1
|
Ziegler M, Böger C, Alecu JE, Kim HM, Saffari A, Davies AK, Sahin M, Ebrahimi-Fakhari D. Arrayed CRISPR/Cas9 Loss-Of-Function Screen in a Neuronal Model of Adaptor Protein Complex 4 Deficiency Identifies Modulators of ATG9A Trafficking. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.22.639634. [PMID: 40027661 PMCID: PMC11870607 DOI: 10.1101/2025.02.22.639634] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/05/2025]
Abstract
Biallelic loss-of-function variants in the adaptor protein complex 4 (AP-4) disrupt trafficking of transmembrane proteins at the trans -Golgi network, including the autophagy-related protein 9A (ATG9A), leading to childhood-onset hereditary spastic paraplegia (AP-4-HSP). AP-4-HSP is characterized by features of both a neurodevelopmental and degenerative neurological disease. To investigate the molecular mechanisms underlying AP-4-HSP and identify potential therapeutic targets, we conducted an arrayed CRISPR/Cas9 loss-of-function screen of 8,478 genes, targeting the 'druggable genome', in a human neuronal model of AP-4 deficiency. Through this phenotypic screen and subsequent experiments, key modulators of ATG9A trafficking were identified, and complementary pathway analyses provided insights into the regulatory landscape of ATG9A transport. Knockdown of ANPEP and NPM1 enhanced ATG9A availability outside the trans -Golgi network, suggesting they regulate ATG9A localization. These findings deepen our understanding of ATG9A trafficking in the context of AP-4 deficiency and offer a framework for the development of targeted interventions for AP-4-HSP.
Collapse
|
2
|
Sabbagh Q, Poblete NH, Angelini C, Hersent C, Benkirane M, Pointaux M, Larrieu L, Castrioto A, Deberge L, Fluchère F, Ramond F, Lesca G, Koenig M, Goizet C. AP4B1 hypomorphic variants cause autosomal recessive adult-onset ataxia. J Neurol 2025; 272:154. [PMID: 39821477 PMCID: PMC11742001 DOI: 10.1007/s00415-025-12889-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Revised: 12/27/2024] [Accepted: 12/28/2024] [Indexed: 01/19/2025]
Affiliation(s)
- Quentin Sabbagh
- Service de Génétique Clinique, Université de Montpellier, Centre de Référence « Anomalies du Développement Et Syndromes Malformatifs », Centre Hospitalier Universitaire de Montpellier, Montpellier, France
| | - Natalia Hernandez Poblete
- Université de Bordeaux, Centre de Référence « Neurogénétique », Centre Hospitalier Universitaire de Bordeaux, Service de Génétique Médicale, Bordeaux, France
| | - Chloé Angelini
- Université de Bordeaux, Centre de Référence « Neurogénétique », Centre Hospitalier Universitaire de Bordeaux, Service de Génétique Médicale, Bordeaux, France
- Université de Bordeaux, CNRS, INCIA, UMR5287, Équipe NRGen, 33000, Bordeaux, France
| | - Clément Hersent
- Laboratoire de Génétique Moléculaire, Université de Montpellier, Institut Universitaire de Recherche Clinique, Centre Hospitalier Universitaire de Montpellier, Montpellier, France
| | - Mehdi Benkirane
- Laboratoire de Génétique Moléculaire, Université de Montpellier, Institut Universitaire de Recherche Clinique, Centre Hospitalier Universitaire de Montpellier, Montpellier, France
| | - Morgane Pointaux
- Laboratoire de Génétique Moléculaire, Université de Montpellier, Institut Universitaire de Recherche Clinique, Centre Hospitalier Universitaire de Montpellier, Montpellier, France
| | - Lise Larrieu
- Laboratoire de Génétique Moléculaire, Université de Montpellier, Institut Universitaire de Recherche Clinique, Centre Hospitalier Universitaire de Montpellier, Montpellier, France
| | - Anna Castrioto
- Département de Neurologie, Université Grenoble Alpes, Centre Hospitalier Universitaire Grenoble-Alpes, Grenoble Institut Neurosciences, Inserm U1216, 38000, Grenoble, France
| | - Louise Deberge
- Service de Médecine Physique Et de Réadaptation, Université de Bordeaux, Centre Hospitalier Universitaire de Bordeaux, Bordeaux, France
| | - Frédérique Fluchère
- Service de Neurologie Et Mouvements Anormaux, Aix-Marseille Université, Assistance Publique -Hôpitaux de Marseille, Marseille, France
| | - Francis Ramond
- Université Jean Monnet, Centre Hospitalier Universitaire de Saint-Etienne, Service de Génétique Médicale, Saint-Etienne, France
| | - Gaetan Lesca
- Département de Génétique Médicale, Université Claude Bernard Lyon 1, Centre de Compétence « Neurogénétique », Hospices Civiles de Lyon, Lyon, France
| | - Michel Koenig
- Laboratoire de Génétique Moléculaire, Université de Montpellier, Institut Universitaire de Recherche Clinique, Centre Hospitalier Universitaire de Montpellier, Montpellier, France
| | - Cyril Goizet
- Université de Bordeaux, Centre de Référence « Neurogénétique », Centre Hospitalier Universitaire de Bordeaux, Service de Génétique Médicale, Bordeaux, France.
- Université de Bordeaux, CNRS, INCIA, UMR5287, Équipe NRGen, 33000, Bordeaux, France.
| |
Collapse
|
3
|
Francisco S, Lamacchia L, Turco A, Ermondi G, Caron G, Rossi Sebastiano M. Restoring adapter protein complex 4 function with small molecules: an in silico approach to spastic paraplegia 50. Protein Sci 2025; 34:e70006. [PMID: 39723768 DOI: 10.1002/pro.70006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2024] [Revised: 11/22/2024] [Accepted: 12/06/2024] [Indexed: 12/28/2024]
Abstract
This study focuses on spastic paraplegia type 50 (SPG50), an adapter protein complex 4 deficiency syndrome caused by mutations in the adapter protein complex 4 subunit mu-1 (AP4M1) gene, and on the downstream alterations of the AP4M1 protein. We applied a battery of heterogeneous computational resources, encompassing two in-house tools described here for the first time, to (a) assess the druggability potential of AP4M1, (b) characterize SPG50-associated mutations and their 3D scenario, (c) identify mutation-tailored drug candidates for SPG50, and (d) elucidate their mechanisms of action by means of structural considerations on homology models of the adapter protein complex 4 core. Altogether, the collected results indicate R367Q as the mutation with the most promising potential of being corrected by small-molecule drugs, and the flavonoid rutin as best candidate for this purpose. Rutin shows promise in rescuing the interaction between the AP4M1 and adapter protein complex subunit beta-1 (AP4B1) subunits by means of a glue-like mode of action. Overall, this approach offers a framework that could be systematically applied to the investigation of mutation-wise molecular mechanisms in different hereditary spastic paraplegias, too.
Collapse
Affiliation(s)
- Serena Francisco
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy
| | - Lorenzo Lamacchia
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy
| | - Attilio Turco
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy
| | - Giuseppe Ermondi
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy
| | - Giulia Caron
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy
| | - Matteo Rossi Sebastiano
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy
| |
Collapse
|
4
|
Wiseman JP, Scarrott JM, Alves-Cruzeiro J, Saffari A, Böger C, Karyka E, Dawes E, Davies AK, Marchi PM, Graves E, Fernandes F, Yang ZL, Coldicott I, Hirst J, Webster CP, Highley JR, Hackett N, Angyal A, Silva TD, Higginbottom A, Shaw PJ, Ferraiuolo L, Ebrahimi-Fakhari D, Azzouz M. Pre-clinical development of AP4B1 gene replacement therapy for hereditary spastic paraplegia type 47. EMBO Mol Med 2024; 16:2882-2917. [PMID: 39358605 PMCID: PMC11554807 DOI: 10.1038/s44321-024-00148-5] [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: 03/15/2024] [Revised: 09/17/2024] [Accepted: 09/19/2024] [Indexed: 10/04/2024] Open
Abstract
Spastic paraplegia 47 (SPG47) is a neurological disorder caused by mutations in the adaptor protein complex 4 β1 subunit (AP4B1) gene leading to AP-4 complex deficiency. SPG47 is characterised by progressive spastic paraplegia, global developmental delay, intellectual disability and epilepsy. Gene therapy aimed at restoring functional AP4B1 protein levels is a rational therapeutic strategy to ameliorate the disease phenotype. Here we report that a single delivery of adeno-associated virus serotype 9 expressing hAP4B1 (AAV9/hAP4B1) into the cisterna magna leads to widespread gene transfer and restoration of various hallmarks of disease, including AP-4 cargo (ATG9A) mislocalisation, calbindin-positive spheroids in the deep cerebellar nuclei, anatomical brain defects and motor dysfunction, in an SPG47 mouse model. Furthermore, AAV9/hAP4B1-based gene therapy demonstrated a restoration of plasma neurofilament light (NfL) levels of treated mice. Encouraged by these preclinical proof-of-concept data, we conducted IND-enabling studies, including immunogenicity and GLP non-human primate (NHP) toxicology studies. Importantly, NHP safety and biodistribution study revealed no significant adverse events associated with the therapeutic intervention. These findings provide evidence of both therapeutic efficacy and safety, establishing a robust basis for the pursuit of an IND application for clinical trials targeting SPG47 patients.
Collapse
Affiliation(s)
- Jessica P Wiseman
- Sheffield Institute for Translational Neuroscience (SITraN), Division of Neuroscience, University of Sheffield, Sheffield, UK
- Neuroscience Institute, University of Sheffield, Western Bank, Sheffield, UK
| | - Joseph M Scarrott
- Sheffield Institute for Translational Neuroscience (SITraN), Division of Neuroscience, University of Sheffield, Sheffield, UK
- Gene Therapy Innovation & Manufacturing Centre (GTIMC), Division of Neuroscience, University of Sheffield, Sheffield, UK
| | - João Alves-Cruzeiro
- Sheffield Institute for Translational Neuroscience (SITraN), Division of Neuroscience, University of Sheffield, Sheffield, UK
| | - Afshin Saffari
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
- Movement Disorders Program, Department of Neurology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
- Division of Child Neurology and Inherited Metabolic Diseases, Heidelberg University Hospital, Heidelberg, Germany
| | - Cedric Böger
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
- Movement Disorders Program, Department of Neurology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Evangelia Karyka
- Sheffield Institute for Translational Neuroscience (SITraN), Division of Neuroscience, University of Sheffield, Sheffield, UK
- Gene Therapy Innovation & Manufacturing Centre (GTIMC), Division of Neuroscience, University of Sheffield, Sheffield, UK
| | - Emily Dawes
- Sheffield Institute for Translational Neuroscience (SITraN), Division of Neuroscience, University of Sheffield, Sheffield, UK
| | - Alexandra K Davies
- School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, UK
- Cambridge Institute for Medical Research, University of Cambridge, Addenbrooke's Hospital, Cambridge, UK
| | - Paolo M Marchi
- Sheffield Institute for Translational Neuroscience (SITraN), Division of Neuroscience, University of Sheffield, Sheffield, UK
| | - Emily Graves
- Sheffield Institute for Translational Neuroscience (SITraN), Division of Neuroscience, University of Sheffield, Sheffield, UK
| | - Fiona Fernandes
- Sheffield Institute for Translational Neuroscience (SITraN), Division of Neuroscience, University of Sheffield, Sheffield, UK
| | - Zih-Liang Yang
- Sheffield Institute for Translational Neuroscience (SITraN), Division of Neuroscience, University of Sheffield, Sheffield, UK
- Neuroscience Institute, University of Sheffield, Western Bank, Sheffield, UK
| | - Ian Coldicott
- Sheffield Institute for Translational Neuroscience (SITraN), Division of Neuroscience, University of Sheffield, Sheffield, UK
- Neuroscience Institute, University of Sheffield, Western Bank, Sheffield, UK
| | - Jennifer Hirst
- Cambridge Institute for Medical Research, University of Cambridge, Addenbrooke's Hospital, Cambridge, UK
| | - Christopher P Webster
- Sheffield Institute for Translational Neuroscience (SITraN), Division of Neuroscience, University of Sheffield, Sheffield, UK
- Neuroscience Institute, University of Sheffield, Western Bank, Sheffield, UK
| | - J Robin Highley
- Sheffield Institute for Translational Neuroscience (SITraN), Division of Neuroscience, University of Sheffield, Sheffield, UK
- Neuroscience Institute, University of Sheffield, Western Bank, Sheffield, UK
| | | | - Adrienn Angyal
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, UK
| | - Thushan de Silva
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, UK
| | - Adrian Higginbottom
- Sheffield Institute for Translational Neuroscience (SITraN), Division of Neuroscience, University of Sheffield, Sheffield, UK
- Neuroscience Institute, University of Sheffield, Western Bank, Sheffield, UK
| | - Pamela J Shaw
- Sheffield Institute for Translational Neuroscience (SITraN), Division of Neuroscience, University of Sheffield, Sheffield, UK
- Neuroscience Institute, University of Sheffield, Western Bank, Sheffield, UK
- Sheffield NIHR Biomedical Research Centre, Sheffield Teaching Hospitals NHS Foundation Trust, Glossop Road, Sheffield, UK
| | - Laura Ferraiuolo
- Sheffield Institute for Translational Neuroscience (SITraN), Division of Neuroscience, University of Sheffield, Sheffield, UK
| | - Darius Ebrahimi-Fakhari
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
- Movement Disorders Program, Department of Neurology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Mimoun Azzouz
- Sheffield Institute for Translational Neuroscience (SITraN), Division of Neuroscience, University of Sheffield, Sheffield, UK.
- Neuroscience Institute, University of Sheffield, Western Bank, Sheffield, UK.
- Gene Therapy Innovation & Manufacturing Centre (GTIMC), Division of Neuroscience, University of Sheffield, Sheffield, UK.
| |
Collapse
|
5
|
Jaramillo Oquendo C, Wai HA, Rich WI, Bunyan DJ, Thomas NS, Hunt D, Lord J, Douglas AGL, Baralle D. Identification of diagnostic candidates in Mendelian disorders using an RNA sequencing-centric approach. Genome Med 2024; 16:110. [PMID: 39252027 PMCID: PMC11382415 DOI: 10.1186/s13073-024-01381-w] [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: 01/15/2024] [Accepted: 08/30/2024] [Indexed: 09/11/2024] Open
Abstract
BACKGROUND RNA sequencing (RNA-seq) is increasingly being used as a complementary tool to DNA sequencing in diagnostics where DNA analysis has been uninformative. RNA-seq enables the identification of aberrant splicing and aberrant gene expression, improving the interpretation of variants of unknown significance (VUSs), and provides the opportunity to scan the transcriptome for aberrant splicing and expression in relevant genes that may be the cause of a patient's phenotype. This work aims to investigate the feasibility of generating new diagnostic candidates in patients without a previously reported VUS using an RNA-seq-centric approach. METHODS We systematically assessed the transcriptomic profiles of 86 patients with suspected Mendelian disorders, 38 of whom had no candidate sequence variant, using RNA from blood samples. Each VUS was visually inspected to search for splicing abnormalities. Once aberrant splicing was identified in cases with VUS, multiple open-source alternative splicing tools were used to investigate if they would identify what was observed in IGV. Expression outliers were detected using OUTRIDER. Diagnoses in cases without a VUS were explored using two separate strategies. RESULTS RNA-seq allowed us to assess 71% of VUSs, detecting aberrant splicing in 14/48 patients with a VUS. We identified four new diagnoses by detecting novel aberrant splicing events in patients with no candidate sequence variants from prior DNA testing (n = 32) or where the candidate VUS did not affect splicing (n = 23). An additional diagnosis was made through the detection of skewed X-inactivation. CONCLUSION This work demonstrates the utility of an RNA-centric approach in identifying novel diagnoses in patients without candidate VUSs. It underscores the utility of blood-based RNA analysis in improving diagnostic yields and highlights optimal approaches for such analyses.
Collapse
Affiliation(s)
- Carolina Jaramillo Oquendo
- Faculty of Medicine, University of Southampton, Southampton General Hospital, Tremona Road, Southampton, SO16 6YD, UK
| | - Htoo A Wai
- Faculty of Medicine, University of Southampton, Southampton General Hospital, Tremona Road, Southampton, SO16 6YD, UK
| | - Wil I Rich
- Faculty of Medicine, University of Southampton, Southampton General Hospital, Tremona Road, Southampton, SO16 6YD, UK
| | - David J Bunyan
- Wessex Genomics Laboratory Service, Salisbury District Hospital, Salisbury, UK
| | - N Simon Thomas
- Wessex Genomics Laboratory Service, Salisbury District Hospital, Salisbury, UK
| | - David Hunt
- Faculty of Medicine, University of Southampton, Southampton General Hospital, Tremona Road, Southampton, SO16 6YD, UK
- Wessex Clinical Genetics Service, University Hospital Southampton NHS Foundation Trust, Southampton, UK
| | - Jenny Lord
- Faculty of Medicine, University of Southampton, Southampton General Hospital, Tremona Road, Southampton, SO16 6YD, UK
| | - Andrew G L Douglas
- Faculty of Medicine, University of Southampton, Southampton General Hospital, Tremona Road, Southampton, SO16 6YD, UK
- Oxford Centre for Genomic Medicine, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | - Diana Baralle
- Faculty of Medicine, University of Southampton, Southampton General Hospital, Tremona Road, Southampton, SO16 6YD, UK.
- Wessex Clinical Genetics Service, University Hospital Southampton NHS Foundation Trust, Southampton, UK.
| |
Collapse
|
6
|
Xu YH, Yuan BY, Ji JL, Wu D, Zhou H, Guo YJ. Novel de novo SPAST mutation in a Han Chinese SPG4 patient: a case report. Front Genet 2024; 15:1410381. [PMID: 39139823 PMCID: PMC11319187 DOI: 10.3389/fgene.2024.1410381] [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: 04/05/2024] [Accepted: 07/12/2024] [Indexed: 08/15/2024] Open
Abstract
Spastic paraplegia type 4 (SPG4), the predominant form of Autosomal Dominant Hereditary spastic paraplegia (AD-HSP), is characterized by variants in the SPAST gene. This study reports a unique case of a late-onset SPG4 in a Han Chinese male, manifesting primarily as gait disturbances from lower extremity spasticity. Uncovered through whole-genome sequencing, a previously undocumented frameshift variant, c.1545dupA in exon 14 of the SPAST gene, was identified. Notably, this variant was absent in asymptomatic parents with confirmed paternity and maternity status, suggesting a de novo variant occurrence. This discovery emphasizes the potential of de novo variants to exhibit a late-onset pure pattern, extending the SPG4 variant spectrum, and consideration of such variants should be given in HSP patients with a negative family history.
Collapse
Affiliation(s)
- Yu-Han Xu
- School of Medicine, Southeast University, Nanjing, China
| | - Bao-Yu Yuan
- School of Medicine, Southeast University, Nanjing, China
- Department of Neurology, Affiliated Zhongda Hospital of Southeast University, Nanjing, China
| | - Jia-Le Ji
- School of Medicine, Southeast University, Nanjing, China
| | - Di Wu
- School of Medicine, Southeast University, Nanjing, China
- Department of Neurology, Affiliated Zhongda Hospital of Southeast University, Nanjing, China
| | - Hong Zhou
- School of Medicine, Southeast University, Nanjing, China
- Department of Neurology, Affiliated Zhongda Hospital of Southeast University, Nanjing, China
| | - Yi-Jing Guo
- School of Medicine, Southeast University, Nanjing, China
- Department of Neurology, Affiliated Zhongda Hospital of Southeast University, Nanjing, China
| |
Collapse
|
7
|
Dowling JJ, Pirovolakis T, Devakandan K, Stosic A, Pidsadny M, Nigro E, Sahin M, Ebrahimi-Fakhari D, Messahel S, Varadarajan G, Greenberg BM, Chen X, Minassian BA, Cohn R, Bonnemann CG, Gray SJ. AAV gene therapy for hereditary spastic paraplegia type 50: a phase 1 trial in a single patient. Nat Med 2024; 30:1882-1887. [PMID: 38942994 PMCID: PMC11271397 DOI: 10.1038/s41591-024-03078-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Accepted: 05/20/2024] [Indexed: 06/30/2024]
Abstract
There are more than 10,000 individual rare diseases and most are without therapy. Personalized genetic therapy represents one promising approach for their treatment. We present a road map for individualized treatment of an ultra-rare disease by establishing a gene replacement therapy developed for a single patient with hereditary spastic paraplegia type 50 (SPG50). Through a multicenter collaboration, an adeno-associated virus-based gene therapy product carrying the AP4M1 gene was created and successfully administered intrathecally to a 4-year-old patient within 3 years of diagnosis as part of a single-patient phase 1 trial. Primary endpoints were safety and tolerability, and secondary endpoints evaluated efficacy. At 12 months after dosing, the therapy was well tolerated. No serious adverse events were observed, with minor events, including transient neutropenia and Clostridioides difficile gastroenteritis, experienced but resolved. Preliminary efficacy measures suggest a stabilization of the disease course. Longer follow-up is needed to confirm the safety and provide additional insights on the efficacy of the therapy. Overall, this report supports the safety of gene therapy for SPG50 and provides insights into precision therapy development for rare diseases. Clinical trial registration: NCT06069687 .
Collapse
Affiliation(s)
- James J Dowling
- Precision Child Health, Hospital for Sick Children, Toronto, Ontario, Canada.
- Division of Neurology and Program for Genetics and Genome Biology, Hospital for Sick Children, Toronto, Ontario, Canada.
- Departments of Paediatrics and Molecular Genetics, University of Toronto, Toronto, Ontario, Canada.
| | | | - Keshini Devakandan
- Precision Child Health, Hospital for Sick Children, Toronto, Ontario, Canada
| | - Ana Stosic
- Precision Child Health, Hospital for Sick Children, Toronto, Ontario, Canada
- Division of Neurology and Program for Genetics and Genome Biology, Hospital for Sick Children, Toronto, Ontario, Canada
| | - Mia Pidsadny
- Precision Child Health, Hospital for Sick Children, Toronto, Ontario, Canada
| | - Elisa Nigro
- Division of Neurology and Program for Genetics and Genome Biology, Hospital for Sick Children, Toronto, Ontario, Canada
| | - Mustafa Sahin
- Department of Neurology, Boston Children's Hospital, Boston, MA, USA
| | | | - Souad Messahel
- Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Ganapathy Varadarajan
- Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Benjamin M Greenberg
- Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Xin Chen
- Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Berge A Minassian
- Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Ronald Cohn
- Precision Child Health, Hospital for Sick Children, Toronto, Ontario, Canada
- Departments of Paediatrics and Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | - Carsten G Bonnemann
- Neuromuscular & Neurogenetic Diseases of Childhood, Neurogenetics Branch (NGB), Bethesda, MD, USA
| | - Steven J Gray
- Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, USA
| |
Collapse
|
8
|
Saffari A, Brechmann B, Böger C, Saber WA, Jumo H, Whye D, Wood D, Wahlster L, Alecu JE, Ziegler M, Scheffold M, Winden K, Hubbs J, Buttermore ED, Barrett L, Borner GHH, Davies AK, Ebrahimi-Fakhari D, Sahin M. High-content screening identifies a small molecule that restores AP-4-dependent protein trafficking in neuronal models of AP-4-associated hereditary spastic paraplegia. Nat Commun 2024; 15:584. [PMID: 38233389 PMCID: PMC10794252 DOI: 10.1038/s41467-023-44264-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Accepted: 12/06/2023] [Indexed: 01/19/2024] Open
Abstract
Unbiased phenotypic screens in patient-relevant disease models offer the potential to detect therapeutic targets for rare diseases. In this study, we developed a high-throughput screening assay to identify molecules that correct aberrant protein trafficking in adapter protein complex 4 (AP-4) deficiency, a rare but prototypical form of childhood-onset hereditary spastic paraplegia characterized by mislocalization of the autophagy protein ATG9A. Using high-content microscopy and an automated image analysis pipeline, we screened a diversity library of 28,864 small molecules and identified a lead compound, BCH-HSP-C01, that restored ATG9A pathology in multiple disease models, including patient-derived fibroblasts and induced pluripotent stem cell-derived neurons. We used multiparametric orthogonal strategies and integrated transcriptomic and proteomic approaches to delineate potential mechanisms of action of BCH-HSP-C01. Our results define molecular regulators of intracellular ATG9A trafficking and characterize a lead compound for the treatment of AP-4 deficiency, providing important proof-of-concept data for future studies.
Collapse
Affiliation(s)
- Afshin Saffari
- Department of Neurology & F.M. Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, MA, 02115, USA
- Division of Child Neurology and Inherited Metabolic Diseases, Heidelberg University Hospital, Heidelberg, Germany
| | - Barbara Brechmann
- Department of Neurology & F.M. Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Cedric Böger
- Department of Neurology & F.M. Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Wardiya Afshar Saber
- Department of Neurology & F.M. Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Hellen Jumo
- Department of Neurology & F.M. Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, MA, 02115, USA
- Rosamund Stone Zander Translational Neuroscience Center, Boston Children's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Dosh Whye
- Rosamund Stone Zander Translational Neuroscience Center, Boston Children's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Delaney Wood
- Rosamund Stone Zander Translational Neuroscience Center, Boston Children's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Lara Wahlster
- Department of Hematology & Oncology, Boston Children's Hospital & Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, 02115, USA
| | - Julian E Alecu
- Department of Neurology & F.M. Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Marvin Ziegler
- Department of Neurology & F.M. Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Marlene Scheffold
- Department of Neurology & F.M. Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Kellen Winden
- Department of Neurology & F.M. Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Jed Hubbs
- Rosamund Stone Zander Translational Neuroscience Center, Boston Children's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Elizabeth D Buttermore
- Rosamund Stone Zander Translational Neuroscience Center, Boston Children's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Lee Barrett
- Rosamund Stone Zander Translational Neuroscience Center, Boston Children's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Georg H H Borner
- Department of Proteomics and Signal Transduction, Max-Planck-Institute of Biochemistry, Martinsried, 82152, Germany
| | - Alexandra K Davies
- Department of Proteomics and Signal Transduction, Max-Planck-Institute of Biochemistry, Martinsried, 82152, Germany
- School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, M13 9PT, UK
| | - Darius Ebrahimi-Fakhari
- Department of Neurology & F.M. Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, MA, 02115, USA.
- Movement Disorders Program, Department of Neurology, Boston Children's Hospital, Harvard Medical School, Boston, MA, 02115, USA.
| | - Mustafa Sahin
- Department of Neurology & F.M. Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, MA, 02115, USA
- Rosamund Stone Zander Translational Neuroscience Center, Boston Children's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| |
Collapse
|
9
|
Li Y, Zhang C, Peng G. Ap4s1 truncation leads to axonal defects in a zebrafish model of spastic paraplegia 52. Int J Dev Neurosci 2023; 83:753-764. [PMID: 37767851 DOI: 10.1002/jdn.10303] [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: 04/17/2023] [Revised: 08/11/2023] [Accepted: 09/08/2023] [Indexed: 09/29/2023] Open
Abstract
Biallelic mutations in AP4S1, the σ4 subunit of the adaptor protein complex 4 (AP-4), lead to autosomal recessive spastic paraplegia 52 (SPG52). It is a subtype of AP-4-associated hereditary spastic paraplegia (AP-4-HSP), a complex childhood-onset neurogenetic disease characterized by progressive spastic paraplegia of the lower limbs. This disease has so far lacked effective treatment, in part due to a lack of suitable animal models. Here, we used CRISPR/Cas9 technology to generate a truncation mutation in the ap4s1 gene in zebrafish. The ap4s1 truncation led to motor impairment, delayed neurodevelopment, and distal axonal degeneration. This animal model is useful for further research into AP-4 and AP-4-HSP.
Collapse
Affiliation(s)
- Yiduo Li
- State Key Laboratory of Medical Neurobiology, Ministry of Education Frontiers Center for Brain Science, and Institutes of Brain Science, Fudan University, Shanghai, China
| | - Cuizhen Zhang
- State Key Laboratory of Medical Neurobiology, Ministry of Education Frontiers Center for Brain Science, and Institutes of Brain Science, Fudan University, Shanghai, China
| | - Gang Peng
- State Key Laboratory of Medical Neurobiology, Ministry of Education Frontiers Center for Brain Science, and Institutes of Brain Science, Fudan University, Shanghai, China
| |
Collapse
|
10
|
Alecu JE, Saffari A, Ziegler M, Jordan C, Tam A, Kim S, Leung E, Szczaluba K, Mierzewska H, King SD, Santorelli FM, Yoon G, Trombetta B, Kivisäkk P, Zhang B, Sahin M, Ebrahimi-Fakhari D. Plasma Neurofilament Light Chain Is Elevated in Adaptor Protein Complex 4-Related Hereditary Spastic Paraplegia. Mov Disord 2023; 38:1742-1750. [PMID: 37482941 PMCID: PMC10529494 DOI: 10.1002/mds.29524] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2023] [Revised: 05/15/2023] [Accepted: 06/09/2023] [Indexed: 07/25/2023] Open
Abstract
BACKGROUND Adaptor protein complex 4-associated hereditary spastic paraplegia (AP-4-HSP) is caused by pathogenic biallelic variants in AP4B1, AP4M1, AP4E1, and AP4S1. OBJECTIVE The aim was to explore blood markers of neuroaxonal damage in AP-4-HSP. METHODS Plasma neurofilament light chain (pNfL) and glial fibrillary acidic protein (GFAP) levels were measured in samples from patients and age- and sex-matched controls (NfL: n = 46 vs. n = 46; GFAP: n = 14 vs. n = 21) using single-molecule array assays. Patients' phenotypes were systematically assessed using the AP-4-HSP natural history study questionnaires, the Spastic Paraplegia Rating Scale, and the SPATAX disability score. RESULTS pNfL levels increased in AP-4-HSP patients, allowing differentiation from controls (Mann-Whitney U test: P = 3.0e-10; area under the curve = 0.87 with a 95% confidence interval of 0.80-0.94). Phenotypic cluster analyses revealed a subgroup of individuals with severe generalized-onset seizures and developmental stagnation, who showed differentially higher pNfL levels (Mann-Whitney U test between two identified clusters: P = 2.5e-6). Plasma GFAP levels were unchanged in patients with AP-4-HSP. CONCLUSIONS pNfL is a potential disease marker in AP-4-HSP and can help differentiate between phenotypic subgroups. © 2023 International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Julian E. Alecu
- Department of Neurology and F.M. Kirby Neurobiology Center, Boston Children’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Friedrich-Alexander-University Erlangen-Nuremberg, Erlangen, Germany
| | - Afshin Saffari
- Department of Neurology and F.M. Kirby Neurobiology Center, Boston Children’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Marvin Ziegler
- Department of Neurology and F.M. Kirby Neurobiology Center, Boston Children’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Catherine Jordan
- Department of Neurology and F.M. Kirby Neurobiology Center, Boston Children’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Amy Tam
- Department of Neurology and F.M. Kirby Neurobiology Center, Boston Children’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Soyoung Kim
- Sozialpaediatrisches Zentrum Frankfurt Mitte, Frankfurt am Main, Germany
| | - Edward Leung
- Department of Pediatrics and Child Health, University of Manitoba, Winnipeg, Manitoba, Canada
| | | | - Hanna Mierzewska
- Department of Neurology, Institute of Mother and Child, Warsaw, Poland
| | - Staci D. King
- Department of Neurology, Texas Children’s Hospital, Houston, Texas, USA
| | | | - Grace Yoon
- Divisions of Clinical and Metabolic Genetics and Neurology, Department of Pediatrics, The Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada
| | - Bianca Trombetta
- Alzheimer’s Clinical and Translational Research Unit, Department of Neurology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Pia Kivisäkk
- Alzheimer’s Clinical and Translational Research Unit, Department of Neurology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Bo Zhang
- Department of Neurology and F.M. Kirby Neurobiology Center, Boston Children’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
- ICCTR Biostatistics and Research Design Center, Boston Children’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Mustafa Sahin
- Department of Neurology and F.M. Kirby Neurobiology Center, Boston Children’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Rosamund Stone Zander Translational Neuroscience Center, Boston Children’s Hospital, Boston, Massachusetts, USA
- Intellectual and Developmental Disabilities Research Center, Boston Children’s Hospital, Boston, Massachusetts, USA
| | - Darius Ebrahimi-Fakhari
- Department of Neurology and F.M. Kirby Neurobiology Center, Boston Children’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Rosamund Stone Zander Translational Neuroscience Center, Boston Children’s Hospital, Boston, Massachusetts, USA
- Intellectual and Developmental Disabilities Research Center, Boston Children’s Hospital, Boston, Massachusetts, USA
- Movement Disorders Program, Department of Neurology, Boston Children’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
11
|
Chen X, Dong T, Hu Y, De Pace R, Mattera R, Eberhardt K, Ziegler M, Pirovolakis T, Sahin M, Bonifacino JS, Ebrahimi-Fakhari D, Gray SJ. Intrathecal AAV9/AP4M1 gene therapy for hereditary spastic paraplegia 50 shows safety and efficacy in preclinical studies. J Clin Invest 2023; 133:e164575. [PMID: 36951961 PMCID: PMC10178841 DOI: 10.1172/jci164575] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Accepted: 03/14/2023] [Indexed: 03/24/2023] Open
Abstract
Spastic paraplegia 50 (SPG50) is an ultrarare childhood-onset neurological disorder caused by biallelic loss-of-function variants in the AP4M1 gene. SPG50 is characterized by progressive spastic paraplegia, global developmental delay, and subsequent intellectual disability, secondary microcephaly, and epilepsy. We preformed preclinical studies evaluating an adeno-associated virus (AAV)/AP4M1 gene therapy for SPG50 and describe in vitro studies that demonstrate transduction of patient-derived fibroblasts with AAV2/AP4M1, resulting in phenotypic rescue. To evaluate efficacy in vivo, Ap4m1-KO mice were intrathecally (i.t.) injected with 5 × 1011, 2.5 × 1011, or 1.25 × 1011 vector genome (vg) doses of AAV9/AP4M1 at P7-P10 or P90. Age- and dose-dependent effects were observed, with early intervention and higher doses achieving the best therapeutic benefits. In parallel, three toxicology studies in WT mice, rats, and nonhuman primates (NHPs) demonstrated that AAV9/AP4M1 had an acceptable safety profile up to a target human dose of 1 × 1015 vg. Of note, similar degrees of minimal-to-mild dorsal root ganglia (DRG) toxicity were observed in both rats and NHPs, supporting the use of rats to monitor DRG toxicity in future i.t. AAV studies. These preclinical results identify an acceptably safe and efficacious dose of i.t.-administered AAV9/AP4M1, supporting an investigational gene transfer clinical trial to treat SPG50.
Collapse
Affiliation(s)
- Xin Chen
- Department of Pediatrics, UT Southwestern Medical Center, Dallas, Texas, USA
| | - Thomas Dong
- Department of Pediatrics, UT Southwestern Medical Center, Dallas, Texas, USA
| | - Yuhui Hu
- Department of Pediatrics, UT Southwestern Medical Center, Dallas, Texas, USA
| | - Raffaella De Pace
- Neurosciences and Cellular and Structural Biology Division, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, Maryland, USA
| | - Rafael Mattera
- Neurosciences and Cellular and Structural Biology Division, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, Maryland, USA
| | - Kathrin Eberhardt
- Department of Neurology and F.M. Kirby Neurobiology Center, Boston Children’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Marvin Ziegler
- Department of Neurology and F.M. Kirby Neurobiology Center, Boston Children’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | | | - Mustafa Sahin
- Department of Neurology and F.M. Kirby Neurobiology Center, Boston Children’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Juan S. Bonifacino
- Neurosciences and Cellular and Structural Biology Division, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, Maryland, USA
| | - Darius Ebrahimi-Fakhari
- Department of Neurology and F.M. Kirby Neurobiology Center, Boston Children’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Steven J. Gray
- Department of Pediatrics, UT Southwestern Medical Center, Dallas, Texas, USA
| |
Collapse
|
12
|
Chu ECP, Huang KHK, Cheng K, Chu VKY, Lin A. Chiropractic Care in a Patient With Hereditary Spastic Paraplegia and Chronic Pain. Cureus 2023; 15:e36602. [PMID: 36968681 PMCID: PMC10037221 DOI: 10.7759/cureus.36602] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/23/2023] [Indexed: 03/25/2023] Open
Abstract
Hereditary spastic paraplegia (HSP) is a rare neurodegenerative disorder, which is challenging to diagnose and is known to be associated with more than 73 genes. Neurodegenerative disorders are characterized by progressive spasticity and weakness of the lower limbs. Here, we report the case of a 13-year-old girl with a history of HSP who presented to a chiropractic clinic with chronic low back pain and rehabilitation for lower extremity weakness. She had been receiving non-steroidal anti-inflammatory drugs and baclofen for spasticity. Full-spine radiography revealed borderline acetabular dysplasia in the right hip. After nine months of chiropractic therapy, the patient reported reduced lower extremity spasticity and pain as well as improved strength and functionality. As non-invasive therapies have minimal side effects, chiropractic therapy can be used alongside or in combination with other treatments as an additional option for the long-term management of HSP.
Collapse
Affiliation(s)
- Eric Chun-Pu Chu
- New York Medical Group (NYMG) Chiropractic Department, EC Healthcare, Hong Kong, HKG
| | - Kevin Hsu Kai Huang
- New York Medical Group (NYMG) Chiropractic Department, EC Healthcare, Hong Kong, HKG
| | - Kenny Cheng
- New York Medical Group (NYMG) Chiropractic Department, EC Healthcare, Hong Kong, HKG
| | - Valerie Kok Yan Chu
- New York Medical Group (NYMG) Chiropractic Department, EC Healthcare, Hong Kong, HKG
| | - Andy Lin
- New York Medical Group (NYMG) Chiropractic Department, EC Healthcare, Hong Kong, HKG
| |
Collapse
|
13
|
Becker A, Felici C, Lambert L, de Saint Martin A, Abi-Warde MT, Schaefer E, Zix C, Zamani M, Sadeghian S, Zeighami J, Seifi T, Azizimalamiri R, Shariati G, Galehdari H, Selig M, Ding C, Duerinckx S, Pirson I, Abramowicz M, Clément G, Leheup B, Jonveaux P, Lefort G, Bronner M, Renaud M, Bonnet C. Putative founder effect of Arg338* AP4M1 (SPG50) variant causing severe intellectual disability, epilepsy and spastic paraplegia: Report of three families. Clin Genet 2023; 103:346-351. [PMID: 36371792 PMCID: PMC10108251 DOI: 10.1111/cge.14264] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Revised: 10/21/2022] [Accepted: 11/06/2022] [Indexed: 11/14/2022]
Abstract
Bi-allelic variants affecting one of the four genes encoding the AP4 subunits are responsible for the "AP4 deficiency syndrome." Core features include hypotonia that progresses to hypertonia and spastic paraplegia, intellectual disability, postnatal microcephaly, epilepsy, and neuroimaging features. Namely, AP4M1 (SPG50) is involved in autosomal recessive spastic paraplegia 50 (MIM#612936). We report on three patients with core features from three unrelated consanguineous families originating from the Middle East. Exome sequencing identified the same homozygous nonsense variant: NM_004722.4(AP4M1):c.1012C>T p.Arg338* (rs146262009). So far, four patients from three other families carrying this homozygous variant have been reported worldwide. We describe their phenotype and compare it to the phenotype of patients with other variants in AP4M1. We construct a shared single-nucleotide polymorphism (SNP) haplotype around AP4M1 in four families and suggest a probable founder effect of Arg338* AP4M1 variant with a common ancestor most likely of Turkish origin.
Collapse
Affiliation(s)
- Aurélie Becker
- CHRU de Nancy, Laboratoire de Génétique, Inserm U1256, Université de Lorraine, Nancy, France
| | - Charlotte Felici
- CHRU de Nancy, Laboratoire de Génétique, Inserm U1256, Université de Lorraine, Nancy, France
| | - Laëtitia Lambert
- CHRU de Nancy, Pôle Enfants, Service de Génétique Clinique, Nancy, France
- Inserm U1256, Université de Lorraine, Nancy, France
| | - Anne de Saint Martin
- Hôpital Hautepierre, Centre de référence des épilepsies rares, Strasbourg, France
| | | | - Elise Schaefer
- HôpService de Génétique médicale, institut de Génétique médicale d'Alsace, CHU Strasbourg, Strasbourg, France
| | | | - Mina Zamani
- Department of Biology, Faculty of Science, Shahid Chamran University of Ahvaz, Ahvaz, Iran
- Narges Medical Genetics and Prenatal Diagnosis Laboratory, Ahvaz, Iran
| | - Saeid Sadeghian
- Department of Pediatric Neurology, Golestan Medical, Educational, and Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Jawaher Zeighami
- Narges Medical Genetics and Prenatal Diagnosis Laboratory, Ahvaz, Iran
| | - Tahereh Seifi
- Department of Biology, Faculty of Science, Shahid Chamran University of Ahvaz, Ahvaz, Iran
- Narges Medical Genetics and Prenatal Diagnosis Laboratory, Ahvaz, Iran
| | - Reza Azizimalamiri
- Department of Pediatric Neurology, Golestan Medical, Educational, and Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Gholamreza Shariati
- Narges Medical Genetics and Prenatal Diagnosis Laboratory, Ahvaz, Iran
- Department of Medical Genetics, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Hamid Galehdari
- Department of Biology, Faculty of Science, Shahid Chamran University of Ahvaz, Ahvaz, Iran
| | - Mareike Selig
- Institute of Human Genetics, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Can Ding
- Institute of Human Genetics, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | | | | | - Marc Abramowicz
- IRIBHM, Université Libre de Bruxelles, Brussels, Belgium
- Genetic Medicine and Development, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Guillemette Clément
- Inserm U1256, Université de Lorraine, Nancy, France
- CHRU de Nancy, Service de neurologie, Nancy, France
| | - Bruno Leheup
- CHRU de Nancy, Pôle Enfants, Service de Génétique Clinique, Nancy, France
- Inserm U1256, Université de Lorraine, Nancy, France
| | - Philippe Jonveaux
- CHRU de Nancy, Laboratoire de Génétique, Inserm U1256, Université de Lorraine, Nancy, France
| | - Geneviève Lefort
- CHRU de Nancy, Laboratoire de Génétique, Inserm U1256, Université de Lorraine, Nancy, France
| | - Myriam Bronner
- CHRU de Nancy, Laboratoire de Génétique, Inserm U1256, Université de Lorraine, Nancy, France
| | - Mathilde Renaud
- CHRU de Nancy, Pôle Enfants, Service de Génétique Clinique, Nancy, France
- Inserm U1256, Université de Lorraine, Nancy, France
| | - Céline Bonnet
- CHRU de Nancy, Laboratoire de Génétique, Inserm U1256, Université de Lorraine, Nancy, France
| |
Collapse
|
14
|
Mercan S, Akcakaya NH, Salman B, Yapici Z, Ozbek U, Ugur Iseri SA. Clinical and genetic analyses in syndromic intellectual disability with primary microcephaly reveal biallelic and de novo variants in patients with parental consanguinity. Genes Genomics 2023; 45:13-21. [PMID: 36371492 DOI: 10.1007/s13258-022-01344-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Accepted: 11/06/2022] [Indexed: 11/13/2022]
Abstract
BACKGROUND Syndromic intellectual disability (ID) with accompanying primary microcephaly is a group of rare neurodevelopmental disorders exhibiting extreme genetic and clinical heterogeneity. This layered heterogeneity can partially be resolved by unbiased genetic approaches targeting the genome with next generation sequencing (NGS) technologies, including exome sequencing (ES). OBJECTIVE This study was performed to dissect the clinical and genetic features in five distinct IDM cases. METHODS Singleton or trio ES approach followed by in-depth variant analysis using alternative inheritance models was performed. RESULTS We have identified biallelic loss of function variants in genes WDR62 and AP4M1 in three families, together with de novo missense variants in genes SOX11 and TRIO in two families. ES based haplotype analysis in two cases upon identification of an identical WDR62 variant in the homozygous state in two cases was suggestive of a small shared haplotype of 0.1 Mb. Additionally, we have shown a paternal origin for the de novo variant in TRIO via a polymorphic tag SNP, which enlightens the mutational mechanism for this variant. CONCLUSION In populations with high parental consanguinity, an autosomal recessive inheritance pattern for data analysis is usually the most obvious choice. Therefore, heterozygous variants may be overlooked in standard NGS analyses in consanguineous families. Our findings underlie the importance of using multiple inheritance models in NGS data analysis.
Collapse
Affiliation(s)
- Sevcan Mercan
- Department of Genetics, Aziz Sancar Institute of Experimental Medicine, Istanbul University, Vakif Gureba Cad., Fatih, 34093, Istanbul, Turkey.,Graduate School of Health Sciences, Istanbul University, Istanbul, Turkey.,Department of Bioengineering, Faculty of Engineering and Architecture, Kafkas University, Kars, Turkey
| | - Nihan Hande Akcakaya
- Department of Genetics, Aziz Sancar Institute of Experimental Medicine, Istanbul University, Vakif Gureba Cad., Fatih, 34093, Istanbul, Turkey.,Graduate School of Health Sciences, Istanbul University, Istanbul, Turkey.,Department of Neurology, Faculty of Medicine, Demiroglu Bilim University, Istanbul, Turkey.,Spastic Children's Foundation of Turkey, Istanbul, Turkey
| | - Baris Salman
- Department of Genetics, Aziz Sancar Institute of Experimental Medicine, Istanbul University, Vakif Gureba Cad., Fatih, 34093, Istanbul, Turkey.,Graduate School of Health Sciences, Istanbul University, Istanbul, Turkey
| | - Zuhal Yapici
- Department of Neurology, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Turkey
| | - Ugur Ozbek
- Department of Medical Genetics, Faculty of Medicine, Acıbadem Mehmet Ali Aydinlar University, Istanbul, Turkey
| | - Sibel Aylin Ugur Iseri
- Department of Genetics, Aziz Sancar Institute of Experimental Medicine, Istanbul University, Vakif Gureba Cad., Fatih, 34093, Istanbul, Turkey.
| |
Collapse
|
15
|
Yu W, He J, Liu X, Wu J, Cai X, Zhang Y, Liu X, Fan D. Clinical features and genetic spectrum of Chinese patients with hereditary spastic paraplegia: A 14-year study. Front Genet 2023; 14:1085442. [PMID: 36923789 PMCID: PMC10008886 DOI: 10.3389/fgene.2023.1085442] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Accepted: 02/13/2023] [Indexed: 03/01/2023] Open
Abstract
Background: Hereditary spastic paraplegia (HSP) constitutes a group of clinically and genetically rare neurodegenerative diseases characterized by progressive corticospinal tract degeneration. The phenotypes and genotypes of HSP are still expanding. In this study, we aimed to analyse the differential diagnosis, clinical features, and genetic distributions of a Chinese HSP patients in a 14-year cohort and to improve our understanding of the disease. Methods: The clinical data of patients with a primary diagnosis of HSP at the initial visit to the Department of the Neurology, Peking University Third Hospital, from 2008 to 2022 were retrospectively collected. Next-generation sequencing gene panels (NGS) combined with a multiplex ligation-amplification assay (MLPA) were conducted. Epidemiological and clinical features and candidate variants in HSP-related genes were analyzed and summarized. Results: 54 cases (probands from 25 different pedigrees and 29 sporadic cases) from 95 patients with a primary diagnosis of HSP were finally confirmed to have a clinical diagnosis of HSP based on clinical criteria, including their clinical findings, family history and long-term follow-up. Earlier disease onset was associated with longer diagnostic delay and longer disease duration and was associated with a lower risk of loss of ability to walk independently. In addition, 20 candidate variants in reported HSP-related genes were identified in these clinically diagnosed HSP patients, including variants in SPAST, ALT1, WASHC5, SPG11, B4GALNT1, and REEP1. The genetic diagnostic rate in these 54 patients was 35.18%. Conclusion: Hereditary spastic paraplegia has high clinical and genetic heterogeneity and is prone to misdiagnosis. Long-term follow-up and genetic testing can partially assist in diagnosing HSP. Our study summarized the clinical features of Chinese HSP patients in a 14-year cohort, expanded the genotype spectrum, and improved our understanding of the disease.
Collapse
Affiliation(s)
- Weiyi Yu
- Department of Neurology, Peking University Third Hospital, Beijing, China.,Beijing Municipal Key Laboratory of Biomarker and Translational Research in Neurodegenerative Diseases, Beijing, China.,Key Laboratory for Neuroscience, National Health Commission, Ministry of Education, Peking University, Beijing, China
| | - Ji He
- Department of Neurology, Peking University Third Hospital, Beijing, China.,Beijing Municipal Key Laboratory of Biomarker and Translational Research in Neurodegenerative Diseases, Beijing, China.,Key Laboratory for Neuroscience, National Health Commission, Ministry of Education, Peking University, Beijing, China
| | - Xiangyi Liu
- Department of Neurology, Peking University Third Hospital, Beijing, China.,Beijing Municipal Key Laboratory of Biomarker and Translational Research in Neurodegenerative Diseases, Beijing, China.,Key Laboratory for Neuroscience, National Health Commission, Ministry of Education, Peking University, Beijing, China
| | - Jieying Wu
- Department of Neurology, Peking University Third Hospital, Beijing, China.,Beijing Municipal Key Laboratory of Biomarker and Translational Research in Neurodegenerative Diseases, Beijing, China.,Key Laboratory for Neuroscience, National Health Commission, Ministry of Education, Peking University, Beijing, China
| | - Xiying Cai
- School of Basic Medical Sciences, Peking University, Beijing, China
| | - Yingshuang Zhang
- Department of Neurology, Peking University Third Hospital, Beijing, China.,Beijing Municipal Key Laboratory of Biomarker and Translational Research in Neurodegenerative Diseases, Beijing, China.,Key Laboratory for Neuroscience, National Health Commission, Ministry of Education, Peking University, Beijing, China
| | - Xiaoxuan Liu
- Department of Neurology, Peking University Third Hospital, Beijing, China.,Beijing Municipal Key Laboratory of Biomarker and Translational Research in Neurodegenerative Diseases, Beijing, China.,Key Laboratory for Neuroscience, National Health Commission, Ministry of Education, Peking University, Beijing, China
| | - Dongsheng Fan
- Department of Neurology, Peking University Third Hospital, Beijing, China.,Beijing Municipal Key Laboratory of Biomarker and Translational Research in Neurodegenerative Diseases, Beijing, China.,Key Laboratory for Neuroscience, National Health Commission, Ministry of Education, Peking University, Beijing, China
| |
Collapse
|
16
|
Wang YM, Ye LQ, Wang MS, Zhang JJ, Khederzadeh S, Irwin DM, Ren XD, Zhang YP, Wu DD. Unveiling the functional and evolutionary landscape of RNA editing in chicken using genomics and transcriptomics. Zool Res 2022; 43:1011-1022. [PMID: 36266925 PMCID: PMC9700494 DOI: 10.24272/j.issn.2095-8137.2022.331] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Accepted: 10/17/2022] [Indexed: 09/10/2024] Open
Abstract
The evolutionary and functional features of RNA editing are well studied in mammals, cephalopods, and insects, but not in birds. Here, we integrated transcriptomic and whole-genomic analyses to exhaustively characterize the expansive repertoire of adenosine-to-inosine (A-to-I) RNA editing sites (RESs) in the chicken. In addition, we investigated the evolutionary status of the chicken editome as a potential mechanism of domestication. We detected the lowest editing level in the liver of chickens, compared to muscles in humans, and found higher editing activity and specificity in the brain than in non-neural tissues, consistent with the brain's functional complexity. To a certain extent, specific editing activity may account for the specific functions of tissues. Our results also revealed that sequences critical to RES secondary structures remained conserved within avian evolution. Furthermore, the RNA editome was shaped by purifying selection during chicken domestication and most RESs may have served as a selection pool for a few functional RESs involved in chicken domestication, including evolution of nervous and immune systems. Regulation of RNA editing in chickens by adenosine deaminase acting on RNA (ADAR) enzymes may be affected by non-ADAR factors whose expression levels changed widely after ADAR knockdown. Collectively, we provide comprehensive lists of candidate RESs and non-ADAR-editing regulators in the chicken, thus contributing to our current understanding of the functions and evolution of RNA editing in animals.
Collapse
Affiliation(s)
- Yun-Mei Wang
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, 650223, China
- Kunming College of Life Science, University of the Chinese Academy of Sciences, Kunming, Yunnan, 650223, China
| | - Ling-Qun Ye
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, 650223, China
- Kunming College of Life Science, University of the Chinese Academy of Sciences, Kunming, Yunnan, 650223, China
| | - Ming-Shan Wang
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, 650223, China
- Kunming College of Life Science, University of the Chinese Academy of Sciences, Kunming, Yunnan, 650223, China
- Department of Ecology and Evolutionary Biology, Howard Hughes Medical Institute, University of California Santa Cruz, Santa Cruz, CA 95064, USA
| | - Jin-Jin Zhang
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, 650223, China
- Kunming College of Life Science, University of the Chinese Academy of Sciences, Kunming, Yunnan, 650223, China
| | - Saber Khederzadeh
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, 650223, China
- Kunming College of Life Science, University of the Chinese Academy of Sciences, Kunming, Yunnan, 650223, China
| | - David M Irwin
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| | - Xiao-Die Ren
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, 650223, China
- Kunming College of Life Science, University of the Chinese Academy of Sciences, Kunming, Yunnan, 650223, China
| | - Ya-Ping Zhang
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, 650223, China
- Center for Excellence in Animal Evolution and Genetics, Chinese Academy of Sciences, Kunming, Yunnan, 650223, China
- Kunming College of Life Science, University of the Chinese Academy of Sciences, Kunming, Yunnan, 650223, China
- KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650201, China. E-mail:
| | - Dong-Dong Wu
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, 650223, China
- Center for Excellence in Animal Evolution and Genetics, Chinese Academy of Sciences, Kunming, Yunnan, 650223, China
- Kunming College of Life Science, University of the Chinese Academy of Sciences, Kunming, Yunnan, 650223, China
- KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650201, China
- Kunming Natural History Museum of Zoology, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650223, China. E-mail:
| |
Collapse
|
17
|
Crook OM, Davies CTR, Breckels LM, Christopher JA, Gatto L, Kirk PDW, Lilley KS. Inferring differential subcellular localisation in comparative spatial proteomics using BANDLE. Nat Commun 2022; 13:5948. [PMID: 36216816 PMCID: PMC9550814 DOI: 10.1038/s41467-022-33570-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Accepted: 09/20/2022] [Indexed: 11/08/2022] Open
Abstract
The steady-state localisation of proteins provides vital insight into their function. These localisations are context specific with proteins translocating between different subcellular niches upon perturbation of the subcellular environment. Differential localisation, that is a change in the steady-state subcellular location of a protein, provides a step towards mechanistic insight of subcellular protein dynamics. High-accuracy high-throughput mass spectrometry-based methods now exist to map the steady-state localisation and re-localisation of proteins. Here, we describe a principled Bayesian approach, BANDLE, that uses these data to compute the probability that a protein differentially localises upon cellular perturbation. Extensive simulation studies demonstrate that BANDLE reduces the number of both type I and type II errors compared to existing approaches. Application of BANDLE to several datasets recovers well-studied translocations. In an application to cytomegalovirus infection, we obtain insights into the rewiring of the host proteome. Integration of other high-throughput datasets allows us to provide the functional context of these data.
Collapse
Affiliation(s)
- Oliver M Crook
- Cambridge Centre for Proteomics, Department of Biochemistry, University of Cambridge, CB2 1GA, Cambridge, UK.
- MRC Biostatistics Unit, School of Clinical Medicine, University of Cambridge, Cambridge, UK.
- Milner Therapeutics Institute, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge, CB2 0AW, UK.
| | - Colin T R Davies
- Cambridge Centre for Proteomics, Department of Biochemistry, University of Cambridge, CB2 1GA, Cambridge, UK
- Milner Therapeutics Institute, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge, CB2 0AW, UK
- Mechanistic Biology and Profiling, Discovery Sciences, R&D, AstraZeneca, Cambridge, UK
| | - Lisa M Breckels
- Cambridge Centre for Proteomics, Department of Biochemistry, University of Cambridge, CB2 1GA, Cambridge, UK
- Milner Therapeutics Institute, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge, CB2 0AW, UK
| | - Josie A Christopher
- Cambridge Centre for Proteomics, Department of Biochemistry, University of Cambridge, CB2 1GA, Cambridge, UK
- Milner Therapeutics Institute, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge, CB2 0AW, UK
| | - Laurent Gatto
- de Duve Institute, Université catholique de Louvain, Avenue Hippocrate 75, 1200, Brussels, Belgium
| | - Paul D W Kirk
- MRC Biostatistics Unit, School of Clinical Medicine, University of Cambridge, Cambridge, UK
- Cambridge Institute of Therapeutic Immunology & Infectious Disease (CITIID), Jeffrey Cheah Biomedical Centre, Cambridge Biomedical Campus, University of Cambridge, Cambridge, UK
| | - Kathryn S Lilley
- Cambridge Centre for Proteomics, Department of Biochemistry, University of Cambridge, CB2 1GA, Cambridge, UK.
- Milner Therapeutics Institute, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge, CB2 0AW, UK.
| |
Collapse
|
18
|
Utz KS, Kohl Z, Marterstock DC, Doerfler A, Winkler J, Schmidt M, Regensburger M. Neuropsychology and MRI correlates of neurodegeneration in SPG11 hereditary spastic paraplegia. Orphanet J Rare Dis 2022; 17:301. [PMID: 35906604 PMCID: PMC9336101 DOI: 10.1186/s13023-022-02451-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Accepted: 07/17/2022] [Indexed: 11/30/2022] Open
Abstract
Background SPG11-linked hereditary spastic paraplegia is characterized by multisystem neurodegeneration leading to a complex clinical and yet incurable phenotype of progressive spasticity and weakness. Severe cognitive symptoms are present in the majority of SPG11 patients, but a systematic and multidimensional analysis of the neuropsychological phenotype in a larger cohort is lacking. While thinning of the corpus callosum is a well-known structural hallmark observed in SPG11 patients, the neuroanatomical pattern of cortical degeneration is less understood. We here aimed to integrate neuropsychological and brain morphometric measures in SPG11. Methods We examined the neuropsychological profile in 16 SPG11 patients using a defined neuropsychological testing battery. Long-term follow up testing was performed in 7 patients. Cortical and subcortical degeneration was analyzed using an approved, artificial intelligence based magnetic resonance imaging brain morphometry, comparing patients to established reference values and to matched controls. Results In SPG11 patients, verbal fluency and memory as well as frontal-executive functions were severely impaired. Later disease stages were associated with a global pattern of impairments. Interestingly, reaction times correlated significantly with disease progression. Brain morphometry showed a significant reduction of cortical and subcortical parenchymal volume following a rostro-caudal gradient in SPG11. Whereas performance in memory tasks correlated with white matter damage, verbal fluency measures showed strong associations with frontal and parietal cortical volumes.
Conclusions The present data will help define neuropsychological and imaging read out parameters in early as well as in advanced clinical stages for future interventional trials in SPG11. Supplementary Information The online version contains supplementary material available at 10.1186/s13023-022-02451-1.
Collapse
Affiliation(s)
- Kathrin S Utz
- Department of Neurology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Zacharias Kohl
- Department of Molecular Neurology, FAU, Schwabachanlage 6, 91054, Erlangen, Germany.,Center for Rare Diseases (ZSEER), University Hospital Erlangen, Erlangen, Germany.,Department of Neurology, University of Regensburg, Regensburg, Germany
| | | | - Arnd Doerfler
- Department of Neuroradiology, FAU, Erlangen, Germany
| | - Jürgen Winkler
- Department of Molecular Neurology, FAU, Schwabachanlage 6, 91054, Erlangen, Germany.,Center for Rare Diseases (ZSEER), University Hospital Erlangen, Erlangen, Germany
| | | | - Martin Regensburger
- Department of Neurology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany. .,Department of Molecular Neurology, FAU, Schwabachanlage 6, 91054, Erlangen, Germany. .,Center for Rare Diseases (ZSEER), University Hospital Erlangen, Erlangen, Germany.
| |
Collapse
|
19
|
Murie M, Peng Y, Rigby MJ, Dieterich IA, Farrugia MA, Endresen A, Bhattacharyya A, Puglielli L. ATase inhibition rescues age-associated proteotoxicity of the secretory pathway. Commun Biol 2022; 5:173. [PMID: 35217767 PMCID: PMC8881600 DOI: 10.1038/s42003-022-03118-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Accepted: 02/04/2022] [Indexed: 01/25/2023] Open
Abstract
Malfunction of autophagy contributes to the progression of many chronic age-associated diseases. As such, improving normal proteostatic mechanisms is an active target for biomedical research and a key focal area for aging research. Endoplasmic reticulum (ER)-based acetylation has emerged as a mechanism that ensures proteostasis within the ER by regulating the induction of ER specific autophagy. ER acetylation is ensured by two ER-membrane bound acetyltransferases, ATase1 and ATase2. Here, we show that ATase inhibitors can rescue ongoing disease manifestations associated with the segmental progeria-like phenotype of AT-1 sTg mice. We also describe a pipeline to reliably identify ATase inhibitors with promising druggability properties. Finally, we show that successful ATase inhibitors can rescue the proteopathy of a mouse model of Alzheimer's disease. In conclusion, our study proposes that ATase-targeting approaches might offer a translational pathway for many age-associated proteopathies affecting the ER/secretory pathway.
Collapse
Affiliation(s)
- Maeghan Murie
- grid.14003.360000 0001 2167 3675Department of Medicine, University of Wisconsin-Madison, Madison, WI USA ,grid.14003.360000 0001 2167 3675Waisman Center, University of Wisconsin-Madison, Madison, WI USA ,grid.14003.360000 0001 2167 3675Neuroscience Training Program, University of Wisconsin-Madison, Madison, WI USA
| | - Yajing Peng
- grid.14003.360000 0001 2167 3675Department of Medicine, University of Wisconsin-Madison, Madison, WI USA ,grid.14003.360000 0001 2167 3675Waisman Center, University of Wisconsin-Madison, Madison, WI USA
| | - Michael J. Rigby
- grid.14003.360000 0001 2167 3675Department of Medicine, University of Wisconsin-Madison, Madison, WI USA ,grid.14003.360000 0001 2167 3675Waisman Center, University of Wisconsin-Madison, Madison, WI USA ,grid.14003.360000 0001 2167 3675Neuroscience Training Program, University of Wisconsin-Madison, Madison, WI USA
| | - Inca A. Dieterich
- grid.14003.360000 0001 2167 3675Department of Medicine, University of Wisconsin-Madison, Madison, WI USA ,grid.14003.360000 0001 2167 3675Waisman Center, University of Wisconsin-Madison, Madison, WI USA ,grid.14003.360000 0001 2167 3675Neuroscience Training Program, University of Wisconsin-Madison, Madison, WI USA
| | - Mark A. Farrugia
- grid.14003.360000 0001 2167 3675Department of Medicine, University of Wisconsin-Madison, Madison, WI USA ,grid.14003.360000 0001 2167 3675Waisman Center, University of Wisconsin-Madison, Madison, WI USA ,grid.17088.360000 0001 2150 1785Present Address: Mark A. Farrugia, Michigan State University, East Lansing, MI USA
| | - Andreas Endresen
- grid.14003.360000 0001 2167 3675Department of Medicine, University of Wisconsin-Madison, Madison, WI USA ,grid.14003.360000 0001 2167 3675Waisman Center, University of Wisconsin-Madison, Madison, WI USA
| | - Anita Bhattacharyya
- grid.14003.360000 0001 2167 3675Waisman Center, University of Wisconsin-Madison, Madison, WI USA ,grid.14003.360000 0001 2167 3675Department of Cell and Regenerative Biology, University of Wisconsin-Madison, Madison, WI USA
| | - Luigi Puglielli
- grid.14003.360000 0001 2167 3675Department of Medicine, University of Wisconsin-Madison, Madison, WI USA ,grid.14003.360000 0001 2167 3675Waisman Center, University of Wisconsin-Madison, Madison, WI USA ,Geriatric Research Education Clinical Center, Veterans Affairs Medical Center, Madison, WI USA
| |
Collapse
|
20
|
Davies AK, Alecu JE, Ziegler M, Vasilopoulou CG, Merciai F, Jumo H, Afshar-Saber W, Sahin M, Ebrahimi-Fakhari D, Borner GHH. AP-4-mediated axonal transport controls endocannabinoid production in neurons. Nat Commun 2022; 13:1058. [PMID: 35217685 PMCID: PMC8881493 DOI: 10.1038/s41467-022-28609-w] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Accepted: 01/08/2022] [Indexed: 01/20/2023] Open
Abstract
The adaptor protein complex AP-4 mediates anterograde axonal transport and is essential for axon health. AP-4-deficient patients suffer from a severe neurodevelopmental and neurodegenerative disorder. Here we identify DAGLB (diacylglycerol lipase-beta), a key enzyme for generation of the endocannabinoid 2-AG (2-arachidonoylglycerol), as a cargo of AP-4 vesicles. During normal development, DAGLB is targeted to the axon, where 2-AG signalling drives axonal growth. We show that DAGLB accumulates at the trans-Golgi network of AP-4-deficient cells, that axonal DAGLB levels are reduced in neurons from a patient with AP-4 deficiency, and that 2-AG levels are reduced in the brains of AP-4 knockout mice. Importantly, we demonstrate that neurite growth defects of AP-4-deficient neurons are rescued by inhibition of MGLL (monoacylglycerol lipase), the enzyme responsible for 2-AG hydrolysis. Our study supports a new model for AP-4 deficiency syndrome in which axon growth defects arise through spatial dysregulation of endocannabinoid signalling. Davies et al. identify a putative mechanism underlying the childhood neurological disorder AP-4 deficiency syndrome. In the absence of AP-4, an enzyme that makes 2-AG is not transported to the axon, leading to axonal growth defects, which can be rescued by inhibition of 2-AG breakdown.
Collapse
Affiliation(s)
- Alexandra K Davies
- Department of Proteomics and Signal Transduction, Max Planck Institute of Biochemistry, Martinsried, 82152, Germany.
| | - Julian E Alecu
- Department of Neurology, The F.M. Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Marvin Ziegler
- Department of Neurology, The F.M. Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, MA, 02115, USA.,Department of Functional Neuroanatomy, Institute of Anatomy and Cell Biology, Heidelberg University, INF 307, Heidelberg, 69120, Germany
| | - Catherine G Vasilopoulou
- Department of Proteomics and Signal Transduction, Max Planck Institute of Biochemistry, Martinsried, 82152, Germany
| | - Fabrizio Merciai
- Department of Proteomics and Signal Transduction, Max Planck Institute of Biochemistry, Martinsried, 82152, Germany.,Department of Pharmacy and PhD Program in Drug Discovery and Development, University of Salerno, 84084, Fisciano, SA, Italy
| | - Hellen Jumo
- Department of Neurology, The F.M. Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Wardiya Afshar-Saber
- Department of Neurology, The F.M. Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Mustafa Sahin
- Department of Neurology, The F.M. Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, MA, 02115, USA.,Rosamund Stone Zander Translational Neuroscience Center, Boston Children's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Darius Ebrahimi-Fakhari
- Department of Neurology, The F.M. Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Georg H H Borner
- Department of Proteomics and Signal Transduction, Max Planck Institute of Biochemistry, Martinsried, 82152, Germany.
| |
Collapse
|
21
|
Shin J, Nile A, Oh JW. Role of adaptin protein complexes in intracellular trafficking and their impact on diseases. Bioengineered 2021; 12:8259-8278. [PMID: 34565296 PMCID: PMC8806629 DOI: 10.1080/21655979.2021.1982846] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2021] [Revised: 09/15/2021] [Accepted: 09/15/2021] [Indexed: 02/07/2023] Open
Abstract
Adaptin proteins (APs) play a crucial role in intracellular cell trafficking. The 'classical' role of APs is carried out by AP1‒3, which bind to clathrin, cargo, and accessory proteins. Accordingly, AP1-3 are crucial for both vesicle formation and sorting. All APs consist of four subunits that are indispensable for their functions. In fact, based on studies using cells, model organism knockdown/knock-out, and human variants, each subunit plays crucial roles and contributes to the specificity of each AP. These studies also revealed that the sorting and intracellular trafficking function of AP can exert varying effects on pathology by controlling features such as cell development, signal transduction related to the apoptosis and proliferation pathways in cancer cells, organelle integrity, receptor presentation, and viral infection. Although the roles and functions of AP1‒3 are relatively well studied, the functions of the less abundant and more recently identified APs, AP4 and AP5, are still to be investigated. Further studies on these APs may enable a better understanding and targeting of specific diseases.APs known or suggested locations and functions.
Collapse
Affiliation(s)
- Juhyun Shin
- Department of Stem Cell and Regenerative Biotechnology and Animal Resources Research Center, Konkuk University, Seoul, Republic of Korea
| | - Arti Nile
- Department of Stem Cell and Regenerative Biotechnology and Animal Resources Research Center, Konkuk University, Seoul, Republic of Korea
| | - Jae-Wook Oh
- Department of Stem Cell and Regenerative Biotechnology and Animal Resources Research Center, Konkuk University, Seoul, Republic of Korea
| |
Collapse
|
22
|
Ebrahimi-Fakhari D, Alecu JE, Ziegler M, Geisel G, Jordan C, D'Amore A, Yeh RC, Akula SK, Saffari A, Prabhu SP, Sahin M, Yang E. Systematic Analysis of Brain MRI Findings in Adaptor Protein Complex 4-Associated Hereditary Spastic Paraplegia. Neurology 2021; 97:e1942-e1954. [PMID: 34544818 DOI: 10.1212/wnl.0000000000012836] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Accepted: 08/23/2021] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND AND OBJECTIVES AP-4-associated hereditary spastic paraplegia (AP-4-HSP: SPG47, SPG50, SPG51, SPG52) is an emerging cause of childhood-onset hereditary spastic paraplegia and mimic of cerebral palsy. This study aims to define the spectrum of brain MRI findings in AP-4-HSP and to investigate radioclinical correlations. METHODS We performed a systematic qualitative and quantitative analysis of 107 brain MRI studies from 76 individuals with genetically confirmed AP-4-HSP and correlation with clinical findings including surrogates of disease severity. RESULTS We define AP-4-HSP as a disorder of gray and white matter and demonstrate that abnormal myelination is common and that metrics of reduced white matter volume correlate with severity of motor symptoms. We identify a common diagnostic imaging signature consisting of (1) a thin splenium of the corpus callosum, (2) an absent or thin anterior commissure, (3) characteristic signal abnormalities of the forceps minor ("ears of the grizzly sign"), and (4) periventricular white matter abnormalities. The presence of 2 or more of these findings has a sensitivity of ∼99% for detecting AP-4-HSP; the combination of all 4 is found in ∼45% of cases. Compared to other HSPs with a thin corpus callosum, the absent anterior commissure appears to be specific to AP-4-HSP. Our analysis identified a subset of patients with polymicrogyria, underscoring the role of AP-4 in early brain development. These patients displayed a higher prevalence of seizures and status epilepticus, many at a young age. DISCUSSION Our findings define the MRI spectrum of AP-4-HSP, providing opportunities for early diagnosis, identification of individuals at risk for complications, and a window into the role of the AP-4 complex in brain development and neurodegeneration.
Collapse
Affiliation(s)
- Darius Ebrahimi-Fakhari
- From the Department of Neurology (D.E.-F., J.E.A., M.Z., G.G., C.J., A.D., A.S., M.S.), and Division of Neuroradiology, Department of Radiology (S.P.P., E.Y.), The Manton Center for Orphan Disease Research (D.E.-F., R.C.Y., S.K.A.), Rosamund Stone Zander Translational Neuroscience Center (M.S.), and Division of Genetics and Genomics (D.E.-F., R.C.Y., S.K.A.), Boston Children's Hospital, Harvard Medical School, MA.
| | - Julian E Alecu
- From the Department of Neurology (D.E.-F., J.E.A., M.Z., G.G., C.J., A.D., A.S., M.S.), and Division of Neuroradiology, Department of Radiology (S.P.P., E.Y.), The Manton Center for Orphan Disease Research (D.E.-F., R.C.Y., S.K.A.), Rosamund Stone Zander Translational Neuroscience Center (M.S.), and Division of Genetics and Genomics (D.E.-F., R.C.Y., S.K.A.), Boston Children's Hospital, Harvard Medical School, MA
| | - Marvin Ziegler
- From the Department of Neurology (D.E.-F., J.E.A., M.Z., G.G., C.J., A.D., A.S., M.S.), and Division of Neuroradiology, Department of Radiology (S.P.P., E.Y.), The Manton Center for Orphan Disease Research (D.E.-F., R.C.Y., S.K.A.), Rosamund Stone Zander Translational Neuroscience Center (M.S.), and Division of Genetics and Genomics (D.E.-F., R.C.Y., S.K.A.), Boston Children's Hospital, Harvard Medical School, MA
| | - Gregory Geisel
- From the Department of Neurology (D.E.-F., J.E.A., M.Z., G.G., C.J., A.D., A.S., M.S.), and Division of Neuroradiology, Department of Radiology (S.P.P., E.Y.), The Manton Center for Orphan Disease Research (D.E.-F., R.C.Y., S.K.A.), Rosamund Stone Zander Translational Neuroscience Center (M.S.), and Division of Genetics and Genomics (D.E.-F., R.C.Y., S.K.A.), Boston Children's Hospital, Harvard Medical School, MA
| | - Catherine Jordan
- From the Department of Neurology (D.E.-F., J.E.A., M.Z., G.G., C.J., A.D., A.S., M.S.), and Division of Neuroradiology, Department of Radiology (S.P.P., E.Y.), The Manton Center for Orphan Disease Research (D.E.-F., R.C.Y., S.K.A.), Rosamund Stone Zander Translational Neuroscience Center (M.S.), and Division of Genetics and Genomics (D.E.-F., R.C.Y., S.K.A.), Boston Children's Hospital, Harvard Medical School, MA
| | - Angelica D'Amore
- From the Department of Neurology (D.E.-F., J.E.A., M.Z., G.G., C.J., A.D., A.S., M.S.), and Division of Neuroradiology, Department of Radiology (S.P.P., E.Y.), The Manton Center for Orphan Disease Research (D.E.-F., R.C.Y., S.K.A.), Rosamund Stone Zander Translational Neuroscience Center (M.S.), and Division of Genetics and Genomics (D.E.-F., R.C.Y., S.K.A.), Boston Children's Hospital, Harvard Medical School, MA
| | - Rebecca C Yeh
- From the Department of Neurology (D.E.-F., J.E.A., M.Z., G.G., C.J., A.D., A.S., M.S.), and Division of Neuroradiology, Department of Radiology (S.P.P., E.Y.), The Manton Center for Orphan Disease Research (D.E.-F., R.C.Y., S.K.A.), Rosamund Stone Zander Translational Neuroscience Center (M.S.), and Division of Genetics and Genomics (D.E.-F., R.C.Y., S.K.A.), Boston Children's Hospital, Harvard Medical School, MA
| | - Shyam K Akula
- From the Department of Neurology (D.E.-F., J.E.A., M.Z., G.G., C.J., A.D., A.S., M.S.), and Division of Neuroradiology, Department of Radiology (S.P.P., E.Y.), The Manton Center for Orphan Disease Research (D.E.-F., R.C.Y., S.K.A.), Rosamund Stone Zander Translational Neuroscience Center (M.S.), and Division of Genetics and Genomics (D.E.-F., R.C.Y., S.K.A.), Boston Children's Hospital, Harvard Medical School, MA
| | - Afshin Saffari
- From the Department of Neurology (D.E.-F., J.E.A., M.Z., G.G., C.J., A.D., A.S., M.S.), and Division of Neuroradiology, Department of Radiology (S.P.P., E.Y.), The Manton Center for Orphan Disease Research (D.E.-F., R.C.Y., S.K.A.), Rosamund Stone Zander Translational Neuroscience Center (M.S.), and Division of Genetics and Genomics (D.E.-F., R.C.Y., S.K.A.), Boston Children's Hospital, Harvard Medical School, MA
| | - Sanjay P Prabhu
- From the Department of Neurology (D.E.-F., J.E.A., M.Z., G.G., C.J., A.D., A.S., M.S.), and Division of Neuroradiology, Department of Radiology (S.P.P., E.Y.), The Manton Center for Orphan Disease Research (D.E.-F., R.C.Y., S.K.A.), Rosamund Stone Zander Translational Neuroscience Center (M.S.), and Division of Genetics and Genomics (D.E.-F., R.C.Y., S.K.A.), Boston Children's Hospital, Harvard Medical School, MA
| | - Mustafa Sahin
- From the Department of Neurology (D.E.-F., J.E.A., M.Z., G.G., C.J., A.D., A.S., M.S.), and Division of Neuroradiology, Department of Radiology (S.P.P., E.Y.), The Manton Center for Orphan Disease Research (D.E.-F., R.C.Y., S.K.A.), Rosamund Stone Zander Translational Neuroscience Center (M.S.), and Division of Genetics and Genomics (D.E.-F., R.C.Y., S.K.A.), Boston Children's Hospital, Harvard Medical School, MA
| | - Edward Yang
- From the Department of Neurology (D.E.-F., J.E.A., M.Z., G.G., C.J., A.D., A.S., M.S.), and Division of Neuroradiology, Department of Radiology (S.P.P., E.Y.), The Manton Center for Orphan Disease Research (D.E.-F., R.C.Y., S.K.A.), Rosamund Stone Zander Translational Neuroscience Center (M.S.), and Division of Genetics and Genomics (D.E.-F., R.C.Y., S.K.A.), Boston Children's Hospital, Harvard Medical School, MA
| | | |
Collapse
|
23
|
Ebrahimi-Fakhari D, Alecu JE, Brechmann B, Ziegler M, Eberhardt K, Jumo H, D’Amore A, Habibzadeh P, Faghihi MA, De Bleecker JL, Vuillaumier-Barrot S, Auvin S, Santorelli FM, Neuser S, Popp B, Yang E, Barrett L, Davies AK, Saffari A, Hirst J, Sahin M. High-throughput imaging of ATG9A distribution as a diagnostic functional assay for adaptor protein complex 4-associated hereditary spastic paraplegia. Brain Commun 2021; 3:fcab221. [PMID: 34729478 PMCID: PMC8557665 DOI: 10.1093/braincomms/fcab221] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Revised: 08/05/2021] [Accepted: 08/19/2021] [Indexed: 01/09/2023] Open
Abstract
Adaptor protein complex 4-associated hereditary spastic paraplegia is caused by biallelic loss-of-function variants in AP4B1, AP4M1, AP4E1 or AP4S1, which constitute the four subunits of this obligate complex. While the diagnosis of adaptor protein complex 4-associated hereditary spastic paraplegia relies on molecular testing, the interpretation of novel missense variants remains challenging. Here, we address this diagnostic gap by using patient-derived fibroblasts to establish a functional assay that measures the subcellular localization of ATG9A, a transmembrane protein that is sorted by adaptor protein complex 4. Using automated high-throughput microscopy, we determine the ratio of the ATG9A fluorescence in the trans-Golgi-network versus cytoplasm and ascertain that this metric meets standards for screening assays (Z'-factor robust >0.3, strictly standardized mean difference >3). The 'ATG9A ratio' is increased in fibroblasts of 18 well-characterized adaptor protein complex 4-associated hereditary spastic paraplegia patients [mean: 1.54 ± 0.13 versus 1.21 ± 0.05 (standard deviation) in controls] and receiver-operating characteristic analysis demonstrates robust diagnostic power (area under the curve: 0.85, 95% confidence interval: 0.849-0.852). Using fibroblasts from two individuals with atypical clinical features and novel biallelic missense variants of unknown significance in AP4B1, we show that our assay can reliably detect adaptor protein complex 4 function. Our findings establish the 'ATG9A ratio' as a diagnostic marker of adaptor protein complex 4-associated hereditary spastic paraplegia.
Collapse
Affiliation(s)
- Darius Ebrahimi-Fakhari
- Department of Neurology, The F.M. Kirby Neurobiology Center, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA
- The Manton Center for Orphan Disease Research, Boston Children’s Hospital, Boston, MA 02115, USA
| | - Julian E Alecu
- Department of Neurology, The F.M. Kirby Neurobiology Center, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Barbara Brechmann
- Department of Neurology, The F.M. Kirby Neurobiology Center, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA
- Division of Neuropediatrics and Inherited Metabolic Diseases, Center for Pediatrics and Adolescent Medicine, University Hospital Heidelberg, 69120 Heidelberg, Germany
| | - Marvin Ziegler
- Department of Neurology, The F.M. Kirby Neurobiology Center, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA
- Ruprecht-Karls University Heidelberg, Medical School, 69120 Heidelberg, Germany
| | - Kathrin Eberhardt
- Department of Neurology, The F.M. Kirby Neurobiology Center, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Hellen Jumo
- Department of Neurology, The F.M. Kirby Neurobiology Center, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Angelica D’Amore
- Department of Neurology, The F.M. Kirby Neurobiology Center, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Parham Habibzadeh
- Persian BayanGene Research and Training Center, Shiraz University of Medical Sciences, 71347 Shiraz, Iran
| | - Mohammad Ali Faghihi
- Department of Psychiatry and Behavioral Sciences, University of Miami, Miami, FL 33136, USA
| | - Jan L De Bleecker
- Department of Neurology, Ghent University Hospital, 9000 Ghent, Belgium
| | | | - Stéphane Auvin
- Pediatric Neurology Department, AP-HP, Robert Debré Hospital, 75019 Paris, France
| | - Filippo M Santorelli
- Department of Molecular Medicine, IRCCS Fondazione Stella Maris, 56128 Pisa, Italy
| | - Sonja Neuser
- Institute of Human Genetics, University of Leipzig Medical Center, 04103 Leipzig, Germany
| | - Bernt Popp
- Institute of Human Genetics, University of Leipzig Medical Center, 04103 Leipzig, Germany
| | - Edward Yang
- Division of Neuroradiology, Department of Radiology, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Lee Barrett
- Department of Neurology, The F.M. Kirby Neurobiology Center, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA
- Rosamund Stone Zander Translational Neuroscience Center, Boston Children’s Hospital, Boston, MA 02115, USA
| | - Alexandra K Davies
- Department of Proteomics and Signal Transduction, Max Planck Institute of Biochemistry, 82152 Martinsried, Germany
| | - Afshin Saffari
- Department of Neurology, The F.M. Kirby Neurobiology Center, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA
- Division of Neuropediatrics and Inherited Metabolic Diseases, Center for Pediatrics and Adolescent Medicine, University Hospital Heidelberg, 69120 Heidelberg, Germany
| | - Jennifer Hirst
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge CB2 0XY, UK
| | - Mustafa Sahin
- Department of Neurology, The F.M. Kirby Neurobiology Center, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA
- Rosamund Stone Zander Translational Neuroscience Center, Boston Children’s Hospital, Boston, MA 02115, USA
| |
Collapse
|
24
|
Jordan C, Geisel G, Alecu JE, Zhang B, Sahin M, Ebrahimi-Fakhari D. Disease Severity and Motor Impairment Correlate With Health-Related Quality of Life in AP-4-Associated Hereditary Spastic Paraplegia. NEUROLOGY-GENETICS 2021; 7:e605. [PMID: 34295967 PMCID: PMC8293284 DOI: 10.1212/nxg.0000000000000605] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Accepted: 04/23/2021] [Indexed: 11/15/2022]
Abstract
Objective AP-4-associated hereditary spastic paraplegia (AP-4-HSP) is a childhood-onset neurogenetic disease and mimic of cerebral palsy. Data on health-related quality of life (HRQoL) are lacking. To establish a metric for HRQoL and caregiver priorities, we used the Caregiver Priorities and Child Health Index of Life with Disabilities (CPCHILD) questionnaire to assess HRQoL in correlation with disease severity in 64 patients with AP-4-HSP. Methods A cross-sectional analysis of caregiver-reported HRQoL was performed using the CPCHILD questionnaire in combination with a detailed clinical characterization. Results HRQoL was impaired in all domains in patients with AP-4-HSP (mean score: 59.6 ± 12.6 [SD]), with no significant difference between the 4 subtypes. Age, as a surrogate for disease duration, and Spastic Paraplegia Rating Scale scores, as an indicator for corticospinal tract dysfunction and motor impairment, correlated with lower CPCHILD scores (Pearson r = −0.31, p = 0.01 and r = −0.52, p < 0.0001, respectively). Patients with tetraplegia showed lower CPCHILD scores compared with individuals with diplegia or no spasticity. Wheelchair dependence reduced HRQoL in all domains. The presence of seizures, including medically refractory epilepsy, was not associated with lower CPCHILD scores. Standardized assessment of caregiver priorities identified several areas of high importance to HRQoL. Conclusions We show that the CPCHILD questionnaire, developed for use in children with cerebral palsy, can be used to assess HRQoL in patients with childhood-onset complex hereditary spastic paraplegia. HRQoL is reduced in patients with AP-4-HSP and correlates with the degree of motor impairment. These results provide a framework for medical decision making and a baseline for the future development of treatment guidelines and interventional trials.
Collapse
Affiliation(s)
- Catherine Jordan
- Department of Neurology and The F.M. Kirby Neurobiology Center (C.J., G.G., J.E.A., M.S., D.E.-F.), Rosamund Stone Zander Translational Neuroscience Center (G.G., M.S.), and ICCTR Biostatistics and Research Design Center (B.Z.), Boston Children's Hospital, Harvard Medical School, MA
| | - Gregory Geisel
- Department of Neurology and The F.M. Kirby Neurobiology Center (C.J., G.G., J.E.A., M.S., D.E.-F.), Rosamund Stone Zander Translational Neuroscience Center (G.G., M.S.), and ICCTR Biostatistics and Research Design Center (B.Z.), Boston Children's Hospital, Harvard Medical School, MA
| | - Julian E Alecu
- Department of Neurology and The F.M. Kirby Neurobiology Center (C.J., G.G., J.E.A., M.S., D.E.-F.), Rosamund Stone Zander Translational Neuroscience Center (G.G., M.S.), and ICCTR Biostatistics and Research Design Center (B.Z.), Boston Children's Hospital, Harvard Medical School, MA
| | - Bo Zhang
- Department of Neurology and The F.M. Kirby Neurobiology Center (C.J., G.G., J.E.A., M.S., D.E.-F.), Rosamund Stone Zander Translational Neuroscience Center (G.G., M.S.), and ICCTR Biostatistics and Research Design Center (B.Z.), Boston Children's Hospital, Harvard Medical School, MA
| | - Mustafa Sahin
- Department of Neurology and The F.M. Kirby Neurobiology Center (C.J., G.G., J.E.A., M.S., D.E.-F.), Rosamund Stone Zander Translational Neuroscience Center (G.G., M.S.), and ICCTR Biostatistics and Research Design Center (B.Z.), Boston Children's Hospital, Harvard Medical School, MA
| | - Darius Ebrahimi-Fakhari
- Department of Neurology and The F.M. Kirby Neurobiology Center (C.J., G.G., J.E.A., M.S., D.E.-F.), Rosamund Stone Zander Translational Neuroscience Center (G.G., M.S.), and ICCTR Biostatistics and Research Design Center (B.Z.), Boston Children's Hospital, Harvard Medical School, MA
| |
Collapse
|
25
|
A novel loss of function mutation in adaptor protein complex 4, subunit mu-1 causing autosomal recessive spastic paraplegia 50. Neurol Sci 2021; 42:5311-5319. [PMID: 33884525 DOI: 10.1007/s10072-021-05262-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Accepted: 04/16/2021] [Indexed: 10/21/2022]
Abstract
BACKGROUND Spastic paraplegia 50 (SPG50) is a rare autosomal recessive inherited disorder characterized by spasticity, severe intellectual disability and delayed or absent speech. Loss-of-function pathogenic mutations in the AP4M1 gene cause SPG50. METHODS In this study, we investigated the clinical and genetic characteristics of a consanguineous family with two male siblings who had infantile hypotonia that progressed to spasticity, paraplegia in one and quadriplegia in the other patient. In addition, the patients also exhibited neurodevelopmental phenotypes including severe intellectual disability, developmental delay, microcephaly and dysmorphism. RESULTS In order to identify the genetic cause, we performed cytogenetics, whole-exome sequencing and Sanger sequencing. Whole-exome sequencing of the affected siblings and unaffected parents revealed a novel exonic frameshift insertion of eight nucleotides (c.341_342insTGAAGTGC) on exon 4 of the AP4M1 gene. CONCLUSION Insertion of these eight nucleotides in the AP4M1 gene is predicted to result in a premature protein product of 132 amino acids. The truncated protein product lacks a signal binding domain which is essential for protein-protein interactions and the transport of cargo proteins to the membrane. Thus, the identified variant is pathogenic and our study expands the knowledge of clinical and genetic features of SPG50.
Collapse
|
26
|
Moreno-De-Luca A, Millan F, Pesacreta DR, Elloumi HZ, Oetjens MT, Teigen C, Wain KE, Scuffins J, Myers SM, Torene RI, Gainullin VG, Arvai K, Kirchner HL, Ledbetter DH, Retterer K, Martin CL. Molecular Diagnostic Yield of Exome Sequencing in Patients With Cerebral Palsy. JAMA 2021; 325:467-475. [PMID: 33528536 PMCID: PMC7856544 DOI: 10.1001/jama.2020.26148] [Citation(s) in RCA: 76] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
IMPORTANCE Cerebral palsy is a common neurodevelopmental disorder affecting movement and posture that often co-occurs with other neurodevelopmental disorders. Individual cases of cerebral palsy are often attributed to birth asphyxia; however, recent studies indicate that asphyxia accounts for less than 10% of cerebral palsy cases. OBJECTIVE To determine the molecular diagnostic yield of exome sequencing (prevalence of pathogenic and likely pathogenic variants) in individuals with cerebral palsy. DESIGN, SETTING, AND PARTICIPANTS A retrospective cohort study of patients with cerebral palsy that included a clinical laboratory referral cohort with data accrued between 2012 and 2018 and a health care-based cohort with data accrued between 2007 and 2017. EXPOSURES Exome sequencing with copy number variant detection. MAIN OUTCOMES AND MEASURES The primary outcome was the molecular diagnostic yield of exome sequencing. RESULTS Among 1345 patients from the clinical laboratory referral cohort, the median age was 8.8 years (interquartile range, 4.4-14.7 years; range, 0.1-66 years) and 601 (45%) were female. Among 181 patients in the health care-based cohort, the median age was 41.9 years (interquartile range, 28.0-59.6 years; range, 4.8-89 years) and 96 (53%) were female. The molecular diagnostic yield of exome sequencing was 32.7% (95% CI, 30.2%-35.2%) in the clinical laboratory referral cohort and 10.5% (95% CI, 6.0%-15.0%) in the health care-based cohort. The molecular diagnostic yield ranged from 11.2% (95% CI, 6.4%-16.2%) for patients without intellectual disability, epilepsy, or autism spectrum disorder to 32.9% (95% CI, 25.7%-40.1%) for patients with all 3 comorbidities. Pathogenic and likely pathogenic variants were identified in 229 genes (29.5% of 1526 patients); 86 genes were mutated in 2 or more patients (20.1% of 1526 patients) and 10 genes with mutations were independently identified in both cohorts (2.9% of 1526 patients). CONCLUSIONS AND RELEVANCE Among 2 cohorts of patients with cerebral palsy who underwent exome sequencing, the prevalence of pathogenic and likely pathogenic variants was 32.7% in a cohort that predominantly consisted of pediatric patients and 10.5% in a cohort that predominantly consisted of adult patients. Further research is needed to understand the clinical implications of these findings.
Collapse
Affiliation(s)
- Andrés Moreno-De-Luca
- Autism & Developmental Medicine Institute, Geisinger, Danville, Pennsylvania
- Genomic Medicine Institute, Geisinger, Danville, Pennsylvania
- Department of Radiology, Geisinger, Danville, Pennsylvania
- Diagnostic Medicine Institute, Geisinger, Danville, Pennsylvania
| | | | - Denis R. Pesacreta
- Autism & Developmental Medicine Institute, Geisinger, Danville, Pennsylvania
| | | | - Matthew T. Oetjens
- Autism & Developmental Medicine Institute, Geisinger, Danville, Pennsylvania
| | | | - Karen E. Wain
- Autism & Developmental Medicine Institute, Geisinger, Danville, Pennsylvania
- Genomic Medicine Institute, Geisinger, Danville, Pennsylvania
| | | | - Scott M. Myers
- Autism & Developmental Medicine Institute, Geisinger, Danville, Pennsylvania
| | | | | | | | - H. Lester Kirchner
- Department of Population Health Sciences, Geisinger, Danville, Pennsylvania
| | - David H. Ledbetter
- Autism & Developmental Medicine Institute, Geisinger, Danville, Pennsylvania
- Genomic Medicine Institute, Geisinger, Danville, Pennsylvania
| | | | - Christa L. Martin
- Autism & Developmental Medicine Institute, Geisinger, Danville, Pennsylvania
- Genomic Medicine Institute, Geisinger, Danville, Pennsylvania
| |
Collapse
|
27
|
Ziegler M, Russell BE, Eberhardt K, Geisel G, D'Amore A, Sahin M, Kornblum HI, Ebrahimi-Fakhari D. Blended Phenotype of Silver-Russell Syndrome and SPG50 Caused by Maternal Isodisomy of Chromosome 7. NEUROLOGY-GENETICS 2020; 7:e544. [PMID: 33553621 PMCID: PMC7862086 DOI: 10.1212/nxg.0000000000000544] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Accepted: 10/22/2020] [Indexed: 11/20/2022]
Abstract
Objective Uniparental isodisomy can lead to blended phenotypes of imprinting disorders and autosomal recessive diseases. To determine whether a complex neurodevelopmental disorder was caused by uniparental isodisomy, a detailed clinical and molecular characterization was performed. Methods A combination of clinical, molecular, and imaging data and functional studies in patient-derived fibroblasts. Results We report a 4-year-old female with a blended, complex phenotype of Silver-Russell syndrome (SRS) and hereditary spastic paraplegia type 50 (SPG50) caused by total maternal isodisomy of chromosome 7 (UPiD(7)mat) and a loss-of-function variant in AP4M1 (NM_00472.3: c.59-1G>C, IVS1-1G>C). Functional studies in patient-derived fibroblasts confirmed the loss of adaptor protein complex 4 function. Distinctive facial features, intrauterine growth restriction, short stature, feeding difficulties, and severe gastroesophageal reflux were consistent with SRS. Features associated with SPG50 included early-onset epilepsy, episodes of stereotypical laughter, and thinning of the corpus callosum and ventriculomegaly on brain MRI. Symptoms shared by both syndromes such as developmental delay, short stature, and axial and appendicular hypotonia were also present. Notably, other common manifestations of SPG50 such as microcephaly or spasticity had not developed yet. Conclusions This case highlights that atypical clinical features in patients with well-described imprinting disorders should lead to investigations for recessive conditions caused by variants in genes that localize to the region of homozygosity.
Collapse
Affiliation(s)
- Marvin Ziegler
- Department of Neurology and The F.M. Kirby Neurobiology Center (M.Z., K.E., G.G., A.D., M.S., D.E.-F.), Boston Children's Hospital, Harvard Medical School, MA; Department of Functional Neuroanatomy (M.Z.), Institute of Anatomy and Cell Biology, Heidelberg University, Germany; Division of Genetics (B.E.R.), Department of Pediatrics, David Geffen School of Medicine at UCLA; Translational Neuroscience Center (G.G., M.S.), Boston Children's Hospital, Harvard Medical School, MA; and Intellectual and Developmental Disabilities Research Center (H.I.K.), Semel Institute for Neuroscience and Human Behavior and Department of Psychiatry and Biobehavioral Sciences (H.I.K.), David Geffen School of Medicine at UCLA
| | - Bianca E Russell
- Department of Neurology and The F.M. Kirby Neurobiology Center (M.Z., K.E., G.G., A.D., M.S., D.E.-F.), Boston Children's Hospital, Harvard Medical School, MA; Department of Functional Neuroanatomy (M.Z.), Institute of Anatomy and Cell Biology, Heidelberg University, Germany; Division of Genetics (B.E.R.), Department of Pediatrics, David Geffen School of Medicine at UCLA; Translational Neuroscience Center (G.G., M.S.), Boston Children's Hospital, Harvard Medical School, MA; and Intellectual and Developmental Disabilities Research Center (H.I.K.), Semel Institute for Neuroscience and Human Behavior and Department of Psychiatry and Biobehavioral Sciences (H.I.K.), David Geffen School of Medicine at UCLA
| | - Kathrin Eberhardt
- Department of Neurology and The F.M. Kirby Neurobiology Center (M.Z., K.E., G.G., A.D., M.S., D.E.-F.), Boston Children's Hospital, Harvard Medical School, MA; Department of Functional Neuroanatomy (M.Z.), Institute of Anatomy and Cell Biology, Heidelberg University, Germany; Division of Genetics (B.E.R.), Department of Pediatrics, David Geffen School of Medicine at UCLA; Translational Neuroscience Center (G.G., M.S.), Boston Children's Hospital, Harvard Medical School, MA; and Intellectual and Developmental Disabilities Research Center (H.I.K.), Semel Institute for Neuroscience and Human Behavior and Department of Psychiatry and Biobehavioral Sciences (H.I.K.), David Geffen School of Medicine at UCLA
| | - Gregory Geisel
- Department of Neurology and The F.M. Kirby Neurobiology Center (M.Z., K.E., G.G., A.D., M.S., D.E.-F.), Boston Children's Hospital, Harvard Medical School, MA; Department of Functional Neuroanatomy (M.Z.), Institute of Anatomy and Cell Biology, Heidelberg University, Germany; Division of Genetics (B.E.R.), Department of Pediatrics, David Geffen School of Medicine at UCLA; Translational Neuroscience Center (G.G., M.S.), Boston Children's Hospital, Harvard Medical School, MA; and Intellectual and Developmental Disabilities Research Center (H.I.K.), Semel Institute for Neuroscience and Human Behavior and Department of Psychiatry and Biobehavioral Sciences (H.I.K.), David Geffen School of Medicine at UCLA
| | - Angelica D'Amore
- Department of Neurology and The F.M. Kirby Neurobiology Center (M.Z., K.E., G.G., A.D., M.S., D.E.-F.), Boston Children's Hospital, Harvard Medical School, MA; Department of Functional Neuroanatomy (M.Z.), Institute of Anatomy and Cell Biology, Heidelberg University, Germany; Division of Genetics (B.E.R.), Department of Pediatrics, David Geffen School of Medicine at UCLA; Translational Neuroscience Center (G.G., M.S.), Boston Children's Hospital, Harvard Medical School, MA; and Intellectual and Developmental Disabilities Research Center (H.I.K.), Semel Institute for Neuroscience and Human Behavior and Department of Psychiatry and Biobehavioral Sciences (H.I.K.), David Geffen School of Medicine at UCLA
| | - Mustafa Sahin
- Department of Neurology and The F.M. Kirby Neurobiology Center (M.Z., K.E., G.G., A.D., M.S., D.E.-F.), Boston Children's Hospital, Harvard Medical School, MA; Department of Functional Neuroanatomy (M.Z.), Institute of Anatomy and Cell Biology, Heidelberg University, Germany; Division of Genetics (B.E.R.), Department of Pediatrics, David Geffen School of Medicine at UCLA; Translational Neuroscience Center (G.G., M.S.), Boston Children's Hospital, Harvard Medical School, MA; and Intellectual and Developmental Disabilities Research Center (H.I.K.), Semel Institute for Neuroscience and Human Behavior and Department of Psychiatry and Biobehavioral Sciences (H.I.K.), David Geffen School of Medicine at UCLA
| | - Harley I Kornblum
- Department of Neurology and The F.M. Kirby Neurobiology Center (M.Z., K.E., G.G., A.D., M.S., D.E.-F.), Boston Children's Hospital, Harvard Medical School, MA; Department of Functional Neuroanatomy (M.Z.), Institute of Anatomy and Cell Biology, Heidelberg University, Germany; Division of Genetics (B.E.R.), Department of Pediatrics, David Geffen School of Medicine at UCLA; Translational Neuroscience Center (G.G., M.S.), Boston Children's Hospital, Harvard Medical School, MA; and Intellectual and Developmental Disabilities Research Center (H.I.K.), Semel Institute for Neuroscience and Human Behavior and Department of Psychiatry and Biobehavioral Sciences (H.I.K.), David Geffen School of Medicine at UCLA
| | - Darius Ebrahimi-Fakhari
- Department of Neurology and The F.M. Kirby Neurobiology Center (M.Z., K.E., G.G., A.D., M.S., D.E.-F.), Boston Children's Hospital, Harvard Medical School, MA; Department of Functional Neuroanatomy (M.Z.), Institute of Anatomy and Cell Biology, Heidelberg University, Germany; Division of Genetics (B.E.R.), Department of Pediatrics, David Geffen School of Medicine at UCLA; Translational Neuroscience Center (G.G., M.S.), Boston Children's Hospital, Harvard Medical School, MA; and Intellectual and Developmental Disabilities Research Center (H.I.K.), Semel Institute for Neuroscience and Human Behavior and Department of Psychiatry and Biobehavioral Sciences (H.I.K.), David Geffen School of Medicine at UCLA
| |
Collapse
|
28
|
Abstract
This scientific commentary refers to ‘Defining the clinical, molecular and imaging spectrum of adaptor protein complex 4-associated hereditary spastic paraplegia’, by Ebrahimi-Fakhari etal. (doi:10.1093/brain/awz307).
Collapse
Affiliation(s)
- Thomas T Warner
- Reta Lila Weston Institute, UCL QueenSquare Institute of Neurology, London, UK
| |
Collapse
|