1
|
Rizvi SZ, Chan WS, Maino E, Steiman S, Forguson G, Klepfish M, Cohn RD, Ivakine EA. Multi-gene duplication removal in an engineered human cellular MECP2 duplication syndrome model with an IRAK1-MECP2 duplication. MOLECULAR THERAPY. NUCLEIC ACIDS 2024; 35:102356. [PMID: 39507402 PMCID: PMC11539574 DOI: 10.1016/j.omtn.2024.102356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Accepted: 10/04/2024] [Indexed: 11/08/2024]
Abstract
Recent progress in genome editing technologies has catalyzed the generation of sophisticated cell models; however, the precise modeling of copy-number variation (CNV) diseases remains a significant challenge despite their substantial prevalence in the human population. To overcome this barrier, we have explored the utility of HAP1 cells for the accurate modeling of disease genomes with large structural variants. As an example, this study details the strategy to generate a novel cell line that serves as a model for the neurological disorder methyl CpG binding protein 2 (MECP2) duplication syndrome (MDS), featuring the critical duplication of both the MECP2 and IRAK1 genes. This model faithfully recapitulates MDS genomic rearrangement, allowing for the mechanistic study of gene overexpression and the development of therapeutic interventions. Employing a single-guide RNA (gRNA) CRISPR-Cas9 strategy, we successfully excised the duplicated genomic segment, notably halving both MECP2 and IRAK1 expression levels. The evidence establishes our model as a crucial tool for research into MDS. Furthermore, the outlined workflow is readily adaptable to model other CNV disorders and subsequently test genomic and pharmacological interventions.
Collapse
Affiliation(s)
- Samar Z. Rizvi
- Program in Genetics and Genome Biology, The Hospital for Sick Children Research Institute, Toronto, ON M5G 0A4, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Wing Suen Chan
- Program in Genetics and Genome Biology, The Hospital for Sick Children Research Institute, Toronto, ON M5G 0A4, Canada
| | - Eleonora Maino
- Biozentrum, The Center for Molecular Life Sciences, University of Basel, Basel 4056, Switzerland
| | - Sydney Steiman
- Program in Genetics and Genome Biology, The Hospital for Sick Children Research Institute, Toronto, ON M5G 0A4, Canada
- Department of Physiology, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Georgiana Forguson
- Program in Genetics and Genome Biology, The Hospital for Sick Children Research Institute, Toronto, ON M5G 0A4, Canada
| | - Maya Klepfish
- Program in Genetics and Genome Biology, The Hospital for Sick Children Research Institute, Toronto, ON M5G 0A4, Canada
| | - Ronald D. Cohn
- Program in Genetics and Genome Biology, The Hospital for Sick Children Research Institute, Toronto, ON M5G 0A4, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada
- Institute of Medical Science, University of Toronto, Toronto, ON M5S 1A8, Canada
- Department of Pediatrics, The Hospital for Sick Children, Toronto, ON M5G 1E8, Canada
| | - Evgueni A. Ivakine
- Program in Genetics and Genome Biology, The Hospital for Sick Children Research Institute, Toronto, ON M5G 0A4, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada
| |
Collapse
|
2
|
Beni R, Hussain SA, Monsell F, Gelfer Y. Management of Legg-Calve-Perthes disease: a scoping review with advice on initial management. Arch Dis Child 2024:archdischild-2024-327891. [PMID: 39613451 DOI: 10.1136/archdischild-2024-327891] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Accepted: 11/09/2024] [Indexed: 12/01/2024]
Abstract
BACKGROUND Legg-Calve-Perthes disease (LCPD) is a developmental disorder causing avascular necrosis of the femoral head in children, with long-term consequences that can extend into adulthood. Early diagnosis and management in primary care are crucial but challenging. AIM This review aims to provide a concise overview of the presentation, differential diagnosis and management of LCPD, offering practical guidance for primary healthcare professionals. METHOD Recent literature and expert opinions were reviewed to summarise the epidemiology, diagnosis and current management of LCPD. RESULTS LCPD commonly presents as a painless limp in children aged between 2 and 14 years, with the diagnosis based on the clinical features and radiographic abnormalities. Management is individualised and includes non-operative care to surgery, which attempts to correct anatomical abnormalities and therefore delay the onset of osteoarthritis. The review highlights the importance of primary care in early detection, appropriate referral and interim management. CONCLUSION LCPD is a rare condition that can lead to long-term disability, affecting a child's physical, mental and social development, often presenting as a painless limp. Diagnosis typically involves plain radiographs, with MRI or hip joint arthrography providing additional details for management, which may include both non-surgical (eg, physiotherapy) and surgical options. Early recognition by primary care providers is crucial for timely referral to orthopaedic services, along with interim support through physiotherapy, pain management and access to mental health and educational resources.
Collapse
Affiliation(s)
- Rebecca Beni
- City St. George's University of London, London, UK
| | - Sabba A Hussain
- St George's University Hospitals NHS Foundation Trust, London, UK
- Royal London Hospital Barts Health NHS Trust, London, UK
| | | | - Yael Gelfer
- City St. George's University of London, London, UK
- St George's University Hospitals NHS Foundation Trust, London, UK
| |
Collapse
|
3
|
Voronin G, Narasimhan J, Gittens J, Sheedy J, Lipari P, Peters M, DeMarco S, Cao L, Varganov Y, Kim MJ, Pear L, Fotouh E, Sinha S, Ray B, Wu MC, Yalamanchili P, Southgate C, Pick J, Saadipour K, Jung S, Lee J, Mollin A, Welch EM, Wu Z, Weetall M. Preclinical studies of gene replacement therapy for CDKL5 deficiency disorder. Mol Ther 2024; 32:3331-3345. [PMID: 39033321 PMCID: PMC11489525 DOI: 10.1016/j.ymthe.2024.07.012] [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: 01/06/2024] [Revised: 04/13/2024] [Accepted: 07/17/2024] [Indexed: 07/23/2024] Open
Abstract
Cyclin-dependent kinase-like 5 (CDKL5) deficiency disorder (CDD) is a rare neurodevelopmental disorder caused by a mutation in the X-linked CDKL5 gene. CDKL5 is a serine/threonine kinase that is critical for axon outgrowth and dendritic morphogenesis as well as synapse formation, maturation, and maintenance. This disorder is characterized by early-onset epilepsy, hypotonia, and failure to reach cognitive and motor developmental milestones. Because the disease is monogenic, delivery of the CDKL5 gene to the brain of patients should provide clinical benefit. To this end, we designed a gene therapy vector, adeno-associated virus (AAV)9.Syn.hCDKL5, in which human CDKL5 gene expression is driven by the synapsin promoter. In biodistribution studies conducted in mice, intracerebroventricular (i.c.v.) injection resulted in broader, more optimal biodistribution than did intra-cisterna magna (i.c.m.) delivery. AAV9.Syn.hCDKL5 treatment increased phosphorylation of EB2, a bona fide CDKL5 substrate, demonstrating biological activity in vivo. Our data provide proof of concept that i.c.v. delivery of AAV9.Syn.hCDKL5 to neonatal male Cdkl5 knockout mice reduces pathology and reduces aberrant behavior. Functional improvements were seen at doses of 3e11 to 5e11 vector genomes/g brain, which resulted in transfection of ≥50% of the neurons. Functional improvements were not seen at lower doses, suggesting a requirement for broad distribution for efficacy.
Collapse
Affiliation(s)
- Gregory Voronin
- PTC Therapeutics, Inc, 500 Warren Corporate Center Drive, Warren, NJ 07059, USA
| | - Jana Narasimhan
- PTC Therapeutics, Inc, 500 Warren Corporate Center Drive, Warren, NJ 07059, USA
| | - Jamila Gittens
- PTC Therapeutics, Inc, 500 Warren Corporate Center Drive, Warren, NJ 07059, USA
| | - Josephine Sheedy
- PTC Therapeutics, Inc, 500 Warren Corporate Center Drive, Warren, NJ 07059, USA
| | - Philip Lipari
- PTC Therapeutics, Inc, 500 Warren Corporate Center Drive, Warren, NJ 07059, USA
| | - Melinda Peters
- PTC Therapeutics, Inc, 500 Warren Corporate Center Drive, Warren, NJ 07059, USA
| | - Steven DeMarco
- PTC Therapeutics, Inc, 500 Warren Corporate Center Drive, Warren, NJ 07059, USA
| | - Liangxian Cao
- PTC Therapeutics, Inc, 500 Warren Corporate Center Drive, Warren, NJ 07059, USA
| | - Yakov Varganov
- PTC Therapeutics, Inc, 500 Warren Corporate Center Drive, Warren, NJ 07059, USA
| | - Min Jung Kim
- PTC Therapeutics, Inc, 500 Warren Corporate Center Drive, Warren, NJ 07059, USA
| | - Lisset Pear
- PTC Therapeutics, Inc, 500 Warren Corporate Center Drive, Warren, NJ 07059, USA
| | - Eman Fotouh
- PTC Therapeutics, Inc, 500 Warren Corporate Center Drive, Warren, NJ 07059, USA
| | - Supriya Sinha
- PTC Therapeutics, Inc, 500 Warren Corporate Center Drive, Warren, NJ 07059, USA
| | - Balmiki Ray
- PTC Therapeutics, Inc, 500 Warren Corporate Center Drive, Warren, NJ 07059, USA
| | - Michael C Wu
- NeuroDigitech, 8400 Miramar Road, Suite 243C, San Diego, CA 092126, USA
| | | | | | - Joseph Pick
- PTC Therapeutics, Inc, 500 Warren Corporate Center Drive, Warren, NJ 07059, USA
| | - Khalil Saadipour
- PTC Therapeutics, Inc, 500 Warren Corporate Center Drive, Warren, NJ 07059, USA
| | - Stephen Jung
- PTC Therapeutics, Inc, 500 Warren Corporate Center Drive, Warren, NJ 07059, USA
| | - Jeanee Lee
- PTC Therapeutics, Inc, 500 Warren Corporate Center Drive, Warren, NJ 07059, USA
| | - Anna Mollin
- PTC Therapeutics, Inc, 500 Warren Corporate Center Drive, Warren, NJ 07059, USA
| | - Ellen M Welch
- PTC Therapeutics, Inc, 500 Warren Corporate Center Drive, Warren, NJ 07059, USA
| | - Zhijian Wu
- PTC Therapeutics, Inc, 500 Warren Corporate Center Drive, Warren, NJ 07059, USA
| | - Marla Weetall
- PTC Therapeutics, Inc, 500 Warren Corporate Center Drive, Warren, NJ 07059, USA.
| |
Collapse
|
4
|
Spencer PS, Valdes Angues R, Palmer VS. Nodding syndrome: A role for environmental biotoxins that dysregulate MECP2 expression? J Neurol Sci 2024; 462:123077. [PMID: 38850769 DOI: 10.1016/j.jns.2024.123077] [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: 10/01/2023] [Revised: 05/15/2024] [Accepted: 05/30/2024] [Indexed: 06/10/2024]
Abstract
Nodding syndrome is an epileptic encephalopathy associated with neuroinflammation and tauopathy. This initially pediatric brain disease, which has some clinical overlap with Methyl-CpG-binding protein 2 (MECP2) Duplication Syndrome, has impacted certain impoverished East African communities coincident with local civil conflict and internal displacement, conditions that forced dependence on contaminated food and water. A potential role in Nodding syndrome for certain biotoxins (freshwater cyanotoxins plus/minus mycotoxins) with neuroinflammatory, excitotoxic, tauopathic, and MECP2-dysregulating properties, is considered here for the first time.
Collapse
Affiliation(s)
- Peter S Spencer
- Department of Neurology, School of Medicine and Oregon Institute of Occupational Health Sciences, Oregon Health & Science University, Portland, OR, USA; Oregon Institute of Occupational Health Sciences, Oregon Health & Science University, Portland, OR, USA; Gulu University School of Medicine, Gulu, Uganda.
| | - Raquel Valdes Angues
- Department of Neurology, School of Medicine and Oregon Institute of Occupational Health Sciences, Oregon Health & Science University, Portland, OR, USA
| | - Valerie S Palmer
- Department of Neurology, School of Medicine and Oregon Institute of Occupational Health Sciences, Oregon Health & Science University, Portland, OR, USA; Gulu University School of Medicine, Gulu, Uganda
| |
Collapse
|
5
|
Guerra M, Medici V, La Sala G, Farini D. Unravelling the Cerebellar Involvement in Autism Spectrum Disorders: Insights into Genetic Mechanisms and Developmental Pathways. Cells 2024; 13:1176. [PMID: 39056758 PMCID: PMC11275240 DOI: 10.3390/cells13141176] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Revised: 07/05/2024] [Accepted: 07/09/2024] [Indexed: 07/28/2024] Open
Abstract
Autism spectrum disorders (ASDs) are complex neurodevelopmental conditions characterized by deficits in social interaction and communication, as well as repetitive behaviors. Although the etiology of ASD is multifactorial, with both genetic and environmental factors contributing to its development, a strong genetic basis is widely recognized. Recent research has identified numerous genetic mutations and genomic rearrangements associated with ASD-characterizing genes involved in brain development. Alterations in developmental programs are particularly harmful during critical periods of brain development. Notably, studies have indicated that genetic disruptions occurring during the second trimester of pregnancy affect cortical development, while disturbances in the perinatal and early postnatal period affect cerebellar development. The developmental defects must be viewed in the context of the role of the cerebellum in cognitive processes, which is now well established. The present review emphasizes the genetic complexity and neuropathological mechanisms underlying ASD and aims to provide insights into the cerebellar involvement in the disorder, focusing on recent advances in the molecular landscape governing its development in humans. Furthermore, we highlight when and in which cerebellar neurons the ASD-associated genes may play a role in the development of cortico-cerebellar circuits. Finally, we discuss improvements in protocols for generating cerebellar organoids to recapitulate the long period of development and maturation of this organ. These models, if generated from patient-induced pluripotent stem cells (iPSC), could provide a valuable approach to elucidate the contribution of defective genes to ASD pathology and inform diagnostic and therapeutic strategies.
Collapse
Affiliation(s)
- Marika Guerra
- Department of Neuroscience, Section of Human Anatomy, Catholic University of the Sacred Hearth, 00168 Rome, Italy; (M.G.); (V.M.)
| | - Vanessa Medici
- Department of Neuroscience, Section of Human Anatomy, Catholic University of the Sacred Hearth, 00168 Rome, Italy; (M.G.); (V.M.)
| | - Gina La Sala
- Institute of Biochemistry and Cell Biology, Italian National Research Council (CNR), 00015 Monterotondo Scalo, Italy
| | - Donatella Farini
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, 00133 Rome, Italy
| |
Collapse
|
6
|
Pascual-Alonso A, Xiol C, Smirnov D, Kopajtich R, Prokisch H, Armstrong J. Multi-omics in MECP2 duplication syndrome patients and carriers. Eur J Neurosci 2024; 60:4004-4018. [PMID: 38746988 DOI: 10.1111/ejn.16389] [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: 01/16/2024] [Revised: 04/16/2024] [Accepted: 04/24/2024] [Indexed: 07/21/2024]
Abstract
MECP2 duplication syndrome (MDS) is an X-linked neurodevelopmental disorder caused by the gain of dose of at least the genes MECP2 and IRAK1 and is characterised by intellectual disability (ID), developmental delay, hypotonia, epilepsy and recurrent infections. It mainly affects males, and females can be affected or asymptomatic carriers. Rett syndrome (RTT) is mainly triggered by loss of function mutations in MECP2 and is a well described syndrome that presents ID, epilepsy, lack of purposeful hand use and impaired speech, among others. As a result of implementing omics technology, altered biological pathways in human RTT samples have been reported, but such molecular characterisation has not been performed in patients with MDS. We gathered human skin fibroblasts from 17 patients with MDS, 10 MECP2 duplication carrier mothers and 21 patients with RTT, and performed multi-omics (RNAseq and proteomics) analysis. Here, we provide a thorough description and compare the shared and specific dysregulated biological processes between the cohorts. We also highlight the genes TMOD2, SRGAP1, COPS2, CNPY2, IGF2BP1, MOB2, VASP, FZD7, ECSIT and KIF3B as biomarker and therapeutic target candidates due to their implication in neuronal functions. Defining the RNA and protein profiles has shown that our four cohorts are less alike than expected by their shared phenotypes.
Collapse
Affiliation(s)
- Ainhoa Pascual-Alonso
- Fundació per la Recerca Sant Joan de Déu, Esplugues de Llobregat, Spain
- Institut de Recerca Sant Joan de Déu, Esplugues de Llobregat, Spain
| | - Clara Xiol
- Fundació per la Recerca Sant Joan de Déu, Esplugues de Llobregat, Spain
- Institut de Recerca Sant Joan de Déu, Esplugues de Llobregat, Spain
| | - Dmitrii Smirnov
- Institute of Human Genetics, Technical University of Munich, Munich, Germany
- Institute of Neurogenomics, Helmholtz Zentrum München, Munich, Germany
| | - Rober Kopajtich
- Institute of Human Genetics, Technical University of Munich, Munich, Germany
- Institute of Neurogenomics, Helmholtz Zentrum München, Munich, Germany
| | - Holger Prokisch
- Institute of Human Genetics, Technical University of Munich, Munich, Germany
- Institute of Neurogenomics, Helmholtz Zentrum München, Munich, Germany
| | - Judith Armstrong
- Institut de Recerca Sant Joan de Déu, Esplugues de Llobregat, Spain
- Instituto de Salud Carlos III (ISCIII), CIBER-ER (Biomedical Network Research Center for Rare Diseases), Madrid, Spain
- Genomic Unit, Molecular and Genetic Medicine Section, Hospital Sant Joan de Déu, Esplugues de Llobregat, Spain
| |
Collapse
|
7
|
Maino E, Scott O, Rizvi SZ, Chan WS, Visuvanathan S, Zablah YB, Li H, Sengar AS, Salter MW, Jia Z, Rossant J, Cohn RD, Gu B, Ivakine EA. An Irak1-Mecp2 tandem duplication mouse model for the study of MECP2 duplication syndrome. Dis Model Mech 2024; 17:dmm050528. [PMID: 38881329 PMCID: PMC11552499 DOI: 10.1242/dmm.050528] [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: 10/16/2023] [Accepted: 06/06/2024] [Indexed: 06/18/2024] Open
Abstract
MECP2 duplication syndrome (MDS) is a neurodevelopmental disorder caused by tandem duplication of the MECP2 locus and its surrounding genes, including IRAK1. Current MDS mouse models involve transgenic expression of MECP2 only, limiting their applicability to the study of the disease. Herein, we show that an efficient and precise CRISPR/Cas9 fusion proximity-based approach can be utilized to generate an Irak1-Mecp2 tandem duplication mouse model ('Mecp2 Dup'). The Mecp2 Dup mouse model recapitulates the genomic landscape of human MDS by harboring a 160 kb tandem duplication encompassing Mecp2 and Irak1, representing the minimal disease-causing duplication, and the neighboring genes Opn1mw and Tex28. The Mecp2 Dup model exhibits neuro-behavioral abnormalities, and an abnormal immune response to infection not previously observed in other mouse models, possibly owing to Irak1 overexpression. The Mecp2 Dup model thus provides a tool to investigate MDS disease mechanisms and develop potential therapies applicable to patients.
Collapse
Affiliation(s)
- Eleonora Maino
- Program in Genetics and Genome Biology, the Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Ori Scott
- Program in Genetics and Genome Biology, the Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
- Division of Clinical Immunology and Allergy, Department of Pediatrics, the Hospital for Sick Children and University of Toronto, Toronto, ON M5G 1E8, Canada
| | - Samar Z. Rizvi
- Program in Genetics and Genome Biology, the Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Wing Suen Chan
- Program in Genetics and Genome Biology, the Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
| | - Shagana Visuvanathan
- Program in Genetics and Genome Biology, the Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
| | - Youssif Ben Zablah
- Department of Physiology, University of Toronto, Toronto, ON M5S 1A8, Canada
- Program in Neuroscience and Mental Health, the Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
| | - Hongbin Li
- Program in Neuroscience and Mental Health, the Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
| | - Ameet S. Sengar
- Program in Neuroscience and Mental Health, the Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
| | - Michael W. Salter
- Department of Physiology, University of Toronto, Toronto, ON M5S 1A8, Canada
- Program in Neuroscience and Mental Health, the Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
| | - Zhengping Jia
- Department of Physiology, University of Toronto, Toronto, ON M5S 1A8, Canada
- Program in Neuroscience and Mental Health, the Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
| | - Janet Rossant
- Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada
- Program in Developmental and Stem Cell Biology, the Hospital for Sick Children Research Institute, Toronto, ON M5G 0A4, Canada
| | - Ronald D. Cohn
- Program in Genetics and Genome Biology, the Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada
- Division of Clinical Immunology and Allergy, Department of Pediatrics, the Hospital for Sick Children and University of Toronto, Toronto, ON M5G 1E8, Canada
- Division of Clinical and Metabolic Genetics, Department of Pediatrics, the Hospital for Sick Children and University of Toronto, Toronto, ON M5G 1X8, Canada
| | - Bin Gu
- Department of Obstetrics, Gynecology and Reproductive Biology, Michigan State University, East Lansing, MI 48824, USA
- Institute for Quantitative Health Science and Engineering, Michigan State University, East Lansing, MI 48824, USA
| | - Evgueni A. Ivakine
- Program in Genetics and Genome Biology, the Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada
- Department of Physiology, University of Toronto, Toronto, ON M5S 1A8, Canada
| |
Collapse
|
8
|
Levy M, Elron E, Shohat M, Lifshitz S, Kahana S, Shani H, Grossman A, Amar S, Narkis G, Sagi-Dain L, Basel-Salmon L, Maya I. Exploring inheritance, and clinical penetrance of distal Xq28 duplication syndrome: insights from 47 new unpublished cases. J Hum Genet 2024; 69:337-343. [PMID: 38632380 PMCID: PMC11199134 DOI: 10.1038/s10038-024-01252-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2023] [Revised: 03/29/2024] [Accepted: 04/01/2024] [Indexed: 04/19/2024]
Abstract
BACKGROUND Distal Xq28 duplication, or int22h1/int22h2-mediated Xq28 duplication syndrome, leads to cognitive impairment, neurobehavioral issues, and facial dysmorphisms. Existing literature has limited information on clinical traits and penetrance. METHODS We identified cases of distal Xq28 duplication (chrX: 154,126,575-154,709,680, GRCh37/hg19) through a review of clinical records and microarray reports from five centers, encompassing both postnatal and prenatal cases, with no prior family knowledge of the duplication. RESULTS Our search found 47 cases across 26 families, with duplications ranging from 208 to 935 Kb. In total, 8 out of 26 index cases featured a 200-300 kb partial duplication, mainly from Armenian/Caucasian Jewish backgrounds. Most prenatal cases showed no major fetal ultrasound malformations. Of cases with known inheritance mode (15 out of 26), maternal inheritance was more common (80%). The study identified seven male carriers of the duplication from six unrelated families, indicating partial penetrance in males. CONCLUSION Our study provides key insights into distal Xq28 duplication. Most prenatal tests showed no major fetal ultrasound issues. Maternal inheritance was common, with unaffected mothers. In the postnatal group, a balanced gender distribution was observed. Among male family members, two fathers had ADHD, one was healthy, and one brother had mild symptoms, indicating partial penetrance in males.
Collapse
Affiliation(s)
- Michal Levy
- The Raphael Recanati Genetics Institute, Rabin Medical Center, Beilinson Campus, Petah Tikva, Israel.
- Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.
| | - Eyal Elron
- The Raphael Recanati Genetics Institute, Rabin Medical Center, Beilinson Campus, Petah Tikva, Israel
- Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Mordechai Shohat
- Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
- Maccabi Genetic Institute & Bioinformatics Unit, Sheba Cancer Research Center, Ramat Gan, Israel
| | - Shira Lifshitz
- Maccabi Genetic Institute & Bioinformatics Unit, Sheba Cancer Research Center, Ramat Gan, Israel
| | - Sarit Kahana
- The Raphael Recanati Genetics Institute, Rabin Medical Center, Beilinson Campus, Petah Tikva, Israel
| | - Hagit Shani
- The Danek Gertner Institute of Human Genetics, Sheba Medical Center, Ramat Gan, Israel
| | - Anat Grossman
- The Danek Gertner Institute of Human Genetics, Sheba Medical Center, Ramat Gan, Israel
| | - Shirly Amar
- Genetic Institute, Soroka Medical Center & Ben Gurion University, Be'er Sheva, Israel
| | - Ginat Narkis
- Genetic Institute, Soroka Medical Center & Ben Gurion University, Be'er Sheva, Israel
| | - Lena Sagi-Dain
- Genetics Institute, Department of Obstetrics and Gynecology, Carmel Medical Center & The Ruth and Bruce Rappaport Faculty of Medicine, Technion, Haifa, Israel
| | - Lina Basel-Salmon
- The Raphael Recanati Genetics Institute, Rabin Medical Center, Beilinson Campus, Petah Tikva, Israel
- Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
- Felsenstein Medical Research Center, Tel Aviv University, Tel Aviv, Israel
| | - Idit Maya
- The Raphael Recanati Genetics Institute, Rabin Medical Center, Beilinson Campus, Petah Tikva, Israel
- Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
9
|
Suter B, Pehlivan D, Ak M, Harris HK, Lyons-Warren AM. Sensory experiences questionnaire unravels differences in sensory profiles between MECP2-related disorders. Autism Res 2024; 17:775-784. [PMID: 38433353 PMCID: PMC11127745 DOI: 10.1002/aur.3112] [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: 10/16/2023] [Accepted: 02/02/2024] [Indexed: 03/05/2024]
Abstract
The methyl CpG-binding protein-2 (MECP2) gene is located on the Xq28 region. Loss of function mutations or increased copies of MECP2 result in Rett syndrome (RTT) and MECP2 duplication syndrome (MDS), respectively. Individuals with both disorders exhibit overlapping autism symptoms, yet few studies have dissected the differences between these gene dosage sensitive disorders. Further, research examining sensory processing patterns in persons with RTT and MDS is largely absent. Thus, the goal of this study was to analyze and compare sensory processing patterns in persons with RTT and MDS. Towards this goal, caregivers of 50 female individuals with RTT and 122 male individuals with MDS, between 1 and 46 years of age, completed a standardized measure of sensory processing, the Sensory Experiences Questionnaire. Patterns detected in both disorders were compared against each other and against normative values. We found sensory processing abnormalities for both hyper- and hypo-sensitivity in both groups. Interestingly, abnormalities in MDS were more pronounced compared with in RTT, particularly with items concerning hypersensitivity and sensory seeking, but not hyposensitivity. Individuals with MDS also exhibited greater sensory symptoms compared with RTT in the areas of tactile and vestibular sensory processing and for both social and nonsocial stimuli. This study provides a first description of sensory symptoms in individuals with RTT and individuals with MDS. Similar to other neurodevelopmental disorders, a variety of sensory processing abnormalities was found. These findings reveal a first insight into sensory processing abnormalities caused by a dosage sensitive gene and may ultimately help guide therapeutic approaches for these disorders.
Collapse
Affiliation(s)
- Bernhard Suter
- Section of Pediatric Neurology and Developmental Neuroscience, Department of Pediatrics, Baylor College of Medicine, Houston, Texas, USA
- Blue Bird Circle Rett Center, Texas Children's Hospital, Houston, Texas, USA
| | - Davut Pehlivan
- Section of Pediatric Neurology and Developmental Neuroscience, Department of Pediatrics, Baylor College of Medicine, Houston, Texas, USA
- Blue Bird Circle Rett Center, Texas Children's Hospital, Houston, Texas, USA
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, Texas, USA
| | - Muharrem Ak
- Section of Pediatric Neurology and Developmental Neuroscience, Department of Pediatrics, Baylor College of Medicine, Houston, Texas, USA
| | - Holly K Harris
- Section of Developmental and Behavioral Pediatrics, Department of Pediatrics, Baylor College of Medicine, Houston, Texas, USA
| | - Ariel M Lyons-Warren
- Section of Pediatric Neurology and Developmental Neuroscience, Department of Pediatrics, Baylor College of Medicine, Houston, Texas, USA
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, Texas, USA
| |
Collapse
|
10
|
Holover G, Adams D, Milligan D, Goldberg R, Rios J, Kornitzer J, Mazzola C. Moya moya vasculopathy and MECP2 duplication syndrome. Childs Nerv Syst 2024; 40:809-812. [PMID: 37804337 DOI: 10.1007/s00381-023-06139-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Accepted: 08/26/2023] [Indexed: 10/09/2023]
Abstract
BACKGROUND Moya moya type vasculopathy (MMV) is a rare disorder in which there is narrowing of bilateral intracranial carotid arteries (Scott and Smith in New Engl J Med 360(12):1226-1237, 2009). MECP2 duplication syndrome (MDS) is a rare genetic disorder that is caused by genetic duplications on Xq28 chromosome (Expanding the clinical picture of the MECP2 duplication syndrome. (Lim et al. in Clin Genet 91(4):557-563, 2017). Both disorders are rare and have not been described together in association. CASE PRESENTATION Interestingly, we present a child with both MDS and MMV. Upon genetic testing, there was found to be a large, de novo duplication sequence in the patient's genome. Possible correlation between our patient's extensive genetic mutation and MMV has been evaluated. CONCLUSION Our literature search disclosed no other known patients with both MDS and MMV. Patients with MDS should be monitored carefully for signs or symptoms of vasculopathy.
Collapse
Affiliation(s)
- Gianna Holover
- School of Arts and Science, Rutgers University, New Brunswick, NJ, USA
| | - Darius Adams
- Department of Genetics, Personalized Genomic Medicine, Morristown, NJ, USA
| | - Dawn Milligan
- Department of Neurological Surgery, New Jersey Pediatric Neuroscience Institute, 131 Madison Ave 3rd Floor, Morristown, NJ, 07960, USA
| | - Rina Goldberg
- Department of Pediatric Comprehensive Epilepsy Center, Institute of Neurology and Neurosurgery, Livingston, NJ, USA
| | - Jose Rios
- Department of Radiology, Atlantic Medical Group Radiology, Morristown, NJ, USA
| | - Jeffrey Kornitzer
- Department of Neurology, New Jersey Pediatric Neuroscience Institute, Morristown, NJ, USA
| | - Catherine Mazzola
- Department of Neurological Surgery, New Jersey Pediatric Neuroscience Institute, 131 Madison Ave 3rd Floor, Morristown, NJ, 07960, USA.
| |
Collapse
|
11
|
Zito A, Lee JT. Variable expression of MECP2, CDKL5, and FMR1 in the human brain: Implications for gene restorative therapies. Proc Natl Acad Sci U S A 2024; 121:e2312757121. [PMID: 38386709 PMCID: PMC10907246 DOI: 10.1073/pnas.2312757121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Accepted: 12/28/2023] [Indexed: 02/24/2024] Open
Abstract
MECP2, CDKL5, and FMR1 are three X-linked neurodevelopmental genes associated with Rett, CDKL5-, and fragile-X syndrome, respectively. These syndromes are characterized by distinct constellations of severe cognitive and neurobehavioral anomalies, reflecting the broad but unique expression patterns of each of the genes in the brain. As these disorders are not thought to be neurodegenerative and may be reversible, a major goal has been to restore expression of the functional proteins in the patient's brain. Strategies have included gene therapy, gene editing, and selective Xi-reactivation methodologies. However, tissue penetration and overall delivery to various regions of the brain remain challenging for each strategy. Thus, gaining insights into how much restoration would be required and what regions/cell types in the brain must be targeted for meaningful physiological improvement would be valuable. As a step toward addressing these questions, here we perform a meta-analysis of single-cell transcriptomics data from the human brain across multiple developmental stages, in various brain regions, and in multiple donors. We observe a substantial degree of expression variability for MECP2, CDKL5, and FMR1 not only across cell types but also between donors. The wide range of expression may help define a therapeutic window, with the low end delineating a minimum level required to restore physiological function and the high end informing toxicology margin. Finally, the inter-cellular and inter-individual variability enable identification of co-varying genes and will facilitate future identification of biomarkers.
Collapse
Affiliation(s)
- Antonino Zito
- Department of Molecular Biology, Massachusetts General Hospital, Boston, MA02114
- Department of Genetics, The Blavatnik Institute, Harvard Medical School, Boston, MA02114
| | - Jeannie T. Lee
- Department of Molecular Biology, Massachusetts General Hospital, Boston, MA02114
- Department of Genetics, The Blavatnik Institute, Harvard Medical School, Boston, MA02114
| |
Collapse
|
12
|
Zeng L, Zhu H, Wang J, Wang Q, Pang Y, Luo Z, Chen A, Qin S, Zhu S. Genetic analysis of a pedigree with MECP2 duplication syndrome in China. BMC Med Genomics 2024; 17:54. [PMID: 38373942 PMCID: PMC10875745 DOI: 10.1186/s12920-024-01831-9] [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: 02/25/2023] [Accepted: 02/10/2024] [Indexed: 02/21/2024] Open
Abstract
BACKGROUND MECP2 duplication syndrome (MDS) is a rare X-linked genomic disorder that primarily affects males. It is characterized by delayed or absent speech development, severe motor and cognitive impairment, and recurrent respiratory infections. MDS is caused by the duplication of a chromosomal region located on chromosome Xq28, which contains the methyl CpG binding protein-2 (MECP2) gene. MECP2 functions as a transcriptional repressor or activator, regulating genes associated with nervous system development. The objective of this study is to provide a clinical description of MDS, including imaging changes observed from the fetal period to the neonatal period. METHODS Conventional G-banding was employed to analyze the chromosome karyotypes of all pedigrees under investigation. Subsequently, whole exome sequencing (WES), advanced biological information analysis, and pedigree validation were conducted, which were further confirmed by copy number variation sequencing (CNV-seq). RESULTS Chromosome karyotype analysis revealed that a male patient had a chromosome karyotype of 46,Y,dup(X)(q27.2q28). Whole-exon duplication in the MECP2 gene was revealed through WES results. CNV-seq validation confirmed the presence of Xq27.1q28 duplicates spanning 14.45 Mb, which was inherited from a mild phenotype mother. Neither the father nor the mother's younger brother carried this duplication. CONCLUSION In this study, we examined a male child in a family who exhibited developmental delay and recurrent respiratory tract infections as the main symptoms. We conducted thorough family investigations and genetic testing to determine the underlying causes of the disease. Our findings will aid in early diagnosis, genetic counseling for male patients in this family, as well as providing prenatal diagnosis and reproductive guidance for female carriers.
Collapse
Affiliation(s)
- Lan Zeng
- Department of Medical Genetics and Prenatal Diagnosis, Sichuan Provincial Maternity and Child Health Care Hospital, Chengdu, Sichuan, China
| | - Hui Zhu
- Department of Pediatrics, Sichuan Provincial Maternity and Child Health Care Hospital, No. 290, Sha Yan West 2Nd Road, Chengdu, 610031, Sichuan, China
| | - Jin Wang
- Department of Medical Genetics and Prenatal Diagnosis, Sichuan Provincial Maternity and Child Health Care Hospital, Chengdu, Sichuan, China
| | - Qiyan Wang
- Department of Radiology, Sichuan Provincial Maternity and Child Health Care Hospital, Chengdu, Sichuan, China
| | - Ying Pang
- Department of Pediatrics, Sichuan Provincial Maternity and Child Health Care Hospital, No. 290, Sha Yan West 2Nd Road, Chengdu, 610031, Sichuan, China
| | - Zemin Luo
- Department of Pediatrics, Sichuan Provincial Maternity and Child Health Care Hospital, No. 290, Sha Yan West 2Nd Road, Chengdu, 610031, Sichuan, China
| | - Ai Chen
- Department of Pediatrics, Sichuan Provincial Maternity and Child Health Care Hospital, No. 290, Sha Yan West 2Nd Road, Chengdu, 610031, Sichuan, China
| | - Shengfang Qin
- Department of Medical Genetics and Prenatal Diagnosis, Sichuan Provincial Maternity and Child Health Care Hospital, Chengdu, Sichuan, China
| | - Shuyao Zhu
- Department of Pediatrics, Sichuan Provincial Maternity and Child Health Care Hospital, No. 290, Sha Yan West 2Nd Road, Chengdu, 610031, Sichuan, China.
| |
Collapse
|
13
|
Bogatova D, Smirnakis SM, Palagina G. Tug-of-Peace: Visual Rivalry and Atypical Visual Motion Processing in MECP2 Duplication Syndrome of Autism. eNeuro 2024; 11:ENEURO.0102-23.2023. [PMID: 37940561 PMCID: PMC10792601 DOI: 10.1523/eneuro.0102-23.2023] [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: 03/23/2023] [Revised: 06/25/2023] [Accepted: 08/12/2023] [Indexed: 11/10/2023] Open
Abstract
Extracting common patterns of neural circuit computations in the autism spectrum and confirming them as a cause of specific core traits of autism is the first step toward identifying cell-level and circuit-level targets for effective clinical intervention. Studies in humans with autism have identified functional links and common anatomic substrates between core restricted behavioral repertoire, cognitive rigidity, and overstability of visual percepts during visual rivalry. To study these processes with single-cell precision and comprehensive neuronal population coverage, we developed the visual bistable perception paradigm for mice based on ambiguous moving plaid patterns consisting of two transparent gratings drifting at an angle of 120°. This results in spontaneous reversals of the perception between local component motion (plaid perceived as two separate moving grating components) and integrated global pattern motion (plaid perceived as a fused moving texture). This robust paradigm does not depend on the explicit report of the mouse, since the direction of the optokinetic nystagmus (OKN) is used to infer the dominant percept. Using this paradigm, we found that the rate of perceptual reversals between global and local motion interpretations is reduced in the methyl-CpG-binding protein 2 duplication syndrome (MECP2-ds) mouse model of autism. Moreover, the stability of local motion percepts is greatly increased in MECP2-ds mice at the expense of global motion percepts. Thus, our model reproduces a subclass of the core features in human autism (reduced rate of visual rivalry and atypical perception of visual motion). This further offers a well-controlled approach for dissecting neuronal circuits underlying these core features.
Collapse
Affiliation(s)
- Daria Bogatova
- Department of Neurology, Brigham and Women's Hospital, Boston, MA 02115
- Department of Biology, Boston University, Boston, MA 02115
- Harvard Medical School, Boston, MA 02115
| | - Stelios M Smirnakis
- Department of Neurology, Brigham and Women's Hospital, Boston, MA 02115
- Harvard Medical School, Boston, MA 02115
- Jamaica Plain Veterans Affairs Hospital, Boston, MA 02130
| | - Ganna Palagina
- Department of Neurology, Brigham and Women's Hospital, Boston, MA 02115
- Harvard Medical School, Boston, MA 02115
- Jamaica Plain Veterans Affairs Hospital, Boston, MA 02130
| |
Collapse
|
14
|
Wu J, Hu Q, Rao X, Zhao H, Tang H, Wang Y. Gut microbiome and metabolic profiles of mouse model for MeCP2 duplication syndrome. Brain Res Bull 2024; 206:110862. [PMID: 38145758 DOI: 10.1016/j.brainresbull.2023.110862] [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: 09/07/2023] [Revised: 12/18/2023] [Accepted: 12/20/2023] [Indexed: 12/27/2023]
Abstract
The extra copy of the methyl-CpG-binding protein 2 (MeCp2) gene causes MeCP2 duplication syndrome (MDS), a neurodevelopmental disorder characterized by intellectual disability and autistic phenotypes. However, the disturbed microbiome and metabolic profiling underlying the autistic-like behavioral deficits of MDS are rarely investigated. Here we aimed to understand the contributions of microbiome disruption and associated metabolic alterations, especially the disturbed neurotransmitters in MDS employing a transgenic mouse model with MeCP2 overexpression. We analyzed metabolic profiles of plasma, urine, and cecum content and microbiome profiles by both 16 s RNA and shotgun metagenomics sequence technology. We found the decreased levels of Firmicutes and increased levels of Bacteroides in the single MeCP2 gene mutation autism-like mouse model, demonstrating the importance of the host genome in a selection of microbiome, leading to the heterogeneity characteristics of microbiome in MDS. Furthermore, the changed levels of several neurotransmitters (such as dopamine, taurine, and glutamate) implied the excitatory-inhibitory imbalance caused by the single gene mutation. Concurrently, a range of microbial metabolisms of aromatic amino acids (such as tryptophan and phenylalanine) were identified in different biological matrices obtained from MeCP2 transgenic mice. Our investigation revealed the importance of genetic variation in accounting for the differences in microbiomes and confirmed the bidirectional regulatory axis of microbiota-gut-brain in studying the role of microbiome on MDS, which could be useful in deeply understanding the microbiome-based treatment in this autistic-like disease.
Collapse
Affiliation(s)
- Junfang Wu
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan 430000, China.
| | - Qingyu Hu
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Human Phenome Institute, Metabonomics and Systems Biology Laboratory at Shanghai International Centre for Molecular Phenomics, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Xiaoping Rao
- State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, Key Laboratory of Magnetic Resonance in Biological Systems, Wuhan Center for Magnetic Resonance, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences, Wuhan 430000, China
| | - Hongyang Zhao
- Department of Pediatrics, Central Hospital Affiliated to Shandong First Medical University, Jinan 250013, China
| | - Huiru Tang
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Human Phenome Institute, Metabonomics and Systems Biology Laboratory at Shanghai International Centre for Molecular Phenomics, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Yulan Wang
- Singapore Phenome Center, Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore 639798, Singapore.
| |
Collapse
|
15
|
Kalani L, Kim BH, Vincent JB, Ausió J. MeCP2 ubiquitination and sumoylation, in search of a function†. Hum Mol Genet 2023; 33:1-11. [PMID: 37694858 DOI: 10.1093/hmg/ddad150] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Revised: 09/06/2023] [Accepted: 09/07/2023] [Indexed: 09/12/2023] Open
Abstract
MeCP2 (Methyl CpG binding protein 2) is an intrinsically disordered protein that binds to methylated genome regions. The protein is a critical transcriptional regulator of the brain, and its mutations account for 95% of Rett syndrome (RTT) cases. Early studies of this neurodevelopmental disorder revealed a close connection with dysregulations of the ubiquitin system (UbS), notably as related to UBE3A, a ubiquitin ligase involved in the proteasome-mediated degradation of proteins. MeCP2 undergoes numerous post-translational modifications (PTMs), including ubiquitination and sumoylation, which, in addition to the potential functional outcomes of their monomeric forms in gene regulation and synaptic plasticity, in their polymeric organization, these modifications play a critical role in proteasomal degradation. UbS-mediated proteasomal degradation is crucial in maintaining MeCP2 homeostasis for proper function and is involved in decreasing MeCP2 in some RTT-causing mutations. However, regardless of all these connections to UbS, the molecular details involved in the signaling of MeCP2 for its targeting by the ubiquitin-proteasome system (UPS) and the functional roles of monomeric MeCP2 ubiquitination and sumoylation remain largely unexplored and are the focus of this review.
Collapse
Affiliation(s)
- Ladan Kalani
- Department of Biochemistry and Microbiology, University of Victoria, 3800 Finnerty Rd, Victoria, BC V8W 2Y2, Canada
| | - Bo-Hyun Kim
- Department of Biochemistry and Microbiology, University of Victoria, 3800 Finnerty Rd, Victoria, BC V8W 2Y2, Canada
| | - John B Vincent
- Molecular Neuropsychiatry & Development (MiND) Lab, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, 250 College St, Toronto, ON M5T 1R8, Canada
- Institute of Medical Science, University of Toronto, 27 King's College Cir, Toronto, ON M5S 1A8, Canada
| | - Juan Ausió
- Department of Biochemistry and Microbiology, University of Victoria, 3800 Finnerty Rd, Victoria, BC V8W 2Y2, Canada
| |
Collapse
|
16
|
Wang Y, Liu Y, Kuo Y, Guan S, Wang N, Lian Y, Huang J, Zhi X, Liu P, Li R, Yan L, Zhu X, Qiao J. Clinical practice and outcomes of preimplantation genetic testing for CMT1A using a novel direct detection method. Heliyon 2023; 9:e22196. [PMID: 38045147 PMCID: PMC10692806 DOI: 10.1016/j.heliyon.2023.e22196] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2023] [Revised: 09/22/2023] [Accepted: 11/06/2023] [Indexed: 12/05/2023] Open
Abstract
Background Charcot-Marie-Tooth type 1A (CMT1A), the most frequent type of Charcot-Marie-Tooth disease, is mainly caused by a 1.4-Mb duplication containing the PMP22 gene. There is no effective treatment other than general supportive care and symptomatic treatment. Preimplantation genetic testing for monogenic defects (PGT-M) is an alternative approach for obtaining healthy babies. Methods A new technology and analysis method based on next-generation sequencing (NGS) was developed to detect duplication mutations directly. Simultaneously, aneuploidy and linkage analyses were performed to achieve a comprehensive and accurate embryo diagnosis. Results Eight couples were recruited in this study; PMP22 duplication was validated in seven couples, and PMP22 splicing mutation was found in one. Forty-five embryos from 12 PGT cycles were successfully detected using this novel method. The direct detection results for all embryos were consistent with the linkage analyses, suggesting a 100 % accuracy rate, and the aneuploidy rate of the biopsied blastocysts was 33.3 %. Eventually, 18 of the 45 diagnosed embryos were deemed suitable for transfer. Four healthy babies from three families were delivered and their genetic status confirmed by amniocentesis. Additionally, there were no adverse effects of anesthesia or increased pregnancy complications during PGT-M in female patients with CMT1A. Conclusions This study provided a simple, reliable, and efficient method that can directly detect PMP22 mutations based on NGS data and does not require positive family members. A clinical workflow for CMT1A interruption in the offspring before embryo implantation is also summarized.
Collapse
Affiliation(s)
- Yuqian Wang
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, 100191, China
- National Clinical Research Center for Obstetrics and Gynecology (Peking University Third Hospital), Beijing, 100191, China
- Key Laboratory of Assisted Reproduction (Peking University), Ministry of Education, Beijing, 100191, China
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, 100191, China
- Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, 100191, China
| | - Yujun Liu
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, 100191, China
- National Clinical Research Center for Obstetrics and Gynecology (Peking University Third Hospital), Beijing, 100191, China
- Key Laboratory of Assisted Reproduction (Peking University), Ministry of Education, Beijing, 100191, China
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, 100191, China
| | - Ying Kuo
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, 100191, China
- National Clinical Research Center for Obstetrics and Gynecology (Peking University Third Hospital), Beijing, 100191, China
- Key Laboratory of Assisted Reproduction (Peking University), Ministry of Education, Beijing, 100191, China
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, 100191, China
| | - Shuo Guan
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, 100191, China
- National Clinical Research Center for Obstetrics and Gynecology (Peking University Third Hospital), Beijing, 100191, China
- Key Laboratory of Assisted Reproduction (Peking University), Ministry of Education, Beijing, 100191, China
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, 100191, China
| | - Nan Wang
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, 100191, China
- National Clinical Research Center for Obstetrics and Gynecology (Peking University Third Hospital), Beijing, 100191, China
- Key Laboratory of Assisted Reproduction (Peking University), Ministry of Education, Beijing, 100191, China
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, 100191, China
| | - Ying Lian
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, 100191, China
- National Clinical Research Center for Obstetrics and Gynecology (Peking University Third Hospital), Beijing, 100191, China
- Key Laboratory of Assisted Reproduction (Peking University), Ministry of Education, Beijing, 100191, China
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, 100191, China
| | - Jin Huang
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, 100191, China
- National Clinical Research Center for Obstetrics and Gynecology (Peking University Third Hospital), Beijing, 100191, China
- Key Laboratory of Assisted Reproduction (Peking University), Ministry of Education, Beijing, 100191, China
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, 100191, China
| | - Xu Zhi
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, 100191, China
- National Clinical Research Center for Obstetrics and Gynecology (Peking University Third Hospital), Beijing, 100191, China
- Key Laboratory of Assisted Reproduction (Peking University), Ministry of Education, Beijing, 100191, China
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, 100191, China
| | - Ping Liu
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, 100191, China
- National Clinical Research Center for Obstetrics and Gynecology (Peking University Third Hospital), Beijing, 100191, China
- Key Laboratory of Assisted Reproduction (Peking University), Ministry of Education, Beijing, 100191, China
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, 100191, China
| | - Rong Li
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, 100191, China
- National Clinical Research Center for Obstetrics and Gynecology (Peking University Third Hospital), Beijing, 100191, China
- Key Laboratory of Assisted Reproduction (Peking University), Ministry of Education, Beijing, 100191, China
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, 100191, China
| | - Liying Yan
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, 100191, China
- National Clinical Research Center for Obstetrics and Gynecology (Peking University Third Hospital), Beijing, 100191, China
- Key Laboratory of Assisted Reproduction (Peking University), Ministry of Education, Beijing, 100191, China
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, 100191, China
| | - Xiaohui Zhu
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, 100191, China
- National Clinical Research Center for Obstetrics and Gynecology (Peking University Third Hospital), Beijing, 100191, China
- Key Laboratory of Assisted Reproduction (Peking University), Ministry of Education, Beijing, 100191, China
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, 100191, China
| | - Jie Qiao
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, 100191, China
- National Clinical Research Center for Obstetrics and Gynecology (Peking University Third Hospital), Beijing, 100191, China
- Key Laboratory of Assisted Reproduction (Peking University), Ministry of Education, Beijing, 100191, China
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, 100191, China
- Beijing Advanced Innovation Center for Genomics, Beijing, 100191, China
- Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, 100191, China
| |
Collapse
|
17
|
Pascual-Alonso A, Xiol C, Smirnov D, Kopajtich R, Prokisch H, Armstrong J. Identification of molecular signatures and pathways involved in Rett syndrome using a multi-omics approach. Hum Genomics 2023; 17:85. [PMID: 37710353 PMCID: PMC10503149 DOI: 10.1186/s40246-023-00532-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Accepted: 09/03/2023] [Indexed: 09/16/2023] Open
Abstract
BACKGROUND Rett syndrome (RTT) is a neurodevelopmental disorder mainly caused by mutations in the methyl-CpG-binding protein 2 gene (MECP2). MeCP2 is a multi-functional protein involved in many cellular processes, but the mechanisms by which its dysfunction causes disease are not fully understood. The duplication of the MECP2 gene causes a distinct disorder called MECP2 duplication syndrome (MDS), highlighting the importance of tightly regulating its dosage for proper cellular function. Additionally, some patients with mutations in genes other than MECP2 exhibit phenotypic similarities with RTT, indicating that these genes may also play a role in similar cellular functions. The purpose of this study was to characterise the molecular alterations in patients with RTT in order to identify potential biomarkers or therapeutic targets for this disorder. METHODS We used a combination of transcriptomics (RNAseq) and proteomics (TMT mass spectrometry) to characterise the expression patterns in fibroblast cell lines from 22 patients with RTT and detected mutation in MECP2, 15 patients with MDS, 12 patients with RTT-like phenotypes and 13 healthy controls. Transcriptomics and proteomics data were used to identify differentially expressed genes at both RNA and protein levels, which were further inspected via enrichment and upstream regulator analyses and compared to find shared features in patients with RTT. RESULTS We identified molecular alterations in cellular functions and pathways that may contribute to the disease phenotype in patients with RTT, such as deregulated cytoskeletal components, vesicular transport elements, ribosomal subunits and mRNA processing machinery. We also compared RTT expression profiles with those of MDS seeking changes in opposite directions that could lead to the identification of MeCP2 direct targets. Some of the deregulated transcripts and proteins were consistently affected in patients with RTT-like phenotypes, revealing potentially relevant molecular processes in patients with overlapping traits and different genetic aetiology. CONCLUSIONS The integration of data in a multi-omics analysis has helped to interpret the molecular consequences of MECP2 dysfunction, contributing to the characterisation of the molecular landscape in patients with RTT. The comparison with MDS provides knowledge of MeCP2 direct targets, whilst the correlation with RTT-like phenotypes highlights processes potentially contributing to the pathomechanism leading these disorders.
Collapse
Affiliation(s)
- Ainhoa Pascual-Alonso
- Fundació Per La Recerca Sant Joan de Déu, Esplugues de Llobregat, Spain
- Institut de Recerca Sant Joan de Déu, Esplugues de Llobregat, Spain
| | - Clara Xiol
- Fundació Per La Recerca Sant Joan de Déu, Esplugues de Llobregat, Spain
- Institut de Recerca Sant Joan de Déu, Esplugues de Llobregat, Spain
| | - Dmitrii Smirnov
- Institute of Human Genetics, Technical University of Munich, Munich, Germany
- Institute of Neurogenomics, Helmholtz Zentrum München, Munich, Germany
| | - Robert Kopajtich
- Institute of Human Genetics, Technical University of Munich, Munich, Germany
- Institute of Neurogenomics, Helmholtz Zentrum München, Munich, Germany
| | - Holger Prokisch
- Institute of Human Genetics, Technical University of Munich, Munich, Germany
- Institute of Neurogenomics, Helmholtz Zentrum München, Munich, Germany
| | - Judith Armstrong
- Institut de Recerca Sant Joan de Déu, Esplugues de Llobregat, Spain.
- CIBER-ER (Biomedical Network Research Center for Rare Diseases), Instituto de Salud Carlos III (ISCIII), Madrid, Spain.
- Genomic Unit, Molecular and Genetic Medicine Section, Hospital Sant Joan de Déu, Barcelona, Spain.
| |
Collapse
|
18
|
John Cherian D, Ta D, Smith J, Downs J, Leonard H. How Families Manage the Complex Medical Needs of Their Children with MECP2 Duplication Syndrome. CHILDREN (BASEL, SWITZERLAND) 2023; 10:1202. [PMID: 37508699 PMCID: PMC10377896 DOI: 10.3390/children10071202] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Revised: 06/27/2023] [Accepted: 07/10/2023] [Indexed: 07/30/2023]
Abstract
MECP2 duplication syndrome (MDS) is a rare, X-linked, neurodevelopmental disorder resulting from the duplication of the methyl-CpG-binding protein 2 (MECP2) gene. The clinical features of MDS include severe intellectual disability, global developmental delay, seizures, recurrent respiratory infections, and gastrointestinal problems. The aim of this qualitative study was to explore how the parents of children with MDS manage their child's seizures, recurrent respiratory infections, and gastrointestinal symptoms, and the impact on them as parents. The data were coded into three categories: (1) complex care needs in the home, (2) highly skilled caregivers, and (3) impact on caregivers and families. Complex 24 h care was required and parents developed complex skillsets to ensure that this was delivered well to their child. The provision of extensive complex medical care in the home had an impact on parent mental and physical health, family dynamics, and finances. This study captures the management of high-burden comorbidities in MDS at home. Investigations into how best to support caregiver wellbeing to reduce their stresses, whilst maintaining optimal child health and wellbeing, are needed.
Collapse
Affiliation(s)
- Dani John Cherian
- School of Human Sciences, University of Western Australia, Perth 6009, Australia
- Telethon Kids Institute, Centre for Child Health Research, University of Western Australia, Perth 6872, Australia
| | - Daniel Ta
- Telethon Kids Institute, Centre for Child Health Research, University of Western Australia, Perth 6872, Australia
- School of Medicine, University of Western Australia, Perth 6009, Australia
| | - Jeremy Smith
- School of Human Sciences, University of Western Australia, Perth 6009, Australia
| | - Jenny Downs
- Telethon Kids Institute, Centre for Child Health Research, University of Western Australia, Perth 6872, Australia
- Curtin School of Allied Health, Curtin University, Perth 6845, Australia
| | - Helen Leonard
- Telethon Kids Institute, Centre for Child Health Research, University of Western Australia, Perth 6872, Australia
| |
Collapse
|
19
|
Singh J, Goodman-Vincent E, Santosh P. Evidence Synthesis of Gene Therapy and Gene Editing from Different Disorders-Implications for Individuals with Rett Syndrome: A Systematic Review. Int J Mol Sci 2023; 24:ijms24109023. [PMID: 37240368 DOI: 10.3390/ijms24109023] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Revised: 05/06/2023] [Accepted: 05/12/2023] [Indexed: 05/28/2023] Open
Abstract
This systematic review and thematic analysis critically evaluated gene therapy trials in amyotrophic lateral sclerosis, haemoglobinopathies, immunodeficiencies, leukodystrophies, lysosomal storage disorders and retinal dystrophies and extrapolated the key clinical findings to individuals with Rett syndrome (RTT). The PRISMA guidelines were used to search six databases during the last decade, followed by a thematic analysis to identify the emerging themes. Thematic analysis across the different disorders revealed four themes: (I) Therapeutic time window of gene therapy; (II) Administration and dosing strategies for gene therapy; (III) Methods of gene therapeutics and (IV) Future areas of clinical interest. Our synthesis of information has further enriched the current clinical evidence base and can assist in optimising gene therapy and gene editing studies in individuals with RTT, but it would also benefit when applied to other disorders. The findings suggest that gene therapies have better outcomes when the brain is not the primary target. Across different disorders, early intervention appears to be more critical, and targeting the pre-symptomatic stage might prevent symptom pathology. Intervention at later stages of disease progression may benefit by helping to clinically stabilise patients and preventing disease-related symptoms from worsening. If gene therapy or editing has the desired outcome, older patients would need concerted rehabilitation efforts to reverse their impairments. The timing of intervention and the administration route would be critical parameters for successful outcomes of gene therapy/editing trials in individuals with RTT. Current approaches also need to overcome the challenges of MeCP2 dosing, genotoxicity, transduction efficiencies and biodistribution.
Collapse
Affiliation(s)
- Jatinder Singh
- Department of Child and Adolescent Psychiatry, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London SE5 8AF, UK
- Centre for Interventional Paediatric Psychopharmacology and Rare Diseases (CIPPRD), South London and Maudsley NHS Foundation Trust, London SE5 8AZ, UK
- Centre for Interventional Paediatric Psychopharmacology (CIPP) Rett Centre, Institute of Psychiatry, Psychology and Neuroscience, King's College London and South London and Maudsley NHS Foundation Trust, London SE5 8AZ, UK
| | - Ella Goodman-Vincent
- Department of Child and Adolescent Psychiatry, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London SE5 8AF, UK
- Centre for Interventional Paediatric Psychopharmacology and Rare Diseases (CIPPRD), South London and Maudsley NHS Foundation Trust, London SE5 8AZ, UK
- Centre for Interventional Paediatric Psychopharmacology (CIPP) Rett Centre, Institute of Psychiatry, Psychology and Neuroscience, King's College London and South London and Maudsley NHS Foundation Trust, London SE5 8AZ, UK
| | - Paramala Santosh
- Department of Child and Adolescent Psychiatry, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London SE5 8AF, UK
- Centre for Interventional Paediatric Psychopharmacology and Rare Diseases (CIPPRD), South London and Maudsley NHS Foundation Trust, London SE5 8AZ, UK
- Centre for Interventional Paediatric Psychopharmacology (CIPP) Rett Centre, Institute of Psychiatry, Psychology and Neuroscience, King's College London and South London and Maudsley NHS Foundation Trust, London SE5 8AZ, UK
| |
Collapse
|
20
|
Fabio RA, Chiarini L, Canegallo V. Pain in Rett syndrome: a pilot study and a single case study on the assessment of pain and the construction of a suitable measuring scale. Orphanet J Rare Dis 2022; 17:356. [PMID: 36104823 PMCID: PMC9476284 DOI: 10.1186/s13023-022-02519-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2022] [Accepted: 09/05/2022] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Rett Syndrome (RTT) is a severe, neurodevelopmental disorder mainly caused by mutations in the MECP2 gene, affecting around 1 in 10,000 female births. Severe physical, language, and social impairments impose a wide range of limitations in the quality of life of the patients with RTT. Comorbidities of patients with RTT are varied and cause a lot of pain, but communicating this suffering is difficult for these patients due to their problems, such as apraxia that does not allow them to express pain in a timely manner, and their difficulties with expressive language that also do not permit them to communicate. Two studies, a pilot study and a single case study, investigate the manifestation of pain of patients with RTT and propose a suitable scale to measure it. AIMS OF THIS STUDY The first aim was to describe pain situations of RTT by collecting information by parents; the second aim was to test and compare existing questionnaires for non-communicating disorders on pain such as Pain assessment in advanced demenzia (PAINAD), the Critical care pain observation tool (CPOT) and the Non-communicating Children's Pain Checklist-Revised (NCCPC-R) to assess which of them is best related to the pain behavior of patients with RTT. The third aim was to identify the specific verbal and non-verbal behaviors that characterize pain in girls with Rett syndrome, discriminating them from non-pain behaviors. METHOD Nineteen participants, eighteen girls with RTT and one girl with RTT with 27 manifestations of pain were video-recorded both in pain and base-line conditions. Two independent observers codified the 90 video-recording (36 and 54) to describe their behavioral characteristics. RESULTS The two studies showed that the most significant pain behaviors expressed by girls with respect to the baseline condition, at the facial level were a wrinkled forehead, wide eyes, grinding, banging teeth, complaining, making sounds, crying and screaming, and the most common manifestations of the body were tremors, forward and backward movement of the torso, tension in the upper limbs, increased movement of the lower limbs and a sprawling movement affecting the whole body. CONCLUSION The results of the two studies helped to create an easy-to-apply scale that healthcare professionals can use to assess pain in patients with Rett's syndrome. This scale used PAINAD as its basic structure, with some changes in the items related to the behavior of patients with RTT.
Collapse
Affiliation(s)
- Rosa Angela Fabio
- Department of Economy, University of Messina, via Dei Verdi, 75, 98123 Messina, Italy
| | - Liliana Chiarini
- Department of Economy, University of Messina, via Dei Verdi, 75, 98123 Messina, Italy
- CARI, (Airett Center Innovation and Research), Vicolo Volto S. Luca, 16, 37100 Verona, Italy
| | - Virginia Canegallo
- Vita-Salute San Raffaele University, Via Olgettina, 58, 20132 Milano, MI Italy
| |
Collapse
|
21
|
Ta D, Downs J, Baynam G, Wilson A, Richmond P, Schmidt A, Decker A, Leonard H. Development of an International Database for a Rare Genetic Disorder: The MECP2 Duplication Database (MDBase). CHILDREN 2022; 9:children9081111. [PMID: 35892614 PMCID: PMC9332564 DOI: 10.3390/children9081111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 07/20/2022] [Accepted: 07/22/2022] [Indexed: 11/16/2022]
Abstract
The natural history of MECP2 duplication syndrome (MDS), a rare X-linked neurodevelopmental disorder with an estimated birth prevalence of 1/150,000 live births, is poorly understood due to a lack of clinical data collected for research. Such information is critical to the understanding of disease progression, therapeutic endpoints and outcome measures for clinical trials, as well as the development of therapies and orphan products. This clinical information can be systematically collected from caregivers through data collation efforts—yet, no such database has existed for MDS before now. Here, in this methodological study, we document the development, launch and management of the international MECP2 Duplication Database (MDBase). The MDBase consists of an extensive family questionnaire that collects information on general medical history, system-specific health problems, medication and hospitalisation records, developmental milestones and function, and quality of life (for individuals with MDS, and their caregivers). Launched in 2020, in its first two years of operation the MDBase has collected clinical data from 154 individuals from 26 countries—the largest sample size to date. The success of this methodology for the establishment and operation of the MDBase may provide insight and aid in the development of databases for other rare neurodevelopmental disorders.
Collapse
Affiliation(s)
- Daniel Ta
- Telethon Kids Institute, University of Western Australia, Perth, WA 6009, Australia; (D.T.); (J.D.); (G.B.); (A.W.); (P.R.)
| | - Jenny Downs
- Telethon Kids Institute, University of Western Australia, Perth, WA 6009, Australia; (D.T.); (J.D.); (G.B.); (A.W.); (P.R.)
- Curtin School of Allied Health, Curtin University, Bentley, WA 6102, Australia
| | - Gareth Baynam
- Telethon Kids Institute, University of Western Australia, Perth, WA 6009, Australia; (D.T.); (J.D.); (G.B.); (A.W.); (P.R.)
- Rare Care Centre, Perth Children’s Hospital, Nedlands, WA 6009, Australia
- Western Australian Register of Developmental Anomalies, King Edward Memorial Hospital, Subiaco, WA 6904, Australia
| | - Andrew Wilson
- Telethon Kids Institute, University of Western Australia, Perth, WA 6009, Australia; (D.T.); (J.D.); (G.B.); (A.W.); (P.R.)
- Curtin School of Allied Health, Curtin University, Bentley, WA 6102, Australia
- North Entrance, Perth Children’s Hospital, 15 Hospital Ave, Nedlands, WA 6009, Australia
- Discipline of Paediatrics, School of Medicine, University of Western Australia, Perth, WA 6009, Australia
| | - Peter Richmond
- Telethon Kids Institute, University of Western Australia, Perth, WA 6009, Australia; (D.T.); (J.D.); (G.B.); (A.W.); (P.R.)
- North Entrance, Perth Children’s Hospital, 15 Hospital Ave, Nedlands, WA 6009, Australia
- Discipline of Paediatrics, School of Medicine, University of Western Australia, Perth, WA 6009, Australia
| | - Aron Schmidt
- MECP2 Duplication Foundation, Tuscon, AZ 85724, USA; (A.S.); (A.D.)
| | - Amelia Decker
- MECP2 Duplication Foundation, Tuscon, AZ 85724, USA; (A.S.); (A.D.)
- Department of Paediatrics, University of Arizona College of Medicine, Tuscon, AZ 85724, USA
| | - Helen Leonard
- Telethon Kids Institute, University of Western Australia, Perth, WA 6009, Australia; (D.T.); (J.D.); (G.B.); (A.W.); (P.R.)
- Correspondence:
| |
Collapse
|
22
|
Collins BE, Neul JL. Rett Syndrome and MECP2 Duplication Syndrome: Disorders of MeCP2 Dosage. Neuropsychiatr Dis Treat 2022; 18:2813-2835. [PMID: 36471747 PMCID: PMC9719276 DOI: 10.2147/ndt.s371483] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Accepted: 11/14/2022] [Indexed: 11/30/2022] Open
Abstract
Rett syndrome (RTT) is a neurodevelopmental disorder caused predominantly by loss-of-function mutations in the gene Methyl-CpG-binding protein 2 (MECP2), which encodes the MeCP2 protein. RTT is a MECP2-related disorder, along with MECP2 duplication syndrome (MDS), caused by gain-of-function duplications of MECP2. Nearly two decades of research have advanced our knowledge of MeCP2 function in health and disease. The following review will discuss MeCP2 protein function and its dysregulation in the MECP2-related disorders RTT and MDS. This will include a discussion of the genetic underpinnings of these disorders, specifically how sporadic X-chromosome mutations arise and manifest in specific populations. We will then review current diagnostic guidelines and clinical manifestations of RTT and MDS. Next, we will delve into MeCP2 biology, describing the dual landscapes of methylated DNA and its reader MeCP2 across the neuronal genome as well as the function of MeCP2 as a transcriptional modulator. Following this, we will outline common MECP2 mutations and genotype-phenotype correlations in both diseases, with particular focus on mutations associated with relatively mild disease in RTT. We will also summarize decades of disease modeling and resulting molecular, synaptic, and behavioral phenotypes associated with RTT and MDS. Finally, we list several therapeutics in the development pipeline for RTT and MDS and available evidence of their safety and efficacy.
Collapse
Affiliation(s)
- Bridget E Collins
- Medical Scientist Training Program, Vanderbilt University, Nashville, TN, USA
| | - Jeffrey L Neul
- Vanderbilt Kennedy Center, Departments of Pediatrics, Pharmacology, and Special Education, Vanderbilt University Medical Center and Vanderbilt University, Nashville, TN, USA
| |
Collapse
|