1
|
Foncuberta ME, Monges S, Medina A, Lubieniecki F, Gravina LP. A novel deep intronic variant in the DMD gene causes Duchenne muscular dystrophy by pseudoexon activation encoding a nonsense codon. Gene 2024; 930:148862. [PMID: 39151676 DOI: 10.1016/j.gene.2024.148862] [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: 05/13/2024] [Revised: 07/30/2024] [Accepted: 08/13/2024] [Indexed: 08/19/2024]
Abstract
Dystrophinopathies are a group of neuromuscular disorders, inherited in an X-linked recessive manner, caused by pathogenic variants in the DMD gene. Copy number variation detection and next generation sequencing allow the detection of around 99 % of the pathogenic variants. However, some patients require mRNA studies from muscle biopsies to identify deep intronic pathogenic variants. Here, we report a child suspected of having Duchenne muscular dystrophy, with a muscle biopsy showing dystrophin deficiency, and negative molecular testing for deletions, duplications, and small variants. mRNA analysis from muscle biopsy revealed a pseudoexon activation that introduce a premature stop codon into the reading frame. gDNA sequencing allowed to identified a novel variant, c.832-186 T>G, which creates a cryptic donor splice site, recognizing the underlying mechanism causing the pseudoexon insertion. This case highlights the usefulness of the mRNA analysis from muscle biopsy when routine genetic testing is negative and clinical suspicion of dystrophinopathies remains the main clinical diagnosis suspicion.
Collapse
Affiliation(s)
- María Eugenia Foncuberta
- Laboratorio de Biología Molecular - Genética, Hospital de Pediatría Garrahan, Buenos Aires, Argentina.
| | - Soledad Monges
- Servicio de Neurología, Hospital de Pediatría Garrahan, Buenos Aires, Argentina
| | - Adriana Medina
- Laboratorio Biología Molecular - Hematogía y Oncología, Hospital de Pediatría Garrahan, Buenos Aires, Argentina
| | - Fabiana Lubieniecki
- Servicio de Patología, Hospital de Pediatría Garrahan, Buenos Aires, Argentina
| | - Luis Pablo Gravina
- Laboratorio de Biología Molecular - Genética, Hospital de Pediatría Garrahan, Buenos Aires, Argentina
| |
Collapse
|
2
|
Forte G, Grossi V, Cariola F, Buonadonna AL, Sanese P, De Marco K, Fasano C, Lepore Signorile M, Disciglio V, Simone C. Identification of a novel germline APC N-terminal pathogenic variant associated with attenuated familial adenomatous polyposis. Genes Dis 2024; 11:101078. [PMID: 39104424 PMCID: PMC11298831 DOI: 10.1016/j.gendis.2023.101078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Revised: 07/18/2023] [Accepted: 07/21/2023] [Indexed: 08/07/2024] Open
Affiliation(s)
- Giovanna Forte
- Medical Genetics, National Institute of Gastroenterology IRCCS “S. de Bellis” Research Hospital, Castellana Grotte, Bari 70013, Italy
| | - Valentina Grossi
- Medical Genetics, National Institute of Gastroenterology IRCCS “S. de Bellis” Research Hospital, Castellana Grotte, Bari 70013, Italy
| | - Filomena Cariola
- Medical Genetics, National Institute of Gastroenterology IRCCS “S. de Bellis” Research Hospital, Castellana Grotte, Bari 70013, Italy
| | - Antonia Lucia Buonadonna
- Medical Genetics, National Institute of Gastroenterology IRCCS “S. de Bellis” Research Hospital, Castellana Grotte, Bari 70013, Italy
| | - Paola Sanese
- Medical Genetics, National Institute of Gastroenterology IRCCS “S. de Bellis” Research Hospital, Castellana Grotte, Bari 70013, Italy
| | - Katia De Marco
- Medical Genetics, National Institute of Gastroenterology IRCCS “S. de Bellis” Research Hospital, Castellana Grotte, Bari 70013, Italy
| | - Candida Fasano
- Medical Genetics, National Institute of Gastroenterology IRCCS “S. de Bellis” Research Hospital, Castellana Grotte, Bari 70013, Italy
| | - Martina Lepore Signorile
- Medical Genetics, National Institute of Gastroenterology IRCCS “S. de Bellis” Research Hospital, Castellana Grotte, Bari 70013, Italy
| | - Vittoria Disciglio
- Medical Genetics, National Institute of Gastroenterology IRCCS “S. de Bellis” Research Hospital, Castellana Grotte, Bari 70013, Italy
| | - Cristiano Simone
- Medical Genetics, National Institute of Gastroenterology IRCCS “S. de Bellis” Research Hospital, Castellana Grotte, Bari 70013, Italy
- Medical Genetics, Department of Precision and Regenerative Medicine and Jonic Area (DiMePRe-J), University of Bari Aldo Moro, Bari 70124, Italy
| |
Collapse
|
3
|
Wei Y, Jiang Y, Lu Y, Hu Q. Histone modifications in Duchenne muscular dystrophy: pathogenesis insights and therapeutic implications. J Med Genet 2024; 61:1003-1010. [PMID: 39327039 DOI: 10.1136/jmg-2024-110045] [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: 04/11/2024] [Accepted: 09/18/2024] [Indexed: 09/28/2024]
Abstract
Duchenne muscular dystrophy (DMD) is a commonly encountered genetic ailment marked by loss-of-function mutations in the Dystrophin gene, ultimately resulting in progressive debilitation of skeletal muscle. The investigation into the pathogenesis of DMD has increasingly converged on the role of histone modifications within the broader context of epigenetic regulation. These modifications, including histone acetylation, methylation and phosphorylation, are catalysed by specific enzymes and play a critical role in gene expression. This article provides an overview of the histone modifications occurring in DMD and analyses the research progress and potential of different types of histone modifications in DMD due to changes in cellular signalling for muscle regeneration, to provide new insights into diagnostic and therapeutic options for DMD.
Collapse
Affiliation(s)
- Yanning Wei
- Department of Cell Biology and Genetics, School of Basic Medical Sciences, Guangxi Medical University, Nanning, Guangxi, China
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Key Laboratory of Biological Molecular Medicine Research of Education, Guangxi Medical University, Nanning, Guangxi, China
| | - Yuanyuan Jiang
- Department of Cell Biology and Genetics, School of Basic Medical Sciences, Guangxi Medical University, Nanning, Guangxi, China
| | - Yufei Lu
- Department of Cell Biology and Genetics, School of Basic Medical Sciences, Guangxi Medical University, Nanning, Guangxi, China
| | - Qiping Hu
- Department of Cell Biology and Genetics, School of Basic Medical Sciences, Guangxi Medical University, Nanning, Guangxi, China
- Key Laboratory of Longevity and Aging-related Diseases, Ministry of Education, Guangxi Medical University, Nanning, Guangxi, China
| |
Collapse
|
4
|
Doisy M, Vacca O, Saoudi A, Goyenvalle A. Levels of Exon-Skipping Are Not Artificially Overestimated Because of the Increased Affinity of Tricyclo-DNA-Modified Antisense Oligonucleotides to the Target DMD Exon. Nucleic Acid Ther 2024; 34:214-220. [PMID: 39046946 DOI: 10.1089/nat.2024.0002] [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: 07/27/2024] Open
Abstract
Antisense oligonucleotides (ASO) are very promising drugs for numerous diseases including neuromuscular disorders such as Duchenne muscular dystrophy (DMD). Several ASO drugs have already been approved by the US Food and Drug Administration for DMD and global efforts are still ongoing to improve further their potency, notably by developing new delivery systems or alternative chemistries. In this context, a recent study investigated the potential of different chemically modified ASO to induce exon-skipping in mouse models of DMD. Importantly, the authors reported a strong discrepancy between exon-skipping and protein restoration levels, which was mainly owing to the high affinity of locked nucleic acid (LNA) modifications to the target RNA, thereby interfering with the amplification of the unskipped product and resulting in artificial overamplification of the exon-skipped product. These findings urged us to verify whether a similar phenomenon could occur with tricyclo-DNA (tcDNA)-ASO that also display high-affinity properties to the target RNA. We thus ran a series of control experiments and demonstrate here that exon-skipping levels are not overestimated owing to an interference of tcDNA-ASO with the unskipped product in contrast to what was observed with LNA-containing ASO.
Collapse
Affiliation(s)
- Mathilde Doisy
- UVSQ, Inserm, END-ICAP, Université Paris-Saclay, Versailles, France
| | - Ophélie Vacca
- UVSQ, Inserm, END-ICAP, Université Paris-Saclay, Versailles, France
| | - Amel Saoudi
- UVSQ, Inserm, END-ICAP, Université Paris-Saclay, Versailles, France
| | | |
Collapse
|
5
|
Van Poucke M, Ledeganck L, Guo LT, Shelton GD, Bhatti SFM, Cornelis I, Peelman L. Exonisation of an intronic L1 element in the dystrophin gene associated with X-linked muscular dystrophy in a Border Collie dog. Anim Genet 2024; 55:733-743. [PMID: 39152696 DOI: 10.1111/age.13470] [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: 06/26/2024] [Accepted: 07/31/2024] [Indexed: 08/19/2024]
Abstract
X-linked recessive dystrophinopathies are the most common muscular dystrophies (MDs) in humans and dogs. To date, 20 breed-specific MD-associated variants are described in the canine dystrophin gene (DMD), including one associated with dystrophin-deficient MD in the Border Collie mixed breed. Here, we report the diagnosis and follow-up of mild dystrophin-deficient MD in a 5-month-old male Border Collie, associated with a novel DMD variant. Diagnosis was based on neurological examination and laboratory evaluations including creatine kinase activity, electromyography and muscle biopsies with immunofluorescent staining. Inspection of the Sashimi plots of the RNA-seq data from the affected muscle biopsy led to the discovery of a 162-bp L1 pseudoexon in DMD intron 63, introducing a frameshift and a premature stop codon (NM_001003343.1: c.9271_9272insN[162] p.(Ala3091fs*21)). Reduced DMD mRNA levels were detected for both the non-pseudoexon (50× less) and pseudoexon (3× less) containing transcripts in the affected muscle, compared with the level of the non-pseudoexon containing transcript in a control muscle, resulting in very low dystrophin protein levels and the upregulation of utrophin. Because the variant was only found in the affected dog, not in the healthy mother and grandmother, or in 108 unrelated Border Collies from the Belgian population (46 males and 62 females), it was considered a de novo variant. Although the prognosis for dystrophinopathy is generally regarded as poor, the dog stabilised at the age of 6 months and is still clinically stable at the age of 2 years.
Collapse
Affiliation(s)
- Mario Van Poucke
- Department of Veterinary and Biosciences, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| | - Liesbet Ledeganck
- Department of Small Animals, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| | - Ling T Guo
- Department of Pathology, School of Medicine, University of California San Diego, La Jolla, California, USA
| | - G Diane Shelton
- Department of Pathology, School of Medicine, University of California San Diego, La Jolla, California, USA
| | - Sofie F M Bhatti
- Department of Small Animals, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| | - Ine Cornelis
- Department of Small Animals, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| | - Luc Peelman
- Department of Veterinary and Biosciences, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| |
Collapse
|
6
|
Blitek M, Phongsavanh X, Goyenvalle A. The bench to bedside journey of tricyclo-DNA antisense oligonucleotides for the treatment of Duchenne muscular dystrophy. RSC Med Chem 2024; 15:3017-3025. [PMID: 39309360 PMCID: PMC11411614 DOI: 10.1039/d4md00394b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Accepted: 07/18/2024] [Indexed: 09/25/2024] Open
Abstract
The development of antisense oligonucleotide (ASO)-based therapeutics has made tremendous progress over the past few years, in particular for the treatment of neuromuscular disorders such as Duchenne muscular dystrophy and spinal muscular atrophy. Several ASO drugs have now reached market approval for these diseases and many more are currently under clinical evaluation. Among them, ASOs made of the tricyclo-DNA originally developed by Christian Leumann have shown particularly interesting properties and demonstrated promise for the treatment of Duchenne muscular dystrophy. In this review, we examine the bench to bedside journey of tricyclo-DNA-ASOs from their early preclinical evaluation as fully phosphorotiated-ASOs to the latest generation of lipid-conjugated-ASOs. Finally we discuss the remaining challenges of ASO-mediated exon-skipping therapy for DMD and future perspectives for this promising chemistry of ASOs.
Collapse
Affiliation(s)
- Mathilde Blitek
- UVSQ, Inserm, END-ICAP, Université Paris-Saclay 78000 Versailles France +33 170429432
| | | | - Aurélie Goyenvalle
- UVSQ, Inserm, END-ICAP, Université Paris-Saclay 78000 Versailles France +33 170429432
- LIA BAHN, CSM-UVSQ Monaco Principality of Monaco
| |
Collapse
|
7
|
Lemoine J, Dubois A, Dorval A, Jaber A, Warthi G, Mamchaoui K, Wang T, Corre G, Bovolenta M, Richard I. Correction of exon 2, exon 2-9 and exons 8-9 duplications in DMD patient myogenic cells by a single CRISPR/Cas9 system. Sci Rep 2024; 14:21238. [PMID: 39261505 PMCID: PMC11390959 DOI: 10.1038/s41598-024-70075-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Accepted: 08/12/2024] [Indexed: 09/13/2024] Open
Abstract
Duchenne Muscular dystrophy (DMD), a yet-incurable X-linked recessive disorder that results in muscle wasting and loss of ambulation is due to mutations in the dystrophin gene. Exonic duplications of dystrophin gene are a common type of mutations found in DMD patients. In this study, we utilized a single guide RNA CRISPR strategy targeting intronic regions to delete the extra duplicated regions in patient myogenic cells carrying duplication of exon 2, exons 2-9, and exons 8-9 in the DMD gene. Immunostaining on CRISPR-corrected derived myotubes demonstrated the rescue of dystrophin protein. Subsequent RNA sequencing of the DMD cells indicated rescue of genes of dystrophin related pathways. Examination of predicted close-match off-targets evidenced no aberrant gene editing at these loci. Here, we further demonstrate the efficiency of a single guide CRISPR strategy capable of deleting multi-exon duplications in the DMD gene without significant off target effect. Our study contributes valuable insights into the safety and efficacy of using single guide CRISPR strategy as a potential therapeutic approach for DMD patients with duplications of variable size.
Collapse
Affiliation(s)
- Juliette Lemoine
- Genethon, 1, bis rue de l'internationale, 91000, Evry, France
- Université Paris-Saclay, Univ Evry, Inserm, Généthon, Integrare Research Unit UMR_S951, 91000, Evry-Courcouronnes, France
| | - Auriane Dubois
- Genethon, 1, bis rue de l'internationale, 91000, Evry, France
- Université Paris-Saclay, Univ Evry, Inserm, Généthon, Integrare Research Unit UMR_S951, 91000, Evry-Courcouronnes, France
| | - Alan Dorval
- Genethon, 1, bis rue de l'internationale, 91000, Evry, France
- Université Paris-Saclay, Univ Evry, Inserm, Généthon, Integrare Research Unit UMR_S951, 91000, Evry-Courcouronnes, France
- ADLIN Science, Pépinière « Genopole Entreprises », 91058, Evry, France
| | - Abbass Jaber
- Genethon, 1, bis rue de l'internationale, 91000, Evry, France
- Université Paris-Saclay, Univ Evry, Inserm, Généthon, Integrare Research Unit UMR_S951, 91000, Evry-Courcouronnes, France
| | - Ganesh Warthi
- Genethon, 1, bis rue de l'internationale, 91000, Evry, France
- Université Paris-Saclay, Univ Evry, Inserm, Généthon, Integrare Research Unit UMR_S951, 91000, Evry-Courcouronnes, France
| | - Kamel Mamchaoui
- Sorbonne Université, Inserm, Institut de Myologie, Centre de Recherche en Myologie, 75013, Paris, France
| | - Tao Wang
- Genethon, 1, bis rue de l'internationale, 91000, Evry, France
- Université Paris-Saclay, Univ Evry, Inserm, Généthon, Integrare Research Unit UMR_S951, 91000, Evry-Courcouronnes, France
| | - Guillaume Corre
- Genethon, 1, bis rue de l'internationale, 91000, Evry, France
- Université Paris-Saclay, Univ Evry, Inserm, Généthon, Integrare Research Unit UMR_S951, 91000, Evry-Courcouronnes, France
| | - Matteo Bovolenta
- Genethon, 1, bis rue de l'internationale, 91000, Evry, France
- Université Paris-Saclay, Univ Evry, Inserm, Généthon, Integrare Research Unit UMR_S951, 91000, Evry-Courcouronnes, France
- Department of Translational Medicine, University of Ferrara, 44121, Ferrara, Italy
| | - Isabelle Richard
- Genethon, 1, bis rue de l'internationale, 91000, Evry, France.
- Université Paris-Saclay, Univ Evry, Inserm, Généthon, Integrare Research Unit UMR_S951, 91000, Evry-Courcouronnes, France.
| |
Collapse
|
8
|
Salame M, Bonnet C, Singh-Estivalet A, Brahim SM, Roux S, Boussaty EC, Hadrami M, Hamed CT, Sidi AM, Veten F, Petit C, Houmeida A. Splice-altering variant of PJVK gene in a Mauritanian family with non-syndromic hearing impairment. J Appl Genet 2024:10.1007/s13353-024-00903-x. [PMID: 39230647 DOI: 10.1007/s13353-024-00903-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Revised: 08/15/2024] [Accepted: 08/16/2024] [Indexed: 09/05/2024]
Abstract
PJVK gene was recently shown to create hypervulnerability to sound in humans and was the first human gene implicated in non-syndromic hearing impairment due to neural defect. Targeted next-generation sequencing of over 150 known deafness genes was performed in the proband. Sanger sequencing was used to validate the PJVK variant and confirm familial segregation of the disease. A minigene-based assay has been performed to assess the impact of the variant on splicing. We identified a novel c.550-6A > G acceptor splice-site variant in the PJVK gene in the homozygous state in a Mauritanian child with severe to profound congenital deafness. The substitution was located in intron 4. The effect of the variation was demonstrated by a minigene assay which showed that the variation, an insertion of an additional 5 bp, created a new splice site resulting in the appearance of a premature stop codon (p.Phe184Tyrfs*26) and likely a truncated protein. This result constitutes a new splice-site variant report in the PJVK gene leading to DFNB59 type associated with autosomal recessive non-syndromic hearing impairment (ARNSHI).
Collapse
Affiliation(s)
- Malak Salame
- Unité de Recherche Sur Les Biomarqueurs Dans La Population Mauritanienne, UN-FST, Nouakchott, Mauritania.
- Institut d'Hépato-Virologie, Nouakchott, Mauritania.
| | - Crystel Bonnet
- Institut de L'Audition, Institut Pasteur, Inserm, Paris, France
| | | | - Selma Mohamed Brahim
- Unité de Recherche Sur Les Biomarqueurs Dans La Population Mauritanienne, UN-FST, Nouakchott, Mauritania
- Centre National d'Oncologie (CNO), Unité de Recherche Et d'Enseignement, Nouakchott, Mauritania
| | - Solene Roux
- Institut de L'Audition, Institut Pasteur, Inserm, Paris, France
| | - Ely Cheikh Boussaty
- Division of Otolaryngology, Department of Surgery, University of California, 9500 Gilman Drive, Mail Code 0666, La Jolla, San Diego, CA, 92093, USA
| | - Mouna Hadrami
- Unité de Recherche Sur Les Biomarqueurs Dans La Population Mauritanienne, UN-FST, Nouakchott, Mauritania
| | | | - Abdellahi M'hamed Sidi
- Unité de Recherche Sur Les Biomarqueurs Dans La Population Mauritanienne, UN-FST, Nouakchott, Mauritania
| | - Fatimetou Veten
- Unité de Recherche Sur Les Biomarqueurs Dans La Population Mauritanienne, UN-FST, Nouakchott, Mauritania
| | - Christine Petit
- Institut de L'Audition, Institut Pasteur, Inserm, Paris, France
- Collège de France, Paris, France
| | - Ahmed Houmeida
- Unité de Recherche Sur Les Biomarqueurs Dans La Population Mauritanienne, UN-FST, Nouakchott, Mauritania
| |
Collapse
|
9
|
Jover I, Ramos MC, Escámez MJ, Lozoya E, Tormo JR, de Prado-Verdún D, Mencía Á, Pont M, Puig C, Larraufie MH, Gutiérrez-Caballero C, Reyes F, Trincado JL, García-González V, Cerrato R, Andrés M, Crespo M, Vicente F, Godessart N, Genilloud O, Larcher F, Nueda A. Identification of novel small molecule-based strategies of COL7A1 upregulation and readthrough activity for the treatment of recessive dystrophic epidermolysis bullosa. Sci Rep 2024; 14:18969. [PMID: 39152155 PMCID: PMC11329504 DOI: 10.1038/s41598-024-67398-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Accepted: 07/10/2024] [Indexed: 08/19/2024] Open
Abstract
Recessive dystrophic epidermolysis bullosa (RDEB) is a rare genetic disease caused by loss of function mutations in the gene coding for collagen VII (C7) due to deficient or absent C7 expression. This disrupts structural and functional skin architecture, leading to blistering, chronic wounds, inflammation, important systemic symptoms affecting the mouth, gastrointestinal tract, cornea, and kidney function, and an increased skin cancer risk. RDEB patients have an extremely poor quality of life and often die at an early age. A frequent class of mutations in RDEB is premature termination codons (PTC), which appear in homozygosity or compound heterozygosity with other mutations. RDEB has no cure and current therapies are mostly palliative. Using patient-derived keratinocytes and a library of 8273 small molecules and 20,160 microbial extracts evaluated in a phenotypic screening interrogating C7 levels, we identified three active chemical series. Two of these series had PTC readthrough activity, and one upregulated C7 mRNA, showing synergistic activity when combined with the reference readthrough molecule gentamicin. These compounds represent novel potential small molecule-based systemic strategies that could complement topical-based treatments for RDEB.
Collapse
Affiliation(s)
- Irene Jover
- R&D Centre, Almirall S.A., Laureà Miró 408-410, 08980, Sant Feliu de Llobregat, Barcelona, Spain
| | - Maria C Ramos
- Fundación MEDINA, Parque Tecnológico de La Salud, Av. Conocimiento 34, 18016, Granada, Spain
| | - María José Escámez
- Departamento de Bioingeniería E Ingeniería Aeroespacial (UC3M), División de Biomedicina Epitelial, Centro de Investigaciones Energéticas, Medioambientales y Tecnológicas (CIEMAT), Centro de Investigación Biomédica en Red de Enfermedades Raras, Universidad Carlos III de Madrid (UC3M), Madrid, Spain
- Unidad de Innovación Biomédica. Centro de Investigaciones Energéticas, U714-CIBER de Enfermedades Raras (CIBERER-ISCIII), Madrid, Spain
- Instituto de Investigación Sanitaria, Fundación Jiménez Díaz (IISFJD), Madrid, Spain
- Centro de Investigaciones Energéticas, Medioambientales y Tecnológicas (CIEMAT), Madrid, Spain
| | - Estrella Lozoya
- R&D Centre, Almirall S.A., Laureà Miró 408-410, 08980, Sant Feliu de Llobregat, Barcelona, Spain
| | - José R Tormo
- Fundación MEDINA, Parque Tecnológico de La Salud, Av. Conocimiento 34, 18016, Granada, Spain
| | - Diana de Prado-Verdún
- Centro de Investigaciones Energéticas, Medioambientales y Tecnológicas (CIEMAT), Madrid, Spain
| | - Ángeles Mencía
- Departamento de Bioingeniería E Ingeniería Aeroespacial (UC3M), División de Biomedicina Epitelial, Centro de Investigaciones Energéticas, Medioambientales y Tecnológicas (CIEMAT), Centro de Investigación Biomédica en Red de Enfermedades Raras, Universidad Carlos III de Madrid (UC3M), Madrid, Spain
- Unidad de Innovación Biomédica. Centro de Investigaciones Energéticas, U714-CIBER de Enfermedades Raras (CIBERER-ISCIII), Madrid, Spain
- Instituto de Investigación Sanitaria, Fundación Jiménez Díaz (IISFJD), Madrid, Spain
- Centro de Investigaciones Energéticas, Medioambientales y Tecnológicas (CIEMAT), Madrid, Spain
| | - Mercè Pont
- R&D Centre, Almirall S.A., Laureà Miró 408-410, 08980, Sant Feliu de Llobregat, Barcelona, Spain
| | - Carles Puig
- R&D Centre, Almirall S.A., Laureà Miró 408-410, 08980, Sant Feliu de Llobregat, Barcelona, Spain
| | - Marie-Helene Larraufie
- R&D Centre, Almirall S.A., Laureà Miró 408-410, 08980, Sant Feliu de Llobregat, Barcelona, Spain
| | | | - Fernando Reyes
- Fundación MEDINA, Parque Tecnológico de La Salud, Av. Conocimiento 34, 18016, Granada, Spain
| | - Juan Luis Trincado
- R&D Centre, Almirall S.A., Laureà Miró 408-410, 08980, Sant Feliu de Llobregat, Barcelona, Spain
| | - Vicente García-González
- R&D Centre, Almirall S.A., Laureà Miró 408-410, 08980, Sant Feliu de Llobregat, Barcelona, Spain
| | - Rosario Cerrato
- R&D Centre, Almirall S.A., Laureà Miró 408-410, 08980, Sant Feliu de Llobregat, Barcelona, Spain
| | - Miriam Andrés
- R&D Centre, Almirall S.A., Laureà Miró 408-410, 08980, Sant Feliu de Llobregat, Barcelona, Spain
| | - Maribel Crespo
- R&D Centre, Almirall S.A., Laureà Miró 408-410, 08980, Sant Feliu de Llobregat, Barcelona, Spain
| | - Francisca Vicente
- Fundación MEDINA, Parque Tecnológico de La Salud, Av. Conocimiento 34, 18016, Granada, Spain
| | - Nuria Godessart
- R&D Centre, Almirall S.A., Laureà Miró 408-410, 08980, Sant Feliu de Llobregat, Barcelona, Spain
| | - Olga Genilloud
- Fundación MEDINA, Parque Tecnológico de La Salud, Av. Conocimiento 34, 18016, Granada, Spain
| | - Fernando Larcher
- Departamento de Bioingeniería E Ingeniería Aeroespacial (UC3M), División de Biomedicina Epitelial, Centro de Investigaciones Energéticas, Medioambientales y Tecnológicas (CIEMAT), Centro de Investigación Biomédica en Red de Enfermedades Raras, Universidad Carlos III de Madrid (UC3M), Madrid, Spain.
- Unidad de Innovación Biomédica. Centro de Investigaciones Energéticas, U714-CIBER de Enfermedades Raras (CIBERER-ISCIII), Madrid, Spain.
- Instituto de Investigación Sanitaria, Fundación Jiménez Díaz (IISFJD), Madrid, Spain.
- Centro de Investigaciones Energéticas, Medioambientales y Tecnológicas (CIEMAT), Madrid, Spain.
| | - Arsenio Nueda
- R&D Centre, Almirall S.A., Laureà Miró 408-410, 08980, Sant Feliu de Llobregat, Barcelona, Spain.
| |
Collapse
|
10
|
Jia X, He X, Huang C, Li J, Dong Z, Liu K. Protein translation: biological processes and therapeutic strategies for human diseases. Signal Transduct Target Ther 2024; 9:44. [PMID: 38388452 PMCID: PMC10884018 DOI: 10.1038/s41392-024-01749-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2023] [Revised: 01/13/2024] [Accepted: 01/18/2024] [Indexed: 02/24/2024] Open
Abstract
Protein translation is a tightly regulated cellular process that is essential for gene expression and protein synthesis. The deregulation of this process is increasingly recognized as a critical factor in the pathogenesis of various human diseases. In this review, we discuss how deregulated translation can lead to aberrant protein synthesis, altered cellular functions, and disease progression. We explore the key mechanisms contributing to the deregulation of protein translation, including functional alterations in translation factors, tRNA, mRNA, and ribosome function. Deregulated translation leads to abnormal protein expression, disrupted cellular signaling, and perturbed cellular functions- all of which contribute to disease pathogenesis. The development of ribosome profiling techniques along with mass spectrometry-based proteomics, mRNA sequencing and single-cell approaches have opened new avenues for detecting diseases related to translation errors. Importantly, we highlight recent advances in therapies targeting translation-related disorders and their potential applications in neurodegenerative diseases, cancer, infectious diseases, and cardiovascular diseases. Moreover, the growing interest lies in targeted therapies aimed at restoring precise control over translation in diseased cells is discussed. In conclusion, this comprehensive review underscores the critical role of protein translation in disease and its potential as a therapeutic target. Advancements in understanding the molecular mechanisms of protein translation deregulation, coupled with the development of targeted therapies, offer promising avenues for improving disease outcomes in various human diseases. Additionally, it will unlock doors to the possibility of precision medicine by offering personalized therapies and a deeper understanding of the molecular underpinnings of diseases in the future.
Collapse
Affiliation(s)
- Xuechao Jia
- Department of Pathophysiology, School of Basic Medical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan, 450000, China
- China-US (Henan) Hormel Cancer Institute, Zhengzhou, Henan, 450000, China
| | - Xinyu He
- Department of Pathophysiology, School of Basic Medical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan, 450000, China
- China-US (Henan) Hormel Cancer Institute, Zhengzhou, Henan, 450000, China
| | - Chuntian Huang
- Department of Pathology and Pathophysiology, Henan University of Chinese Medicine, Zhengzhou, Henan, 450000, China
| | - Jian Li
- China-US (Henan) Hormel Cancer Institute, Zhengzhou, Henan, 450000, China
| | - Zigang Dong
- Department of Pathophysiology, School of Basic Medical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan, 450000, China.
- China-US (Henan) Hormel Cancer Institute, Zhengzhou, Henan, 450000, China.
- Tianjian Laboratory of Advanced Biomedical Sciences, Zhengzhou, Henan, 450052, China.
- Research Center for Basic Medicine Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, 450052, Henan, China.
- Provincial Cooperative Innovation Center for Cancer Chemoprevention, Zhengzhou University, Zhengzhou, Henan, 450000, China.
| | - Kangdong Liu
- Department of Pathophysiology, School of Basic Medical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan, 450000, China.
- China-US (Henan) Hormel Cancer Institute, Zhengzhou, Henan, 450000, China.
- Tianjian Laboratory of Advanced Biomedical Sciences, Zhengzhou, Henan, 450052, China.
- Research Center for Basic Medicine Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, 450052, Henan, China.
- Provincial Cooperative Innovation Center for Cancer Chemoprevention, Zhengzhou University, Zhengzhou, Henan, 450000, China.
- State Key Laboratory of Esophageal Cancer Prevention and Treatment, Zhengzhou University, Zhengzhou, Henan, 450000, China.
- The Collaborative Innovation Center of Henan Province for Cancer Chemoprevention, Zhengzhou, Henan, 450000, China.
| |
Collapse
|
11
|
Martinsen E, Jinnurine T, Subramani S, Rogne M. Advances in RNA therapeutics for modulation of 'undruggable' targets. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2024; 204:249-294. [PMID: 38458740 DOI: 10.1016/bs.pmbts.2023.12.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/10/2024]
Abstract
Over the past decades, drug discovery utilizing small pharmacological compounds, fragment-based therapeutics, and antibody therapy have significantly advanced treatment options for many human diseases. However, a major bottleneck has been that>70% of human proteins/genomic regions are 'undruggable' by the above-mentioned approaches. Many of these proteins constitute essential drug targets against complex multifactorial diseases like cancer, immunological disorders, and neurological diseases. Therefore, alternative approaches are required to target these proteins or genomic regions in human cells. RNA therapeutics is a promising approach for many of the traditionally 'undruggable' targets by utilizing methods such as antisense oligonucleotides, RNA interference, CRISPR/Cas-based genome editing, aptamers, and the development of mRNA therapeutics. In the following chapter, we will put emphasis on recent advancements utilizing these approaches against challenging drug targets, such as intranuclear proteins, intrinsically disordered proteins, untranslated genomic regions, and targets expressed in inaccessible tissues.
Collapse
Affiliation(s)
| | | | - Saranya Subramani
- Pioneer Research AS, Oslo Science Park, Oslo, Norway; Department of Pharmacy, Section for Pharmacology and Pharmaceutical Biosciences, University of Oslo, Oslo, Norway
| | - Marie Rogne
- Pioneer Research AS, Oslo Science Park, Oslo, Norway; Department of Molecular Medicine, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, Oslo, Norway.
| |
Collapse
|
12
|
Ling C, Dai Y, Geng C, Pan S, Quan W, Ding Q, Yang X, Shen D, Tao Q, Li J, Li J, Wang Y, Jiang S, Wang Y, Chen L, Cui L, Wang D. Uncovering the true features of dystrophin gene rearrangement and improving the molecular diagnosis of Duchenne and Becker muscular dystrophies. iScience 2023; 26:108365. [PMID: 38047063 PMCID: PMC10690541 DOI: 10.1016/j.isci.2023.108365] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Revised: 09/19/2023] [Accepted: 10/26/2023] [Indexed: 12/05/2023] Open
Abstract
Duchenne and Becker muscular dystrophies (DMD/BMD) are caused by complex mutations in the dystrophin gene (DMD). Currently, there is no integrative method for the precise detection of all potential DMD variants, a gap which we aimed to address using long-read sequencing. The captured long-read sequencing panel developed in this study was applied to 129 subjects, including 11 who had previously unsolved cases. The results showed that this method accurately detected DMD mutations, ranging from single-nucleotide variations to structural variations. Furthermore, our findings revealed that continuous exon duplication/deletion in the DMD/BMD cohort may be attributed to complex segmental rearrangements and that noncontiguous duplication/deletion is generally attributed to intragenic inversion or interchromosome translocation. Mutations in the deep introns were confirmed to produce a pseudoexon. Moreover, variations in female carriers were precisely identified. The integrated and precise DMD gene screening method proposed in this study could improve the molecular diagnosis of DMD/BMD.
Collapse
Affiliation(s)
- Chao Ling
- The Laboratory of Clinical Genetics, Medical Research Center, Peking Union Medical College Hospital, Chinese Academy of Medical Science & Peking Union Medical College, Beijing 100730, China
- State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science & Peking Union Medical College, Beijing 100730, China
| | - Yi Dai
- Department of Neurology, Peking Union Medical College Hospital, Chinese Academy of Medical Science & Peking Union Medical College, Beijing 100730, China
| | - Chang Geng
- Department of Neurology, Peking Union Medical College Hospital, Chinese Academy of Medical Science & Peking Union Medical College, Beijing 100730, China
| | - Shirang Pan
- Grandomics Biosciences, Beijing 102200, China
| | | | - Qingyun Ding
- Department of Neurology, Peking Union Medical College Hospital, Chinese Academy of Medical Science & Peking Union Medical College, Beijing 100730, China
| | - Xunzhe Yang
- Department of Neurology, Peking Union Medical College Hospital, Chinese Academy of Medical Science & Peking Union Medical College, Beijing 100730, China
| | - Dongchao Shen
- Department of Neurology, Peking Union Medical College Hospital, Chinese Academy of Medical Science & Peking Union Medical College, Beijing 100730, China
| | - Qing Tao
- Grandomics Biosciences, Beijing 102200, China
| | - Jingjing Li
- Grandomics Biosciences, Beijing 102200, China
| | - Jia Li
- Department of Neurology, Peking Union Medical College Hospital, Chinese Academy of Medical Science & Peking Union Medical College, Beijing 100730, China
| | - Yinbing Wang
- Department of Neurology, Peking Union Medical College Hospital, Chinese Academy of Medical Science & Peking Union Medical College, Beijing 100730, China
| | - Shan Jiang
- Department of Neurology, Peking Union Medical College Hospital, Chinese Academy of Medical Science & Peking Union Medical College, Beijing 100730, China
| | - Yang Wang
- Grandomics Biosciences, Beijing 102200, China
| | - Lin Chen
- Department of Neurology, Peking Union Medical College Hospital, Chinese Academy of Medical Science & Peking Union Medical College, Beijing 100730, China
| | - Liying Cui
- Department of Neurology, Peking Union Medical College Hospital, Chinese Academy of Medical Science & Peking Union Medical College, Beijing 100730, China
| | - Depeng Wang
- Grandomics Biosciences, Beijing 102200, China
| |
Collapse
|
13
|
Doisy M, Vacca O, Fergus C, Gileadi T, Verhaeg M, Saoudi A, Tensorer T, Garcia L, Kelly VP, Montanaro F, Morgan JE, van Putten M, Aartsma-Rus A, Vaillend C, Muntoni F, Goyenvalle A. Networking to Optimize Dmd exon 53 Skipping in the Brain of mdx52 Mouse Model. Biomedicines 2023; 11:3243. [PMID: 38137463 PMCID: PMC10741439 DOI: 10.3390/biomedicines11123243] [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: 09/19/2023] [Revised: 11/28/2023] [Accepted: 12/04/2023] [Indexed: 12/24/2023] Open
Abstract
Duchenne muscular dystrophy (DMD) is caused by mutations in the DMD gene that disrupt the open reading frame and thus prevent production of functional dystrophin proteins. Recent advances in DMD treatment, notably exon skipping and AAV gene therapy, have achieved some success aimed at alleviating the symptoms related to progressive muscle damage. However, they do not address the brain comorbidities associated with DMD, which remains a critical aspect of the disease. The mdx52 mouse model recapitulates one of the most frequent genetic pathogenic variants associated with brain involvement in DMD. Deletion of exon 52 impedes expression of two brain dystrophins, Dp427 and Dp140, expressed from distinct promoters. Interestingly, this mutation is eligible for exon skipping strategies aimed at excluding exon 51 or 53 from dystrophin mRNA. We previously showed that exon 51 skipping can restore partial expression of internally deleted yet functional Dp427 in the brain following intracerebroventricular (ICV) injection of antisense oligonucleotides (ASO). This was associated with a partial improvement of anxiety traits, unconditioned fear response, and Pavlovian fear learning and memory in the mdx52 mouse model. In the present study, we investigated in the same mouse model the skipping of exon 53 in order to restore expression of both Dp427 and Dp140. However, in contrast to exon 51, we found that exon 53 skipping was particularly difficult in mdx52 mice and a combination of multiple ASOs had to be used simultaneously to reach substantial levels of exon 53 skipping, regardless of their chemistry (tcDNA, PMO, or 2'MOE). Following ICV injection of a combination of ASO sequences, we measured up to 25% of exon 53 skipping in the hippocampus of treated mdx52 mice, but this did not elicit significant protein restoration. These findings indicate that skipping mouse dystrophin exon 53 is challenging. As such, it has not yet been possible to answer the pertinent question whether rescuing both Dp427 and Dp140 in the brain is imperative to more optimal treatment of neurological aspects of dystrophinopathy.
Collapse
Affiliation(s)
- Mathilde Doisy
- Université Paris-Saclay, UVSQ, Inserm, END-ICAP, 78000 Versailles, France; (M.D.); (O.V.); (A.S.)
| | - Ophélie Vacca
- Université Paris-Saclay, UVSQ, Inserm, END-ICAP, 78000 Versailles, France; (M.D.); (O.V.); (A.S.)
| | - Claire Fergus
- School of Biochemistry & Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, D02 R590 Dublin, Ireland; (C.F.)
| | - Talia Gileadi
- Dubowitz Neuromuscular Centre, UCL Great Ormond Street Institute of Child Health, 30 Guildford Street, London WC1N 1EH, UK; (T.G.); (F.M.); (J.E.M.); (F.M.)
- National Institute for Health Research, Great Ormond Street Institute of Child Health Biomedical Research Centre, University College London, London WC1N 1EH, UK
| | - Minou Verhaeg
- Department of Human Genetics, Leiden University Medical Center, 2333ZA Leiden, The Netherlands; (M.V.); (M.v.P.); (A.A.-R.)
| | - Amel Saoudi
- Université Paris-Saclay, UVSQ, Inserm, END-ICAP, 78000 Versailles, France; (M.D.); (O.V.); (A.S.)
- Université Paris-Saclay, CNRS, Institut des Neurosciences Paris-Saclay, 91400 Saclay, France;
| | - Thomas Tensorer
- SQY Therapeutics-Synthena, UVSQ, 78180 Montigny le Bretonneux, France
| | - Luis Garcia
- Université Paris-Saclay, UVSQ, Inserm, END-ICAP, 78000 Versailles, France; (M.D.); (O.V.); (A.S.)
| | - Vincent P. Kelly
- School of Biochemistry & Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, D02 R590 Dublin, Ireland; (C.F.)
| | - Federica Montanaro
- Dubowitz Neuromuscular Centre, UCL Great Ormond Street Institute of Child Health, 30 Guildford Street, London WC1N 1EH, UK; (T.G.); (F.M.); (J.E.M.); (F.M.)
- National Institute for Health Research, Great Ormond Street Institute of Child Health Biomedical Research Centre, University College London, London WC1N 1EH, UK
| | - Jennifer E. Morgan
- Dubowitz Neuromuscular Centre, UCL Great Ormond Street Institute of Child Health, 30 Guildford Street, London WC1N 1EH, UK; (T.G.); (F.M.); (J.E.M.); (F.M.)
- National Institute for Health Research, Great Ormond Street Institute of Child Health Biomedical Research Centre, University College London, London WC1N 1EH, UK
| | - Maaike van Putten
- Department of Human Genetics, Leiden University Medical Center, 2333ZA Leiden, The Netherlands; (M.V.); (M.v.P.); (A.A.-R.)
| | - Annemieke Aartsma-Rus
- Department of Human Genetics, Leiden University Medical Center, 2333ZA Leiden, The Netherlands; (M.V.); (M.v.P.); (A.A.-R.)
| | - Cyrille Vaillend
- Université Paris-Saclay, CNRS, Institut des Neurosciences Paris-Saclay, 91400 Saclay, France;
| | - Francesco Muntoni
- Dubowitz Neuromuscular Centre, UCL Great Ormond Street Institute of Child Health, 30 Guildford Street, London WC1N 1EH, UK; (T.G.); (F.M.); (J.E.M.); (F.M.)
- National Institute for Health Research, Great Ormond Street Institute of Child Health Biomedical Research Centre, University College London, London WC1N 1EH, UK
| | - Aurélie Goyenvalle
- Université Paris-Saclay, UVSQ, Inserm, END-ICAP, 78000 Versailles, France; (M.D.); (O.V.); (A.S.)
| |
Collapse
|
14
|
Falzarano MS, Mietto M, Fortunato F, Farnè M, Martini F, Ala P, Selvatici R, Muntoni F, Ferlini A. mRNA in situ hybridization exhibits unbalanced nuclear/cytoplasmic dystrophin transcript repartition in Duchenne myogenic cells and skeletal muscle biopsies. Sci Rep 2023; 13:15942. [PMID: 37743371 PMCID: PMC10518324 DOI: 10.1038/s41598-023-43134-6] [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: 05/04/2023] [Accepted: 09/20/2023] [Indexed: 09/26/2023] Open
Abstract
To gain insight on dystrophin (DMD) gene transcription dynamics and spatial localization, we assayed the DMD mRNA amount and defined its compartmentalization in myoblasts, myotubes, and skeletal muscle biopsies of Duchenne muscular dystrophy (DMD) patients. Using droplet digital PCR, Real-time PCR, and RNAscope in situ hybridization, we showed that the DMD transcript amount is extremely reduced in both DMD patients' cells and muscle biopsies and that mutation-related differences occur. We also found that, compared to controls, DMD transcript is dramatically reduced in the cytoplasm, as up to 90% of it is localized in nuclei, preferentially at the perinuclear region. Using RNA/protein colocalization experiments, we showed that about 40% of nuclear DMD mRNA is localized in the nucleoli in both control and DMD myogenic cells. Our results clearly show that mutant DMD mRNA quantity is strongly reduced in the patients' myogenic cells and muscle biopsies. Furthermore, mutant DMD mRNA compartmentalization is spatially unbalanced due to a shift in its localization towards the nuclei. This abnormal transcript repartition contributes to the poor abundance and availability of the dystrophin messenger in cytoplasm. This novel finding also has important repercussions for RNA-targeted therapies.
Collapse
Affiliation(s)
- Maria Sofia Falzarano
- Department of Medical Sciences, Unit of Medical Genetics, University of Ferrara, Ferrara, Italy
| | - Martina Mietto
- Department of Medical Sciences, Unit of Medical Genetics, University of Ferrara, Ferrara, Italy
| | - Fernanda Fortunato
- Department of Medical Sciences, Unit of Medical Genetics, University of Ferrara, Ferrara, Italy
| | - Marianna Farnè
- Department of Medical Sciences, Unit of Medical Genetics, University of Ferrara, Ferrara, Italy
| | - Fernanda Martini
- Department of Medical Sciences, Section of Experimental Medicine, University of Ferrara, Ferrara, Italy
| | - Pierpaolo Ala
- Dubowitz Neuromuscular Centre and National Institute for Health Research, Great Ormond Street Institute of Child Health, Biomedical Research Centre, University College London, London, UK
| | - Rita Selvatici
- Department of Medical Sciences, Unit of Medical Genetics, University of Ferrara, Ferrara, Italy
| | - Francesco Muntoni
- Dubowitz Neuromuscular Centre and National Institute for Health Research, Great Ormond Street Institute of Child Health, Biomedical Research Centre, University College London, London, UK
| | - Alessandra Ferlini
- Department of Medical Sciences, Unit of Medical Genetics, University of Ferrara, Ferrara, Italy.
- Dubowitz Neuromuscular Centre and National Institute for Health Research, Great Ormond Street Institute of Child Health, Biomedical Research Centre, University College London, London, UK.
| |
Collapse
|
15
|
Hildyard JCW, Piercy RJ. When Size Really Matters: The Eccentricities of Dystrophin Transcription and the Hazards of Quantifying mRNA from Very Long Genes. Biomedicines 2023; 11:2082. [PMID: 37509720 PMCID: PMC10377302 DOI: 10.3390/biomedicines11072082] [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: 06/27/2023] [Revised: 07/10/2023] [Accepted: 07/14/2023] [Indexed: 07/30/2023] Open
Abstract
At 2.3 megabases in length, the dystrophin gene is enormous: transcription of a single mRNA requires approximately 16 h. Principally expressed in skeletal muscle, the dystrophin protein product protects the muscle sarcolemma against contraction-induced injury, and dystrophin deficiency results in the fatal muscle-wasting disease, Duchenne muscular dystrophy. This gene is thus of key clinical interest, and therapeutic strategies aimed at eliciting dystrophin restoration require quantitative analysis of its expression. Approaches for quantifying dystrophin at the protein level are well-established, however study at the mRNA level warrants closer scrutiny: measured expression values differ in a sequence-dependent fashion, with significant consequences for data interpretation. In this manuscript, we discuss these nuances of expression and present evidence to support a transcriptional model whereby the long transcription time is coupled to a short mature mRNA half-life, with dystrophin transcripts being predominantly nascent as a consequence. We explore the effects of such a model on cellular transcriptional dynamics and then discuss key implications for the study of dystrophin gene expression, focusing on both conventional (qPCR) and next-gen (RNAseq) approaches.
Collapse
Affiliation(s)
- John C. W. Hildyard
- Comparative Neuromuscular Disease Laboratory, Department of Clinical Science and Services, Royal Veterinary College, London NW1 0TU, UK;
| | | |
Collapse
|
16
|
Saoudi A, Barberat S, le Coz O, Vacca O, Doisy Caquant M, Tensorer T, Sliwinski E, Garcia L, Muntoni F, Vaillend C, Goyenvalle A. Partial restoration of brain dystrophin by tricyclo-DNA antisense oligonucleotides alleviates emotional deficits in mdx52 mice. MOLECULAR THERAPY - NUCLEIC ACIDS 2023; 32:173-188. [PMID: 37078061 PMCID: PMC10106732 DOI: 10.1016/j.omtn.2023.03.009] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Accepted: 03/16/2023] [Indexed: 04/05/2023]
Abstract
The mdx52 mouse model recapitulates a frequent mutation profile associated with brain involvement in Duchenne muscular dystrophy. Deletion of exon 52 impedes expression of two dystrophins (Dp427, Dp140) expressed in brain, and is eligible for therapeutic exon-skipping strategies. We previously showed that mdx52 mice display enhanced anxiety and fearfulness, and impaired associative fear learning. In this study, we examined the reversibility of these phenotypes using exon 51 skipping to restore exclusively Dp427 expression in the brain of mdx52 mice. We first show that a single intracerebroventricular administration of tricyclo-DNA antisense oligonucleotides targeting exon 51 restores 5%-15% of dystrophin protein expression in the hippocampus, cerebellum, and cortex, at stable levels between 7 and 11 week after injection. Anxiety and unconditioned fear were significantly reduced in treated mdx52 mice and acquisition of fear conditioning appeared fully rescued, while fear memory tested 24 h later was only partially improved. Additional restoration of Dp427 in skeletal and cardiac muscles by systemic treatment did not further improve the unconditioned fear response, confirming the central origin of this phenotype. These findings indicate that some emotional and cognitive deficits associated with dystrophin deficiency may be reversible or at least improved by partial postnatal dystrophin rescue.
Collapse
Affiliation(s)
- Amel Saoudi
- Université Paris-Saclay, UVSQ, Inserm, END-ICAP, 78000 Versailles, France
- Université Paris-Saclay, CNRS, Institut des Neurosciences Paris-Saclay, 91400 Saclay, France
| | - Sacha Barberat
- Université Paris-Saclay, UVSQ, Inserm, END-ICAP, 78000 Versailles, France
| | - Olivier le Coz
- Université Paris-Saclay, UVSQ, Inserm, END-ICAP, 78000 Versailles, France
| | - Ophélie Vacca
- Université Paris-Saclay, UVSQ, Inserm, END-ICAP, 78000 Versailles, France
| | | | - Thomas Tensorer
- SQY Therapeutics – Synthena, UVSQ, 78180 Montigny le Bretonneux, France
| | - Eric Sliwinski
- SQY Therapeutics – Synthena, UVSQ, 78180 Montigny le Bretonneux, France
| | - Luis Garcia
- Université Paris-Saclay, UVSQ, Inserm, END-ICAP, 78000 Versailles, France
| | - Francesco Muntoni
- The Dubowitz Neuromuscular Centre, Developmental Neurosciences Research and Teaching Department, Great Ormond Street Institute of Child Health, University College London, WC1N 1EH London, UK
| | - Cyrille Vaillend
- Université Paris-Saclay, CNRS, Institut des Neurosciences Paris-Saclay, 91400 Saclay, France
- Corresponding author Cyrille Vaillend, Université Paris-Saclay, CNRS, Institut des Neurosciences Paris-Saclay, 91400 Saclay, France.
| | - Aurélie Goyenvalle
- Université Paris-Saclay, UVSQ, Inserm, END-ICAP, 78000 Versailles, France
- Corresponding author Aurélie Goyenvalle, Université Paris-Saclay, UVSQ, Inserm, END-ICAP, 78000 Versailles, France.
| |
Collapse
|
17
|
Dystrophin myonuclear domain restoration governs treatment efficacy in dystrophic muscle. Proc Natl Acad Sci U S A 2023; 120:e2206324120. [PMID: 36595689 PMCID: PMC9926233 DOI: 10.1073/pnas.2206324120] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Dystrophin is essential for muscle health: its sarcolemmal absence causes the fatal, X-linked condition, Duchenne muscular dystrophy (DMD). However, its normal, spatial organization remains poorly understood, which hinders the interpretation of efficacy of its therapeutic restoration. Using female reporter mice heterozygous for fluorescently tagged dystrophin (DmdEGFP), we here reveal that dystrophin distribution is unexpectedly compartmentalized, being restricted to myonuclear-defined sarcolemmal territories extending ~80 µm, which we called "basal sarcolemmal dystrophin units (BSDUs)." These territories were further specialized at myotendinous junctions, where both Dmd transcripts and dystrophin protein were enriched. Genome-level correction in X-linked muscular dystrophy mice via CRISPR/Cas9 gene editing restored a mosaic of separated dystrophin domains, whereas transcript-level Dmd correction, following treatment with tricyclo-DNA antisense oligonucleotides, restored dystrophin initially at junctions before extending along the entire fiber-with levels ~2% sufficient to moderate the dystrophic process. We conclude that widespread restoration of fiber dystrophin is likely critical for therapeutic success in DMD, perhaps most importantly, at muscle-tendon junctions.
Collapse
|
18
|
Chey YCJ, Arudkumar J, Aartsma-Rus A, Adikusuma F, Thomas PQ. CRISPR applications for Duchenne muscular dystrophy: From animal models to potential therapies. WIREs Mech Dis 2023; 15:e1580. [PMID: 35909075 PMCID: PMC10078488 DOI: 10.1002/wsbm.1580] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Revised: 04/28/2022] [Accepted: 06/30/2022] [Indexed: 01/31/2023]
Abstract
CRISPR gene-editing technology creates precise and permanent modifications to DNA. It has significantly advanced our ability to generate animal disease models for use in biomedical research and also has potential to revolutionize the treatment of genetic disorders. Duchenne muscular dystrophy (DMD) is a monogenic muscle-wasting disease that could potentially benefit from the development of CRISPR therapy. It is commonly associated with mutations that disrupt the reading frame of the DMD gene that encodes dystrophin, an essential scaffolding protein that stabilizes striated muscles and protects them from contractile-induced damage. CRISPR enables the rapid generation of various animal models harboring mutations that closely simulates the wide variety of mutations observed in DMD patients. These models provide a platform for the testing of sequence-specific interventions like CRISPR therapy that aim to reframe or skip DMD mutations to restore functional dystrophin expression. This article is categorized under: Congenital Diseases > Genetics/Genomics/Epigenetics.
Collapse
Affiliation(s)
- Yu C J Chey
- School of Biomedicine and Robinson Research Institute, University of Adelaide, Adelaide, South Australia, Australia.,Genome Editing Program, South Australian Health and Medical Research Institute (SAHMRI), Adelaide, South Australia, Australia
| | - Jayshen Arudkumar
- School of Biomedicine and Robinson Research Institute, University of Adelaide, Adelaide, South Australia, Australia.,Genome Editing Program, South Australian Health and Medical Research Institute (SAHMRI), Adelaide, South Australia, Australia
| | - Annemieke Aartsma-Rus
- Department of Human Genetics, Leiden University Medical Center, Leiden, The Netherlands
| | - Fatwa Adikusuma
- School of Biomedicine and Robinson Research Institute, University of Adelaide, Adelaide, South Australia, Australia.,Genome Editing Program, South Australian Health and Medical Research Institute (SAHMRI), Adelaide, South Australia, Australia.,CSIRO Synthetic Biology Future Science Platform, Canberra, Australia
| | - Paul Q Thomas
- School of Biomedicine and Robinson Research Institute, University of Adelaide, Adelaide, South Australia, Australia.,Genome Editing Program, South Australian Health and Medical Research Institute (SAHMRI), Adelaide, South Australia, Australia.,South Australian Genome Editing (SAGE), South Australian Health and Medical Research Institute (SAHMRI), Adelaide, South Australia, Australia
| |
Collapse
|
19
|
Goossens R, Verwey N, Ariyurek Y, Schnell F, Aartsma-Rus A. DMD antisense oligonucleotide mediated exon skipping efficiency correlates with flanking intron retention time and target position within the exon. RNA Biol 2023; 20:693-702. [PMID: 37667454 PMCID: PMC10481881 DOI: 10.1080/15476286.2023.2254041] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Revised: 08/25/2023] [Accepted: 08/28/2023] [Indexed: 09/06/2023] Open
Abstract
Mutations in the DMD gene are causative for Duchenne muscular dystrophy (DMD). Antisense oligonucleotide (AON) mediated exon skipping to restore disrupted dystrophin reading frame is a therapeutic approach that allows production of a shorter but functional protein. As DMD causing mutations can affect most of the 79 exons encoding dystrophin, a wide variety of AONs are needed to treat the patient population. Design of AONs is largely guided by trial-and-error, and it is yet unclear what defines the skippability of an exon. Here, we use a library of phosphorodiamidate morpholino oligomer (PMOs) AONs of similar physical properties to test the skippability of a large number of DMD exons. The DMD transcript is non-sequentially spliced, meaning that certain introns are retained longer in the transcript than downstream introns. We tested whether the relative intron retention time has a significant effect on AON efficiency, and found that targeting an out-of-frame exon flanked at its 5'-end by an intron that is retained in the transcript longer ('slow' intron) leads to overall higher exon skipping efficiency than when the 5'-end flanking intron is 'fast'. Regardless of splicing speed of flanking introns, we find that positioning an AON closer to the 5'-end of the target exon leads to higher exon skipping efficiency opposed to targeting an exons 3'-end. The data enclosed herein can be of use to guide future target selection and preferential AON binding sites for both DMD and other disease amenable by exon skipping therapies.
Collapse
Affiliation(s)
- Remko Goossens
- Department of Human Genetics, Leiden University Medical Center, Leiden, The Netherlands
| | - Nisha Verwey
- Department of Human Genetics, Leiden University Medical Center, Leiden, The Netherlands
| | - Yavuz Ariyurek
- Department of Human Genetics, Leiden University Medical Center, Leiden, The Netherlands
- Leiden Genome Technology Center, Leiden University Medical Center, Leiden, The Netherlands
| | | | - Annemieke Aartsma-Rus
- Department of Human Genetics, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
20
|
Kaur B, Kaur J, Kashyap N, Arora JS, Mukhopadhyay CS. A comprehensive review of genomic perspectives of canine diseases as a model to study human disorders. CANADIAN JOURNAL OF VETERINARY RESEARCH = REVUE CANADIENNE DE RECHERCHE VETERINAIRE 2023; 87:3-8. [PMID: 36606040 PMCID: PMC9808881] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Accepted: 08/18/2022] [Indexed: 01/07/2023]
Abstract
The domestic dog has been given considerable attention as a system for investigating the genetics of human diseases. Population diversity and breed structure are unique features that make dogs particularly amenable to genetic studies. Dogs show distinguished features of breed-specific homogeneity, which is associated with striking interbreed heterogeneity. This review discusses the significance of studying the genetic maps, genome-wide association studies (GWAS), and usefulness of this species as an animal model. Most canine genetic disorders are similar to those of humans, including inherited, psychiatric, and genetic disorders. In addition to revealing new candidate genes, canine models allow access to experimental resources, such as cells, tissues, and even live animals, for research and intervention purposes.
Collapse
|
21
|
Histone deacetylase inhibitors improve antisense-mediated exon-skipping efficacy in mdx mice. MOLECULAR THERAPY. NUCLEIC ACIDS 2022; 30:606-620. [PMID: 36514350 PMCID: PMC9722397 DOI: 10.1016/j.omtn.2022.11.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Accepted: 11/17/2022] [Indexed: 11/23/2022]
Abstract
Antisense-mediated exon skipping is one of the most promising therapeutic strategies for Duchenne muscular dystrophy (DMD), and some antisense oligonucleotide (ASO) drugs have already been approved by the US FDA despite their low efficacy. The potential of this therapy is still limited by several challenges, including the reduced expression of the dystrophin transcript and the strong 5'-3' imbalance in mutated transcripts. We therefore hypothesize that increasing histone acetylation using histone deacetylase inhibitors (HDACi) could correct the transcript imbalance, offering more available pre-mRNA target and ultimately increasing dystrophin rescue. Here, we evaluated the impact of such a combined therapy on the Dmd transcript imbalance phenomenon and on dystrophin restoration levels in mdx mice. Analysis of the Dmd transcript levels at different exon-exon junctions revealed a tendency to correct the 5'-3' imbalance phenomenon following treatment with HDACi. Significantly higher levels of dystrophin restoration (up to 74% increase) were obtained with givinostat and valproic acid compared with mice treated with ASO alone. Additionally, we demonstrate an increase in H3K9 acetylation in human myocytes after treatment with valproic acid. These findings indicate that HDACi can improve the therapeutic potential of exon-skipping approaches, offering promising perspectives for the treatment of DMD.
Collapse
|
22
|
Desjardins CA, Yao M, Hall J, O’Donnell E, Venkatesan R, Spring S, Wen A, Hsia N, Shen P, Russo R, Lan B, Picariello T, Tang K, Weeden T, Zanotti S, Subramanian R, Ibraghimov-Beskrovnaya O. Enhanced exon skipping and prolonged dystrophin restoration achieved by TfR1-targeted delivery of antisense oligonucleotide using FORCE conjugation in mdx mice. Nucleic Acids Res 2022; 50:11401-11414. [PMID: 35944903 PMCID: PMC9723632 DOI: 10.1093/nar/gkac641] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 06/15/2022] [Accepted: 07/20/2022] [Indexed: 12/24/2022] Open
Abstract
Current therapies for Duchenne muscular dystrophy (DMD) use phosphorodiamidate morpholino oligomers (PMO) to induce exon skipping in the dystrophin pre-mRNA, enabling the translation of a shortened but functional dystrophin protein. This strategy has been hampered by insufficient delivery of PMO to cardiac and skeletal muscle. To overcome these limitations, we developed the FORCETM platform consisting of an antigen-binding fragment, which binds the transferrin receptor 1, conjugated to an oligonucleotide. We demonstrate that a single dose of the mouse-specific FORCE-M23D conjugate enhances muscle delivery of exon skipping PMO (M23D) in mdx mice, achieving dose-dependent and robust exon skipping and durable dystrophin restoration. FORCE-M23D-induced dystrophin expression reached peaks of 51%, 72%, 62%, 90% and 77%, of wild-type levels in quadriceps, tibialis anterior, gastrocnemius, diaphragm, and heart, respectively, with a single 30 mg/kg PMO-equivalent dose. The shortened dystrophin localized to the sarcolemma, indicating expression of a functional protein. Conversely, a single 30 mg/kg dose of unconjugated M23D displayed poor muscle delivery resulting in marginal levels of exon skipping and dystrophin expression. Importantly, FORCE-M23D treatment resulted in improved functional outcomes compared with administration of unconjugated M23D. Our results suggest that FORCE conjugates are a potentially effective approach for the treatment of DMD.
Collapse
Affiliation(s)
| | - Monica Yao
- Research Department, Dyne Therapeutics Inc., Waltham, MA 02451, USA
| | - John Hall
- Research Department, Dyne Therapeutics Inc., Waltham, MA 02451, USA
| | - Emma O’Donnell
- Research Department, Dyne Therapeutics Inc., Waltham, MA 02451, USA
| | | | - Sean Spring
- Research Department, Dyne Therapeutics Inc., Waltham, MA 02451, USA
| | - Aiyun Wen
- Research Department, Dyne Therapeutics Inc., Waltham, MA 02451, USA
| | - Nelson Hsia
- Research Department, Dyne Therapeutics Inc., Waltham, MA 02451, USA
| | - Peiyi Shen
- Research Department, Dyne Therapeutics Inc., Waltham, MA 02451, USA
| | - Ryan Russo
- Research Department, Dyne Therapeutics Inc., Waltham, MA 02451, USA
| | - Bo Lan
- Research Department, Dyne Therapeutics Inc., Waltham, MA 02451, USA
| | - Tyler Picariello
- Research Department, Dyne Therapeutics Inc., Waltham, MA 02451, USA
| | - Kim Tang
- Research Department, Dyne Therapeutics Inc., Waltham, MA 02451, USA
| | - Timothy Weeden
- Research Department, Dyne Therapeutics Inc., Waltham, MA 02451, USA
| | - Stefano Zanotti
- Research Department, Dyne Therapeutics Inc., Waltham, MA 02451, USA
| | | | | |
Collapse
|
23
|
Mollard A, Peccate C, Forand A, Chassagne J, Julien L, Meunier P, Guesmia Z, Marais T, Bitoun M, Piétri-Rouxel F, Benkhelifa-Ziyyat S, Lorain S. Muscle regeneration affects Adeno Associated Virus 1 mediated transgene transcription. Sci Rep 2022; 12:9674. [PMID: 35690627 PMCID: PMC9188557 DOI: 10.1038/s41598-022-13405-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Accepted: 05/24/2022] [Indexed: 12/16/2022] Open
Abstract
Duchenne muscular dystrophy is a severe neuromuscular disease causing a progressive muscle wasting due to mutations in the DMD gene that lead to the absence of dystrophin protein. Adeno-associated virus (AAV)-based therapies aiming to restore dystrophin in muscles, by either exon skipping or microdystrophin expression, are very promising. However, the absence of dystrophin induces cellular perturbations that hinder AAV therapy efficiency. We focused here on the impact of the necrosis-regeneration process leading to nuclear centralization in myofiber, a common feature of human myopathies, on AAV transduction efficiency. We generated centronucleated myofibers by cardiotoxin injection in wild-type muscles prior to AAV injection. Intramuscular injections of AAV1 vectors show that transgene expression was drastically reduced in regenerated muscles, even when the AAV injection occurred 10 months post-regeneration. We show also that AAV genomes were not lost from cardiotoxin regenerated muscle and were properly localised in the myofiber nuclei but were less transcribed leading to muscle transduction defect. A similar defect was observed in muscles of the DMD mouse model mdx. Therefore, the regeneration process per se could participate to the AAV-mediated transduction defect observed in dystrophic muscles which may limit AAV-based therapies.
Collapse
Affiliation(s)
- Amédée Mollard
- Sorbonne Université, Inserm, Institut de Myologie, Centre de Recherche en Myologie, 75013, Paris, France
| | - Cécile Peccate
- Sorbonne Université, Inserm, Institut de Myologie, Centre de Recherche en Myologie, 75013, Paris, France
| | - Anne Forand
- Sorbonne Université, Inserm, Institut de Myologie, Centre de Recherche en Myologie, 75013, Paris, France
| | - Julie Chassagne
- Sorbonne Université, Inserm, Institut de Myologie, Centre de Recherche en Myologie, 75013, Paris, France
| | - Laura Julien
- Sorbonne Université, Inserm, Institut de Myologie, Centre de Recherche en Myologie, 75013, Paris, France
| | - Pierre Meunier
- Sorbonne Université, Inserm, Institut de Myologie, Centre de Recherche en Myologie, 75013, Paris, France
| | - Zoheir Guesmia
- Sorbonne Université, Inserm, Institut de Myologie, Centre de Recherche en Myologie, 75013, Paris, France
| | - Thibaut Marais
- Sorbonne Université, Inserm, Institut de Myologie, Centre de Recherche en Myologie, 75013, Paris, France
| | - Marc Bitoun
- Sorbonne Université, Inserm, Institut de Myologie, Centre de Recherche en Myologie, 75013, Paris, France
| | - France Piétri-Rouxel
- Sorbonne Université, Inserm, Institut de Myologie, Centre de Recherche en Myologie, 75013, Paris, France
| | - Sofia Benkhelifa-Ziyyat
- Sorbonne Université, Inserm, Institut de Myologie, Centre de Recherche en Myologie, 75013, Paris, France.
| | - Stéphanie Lorain
- Sorbonne Université, Inserm, Institut de Myologie, Centre de Recherche en Myologie, 75013, Paris, France.,AFM-Téléthon, 1 rue de l'Internationale, BP59, 91002, Evry, France
| |
Collapse
|
24
|
Stefano MED, Ferretti V, Mozzetta C. Synaptic alterations as a neurodevelopmental trait of Duchenne muscular dystrophy. Neurobiol Dis 2022; 168:105718. [PMID: 35390481 DOI: 10.1016/j.nbd.2022.105718] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Revised: 03/28/2022] [Accepted: 03/29/2022] [Indexed: 01/14/2023] Open
Abstract
Dystrophinopaties, e.g., Duchenne muscular dystrophy (DMD), Becker muscular dystrophy and X-linked dilated cardiomyopathy are inherited neuromuscular diseases, characterized by progressive muscular degeneration, which however associate with a significant impact on general system physiology. The more severe is the pathology and its diversified manifestations, the heavier are its effects on organs, systems, and tissues other than muscles (skeletal, cardiac and smooth muscles). All dystrophinopaties are characterized by mutations in a single gene located on the X chromosome encoding dystrophin (Dp427) and its shorter isoforms, but DMD is the most devasting: muscular degenerations manifests within the first 4 years of life, progressively affecting motility and other muscular functions, and leads to a fatal outcome between the 20s and 40s. To date, after years of studies on both DMD patients and animal models of the disease, it has been clearly demonstrated that a significant percentage of DMD patients are also afflicted by cognitive, neurological, and autonomic disorders, of varying degree of severity. The anatomical correlates underlying neural functional damages are established during embryonic development and the early stages of postnatal life, when brain circuits, sensory and motor connections are still maturing. The impact of the absence of Dp427 on the development, differentiation, and consolidation of specific cerebral circuits (hippocampus, cerebellum, prefrontal cortex, amygdala) is significant, and amplified by the frequent lack of one or more of its lower molecular mass isoforms. The most relevant aspect, which characterizes DMD-associated neurological disorders, is based on morpho-functional alterations of selective synaptic connections within the affected brain areas. This pathological feature correlates neurological conditions of DMD to other severe neurological disorders, such as schizophrenia, epilepsy and autistic spectrum disorders, among others. This review discusses the organization and the role of the dystrophin-dystroglycan complex in muscles and neurons, focusing on the neurological aspect of DMD and on the most relevant morphological and functional synaptic alterations, in both central and autonomic nervous systems, described in the pathology and its animal models.
Collapse
Affiliation(s)
- Maria Egle De Stefano
- Department of Biology and Biotechnology "Charles Darwin", Sapienza University of Rome, 00185 Rome, Italy; Center for Research in Neurobiology Daniel Bovet, Sapienza University of Rome, 00185 Rome, Italy.
| | - Valentina Ferretti
- Department of Biology and Biotechnology "Charles Darwin", Sapienza University of Rome, 00185 Rome, Italy; Center for Research in Neurobiology Daniel Bovet, Sapienza University of Rome, 00185 Rome, Italy
| | - Chiara Mozzetta
- Institute of Molecular Biology and Pathology (IBPM), National Research Council (CNR) of Italy c/o Department of Biology and Biotechnology "Charles Darwin", Sapienza University of Rome, 00185 Rome, Italy
| |
Collapse
|
25
|
Waldrop MA, Moore SA, Mathews KD, Darbro BW, Medne L, Finkel R, Connolly AM, Crawford TO, Drachman D, Wein N, Habib AA, Krzesniak-Swinarska MA, Zaidman CM, Collins JJ, Jokela M, Udd B, Day JW, Ortiz-Guerrero G, Statland J, Butterfield RJ, Dunn DM, Weiss RB, Flanigan KM. Intron mutations and early transcription termination in Duchenne and Becker muscular dystrophy. Hum Mutat 2022; 43:511-528. [PMID: 35165973 PMCID: PMC9901284 DOI: 10.1002/humu.24343] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Revised: 02/05/2022] [Accepted: 02/07/2022] [Indexed: 11/11/2022]
Abstract
DMD pathogenic variants for Duchenne and Becker muscular dystrophy are detectable with high sensitivity by standard clinical exome analyses of genomic DNA. However, up to 7% of DMD mutations are deep intronic and analysis of muscle-derived RNA is an important diagnostic step for patients who have negative genomic testing but abnormal dystrophin expression in muscle. In this study, muscle biopsies were evaluated from 19 patients with clinical features of a dystrophinopathy, but negative clinical DMD mutation analysis. Reverse transcription-polymerase chain reaction or high-throughput RNA sequencing methods identified 19 mutations with one of three pathogenic pseudoexon types: deep intronic point mutations, deletions or insertions, and translocations. In association with point mutations creating intronic splice acceptor sites, we observed the first examples of DMD pseudo 3'-terminal exon mutations causing high efficiency transcription termination within introns. This connection between splicing and premature transcription termination is reminiscent of U1 snRNP-mediating telescripting in sustaining RNA polymerase II elongation across large genes, such as DMD. We propose a novel classification of three distinct types of mutations identifiable by muscle RNA analysis, each of which differ in potential treatment approaches. Recognition and appropriate characterization may lead to therapies directed toward full-length dystrophin expression for some patients.
Collapse
Affiliation(s)
- Megan A. Waldrop
- The Center for Gene Therapy, Nationwide Children’s Hospital, Columbus, OH 43205,Department of Neurology, The Ohio State University, Columbus, OH 43205,Department of Pediatrics, The Ohio State University, Columbus, OH 43205
| | - Steven A. Moore
- Department of Pathology, The University of Iowa, Iowa City, IA, 52242
| | | | | | - Livja Medne
- Children’s Hospital of Philadelphia, Philadelphia, PA 19104
| | | | - Anne M. Connolly
- Department of Neurology, Washington University, Saint Louis, MO 63110
| | | | | | - Nicolas Wein
- The Center for Gene Therapy, Nationwide Children’s Hospital, Columbus, OH 43205
| | | | | | - Craig M. Zaidman
- Department of Neurology, Washington University, Saint Louis, MO 63110
| | - James J. Collins
- Department of Pediatric Neurology, Mercy Hospitals, Springfield, MO 65804
| | - Manu Jokela
- Neuromuscular Research Center, Tampere University Hospital and University of Tampere, Tampere, Finland,Division of Clinical Neurosciences, Turku University Hospital and University of Turku, Turku, Finland
| | - Bjarne Udd
- Neuromuscular Research Center, Tampere University Hospital and University of Tampere, Tampere, Finland
| | - John W. Day
- Department of Neurology, University of Minnesota Medical Center, Minneapolis, MN 55454
| | | | - Jeff Statland
- Department of Neurology, University of Kansas, Kansas City, KS
| | - Russell J. Butterfield
- Department of Pediatrics, The University of Utah School of Medicine, Salt Lake City, UT 84112
| | - Diane M. Dunn
- Department of Human Genetics, The University of Utah School of Medicine, Salt Lake City, UT 84112
| | - Robert B. Weiss
- Department of Pediatrics, The University of Utah School of Medicine, Salt Lake City, UT 84112,Department of Human Genetics, The University of Utah School of Medicine, Salt Lake City, UT 84112
| | - Kevin M. Flanigan
- The Center for Gene Therapy, Nationwide Children’s Hospital, Columbus, OH 43205,Department of Neurology, The Ohio State University, Columbus, OH 43205,Department of Pediatrics, The Ohio State University, Columbus, OH 43205
| |
Collapse
|
26
|
Passos-Bueno MR, Costa CIS, Zatz M. Dystrophin genetic variants and autism. DISCOVER MENTAL HEALTH 2022; 2:4. [PMID: 37861890 PMCID: PMC10501027 DOI: 10.1007/s44192-022-00008-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Accepted: 02/07/2022] [Indexed: 10/21/2023]
Abstract
Loss-of-function variants in the dystrophin gene, a well-known cause of muscular dystrophies, have emerged as a mutational risk mechanism for autism spectrum disorder (ASD), which in turn is a highly prevalent (~ 1%) genetically heterogeneous neurodevelopmental disorder. Although the association of intellectual disability with the dystrophinopathies Duchenne (DMD) and Becker muscular dystrophy (BMD) has been long established, their association with ASD is more recent, and the dystrophin genotype-ASD phenotype correlation is unclear. We therefore present a review of the literature focused on the ASD prevalence among dystrophinopathies, the relevance of the dystrophin isoforms, and most particularly the relevance of the genetic background to the etiology of ASD in these patients. Four families with ASD-DMD/BMD patients are also reported here for the first time. These include a single ASD individual, ASD-discordant and ASD-concordant monozygotic twins, and non-identical ASD triplets. Notably, two unrelated individuals, which were first ascertained because of the ASD phenotype at ages 15 and 5 years respectively, present rare dystrophin variants still poorly characterized, suggesting that some dystrophin variants may compromise the brain more prominently. Whole exome sequencing in these ASD-DMD/BMD individuals together with the literature suggest, although based on preliminary data, a complex and heterogeneous genetic architecture underlying ASD in dystrophinopathies, that include rare variants of large and medium effect. The need for the establishment of a consortia for genomic investigation of ASD-DMD/BMD patients, which may shed light on the genetic architecture of ASD, is discussed.
Collapse
Affiliation(s)
- Maria Rita Passos-Bueno
- Departamento de Genética e Biologia Evolutiva, Centro de Estudos do Genoma Humano e Células-Tronco, Instituto de Biociências, Universidade de São Paulo, São Paulo, SP, Brazil.
| | - Claudia Ismania Samogy Costa
- Departamento de Genética e Biologia Evolutiva, Centro de Estudos do Genoma Humano e Células-Tronco, Instituto de Biociências, Universidade de São Paulo, São Paulo, SP, Brazil
| | - Mayana Zatz
- Departamento de Genética e Biologia Evolutiva, Centro de Estudos do Genoma Humano e Células-Tronco, Instituto de Biociências, Universidade de São Paulo, São Paulo, SP, Brazil
| |
Collapse
|
27
|
Mei H, Boom J, El Abdellaoui S, Abdelmohsen K, Munk R, Martindale JL, Kloet S, Kielbasa SM, Sharp TH, Gorospe M, Raz V. Alternative polyadenylation utilization results in ribosome assembly and mRNA translation deficiencies in a model for muscle aging. J Gerontol A Biol Sci Med Sci 2022; 77:1130-1140. [PMID: 35245938 PMCID: PMC9159670 DOI: 10.1093/gerona/glac058] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Indexed: 11/15/2022] Open
Abstract
Aging-associated muscle wasting is regulated by multiple molecular processes, whereby aberrant mRNA processing regulation induces muscle wasting. The poly(A)-binding protein nuclear 1 (PABPN1) regulates polyadenylation site (PAS) utilization, in the absence of PABPN1 the alternative PAS (APA) is utilized. Reduced PABPN1 levels induce muscle wasting where the expression of cellular processes regulating protein homeostasis, the ubiquitin-proteasome system, and translation, are robustly dysregulated. Translation is impacted by mRNA levels, but PABPN1 impact on translation is not fully understood. Here we show that a persistent reduction in PABPN1 levels led to a significant loss of translation efficiency. RNA sequencing of rRNA-depleted libraries from polysome traces revealed reduced mRNA abundance across ribosomal fractions, as well as reduced levels of small RNAs. We show that the abundance of translated mRNAs in the polysomes correlated with PAS switches at the 3'-UTR. Those mRNAs are enriched in cellular processes that are essential for proper muscle function. This study suggests that the effect of PABPN1 on translation efficiency impacts protein homeostasis in aging-associated muscle atrophy.
Collapse
Affiliation(s)
- Hailiang Mei
- Sequencing Analysis Support Core, Leiden University Medical Centre, Leiden, The Netherlands
| | - Jasper Boom
- Sequencing Analysis Support Core, Leiden University Medical Centre, Leiden, The Netherlands
| | - Salma El Abdellaoui
- Department of Human Genetics, Leiden University Medical Centre, Leiden, The Netherlands
| | - Kotb Abdelmohsen
- Laboratory of Genetics and Genomics, National Institute on Aging, National Institutes of Health, Baltimore, Maryland, USA
| | - Rachel Munk
- Laboratory of Genetics and Genomics, National Institute on Aging, National Institutes of Health, Baltimore, Maryland, USA
| | - Jennifer L Martindale
- Laboratory of Genetics and Genomics, National Institute on Aging, National Institutes of Health, Baltimore, Maryland, USA
| | - Susan Kloet
- Department of Human Genetics, Leiden University Medical Centre, Leiden, The Netherlands
| | - Szymone M Kielbasa
- Sequencing Analysis Support Core, Leiden University Medical Centre, Leiden, The Netherlands
| | - Thomas H Sharp
- Department of Cell and Chemical Biology, Leiden University Medical Centre, Leiden The Netherlands
| | - Myriam Gorospe
- Laboratory of Genetics and Genomics, National Institute on Aging, National Institutes of Health, Baltimore, Maryland, USA
| | - Vered Raz
- Department of Human Genetics, Leiden University Medical Centre, Leiden, The Netherlands
| |
Collapse
|
28
|
Xie J, Zhang X, Zheng J, Hong X, Tong X, Liu X, Xue Y, Wang X, Zhang Y, Liu S. Two novel RNA-binding proteins identification through computational prediction and experimental validation. Genomics 2021; 114:149-160. [PMID: 34921931 DOI: 10.1016/j.ygeno.2021.12.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Revised: 08/05/2021] [Accepted: 12/13/2021] [Indexed: 11/16/2022]
Abstract
Since RBPs play important roles in the cell, it's particularly important to find new RBPs. We performed iRIP-seq and CLIP-seq to verify two proteins, CLIP1 and DMD, predicted by RBPPred whether are RBPs or not. The experimental results confirm that these two proteins have RNA-binding activity. We identified significantly enriched binding motifs UGGGGAGG, CUUCCG and CCCGU for CLIP1 (iRIP-seq), DMD (iRIP-seq) and DMD (CLIP-seq), respectively. The computational KEGG and GO analysis show that the CLIP1 and DMD share some biological processes and functions. Besides, we found that the SNPs between DMD and its RNA partners may be associated with Becker muscular dystrophy, Duchenne muscular dystrophy, Dilated cardiomyopathy 3B and Cardiovascular phenotype. Among the thirteen cancers data, CLIP1 and another 300 oncogenes always co-occur, and 123 of these 300 genes interact with CLIP1. These cancers may be associated with the mutations occurred in both CLIP1 and the genes it interacts with.
Collapse
Affiliation(s)
- Juan Xie
- School of Physics, Huazhong University of Science and Technology, Wuhan, Hubei 430074, China
| | - Xiaoli Zhang
- School of Physics, Huazhong University of Science and Technology, Wuhan, Hubei 430074, China
| | - Jinfang Zheng
- School of Physics, Huazhong University of Science and Technology, Wuhan, Hubei 430074, China
| | - Xu Hong
- School of Physics, Huazhong University of Science and Technology, Wuhan, Hubei 430074, China
| | - Xiaoxue Tong
- School of Physics, Huazhong University of Science and Technology, Wuhan, Hubei 430074, China
| | - Xudong Liu
- School of Physics, Huazhong University of Science and Technology, Wuhan, Hubei 430074, China
| | - Yaqiang Xue
- Laboratory for Genome Regulation and Human Health, ABLife Inc., Wuhan, Hubei 430075, China
| | - Xuelian Wang
- ABLife BioBigData Institute, Wuhan, Hubei 430075, China
| | - Yi Zhang
- ABLife BioBigData Institute, Wuhan, Hubei 430075, China
| | - Shiyong Liu
- School of Physics, Huazhong University of Science and Technology, Wuhan, Hubei 430074, China.
| |
Collapse
|
29
|
Johnston JR, McNally EM. Genetic correction strategies for Duchenne Muscular Dystrophy and their impact on the heart. PROGRESS IN PEDIATRIC CARDIOLOGY 2021; 63. [PMID: 34898968 DOI: 10.1016/j.ppedcard.2021.101460] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Background Duchenne muscular dystrophy (DMD) is an X-linked recessive disorder with early childhood onset characterized by profound loss of muscle strength and associated cardiomyopathy. DMD affects is most often caused by deletions involving single or multiple exons that disrupt the open reading frame of the DMD gene. Mutations causing loss or premature truncation of dystrophin result in dystrophin protein deficiency, which renders the plasma membrane of skeletal myofibers and cardiomyocytes weakened. Aim of Review Genetic correction is in use to treat DMD, since several drugs have been already approved which partially restore dystrophin production through the use of antisense oligonucleotides. There are multiple ongoing clinical trials to evaluate the efficacy of treating DMD with micro-dystrophins delivered by adeno-associated viruses. Future approaches entail gene editing to target the single copy of the DMD gene on the X-chromosome. The primary, near-term goal is restoration of skeletal muscle dystrophin, and for some of these treatments, the efficacy in the heart is not fully known. Here, we discuss the anticipated cardiac outcomes of dystrophin-targeted therapies, and how this information informs genomic medicine for cardiomyopathies, especially in childhood. Key Scientific Concepts of Review Many genetic treatment strategies are being implemented to treat DMD. Since most preclinical testing has focused on skeletal muscle, there is a gap in knowledge about the expected effects of these approaches on cardiac genetic correction and cardiomyopathy progression in DMD. Additional study is needed.
Collapse
Affiliation(s)
- Jamie R Johnston
- Center for Genetic Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Elizabeth M McNally
- Center for Genetic Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| |
Collapse
|
30
|
Paredes-Redondo A, Harley P, Maniati E, Ryan D, Louzada S, Meng J, Kowala A, Fu B, Yang F, Liu P, Marino S, Pourquié O, Muntoni F, Wang J, Lieberam I, Lin YY. Optogenetic modeling of human neuromuscular circuits in Duchenne muscular dystrophy with CRISPR and pharmacological corrections. SCIENCE ADVANCES 2021; 7:eabi8787. [PMID: 34516770 PMCID: PMC8442926 DOI: 10.1126/sciadv.abi8787] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Accepted: 07/20/2021] [Indexed: 05/13/2023]
Abstract
Duchenne muscular dystrophy (DMD) is caused by dystrophin gene mutations leading to skeletal muscle weakness and wasting. Dystrophin is enriched at the neuromuscular junction (NMJ), but how NMJ abnormalities contribute to DMD pathogenesis remains unclear. Here, we combine transcriptome analysis and modeling of DMD patient-derived neuromuscular circuits with CRISPR-corrected isogenic controls in compartmentalized microdevices. We show that NMJ volumes and optogenetic motor neuron–stimulated myofiber contraction are compromised in DMD neuromuscular circuits, which can be rescued by pharmacological inhibition of TGFβ signaling, an observation validated in a 96-well human neuromuscular circuit coculture assay. These beneficial effects are associated with normalization of dysregulated gene expression in DMD myogenic transcriptomes affecting NMJ assembly (e.g., MUSK) and axon guidance (e.g., SLIT2 and SLIT3). Our study provides a new human microphysiological model for investigating NMJ defects in DMD and assessing candidate drugs and suggests that enhancing neuromuscular connectivity may be an effective therapeutic strategy.
Collapse
Affiliation(s)
- Amaia Paredes-Redondo
- Centre for Genomics and Child Health, Blizard
Institute, Barts and the London School of Medicine and Dentistry, Queen Mary
University of London, 4 Newark Street, London E1 2AT, UK
- Stem Cell Laboratory, National Bowel Research Centre,
Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen
Mary University of London, 2 Newark Street, London E1 2AT, UK
- Centre for Predictive in vitro Model, Queen Mary
University of London, Mile End Road, London E1 4NS, UK
| | - Peter Harley
- Centre for Stem Cells and Regenerative Medicine, MRC
Centre for Neurodevelopmental Disorders, and Centre for Developmental
Neurobiology, King’s College London, London, UK
| | - Eleni Maniati
- Centre for Cancer Genomics and Computational Biology,
Barts Cancer Institute, Queen Mary University of London, London, UK
| | - David Ryan
- Wellcome Sanger Institute, Wellcome Genome Campus,
Hinxton, Cambridge CB10 1SA, UK
| | - Sandra Louzada
- Wellcome Sanger Institute, Wellcome Genome Campus,
Hinxton, Cambridge CB10 1SA, UK
| | - Jinhong Meng
- UCL Great Ormond Street Institute of Child Health, 30
Guilford Street, London WC1N 1EH, UK
| | - Anna Kowala
- Centre for Genomics and Child Health, Blizard
Institute, Barts and the London School of Medicine and Dentistry, Queen Mary
University of London, 4 Newark Street, London E1 2AT, UK
- Stem Cell Laboratory, National Bowel Research Centre,
Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen
Mary University of London, 2 Newark Street, London E1 2AT, UK
- Centre for Predictive in vitro Model, Queen Mary
University of London, Mile End Road, London E1 4NS, UK
| | - Beiyuan Fu
- Wellcome Sanger Institute, Wellcome Genome Campus,
Hinxton, Cambridge CB10 1SA, UK
| | - Fengtang Yang
- Wellcome Sanger Institute, Wellcome Genome Campus,
Hinxton, Cambridge CB10 1SA, UK
| | - Pentao Liu
- School of Biomedical Sciences, Stem Cell and
Regenerative Medicine Consortium, Li Ka Shing Faculty of Medicine, The
University of Hong Kong, Hong Kong, China
| | - Silvia Marino
- Centre for Genomics and Child Health, Blizard
Institute, Barts and the London School of Medicine and Dentistry, Queen Mary
University of London, 4 Newark Street, London E1 2AT, UK
| | - Olivier Pourquié
- Department of Genetics and Department of Pathology,
Brigham and Women’s Hospital, Harvard Medical School, 60 Fenwood Road,
Boston, MA, USA
| | - Francesco Muntoni
- UCL Great Ormond Street Institute of Child Health, 30
Guilford Street, London WC1N 1EH, UK
- NIHR Biomedical Research Centre, Great Ormond
Street Hospital, Great Ormond Street, London, UK
| | - Jun Wang
- Centre for Cancer Genomics and Computational Biology,
Barts Cancer Institute, Queen Mary University of London, London, UK
| | - Ivo Lieberam
- Centre for Stem Cells and Regenerative Medicine, MRC
Centre for Neurodevelopmental Disorders, and Centre for Developmental
Neurobiology, King’s College London, London, UK
| | - Yung-Yao Lin
- Centre for Genomics and Child Health, Blizard
Institute, Barts and the London School of Medicine and Dentistry, Queen Mary
University of London, 4 Newark Street, London E1 2AT, UK
- Stem Cell Laboratory, National Bowel Research Centre,
Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen
Mary University of London, 2 Newark Street, London E1 2AT, UK
- Centre for Predictive in vitro Model, Queen Mary
University of London, Mile End Road, London E1 4NS, UK
| |
Collapse
|
31
|
Abstract
Duchenne muscular dystrophy (DMD) is a devastating, rare disease. While clinically described in the 19th century, the genetic foundation of DMD was not discovered until more than 100 years later. This genetic understanding opened the door to the development of genetic treatments for DMD. Over the course of the last 30 years, the research that supports this development has moved into the realm of clinical trials and regulatory drug approvals. Exon skipping to therapeutically restore the frame of an out-of-frame dystrophin mutation has taken center stage in drug development for DMD. The research reviewed here focuses on the clinical development of exon skipping for the treatment of DMD. In addition to the generation of clinical treatments that are being used for patient care, this research sets the stage for future therapeutic development with a focus on increasing efficacy while providing safety and addressing the multi-systemic aspects of DMD.
Collapse
Affiliation(s)
- Shin'ichi Takeda
- Honorary Director General, National Institute of Neuroscience, National Center of Neurology and Psychiatry (NCNP), Kodaira, Japan
| | - Paula R Clemens
- Professor and Vice Chair of VA Affairs, Department of Neurology, University of Pittsburgh School of Medicine, Division Chief, Neurology, Medical Service Line, VA Pittsburgh Healthcare System, Pittsburgh, PA USA
| | - Eric P Hoffman
- Professor, Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, Binghamton University - State University of New York, Binghamton, NY USA
| |
Collapse
|
32
|
Aziz MC, Schneider PN, Carvill GL. Targeting Poison Exons to Treat Developmental and Epileptic Encephalopathy. Dev Neurosci 2021; 43:241-246. [PMID: 33971653 DOI: 10.1159/000516143] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Accepted: 03/16/2021] [Indexed: 11/19/2022] Open
Abstract
Developmental and epileptic encephalopathies (DEEs) describe a subset of neurodevelopmental disorders categorized by refractory epilepsy that is often associated with intellectual disability and autism spectrum disorder. The majority of DEEs are now known to have a genetic basis with de novo coding variants accounting for the majority of cases. More recently, a small number of individuals have been identified with intronic SCN1A variants that result in alternative splicing events that lead to ectopic inclusion of poison exons (PEs). PEs are short highly conserved exons that contain a premature truncation codon, and when spliced into the transcript, lead to premature truncation and subsequent degradation by nonsense-mediated decay. The reason for the inclusion/exclusion of these PEs is not entirely clear, but research suggests an autoregulatory role in gene expression and protein abundance. This is seen in proteins such as RNA-binding proteins and serine/arginine-rich proteins. Recent studies have focused on targeting these PEs as a method for therapeutic intervention. Targeting PEs using antisense oligonucleotides (ASOs) has shown to be effective in modulating alternative splicing events by decreasing the amount of transcripts harboring PEs, thus increasing the abundance of full-length transcripts and thereby the amount of protein in haploinsufficient genes implicated in DEE. In the age of personalized medicine, cellular and animal models of the genetic epilepsies have become essential in developing and testing novel precision therapeutics, including PE-targeting ASOs in a subset of DEEs.
Collapse
Affiliation(s)
- Miriam C Aziz
- Ken and Ruth Davee Department of Neurology, Northwestern University School of Medicine, Chicago, Illinois, USA
| | - Patricia N Schneider
- Ken and Ruth Davee Department of Neurology, Northwestern University School of Medicine, Chicago, Illinois, USA.,Instituto de Ciencias Biologicas, Universidade Federal do Para, Belem, Brazil
| | - Gemma L Carvill
- Ken and Ruth Davee Department of Neurology, Northwestern University School of Medicine, Chicago, Illinois, USA.,Department of Pharmacology and Pediatrics, Northwestern University School of Medicine, Chicago, Illinois, USA
| |
Collapse
|
33
|
Abstract
Duchenne muscular dystrophy is a severe, progressive, muscle-wasting disease that leads to difficulties with movement and, eventually, to the need for assisted ventilation and premature death. The disease is caused by mutations in DMD (encoding dystrophin) that abolish the production of dystrophin in muscle. Muscles without dystrophin are more sensitive to damage, resulting in progressive loss of muscle tissue and function, in addition to cardiomyopathy. Recent studies have greatly deepened our understanding of the primary and secondary pathogenetic mechanisms. Guidelines for the multidisciplinary care for Duchenne muscular dystrophy that address obtaining a genetic diagnosis and managing the various aspects of the disease have been established. In addition, a number of therapies that aim to restore the missing dystrophin protein or address secondary pathology have received regulatory approval and many others are in clinical development.
Collapse
Affiliation(s)
- Dongsheng Duan
- Department of Molecular Microbiology and Immunology and Department of Neurology, School of Medicine; Department of Biomedical Sciences, College of Veterinary Medicine; Department of Biomedical, Biological & Chemical Engineering, College of Engineering, University of Missouri, Columbia, MO, USA
| | - Nathalie Goemans
- Department of Child Neurology, University Hospitals Leuven, Leuven, Belgium
| | | | - Eugenio Mercuri
- Centro Clinico Nemo, Policlinico Gemelli, Rome, Italy
- Peadiatric Neurology, Catholic University, Rome, Italy
| | - Annemieke Aartsma-Rus
- Department of Human Genetics, Leiden University Medical Center, Leiden, The Netherlands.
| |
Collapse
|
34
|
Schneider AFE, Aartsma-Rus A. Developments in reading frame restoring therapy approaches for Duchenne muscular dystrophy. Expert Opin Biol Ther 2020; 21:343-359. [PMID: 33074029 DOI: 10.1080/14712598.2021.1832462] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
INTRODUCTION Exon skipping compounds restoring the dystrophin transcript reading frame have received regulatory approval for Duchenne muscular dystrophy (DMD). Recently, focus shifted to developing compounds to skip additional exons, improving delivery to skeletal muscle, and to genome editing, to restore the reading frame on DNA level. AREAS COVERED We outline developments for reading frame restoring approaches, challenges of mutation specificity, and optimizing delivery. Also, we highlight ongoing efforts to better detect exon skipping therapeutic effects in clinical trials. Searches on relevant terms were performed, focusing on recent publications (<3 years). EXPERT OPINION Currently, 3 AONS are approved. Whether dystrophin levels are sufficient to slowdown disease progression needs to be confirmed. Enhancing AON uptake by muscles is currently under investigation. Gene editing is an alternative, but one that involves practical and ethical concerns. Given the field's momentum, we believe the efficiency of frame-restoring approaches will improve.
Collapse
Affiliation(s)
| | - Annemieke Aartsma-Rus
- Department of Human Genetics, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|