM, Institut de MyologieParisFrance
| |
Collapse
|
3
|
Findlay AR. Dominantly inherited muscle disorders: understanding their complexity and exploring therapeutic approaches. Dis Model Mech 2024; 17:dmm050720. [PMID: 39501809 PMCID: PMC11574355 DOI: 10.1242/dmm.050720] [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] [Indexed: 11/21/2024] Open
Abstract
Treatments for disabling and life-threatening hereditary muscle disorders are finally close to becoming a reality. Research has thus far focused primarily on recessive forms of muscle disease. The gene replacement strategies that are commonly employed for recessive, loss-of-function disorders are not readily translatable to most dominant myopathies owing to the presence of a normal chromosome in each nucleus, hindering the development of novel treatments for these dominant disorders. This is largely due to their complex, heterogeneous disease mechanisms that require unique therapeutic approaches. However, as viral and RNA interference-based therapies enter clinical use, key tools are now in place to develop treatments for dominantly inherited disorders of muscle. This article will review what is known about dominantly inherited disorders of muscle, specifically their genetic basis, how mutations lead to disease, and the pathomechanistic implications for therapeutic approaches.
Collapse
Affiliation(s)
- Andrew R Findlay
- Washington University Saint Louis, Neuromuscular Disease Center, 660 S. Euclid Ave., St Louis, MO 63110, USA
| |
Collapse
|
4
|
Cubero Z, Chiou CA, Mukharesh L, Rizzo JF. A 61-Year-Old Man With Blepharoptosis, Ophthalmoplegia, Dysphagia, and Trouble Focusing His Eyes. J Neuroophthalmol 2024; 44:e431-e432. [PMID: 37171902 DOI: 10.1097/wno.0000000000001882] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/14/2023]
Affiliation(s)
- Zoe Cubero
- Juan Bautista School of Medicine (ZC), Caguas, Puerto Rico; Neuro-Ophthalmology Service (CAC, LM, JFR), Massachusetts Eye and Ear, Harvard Medical School, Boston, Massachusetts; and Department of Neurology (LM), University of North Carolina, Chapel Hill, North Carolina
| | | | | | | |
Collapse
|
5
|
Mak G, Tarnopolsky M, Lu JQ. Secondary mitochondrial dysfunction across the spectrum of hereditary and acquired muscle disorders. Mitochondrion 2024; 78:101945. [PMID: 39134108 DOI: 10.1016/j.mito.2024.101945] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2024] [Revised: 07/15/2024] [Accepted: 08/08/2024] [Indexed: 08/23/2024]
Abstract
Mitochondria form a dynamic network within skeletal muscle. This network is not only responsible for producing adenosine triphosphate (ATP) through oxidative phosphorylation, but also responds through fission, fusion and mitophagy to various factors, such as increased energy demands, oxidative stress, inflammation, and calcium dysregulation. Mitochondrial dysfunction in skeletal muscle not only occurs in primary mitochondrial myopathies, but also other hereditary and acquired myopathies. As such, this review attempts to highlight the clinical and histopathologic aspects of mitochondrial dysfunction seen in hereditary and acquired myopathies, as well as discuss potential mechanisms leading to mitochondrial dysfunction and therapies to restore mitochondrial function.
Collapse
Affiliation(s)
- Gloria Mak
- University of Alberta, Department of Neurology, Edmonton, Alberta, Canada
| | - Mark Tarnopolsky
- McMaster University, Department of Medicine and Pediatrics, Hamilton, Ontario, Canada
| | - Jian-Qiang Lu
- McMaster University, Department of Pathology and Molecular Medicine, Hamilton, Ontario, Canada.
| |
Collapse
|
6
|
McCulloch MK, Mehryab F, Rashnonejad A. Navigating the Landscape of CMT1B: Understanding Genetic Pathways, Disease Models, and Potential Therapeutic Approaches. Int J Mol Sci 2024; 25:9227. [PMID: 39273178 PMCID: PMC11395143 DOI: 10.3390/ijms25179227] [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: 06/16/2024] [Revised: 08/13/2024] [Accepted: 08/19/2024] [Indexed: 09/15/2024] Open
Abstract
Charcot-Marie-Tooth type 1B (CMT1B) is a peripheral neuropathy caused by mutations in the gene encoding myelin protein zero (MPZ), a key component of the myelin sheath in Schwann cells. Mutations in the MPZ gene can lead to protein misfolding, unfolded protein response (UPR), endoplasmic reticulum (ER) stress, or protein mistrafficking. Despite significant progress in understanding the disease mechanisms, there is currently no effective treatment for CMT1B, with therapeutic strategies primarily focused on supportive care. Gene therapy represents a promising therapeutic approach for treating CMT1B. To develop a treatment and better design preclinical studies, an in-depth understanding of the pathophysiological mechanisms and animal models is essential. In this review, we present a comprehensive overview of the disease mechanisms, preclinical models, and recent advancements in therapeutic research for CMT1B, while also addressing the existing challenges in the field. This review aims to deepen the understanding of CMT1B and to encourage further research towards the development of effective treatments for CMT1B patients.
Collapse
Affiliation(s)
- Mary Kate McCulloch
- Center for Gene Therapy, The Abigail Wexner Research Institute at Nationwide Children’s Hospital, 575 Children’s Crossroad, Columbus, OH 43215, USA
- Molecular, Cellular, and Developmental Biology Program, The Ohio State University, Columbus, OH 43210, USA
| | - Fatemeh Mehryab
- Center for Gene Therapy, The Abigail Wexner Research Institute at Nationwide Children’s Hospital, 575 Children’s Crossroad, Columbus, OH 43215, USA
| | - Afrooz Rashnonejad
- Center for Gene Therapy, The Abigail Wexner Research Institute at Nationwide Children’s Hospital, 575 Children’s Crossroad, Columbus, OH 43215, USA
- Molecular, Cellular, and Developmental Biology Program, The Ohio State University, Columbus, OH 43210, USA
| |
Collapse
|
7
|
Shademan M, Mei H, van Engelen B, Ariyurek Y, Kloet S, Raz V. PABPN1 loss-of-function causes APA-shift in oculopharyngeal muscular dystrophy. HGG ADVANCES 2024; 5:100269. [PMID: 38213032 PMCID: PMC10840355 DOI: 10.1016/j.xhgg.2024.100269] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Revised: 01/05/2024] [Accepted: 01/05/2024] [Indexed: 01/13/2024] Open
Abstract
Alternative polyadenylation (APA) at the 3' UTR of transcripts contributes to the cell transcriptome. APA is suppressed by the nuclear RNA-binding protein PABPN1. Aging-associated reduced PABPN1 levels in skeletal muscles lead to muscle wasting. Muscle weakness in oculopharyngeal muscular dystrophy (OPMD) is caused by short alanine expansion in PABPN1 exon1. The expanded PABPN1 forms nuclear aggregates, an OPMD hallmark. Whether the expanded PABPN1 affects APA and how it contributes to muscle pathology is unresolved. To investigate these questions, we developed a procedure including RNA library preparation and a simple pipeline calculating the APA-shift ratio as a readout for PABPN1 activity. Comparing APA-shift results to previously published PAS utilization and APA-shift results, we validated this procedure. The procedure was then applied on the OPMD cell model and on RNA from OPMD muscles. APA-shift was genome-wide in the mouse OPMD model, primarily affecting muscle transcripts. In OPMD individuals, APA-shift was enriched with muscle transcripts. In an OPMD cell model APA-shift was not significant. APA-shift correlated with reduced expression levels of a subset of PABPN1 isoforms, whereas the expression of the expanded PABPN1 did not correlate with APA-shift. PABPN1 activity is not affected by the expression of expanded PABPN1, but rather by reduced PABPN1 expression levels. In muscles, PABPN1 activity initially affects muscle transcripts. We suggest that muscle weakness in OPMD is caused by PABPN1 loss-of-function leading to APA-shift that primarily affects in muscle transcripts.
Collapse
Affiliation(s)
- Milad Shademan
- Department of Human Genetics, Leiden University Medical Centre, Leiden, the Netherlands
| | - Hailiang Mei
- Department of Biomedical Data Sciences, Leiden University Medical Centre, Leiden, the Netherlands
| | - Baziel van Engelen
- Department of Neurology, Radboud University Medical Centre, Nijmegen, the Netherlands
| | - Yavuz Ariyurek
- Department of Human Genetics, Leiden University Medical Centre, Leiden, the Netherlands
| | - Susan Kloet
- Department of Human Genetics, Leiden University Medical Centre, Leiden, the Netherlands
| | - Vered Raz
- Department of Human Genetics, Leiden University Medical Centre, Leiden, the Netherlands.
| |
Collapse
|
8
|
Smith IC, Chakraborty S, Bourque PR, Sampaio ML, Melkus G, Lochmüller H, Woulfe J, Parks RJ, Brais B, Warman-Chardon J. Emerging and established biomarkers of oculopharyngeal muscular dystrophy. Neuromuscul Disord 2023; 33:824-834. [PMID: 37926637 DOI: 10.1016/j.nmd.2023.09.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Revised: 09/25/2023] [Accepted: 09/27/2023] [Indexed: 11/07/2023]
Abstract
Oculopharyngeal muscular dystrophy (OPMD) is a rare, primarily autosomal dominant, late onset muscular dystrophy commonly presenting with ptosis, dysphagia, and subsequent weakness of proximal muscles. Although OPMD diagnosis can be confirmed with high confidence by genetic testing, the slow progression of OPMD poses a significant challenge to clinical monitoring and a barrier to assessing the efficacy of treatments during clinical trials. Accordingly, there is a pressing need for more sensitive measures of OPMD progression, particularly those which do not require a muscle biopsy. This review provides an overview of progress in OPMD biomarkers from clinical assessment, quantitative imaging, histological assessments, and genomics, as well as hypothesis-generating "omics" approaches. The ongoing search for biomarkers relevant to OPMD progression needs an integrative, longitudinal approach combining validated and experimental approaches which may include clinical, imaging, demographic, and biochemical assessment methods. A multi-omics approach to biochemical biomarker discovery could help provide context for differences found between individuals with varying levels of disease activity and provide insight into pathomechanisms and prognosis of OPMD.
Collapse
Affiliation(s)
- Ian C Smith
- The Ottawa Hospital Research Institute, Ottawa, ON K1Y 4E9, Canada
| | | | - Pierre R Bourque
- Faculty of Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada; Department of Medicine, The Ottawa Hospital/University of Ottawa, Ottawa, ON K1H 8L6, Canada; Eric Poulin Centre for Neuromuscular Disease, University of Ottawa, Ottawa, ON K1Y 4E9, Canada
| | - Marcos L Sampaio
- Faculty of Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada; Eric Poulin Centre for Neuromuscular Disease, University of Ottawa, Ottawa, ON K1Y 4E9, Canada; Department of Medical Imaging, The Ottawa Hospital, Ottawa, Ontario K1Y 4E9, Canada; Department of Radiology, Radiation Oncology and Medical Physics, University of Ottawa, Ottawa, ON K1H 8L6, Canada
| | - Gerd Melkus
- Faculty of Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada; Eric Poulin Centre for Neuromuscular Disease, University of Ottawa, Ottawa, ON K1Y 4E9, Canada; Department of Medical Imaging, The Ottawa Hospital, Ottawa, Ontario K1Y 4E9, Canada; Department of Physics, Carleton University, Ottawa, ON K1S 5B6, Canada
| | - Hanns Lochmüller
- The Ottawa Hospital Research Institute, Ottawa, ON K1Y 4E9, Canada; Faculty of Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada; Department of Medicine, The Ottawa Hospital/University of Ottawa, Ottawa, ON K1H 8L6, Canada; Eric Poulin Centre for Neuromuscular Disease, University of Ottawa, Ottawa, ON K1Y 4E9, Canada; Genetics, Children's Hospital of Eastern Ontario, Ottawa, ON K1H 8L1, Canada
| | - John Woulfe
- The Ottawa Hospital Research Institute, Ottawa, ON K1Y 4E9, Canada; Eric Poulin Centre for Neuromuscular Disease, University of Ottawa, Ottawa, ON K1Y 4E9, Canada; Department of Pathology and Laboratory Medicine, The Ottawa Hospital, Ottawa, ON K1Y 4E9, Canada
| | - Robin J Parks
- The Ottawa Hospital Research Institute, Ottawa, ON K1Y 4E9, Canada; Department of Medicine, The Ottawa Hospital/University of Ottawa, Ottawa, ON K1H 8L6, Canada; Eric Poulin Centre for Neuromuscular Disease, University of Ottawa, Ottawa, ON K1Y 4E9, Canada
| | - Bernard Brais
- Montreal Neurological Institute and Hospital, McGill University, Montreal, Quebec H3A 2B4, Canada
| | - Jodi Warman-Chardon
- The Ottawa Hospital Research Institute, Ottawa, ON K1Y 4E9, Canada; Faculty of Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada; Department of Medicine, The Ottawa Hospital/University of Ottawa, Ottawa, ON K1H 8L6, Canada; Eric Poulin Centre for Neuromuscular Disease, University of Ottawa, Ottawa, ON K1Y 4E9, Canada; Genetics, Children's Hospital of Eastern Ontario, Ottawa, ON K1H 8L1, Canada; Montreal Neurological Institute and Hospital, McGill University, Montreal, Quebec H3A 2B4, Canada.
| |
Collapse
|
9
|
Maze D, Girardin C, Benz N, Montier T, Pichon C, Midoux P. CFTR and dystrophin encoding plasmids carrying both luciferase reporter gene, nuclear import specific sequences and triple helix sites. Plasmid 2023; 127:102686. [PMID: 37207938 DOI: 10.1016/j.plasmid.2023.102686] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 04/26/2023] [Accepted: 05/16/2023] [Indexed: 05/21/2023]
Abstract
Duchenne Muscular Dystrophy and Cystic Fibrosis are two major monogenetic diseases which could be treated by non-viral gene therapy. For this purpose, plasmid DNA (pDNA) coding for the functional genes requires its equipment with signal molecules favouring its intracellular trafficking and delivery in the nucleus of the target cells. Here, two novel constructions of large pDNAs encoding the Cystic Fibrosis Transmembrane Conductance Regulator (CFTR) and full-length dystrophin (DYS) genes are reported. The expression of CFTR and DYS genes are driven respectively by the hCEF1 airway epithelial cells and spc5-12 muscle cells specific promoter. Those pDNAs encode also the luciferase reporter gene driven by the CMV promoter to evaluate gene delivery in animals by bioluminescence. In addition, oligopurine • oligopyrimidine sequences are inserted to enable equipment of pDNAs with peptides conjugated with a triple helix forming oligonucleotide (TFO). Furthermore, specific κB sequences are also inserted to promote their NFκB-mediated nuclear import. pDNA constructions are reported; transfection efficiency, tissue specific expression of CFTR and dystrophin in target cells, and triple helix formation are demonstrated. These plasmids are tools of interest to develop non-viral gene therapy of Cystic Fibrosis and Duchenne Muscular Dystrophy.
Collapse
Affiliation(s)
- Delphine Maze
- Centre de Biophysique Moléculaire, CNRS UPR4301, Inserm and University of Orléans, 45071 Orléans cedex 02, France
| | - Caroline Girardin
- Centre de Biophysique Moléculaire, CNRS UPR4301, Inserm and University of Orléans, 45071 Orléans cedex 02, France
| | - Nathalie Benz
- Univ Brest, INSERM, EFS, UMR 1078, GGB - GTCA Team, Brest F-29200, France
| | - Tristan Montier
- Univ Brest, INSERM, EFS, UMR 1078, GGB - GTCA Team, Brest F-29200, France; Service de Génétique Médicale et Biologie de la Reproduction, Centre de référence des maladies rares 'Maladies neuromusculaires', CHRU de Brest, Brest F-29200, France
| | - Chantal Pichon
- Centre de Biophysique Moléculaire, CNRS UPR4301, Inserm and University of Orléans, 45071 Orléans cedex 02, France
| | - Patrick Midoux
- Centre de Biophysique Moléculaire, CNRS UPR4301, Inserm and University of Orléans, 45071 Orléans cedex 02, France.
| |
Collapse
|
10
|
Harish P, Malerba A, Kroon RHMJM, Shademan M, van Engelan B, Raz V, Popplewell L, Snowden SG. Novel Metabolomic Approach for Identifying Pathology-Specific Biomarkers in Rare Diseases: A Case Study in Oculopharyngeal Muscular Dystrophy (OPMD). Metabolites 2023; 13:769. [PMID: 37367926 DOI: 10.3390/metabo13060769] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Revised: 06/02/2023] [Accepted: 06/15/2023] [Indexed: 06/28/2023] Open
Abstract
The identification of metabolomic biomarkers relies on the analysis of large cohorts of patients compared to healthy controls followed by the validation of markers in an independent sample set. Indeed, circulating biomarkers should be causally linked to pathology to ensure that changes in the marker precede changes in the disease. However, this approach becomes unfeasible in rare diseases due to the paucity of samples, necessitating the development of new methods for biomarker identification. The present study describes a novel approach that combines samples from both mouse models and human patients to identify biomarkers of OPMD. We initially identified a pathology-specific metabolic fingerprint in murine dystrophic muscle. This metabolic fingerprint was then translated into (paired) murine serum samples and then to human plasma samples. This study identified a panel of nine candidate biomarkers that could predict muscle pathology with a sensitivity of 74.3% and specificity of 100% in a random forest model. These findings demonstrate that the proposed approach can identify biomarkers with good predictive performance and a higher degree of confidence in their relevance to pathology than markers identified in a small cohort of human samples alone. Therefore, this approach has a high potential utility for identifying circulating biomarkers in rare diseases.
Collapse
Affiliation(s)
- Pradeep Harish
- Department of Pharmacology and Therapeutics, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool L69 3GE, UK
| | - Alberto Malerba
- Department of Biological Sciences, Royal Holloway University of London, Egham TW20 0EX, Surrey, UK
| | - Rosemarie H M J M Kroon
- Department of Rehabilitation, Donder Institute for Brain, Cognition and Behaviour, Radboud University Medical Centre, 6525 AJ Nijmegen, The Netherlands
| | - Milad Shademan
- Department of Human Genetics, Leiden University Medical Centre, 2333 ZC Leiden, The Netherlands
| | - Baziel van Engelan
- Department of Rehabilitation, Donder Institute for Brain, Cognition and Behaviour, Radboud University Medical Centre, 6525 AJ Nijmegen, The Netherlands
| | - Vered Raz
- Department of Human Genetics, Leiden University Medical Centre, 2333 ZC Leiden, The Netherlands
| | - Linda Popplewell
- Department of Biological Sciences, Royal Holloway University of London, Egham TW20 0EX, Surrey, UK
- National Horizons Centre, Teesside University, Darlington DL1 1HG, County Durham, UK
| | - Stuart G Snowden
- Department of Biological Sciences, Royal Holloway University of London, Egham TW20 0EX, Surrey, UK
| |
Collapse
|
11
|
Viral Vectors in Gene Therapy: Where Do We Stand in 2023? Viruses 2023; 15:v15030698. [PMID: 36992407 PMCID: PMC10059137 DOI: 10.3390/v15030698] [Citation(s) in RCA: 52] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Revised: 02/23/2023] [Accepted: 03/02/2023] [Indexed: 03/11/2023] Open
Abstract
Viral vectors have been used for a broad spectrum of gene therapy for both acute and chronic diseases. In the context of cancer gene therapy, viral vectors expressing anti-tumor, toxic, suicide and immunostimulatory genes, such as cytokines and chemokines, have been applied. Oncolytic viruses, which specifically replicate in and kill tumor cells, have provided tumor eradication, and even cure of cancers in animal models. In a broader meaning, vaccine development against infectious diseases and various cancers has been considered as a type of gene therapy. Especially in the case of COVID-19 vaccines, adenovirus-based vaccines such as ChAdOx1 nCoV-19 and Ad26.COV2.S have demonstrated excellent safety and vaccine efficacy in clinical trials, leading to Emergency Use Authorization in many countries. Viral vectors have shown great promise in the treatment of chronic diseases such as severe combined immunodeficiency (SCID), muscular dystrophy, hemophilia, β-thalassemia, and sickle cell disease (SCD). Proof-of-concept has been established in preclinical studies in various animal models. Clinical gene therapy trials have confirmed good safety, tolerability, and therapeutic efficacy. Viral-based drugs have been approved for cancer, hematological, metabolic, neurological, and ophthalmological diseases as well as for vaccines. For example, the adenovirus-based drug Gendicine® for non-small-cell lung cancer, the reovirus-based drug Reolysin® for ovarian cancer, the oncolytic HSV T-VEC for melanoma, lentivirus-based treatment of ADA-SCID disease, and the rhabdovirus-based vaccine Ervebo against Ebola virus disease have been approved for human use.
Collapse
|
12
|
Wang Y, Huang L, Zhu Y, An X, Li J, Zhen J, Yu J. De novo variations of ANK1 gene caused hereditary spherocytosis in two Chinese children by affecting pre-mRNA splicing. BMC Pediatr 2023; 23:23. [PMID: 36647015 PMCID: PMC9841706 DOI: 10.1186/s12887-022-03795-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Accepted: 12/09/2022] [Indexed: 01/18/2023] Open
Abstract
BACKGROUND AND AIMS Hereditary spherocytosis (HS) is one of the most common hereditary haemolytic disorders. Here, two unrelated families with the probands displaying typical manifestations of HS were enrolled. Our study aimed to characterize the effect of two novel variants in HS patients on gene splicing to help minimize the rate of misdiagnosis of HS and enhance clinicians' understanding of the disease. PARTICIPANTS AND METHODS A retrospective review was conducted. Peripheral blood samples were collected from all the family members, and genomic DNA was extracted for genetic diagnostics. First, high-throughput sequencing technology was used for the preliminary screening of candidate causative variants. Thereafter, the variants were verified via Sanger sequencing. Furthermore, a pathogenicity analysis of the detected variants was performed including in silico prediction and in vitro experiments. We constructed matched wild-type and mutant-type minigene plasmid of ANK1 based on HEK293T cells to address the effects of variants on mRNA splicing. RESULTS The c.1305 + 2 T > A (family1) and c.1305 + 2del (family2) variants were detected in the ANK1 gene. These two de novo mutations described by us which have not been reported prior to this study. Moreover, the validation results of splicing reporter systems revealed that the intronic mutations resulted in abnormal pre-mRNA splicing. Specifically, the minigene plasmid expressing the c.1305 + 2 T > A variant transcribed the two aberrant transcripts: r.1305_1306ins1305 + 1_1305 + 229 and r.1305_1306ins1305 + 1_1305 + 552. The minigene plasmid expressing c.1305 + 2del transcribed the two aberrant transcripts: r.1305_1306ins1305 + 1_1305 + 228 and r.1305_1306ins1305 + 1_1305 + 551. CONCLUSION The two de novo variants identified in the ANK1 gene were the genetic etiology of the probands with HS in our study. Our findings further enrich the HS genotype database and provide a basis for genetic counselling and molecular diagnosis.
Collapse
Affiliation(s)
- Yang Wang
- grid.488412.3Department of Hematology and Oncology, Children’s Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Children’s Hospital of Chongqing Medical University, 136 Zhong shan er lu, Yu zhong district, Chongqing, 400014 China ,grid.488412.3Chongqing Key Laboratory of Pediatrics, Chongqing, China
| | - Lan Huang
- grid.488412.3Department of Hematology and Oncology, Children’s Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Children’s Hospital of Chongqing Medical University, 136 Zhong shan er lu, Yu zhong district, Chongqing, 400014 China ,grid.488412.3Chongqing Key Laboratory of Pediatrics, Chongqing, China
| | - Yao Zhu
- grid.488412.3Department of Hematology and Oncology, Children’s Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Children’s Hospital of Chongqing Medical University, 136 Zhong shan er lu, Yu zhong district, Chongqing, 400014 China ,grid.488412.3Chongqing Key Laboratory of Pediatrics, Chongqing, China
| | - Xizhou An
- grid.488412.3Department of Hematology and Oncology, Children’s Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Children’s Hospital of Chongqing Medical University, 136 Zhong shan er lu, Yu zhong district, Chongqing, 400014 China
| | - Jiacheng Li
- grid.488412.3Department of Hematology and Oncology, Children’s Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Children’s Hospital of Chongqing Medical University, 136 Zhong shan er lu, Yu zhong district, Chongqing, 400014 China ,grid.488412.3Chongqing Key Laboratory of Pediatrics, Chongqing, China
| | - Jiangwei Zhen
- grid.440186.fDepartment of Endocrinology, Shenzhen Samii International Medical Center, Shenzhen, 518000 China
| | - Jie Yu
- Department of Hematology and Oncology, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Children's Hospital of Chongqing Medical University, 136 Zhong shan er lu, Yu zhong district, Chongqing, 400014, China. .,Chongqing Key Laboratory of Pediatrics, Chongqing, China.
| |
Collapse
|
13
|
Lundstrom K. Gene Therapy Cargoes Based on Viral Vector Delivery. Curr Gene Ther 2023; 23:111-134. [PMID: 36154608 DOI: 10.2174/1566523222666220921112753] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2022] [Revised: 07/13/2022] [Accepted: 08/05/2022] [Indexed: 11/22/2022]
Abstract
Viral vectors have been proven useful in a broad spectrum of gene therapy applications due to their possibility to accommodate foreign genetic material for both local and systemic delivery. The wide range of viral vectors has enabled gene therapy applications for both acute and chronic diseases. Cancer gene therapy has been addressed by the delivery of viral vectors expressing anti-tumor, toxic, and suicide genes for the destruction of tumors. Delivery of immunostimulatory genes such as cytokines and chemokines has also been applied for cancer therapy. Moreover, oncolytic viruses specifically replicating in and killing tumor cells have been used as such for tumor eradication or in combination with tumor killing or immunostimulatory genes. In a broad meaning, vaccines against infectious diseases and various cancers can be considered gene therapy, which has been highly successful, not the least for the development of effective COVID-19 vaccines. Viral vector-based gene therapy has also demonstrated encouraging and promising results for chronic diseases such as severe combined immunodeficiency (SCID), muscular dystrophy, and hemophilia. Preclinical gene therapy studies in animal models have demonstrated proof-of-concept for a wide range of disease indications. Clinical evaluation of drugs and vaccines in humans has showed high safety levels, good tolerance, and therapeutic efficacy. Several gene therapy drugs such as the adenovirus-based drug Gendicine® for non-small-cell lung cancer, the reovirus-based drug Reolysin® for ovarian cancer, lentivirus-based treatment of SCID-X1 disease, and the rhabdovirus-based vaccine Ervebo against Ebola virus disease, and adenovirus-based vaccines against COVID-19 have been developed.
Collapse
|
14
|
Malerba A, Harish P, Popplewell L. Systemic Delivery of a Monoclonal Antibody to Immunologically Block Myostatin in the A17 Mouse Model of OPMD. Methods Mol Biol 2023; 2587:557-568. [PMID: 36401050 DOI: 10.1007/978-1-0716-2772-3_30] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
Oculopharyngeal muscular dystrophy (OPMD) is a late-onset rare muscle disease affecting approximately 1 in 80,000 individuals worldwide. However, it can affect as much as 1:600 individuals in some populations due to a strong founder effect. The muscle pathology is characterized by progressive eyelid drooping (ptosis), swallowing difficulties (dysphagia), and limb weakness at later stages of disease progression. The genetic defect is associated with significant fibrotic deposition and atrophy in affected muscles. No treatments are available to cure the disease. Only surgical techniques to correct ptosis and swallowing are currently possible, though they carry a risk of recurrence. Myostatin is a negative regulator of muscle growth, and several strategies to downregulate its expression have been developed with the aim of improving muscle mass and strength in muscular pathologies. We recently showed that weekly systemic treatment of the A17 murine model of OPMD with a monoclonal antibody for myostatin improves body and muscle mass, increases muscle strength, and reduces muscle fibrosis. Here, we describe the methodology for repeated intraperitoneal delivery of myostatin antibody in the murine model. Furthermore, we detail the most relevant analyses to assess histopathological and functional improvements of this treatment in this mouse model.
Collapse
Affiliation(s)
- Alberto Malerba
- Department of Biological Sciences, Royal Holloway University of London, Egham, Surrey, UK
| | - Pradeep Harish
- College of Health, Medicine and Life Science, Biosciences, Brunel University London, Uxbridge, UK
| | - Linda Popplewell
- Department of Biological Sciences, Royal Holloway University of London, Egham, Surrey, UK.
- National Horizons Centre, Teesside University, Darlington, UK.
| |
Collapse
|
15
|
Assessment of PABPN1 nuclear inclusions on a large cohort of patients and in a human xenograft model of oculopharyngeal muscular dystrophy. Acta Neuropathol 2022; 144:1157-1170. [PMID: 36197469 PMCID: PMC9637588 DOI: 10.1007/s00401-022-02503-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Revised: 08/26/2022] [Accepted: 09/17/2022] [Indexed: 01/26/2023]
Abstract
Oculopharyngeal muscular dystrophy (OPMD) is a rare muscle disease characterized by an onset of weakness in the pharyngeal and eyelid muscles. The disease is caused by the extension of a polyalanine tract in the Poly(A) Binding Protein Nuclear 1 (PABPN1) protein leading to the formation of intranuclear inclusions or aggregates in the muscle of OPMD patients. Despite numerous studies stressing the deleterious role of nuclear inclusions in cellular and animal OPMD models, their exact contribution to human disease is still unclear. In this study, we used a large and unique collection of human muscle biopsy samples to perform an in-depth analysis of PABPN1 aggregates in relation to age, genotype and muscle status with the final aim to improve our understanding of OPMD physiopathology. Here we demonstrate that age and genotype influence PABPN1 aggregates: the percentage of myonuclei containing PABPN1 aggregates increases with age and the chaperone HSP70 co-localize more frequently with PABPN1 aggregates with a larger polyalanine tract. In addition to the previously described PRMT1 and HSP70 co-factors, we identified new components of PABPN1 aggregates including GRP78/BiP, RPL24 and p62. We also observed that myonuclei containing aggregates are larger than myonuclei without. When comparing two muscles from the same patient, a similar amount of aggregates is observed in different muscles, except for the pharyngeal muscle where fewer aggregates are observed. This could be due to the peculiar nature of this muscle which has a low level of PAPBN1 and contains regenerating fibers. To confirm the fate of PABPN1 aggregates in a regenerating muscle, we generated a xenograft model by transplanting human OPMD muscle biopsy samples into the hindlimb of an immunodeficient mouse. Xenografts from subjects with OPMD displayed regeneration of human myofibers and PABPN1 aggregates were rapidly present-although to a lower extent-after muscle fiber regeneration. Our data obtained on human OPMD samples add support to the dual non-exclusive models in OPMD combining toxic PABPN1 intranuclear inclusions together with PABPN1 loss of function which altogether result in this late-onset and muscle selective disease.
Collapse
|
16
|
Boulinguiez A, Roth F, Mouigni HR, Butler-Browne G, Mouly V, Trollet C. [Nuclear aggregates in oculopharyngeal muscular dystrophy]. Med Sci (Paris) 2022; 38 Hors série n° 1:13-16. [PMID: 36649629 DOI: 10.1051/medsci/2022175] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Oculopharyngeal muscular dystrophy (OPMD) is one of the diseases related to pathological expansions of trinucleotides. Its pathogenesis remains unclear although the presence of aggregates within the nuclei of the muscle fiber seems to play an important role. The basic research studies presented here help understand their composition and their deleterious role. These elements may result in new therapeutic avenues.
Collapse
Affiliation(s)
- Alexis Boulinguiez
- Sorbonne Université-Inserm, Centre de Recherche en Myologie, Institut de Myologie, Paris, France
| | - Fany Roth
- Sorbonne Université-Inserm, Centre de Recherche en Myologie, Institut de Myologie, Paris, France
| | - Hadidja Rose Mouigni
- Sorbonne Université-Inserm, Centre de Recherche en Myologie, Institut de Myologie, Paris, France
| | - Gillian Butler-Browne
- Sorbonne Université-Inserm, Centre de Recherche en Myologie, Institut de Myologie, Paris, France
| | - Vincent Mouly
- Sorbonne Université-Inserm, Centre de Recherche en Myologie, Institut de Myologie, Paris, France
| | - Capucine Trollet
- Sorbonne Université-Inserm, Centre de Recherche en Myologie, Institut de Myologie, Paris, France
| |
Collapse
|
17
|
Zhang Y, Zeuthen C, Zhu C, Wu F, Mezzell AT, Whitlow TJ, Choo HJ, Vest KE. Pharyngeal pathology in a mouse model of oculopharyngeal muscular dystrophy is associated with impaired basal autophagy in myoblasts. Front Cell Dev Biol 2022; 10:986930. [PMID: 36313551 PMCID: PMC9614327 DOI: 10.3389/fcell.2022.986930] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Accepted: 09/26/2022] [Indexed: 11/05/2023] Open
Abstract
Oculopharyngeal muscular dystrophy (OPMD) is a late-onset dominant disease that primarily affects craniofacial muscles. Despite the fact that the genetic cause of OPMD is known to be expansion mutations in the gene encoding the nuclear polyadenosine RNA binding protein PABPN1, the molecular mechanisms of pathology are unknown and no pharmacologic treatments are available. Due to the limited availability of patient tissues, several animal models have been employed to study the pathology of OPMD. However, none of these models have demonstrated functional deficits in the muscles of the pharynx, which are predominantly affected by OPMD. Here, we used a knock-in mouse model of OPMD, Pabpn1 +/A17 , that closely genocopies patients. In Pabpn1 +/A17 mice, we detected impaired pharyngeal muscle function, and impaired pharyngeal satellite cell proliferation and fusion. Molecular studies revealed that basal autophagy, which is required for normal satellite cell function, is higher in pharynx-derived myoblasts than in myoblasts derived from limb muscles. Interestingly, basal autophagy is impaired in cells derived from Pabpn1 +/A17 mice. Pabpn1 knockdown in pharyngeal myoblasts failed to recapitulate the autophagy defect detected in Pabpn1 +/A17 myoblasts suggesting that loss of PABPN1 function does not contribute to the basal autophagy defect. Taken together, these studies provide the first evidence for pharyngeal muscle and satellite cell pathology in a mouse model of OPMD and suggest that aberrant gain of PABPN1 function contributes to the craniofacial pathology in OPMD.
Collapse
Affiliation(s)
- Yu Zhang
- Department of Molecular Genetics, Biochemistry and Microbiology, University of Cincinnati College of Medicine, Cincinnati, OH, United States
| | - Christopher Zeuthen
- Department of Cell Biology, Emory University School of Medicine, Atlanta, GA, United States
| | - Carol Zhu
- Department of Cell Biology, Emory University School of Medicine, Atlanta, GA, United States
| | - Fang Wu
- Department of Cell Biology, Emory University School of Medicine, Atlanta, GA, United States
| | - Allison T. Mezzell
- Department of Molecular Genetics, Biochemistry and Microbiology, University of Cincinnati College of Medicine, Cincinnati, OH, United States
| | - Thomas J. Whitlow
- Department of Molecular Genetics, Biochemistry and Microbiology, University of Cincinnati College of Medicine, Cincinnati, OH, United States
| | - Hyojung J. Choo
- Department of Cell Biology, Emory University School of Medicine, Atlanta, GA, United States
| | - Katherine E. Vest
- Department of Molecular Genetics, Biochemistry and Microbiology, University of Cincinnati College of Medicine, Cincinnati, OH, United States
| |
Collapse
|
18
|
Richard P, Stojkovic T, Metay C, Lacau St Guily J, Trollet C. Distrofia muscolare oculofaringea. Neurologia 2022. [DOI: 10.1016/s1634-7072(22)46725-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
|
19
|
Guo Y, Kratky V, Xie H, Shentu X, Man X, Wang Y, Wen W, Rokohl AC, Heindl LM. Grand Challenges and Opportunities in Surgical Ophthalmology: Together for a Shared Future. FRONTIERS IN OPHTHALMOLOGY 2022; 2:922240. [PMID: 38983527 PMCID: PMC11182242 DOI: 10.3389/fopht.2022.922240] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/17/2022] [Accepted: 06/06/2022] [Indexed: 07/11/2024]
Affiliation(s)
- Yongwei Guo
- Eye Center, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Zhejiang University Eye Hospital, Hangzhou, China
- Zhejiang Provincial Key Lab of Ophthalmology, Hangzhou, China
| | - Vladimir Kratky
- Department of Ophthalmology, Queen's University, Kingston, ON, Canada
| | - Huatao Xie
- Department of Ophthalmology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xingchao Shentu
- Eye Center, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Zhejiang University Eye Hospital, Hangzhou, China
- Zhejiang Provincial Key Lab of Ophthalmology, Hangzhou, China
| | - Xiaofei Man
- Department of Ophthalmology, Xinhua Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Yanling Wang
- Department of Ophthalmology, Beijing Friendship Hospital Affiliated to Capital Medical University, Beijing, China
| | - Wen Wen
- Department of Ophthalmology and Visual Science, Eye and ENT Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| | - Alexander C Rokohl
- Department of Ophthalmology, University of Cologne, Faculty of Medicine and University Hospital of Cologne, Cologne, Germany
- Center for Integrated Oncology (CIO) Aachen-Bonn-Cologne-Duesseldorf, Cologne, Germany
| | - Ludwig M Heindl
- Department of Ophthalmology, University of Cologne, Faculty of Medicine and University Hospital of Cologne, Cologne, Germany
- Center for Integrated Oncology (CIO) Aachen-Bonn-Cologne-Duesseldorf, Cologne, Germany
| |
Collapse
|
20
|
Implications of Poly(A) Tail Processing in Repeat Expansion Diseases. Cells 2022; 11:cells11040677. [PMID: 35203324 PMCID: PMC8870147 DOI: 10.3390/cells11040677] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Revised: 02/11/2022] [Accepted: 02/13/2022] [Indexed: 11/21/2022] Open
Abstract
Repeat expansion diseases are a group of more than 40 disorders that affect mainly the nervous and/or muscular system and include myotonic dystrophies, Huntington’s disease, and fragile X syndrome. The mutation-driven expanded repeat tract occurs in specific genes and is composed of tri- to dodeca-nucleotide-long units. Mutant mRNA is a pathogenic factor or important contributor to the disease and has great potential as a therapeutic target. Although repeat expansion diseases are quite well known, there are limited studies concerning polyadenylation events for implicated transcripts that could have profound effects on transcript stability, localization, and translation efficiency. In this review, we briefly present polyadenylation and alternative polyadenylation (APA) mechanisms and discuss their role in the pathogenesis of selected diseases. We also discuss several methods for poly(A) tail measurement (both transcript-specific and transcriptome-wide analyses) and APA site identification—the further development and use of which may contribute to a better understanding of the correlation between APA events and repeat expansion diseases. Finally, we point out some future perspectives on the research into repeat expansion diseases, as well as APA studies.
Collapse
|
21
|
Galli F, Mouly V, Butler-Browne G, Cossu G. Challenges in cell transplantation for muscular dystrophy. Exp Cell Res 2021; 409:112908. [PMID: 34736920 DOI: 10.1016/j.yexcr.2021.112908] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Revised: 09/21/2021] [Accepted: 10/29/2021] [Indexed: 11/28/2022]
Abstract
For decades now, cell transplantation has been considered a possible therapeutic strategy for muscular dystrophy, but failures have largely outnumbered success or at least encouraging outcomes. In this review we will briefly recall the history of cell transplantation, discuss the peculiar features of skeletal muscle, and dystrophic skeletal muscle in particular, that make the procedure complicated and inefficient. As there are many recent and exhaustive reviews on the various myogenic cell types that have been or will be transplanted, we will only briefly describe them and refer the reader to these reviews. Finally, we will discuss possible strategies to overcome the hurdles that prevent biological efficacy and hence clinical success.
Collapse
Affiliation(s)
- Francesco Galli
- Division of Cell Matrix Biology & Regenerative Medicine, University of Manchester, UK
| | - Vincent Mouly
- Sorbonne Université, Inserm, Institut de Myologie, Centre de Recherche en Myologie, Paris, France
| | - Gillian Butler-Browne
- Sorbonne Université, Inserm, Institut de Myologie, Centre de Recherche en Myologie, Paris, France
| | - Giulio Cossu
- Division of Cell Matrix Biology & Regenerative Medicine, University of Manchester, UK; Muscle Research Unit, Charité Medical Faculty and Max Delbrück Center, Berlin, Germany; Division of Neuroscience, IRCCS Ospedale San Raffaele, Milan, Italy.
| |
Collapse
|
22
|
Hakim CH, Kumar SRP, Pérez-López DO, Wasala NB, Zhang D, Yue Y, Teixeira J, Pan X, Zhang K, Million ED, Nelson CE, Metzger S, Han J, Louderman JA, Schmidt F, Feng F, Grimm D, Smith BF, Yao G, Yang NN, Gersbach CA, Chen SJ, Herzog RW, Duan D. Cas9-specific immune responses compromise local and systemic AAV CRISPR therapy in multiple dystrophic canine models. Nat Commun 2021; 12:6769. [PMID: 34819506 PMCID: PMC8613397 DOI: 10.1038/s41467-021-26830-7] [Citation(s) in RCA: 86] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Accepted: 10/21/2021] [Indexed: 11/12/2022] Open
Abstract
Adeno-associated virus (AAV)-mediated CRISPR-Cas9 editing holds promise to treat many diseases. The immune response to bacterial-derived Cas9 has been speculated as a hurdle for AAV-CRISPR therapy. However, immunological consequences of AAV-mediated Cas9 expression have thus far not been thoroughly investigated in large mammals. We evaluate Cas9-specific immune responses in canine models of Duchenne muscular dystrophy (DMD) following intramuscular and intravenous AAV-CRISPR therapy. Treatment results initially in robust dystrophin restoration in affected dogs but also induces muscle inflammation, and Cas9-specific humoral and cytotoxic T-lymphocyte (CTL) responses that are not prevented by the muscle-specific promoter and transient prednisolone immune suppression. In normal dogs, AAV-mediated Cas9 expression induces similar, though milder, immune responses. In contrast, other therapeutic (micro-dystrophin and SERCA2a) and reporter (alkaline phosphatase, AP) vectors result in persistent expression without inducing muscle inflammation. Our results suggest Cas9 immunity may represent a critical barrier for AAV-CRISPR therapy in large mammals.
Collapse
Affiliation(s)
- Chady H Hakim
- Department of Molecular Microbiology and Immunology, The University of Missouri, Columbia, MO, USA
- National Center for Advancing Translational Sciences, NIH, Rockville, MD, USA
| | - Sandeep R P Kumar
- Department of Pediatrics, Indiana University, Indianapolis, IN, USA
- Herman B Wells Center for Pediatric Research, Indiana University, Indianapolis, IN, USA
| | - Dennis O Pérez-López
- Department of Molecular Microbiology and Immunology, The University of Missouri, Columbia, MO, USA
| | - Nalinda B Wasala
- Department of Molecular Microbiology and Immunology, The University of Missouri, Columbia, MO, USA
| | - Dong Zhang
- Department of Physics, The University of Missouri, Columbia, MO, USA
- Department of Biochemistry, The University of Missouri, Columbia, MO, USA
- Institute for Data Science and Informatics, The University of Missouri, Columbia, MO, USA
| | - Yongping Yue
- Department of Molecular Microbiology and Immunology, The University of Missouri, Columbia, MO, USA
| | - James Teixeira
- Department of Molecular Microbiology and Immunology, The University of Missouri, Columbia, MO, USA
| | - Xiufang Pan
- Department of Molecular Microbiology and Immunology, The University of Missouri, Columbia, MO, USA
| | - Keqing Zhang
- Department of Molecular Microbiology and Immunology, The University of Missouri, Columbia, MO, USA
| | - Emily D Million
- Department of Molecular Microbiology and Immunology, The University of Missouri, Columbia, MO, USA
| | - Christopher E Nelson
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
- Center for Advanced Genomic Technologies Biology, Duke University, Durham, NC, USA
| | - Samantha Metzger
- Department of Molecular Microbiology and Immunology, The University of Missouri, Columbia, MO, USA
| | - Jin Han
- Department of Molecular Microbiology and Immunology, The University of Missouri, Columbia, MO, USA
| | - Jacqueline A Louderman
- Department of Molecular Microbiology and Immunology, The University of Missouri, Columbia, MO, USA
| | - Florian Schmidt
- Department of Infectious Diseases/Virology, University of Heidelberg, Heidelberg, Germany
- Cluster of Excellence CellNetworks, University of Heidelberg, Heidelberg, Germany
- BioQuant, University of Heidelberg, Heidelberg, Germany
| | - Feng Feng
- Department of Molecular Microbiology and Immunology, The University of Missouri, Columbia, MO, USA
| | - Dirk Grimm
- Department of Infectious Diseases/Virology, University of Heidelberg, Heidelberg, Germany
- Cluster of Excellence CellNetworks, University of Heidelberg, Heidelberg, Germany
- BioQuant, University of Heidelberg, Heidelberg, Germany
| | - Bruce F Smith
- Department of Pathobiology, Auburn University, Auburn, AL, USA
- Scott-Ritchey Research Center, Auburn University, Auburn, AL, USA
| | - Gang Yao
- Department of Biomedical, Biological & Chemical Engineering, The University of Missouri, Columbia, MO, USA
| | - N Nora Yang
- National Center for Advancing Translational Sciences, NIH, Rockville, MD, USA
| | - Charles A Gersbach
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
- Center for Advanced Genomic Technologies Biology, Duke University, Durham, NC, USA
- Department of Surgery, Duke University Medical Center, Durham, NC, USA
| | - Shi-Jie Chen
- Department of Physics, The University of Missouri, Columbia, MO, USA
- Department of Biochemistry, The University of Missouri, Columbia, MO, USA
- Institute for Data Science and Informatics, The University of Missouri, Columbia, MO, USA
| | - Roland W Herzog
- Department of Pediatrics, Indiana University, Indianapolis, IN, USA
- Herman B Wells Center for Pediatric Research, Indiana University, Indianapolis, IN, USA
| | - Dongsheng Duan
- Department of Molecular Microbiology and Immunology, The University of Missouri, Columbia, MO, USA.
- Department of Biomedical, Biological & Chemical Engineering, The University of Missouri, Columbia, MO, USA.
- Department of Neurology, The University of Missouri, Columbia, MO, USA.
- Department of Biomedical Sciences, The University of Missouri, Columbia, MO, USA.
| |
Collapse
|
23
|
Kao BR, Malerba A, Lu-Nguyen NB, Harish P, McCarthy JJ, Dickson G, Popplewell LJ. Knockdown of Muscle-Specific Ribosomal Protein L3-Like Enhances Muscle Function in Healthy and Dystrophic Mice. Nucleic Acid Ther 2021; 31:457-464. [PMID: 34081545 DOI: 10.1089/nat.2020.0928] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Ribosomal protein L3-like (RPL3L) is a poorly characterized ribosomal protein that is exclusively expressed in skeletal and cardiac muscle. RPL3L is also downregulated in Duchenne muscular dystrophy (DMD), suggesting that it may play an important role in muscle biology. In this study, we investigated the role of RPL3L in skeletal muscle of healthy C57 and dystrophic mdx mice. We show that RPL3L is developmentally regulated and that intramuscular adeno-associated virus (AAV)-mediated RPL3L knockdown in the tibialis anterior of C57 and mdx mice results in increased specific force with improved resistance to eccentric contraction induced muscle damage in dystrophic muscles. The mechanism by which RPL3L knockdown improves muscle function remains unclear. Histological observations showed a significant increase in muscle length and decrease in muscle cross-sectional area after RPL3L inhibition suggesting that this ribosomal protein may play a role in myofiber morphology. The endogenous downregulation of RPL3L in DMD may be a protective mechanism that attempts to improve skeletal muscle function and counteract the dystrophic phenotype.
Collapse
Affiliation(s)
- Betty R Kao
- Department of Biological Sciences, School of Life Sciences and the Environment, Royal Holloway University of London, Egham, United Kingdom
| | - Alberto Malerba
- Department of Biological Sciences, School of Life Sciences and the Environment, Royal Holloway University of London, Egham, United Kingdom
| | - Ngoc B Lu-Nguyen
- Department of Biological Sciences, School of Life Sciences and the Environment, Royal Holloway University of London, Egham, United Kingdom
| | - Pradeep Harish
- Department of Biological Sciences, School of Life Sciences and the Environment, Royal Holloway University of London, Egham, United Kingdom
| | - John J McCarthy
- Department of Physiology, University of Kentucky, Lexington, Kentucky, USA
| | - George Dickson
- Department of Biological Sciences, School of Life Sciences and the Environment, Royal Holloway University of London, Egham, United Kingdom
| | - Linda J Popplewell
- Department of Biological Sciences, School of Life Sciences and the Environment, Royal Holloway University of London, Egham, United Kingdom
| |
Collapse
|
24
|
Yamashita S. Recent Progress in Oculopharyngeal Muscular Dystrophy. J Clin Med 2021; 10:jcm10071375. [PMID: 33805441 PMCID: PMC8036457 DOI: 10.3390/jcm10071375] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Revised: 03/18/2021] [Accepted: 03/26/2021] [Indexed: 12/23/2022] Open
Abstract
Oculopharyngeal muscular dystrophy (OPMD) is a late-onset intractable myopathy, characterized by slowly progressive ptosis, dysphagia, and proximal limb weakness. It is caused by the abnormal expansion of the alanine-encoding (GCN)n trinucleotide repeat in the exon 1 of the polyadenosine (poly[A]) binding protein nuclear 1 gene (11-18 repeats in OPMD instead of the normal 10 repeats). As the disease progresses, the patients gradually develop a feeling of suffocation, regurgitation of food, and aspiration pneumonia, although the initial symptoms and the progression patterns vary among the patients. Autologous myoblast transplantation may provide therapeutic benefits by reducing swallowing problems in these patients. Therefore, it is important to assemble information on such patients for the introduction of effective treatments in nonendemic areas. Herein, we present a concise review of recent progress in clinical and pathological studies of OPMD and introduce an idea for setting up a nation-wide OPMD disease registry in Japan. Since it is important to understand patients' unmet medical needs, realize therapeutically targetable symptoms, and identify indices of therapeutic efficacy, our attempt to establish a unique patient registry of OPMD will be a helpful tool to address these urgent issues.
Collapse
Affiliation(s)
- Satoshi Yamashita
- Department of Neurology, Graduate School of Medical Sciences, Kumamoto University, Kumamoto 860-8556, Japan
| |
Collapse
|
25
|
Carraro U, Yablonka-Reuveni Z. Translational research on Myology and Mobility Medicine: 2021 semi-virtual PDM3 from Thermae of Euganean Hills, May 26 - 29, 2021. Eur J Transl Myol 2021; 31:9743. [PMID: 33733717 PMCID: PMC8056169 DOI: 10.4081/ejtm.2021.9743] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Accepted: 03/17/2021] [Indexed: 02/08/2023] Open
Abstract
On 19-21 November 2020, the meeting of the 30 years of the Padova Muscle Days was virtually held while the SARS-CoV-2 epidemic was hitting the world after a seemingly quiet summer. During the 2020-2021 winter, the epidemic is still active, despite the start of vaccinations. The organizers hope to hold the 2021 Padua Days on Myology and Mobility Medicine in a semi-virtual form (2021 S-V PDM3) from May 26 to May 29 at the Thermae of Euganean Hills, Padova, Italy. Here the program and the Collection of Abstracts are presented. Despite numerous world problems, the number of submitted/selected presentations (lectures and oral presentations) has increased, prompting the organizers to extend the program to four dense days.
Collapse
Affiliation(s)
- Ugo Carraro
- Department of Biomedical Sciences of the University of Padova, Italy; CIR-Myo - Myology Centre, University of Padova, Italy; A-C Mioni-Carraro Foundation for Translational Myology, Padova.
| | - Zipora Yablonka-Reuveni
- Department of Biological Structure, University of Washington School of Medicine, Seattle, WA.
| |
Collapse
|
26
|
Baker JD, Uhrich RL, Strovas TJ, Saxton AD, Kraemer BC. AlphaScreen Identifies MSUT2 Inhibitors for Tauopathy-Targeting Therapeutic Discovery. SLAS DISCOVERY : ADVANCING LIFE SCIENCES R & D 2021; 26:400-409. [PMID: 32981422 PMCID: PMC8592089 DOI: 10.1177/2472555220958387] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Tauopathies are neurological disorders characterized by intracellular tau deposits forming neurofibrillary tangles, neuropil threads, or other disease-specific aggregates composed of the protein tau. Tauopathy disorders include frontotemporal lobar degeneration, corticobasal degeneration, Pick's disease, and the largest cause of dementia, Alzheimer's disease. The lack of disease-modifying therapeutic strategies to address tauopathies remains a critical unmet need in dementia care. Thus, novel broad-spectrum tau-targeted therapeutics could have a profound impact in multiple tauopathy disorders, including Alzheimer's disease. Here we have designed a drug discovery paradigm to identify inhibitors of the pathological tau-enabling protein, MSUT2. We previously showed that activity of the RNA-binding protein MSUT2 drives tauopathy, including tau-mediated neurodegeneration and cognitive dysfunction, in mouse models. Thus, we hypothesized that MSUT2 inhibitors could be therapeutic for tauopathy disorders. Our pipeline for MSUT2 inhibitory compound identification included a primary AlphaScreen, followed by dose-response validation, a secondary fluorescence polarization orthogonal assay, a tertiary specificity screen, and a preliminary toxicity screen. Our work here serves as a proof-of-principle methodology for finding specific inhibitors of the poly(A) RNA-binding protein MSUT2 interaction. Here we identify 4,4'-diisothiocyanostilbene-2,2'-sulfonic acid (DIDS) as a potential tool compound for future work probing the mechanism of MSUT2-induced tau pathology.
Collapse
Affiliation(s)
- Jeremy D. Baker
- Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Washington, Seattle, WA, USA
- Geriatric Research Education and Clinical Center, Veterans Affairs Puget Sound Health Care System, Seattle, WA, USA
| | - Rikki L. Uhrich
- Geriatric Research Education and Clinical Center, Veterans Affairs Puget Sound Health Care System, Seattle, WA, USA
| | - Timothy J. Strovas
- Geriatric Research Education and Clinical Center, Veterans Affairs Puget Sound Health Care System, Seattle, WA, USA
| | - Aleen D. Saxton
- Geriatric Research Education and Clinical Center, Veterans Affairs Puget Sound Health Care System, Seattle, WA, USA
| | - Brian C. Kraemer
- Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Washington, Seattle, WA, USA
- Geriatric Research Education and Clinical Center, Veterans Affairs Puget Sound Health Care System, Seattle, WA, USA
- Department of Psychiatry and Behavioral Sciences, University of Washington, Seattle, WA, USA
- Department of Pathology, University of Washington, Seattle, WA, USA
| |
Collapse
|
27
|
Strings-Ufombah V, Malerba A, Kao SC, Harbaran S, Roth F, Cappellari O, Lu-Nguyen N, Takahashi K, Mukadam S, Kilfoil G, Kloth C, Roelvink P, Dickson G, Trollet C, Suhy D. BB-301: a silence and replace AAV-based vector for the treatment of oculopharyngeal muscular dystrophy. MOLECULAR THERAPY-NUCLEIC ACIDS 2021; 24:67-78. [PMID: 33738139 PMCID: PMC7940701 DOI: 10.1016/j.omtn.2021.02.017] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Accepted: 02/14/2021] [Indexed: 11/08/2022]
Abstract
Oculopharyngeal muscular dystrophy (OPMD) is a rare autosomal dominant disease that results from an alanine expansion in the N-terminal domain of Poly-A Binding Protein Nuclear-1 (PABPN1). We have recently demonstrated that a two-vector gene therapy strategy significantly ameliorated the pathology in a mouse model of OPMD. This approach entailed intramuscular injection of two recombinant adeno-associated viruses (AAVs), one expressing three short hairpin RNAs (shRNAs) to silence both mutant and wild-type PABPN1 and one expressing a codon-optimized version of PABPN1 that is insensitive to RNA interference. Here we report the continued development of this therapeutic strategy by delivering “silence and replace” sequences in a single AAV vector named BB-301. This construct is composed of a modified AAV serotype 9 (AAV9) capsid that expresses a unique single bifunctional construct under the control of the muscle-specific Spc5-12 promoter for the co-expression of both the codon-optimized PABPN1 protein and two small inhibitory RNAs (siRNAs) against PABPN1 modeled into microRNA (miRNA) backbones. A single intramuscular injection of BB-301 results in robust inhibition of mutant PABPN1 and concomitant replacement of the codon-optimized PABPN1 protein. The treatment restores muscle strength and muscle weight to wild-type levels as well as improving other physiological hallmarks of the disease in a mouse model of OPMD.
Collapse
Affiliation(s)
| | - Alberto Malerba
- Department of Biological Sciences, School of Life Sciences and the Environment, Royal Holloway University of London, Egham, Surrey TW20 0EX, UK
| | | | | | - Fanny Roth
- Sorbonne Université, INSERM, Association Institut de Myologie, Centre de Recherche en Myologie, 75013 Paris, France
| | - Ornella Cappellari
- Department of Biological Sciences, School of Life Sciences and the Environment, Royal Holloway University of London, Egham, Surrey TW20 0EX, UK
| | - Ngoc Lu-Nguyen
- Department of Biological Sciences, School of Life Sciences and the Environment, Royal Holloway University of London, Egham, Surrey TW20 0EX, UK
| | | | | | | | | | | | - George Dickson
- Department of Biological Sciences, School of Life Sciences and the Environment, Royal Holloway University of London, Egham, Surrey TW20 0EX, UK
| | - Capucine Trollet
- Sorbonne Université, INSERM, Association Institut de Myologie, Centre de Recherche en Myologie, 75013 Paris, France
| | - David Suhy
- Benitec Biopharma, Inc., Hayward, CA 94545, USA
| |
Collapse
|
28
|
Skopenkova VV, Egorova TV, Bardina MV. Muscle-Specific Promoters for Gene Therapy. Acta Naturae 2021; 13:47-58. [PMID: 33959386 PMCID: PMC8084301 DOI: 10.32607/actanaturae.11063] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2020] [Accepted: 07/30/2020] [Indexed: 12/19/2022] Open
Abstract
Many genetic diseases that are responsible for muscular disorders have been described to date. Gene replacement therapy is a state-of-the-art strategy used to treat such diseases. In this approach, the functional copy of a gene is delivered to the affected tissues using viral vectors. There is an urgent need for the design of short, regulatory sequences that would drive a high and robust expression of a therapeutic transgene in skeletal muscles, the diaphragm, and the heart, while exhibiting limited activity in non-target tissues. This review focuses on the development and improvement of muscle-specific promoters based on skeletal muscle α-actin, muscle creatine kinase, and desmin genes, as well as other genes expressed in muscles. The current approaches used to engineer synthetic muscle-specific promoters are described. Other elements of the viral vectors that contribute to tissue-specific expression are also discussed. A special feature of this review is the presence of up-to-date information on the clinical and preclinical trials of gene therapy drug candidates that utilize muscle-specific promoters.
Collapse
Affiliation(s)
- V. V. Skopenkova
- Institute of Gene Biology, Russian Academy of Sciences, Moscow, 119334 Russia
- Marlin Biotech LLC, Moscow, 121205 Russia
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Institute of Gene Biology, Russian Academy of Sciences, Moscow, 119334 Russia
| | - T. V. Egorova
- Institute of Gene Biology, Russian Academy of Sciences, Moscow, 119334 Russia
- Marlin Biotech LLC, Moscow, 121205 Russia
| | - M. V. Bardina
- Institute of Gene Biology, Russian Academy of Sciences, Moscow, 119334 Russia
- Marlin Biotech LLC, Moscow, 121205 Russia
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Institute of Gene Biology, Russian Academy of Sciences, Moscow, 119334 Russia
| |
Collapse
|
29
|
Weskamp K, Olwin BB, Parker R. Post-Transcriptional Regulation in Skeletal Muscle Development, Repair, and Disease. Trends Mol Med 2020; 27:469-481. [PMID: 33384234 DOI: 10.1016/j.molmed.2020.12.002] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Revised: 11/30/2020] [Accepted: 12/02/2020] [Indexed: 12/14/2022]
Abstract
Skeletal muscle formation is a complex process that requires tight spatiotemporal control of key myogenic factors. Emerging evidence suggests that RNA processing is crucial for the regulation of these factors, and that multiple post-transcriptional regulatory pathways work dependently and independently of one another to enable precise control of transcripts throughout muscle development and repair. Moreover, disruption of these pathways is implicated in neuromuscular disease, and the recent development of RNA-mediated therapies shows enormous promise in the treatment of these disorders. We discuss the overlapping post-transcriptional regulatory pathways that mediate muscle development, how these pathways are disrupted in neuromuscular disorders, and advances in RNA-mediated therapies that present a novel approach to the treatment of these diseases.
Collapse
Affiliation(s)
- Kaitlin Weskamp
- Department of Biochemistry, University of Colorado Boulder, Boulder, CO, USA.
| | - Bradley B Olwin
- Department of Molecular, Cellular and Developmental Biology, University of Colorado Boulder, Boulder, CO, USA
| | - Roy Parker
- Department of Biochemistry, University of Colorado Boulder, Boulder, CO, USA; Howard Hughes Medical Institute, Chevy Chase, MD, USA.
| |
Collapse
|
30
|
The Impact of Mitochondrial Deficiencies in Neuromuscular Diseases. Antioxidants (Basel) 2020; 9:antiox9100964. [PMID: 33050147 PMCID: PMC7600520 DOI: 10.3390/antiox9100964] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Revised: 09/23/2020] [Accepted: 09/24/2020] [Indexed: 02/06/2023] Open
Abstract
Neuromuscular diseases (NMDs) are a heterogeneous group of acquired or inherited rare disorders caused by injury or dysfunction of the anterior horn cells of the spinal cord (lower motor neurons), peripheral nerves, neuromuscular junctions, or skeletal muscles leading to muscle weakness and waste. Unfortunately, most of them entail serious or even fatal consequences. The prevalence rates among NMDs range between 1 and 10 per 100,000 population, but their rarity and diversity pose difficulties for healthcare and research. Some molecular hallmarks are being explored to elucidate the mechanisms triggering disease, to set the path for further advances. In fact, in the present review we outline the metabolic alterations of NMDs, mainly focusing on the role of mitochondria. The aim of the review is to discuss the mechanisms underlying energy production, oxidative stress generation, cell signaling, autophagy, and inflammation triggered or conditioned by the mitochondria. Briefly, increased levels of inflammation have been linked to reactive oxygen species (ROS) accumulation, which is key in mitochondrial genomic instability and mitochondrial respiratory chain (MRC) dysfunction. ROS burst, impaired autophagy, and increased inflammation are observed in many NMDs. Increasing knowledge of the etiology of NMDs will help to develop better diagnosis and treatments, eventually reducing the health and economic burden of NMDs for patients and healthcare systems.
Collapse
|
31
|
Baker JD, Uhrich RL, Strovas TJ, Saxton AD, Kraemer BC. Targeting Pathological Tau by Small Molecule Inhibition of the Poly(A):MSUT2 RNA-Protein Interaction. ACS Chem Neurosci 2020; 11:2277-2285. [PMID: 32589834 PMCID: PMC8629322 DOI: 10.1021/acschemneuro.0c00214] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Neurofibrillary tangles composed of aberrantly aggregating tau protein are a hallmark of Alzheimer's disease and related dementia disorders. Recent work has shown that mammalian suppressor of tauopathy 2 (MSUT2), also named ZC3H14 (Zinc Finger CCCH-Type Containing 14), controls accumulation of pathological tau in cultured human cells and mice. Knocking out MSUT2 protects neurons from neurodegenerative tauopathy and preserves learning and memory. MSUT2 protein functions to bind polyadenosine [poly(A)] tails of mRNA through its C-terminal CCCH type zinc finger domains, and loss of CCCH domain function suppresses tauopathy in Caenorhabditis elegans and mice. Thus, we hypothesized that inhibiting the poly(A):MSUT2 RNA-protein interaction would ameliorate pathological tau accumulation. Here we present a high-throughput screening method for the identification of small molecules inhibiting the poly(A):MSUT2 RNA-protein interaction. We employed a fluorescent polarization assay for initial small molecule discovery with the intention to repurpose hits identified from the NIH Clinical Collection (NIHCC). Our drug repurposing development workflow included validation of hits by dose-response analysis, specificity testing, orthogonal assays of activity, and cytotoxicity. Validated compounds passing through this screening funnel will be evaluated for translational effectiveness in future studies. This preclinical drug development pipeline identified diverse FDA approved drugs duloxetine, saquinavir, and clofazimine as potential repurposing candidates for reducing pathological tau accumulation.
Collapse
Affiliation(s)
- Jeremy D Baker
- Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Washington, Seattle, Washington 98104, United States
- Geriatrics Research Education and Clinical Center, Veterans Affairs Puget Sound Health Care System, Seattle, Washington 98108, United States
| | - Rikki L Uhrich
- Geriatrics Research Education and Clinical Center, Veterans Affairs Puget Sound Health Care System, Seattle, Washington 98108, United States
| | - Timothy J Strovas
- Geriatrics Research Education and Clinical Center, Veterans Affairs Puget Sound Health Care System, Seattle, Washington 98108, United States
| | - Aleen D Saxton
- Geriatrics Research Education and Clinical Center, Veterans Affairs Puget Sound Health Care System, Seattle, Washington 98108, United States
| | - Brian C Kraemer
- Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Washington, Seattle, Washington 98104, United States
- Geriatrics Research Education and Clinical Center, Veterans Affairs Puget Sound Health Care System, Seattle, Washington 98108, United States
- Department of Psychiatry and Behavioral Sciences, University of Washington, Seattle, Washington 98195, United States
- Department of Pathology, University of Washington, Seattle, Washington 98195, United States
| |
Collapse
|
32
|
Kariyawasam D, Alexander IE, Kurian M, Farrar MA. Great expectations: virus-mediated gene therapy in neurological disorders. J Neurol Neurosurg Psychiatry 2020; 91:849-860. [PMID: 32503884 DOI: 10.1136/jnnp-2019-322327] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Revised: 04/21/2020] [Accepted: 04/22/2020] [Indexed: 12/25/2022]
Abstract
Gene therapy (GT) has tremendous potential for the treatment of neurological disorders to transform patient care. The successful application of virus-mediated GT to treat spinal muscular atrophy is a significant milestone, serving to accelerate similar progress in a spectrum of neurological conditions, with more than 50 clinical trials currently underway, across neurodevelopmental, neurodegenerative, muscular dystrophy, epilepsy, chronic pain and neoplastic diseases. This review provides an overview of the key features of virus-mediated GT, paradigms of delivery and dosing, potential risks and highlights ongoing research to optimise safe and effective delivery of vectors into the nervous system. Examples of the application of GT in various neurological diseases alongside clinical development challenges will be presented. As the development and translation of GTs gain pace, success can only ultimately be realised for patients following implementation in the health system. The challenges and controversies of daunting costs, ethics, early diagnosis and health system readiness will require innovative pricing schemes, regulatory policies, education and organisation of a skilled workforce to deliver of high-quality care in clinical practice as we prepare for advanced therapeutics in neurology.
Collapse
Affiliation(s)
- Didu Kariyawasam
- Neurology, Sydney Children's Hospital Randwick, Randwick, New South Wales, Australia.,School of Women's and Children's Health, UNSW Medicine, University of New South Wales, Sydney, New South Wales, Australia
| | - Ian E Alexander
- Discipline of Child and Adolescent Health, Sydney Medical School, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia.,Gene Therapy Unit, Children's Medical Research Institute, Westmead, New South Wales, Australia
| | - Manju Kurian
- Neurosciences Unit, Institute of Child Health, University College London, London, UK.,Neurology, Great Ormond Street Hospital for Children, London, UK
| | - Michelle Anne Farrar
- Neurology, Sydney Children's Hospital Randwick, Randwick, New South Wales, Australia .,School of Women's and Children's Health, UNSW Medicine, University of New South Wales, Sydney, New South Wales, Australia
| |
Collapse
|
33
|
Oculopharyngeal Muscular Dystrophy, an Often Misdiagnosed Neuromuscular Disorder: A Southern California Experience. J Clin Neuromuscul Dis 2020; 21:61-68. [PMID: 31743248 DOI: 10.1097/cnd.0000000000000271] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
OBJECTIVE Oculopharyngeal muscular dystrophy (OPMD) is a rare neuromuscular disorder characterized by late-onset development of bilateral eyelid ptosis, ophthalmoparesis and dysphagia with further progression to proximal limb muscle weakness that is an under recognized condition. The mode of inheritance is usually autosomal dominant, but a recessive form has been reported. OPMD is caused by a short expansion of the alanine repeat (GCN trinucleotide) in the poly(adenylate)-binding protein nuclear1 (PABPN1) gene. METHODS We performed a retrospective review of undiagnosed cases that initially presented with ptosis, diplopia, dysphagia, muscle weakness, muscular dystrophy and/or myasthenia gravis from 2000 to 2015 at two institutions in Southern California. RESULTS Twenty-five patients were identified to have OPMD with genetic confirmation. CONCLUSIONS Even though a rare condition, the prevalence is disproportionally frequent in certain ethnic groups and in certain regions; thus, we report our experience of OPMD patients in Southern California.
Collapse
|
34
|
Malerba A, Klein P, Lu-Nguyen N, Cappellari O, Strings-Ufombah V, Harbaran S, Roelvink P, Suhy D, Trollet C, Dickson G. Established PABPN1 intranuclear inclusions in OPMD muscle can be efficiently reversed by AAV-mediated knockdown and replacement of mutant expanded PABPN1. Hum Mol Genet 2020; 28:3301-3308. [PMID: 31294444 DOI: 10.1093/hmg/ddz167] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2019] [Revised: 06/21/2019] [Accepted: 07/08/2019] [Indexed: 11/12/2022] Open
Abstract
Oculopharyngeal muscular dystrophy (OPMD) is a rare autosomal dominant late-onset muscular dystrophy affecting approximately 1:100 000 individuals in Europe. OPMD is mainly characterized by progressive eyelid drooping (ptosis) and dysphagia although muscles of the limbs can also be affected late in life. This muscle disease is due to a trinucleotide repeat expansion in the polyA-binding protein nuclear-1 gene. Patients express a protein with an 11-18 alanine tract that is misfolded and prone to form intranuclear inclusions, which are the hallmark of the disease. Other features of OPMD include muscle fibrosis and atrophy in affected muscles. Currently, no pharmacological treatments are available, and OPMD patients can only be referred to surgeons for cricopharyngeal myotomy or corrective surgery of extraocular muscles to ease ptosis. We recently tested a two-AAV `silence' and `replace' vector-based gene therapy treatment in a mouse model of OPMD. We demonstrate here that this gene therapy approach can revert already established insoluble aggregates and partially rescues the muscle from atrophy, which are both crucially important since in most cases OPMD patients already have an established disease when diagnosed. This strategy also prevents the formation of muscle fibrosis and stabilizes the muscle strength to the level of healthy muscles. Furthermore, we show here that similar results can be obtained using a single AAV vector incorporating both the `silence' and `replace' cassettes. These results further support the application of a gene therapy approach as a novel treatment for OPMD in humans.
Collapse
Affiliation(s)
- Alberto Malerba
- Centres of Gene and Cell Therapy and Biomedical Sciences, School of Biological Sciences, Royal Holloway, University of London, Egham TW20 0EX, Surrey, UK
| | - Pierre Klein
- Sorbonne Université, INSERM, Association Institut de Myologie, Centre de Recherche en Myologie, UMRS974, 47 bd de l'Hôpital, 75013 Paris, France
| | - Ngoc Lu-Nguyen
- Centres of Gene and Cell Therapy and Biomedical Sciences, School of Biological Sciences, Royal Holloway, University of London, Egham TW20 0EX, Surrey, UK
| | - Ornella Cappellari
- Comparative Biomedical Sciences, Royal Veterinary College, London NW1 0TU, UK
| | | | | | | | - David Suhy
- Benitec Biopharma, Hayward, CA 94545, USA
| | - Capucine Trollet
- Sorbonne Université, INSERM, Association Institut de Myologie, Centre de Recherche en Myologie, UMRS974, 47 bd de l'Hôpital, 75013 Paris, France
| | - George Dickson
- Centres of Gene and Cell Therapy and Biomedical Sciences, School of Biological Sciences, Royal Holloway, University of London, Egham TW20 0EX, Surrey, UK
| |
Collapse
|
35
|
Shukla TN, Song J, Campbell ZT. Molecular entrapment by RNA: an emerging tool for disrupting protein-RNA interactions in vivo. RNA Biol 2020; 17:417-424. [PMID: 31957541 PMCID: PMC7237136 DOI: 10.1080/15476286.2020.1717059] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2019] [Revised: 12/09/2019] [Accepted: 01/12/2020] [Indexed: 10/25/2022] Open
Abstract
mRNA function is controlled by RNA-binding proteins. The specificity of RNA-binding factors for their targets is critical in that it enables all subsequent regulation. Despite widespread recognition of the pervasive role RNA-binding proteins play in development and disease, they remain challenging to target with small molecules. A renaissance in RNA therapeutics has led to the identification of modifications that substantially increase RNA stability. When combined with information regarding specificity, a new class of oligonucleotide mimics has emerged as a means to competitively disrupt the regulation of endogenous substrates. These decoys have been used to inhibit RNA-binding proteins in living animals. Decoys will likely provide new insights into the expansive roles of RNA-binding proteins in biology and disease. Here, we describe examples where they have been used and discuss how they could be applied to new targets.
Collapse
Affiliation(s)
- Tarjani N. Shukla
- The Department of Biological Sciences, University of Texas-Dallas, Richardson, TX, USA
| | - Jane Song
- The Department of Biological Sciences, University of Texas-Dallas, Richardson, TX, USA
| | - Zachary T. Campbell
- The Department of Biological Sciences, University of Texas-Dallas, Richardson, TX, USA
| |
Collapse
|
36
|
Harish P, Forrest L, Herath S, Dickson G, Malerba A, Popplewell L. Inhibition of Myostatin Reduces Collagen Deposition in a Mouse Model of Oculopharyngeal Muscular Dystrophy (OPMD) With Established Disease. Front Physiol 2020; 11:184. [PMID: 32194441 PMCID: PMC7066371 DOI: 10.3389/fphys.2020.00184] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Accepted: 02/17/2020] [Indexed: 12/12/2022] Open
Abstract
Background Oculopharyngeal muscular dystrophy (OPMD) is a late-onset muscle disease presented by ptosis, dysphagia, and limb weakness. Affected muscles display increased fibrosis and atrophy, with characteristic inclusion bodies in the nucleus. Myostatin is a negative regulator of muscle mass, and inhibition of myostatin has been demonstrated to improve symptoms in models of muscular dystrophy. Methods We systemically administered a monoclonal antibody to block myostatin in the A17 mouse model of OPMD at 42 weeks of age. The mice were administered a weekly dose of 10 mg/kg RK35 intraperitonially for 10 weeks, following which serum and histological analyses were performed on muscle samples. Results The administration of the antibody resulted in a significant decrease in serum myostatin and collagen deposition in muscles. However, minimal effects on body mass, muscle mass and myofiber diameter, or the density of intranuclear inclusions (INIs) (a hallmark of disease progression of OPMD) were observed. Conclusion This study demonstrates that inhibition of myostatin does not revert muscle atrophy in a mouse model with established OPMD disease, but is effective at reducing observed histological markers of fibrosis in the treated muscles.
Collapse
Affiliation(s)
- Pradeep Harish
- Department of Biological Sciences, Centre of Gene and Cell Therapy and Biomedical Sciences, Royal Holloway, University of London, Egham, United Kingdom
| | - Leysa Forrest
- Department of Biological Sciences, Centre of Gene and Cell Therapy and Biomedical Sciences, Royal Holloway, University of London, Egham, United Kingdom
| | - Shanti Herath
- Department of Biological Sciences, Centre of Gene and Cell Therapy and Biomedical Sciences, Royal Holloway, University of London, Egham, United Kingdom
| | - George Dickson
- Department of Biological Sciences, Centre of Gene and Cell Therapy and Biomedical Sciences, Royal Holloway, University of London, Egham, United Kingdom
| | - Alberto Malerba
- Department of Biological Sciences, Centre of Gene and Cell Therapy and Biomedical Sciences, Royal Holloway, University of London, Egham, United Kingdom
| | - Linda Popplewell
- Department of Biological Sciences, Centre of Gene and Cell Therapy and Biomedical Sciences, Royal Holloway, University of London, Egham, United Kingdom
| |
Collapse
|
37
|
Gonzalez-Alegre P. Recent advances in molecular therapies for neurological disease: triplet repeat disorders. Hum Mol Genet 2020; 28:R80-R87. [PMID: 31227833 DOI: 10.1093/hmg/ddz138] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2019] [Revised: 04/03/2019] [Accepted: 06/11/2019] [Indexed: 02/06/2023] Open
Abstract
Triplet repeat diseases (TRDs) are caused by pathogenic expansions of trinucleotide sequence repeats within coding and non-coding regions of different genes. They are typically progressive, very disabling and frequently involve the nervous system. Currently available symptomatic therapies provide modest benefit at best. The development of interventions that interfere with the natural history of these diseases is a priority. A common pathogenic process shared by most TRDs is the presence of toxicity from the messenger RNA or protein encoded by the gene harboring the abnormal expansion. Strategies to interfere with the expression of these genes using different molecular approaches are being pursued and have reached the clinical stage. This review will summarize the significant progress made in this field in the last few years, focusing on three main areas: the discovery of biomarkers of disease progression and target engagement, advances in preclinical studies for the polyglutamine ataxias and the initial clinical application in myotonic dystrophy type 1 and Huntington's disease.
Collapse
Affiliation(s)
- Pedro Gonzalez-Alegre
- Department of Neurology, the University of Pennsylvania, Philadelphia, PA 19104, USA.,Raymond G. Perelman Center for Cellular and Molecular Therapy, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| |
Collapse
|
38
|
Malerba A, Roth F, Harish P, Dhiab J, Lu-Nguyen N, Cappellari O, Jarmin S, Mahoudeau A, Ythier V, Lainé J, Negroni E, Abgueguen E, Simonelig M, Guedat P, Mouly V, Butler-Browne G, Voisset C, Dickson G, Trollet C. Pharmacological modulation of the ER stress response ameliorates oculopharyngeal muscular dystrophy. Hum Mol Genet 2020; 28:1694-1708. [PMID: 30649389 DOI: 10.1093/hmg/ddz007] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2018] [Revised: 01/03/2019] [Accepted: 01/03/2019] [Indexed: 12/23/2022] Open
Abstract
Oculopharyngeal muscular dystrophy (OPMD) is a rare late onset genetic disease leading to ptosis, dysphagia and proximal limb muscles at later stages. A short abnormal (GCN) triplet expansion in the polyA-binding protein nuclear 1 (PABPN1) gene leads to PABPN1-containing aggregates in the muscles of OPMD patients. Here we demonstrate that treating mice with guanabenz acetate (GA), an FDA-approved antihypertensive drug, reduces the size and number of nuclear aggregates, improves muscle force, protects myofibers from the pathology-derived turnover and decreases fibrosis. GA targets various cell processes, including the unfolded protein response (UPR), which acts to attenuate endoplasmic reticulum (ER) stress. We demonstrate that GA increases both the phosphorylation of the eukaryotic translation initiation factor 2α subunit and the splicing of Xbp1, key components of the UPR. Altogether these data show that modulation of protein folding regulation is beneficial for OPMD and promote the further development of GA or its derivatives for treatment of OPMD in humans. Furthermore, they support the recent evidences that treating ER stress could be therapeutically relevant in other more common proteinopathies.
Collapse
Affiliation(s)
- Alberto Malerba
- School of Biological Sciences, Centers of Gene and Cell Therapy and Biomedical Sciences, Royal Holloway, University of London, TW20 OEX Surrey, UK
| | - Fanny Roth
- Sorbonne Université, INSERM, Association Institut de Myologie, Centre de Recherche en Myologie, UMRS974, 47 bd de l'Hôpital, Paris, France
| | - Pradeep Harish
- School of Biological Sciences, Centers of Gene and Cell Therapy and Biomedical Sciences, Royal Holloway, University of London, TW20 OEX Surrey, UK
| | - Jamila Dhiab
- Sorbonne Université, INSERM, Association Institut de Myologie, Centre de Recherche en Myologie, UMRS974, 47 bd de l'Hôpital, Paris, France
| | - Ngoc Lu-Nguyen
- School of Biological Sciences, Centers of Gene and Cell Therapy and Biomedical Sciences, Royal Holloway, University of London, TW20 OEX Surrey, UK
| | - Ornella Cappellari
- Comparative Biomedical Sciences, The Royal Veterinary College, London, UK
| | - Susan Jarmin
- School of Biological Sciences, Centers of Gene and Cell Therapy and Biomedical Sciences, Royal Holloway, University of London, TW20 OEX Surrey, UK
| | - Alexandrine Mahoudeau
- Sorbonne Université, INSERM, Association Institut de Myologie, Centre de Recherche en Myologie, UMRS974, 47 bd de l'Hôpital, Paris, France
| | - Victor Ythier
- Sorbonne Université, INSERM, Association Institut de Myologie, Centre de Recherche en Myologie, UMRS974, 47 bd de l'Hôpital, Paris, France
| | - Jeanne Lainé
- Sorbonne Université, INSERM, Association Institut de Myologie, Centre de Recherche en Myologie, UMRS974, 47 bd de l'Hôpital, Paris, France
| | - Elisa Negroni
- Sorbonne Université, INSERM, Association Institut de Myologie, Centre de Recherche en Myologie, UMRS974, 47 bd de l'Hôpital, Paris, France
| | | | - Martine Simonelig
- Institute of Human Genetics, CNRS UMR9002-University of Montpellier, mRNA Regulation and Development, Montpellier, France
| | | | - Vincent Mouly
- Sorbonne Université, INSERM, Association Institut de Myologie, Centre de Recherche en Myologie, UMRS974, 47 bd de l'Hôpital, Paris, France
| | - Gillian Butler-Browne
- Sorbonne Université, INSERM, Association Institut de Myologie, Centre de Recherche en Myologie, UMRS974, 47 bd de l'Hôpital, Paris, France
| | - Cécile Voisset
- UMR1078 'Genetic, Functional Genomic and Biotechnologies', INSERM, EFS, Brest University, IBSAM, Brest, France
| | - George Dickson
- School of Biological Sciences, Centers of Gene and Cell Therapy and Biomedical Sciences, Royal Holloway, University of London, TW20 OEX Surrey, UK
| | - Capucine Trollet
- Sorbonne Université, INSERM, Association Institut de Myologie, Centre de Recherche en Myologie, UMRS974, 47 bd de l'Hôpital, Paris, France
| |
Collapse
|
39
|
Muraine L, Bensalah M, Dhiab J, Cordova G, Arandel L, Marhic A, Chapart M, Vasseur S, Benkhelifa-Ziyyat S, Bigot A, Butler-Browne G, Mouly V, Negroni E, Trollet C. Transduction Efficiency of Adeno-Associated Virus Serotypes After Local Injection in Mouse and Human Skeletal Muscle. Hum Gene Ther 2020; 31:233-240. [PMID: 31880951 DOI: 10.1089/hum.2019.173] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
The adeno-associated virus (AAV) vector is an efficient tool for gene delivery in skeletal muscle. AAV-based therapies show promising results for treatment of various genetic disorders, including muscular dystrophy. These dystrophies represent a heterogeneous group of diseases affecting muscles and typically characterized by progressive skeletal muscle wasting and weakness and the development of fibrosis. The tropism of each AAV serotype has been extensively studied using systemic delivery routes, but very few studies have compared their transduction efficiency through direct intramuscular injection. Yet, in some muscular dystrophies, where only a few muscles are primarily affected, a local intramuscular injection to target these muscles would be the most appropriate route. A comprehensive comparison between different recombinant AAV (rAAV) serotypes is therefore needed. In this study, we investigated the transduction efficiency of rAAV serotypes 1-10 by local injection in skeletal muscle of control C57BL/6 mice. We used a CMV-nls-LacZ reporter cassette allowing nuclear expression of LacZ to easily localize targeted cells. Detection of β-galactosidase activity on muscle cryosections demonstrated that rAAV serotypes 1, 7, 8, 9, and 10 were more efficient than the others, with rAAV9 being the most efficient in mice. Furthermore, using a model of human muscle xenograft in immunodeficient mice, we observed that in human muscle, rAAV8 and rAAV9 had similar transduction efficiency. These findings demonstrate for the first time that the human muscle xenograft can be used to evaluate AAV-based therapeutical approaches in a human context.
Collapse
Affiliation(s)
- Laura Muraine
- Sorbonne Université, Inserm, Institut de Myologie, U974, Centre de Recherche en Myologie, Paris, France
| | - Mona Bensalah
- Sorbonne Université, Inserm, Institut de Myologie, U974, Centre de Recherche en Myologie, Paris, France
| | - Jamila Dhiab
- Sorbonne Université, Inserm, Institut de Myologie, U974, Centre de Recherche en Myologie, Paris, France
| | - Gonzalo Cordova
- Sorbonne Université, Inserm, Institut de Myologie, U974, Centre de Recherche en Myologie, Paris, France
| | - Ludovic Arandel
- Sorbonne Université, Inserm, Institut de Myologie, U974, Centre de Recherche en Myologie, Paris, France
| | - Alix Marhic
- Sorbonne Université, Inserm, Institut de Myologie, U974, Centre de Recherche en Myologie, Paris, France
| | | | | | - Sofia Benkhelifa-Ziyyat
- Sorbonne Université, Inserm, Institut de Myologie, U974, Centre de Recherche en Myologie, Paris, France
| | - Anne Bigot
- Sorbonne Université, Inserm, Institut de Myologie, U974, Centre de Recherche en Myologie, Paris, France
| | - Gillian Butler-Browne
- Sorbonne Université, Inserm, Institut de Myologie, U974, Centre de Recherche en Myologie, Paris, France
| | - Vincent Mouly
- Sorbonne Université, Inserm, Institut de Myologie, U974, Centre de Recherche en Myologie, Paris, France
| | - Elisa Negroni
- Sorbonne Université, Inserm, Institut de Myologie, U974, Centre de Recherche en Myologie, Paris, France
| | - Capucine Trollet
- Sorbonne Université, Inserm, Institut de Myologie, U974, Centre de Recherche en Myologie, Paris, France
| |
Collapse
|
40
|
Galimberti V, Tironi R, Lerario A, Scali M, Del Bo R, Rodolico C, Brizzi T, Gibertini S, Maggi L, Mora M, Toscano A, Comi GP, Sciacco M, Moggio M, Peverelli L. Value of insoluble PABPN1 accumulation in the diagnosis of oculopharyngeal muscular dystrophy. Eur J Neurol 2019; 27:709-715. [DOI: 10.1111/ene.14131] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2019] [Accepted: 11/18/2019] [Indexed: 12/13/2022]
Affiliation(s)
- V. Galimberti
- Neuromuscular and Rare Disease Unit Department of Neuroscience Foundation IRCCS Ca’ Granda Ospedale Maggiore Policlinico Milano Italy
| | - R. Tironi
- Neuromuscular and Rare Disease Unit Department of Neuroscience Foundation IRCCS Ca’ Granda Ospedale Maggiore Policlinico Milano Italy
| | - A. Lerario
- Neuromuscular and Rare Disease Unit Department of Neuroscience Foundation IRCCS Ca’ Granda Ospedale Maggiore Policlinico Milano Italy
| | - M. Scali
- Neuromuscular and Rare Disease Unit Department of Neuroscience Foundation IRCCS Ca’ Granda Ospedale Maggiore Policlinico Milano Italy
| | - R. Del Bo
- Dino Ferrari Centre Neuroscience Section Department of Pathophysiology and Transplantation (DEPT) University of Milan MilanoItaly
- Neurology Unit IRCCS Foundation Ca’ Granda Ospedale Maggiore Policlinico Milano Italy
| | - C. Rodolico
- Neurology and Neuromuscular Diseases Unit Department of Clinical and Experimental Medicine University of Messina Messina Italy
| | - T. Brizzi
- Neurology and Neuromuscular Diseases Unit Department of Clinical and Experimental Medicine University of Messina Messina Italy
- DIBIMIS University of Palermo Palermo Italy
| | - S. Gibertini
- Neuromuscular Diseases and Neuroimmunology Unit Fondazione IRCCS Istituto Neurologico Carlo Besta Milano Italy
| | - L. Maggi
- Neuromuscular Diseases and Neuroimmunology Unit Fondazione IRCCS Istituto Neurologico Carlo Besta Milano Italy
| | - M. Mora
- Neuromuscular Diseases and Neuroimmunology Unit Fondazione IRCCS Istituto Neurologico Carlo Besta Milano Italy
| | - A. Toscano
- Neurology and Neuromuscular Diseases Unit Department of Clinical and Experimental Medicine University of Messina Messina Italy
| | - G. P. Comi
- Dino Ferrari Centre Neuroscience Section Department of Pathophysiology and Transplantation (DEPT) University of Milan MilanoItaly
- Neurology Unit IRCCS Foundation Ca’ Granda Ospedale Maggiore Policlinico Milano Italy
| | - M. Sciacco
- Neuromuscular and Rare Disease Unit Department of Neuroscience Foundation IRCCS Ca’ Granda Ospedale Maggiore Policlinico Milano Italy
| | - M. Moggio
- Neuromuscular and Rare Disease Unit Department of Neuroscience Foundation IRCCS Ca’ Granda Ospedale Maggiore Policlinico Milano Italy
| | - L. Peverelli
- Neuromuscular and Rare Disease Unit Department of Neuroscience Foundation IRCCS Ca’ Granda Ospedale Maggiore Policlinico Milano Italy
| |
Collapse
|
41
|
Dupont JB, Guo J, Renaud-Gabardos E, Poulard K, Latournerie V, Lawlor MW, Grange RW, Gray JT, Buj-Bello A, Childers MK, Mack DL. AAV-Mediated Gene Transfer Restores a Normal Muscle Transcriptome in a Canine Model of X-Linked Myotubular Myopathy. Mol Ther 2019; 28:382-393. [PMID: 31784415 DOI: 10.1016/j.ymthe.2019.10.018] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2019] [Revised: 09/13/2019] [Accepted: 10/31/2019] [Indexed: 12/27/2022] Open
Abstract
Multiple clinical trials employing recombinant adeno-associated viral (rAAV) vectors have been initiated for neuromuscular disorders, including Duchenne and limb-girdle muscular dystrophies, spinal muscular atrophy, and recently X-linked myotubular myopathy (XLMTM). Our previous work on a canine model of XLMTM showed that a single rAAV8-cMTM1 systemic infusion corrected structural abnormalities within the muscle and restored contractile function, with affected dogs surviving more than 4 years post injection. This remarkable therapeutic efficacy presents a unique opportunity to identify the downstream molecular drivers of XLMTM pathology and to what extent the whole muscle transcriptome is restored to normal after gene transfer. Herein, RNA-sequencing was used to examine the transcriptomes of the Biceps femoris and Vastus lateralis in a previously described canine cohort that showed dose-dependent clinical improvements after rAAV8-cMTM1 gene transfer. Our analysis confirmed several dysregulated genes previously observed in XLMTM mice but also identified transcripts linked to XLMTM pathology. We demonstrated XLMTM transcriptome remodeling and dose-dependent normalization of gene expression after gene transfer and created metrics to pinpoint potential biomarkers of disease progression and correction.
Collapse
Affiliation(s)
- Jean-Baptiste Dupont
- Department of Rehabilitation Medicine, Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA 98109, USA
| | - Jianjun Guo
- Audentes Therapeutics, San Francisco, CA 94108, USA
| | - Edith Renaud-Gabardos
- Genethon, INSERM UMR S951, Université Evry Val-d'Essone, Université Paris-Saclay, 91000 Evry, France
| | - Karine Poulard
- Genethon, INSERM UMR S951, Université Evry Val-d'Essone, Université Paris-Saclay, 91000 Evry, France
| | - Virginie Latournerie
- Genethon, INSERM UMR S951, Université Evry Val-d'Essone, Université Paris-Saclay, 91000 Evry, France
| | - Michael W Lawlor
- Department of Pathology and Laboratory Medicine and Neuroscience Research Center, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Robert W Grange
- Department of Human Nutrition, Foods, and Exercise, Virginia Polytechnic Institute and State University, Blacksburg, VA 24060, USA
| | - John T Gray
- Audentes Therapeutics, San Francisco, CA 94108, USA
| | - Ana Buj-Bello
- Genethon, INSERM UMR S951, Université Evry Val-d'Essone, Université Paris-Saclay, 91000 Evry, France
| | - Martin K Childers
- Department of Rehabilitation Medicine, Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA 98109, USA
| | - David L Mack
- Department of Rehabilitation Medicine, Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA 98109, USA; Department of Bioengineering, University of Washington, Seattle, WA 98109, USA.
| |
Collapse
|
42
|
|
43
|
Gois Beghini D, Iwao Horita S, Monteiro da Fonseca Cardoso L, Anastacio Alves L, Nagaraju K, Henriques-Pons A. A Promising Future for Stem-Cell-Based Therapies in Muscular Dystrophies-In Vitro and In Vivo Treatments to Boost Cellular Engraftment. Int J Mol Sci 2019; 20:ijms20215433. [PMID: 31683627 PMCID: PMC6861917 DOI: 10.3390/ijms20215433] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2019] [Revised: 08/28/2019] [Accepted: 09/23/2019] [Indexed: 02/06/2023] Open
Abstract
Muscular dystrophies (MD) are a group of genetic diseases that lead to skeletal muscle wasting and may affect many organs (multisystem). Unfortunately, no curative therapies are available at present for MD patients, and current treatments mainly address the symptoms. Thus, stem-cell-based therapies may present hope for improvement of life quality and expectancy. Different stem cell types lead to skeletal muscle regeneration and they have potential to be used for cellular therapies, although with several limitations. In this review, we propose a combination of genetic, biochemical, and cell culture treatments to correct pathogenic genetic alterations and to increase proliferation, dispersion, fusion, and differentiation into new or hybrid myotubes. These boosted stem cells can also be injected into pretreate recipient muscles to improve engraftment. We believe that this combination of treatments targeting the limitations of stem-cell-based therapies may result in safer and more efficient therapies for MD patients. Matricryptins have also discussed.
Collapse
Affiliation(s)
- Daniela Gois Beghini
- Laboratório de Inovações em Terapias, Ensino e Bioprodutos, Instituto Oswaldo Cruz, Fiocruz, Rio de Janeiro (RJ) 21040-900, Brazil.
| | - Samuel Iwao Horita
- Laboratório de Inovações em Terapias, Ensino e Bioprodutos, Instituto Oswaldo Cruz, Fiocruz, Rio de Janeiro (RJ) 21040-900, Brazil.
| | | | - Luiz Anastacio Alves
- Laboratório de Comunicação Celular, Instituto Oswaldo Cruz, Fiocruz, Rio de Janeiro (RJ) 21040-900, Brazil.
| | - Kanneboyina Nagaraju
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, Binghamton University, New York, NY 13902, USA.
| | - Andrea Henriques-Pons
- Laboratório de Inovações em Terapias, Ensino e Bioprodutos, Instituto Oswaldo Cruz, Fiocruz, Rio de Janeiro (RJ) 21040-900, Brazil.
| |
Collapse
|
44
|
Harish P, Malerba A, Lu-Nguyen N, Forrest L, Cappellari O, Roth F, Trollet C, Popplewell L, Dickson G. Inhibition of myostatin improves muscle atrophy in oculopharyngeal muscular dystrophy (OPMD). J Cachexia Sarcopenia Muscle 2019; 10:1016-1026. [PMID: 31066242 PMCID: PMC6818462 DOI: 10.1002/jcsm.12438] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Accepted: 03/21/2019] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Oculopharyngeal muscular dystrophy (OPMD) is a late-onset muscle disease affecting one per 80 000 of the general population characterized by profound dysphagia and ptosis, and limb weakness at later stages. Affected muscles are characterized by increased fibrosis and atrophy. Myostatin is a negative regulator of muscle mass, and inhibition of myostatin has been demonstrated to ameliorate symptoms in dystrophic muscles. METHODS In this study, we performed a systemic delivery of a monoclonal antibody to immunologically block myostatin in the A17 mouse model of OPMD. The mice were administered a weekly dose of 10 mg/kg RK35 intraperitonially for 10 weeks, following which histological analyses were performed on the samples. RESULTS This treatment significantly (P < 0.01) improved body mass (11%) and muscle mass (for the tibialis anterior and extensor digitorum longus by 19% and 41%) in the A17 mice treated with RK35 when compared to saline controls. Similarly, a significantly (P < 0.01) increased muscle strength (18% increase in maximal tetanic force) and myofibre diameter (17% and 44% for the tibialis anterior and extensor digitorum longus), and reduced expression of markers of muscle fibrosis (40% reduction in area of expression), was also observed. No change in the density of intranuclear inclusions (a hallmark of disease progression of OPMD) was however observed. CONCLUSIONS Our study supports the clinical translation of such antibody-mediated inhibition of myostatin as a treatment of OPMD. This strategy has implications to be used as adjuvant therapies with gene therapy based approaches, or to stabilize the muscle prior to myoblast transplantation.
Collapse
Affiliation(s)
- Pradeep Harish
- Centres of Gene and Cell Therapy and Biomedical Sciences, School of Biological Sciences, Royal Holloway-University of London, Surrey, UK
| | - Alberto Malerba
- Centres of Gene and Cell Therapy and Biomedical Sciences, School of Biological Sciences, Royal Holloway-University of London, Surrey, UK
| | - Ngoc Lu-Nguyen
- Centres of Gene and Cell Therapy and Biomedical Sciences, School of Biological Sciences, Royal Holloway-University of London, Surrey, UK
| | - Leysa Forrest
- Centres of Gene and Cell Therapy and Biomedical Sciences, School of Biological Sciences, Royal Holloway-University of London, Surrey, UK
| | | | - Fanny Roth
- Association Institut de Myologie, Centre de Recherche en Myologie UMRS974, Sorbonne Université, INSERM, Paris, France
| | - Capucine Trollet
- Association Institut de Myologie, Centre de Recherche en Myologie UMRS974, Sorbonne Université, INSERM, Paris, France
| | - Linda Popplewell
- Centres of Gene and Cell Therapy and Biomedical Sciences, School of Biological Sciences, Royal Holloway-University of London, Surrey, UK
| | - George Dickson
- Centres of Gene and Cell Therapy and Biomedical Sciences, School of Biological Sciences, Royal Holloway-University of London, Surrey, UK
| |
Collapse
|
45
|
RNA-Based Therapy Utilizing Oculopharyngeal Muscular Dystrophy Transcript Knockdown and Replacement. MOLECULAR THERAPY. NUCLEIC ACIDS 2019; 15:12-25. [PMID: 30831428 PMCID: PMC6403420 DOI: 10.1016/j.omtn.2019.02.003] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/02/2018] [Revised: 02/08/2019] [Accepted: 02/10/2019] [Indexed: 11/23/2022]
Abstract
Oculopharyngeal muscular dystrophy (OPMD) is caused by a small expansion of a short polyalanine (polyAla) tract in the poly(A)-binding protein nuclear 1 protein (PABPN1). Despite the monogenic nature of OPMD, no treatment is currently available. Here we report an RNA replacement strategy that has therapeutic potential in cell and C. elegans OPMD models. We develop selective microRNAs (miRNAs) against PABPN1, and we report that miRNAs and our previously developed hammerhead ribozymes (hhRzs) are capable of reducing the expression of both the mRNA and protein levels of PABPN1 by as much as 90%. Since OPMD derives from a very small expansion of GCG within the polyAla tract, our hhRz and miRNA molecules cannot distinguish between the wild-type and mutant mRNAs of PABPN1. Therefore, we designed an optimized-codon wild-type PABPN1 (opt-PABPN1) that is resistant to cleavage by hhRzs and miRNAs. Co-expression of opt-PABPN1 with either our hhRzs or miRNAs restored the level of PABPN1, concomitantly with a reduction in expanded PABPN1-associated cell death in a stable C2C12 OPMD model. Interestingly, knockdown of the PABPN1 by selective hhRzs in the C. elegans OPMD model significantly improved the motility of the PABPN1-13Ala worms. Taken together, RNA replacement therapy represents an exciting approach for OPMD treatment.
Collapse
|
46
|
Harish P, Dickson G, Malerba A. Advances in emerging therapeutics for oculopharyngeal muscular dystrophy. Expert Opin Orphan Drugs 2018. [DOI: 10.1080/21678707.2018.1536542] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
Affiliation(s)
- Pradeep Harish
- School of Biological Sciences, Centres of Gene and Cell therapy and Biomedical sciences, Royal Holloway University of London, Egham, Surrey, UK
| | - George Dickson
- School of Biological Sciences, Centres of Gene and Cell therapy and Biomedical sciences, Royal Holloway University of London, Egham, Surrey, UK
| | - Alberto Malerba
- School of Biological Sciences, Centres of Gene and Cell therapy and Biomedical sciences, Royal Holloway University of London, Egham, Surrey, UK
| |
Collapse
|
47
|
Chen AWG, Wu SL, Cheng WL, Chuang CS, Chen CH, Chen MK, Liu CS. Dysphagia with fatal choking in oculopharyngeal muscular dystrophy: Case report. Medicine (Baltimore) 2018; 97:e12935. [PMID: 30412104 PMCID: PMC6221716 DOI: 10.1097/md.0000000000012935] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
RATIONALE Oculopharyngeal muscular dystrophy (OPMD) is an autosomal dominant late-onset progressive muscle disorder typically characterized by ptosis, difficulty in swallowing, and proximal limb weakness. Underdiagnosis of OPMD is common in Asian countries and results in delayed diagnoses and fatal events. PATIENT CONCERNS Here, we report the case of a 53-year-old female who suffered from progressive dysphagia and experienced several choking events involving solid material. An extensive family history of dysphagia was noted, and 2 family members had died as a result of aspiration. DIAGNOSES PABPN1 genotyping and DNA sequence analysis revealed a heterozygous (GCG)10(GCA)3GCG mutation that led to the diagnosis of OPMD. INTERVENTIONS Rehabilitation exercises, namely, the Shaker exercise and the Masako maneuver, were suggested. OUTCOMES Improved swallowing ability with safe food intake was noted after 2 months of training. Surgical intervention will be considered when progression of the disease is noted. LESSONS Underdiagnosis and a lack of awareness of OPMD may lead to choking, aspiration pneumonia, and death in multiple members of affected families. Currently, there is no definitive treatment for OPMD, but rehabilitation exercises and surgical intervention are helpful in relieving dysphagia.
Collapse
Affiliation(s)
| | | | | | | | - Chih-Hua Chen
- Department of Otorhinolaryngology, Head and Neck Surgery
| | - Mu-Kuan Chen
- Department of Otorhinolaryngology, Head and Neck Surgery
| | - Chin-San Liu
- Department of Neurology
- Vascular and Genomics Center
- Institution of ATP, International Medical Industry Alliance, Changhua Christian Hospital, Changhua
- Graduate Institute of Integrative Medicine, China Medical University, Taichung, Taiwan
| |
Collapse
|
48
|
Pavone P, Cho SY, Praticò A, Falsaperla R, Ruggieri M, Jin DK. Ptosis in childhood: A clinical sign of several disorders: Case series reports and literature review. Medicine (Baltimore) 2018; 97:e12124. [PMID: 30200099 PMCID: PMC6133583 DOI: 10.1097/md.0000000000012124] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Blepharoptosis (ptosis) is a common but often overlooked sign that may serve as a sign/manifestation of other conditions, ranging from a mild and purely cosmetic presentation to a severe and occasionally progressive disorder. Ptosis may show an acute onset or may manifest as a chronic disorder. Its presentation may vary: unilateral versus bilateral, progressive versus non-progressive, isolated versus complex which occurs in association with other symptoms, and congenital versus acquired (often concomitant with neuromuscular disorders).Congenital ptosis includes the isolated type-the congenital cranial dysinnervation disorders, which are further, distinguished into different subtypes such as Horner syndrome (HS), and ptosis as a sign/manifestation of various congenital malformation syndromes.In this article, we review the primary causes of ptosis occurring in childhood, and its various clinical presentations, including a short report on selected cases observed in our institution: a classical isolated familial ptosis comprising 14 members over 5 generations, 3 sibling with isolated congenital ptosis who in addition suffered by episodes of febrile seizures, a patient with Duane retraction syndrome who presented congenital skin and hair anomalies, and a girl with HS who showed a history of congenital imperforate hymen. A flowchart outlining the congenital and acquired type of ptosis and the clinical approach to the management and treatment of children with this anomaly is reported.
Collapse
Affiliation(s)
- P. Pavone
- University-Hospital Policlinico-Vittorio Emanuele
| | - Sung Yoon Cho
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - A.D. Praticò
- Department of Clinical and Experimental Medicine, Section of Pediatrics and Child Neuropsychiatry, University of Catania, Italy
| | | | - M. Ruggieri
- Department of Clinical and Experimental Medicine, Section of Pediatrics and Child Neuropsychiatry, University of Catania, Italy
| | - Dong-Kyu Jin
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| |
Collapse
|
49
|
Aguti S, Malerba A, Zhou H. The progress of AAV-mediated gene therapy in neuromuscular disorders. Expert Opin Biol Ther 2018; 18:681-693. [DOI: 10.1080/14712598.2018.1479739] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Affiliation(s)
- Sara Aguti
- The Dubowitz Neuromuscular Centre, Developmental Neurosciences Programme, Great Ormond Street Institute of Child Health, University College London, London, UK
| | - Alberto Malerba
- School of Biological Sciences, Royal Holloway, University of London, Egham, Surrey, UK
| | - Haiyan Zhou
- The Dubowitz Neuromuscular Centre, Developmental Neurosciences Programme, Great Ormond Street Institute of Child Health, University College London, London, UK
- Genetics and Genomic Medicine Programme, Great Ormond Street Institute of Child Health, University College London, London, UK
- NIHR Great Ormond Street Hospital Biomedical Research Centre, London, UK
| |
Collapse
|
50
|
Pre-clinical Safety and Off-Target Studies to Support Translation of AAV-Mediated RNAi Therapy for FSHD. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2017; 8:121-130. [PMID: 29387734 PMCID: PMC5787672 DOI: 10.1016/j.omtm.2017.12.005] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/07/2017] [Accepted: 12/19/2017] [Indexed: 11/22/2022]
Abstract
RNAi emerged as a prospective molecular therapy nearly 15 years ago. Since then, two major RNAi platforms have been under development: oligonucleotides and gene therapy. Oligonucleotide-based approaches have seen more advancement, with some promising therapies that may soon reach market. In contrast, vector-based approaches for RNAi therapy have remained largely in the pre-clinical realm, with limited clinical safety and efficacy data to date. We are developing a gene therapy approach to treat the autosomal-dominant disorder facioscapulohumeral muscular dystrophy. Our strategy involves silencing the myotoxic gene DUX4 using adeno-associated viral vectors to deliver targeted microRNA expression cassettes (miDUX4s). We previously demonstrated proof of concept for this approach in mice, and we are now taking additional steps here to assess safety issues related to miDUX4 overexpression and sequence-specific off-target silencing. In this study, we describe improvements in vector design and expansion of our miDUX4 sequence repertoire and report differential toxicity elicited by two miDUX4 sequences, of which one was toxic and the other was not. This study provides important data to help advance our goal of translating RNAi gene therapy for facioscapulohumeral muscular dystrophy.
Collapse
|