1
|
Smith M, Dodis GE, Vanderplow AM, Gonzalez S, Rhee Y, Scrogin K, Gogliotti RG. Potentiation of the M 1 muscarinic acetylcholine receptor normalizes neuronal activation patterns and improves apnea severity in Mecp2 +/- mice. Neurobiol Dis 2025; 208:106859. [PMID: 40021095 DOI: 10.1016/j.nbd.2025.106859] [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: 11/26/2024] [Revised: 02/07/2025] [Accepted: 02/24/2025] [Indexed: 03/03/2025] Open
Abstract
Rett syndrome (RTT) is a neurodevelopmental disorder that is caused by loss-of-function mutations in the methyl-CpG binding protein 2 (MeCP2) gene. RTT patients experience a myriad of debilitating symptoms, which include respiratory phenotypes that are often associated with lethality. Our previous work established that expression of the M1 muscarinic acetylcholine receptor (mAchR) is decreased in RTT autopsy samples, and that potentiation of the M1 receptor improves apneas in a mouse model of RTT; however, the population of neurons driving this rescue is unclear. Loss of Mecp2 correlates with excessive neuronal activity in cardiorespiratory nuclei. Since M1 is found on cholinergic interneurons, we hypothesized that M1-potentiating compounds decrease apnea frequency by tempering brainstem hyperactivity. To test this, Mecp2+/- and Mecp2+/+ mice were screened for apneas before and after administration of the M1 positive allosteric modulator (PAM) VU0453595 (VU595). Brains from the same mice were then imaged for c-Fos, ChAT, and Syto16 using whole-brain light-sheet microscopy to establish genotype and drug-dependent activation patterns that could be correlated with VU595's efficacy on apneas. The vehicle-treated Mecp2+/- brain exhibited broad hyperactivity when coupled with the phenotypic prescreen, which was significantly decreased by administration of VU595, particularly in regions known to modulate the activity of respiratory nuclei (i.e. hippocampus and striatum). Further, the extent of apnea rescue in each mouse showed a significant positive correlation with c-Fos expression in non-cholinergic neurons in the striatum, thalamus, dentate gyrus, and within the cholinergic neurons of the brainstem. These results indicate that Mecp2+/- mice are prone to hyperactivity in brain regions that regulate respiration, which can be normalized through M1 potentiation.
Collapse
Affiliation(s)
- Mackenzie Smith
- Department of Molecular Pharmacology and Neuroscience, Loyola University Chicago, 2160 S 1st Avenue, Maywood, IL 60153, USA; Edward Hines Jr. VA Hospital, 5000 5th Ave, Hines, IL 60141, USA
| | - Grace E Dodis
- Department of Molecular Pharmacology and Neuroscience, Loyola University Chicago, 2160 S 1st Avenue, Maywood, IL 60153, USA; Edward Hines Jr. VA Hospital, 5000 5th Ave, Hines, IL 60141, USA
| | - Amanda M Vanderplow
- Department of Molecular Pharmacology and Neuroscience, Loyola University Chicago, 2160 S 1st Avenue, Maywood, IL 60153, USA; Edward Hines Jr. VA Hospital, 5000 5th Ave, Hines, IL 60141, USA
| | - Sonia Gonzalez
- Department of Molecular Pharmacology and Neuroscience, Loyola University Chicago, 2160 S 1st Avenue, Maywood, IL 60153, USA; Edward Hines Jr. VA Hospital, 5000 5th Ave, Hines, IL 60141, USA
| | - Yewon Rhee
- Department of Molecular Pharmacology and Neuroscience, Loyola University Chicago, 2160 S 1st Avenue, Maywood, IL 60153, USA; Edward Hines Jr. VA Hospital, 5000 5th Ave, Hines, IL 60141, USA
| | - Karie Scrogin
- Department of Molecular Pharmacology and Neuroscience, Loyola University Chicago, 2160 S 1st Avenue, Maywood, IL 60153, USA; Edward Hines Jr. VA Hospital, 5000 5th Ave, Hines, IL 60141, USA
| | - Rocco G Gogliotti
- Department of Molecular Pharmacology and Neuroscience, Loyola University Chicago, 2160 S 1st Avenue, Maywood, IL 60153, USA; Edward Hines Jr. VA Hospital, 5000 5th Ave, Hines, IL 60141, USA.
| |
Collapse
|
2
|
Lai YYL, Zafar S, Leonard HM, Walsh LJ, Downs JA. Access to Oral Healthcare in Individuals With Rett Syndrome: A Qualitative Study of Parent Perspectives. JOURNAL OF INTELLECTUAL DISABILITY RESEARCH : JIDR 2025; 69:403-416. [PMID: 40033823 DOI: 10.1111/jir.13222] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Revised: 02/02/2025] [Accepted: 02/04/2025] [Indexed: 03/05/2025]
Abstract
BACKGROUND Intellectual and developmental disabilities (IDD) are varied in their nature and presentation. Barriers to oral healthcare are reported in studies of general populations with IDD but these may not reflect the barriers experienced by individuals with rare disorders such as Rett syndrome (RTT). There are also few peer-reviewed studies in the Australian context exploring barriers to dental care access for patients living with a disability. This qualitative study explored caregivers' perceptions and experiences regarding oral health and access to dental care for those with RTT in Australia. METHODS Parents of 31 individuals with a confirmed MECP2 mutation were sampled purposively from the Australian Rett Syndrome Database. Interview questions were based on earlier studies used in other disability populations and queried identification and management of dental pain and influence of other comorbidities in their child's oral care. Interviews were audio-recorded, transcribed and analysed using NVivo (Version 12 Plus). Directed content analysis was used to code data to a framework constructed from a literature review of factors affecting access to professional oral healthcare systems and factors affecting access to optimal at-home oral care in disability. RESULTS The most frequently cited barriers to professional dental care were dentist-related, while caregiver related financial barriers were cited by a minority of families. Dentist-related financial barriers were not present in these data. Most factors affecting access to optimal at-home oral care coded to the existing framework, with further enablers identified under training for the caregiver or parent. CONCLUSIONS The findings of this study provide a point of reference to understand factors affecting provision of at-home dental care and professional services to enable optimal oral health in RTT. Future research could explore the provision of targeted oral health information on RTT to carers and clinicians.
Collapse
Affiliation(s)
- Yvonne Yee Lok Lai
- UQ Oral Health Centre, University of Queensland School of Dentistry, Brisbane, Queensland, Australia
- Child Disability, The Kids Research Institute Australia, Perth, Western Australia, Australia
- Paediatric Dental Department, Women's and Children's Hospital, Adelaide, South Australia, Australia
| | - Sobia Zafar
- UQ Oral Health Centre, University of Queensland School of Dentistry, Brisbane, Queensland, Australia
| | - Helen Margaret Leonard
- Child Disability, The Kids Research Institute Australia, Perth, Western Australia, Australia
| | - Laurence James Walsh
- UQ Oral Health Centre, University of Queensland School of Dentistry, Brisbane, Queensland, Australia
| | - Jenny Anne Downs
- Child Disability, The Kids Research Institute Australia, Perth, Western Australia, Australia
- Curtin School of Allied Health, Curtin University, Perth, Western Australia, Australia
| |
Collapse
|
3
|
Wang M, Liu K, Guo D, Lv Y, Wang X. Arbovirus Infections and Epigenetic Mechanisms; a Potential Therapeutic Target. Rev Med Virol 2025; 35:e70033. [PMID: 40155348 DOI: 10.1002/rmv.70033] [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: 02/07/2025] [Revised: 03/06/2025] [Accepted: 03/11/2025] [Indexed: 04/01/2025]
Abstract
Arboviruses are a group of arthropod-borne viral pathogens that pose a significant threat to the public health system. The clinical manifestations associated with these viruses range from self-limiting infections to life-threatening disorders. As a group of systemic viral infections, arboviruses can affect various parts of human organ systems, such as the nervous system. In the nervous system, epigenetic mechanisms are involved in various mechanisms including adult neurogenesis, neuronal-glial differentiation, the regulation of neural behaviour and neural plasticity, as well as other brain functions such as memory, and cognition. Hence, epigenetic deregulation is a key factor in the aetiology of different neurological disorders that highlights the importance of studying the underlying mechanisms and risk factors to introduce effective therapeutic approaches. There is mounting evidence that arboviruses that affect the nervous system take advantage of various mechanisms to modulate epigenetic processes to regulate their life cycles. This phenomenon may affect the nervous system leading to neurotropic arboviral infection-associated neurological disorders. Hence, it is important to understand reciprocal interplays between neurotropic arboviral pathogens and epigenetic processes to better control these disorders. The present review provides an overview of different interactions of arboviruses with epigenetic mechanisms during neurotropic arboviral infections. It uniquely focuses on the interplay between epigenetic modifications and arboviral neurotropism, shedding light on potential therapeutic strategies that have not been comprehensively addressed before. Targeting virus-induced epigenetic alterations, such as miRNA regulation, could lead to novel antiviral therapies aimed at mitigating neuroinflammation and disease severity.
Collapse
Affiliation(s)
- Manhong Wang
- University Hospital, Jilin Normal University, Siping, China
| | - Kexin Liu
- Department of Pathology, Siping City Centeral People's Hospital, Siping, China
| | - Dan Guo
- University Hospital, Jilin Normal University, Siping, China
| | - Youjia Lv
- Department of Hepatology, Siping City Infectious Disease Hospital, Siping, China
| | - Xin Wang
- Student Affairs Office, Jilin Normal University, Siping, China
| |
Collapse
|
4
|
Oshizuki S, Masaki S, Tanaka S, Kataoka N. SRSF9-Mediated Exon Recognition Promotes Exon 2 Inclusion in Mecp2 Pre-mRNA Alternative Splicing. Int J Mol Sci 2025; 26:3319. [PMID: 40244165 PMCID: PMC11989674 DOI: 10.3390/ijms26073319] [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: 03/13/2025] [Revised: 03/31/2025] [Accepted: 04/01/2025] [Indexed: 04/18/2025] Open
Abstract
Alternative splicing is one of the processes that contributes to producing a vast protein diversity from the limited number of protein-coding genes in higher eukaryotes. The Methyl CpG Binding Protein 2 (Mecp2) gene, whose mutations cause Rett syndrome, generates two protein isoforms, MeCP2E1 and MeCP2E2, by alternative splicing. These isoforms likely possess non-redundant functions. However, the molecular mechanism for Mecp2 pre-mRNA alternative splicing remains to be understood. Here, we analyzed the alternative splicing mechanism of MeCP2 pre-mRNA and found that exon 2 is efficiently recognized through adjacent strong splice sites. In addition, exonic splicing enhancer (ESE) in exon 2 plays an important role in exon 2 inclusion, which is highly likely to be mediated by SRSF9.
Collapse
Affiliation(s)
| | | | | | - Naoyuki Kataoka
- Laboratory of Cellular Biochemistry, Department of Animal Resource Sciences, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo 113-8657, Japan
| |
Collapse
|
5
|
Ross PD, Gadalla KKE, Thomson SR, Selfridge J, Bahey NG, Benito J, Burstein SR, McMinn R, Bolon B, Hector RD, Cobb SR. Self-regulating gene therapy ameliorates phenotypes and overcomes gene dosage sensitivity in a mouse model of Rett syndrome. Sci Transl Med 2025; 17:eadq3614. [PMID: 40173263 DOI: 10.1126/scitranslmed.adq3614] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Revised: 09/27/2024] [Accepted: 03/12/2025] [Indexed: 04/04/2025]
Abstract
Conventional methods of gene transfer lead to inconsistent transgene expression within cells. This variability can be problematic, particularly in conditions like Rett syndrome (RTT), a neurological disorder caused by mutations in the MECP2 (methyl-CpG binding protein 2) gene, because overexpression of MECP2 can also cause adverse effects. To address these challenges, we devised a gene regulation system called Expression Attenuation via Construct Tuning (EXACT), which uses a self-contained, microRNA-based feed-forward loop that not only ensures more consistent transgene expression but also protects against excessive expression. Through cell-based screening assays, we demonstrated the ability of the EXACT circuit to modulate the expression of full-length human MeCP2. Compared with a conventional construct, an EXACT-MECP2 construct exhibited a narrower range of cellular protein abundance. Furthermore, the degree of regulation by the EXACT circuit increased with higher transgene doses in vitro and in wild-type mice and mice modeling RTT. On the basis of cellular and in vivo testing, we identified an optimal configuration for the adeno-associated virus serotype 9 (AAV9) construct for self-regulated MECP2 gene therapy, designated NGN-401. Delivery of NGN-401 to neonatal male Mecp2-/y hemizygous mice via intracerebroventricular injection resulted in prolonged survival and amelioration of RTT-like phenotypes compared with vehicle-treated animals. NGN-401 was also well tolerated by female Mecp2+/- mice and healthy juvenile nonhuman primates, in contrast with a conventional construct, which caused toxicity. The results from these studies underpin a first-in-human pediatric trial of NGN-401 in RTT (ClinicalTrials.gov, NCT05898620).
Collapse
Affiliation(s)
- Paul D Ross
- Simons Initiative for the Developing Brain, Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh EH8 9XD, UK
| | - Kamal K E Gadalla
- Simons Initiative for the Developing Brain, Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh EH8 9XD, UK
| | - Sophie R Thomson
- Simons Initiative for the Developing Brain, Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh EH8 9XD, UK
| | - Jim Selfridge
- Simons Initiative for the Developing Brain, Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh EH8 9XD, UK
| | - Noha G Bahey
- Simons Initiative for the Developing Brain, Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh EH8 9XD, UK
| | | | | | | | | | - Ralph D Hector
- Simons Initiative for the Developing Brain, Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh EH8 9XD, UK
| | - Stuart R Cobb
- Simons Initiative for the Developing Brain, Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh EH8 9XD, UK
- Neurogene Inc., New York, NY 10011, USA
| |
Collapse
|
6
|
Ette EI, Fadiran EO, Missling C, Hammond E. The new big is small: Leveraging knowledge from small trials for rare disease drug development: Blarcamesine for Rett syndrome. Br J Clin Pharmacol 2025; 91:1049-1063. [PMID: 37429704 PMCID: PMC11992655 DOI: 10.1111/bcp.15843] [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: 01/09/2023] [Revised: 05/10/2023] [Accepted: 06/01/2023] [Indexed: 07/12/2023] Open
Abstract
Big data in drug development may not satisfactorily address the demands of precision medicine in a rare disease population, making the use of smaller clinical trials necessary. Consequently, the use of innovative design and analysis of these clinical trials using model-informed approaches have become indispensable. This requires informative exposure-outcome analysis, together with formal statistical analysis, which should include the strength of evidence for a study outcome. We demonstrate how knowledge can be gained, with supporting strength of evidence, from a small (data) clinical trial with a low dose of blarcamesine in the treatment of Rett syndrome. Based on a small data paradigm, pharmacometrics item response theory modelling and Bayes factor analysis were used to demonstrate the efficacy of blarcamesine in Rett syndrome.
Collapse
|
7
|
Narita H, Natsume J, Suzuki T, Shiohama T, Kawaguchi M, Okazaki M, Hashizume A, Naganawa S, Ito Y, Yamamoto H, Nakata T, Kidokoro H, Takahashi Y, Takahashi S, Tsujimura K. Diffuse but Non-homogeneous Brain Atrophy: Identification of Specific Brain Regions and Their Correlation with Clinical Severity in Rett Syndrome. Brain Dev 2025; 47:104348. [PMID: 40147315 DOI: 10.1016/j.braindev.2025.104348] [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: 11/02/2024] [Revised: 02/27/2025] [Accepted: 03/09/2025] [Indexed: 03/29/2025]
Abstract
BACKGROUND Rett syndrome is a genetic neurodevelopmental disorder that predominantly affects girls. While microcephaly is a common feature, there is limited information on the detailed structural changes in the brain. This study aimed to identify regional brain volume abnormalities and explore the correlation between brain volume and clinical characteristics. METHODS We compared the regional brain volumes of 20 female children with Rett syndrome to those of 25 healthy female children. Additionally, we assessed the correlation between regional brain volume, Clinical Severity Scores, and epilepsy status. RESULTS Significantly smaller volumes were observed in all brain regions, including the cerebral cortex, cerebral white matter, subcortical gray matter, cerebellum, and brainstem. Within the cortical regions, volume reduction was prominent in the left precentral, right lateral occipital, left precuneus, left inferior parietal, and right medial orbitofrontal cortices. After correcting for intracranial volumes, volume reduction was more prominent in the cerebral cortices than in the cerebral white matter. Small volumes were consistently observed, regardless of age. Negative correlations were observed between the volumes of multiple regions and the Clinical Severity Scores. There were no correlations among regional brain volume, seizure control, or duration of epilepsy. CONCLUSION The mechanism underlying the cortical-dominant volume reduction remains unclear; however, it may be caused by altered synapse development associated with methyl-CpG-binding protein 2 gene abnormalities. Characteristic impairments in visual recognition and deterioration of motor function in Rett syndrome may be associated with significant volume reduction in specific cortical regions, such as the lateral occipital cortex, precuneus, and precentral gyrus.
Collapse
Affiliation(s)
- Hajime Narita
- Department of Pediatrics, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Jun Natsume
- Department of Pediatrics, Nagoya University Graduate School of Medicine, Nagoya, Japan; Department of Developmental Disability Medicine, Nagoya University Graduate School of Medicine, Nagoya, Japan.
| | - Takeshi Suzuki
- Department of Pediatrics, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Tadashi Shiohama
- Department of Pediatrics, Chiba University Graduate School of Medicine, Chiba, Japan
| | - Masahiro Kawaguchi
- Department of Pediatrics, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Masaki Okazaki
- Department of Clinical Research Education, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Atsushi Hashizume
- Department of Clinical Research Education, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Shinji Naganawa
- Department of Radiology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yuji Ito
- Department of Pediatrics, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Hiroyuki Yamamoto
- Department of Pediatrics, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Tomohiko Nakata
- Department of Pediatrics, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Hiroyuki Kidokoro
- Department of Pediatrics, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yoshiyuki Takahashi
- Department of Pediatrics, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Satoru Takahashi
- Department of Pediatrics, Asahikawa Medical University, Asahikawa, Japan
| | - Keita Tsujimura
- Group of Brain Function and Development, Nagoya University Neuroscience Institute of the Graduate School of Science, Nagoya, Japan; Research Unit for Developmental Disorders, Institute for Advanced Research, Nagoya University, Nagoya, Japan
| |
Collapse
|
8
|
Thom RP, Warren TL, Khan S, Muhle RA, Wang PP, Brennand K, Zürcher NR, Veenstra-VanderWeele J, Hoffman EJ. A Blueprint for Translational Precision Medicine in Autism Spectrum Disorder and Related Neurogenetic Syndromes. J Child Adolesc Psychopharmacol 2025. [PMID: 40138183 DOI: 10.1089/cap.2025.0023] [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] [Indexed: 03/29/2025]
Abstract
Objectives: Despite growing knowledge of the underlying neurobiology of autism spectrum disorder (ASD) and related neurogenetic syndromes, treatment discovery has remained elusive. In this review, we provide a blueprint for translational precision medicine in ASD and related neurogenetic syndromes. Methods: The discovery of trofinetide for Rett syndrome (RTT) is described, and the role of nonmammalian, mammalian, and stem cell model systems in the identification of molecular targets and drug screening is discussed. We then provide a framework for translating preclinical findings to human clinical trials, including the role of biomarkers in selecting molecular targets and evaluating target engagement, and discuss how to leverage these findings for future ASD drug development. Results: Multiple preclinical model systems for ASD have been developed, each with tradeoffs with regard to suitability for high-throughput small molecule screening, conservation across species, and behavioral face validity. Future clinical trials should incorporate biomarkers and intermediate phenotypes to demonstrate target engagement. Factors that contributed to the approval of trofinetide for RTT included replicated findings in mouse models, a well-studied natural history of the syndrome, development of RTT-specific outcome measures, and strong engagement of the RTT family community. Conclusions: The translation of our growing understanding of the neurobiology of ASD to human drug discovery will require a precision medicine approach, including the use of multiple model systems for molecular target selection, evaluation of target engagement, and clinical trial design strategies that address heterogeneity, power, and the placebo response.
Collapse
Affiliation(s)
- Robyn P Thom
- Massachusetts General Hospital Lurie Center for Autism, Harvard Medical School, Lexington, Massachusetts, USA
| | | | - Suha Khan
- Child Study Center, Yale School of Medicine, New Haven, Connecticut, USA
| | - Rebecca A Muhle
- Columbia University and New York State Psychiatric Institute, New York, New York, USA
| | - Paul P Wang
- Yale School of Medicine, New Haven, Connecticut, USA
- Simons Foundation Autism Research Initiative, New Haven, Connecticut, USA
| | | | - Nicole R Zürcher
- Massachusetts General Hospital Lurie Center for Autism, Harvard Medical School, Lexington, Massachusetts, USA
- Athinoula A. Martinos Center for Biomedical Imaging, Boston, Massachusetts, USA
| | | | - Ellen J Hoffman
- Child Study Center, Yale School of Medicine, New Haven, Connecticut, USA
- Department of Neuroscience, Yale School of Medicine, New Haven, Connecticut, USA
| |
Collapse
|
9
|
Monteiro-Fernandes D, Charles I, Guerreiro S, Cunha-Garcia D, Pereira-Sousa J, Oliveira S, Teixeira-Castro A, Varney MA, Kleven MS, Newman-Tancredi A, P Sheikh Abdala A, Duarte-Silva S, Maciel P. Rescue of respiratory and cognitive impairments in Rett Syndrome mice using NLX-101, a selective 5-HT 1A receptor biased agonist. Biomed Pharmacother 2025; 186:117989. [PMID: 40121895 DOI: 10.1016/j.biopha.2025.117989] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Revised: 03/12/2025] [Accepted: 03/13/2025] [Indexed: 03/25/2025] Open
Abstract
Rett Syndrome (RTT) is a neurodevelopmental disorder caused by mutations in the X-linked gene encoding the methyl-CpG-binding protein 2 (MECP2). Impaired function of this transcriptional regulator leads to profound neurological defects, among which respiratory distress, motor function and cognitive disorders are prominent. Despite great advances in understanding RTT neurobiology, therapies that can meaningfully improve patients' symptoms are still needed. Here, we focused on 5-HT1A receptor-mediated serotonergic signaling as a potential therapeutical route for RTT. We report the effects of a drug candidate, NLX-101, a highly selective, biased agonist of 5-HT1A post-synaptic receptors at brainstem and cortical regions, on key phenotypes of RTT. Unrestrained whole-body plethysmography studies confirmed and extended the previous observation that single i.p. administration of NLX-101 dose-dependently reduced the occurrence and length of apneic events in Mecp2tm1.1Bird heterozygous female mice and largely corrected respiratory irregularity. Although no preservation of motor function was observed, early onset chronic administration of NLX-101 entirely prevented the cognitive deficits of the Mecp2tm1.1Bird mice both in the short and the long-term memory paradigms of the Novel Object Recognition upon 10 weeks of treatment, an effect that was maintained throughout animals' age. Similar effects were observed in the Fear Conditioning paradigm, with treated Rett mice performing as well as wild-type controls, highlighting the procognitive properties of NLX-101. This work provides compelling evidence of the therapeutic potential of targeting post-synaptic 5-HT1A receptors to improve cognitive function in patients with RTT while supporting its respiratory-rescue properties.
Collapse
Affiliation(s)
- Daniela Monteiro-Fernandes
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal; ICVS/3B's - PT Government Associate Laboratory, Guimarães, Braga, Portugal
| | - Ian Charles
- School of Physiology, Pharmacology & Neuroscience, Faculty of Life Sciences, University of Bristol, Bristol, United Kingdom
| | - Sara Guerreiro
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal; ICVS/3B's - PT Government Associate Laboratory, Guimarães, Braga, Portugal
| | - Daniela Cunha-Garcia
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal; ICVS/3B's - PT Government Associate Laboratory, Guimarães, Braga, Portugal
| | - Joana Pereira-Sousa
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal; ICVS/3B's - PT Government Associate Laboratory, Guimarães, Braga, Portugal
| | - Stéphanie Oliveira
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal; ICVS/3B's - PT Government Associate Laboratory, Guimarães, Braga, Portugal
| | - Andreia Teixeira-Castro
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal; ICVS/3B's - PT Government Associate Laboratory, Guimarães, Braga, Portugal
| | | | | | | | - Ana P Sheikh Abdala
- School of Physiology, Pharmacology & Neuroscience, Faculty of Life Sciences, University of Bristol, Bristol, United Kingdom
| | - Sara Duarte-Silva
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal; ICVS/3B's - PT Government Associate Laboratory, Guimarães, Braga, Portugal
| | - Patrícia Maciel
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal; ICVS/3B's - PT Government Associate Laboratory, Guimarães, Braga, Portugal.
| |
Collapse
|
10
|
Levy-Shraga Y, Goldmann S, Gruber N, Tripto-Shkolnik L, Modan-Moses D, Givon U, Ben-Zeev B. Bone health and bisphosphonate treatment in females with Rett syndrome in a national center. Pediatr Res 2025:10.1038/s41390-025-04001-4. [PMID: 40119038 DOI: 10.1038/s41390-025-04001-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Revised: 02/19/2025] [Accepted: 02/25/2025] [Indexed: 03/24/2025]
Abstract
BACKGROUND Impaired bone health is a common morbidity in Rett syndrome (RTT). We aimed to assess lumbar bone mineral density (BMD) and trabecular bone score (TBS) in females with RTT, and to evaluate the effectiveness of bisphosphonate treatment. METHODS This retrospective study included 40 females with RTT, aged 5-22 years, who underwent dual-energy X-ray absorptiometry (DXA) scans during 2019-2024 at a national center for RTT. Data collected included medical treatment, anthropometric measurements, and functional scores. RESULTS The median age at the first DXA scan was 10.8 years. The mean L1-4 BMD Z-score was -2.1 ± 1.4, and the mean TBS Z-score was -0.4 ± 1.3. The L1-4 BMD Z-score correlated with height (r = 0.407, p = 0.009), weight (r = 0.551, p < 0.001), BMI (r = 0.644, p < 0.001), and TBS Z-scores (r = 0.594, p = 0.009). Poor L1-4 BMD Z-scores were associated with poor mobility scores (p = 0.05) and valproate treatment (p = 0.016). Nine patients (23%) received zoledronate, for a mean 2 years. The mean age at zoledronate initiation was 9.7 ± 2.3 years. Four completed two DXA scans (pre- and post-treatment); the mean BMD Z-score improved from -2.2 ± 0.9 to -1.4 ± 0.9 after treatment. CONCLUSIONS Females with RTT have reduced lumbar BMD, which was associated with anthropometric factors, TBS, mobility, and valproate use. Zoledronate may be effective for some patients. IMPACT In a retrospective study of 40 females with Rett syndrome (RTT), low bone mineral density (BMD) correlated with lower anthropometric measurements, impaired mobility, and valproic acid use. The association between BMD and trabecular bone score (TBS) in the context of RTT is a novel finding. Our preliminary data support the effectiveness and safety of zoledronate for treating osteoporosis in patients with RTT. Our findings are important in light of the increasing life expectancy of individuals with RTT, and the consequent need to prioritize bone health in this population.
Collapse
Affiliation(s)
- Yael Levy-Shraga
- Faculty of Medical and Health Sciences, Tel Aviv University, Tel Aviv, Israel.
- Pediatric Endocrinology and Diabetes Unit, The Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Tel-Hashomer, Ramat Gan, Israel.
| | - Simon Goldmann
- Faculty of Medical and Health Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Noah Gruber
- Faculty of Medical and Health Sciences, Tel Aviv University, Tel Aviv, Israel
- Pediatric Endocrinology and Diabetes Unit, The Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Tel-Hashomer, Ramat Gan, Israel
| | - Liana Tripto-Shkolnik
- Faculty of Medical and Health Sciences, Tel Aviv University, Tel Aviv, Israel
- Division of Endocrinology, Diabetes and Metabolism, Sheba Medical Center, Tel Hashomer, Ramat Gan, Israel
| | - Dalit Modan-Moses
- Faculty of Medical and Health Sciences, Tel Aviv University, Tel Aviv, Israel
- Pediatric Endocrinology and Diabetes Unit, The Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Tel-Hashomer, Ramat Gan, Israel
| | - Uri Givon
- Faculty of Medical and Health Sciences, Tel Aviv University, Tel Aviv, Israel
- Pediatric Orthopedics Unit, The Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Tel-Hashomer, Ramat Gan, Israel
| | - Bruria Ben-Zeev
- Faculty of Medical and Health Sciences, Tel Aviv University, Tel Aviv, Israel
- Division of Pediatric Neurology, The Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Tel-Hashomer, Ramat Gan, Israel
| |
Collapse
|
11
|
Weekley BH, Ahmed NI, Maze I. Elucidating neuroepigenetic mechanisms to inform targeted therapeutics for brain disorders. iScience 2025; 28:112092. [PMID: 40160416 PMCID: PMC11951040 DOI: 10.1016/j.isci.2025.112092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/02/2025] Open
Abstract
The evolving field of neuroepigenetics provides important insights into the molecular foundations of brain function. Novel sequencing technologies have identified patient-specific mutations and gene expression profiles involved in shaping the epigenetic landscape during neurodevelopment and in disease. Traditional methods to investigate the consequences of chromatin-related mutations provide valuable phenotypic insights but often lack information on the biochemical mechanisms underlying these processes. Recent studies, however, are beginning to elucidate how structural and/or functional aspects of histone, DNA, and RNA post-translational modifications affect transcriptional landscapes and neurological phenotypes. Here, we review the identification of epigenetic regulators from genomic studies of brain disease, as well as mechanistic findings that reveal the intricacies of neuronal chromatin regulation. We then discuss how these mechanistic studies serve as a guideline for future neuroepigenetics investigations. We end by proposing a roadmap to future therapies that exploit these findings by coupling them to recent advances in targeted therapeutics.
Collapse
Affiliation(s)
- Benjamin H. Weekley
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Newaz I. Ahmed
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Ian Maze
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Howard Hughes Medical Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| |
Collapse
|
12
|
Narusawa H, Ogawa T, Yagasaki H, Nagasaki K, Urakawa T, Saito T, Soneda S, Kinjo S, Sano S, Mamada M, Terashita S, Dateki S, Narumi S, Naiki Y, Horikawa R, Ogata T, Fukami M, Kagami M. Comprehensive Study on Central Precocious Puberty: Molecular and Clinical Analyses in 90 Patients. J Clin Endocrinol Metab 2025; 110:1023-1036. [PMID: 39324648 DOI: 10.1210/clinem/dgae666] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Revised: 08/25/2024] [Accepted: 09/24/2024] [Indexed: 09/27/2024]
Abstract
CONTEXT Defects in MKRN3, DLK1, KISS1, and KISS1R and some disorders, such as Temple syndrome (TS14), cause central precocious puberty (CPP). Recently, pathogenic variants (PVs) in MECP2 have been reported to be associated with CPP. OBJECTIVE We aimed to clarify the contribution of (epi)genetic abnormalities to CPP and clinical and hormonal features in each etiology. METHODS We conducted targeted sequencing for MKRN3, DLK1, MECP2, KISS1, and KISS1R and methylation analysis for screening of imprinting disorders such as TS14 associated with CPP in 90 patients with CPP (no history of brain injuries and negative brain magnetic resonance imaging) and collected their clinical and laboratory data. We measured serum DLK1 levels in 3 patients with TS14 and serum MKRN3 levels in 2 patients with MKRN3 genetic defects, together with some etiology-unknown patients with CPP and controls. RESULTS We detected 8 patients with TS14 (6, epimutation; 1, mosaic maternal uniparental disomy chromosome 14; 1, microdeletion) and 3 patients with MKRN3 genetic defects (1, PV; 1, 13-bp deletion in the 5'-untranslated region [5'-UTR]; 1, microdeletion) with family histories of paternal early puberty. There were no patients with PVs identified in MECP2, KISS1, or KISS1R. We confirmed low serum MKRN3 level in the patient with a deletion in 5'-UTR. The median height at initial evaluation of TS14 patients was lower than that of all patients. Six patients with TS14 were born small for gestational age (SGA). CONCLUSION (Epi)genetic causes were identified in 12.2% of patients with CPP at our center. For patients with CPP born SGA or together with family histories of paternal early puberty, (epi)genetic testing for TS14 and MKRN3 genetic defects should be considered.
Collapse
Affiliation(s)
- Hiromune Narusawa
- Department of Molecular Endocrinology, National Research Institute for Child Health and Development, Tokyo 157-8535, Japan
- Department of Pediatrics, Faculty of Medicine, University of Yamanashi, Chuo 409-3898, Japan
| | - Tomoe Ogawa
- Department of Molecular Endocrinology, National Research Institute for Child Health and Development, Tokyo 157-8535, Japan
| | - Hideaki Yagasaki
- Department of Pediatrics, Faculty of Medicine, University of Yamanashi, Chuo 409-3898, Japan
| | - Keisuke Nagasaki
- Division of Pediatrics, Department of Homeostatic Regulation and Development, Niigata University Graduate School of Medical and Dental Science, Niigata 951-8510, Japan
| | - Tatsuki Urakawa
- Department of Molecular Endocrinology, National Research Institute for Child Health and Development, Tokyo 157-8535, Japan
| | - Tomohiro Saito
- Department of Pediatrics, Yamanashi Prefectural Central Hospital, Kofu 400-0027, Japan
| | - Shun Soneda
- Tanaka Growth Clinic, Tokyo 158-0097, Japan
- Department of Pediatrics, St. Marianna University School of Medicine, Kawasaki 216-0015, Japan
| | - Saori Kinjo
- Department of Pediatrics, Okinawa Chubu Hospital, Uruma 904-2293, Japan
| | - Shinichiro Sano
- Department of Pediatric Endocrinology and Metabolism, Shizuoka Children's Hospital, Shizuoka 420-0953, Japan
| | - Mitsukazu Mamada
- Department of Pediatrics, Japanese Red Cross Wakayama Medical Center, Wakayama 640-8558, Japan
| | - Shintaro Terashita
- Department of Pediatrics, Faculty of Medicine, University of Toyama, Toyama 930-0194, Japan
| | - Sumito Dateki
- Department of Pediatrics, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki 852-8501, Japan
| | - Satoshi Narumi
- Department of Molecular Endocrinology, National Research Institute for Child Health and Development, Tokyo 157-8535, Japan
| | - Yasuhiro Naiki
- Division of Endocrinology and Metabolism, National Center for Child Health and Development, Tokyo 157-8535, Japan
| | - Reiko Horikawa
- Division of Endocrinology and Metabolism, National Center for Child Health and Development, Tokyo 157-8535, Japan
| | - Tsutomu Ogata
- Department of Biochemistry, Hamamatsu University School of Medicine, Hamamatsu 431-3192, Japan
- Department of Pediatrics, Hamamatsu Medical Center, Hamamatsu 432-8580, Japan
| | - Maki Fukami
- Department of Molecular Endocrinology, National Research Institute for Child Health and Development, Tokyo 157-8535, Japan
| | - Masayo Kagami
- Department of Molecular Endocrinology, National Research Institute for Child Health and Development, Tokyo 157-8535, Japan
| |
Collapse
|
13
|
Romano A, Rodocanachi Roidi ML, Savini MN, Viganò I, Dziubak M, Pietrogrande L, Moran DS, Lotan M. Effects of a Supervised-As-Needed Home Exercise Program on Scoliosis and Motor Function in Rett Syndrome: A Multiple-Baseline Study. J Clin Med 2025; 14:1873. [PMID: 40142681 PMCID: PMC11943425 DOI: 10.3390/jcm14061873] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2025] [Revised: 03/01/2025] [Accepted: 03/05/2025] [Indexed: 03/28/2025] Open
Abstract
Background/Objectives: Scoliosis is a prevalent comorbidity in Rett syndrome (RTT), often necessitating surgical intervention. This study investigated the impact of a 10-month individualized home exercise program (HEP) on scoliosis progression and gross motor function in girls aged six to 16 years with RTT. Methods: A multiple-baseline single-case design (AABA) was employed with 20 participants. A remotely supervised HEP, based on established principles focused on posture and physical activity, was implemented daily for at least one hour. The primary outcome was the rate of scoliosis progression assessed through the Cobb angle change measured via spinal radiographs at baseline, pre-intervention, and post-intervention. The secondary outcome was the gross motor function. Results: The HEP did not significantly reduce the rate of scoliosis progression. However, individual responses varied, with three participants showing scoliosis reduction. Significant improvements were observed in gross motor function, particularly in standing, walking, and stair-climbing abilities. Conclusions: The HEP did not significantly impact overall scoliosis progression, but a significant improvement was found in gross motor function. Further research into larger sample sizes is needed to confirm the effectiveness of exercise interventions in people with RTT.
Collapse
Affiliation(s)
- Alberto Romano
- Department of Health System Management, Ariel University, Ariel 4070000, Israel
| | | | - Miriam Nella Savini
- Developmental Age Neurology, Epilepsy Center, San Paolo Hospital, 20142 Milan, Italy
| | - Ilaria Viganò
- Developmental Age Neurology, Epilepsy Center, San Paolo Hospital, 20142 Milan, Italy
| | - Michal Dziubak
- Orthopedic and Traumatology, San Paolo Hospital, 20142 Milan, Italy
| | - Luca Pietrogrande
- Department of Health Sciences, University of Milan, 20142 Milan, Italy
| | - Daniel Sender Moran
- Department of Health System Management, Ariel University, Ariel 4070000, Israel
| | - Meir Lotan
- Department of Physiotherapy, Ariel University, Ariel 4070000, Israel
- Israeli Rett Syndrome National Evaluation Team, Ramat Gan 5200100, Israel
| |
Collapse
|
14
|
Raspa M, Gwaltney A, Bann C, von Hehn J, Benke TA, Marsh ED, Peters SU, Ananth A, Percy AK, Neul JL. Psychometric Assessment of the Rett Syndrome Caregiver Assessment of Symptom Severity (RCASS). J Autism Dev Disord 2025; 55:997-1009. [PMID: 38438817 PMCID: PMC11374935 DOI: 10.1007/s10803-024-06238-0] [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] [Accepted: 01/03/2024] [Indexed: 03/06/2024]
Abstract
Rett syndrome is a severe neurodevelopmental disorder that affects about 1 in 10,000 females. Clinical trials of disease modifying therapies are on the rise, but there are few psychometrically sound caregiver-reported outcome measures available to assess treatment benefit. We report on a new caregiver-reported outcome measure, the Rett Caregiver Assessment of Symptom Severity (RCASS). Using data from the Rett Natural History Study (n = 649), we examined the factor structure, using both exploratory and confirmatory factor analysis, and the reliability and validity of the RCASS. The four-factor model had the best overall fit, which covered movement, communication, behavior, and Rett-specific symptoms. The RCASS had moderate internal consistency. Strong face validity was found with age and mutation type, and convergent validity was established with other similar measures, including the Revised Motor-Behavior Assessment Scale, Clinical Severity Scale, Clinical Global Impression Scale, and the Child Health Questionnaire. These data provide initial evidence that the RCASS is a viable caregiver-outcome measure for use in clinical trials in Rett syndrome. Future work to assess sensitivity to change and other measures of reliability, such as test-retest and inter-rater agreement, are needed.
Collapse
Affiliation(s)
- Melissa Raspa
- RTI International, 3040 East Cornwallis Road, Research Triangle Park, NC, 27708, USA.
| | - Angela Gwaltney
- RTI International, 3040 East Cornwallis Road, Research Triangle Park, NC, 27708, USA
| | - Carla Bann
- RTI International, 3040 East Cornwallis Road, Research Triangle Park, NC, 27708, USA
| | | | - Timothy A Benke
- Children's Hospital Colorado and University of Colorado School of Medicine, Aurora, USA
| | - Eric D Marsh
- Children's Hospital of Philadelphia, University of Pennsylvania Perelman School of Medicine, Philadelphia, USA
| | - Sarika U Peters
- Vanderbilt Kennedy Center, Vanderbilt University Medical Center, Nashville, USA
| | - Amitha Ananth
- University of Alabama at Birmingham, Birmingham, USA
| | - Alan K Percy
- University of Alabama at Birmingham, Birmingham, USA
| | - Jeffrey L Neul
- Vanderbilt Kennedy Center, Vanderbilt University Medical Center, Nashville, USA.
| |
Collapse
|
15
|
Wu X, Wu H, Zhong M, Chen Y, Su W, Li P. Epigenetic regulation by naringenin and naringin: A literature review focused on the mechanisms underlying its pharmacological effects. Fitoterapia 2025; 181:106353. [PMID: 39706348 DOI: 10.1016/j.fitote.2024.106353] [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: 08/30/2024] [Revised: 12/06/2024] [Accepted: 12/15/2024] [Indexed: 12/23/2024]
Abstract
Epigenetics refers to heritable changes in gene expression or phenotypic changes that occur without changing the gene sequence. The main methods of epigenetics include non-coding RNA, histone modification, and DNA modification, which play an essential role in gene expression regulation and even the occurrence of diverse diseases. Naringenin, the aglycone form of naringin, is a natural flavonoid compound mainly found in fruits or plant derivatives such as citrus, tomatoes, and cherries. Naringenin and naringin exhibit a broad spectrum of biological activities and pharmacological effects, including anti-cancer, cardiovascular disease improving, anti-inflammatory, and anti-oxidant activities, all of which are advantageous for human health. Recent studies have uncovered that naringenin and naringin influence gene expression by modulating epigenetic pathways, including microRNA (miRNA) regulation. This mechanism plays a crucial role in the therapeutic potential for various diseases. This paper reviews the epigenetic researches on the physiological activities of naringenin and naringin. It highlights how these compounds can exert diverse effects through different signaling pathways, thereby ameliorating associated diseases. These findings provide valuable insights for the future applications of naringenin and naringin.
Collapse
Affiliation(s)
- Xiao Wu
- Guangdong Engineering and Technology Research Center for Quality and Efficacy Re-Evaluation of Post-Marketed TCM, Guangdong Provincial Key Laboratory of Plant Stress Biology, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou 510275, PR China
| | - Hao Wu
- Guangdong Engineering and Technology Research Center for Quality and Efficacy Re-Evaluation of Post-Marketed TCM, Guangdong Provincial Key Laboratory of Plant Stress Biology, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou 510275, PR China
| | - Mengli Zhong
- Guangdong Engineering and Technology Research Center for Quality and Efficacy Re-Evaluation of Post-Marketed TCM, Guangdong Provincial Key Laboratory of Plant Stress Biology, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou 510275, PR China
| | - Yixuan Chen
- Guangdong Engineering and Technology Research Center for Quality and Efficacy Re-Evaluation of Post-Marketed TCM, Guangdong Provincial Key Laboratory of Plant Stress Biology, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou 510275, PR China
| | - Weiwei Su
- Guangdong Engineering and Technology Research Center for Quality and Efficacy Re-Evaluation of Post-Marketed TCM, Guangdong Provincial Key Laboratory of Plant Stress Biology, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou 510275, PR China
| | - Peibo Li
- Guangdong Engineering and Technology Research Center for Quality and Efficacy Re-Evaluation of Post-Marketed TCM, Guangdong Provincial Key Laboratory of Plant Stress Biology, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou 510275, PR China.
| |
Collapse
|
16
|
Barrett AM, Olayinka-Amao O, Martin S, Doshi D, Bishop KM, Youakim JM. Assessing Experiences With Trofinetide for Rett Syndrome: Interviews With Caregivers of Participants in Clinical Trials. Clin Ther 2025; 47:181-188. [PMID: 39824747 DOI: 10.1016/j.clinthera.2024.12.012] [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: 09/11/2024] [Revised: 11/18/2024] [Accepted: 12/20/2024] [Indexed: 01/20/2025]
Abstract
PURPOSE Rett syndrome (RTT) is a rare neurodevelopmental disorder that mainly affects girls and women. Trofinetide is approved for the treatment of RTT in adults and children aged ≥2 years. To gain insight into experiences with RTT and effects of trofinetide treatment at different stages of RTT, interviews with caregivers of individuals with RTT were conducted upon their exit from the open-label trofinetide trials. METHODS Interviews were conducted with caregivers of participants in the LILAC/LILAC-2 open-label extension trials of the phase 3 LAVENDER trial in participants aged 5 to 20 years, and in DAFFODIL, an open-label trial in participants aged 2 to 4 years. Caregivers were asked about the RTT effects, experiences with trofinetide, meaningfulness of treatment effects, and satisfaction. Qualitative thematic analysis was performed. FINDINGS Caregivers of 33 participants from the open-label trials were interviewed, including 26 from LILAC/LILAC-2 (mean age, 12.3 years) and 7 from DAFFODIL (mean age, 4.5 years). The most commonly reported effects of RTT in LILAC/LILAC-2 were no verbal communication (24/26 [92.3%]), unable to use hands (15/26 [57.7%]), repetitive hand movements (15/26 [57.7%]), unable to walk (15/26 [57.7%]), and seizures (14/26 [53.8%]). In DAFFODIL, the most commonly reported effects of RTT were no verbal communication (7/7 [100%]), impaired balance (4/7 [57.1%]), unable to use hands (3/7 [42.9%]), repetitive hand movements (3/7 [42.9%]), mood disturbance (3/7 [42.9%]), constipation (3/7 [42.9%]), and limited ability to use hands (3/7 [42.9%]). Caregivers most commonly reported improvements in hand use (11/26 [42.3%]), engagement with others (11/26 [42.3%]), eye gaze (8/26 [30.8%]), use of the Tobii eye tracking device (7/26 [26.9%]), and attention/focus/concentration (7/26 [26.9%]) in LILAC/LILAC-2. In DAFFODIL, caregivers reported improvements in new words (5/7 [71.4%]), hand use (4/7 [57.1%]), and eye contact (4/7 [57.1%]). Nearly all (31/32) caregivers were very satisfied or satisfied with trofinetide. IMPLICATIONS Caregivers of participants in open-label trofinetide trials reported improvements in RTT with meaningful impact in areas of motor function, communication, and engagement.
Collapse
Affiliation(s)
- Amy M Barrett
- RTI Health Solutions, Research Triangle Park, North Carolina
| | | | - Susan Martin
- RTI Health Solutions, Research Triangle Park, North Carolina
| | - Dilesh Doshi
- Acadia Pharmaceuticals, Inc., San Diego, California
| | | | | |
Collapse
|
17
|
Mattingly Z, Chetty S. Untangling the Molecular Mechanisms Contributing to Autism Spectrum Disorder Using Stem Cells. Autism Res 2025; 18:476-485. [PMID: 39989339 DOI: 10.1002/aur.70005] [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: 08/12/2024] [Revised: 02/06/2025] [Accepted: 02/08/2025] [Indexed: 02/25/2025]
Abstract
Autism spectrum disorder (ASD) is a complex neuro developmental condition characterized by significant genetic and phenotypic variability, making diagnosis and treatment challenging. The heterogeneity of ASD-associated genetic variants and the absence of clear causal factors in many cases complicate personalized care. Traditional models, such as postmortem brain tissue and animal studies, have provided valuable insights but are limited in capturing the dynamic processes and human-specific aspects of ASD pathology. Recent advances in human induced pluripotent stem cell (iPSC) technology have transformed ASD research by enabling the generation of patient-derived neural cells in both two-dimensional cultures and three-dimensional brain organoid models. These models retain the donor's genetic background, allowing researchers to investigate disease-specific cellular and molecular mechanisms while identifying potential therapeutic targets tailored to individual patients. This commentary highlights how stem cell-based approaches are advancing our understanding of ASD and paving the way for more personalized diagnostic and therapeutic strategies.
Collapse
Affiliation(s)
- Zoe Mattingly
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Sundari Chetty
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
- Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Harvard Stem Cell Institute, Cambridge, Massachusetts, USA
- Lurie Center for Autism, Massachusetts General Hospital, Boston, Massachusetts, USA
| |
Collapse
|
18
|
Liu S, Wang CY, Zheng P, Jia BB, Zemke NR, Ren P, Park HL, Ren B, Zhuang X. Cell type-specific 3D-genome organization and transcription regulation in the brain. SCIENCE ADVANCES 2025; 11:eadv2067. [PMID: 40009678 PMCID: PMC11864200 DOI: 10.1126/sciadv.adv2067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Accepted: 01/23/2025] [Indexed: 02/28/2025]
Abstract
3D organization of the genome plays a critical role in regulating gene expression. How 3D-genome organization differs among different cell types and relates to cell type-dependent transcriptional regulation remains unclear. Here, we used genome-scale DNA and RNA imaging to investigate 3D-genome organization in transcriptionally distinct cell types in the mouse cerebral cortex. We uncovered a wide spectrum of differences in the nuclear architecture and 3D-genome organization among different cell types, ranging from the size of the cell nucleus to higher-order chromosome structures and radial positioning of chromatin loci within the nucleus. These cell type-dependent variations in nuclear architecture and chromatin organization exhibit strong correlations with both the total transcriptional activity of the cell and transcriptional regulation of cell type-specific marker genes. Moreover, we found that the methylated DNA binding protein MeCP2 promotes active-inactive chromatin segregation and regulates transcription in a nuclear radial position-dependent manner that is highly correlated with its function in modulating active-inactive chromatin compartmentalization.
Collapse
Affiliation(s)
- Shiwei Liu
- Howard Hughes Medical Institute, Department of Chemistry and Chemical Biology, Department of Physics, Harvard University, Cambridge, MA, USA
| | - Cosmos Yuqi Wang
- Howard Hughes Medical Institute, Department of Chemistry and Chemical Biology, Department of Physics, Harvard University, Cambridge, MA, USA
| | - Pu Zheng
- Howard Hughes Medical Institute, Department of Chemistry and Chemical Biology, Department of Physics, Harvard University, Cambridge, MA, USA
- Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA, USA
| | - Bojing Blair Jia
- Bioinformatics and Systems Biology Graduate Program, Medical Scientist Training Program, University of California San Diego, La Jolla, CA, USA
| | - Nathan R. Zemke
- Department of Cellular and Molecular Medicine and Center for Epigenomics, University of California, San Diego School of Medicine, La Jolla, CA, USA
| | - Peter Ren
- Howard Hughes Medical Institute, Department of Chemistry and Chemical Biology, Department of Physics, Harvard University, Cambridge, MA, USA
- Graduate Program in Biophysics, Harvard University, Cambridge, MA, USA
| | - Hannah L. Park
- Howard Hughes Medical Institute, Department of Chemistry and Chemical Biology, Department of Physics, Harvard University, Cambridge, MA, USA
| | - Bing Ren
- Department of Cellular and Molecular Medicine and Center for Epigenomics, University of California, San Diego School of Medicine, La Jolla, CA, USA
| | - Xiaowei Zhuang
- Howard Hughes Medical Institute, Department of Chemistry and Chemical Biology, Department of Physics, Harvard University, Cambridge, MA, USA
| |
Collapse
|
19
|
Lang S, Marschik PB, Laudańska Z, Wilken B, Schaaf CP, Hahn A, Kulvicius T, Sigafoos J, Bölte S, Poustka L, Neul JL, Zhang D. Shared Environment - Different Genes: Speech-Language Development in a Pair of Dizygotic Twins with and Without MECP2 Mutation. J Genet Psychol 2025:1-16. [PMID: 39973088 DOI: 10.1080/00221325.2025.2465788] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Accepted: 02/06/2025] [Indexed: 02/21/2025]
Abstract
This retrospective study compared the speech-language development of a pair of dizygotic twin girls during the first 2 years of life: one with typical development (Twin A) and one with atypical development (Twin B), who was later diagnosed with Rett syndrome (RTT). Audio snippets were extracted from home videos, with nearly equal representation from each child. The audio analysis focused on articulatory complexity, voice characteristics, and linguistic variability. Despite sharing the same social-communicative environment, the twins' speech-language development diverged. From the first to second year of life, articulatory complexity and variability increased in the typically developing Twin A. In contrast, Twin B produced a high number of vocalizations in the 7th month, including canonical sounds with substantial variability. However, her vocalization quantity, complexity, and variability subsequently decreased, resulting in only sporadically discernable canonical vocalizations during her second year. This developmental trajectory points to very early stagnation and regression in Twin B, occurring earlier than typically observed. While Twin B displayed a range of typical vocalization features, deviations in the density and distribution of inspiratory and high-pitched vocalizations during the first year further suggest early speech-language abnormalities in RTT, preceding frank developmental stagnation and regression. As the study relied on limited retrospective data, the findings should be interpreted with caution, and further investigation is needed. Nevertheless, this twin study provides a unique perspective that deepens our understanding of early speech-language developmental profiles in RTT, especially in light of the intertwinement of genetic, individual, and contextual factors.
Collapse
Affiliation(s)
- Sigrun Lang
- Child and Adolescent Psychiatry and Psychotherapy, University Medical Center Göttingen, German Center for Child and Adolescent Health (DZKJ) and Leibniz ScienceCampus Primate Cognition, Göttingen, Germany
| | - Peter B Marschik
- Child and Adolescent Psychiatry and Psychotherapy, University Medical Center Göttingen, German Center for Child and Adolescent Health (DZKJ) and Leibniz ScienceCampus Primate Cognition, Göttingen, Germany
- Department of Child and Adolescent Psychiatry, University Hospital Heidelberg, Heidelberg University, Heidelberg, Germany
- Interdisciplinary Developmental Neuroscience (iDN), Division of Phoniatrics, Medical University of Graz, Graz, Austria
- Center of Neurodevelopmental Disorders (KIND), Centre for Psychiatry Research, Department of Women's and Children's Health, Child and Adolescent Psychiatry, Region Stockholm, Karolinska Institutet & Stockholm Health Care Services, Stockholm, Sweden
| | - Zuzanna Laudańska
- Department of Child and Adolescent Psychiatry, University Hospital Heidelberg, Heidelberg University, Heidelberg, Germany
| | - Bernd Wilken
- Department of Pediatric Neurology, Hospital Kassel, Kassel, Germany
| | - Christian P Schaaf
- Institute of Human Genetics, University Hospital Heidelberg, Heidelberg University, Heidelberg, Germany
| | - Alisa Hahn
- Child and Adolescent Psychiatry and Psychotherapy, University Medical Center Göttingen, German Center for Child and Adolescent Health (DZKJ) and Leibniz ScienceCampus Primate Cognition, Göttingen, Germany
| | - Tomas Kulvicius
- Child and Adolescent Psychiatry and Psychotherapy, University Medical Center Göttingen, German Center for Child and Adolescent Health (DZKJ) and Leibniz ScienceCampus Primate Cognition, Göttingen, Germany
- Department of Child and Adolescent Psychiatry, University Hospital Heidelberg, Heidelberg University, Heidelberg, Germany
| | - Jeff Sigafoos
- School of Education, Victoria University of Wellington, Wellington, New Zealand
| | - Sven Bölte
- Center of Neurodevelopmental Disorders (KIND), Centre for Psychiatry Research, Department of Women's and Children's Health, Child and Adolescent Psychiatry, Region Stockholm, Karolinska Institutet & Stockholm Health Care Services, Stockholm, Sweden
- Curtin Autism Research Group, Curtin School of Allied Health, Curtin University, Perth, Washington, Australia
| | - Luise Poustka
- Department of Child and Adolescent Psychiatry, University Hospital Heidelberg, Heidelberg University, Heidelberg, Germany
| | - Jeffrey L Neul
- Department of Pediatrics (Neurology), Vanderbilt Kennedy Center, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Dajie Zhang
- Department of Child and Adolescent Psychiatry, University Hospital Heidelberg, Heidelberg University, Heidelberg, Germany
- Interdisciplinary Developmental Neuroscience (iDN), Division of Phoniatrics, Medical University of Graz, Graz, Austria
| |
Collapse
|
20
|
Premachandran S, Ocay DD, Beaulieu C, Balduzzi J, Ye DL, Davidova A, Bokhari R, Ouellet JA, Orlowski J, Gendron L, Sharif-Naeini R, Ferland CE. Pain experience of children with Christianson syndrome. Pain 2025:00006396-990000000-00822. [PMID: 39945731 DOI: 10.1097/j.pain.0000000000003522] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2024] [Accepted: 11/01/2024] [Indexed: 02/20/2025]
Abstract
ABSTRACT Children with Christianson syndrome (CS), an X-linked neurodevelopmental disorder caused by loss-of-function mutations in the alkali cation/proton exchanger SLC9A6/NHE6, display severe cognitive impairments, mutism, and sensory abnormalities such as hyposensitivity to pain. However, it is unclear whether these children display other sensory abnormalities and whether their pain hyposensitivity is the result of an elevated pain threshold or a complete insensitivity to pain. To better characterize the sensory abnormalities in this disorder, we used a combination of a mouse model of CS and pain questionnaires directed at nonverbal patients with CS. We recruited 14 young male participants with CS and subjected them to a novel observational tool, the Pain Sensory and Painful Situations Questionnaire (PSQ), which takes multiple painful situations into account to broaden the description of pain expression. By analyzing social expressive behaviours of pain in these nonverbal patients, the PSQ documented that over 60% of the participants were unaffected by mechanical or inflammatory painful stimuli. This reduced pain sensitivity was also observed in the mouse CS model. Surprisingly, CS mice also displayed aversive reactions to innocuous stimuli, which prompted us to examine whether such reactions were also present in children with CS. Indeed, the results from the PSQ revealed that 30% to 50% of these patients showed an aversive response to normally innocuous stimuli like light touch and gusts of air. Our results demonstrate that children with CS have aversive reactions to innocuous stimuli and are hyposensitive to painful stimuli, the latter making them at risk for developing complications from unreported injuries.
Collapse
Affiliation(s)
- Shajenth Premachandran
- Department of Physiology, McGill University, Montreal, QC, Canada
- Alan Edwards Center for Research on Pain, Montreal, QC, Canada
| | - Don Daniel Ocay
- Department of Anesthesiology, Critical Care, and Pain Medicine, Boston Children's Hospital, Boston, MA, United States
- Department of Anaesthesia, Harvard Medical School, Boston, MA, United States
| | - Claudie Beaulieu
- Department of Pharmacology-Physiology, Université de Sherbrooke, Sherbrooke, QC, Canada
- Institut de Pharmacologie de Sherbrooke, Sherbrooke, QC, Canada
- Centre de Recherche du Centre Hospitalier Universitaire de Sherbrooke (CHUS), Sherbrooke, QC, Canada
- Quebec Pain Research Network, Sherbrooke, QC, Canada
| | - Jonathan Balduzzi
- Department of Physiology, McGill University, Montreal, QC, Canada
- Alan Edwards Center for Research on Pain, Montreal, QC, Canada
| | - Diana-Luk Ye
- Department of Experimental Surgery, McGill University, Montreal, QC, Canada
- Shriners Hospitals for Children Canada, Montreal, QC, Canada
| | - Albena Davidova
- Department of Physiology, McGill University, Montreal, QC, Canada
- Alan Edwards Center for Research on Pain, Montreal, QC, Canada
| | - Rakan Bokhari
- Department of Experimental Surgery, McGill University, Montreal, QC, Canada
- Shriners Hospitals for Children Canada, Montreal, QC, Canada
| | - Jean A Ouellet
- Department of Experimental Surgery, McGill University, Montreal, QC, Canada
- Shriners Hospitals for Children Canada, Montreal, QC, Canada
| | - John Orlowski
- Department of Physiology, McGill University, Montreal, QC, Canada
| | - Louis Gendron
- Department of Pharmacology-Physiology, Université de Sherbrooke, Sherbrooke, QC, Canada
- Institut de Pharmacologie de Sherbrooke, Sherbrooke, QC, Canada
- Centre de Recherche du Centre Hospitalier Universitaire de Sherbrooke (CHUS), Sherbrooke, QC, Canada
- Quebec Pain Research Network, Sherbrooke, QC, Canada
| | - Reza Sharif-Naeini
- Department of Physiology, McGill University, Montreal, QC, Canada
- Alan Edwards Center for Research on Pain, Montreal, QC, Canada
- Quebec Pain Research Network, Sherbrooke, QC, Canada
| | - Catherine E Ferland
- Department of Experimental Surgery, McGill University, Montreal, QC, Canada
- Shriners Hospitals for Children Canada, Montreal, QC, Canada
- Department of Anesthesia, McGill University, Montreal, QC, Canada
- Research Institute-McGill University Health Center, Montreal, QC, Canada
| |
Collapse
|
21
|
Ährlund-Richter S, Harpe J, Fernandes G, Lam R, Sur M. Persistent Disruptions in Prefrontal Connectivity Despite Behavioral Rescue by Environmental Enrichment in a Mouse Model of Rett Syndrome. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.10.637474. [PMID: 39990439 PMCID: PMC11844379 DOI: 10.1101/2025.02.10.637474] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/25/2025]
Abstract
Rett Syndrome, a neurodevelopmental disorder caused by loss-of-function mutations in the MECP2 gene, is characterized by severe motor, cognitive and emotional impairments. Some of the deficits may result from changes in cortical connections, especially downstream projections of the prefrontal cortex, which may also be targets of restoration following rearing conditions such as environmental enrichment that alleviate specific symptoms. Here, using a heterozygous Mecp2 +/- female mouse model closely analogous to human Rett Syndrome, we investigated the impact of early environmental enrichment on behavioral deficits and prefrontal cortex connectivity. Behavioral analyses revealed that enriched housing rescued fine motor deficits and reduced anxiety, with enrichment-housed Mecp2 +/- mice performing comparably to wild-type (WT) controls in rotarod and open field assays. Anatomical mapping of top-down anterior cingulate cortex (ACA) projections demonstrated altered prefrontal cortex connectivity in Mecp2 +/- mice, with increased axonal density in the somatosensory cortex and decreased density in the motor cortex compared to WT controls. ACA axons revealed shifts in hemispheric distribution, particularly in the medial network regions, with Mecp2 +/- mice exhibiting reduced ipsilateral dominance. These changes were unaffected by enriched housing, suggesting that structural abnormalities in prefrontal cortex connectivity persist despite behavioral improvements. Enriched housing rescued brain-derived neurotrophic factor (BDNF) levels in the hippocampus but failed to restore BDNF levels in the prefrontal cortex, consistent with the persistent deficits observed in prefrontal axonal projections. These findings highlight the focal nature of changes induced by reduction of MeCP2 and by exposure to environmental enrichment, and suggest that environmental enrichment starting in adolescence can alleviate behavioral deficits without reversing abnormalities in large-scale cortical connectivity.
Collapse
Affiliation(s)
- Sofie Ährlund-Richter
- The Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Jonathan Harpe
- The Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
- Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Giselle Fernandes
- The Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Ruby Lam
- The Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Mriganka Sur
- The Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
- Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| |
Collapse
|
22
|
Siqueira E, Velasco C, Tarrasón A, Soler M, Srinivas T, Setién F, Oliveira-Mateos C, Casado-Pelaez M, Martinez-Verbo L, Armstrong J, Esteller M, Alves L, Llobet A, Guil S. NEAT1-mediated regulation of proteostasis and mRNA localization impacts autophagy dysregulation in Rett syndrome. Nucleic Acids Res 2025; 53:gkaf074. [PMID: 39970285 PMCID: PMC11806351 DOI: 10.1093/nar/gkaf074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 01/21/2025] [Accepted: 01/28/2025] [Indexed: 02/10/2025] Open
Abstract
Rett syndrome (RTT) is a severe neurodevelopmental disorder primarily caused by loss-of-function mutations in the MECP2 gene, resulting in diverse cellular dysfunctions. Here, we investigated the role of the long noncoding RNA (lncRNA) NEAT1 in the context of MeCP2 deficiency using human neural cells and RTT patient samples. Through single-cell RNA sequencing and molecular analyses, we found that NEAT1 is markedly downregulated in MECP2 knockout (KO) cells at various stages of neural differentiation. NEAT1 downregulation correlated with aberrant activation of the mTOR pathway, abnormal protein metabolism, and dysregulated autophagy, contributing to the accumulation of protein aggregates and impaired mitochondrial function. Reactivation of NEAT1 in MECP2-KO cells rescued these phenotypes, indicating its critical role downstream of MECP2. Furthermore, direct RNA-RNA interaction was revealed as the key process for NEAT1 influence on autophagy genes, leading to altered subcellular localization of specific autophagy-related messenger RNAs and impaired biogenesis of autophagic complexes. Importantly, NEAT1 restoration rescued the morphological defects observed in MECP2-KO neurons, highlighting its crucial role in neuronal maturation. Overall, our findings elucidate lncRNA NEAT1 as a key mediator of MeCP2 function, regulating essential pathways involved in protein metabolism, autophagy, and neuronal morphology.
Collapse
Affiliation(s)
- Edilene Siqueira
- Josep Carreras Leukaemia Research Institute (IJC), Genesis of cancer Program, 08916 Badalona,Catalonia, Spain
- Conselho Nacional de Desenvolvimento Cientifico e Tecnológico (CNPq), 70.070-010 Brasilia, Brazil
| | - Cecilia D Velasco
- Laboratory of Neurobiology, Department of Pathology and Experimental Therapy, Institute of Neurosciences, University of Barcelona, 08907L’Hospitalet de Llobregat, Catalonia, Spain
- Bellvitge Biomedical Research Institute (IDIBELL), 08907 L’Hospitalet de Llobregat, Catalonia, Spain
| | - Ariadna Tarrasón
- Josep Carreras Leukaemia Research Institute (IJC), Genesis of cancer Program, 08916 Badalona,Catalonia, Spain
| | - Marta Soler
- Josep Carreras Leukaemia Research Institute (IJC), Genesis of cancer Program, 08916 Badalona,Catalonia, Spain
| | - Tara Srinivas
- Josep Carreras Leukaemia Research Institute (IJC), Genesis of cancer Program, 08916 Badalona,Catalonia, Spain
| | - Fernando Setién
- Josep Carreras Leukaemia Research Institute (IJC), Genesis of cancer Program, 08916 Badalona,Catalonia, Spain
| | - Cristina Oliveira-Mateos
- Josep Carreras Leukaemia Research Institute (IJC), Genesis of cancer Program, 08916 Badalona,Catalonia, Spain
| | - Marta Casado-Pelaez
- Josep Carreras Leukaemia Research Institute (IJC), Genesis of cancer Program, 08916 Badalona,Catalonia, Spain
| | - Laura Martinez-Verbo
- Josep Carreras Leukaemia Research Institute (IJC), Genesis of cancer Program, 08916 Badalona,Catalonia, Spain
| | - Judith Armstrong
- Institut de Recerca Pediàtrica, Hospital Sant Joan de Déu, 08950 Barcelona, Catalonia, Spain
- Servei de Medicina Genètica i Molecular, Hospital Sant Joan de Déu, 08950 Barcelona, Catalonia, Spain
- CIBER-ER (Biomedical Network Research Center for Rare Diseases), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Manel Esteller
- Josep Carreras Leukaemia Research Institute (IJC), Genesis of cancer Program, 08916 Badalona,Catalonia, Spain
- Centro de Investigación Biomedica en Red Cancer (CIBERONC), 28029 Madrid, Spain
- Institució Catalana de Recerca i Estudis Avançats (ICREA), 08010 Barcelona, Catalonia, Spain
- Physiological Sciences Department, School of Medicine and Health Sciences, University of Barcelona (UB), 08907 Barcelona, Catalonia, Spain
| | - Letícia F Alves
- Josep Carreras Leukaemia Research Institute (IJC), Genesis of cancer Program, 08916 Badalona,Catalonia, Spain
| | - Artur Llobet
- Laboratory of Neurobiology, Department of Pathology and Experimental Therapy, Institute of Neurosciences, University of Barcelona, 08907L’Hospitalet de Llobregat, Catalonia, Spain
- Bellvitge Biomedical Research Institute (IDIBELL), 08907 L’Hospitalet de Llobregat, Catalonia, Spain
| | - Sonia Guil
- Josep Carreras Leukaemia Research Institute (IJC), Genesis of cancer Program, 08916 Badalona,Catalonia, Spain
- Germans Trias i Pujol Health Science Research Institute, 08916 Badalona, Catalonia, Spain
| |
Collapse
|
23
|
Kaufmann WE, Oberman LM, Downs J, Leonard H, Barnes KV. Rett Syndrome Behaviour Questionnaire: Variability of Scores and Related Factors. J Child Adolesc Psychopharmacol 2025. [PMID: 39907092 DOI: 10.1089/cap.2024.0128] [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] [Indexed: 02/06/2025]
Abstract
Objective: Rett syndrome (RTT) is a severe neurodevelopmental disorder affecting predominantly females and associated with variants in the MECP2 gene. Recent success in clinical trials have resulted in an expanded use of the Rett Syndrome Behaviour Questionnaire (RSBQ) for clinical and research purposes. Implementation of the RSBQ as a global clinical severity scale has raised concerns about its construct validity considering its content, structure, and psychometric features. To further understand RSBQ data, we analyzed RSBQ scores available in the literature with a focus on variability and influencing factors. Methods: We identified publications reporting RSBQ total and/or subscale scores and summarized relevant study information, such as type of investigation, administration method, and descriptive data. We then analyzed means and standard deviations, calculating variance-to-mean ratios (VMR), as a measure of variability, when raw score descriptive statistics were available. Where appropriate, we compared means and VMRs by Welch t-tests. Results: Of the 14 publications identified, raw total scores from 5 observational studies and 4 clinical trials (baseline) were available. Raw subscale scores from four of the five observational studies were also available. We found a wide but comparable range of mean total scores for observational studies and clinical trials. However, VMRs were significantly higher in observational studies. Subscale scores showed either high (i.e., General Mood, Breathing Problems) or low (e.g., Hand Behaviours, Body Rocking and Expressionless Face) variability. Available data demonstrated greater variability in pediatric than adult groups and less variability when using interviews or electronic RSBQ administration compared with paper forms. Total score changes over time did not affect variability. Although certain studies offered insight into the relationship between the RSBQ and other measures, overall, data were insufficient for characterizing how RSBQ variability relates to other factors. Conclusions: Our findings on score variability support the need for more comprehensive reporting of RSBQ data, cohort characterization, and methodology; and the deployment of standardized RSBQ administration methods, such as advanced data capture systems. There is potential for use of subscales as outcome measures, subject to further psychometric validation studies, including prospective investigations testing the stability of RSBQ scores and influencing factors. Further examining the relationship between RSBQ scores and other instruments will aid in its interpretation as a clinical outcome measure.
Collapse
Affiliation(s)
- Walter E Kaufmann
- Department of Neurology, Boston Children's Hospital, Boston, Massachusetts, USA
- Department of Human Genetics, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Lindsay M Oberman
- National Institute of Mental Health Intramural Research Program, National Institutes of Health, Bethesda, Maryland, USA
| | - Jenny Downs
- The Kids Research Institute Australia, The University of Western Australia, Nedlands, Western Australia, Australia
| | - Helen Leonard
- The Kids Research Institute Australia, The University of Western Australia, Nedlands, Western Australia, Australia
| | | |
Collapse
|
24
|
Bajikar SS, Zhou J, O'Hara R, Tirumala HP, Durham MA, Trostle AJ, Dias M, Shao Y, Chen H, Wang W, Yalamanchili HK, Wan YW, Banaszynski LA, Liu Z, Zoghbi HY. Acute MeCP2 loss in adult mice reveals transcriptional and chromatin changes that precede neurological dysfunction and inform pathogenesis. Neuron 2025; 113:380-395.e8. [PMID: 39689710 PMCID: PMC11802321 DOI: 10.1016/j.neuron.2024.11.006] [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: 01/22/2024] [Revised: 09/25/2024] [Accepted: 11/08/2024] [Indexed: 12/19/2024]
Abstract
Mutations in the X-linked methyl-CpG-binding protein 2 (MECP2) gene cause Rett syndrome, a severe childhood neurological disorder. MeCP2 is a well-established transcriptional repressor, yet upon its loss, hundreds of genes are dysregulated in both directions. To understand what drives such dysregulation, we deleted Mecp2 in adult mice, circumventing developmental contributions and secondary pathogenesis. We performed time series transcriptional, chromatin, and phenotypic analyses of the hippocampus to determine the immediate consequences of MeCP2 loss and the cascade of pathogenesis. We find that loss of MeCP2 causes immediate and bidirectional progressive dysregulation of the transcriptome. To understand what drives gene downregulation, we profiled genome-wide histone modifications and found that a decrease in histone H3 acetylation (ac) at downregulated genes is among the earliest molecular changes occurring well before any measurable deficiencies in electrophysiology and neurological function. These data reveal a molecular cascade that drives disease independent of any developmental contributions or secondary pathogenesis.
Collapse
Affiliation(s)
- Sameer S Bajikar
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA; Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital, Houston, TX 77030, USA
| | - Jian Zhou
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA; Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital, Houston, TX 77030, USA
| | - Ryan O'Hara
- Cecil H. and Ida Green Center for Reproductive Biology Sciences, Children's Medical Center Research Institute, Department of Obstetrics and Gynecology, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Harini P Tirumala
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA; Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital, Houston, TX 77030, USA
| | - Mark A Durham
- Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital, Houston, TX 77030, USA; Program in Developmental Biology, Baylor College of Medicine, Houston, TX 77030, USA; Medical Scientist Training Program, Baylor College of Medicine, Houston, TX 77030, USA
| | - Alexander J Trostle
- Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital, Houston, TX 77030, USA; Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Michelle Dias
- Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital, Houston, TX 77030, USA; Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Yingyao Shao
- Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital, Houston, TX 77030, USA; Program in Developmental Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Hu Chen
- Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital, Houston, TX 77030, USA; Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Wei Wang
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA; Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital, Houston, TX 77030, USA
| | - Hari Krishna Yalamanchili
- Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital, Houston, TX 77030, USA; Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA; USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Ying-Wooi Wan
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA; Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital, Houston, TX 77030, USA
| | - Laura A Banaszynski
- Cecil H. and Ida Green Center for Reproductive Biology Sciences, Children's Medical Center Research Institute, Department of Obstetrics and Gynecology, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Zhandong Liu
- Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital, Houston, TX 77030, USA; Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Huda Y Zoghbi
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA; Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital, Houston, TX 77030, USA; Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA; Howard Hughes Medical Institute, Baylor College of Medicine, Houston, TX 77030, USA.
| |
Collapse
|
25
|
Ishido M, Higashi K, Mori H, Ueno M, Kurokawa K. DNA methylation profiles of transgenerational rat hyperactivity primed by silver nanoparticles: Comparison with valproate model rats of autism. Behav Brain Res 2025; 477:115293. [PMID: 39419183 DOI: 10.1016/j.bbr.2024.115293] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Revised: 09/23/2024] [Accepted: 10/14/2024] [Indexed: 10/19/2024]
Abstract
There is an increasing body of evidence suggesting that a single exposure to certain chemicals can have transgenerational effects, with the underlying mechanism believed to be epigenetic. However, it remains largely unknown whether psychiatric conditions like ADHD or autism, induced by environmental chemicals, can be inherited across generations. Pregnant rats were purchased from a commercial breeder. On the 7th day of gestation (E7), they were divided into two groups: one group was orally exposed to silver nanoparticles (AgNP; 4 mg/kg), while the control group received vehicle alone. The subsequent generation (F1) underwent spontaneous motor activity (SMA) measurements at 8-11 weeks of age. For breeding at 26 weeks of age, rats with higher SMA were selected from hyperactive litters, while untreated rats were randomly selected. This process was continued for four generations in both groups. The AgNP-primed rats at 4th generation displayed significantly higher SMA, 1.8 times greater than that of untreated rats. Intraperitoneal injection of valproic acid (150 mg/kg), an epigenetic modifier to 5-day-old rats causes adult hyperactivity (1.4-fold), suggesting that epigenetic modification contributes to rat hyperactivity. Global DNA methylation profiles in the mesencephalon were positively correlated in both groups of hyperactive rats. Furthermore, there were 7-8 common genes showing both hypermethylation and hypomethylation, which are involved in neuronal development, neuronal function, transcriptional activity, DNA binding activity, cell differentiation, ubiquitination processes, or histone modification, including Pax 6 and Mecp 2. Thus, it is most likely that rats retain hyperactivity through mesencephalic DNA methylation status across transgeneration.
Collapse
Affiliation(s)
- Masami Ishido
- Center for Environmental Risk & Health Research, National Institute for Environmental Studies, Tsukuba 305-8506, Japan.
| | - Kouichi Higashi
- Center for Information Biology, National Institute of Genetics, Mishima, Shizuoka 411-8540, Japan
| | - Hiroshi Mori
- Center for Information Biology, National Institute of Genetics, Mishima, Shizuoka 411-8540, Japan
| | - Masaki Ueno
- Department of Pathology and Host Defense, Faculty of Medicine, Kagawa University, 1750-1 Ikenobe, Miki-cho, Kita-gun, Kagawa 761-0793, Japan
| | - Ken Kurokawa
- Center for Information Biology, National Institute of Genetics, Mishima, Shizuoka 411-8540, Japan
| |
Collapse
|
26
|
Khaliulin I, Hamoudi W, Amal H. The multifaceted role of mitochondria in autism spectrum disorder. Mol Psychiatry 2025; 30:629-650. [PMID: 39223276 PMCID: PMC11753362 DOI: 10.1038/s41380-024-02725-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2024] [Revised: 08/21/2024] [Accepted: 08/23/2024] [Indexed: 09/04/2024]
Abstract
Normal brain functioning relies on high aerobic energy production provided by mitochondria. Failure to supply a sufficient amount of energy, seen in different brain disorders, including autism spectrum disorder (ASD), may have a significant negative impact on brain development and support of different brain functions. Mitochondrial dysfunction, manifested in the abnormal activities of the electron transport chain and impaired energy metabolism, greatly contributes to ASD. The aberrant functioning of this organelle is of such high importance that ASD has been proposed as a mitochondrial disease. It should be noted that aerobic energy production is not the only function of the mitochondria. In particular, these organelles are involved in the regulation of Ca2+ homeostasis, different mechanisms of programmed cell death, autophagy, and reactive oxygen and nitrogen species (ROS and RNS) production. Several syndromes originated from mitochondria-related mutations display ASD phenotype. Abnormalities in Ca2+ handling and ATP production in the brain mitochondria affect synaptic transmission, plasticity, and synaptic development, contributing to ASD. ROS and Ca2+ regulate the activity of the mitochondrial permeability transition pore (mPTP). The prolonged opening of this pore affects the redox state of the mitochondria, impairs oxidative phosphorylation, and activates apoptosis, ultimately leading to cell death. A dysregulation between the enhanced mitochondria-related processes of apoptosis and the inhibited autophagy leads to the accumulation of toxic products in the brains of individuals with ASD. Although many mitochondria-related mechanisms still have to be investigated, and whether they are the cause or consequence of this disorder is still unknown, the accumulating data show that the breakdown of any of the mitochondrial functions may contribute to abnormal brain development leading to ASD. In this review, we discuss the multifaceted role of mitochondria in ASD from the various aspects of neuroscience.
Collapse
Affiliation(s)
- Igor Khaliulin
- Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Wajeha Hamoudi
- Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Haitham Amal
- Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel.
| |
Collapse
|
27
|
Kitazawa S, Haraguchi R, Kitazawa R. Roles of osteoclasts in pathological conditions. Pathol Int 2025; 75:55-68. [PMID: 39704061 PMCID: PMC11849001 DOI: 10.1111/pin.13500] [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: 11/06/2024] [Revised: 11/26/2024] [Accepted: 11/29/2024] [Indexed: 12/21/2024]
Abstract
Bone is a unique organ crucial for locomotion, mineral metabolism, and hematopoiesis. It maintains homeostasis through a balance between bone formation by osteoblasts and bone resorption by osteoclasts, which is regulated by the basic multicellular unit (BMU). Abnormal bone metabolism arises from an imbalance in the BMU. Osteoclasts, derived from the monocyte-macrophage lineage, are regulated by the RANKL-RANK-OPG system, which is a key factor in osteoclast differentiation. RANKL activates osteoclasts through its receptor RANK, while OPG acts as a decoy receptor that inhibits RANKL. In trabecular bone, high turnover involves rapid bone formation and resorption, influenced by conditions such as malignancy and inflammatory cytokines that increase RANKL expression. Cortical bone remodeling, regulated by aged osteocytes expressing RANKL, is less understood, despite ongoing research into how Rett syndrome, characterized by MeCP2 abnormalities, affects RANKL expression. Balancing trabecular and cortical bone involves mechanisms that preserve cortical bone, despite overall bone mass reduction due to aging or oxidative stress. Research into genes like sFRP4, which modulates bone mass, highlights the complex regulation by BMUs. The roles of the RANKL-RANK-OPG system extend beyond bone, affecting processes such as aortic valve formation and temperature regulation, which highlight the interconnected nature of biological research.
Collapse
Affiliation(s)
- Sohei Kitazawa
- Department of Molecular PathologyEhime University Graduate School of Medicine, ShitsukawaToon CityJapan
| | - Ryuma Haraguchi
- Department of Molecular PathologyEhime University Graduate School of Medicine, ShitsukawaToon CityJapan
| | - Riko Kitazawa
- Department of Molecular PathologyEhime University Graduate School of Medicine, ShitsukawaToon CityJapan
- Division of Diagnostic PathologyEhime University Hospital, ShitsukawaToon CityJapan
| |
Collapse
|
28
|
Lötjönen J, Kurra V, Laivuori H, Bjelogrlić N. Broadening the Phenotype Spectrum of MECP2 Variants in Men. Mol Genet Genomic Med 2025; 13:e70056. [PMID: 39887655 PMCID: PMC11780493 DOI: 10.1002/mgg3.70056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Revised: 11/25/2024] [Accepted: 01/07/2025] [Indexed: 02/01/2025] Open
Abstract
BACKGROUND MECP2 variants cause X-chromosome-linked rare developmental syndromes. Typically, the mutation is sporadic, occurs in females and is fatal in men. Accurate genetic and clinical diagnostics are considered essential for the management of symptoms and the development of new treatments. These aims may be difficult to reach before more is known about factors resulting in highly variable clinical pictures among patients carrying the same MECP2 variant. We describe the clinical picture of two brothers carrying the same MECP2 variant and compare them with cases published in the literature. METHODS Most of the MECP2 mutations are known to be de novo mutations, which is why the recurrence of the mutation in the couple's other children is unlikely. Unexpectedly, our routine genetic testing revealed a 23-year-old man (P1) and his younger brother (P2) to carry the same hemizygous pathogenic missense variant c.419C>T, p.(Ala140Val) (transcript NM_004992.3) of MECP2, which was found to be inherited from their presumably asymptomatic mother. Thus, further clinical evaluation and comparison with literature cases was considered necessary. RESULTS The P1 has a severe syndromic intellectual disorder (ID), whereas his brother has a substantially milder ID predominantly limited to problems in verbal skills. Neither P1 nor his younger brother has been diagnosed with Rett syndrome. The P1 (unlike his younger brother) has several lingual, social and motor difficulties; disruptive behavior was the most difficult symptom to treat. P1's response to several medical and non-medical treatment trials has remained inadequate, thus requiring the patient to be hospitalised for a long time. The literature review revealed that apart from our family, there are five other families with more than one male carrying the same MECP2 p.Ala140Val mutation, such as P1 and P2. The phenotypes of all 24 men from us (n = 2) and others (n = 22) carrying the same, presumably non-lethal mutation show great variability. CONCLUSIONS The p.Ala140Val mutation of MECP2 in males is associated with a rare X-chromosomal developmental disorder with highly variable phenotypes. Further studies are needed to better understand all those influencing factors that can explain phenotypic differences within the same genotype to find optimal medicinal therapies.
Collapse
Affiliation(s)
- Johannes Lötjönen
- Faculty of Medicine and Health TechnologyTampere UniversityTampereFinland
| | - Venla Kurra
- Department of Clinical GeneticsTampere University HospitalTampereFinland
- Center for Child, Adolescent and Maternal Health Research, Faculty of Medicine and Health TechnologyTampere UniversityTampereFinland
| | - Hannele Laivuori
- Center for Child, Adolescent and Maternal Health Research, Faculty of Medicine and Health TechnologyTampere UniversityTampereFinland
- Department of Obstetrics and GynaecologyTampere University HospitalTampereFinland
| | - Nina Bjelogrlić
- Tampere University HospitalWellbeing Services County of PirkanmaaTampereFinland
| |
Collapse
|
29
|
Pehlivan D, Huang C, Harris HK, Coquery C, Mahat A, Maletic‐Savatic M, Mignon L, Aras S, Glaze DG, Layne CS, Sahelijo L, Zoghbi HY, McGinley MJ, Suter B. Comprehensive assessment reveals numerous clinical and neurophysiological differences between MECP2-allelic disorders. Ann Clin Transl Neurol 2025; 12:433-447. [PMID: 39838601 PMCID: PMC11822789 DOI: 10.1002/acn3.52269] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 10/27/2024] [Accepted: 11/21/2024] [Indexed: 01/23/2025] Open
Abstract
OBJECTIVE Rett syndrome (RTT) and MECP2 duplication syndrome (MDS) result from under- and overexpression of MECP2, respectively. Preclinical studies using genetic-based treatment showed robust phenotype recovery for both MDS and RTT. However, there is a risk of converting MDS to RTT, or vice versa, if accurate MeCP2 levels are not achieved. The aim of this study was to identify biomarkers distinguishing RTT from MDS. MATERIALS AND METHODS We prospectively enrolled 11 MDS and 6 male RTT like (MRL) individuals for a panel of clinical and neurophysiological assessments over two visits, 8-10 months apart. RESULTS We identified numerous clinical and physiological features as promising biomarkers. MRL individuals exhibited large amplitude whole body tremor, midline stereotypies (vs. hand flapping at sides in MDS), earlier neuromotor regression, and earlier onset but less commonly refractory epilepsy. In the neurophysiological domain, we observed several marked differences in sleep physiology between MDS/MRL and typically developing (TD) individuals including reduced sleeping time, increased delta power during rapid eye movement (REM) sleep, decreased occipital alpha and increased brain-wide delta power during wakefulness, and reduced spindle density and duration. MRL individuals also had much lower delta power during NREM 2 and 3 stages than the TD group. We found differences in spindle duration in the temporal lobes and spindle amplitude in the frontal lobes between MDS and MRL. DISCUSSION Our study revealed distinct clinical features of MDS and MRL that can be monitored during a clinical trial and may serve as target engagement, disease progression, or safety biomarkers for interventional studies.
Collapse
Affiliation(s)
- Davut Pehlivan
- Section of Pediatric Neurology and Developmental Neuroscience, Department of PediatricsBaylor College of MedicineHoustonTexas77030USA
- Blue Bird Circle Rett CenterTexas Children's HospitalHoustonTexas77030USA
- Texas Children's HospitalHoustonTexas77030USA
- Jan and Dan Duncan Neurological Research Institute at Texas Children's HospitalHoustonTexas77030USA
| | - Chengjun Huang
- Jan and Dan Duncan Neurological Research Institute at Texas Children's HospitalHoustonTexas77030USA
- Present address:
University of Health and Rehabilitation SciencesQingdao CityShandong ProvinceChina
| | - Holly K. Harris
- Texas Children's HospitalHoustonTexas77030USA
- Section of Developmental Pediatrics, Department of PediatricsBaylor College of MedicineHoustonTexas77054USA
| | | | - Aditya Mahat
- Jan and Dan Duncan Neurological Research Institute at Texas Children's HospitalHoustonTexas77030USA
| | - Mirjana Maletic‐Savatic
- Section of Pediatric Neurology and Developmental Neuroscience, Department of PediatricsBaylor College of MedicineHoustonTexas77030USA
- Jan and Dan Duncan Neurological Research Institute at Texas Children's HospitalHoustonTexas77030USA
| | | | - Sukru Aras
- Section of Pediatric Neurology and Developmental Neuroscience, Department of PediatricsBaylor College of MedicineHoustonTexas77030USA
- Jan and Dan Duncan Neurological Research Institute at Texas Children's HospitalHoustonTexas77030USA
| | - Daniel G. Glaze
- Section of Pediatric Neurology and Developmental Neuroscience, Department of PediatricsBaylor College of MedicineHoustonTexas77030USA
- Blue Bird Circle Rett CenterTexas Children's HospitalHoustonTexas77030USA
- Texas Children's HospitalHoustonTexas77030USA
| | - Charles S. Layne
- Department of Health and Human PerformanceUniversity of HoustonHoustonTexasUSA
- Center for Neuromotor and Biomechanics ResearchUniversity of HoustonHoustonTexasUSA
- Center for NeuroEngineering and Cognitive ScienceUniversity of HoustonHoustonTexasUSA
| | | | - Huda Y. Zoghbi
- Section of Pediatric Neurology and Developmental Neuroscience, Department of PediatricsBaylor College of MedicineHoustonTexas77030USA
- Blue Bird Circle Rett CenterTexas Children's HospitalHoustonTexas77030USA
- Texas Children's HospitalHoustonTexas77030USA
- Jan and Dan Duncan Neurological Research Institute at Texas Children's HospitalHoustonTexas77030USA
| | - Matthew J. McGinley
- Jan and Dan Duncan Neurological Research Institute at Texas Children's HospitalHoustonTexas77030USA
- Department of NeuroscienceBaylor College of MedicineHoustonTexas77030USA
| | - Bernhard Suter
- Section of Pediatric Neurology and Developmental Neuroscience, Department of PediatricsBaylor College of MedicineHoustonTexas77030USA
- Blue Bird Circle Rett CenterTexas Children's HospitalHoustonTexas77030USA
- Texas Children's HospitalHoustonTexas77030USA
| |
Collapse
|
30
|
Darwish M, Passarell J, Maxwell K, Bradley H, Bishop KM, Youakim JM. Population Pharmacokinetics of Trofinetide in a Pediatric Population Aged 2-4 Years with Rett Syndrome. Adv Ther 2025; 42:1009-1025. [PMID: 39692837 PMCID: PMC11787236 DOI: 10.1007/s12325-024-03058-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Accepted: 10/25/2024] [Indexed: 12/19/2024]
Abstract
INTRODUCTION Weight-banded trofinetide dosing improved physician- and caregiver-rated efficacy measures and had acceptable tolerability in patients aged 2‒4 years (DAFFODIL study) and 5‒20 years (LAVENDER study) with Rett syndrome (RTT). Selection of weight-banded dosing regimens for these studies was based on population pharmacokinetic (popPK) modeling and exposure simulations. This study applied an updated popPK model to confirm steady-state trofinetide exposures achieved in DAFFODIL patients were within target range. METHODS A popPK model was developed using data from 14 clinical studies of trofinetide in healthy volunteers and pediatric and adult patients, including the LAVENDER and DAFFODIL studies. Individual exposure measures (area under concentration-time curve over 0-12 h [AUC0-12] were generated via integration of the predicted concentration-time profile for each DAFFODIL study participant based on the popPK model and individual empiric Bayesian pharmacokinetic parameter estimates. Distributions of steady-state AUC0-12 values for each body-weight group were compared against target exposure (AUC0-12 = 800‒1200 µg·h/mL). RESULTS Distribution and box plots of simulated steady-state AUC0-12 values achieved with the weight-banded DAFFODIL dosing regimen used in younger individuals aged 2-4 years with RTT (twice daily trofinetide 5 g [≥ 9 to < 12 kg] or 6 g [≥ 12 to < 20 kg]) indicated good overlap with the target exposure range. Median steady-state AUC0-12 values for both body-weight bands fell within the target exposure range. CONCLUSIONS PopPK model-based simulations confirm that the weight-banded dosing regimen used in DAFFODIL is adequate to achieve target trofinetide exposure in 2- to 4-year-olds with RTT.
Collapse
Affiliation(s)
- Mona Darwish
- Acadia Pharmaceuticals Inc., 12830 El Camino Real, Suite 400, San Diego, CA, 92130, USA.
| | - Julie Passarell
- Clinical Pharmacology and Pharmacometrics Division, Simulations Plus Inc., Buffalo, NY, USA
| | - Kelly Maxwell
- Clinical Pharmacology and Pharmacometrics Division, Simulations Plus Inc., Buffalo, NY, USA
| | - Heather Bradley
- Acadia Pharmaceuticals Inc., 12830 El Camino Real, Suite 400, San Diego, CA, 92130, USA
| | - Kathie M Bishop
- Acadia Pharmaceuticals Inc., 12830 El Camino Real, Suite 400, San Diego, CA, 92130, USA
| | - James M Youakim
- Acadia Pharmaceuticals Inc., 12830 El Camino Real, Suite 400, San Diego, CA, 92130, USA
| |
Collapse
|
31
|
Gioiosa S, Gasparini S, Presutti C, Rinaldi A, Castrignanò T, Mannironi C. Integrated gene expression and alternative splicing analysis in human and mouse models of Rett syndrome. Sci Rep 2025; 15:2778. [PMID: 39843543 PMCID: PMC11754816 DOI: 10.1038/s41598-025-86114-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Accepted: 01/08/2025] [Indexed: 01/24/2025] Open
Abstract
Mutations of the MECP2 gene lead to Rett syndrome (RTT), a rare developmental disease causing severe intellectual and physical disability. How the loss or defective function of MeCP2 mediates RTT is still poorly understood. MeCP2 is a global gene expression regulator, acting at transcriptional and post-transcriptional levels. Little attention has been given so far to the contribution of alternative splicing (AS) dysregulation to RTT pathophysiology. To perform a comparative analysis of publicly available RNA sequencing (RNA-seq) studies and generate novel data resources for AS, we explored 100 human datasets and 130 mouse datasets from Mecp2-mutant models, processing data for gene expression and alternative splicing. Our comparative analysis across studies indicates common species-specific differentially expressed genes (DEGs) and differentially alternatively spliced (DAS) genes. Human and mouse dysregulated genes are involved in two main functional categories: cell-extracellular matrix adhesion regulation and synaptic functions, the first category more significantly enriched in human datasets. Our extensive bioinformatics study indicates, for the first time, a significant dysregulation of AS in human RTT datasets, suggesting the crucial contribution of altered RNA processing to the pathophysiology of RTT.
Collapse
Affiliation(s)
- Silvia Gioiosa
- CINECA, SuperComputing Applications and Innovation Department, Via dei Tizii 6, 00185, Rome, Italy.
| | - Silvia Gasparini
- Institute of Molecular Biology and Pathology, National Research Council, 00185, Rome, Italy
| | - Carlo Presutti
- Department of Biology and Biotechnology "C. Darwin", Sapienza University of Rome, 00185, Rome, Italy
| | - Arianna Rinaldi
- Department of Biology and Biotechnology "C. Darwin", Sapienza University of Rome, 00185, Rome, Italy
- Center for Research in Neurobiology "D. Bovet", University of Tuscia, Sapienza University of Rome, 00185, Rome, Italy
| | - Tiziana Castrignanò
- Department of Ecological and Biological Sciences (DEB), University of Tuscia, Largo Università snc, 01100, Viterbo, Italy
| | - Cecilia Mannironi
- Institute of Molecular Biology and Pathology, National Research Council, 00185, Rome, Italy.
| |
Collapse
|
32
|
Gabriel GC, Yagi H, Tan T, Bais A, Glennon BJ, Stapleton MC, Huang L, Reynolds WT, Shaffer MG, Ganapathiraju M, Simon D, Panigrahy A, Wu YL, Lo CW. Mitotic block and epigenetic repression underlie neurodevelopmental defects and neurobehavioral deficits in congenital heart disease. Nat Commun 2025; 16:469. [PMID: 39774941 PMCID: PMC11707140 DOI: 10.1038/s41467-024-55741-6] [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: 02/27/2024] [Accepted: 12/24/2024] [Indexed: 01/11/2025] Open
Abstract
Hypoplastic left heart syndrome (HLHS) is a severe congenital heart disease associated with microcephaly and poor neurodevelopmental outcomes. Here we show that the Ohia HLHS mouse model, with mutations in Sap130, a chromatin modifier, and Pcdha9, a cell adhesion protein, also exhibits microcephaly associated with mitotic block and increased apoptosis leading to impaired cortical neurogenesis. Transcriptome profiling, DNA methylation, and Sap130 ChIPseq analyses all demonstrate dysregulation of genes associated with autism and cognitive impairment. This includes perturbation of REST transcriptional regulation of neurogenesis, disruption of CREB signaling regulating synaptic plasticity, and defects in neurovascular coupling mediating cerebral blood flow. Adult mice harboring either the Pcdha9 mutation, which show normal brain anatomy, or forebrain-specific Sap130 deletion via Emx1-Cre, which show microcephaly, both demonstrate learning and memory deficits and autism-like behavior. These findings provide mechanistic insights indicating the adverse neurodevelopment in HLHS may involve cell autonomous/nonautonomous defects and epigenetic dysregulation.
Collapse
Affiliation(s)
- George C Gabriel
- Department of Pediatrics and Department of Developmental Biology, University of Pittsburgh, Pittsburgh, USA
| | - Hisato Yagi
- Department of Pediatrics and Department of Developmental Biology, University of Pittsburgh, Pittsburgh, USA
| | - Tuantuan Tan
- Department of Pediatrics and Department of Developmental Biology, University of Pittsburgh, Pittsburgh, USA
| | - Abha Bais
- Department of Pediatrics and Department of Developmental Biology, University of Pittsburgh, Pittsburgh, USA
| | - Benjamin J Glennon
- Department of Pediatrics and Department of Developmental Biology, University of Pittsburgh, Pittsburgh, USA
| | - Margaret C Stapleton
- Department of Pediatrics and Department of Developmental Biology, University of Pittsburgh, Pittsburgh, USA
| | - Lihua Huang
- Chinese University of Hong Kong, Hong Kong, China
| | - William T Reynolds
- Department of Pediatrics and Department of Developmental Biology, University of Pittsburgh, Pittsburgh, USA
| | - Marla G Shaffer
- Department of Pediatrics and Department of Developmental Biology, University of Pittsburgh, Pittsburgh, USA
| | | | - Dennis Simon
- Department of Critical Care Medicine, University of Pittsburgh, Pittsburgh, USA
| | - Ashok Panigrahy
- Department of Radiology, University of Pittsburgh, Pittsburgh, USA
| | - Yijen L Wu
- Department of Pediatrics and Department of Developmental Biology, University of Pittsburgh, Pittsburgh, USA
| | - Cecilia W Lo
- Department of Pediatrics and Department of Developmental Biology, University of Pittsburgh, Pittsburgh, USA.
| |
Collapse
|
33
|
Dionne O, Sabatié S, Laurent B. Deciphering the physiopathology of neurodevelopmental disorders using brain organoids. Brain 2025; 148:12-26. [PMID: 39222411 PMCID: PMC11706293 DOI: 10.1093/brain/awae281] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2024] [Revised: 07/25/2024] [Accepted: 08/09/2024] [Indexed: 09/04/2024] Open
Abstract
Neurodevelopmental disorders (NDD) encompass a range of conditions marked by abnormal brain development in conjunction with impaired cognitive, emotional and behavioural functions. Transgenic animal models, mainly rodents, traditionally served as key tools for deciphering the molecular mechanisms driving NDD physiopathology and significantly contributed to the development of pharmacological interventions aimed at treating these disorders. However, the efficacy of these treatments in humans has proven to be limited, due in part to the intrinsic constraint of animal models to recapitulate the complex development and structure of the human brain but also to the phenotypic heterogeneity found between affected individuals. Significant advancements in the field of induced pluripotent stem cells (iPSCs) offer a promising avenue for overcoming these challenges. Indeed, the development of advanced differentiation protocols for generating iPSC-derived brain organoids gives an unprecedented opportunity to explore human neurodevelopment. This review provides an overview of how 3D brain organoids have been used to investigate various NDD (i.e. Fragile X syndrome, Rett syndrome, Angelman syndrome, microlissencephaly, Prader-Willi syndrome, Timothy syndrome, tuberous sclerosis syndrome) and elucidate their pathophysiology. We also discuss the benefits and limitations of employing such innovative 3D models compared to animal models and 2D cell culture systems in the realm of personalized medicine.
Collapse
Affiliation(s)
- Olivier Dionne
- Research Center on Aging, Centre Intégré Universitaire de Santé et Services Sociaux de l'Estrie-Centre Hospitalier Universitaire de Sherbrooke, Sherbrooke, QC J1H 4C4, Canada
| | - Salomé Sabatié
- Research Center on Aging, Centre Intégré Universitaire de Santé et Services Sociaux de l'Estrie-Centre Hospitalier Universitaire de Sherbrooke, Sherbrooke, QC J1H 4C4, Canada
| | - Benoit Laurent
- Research Center on Aging, Centre Intégré Universitaire de Santé et Services Sociaux de l'Estrie-Centre Hospitalier Universitaire de Sherbrooke, Sherbrooke, QC J1H 4C4, Canada
- Department of Biochemistry and Functional Genomics, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC J1H 5H4, Canada
| |
Collapse
|
34
|
Canton APM, Macedo DB, Abreu AP, Latronico AC. Genetics and Epigenetics of Human Pubertal Timing: The Contribution of Genes Associated With Central Precocious Puberty. J Endocr Soc 2025; 9:bvae228. [PMID: 39839367 PMCID: PMC11746960 DOI: 10.1210/jendso/bvae228] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Indexed: 01/23/2025] Open
Abstract
Human puberty is a dynamic biological process determined by the increase in the pulsatile secretion of GnRH triggered by distinct factors not fully understood. Current knowledge reveals fine tuning between an increase in stimulatory factors and a decrease in inhibitory factors, where genetic and epigenetic factors have been indicated as key players in the regulation of puberty onset by distinct lines of evidence. Central precocious puberty (CPP) results from the premature reactivation of pulsatile secretion of GnRH. In the past decade, the identification of genetic causes of CPP has largely expanded, revealing hypothalamic regulatory factors of pubertal timing. Among them, 3 genes associated with CPP are linked to mechanisms involving DNA methylation, reinforcing the strong role of epigenetics underlying this disorder. Loss-of-function mutations in Makorin Ring-Finger Protein 3 (MKRN3) and Delta-Like Non-Canonical Notch Ligand 1 (DLK1), 2 autosomal maternally imprinted genes, have been described as relevant monogenic causes of CPP with the phenotype exclusively associated with paternal transmission. MKRN3 has proven to be a key component of the hypothalamic inhibitory input on GnRH neurons through different mechanisms. Additionally, rare heterozygous variants in the Methyl-CpG-Binding Protein 2 (MECP2), an X-linked gene that is a key factor of DNA methylation machinery, were identified in girls with sporadic CPP with or without neurodevelopmental disorders. In this mini-review, we focus on how the identification of genetic causes of CPP has revealed epigenetic regulators of human pubertal timing, summarizing the latest knowledge on the associations of puberty with MKRN3, DLK1, and MECP2.
Collapse
Affiliation(s)
- Ana Pinheiro Machado Canton
- Cellular and Molecular Endocrinology Laboratory LIM/25, Division of Endocrinology and Metabolism, Clinicas Hospital, School of Medicine, University of Sao Paulo, 01246-903 Sao Paulo, Brazil
| | - Delanie Bulcao Macedo
- Integrated Medical Care Center, Center for Health Sciences, University of Fortaleza (Unifor), Fortaleza 60811-905, Brazil
| | - Ana Paula Abreu
- Division of Endocrinology, Diabetes and Hypertension, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Ana Claudia Latronico
- Cellular and Molecular Endocrinology Laboratory LIM/25, Division of Endocrinology and Metabolism, Clinicas Hospital, School of Medicine, University of Sao Paulo, 01246-903 Sao Paulo, Brazil
- Discipline of Endocrinology and Metabolism, Department of Internal Medicine, School of Medicine, University of Sao Paulo, 05403-000, Sao Paulo, Brazil
| |
Collapse
|
35
|
Caffarelli C, Gonnelli S. The Management of Bone Defects in Rett Syndrome. Calcif Tissue Int 2025; 116:11. [PMID: 39751871 DOI: 10.1007/s00223-024-01322-0] [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: 04/29/2024] [Accepted: 10/29/2024] [Indexed: 01/04/2025]
Abstract
Rett syndrome (RS) is a rare neurodevelopmental disorder primarily caused by mutations in the X-linked methyl-CpG binding protein 2 (MECP2) gene, responsible for encoding MECP2 which plays a pivotal role in regulating gene expression. The neurological and non-neurological manifestations of RS vary widely in severity depending on the specific mutation type. Bone complications, mostly scoliosis but also osteoporosis, hip displacement, and a high rate of fractures, are among the most prevalent non-neurological comorbidities observed in girls with RS. Low bone mineral density (BMD) is primarily due to a slow rate of bone formation due to dysfunctional osteoblast activity. The use of anticonvulsants, immobilization, low physical activity, poor nutrition, and inadequate vitamin D intake all significantly hamper skeletal maturation and the accumulation of bone mass in RS girls, making them more susceptible to fragility fractures. In RS patients, the upper and lower limbs are the most common sites for fractures which are due to both a reduced BMD and a diminished bone size. This review summarizes the knowledge on risk factors for fragility fracture in patients with RS and proposes a potential diagnostic and therapeutic pathway to enhance low BMD and mitigate the risk of fragility fractures. In particular, this review focused on the importance of clinical and instrumental evaluation of bone status as a basis for adequate planning of nutritional, pharmacological, and surgical interventions to be undertaken. Additionally, the management of bone defects in individuals with RS should be customized to meet each person's specific needs, abilities, and general health.
Collapse
Affiliation(s)
- Carla Caffarelli
- Section of Internal Medicine, Department of Medicine, Surgery and Neuroscience, University of Siena, Policlinico Le Scotte, Viale Bracci 2, 53100, Siena, Italy.
| | - Stefano Gonnelli
- Department of Medicine, Surgery and Neuroscience, University of Siena, Siena, Italy
| |
Collapse
|
36
|
Lesire S, Lata R, Hoogvliets Y, Herrebosch K, Van De Velde P, Speleers A, Christ F, Van Belle S, Debyser Z. LEDGF interacts with the NID domain of MeCP2 and modulates MeCP2 condensates. Structure 2025; 33:78-90.e6. [PMID: 39500316 DOI: 10.1016/j.str.2024.10.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2024] [Revised: 08/20/2024] [Accepted: 10/09/2024] [Indexed: 01/06/2025]
Abstract
Methyl-CpG-binding protein 2 (MeCP2) is a ubiquitously expressed nuclear protein involved in transcriptional regulation and chromatin remodeling. MeCP2 exists in two isoforms, MeCP2 E1 and E2, which share the same functional domains. Loss-of-function mutations in the MeCP2 gene are the main cause of Rett syndrome (RTT). Previous studies identified a complex formation between MeCP2 and lens epithelium derived growth factor (LEDGF), a transcriptional regulator that exists in two isoforms, LEDGF/p75 and LEDGF/p52. Here, we characterized the molecular and functional interaction between MeCP2 and LEDGF. The NCoR interaction domain (NID) domain in MeCP2 is essential for the direct binding to the PWWP-CR1 region of LEDGF. Introduction of R306C, an RTT mutation in the NID of MeCP2, reduced the interaction with LEDGF. Our data reveal mutual inhibition of MeCP2 and LEDGF multimerization due to overlapping binding sites. Aligning with this observation, LEDGF depletion resulted in larger MeCP2 and DNA foci in NIH3T3 cells, suggesting a role for the MeCP2-LEDGF complex in chromatin organization.
Collapse
Affiliation(s)
- Saskia Lesire
- KU Leuven, Department of Pharmaceutical and Pharmacological Sciences, 3000 Leuven, Flanders, Belgium
| | - Rodrigo Lata
- KU Leuven, Department of Pharmaceutical and Pharmacological Sciences, 3000 Leuven, Flanders, Belgium
| | - Yannick Hoogvliets
- KU Leuven, Department of Pharmaceutical and Pharmacological Sciences, 3000 Leuven, Flanders, Belgium
| | - Kune Herrebosch
- KU Leuven, Department of Pharmaceutical and Pharmacological Sciences, 3000 Leuven, Flanders, Belgium
| | - Paulien Van De Velde
- KU Leuven, Department of Pharmaceutical and Pharmacological Sciences, 3000 Leuven, Flanders, Belgium
| | - Anouk Speleers
- KU Leuven, Department of Pharmaceutical and Pharmacological Sciences, 3000 Leuven, Flanders, Belgium
| | - Frauke Christ
- KU Leuven, Department of Pharmaceutical and Pharmacological Sciences, 3000 Leuven, Flanders, Belgium
| | - Siska Van Belle
- KU Leuven, Department of Pharmaceutical and Pharmacological Sciences, 3000 Leuven, Flanders, Belgium
| | - Zeger Debyser
- KU Leuven, Department of Pharmaceutical and Pharmacological Sciences, 3000 Leuven, Flanders, Belgium.
| |
Collapse
|
37
|
Rana S, Fusco AF, Witkin JM, Radin DP, Cerne R, Lippa A, Fuller DD. Pharmacological modulation of respiratory control: Ampakines as a therapeutic strategy. Pharmacol Ther 2025; 265:108744. [PMID: 39521442 PMCID: PMC11849399 DOI: 10.1016/j.pharmthera.2024.108744] [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: 08/04/2024] [Revised: 10/18/2024] [Accepted: 10/29/2024] [Indexed: 11/16/2024]
Abstract
Ampakines are a class of compounds that are positive allosteric modulators of α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptors and enhance glutamatergic neurotransmission. Glutamatergic synaptic transmission and AMPA receptor activation are fundamentally important to the genesis and propagation of the neural impulses driving breathing, including respiratory motoneuron depolarization. Ampakines therefore have the potential to modulate the neural control of breathing. In this paper, we describe the influence of ampakines on respiratory motor output in health and disease. We dissect the molecular mechanisms underlying ampakine action, delineate the diverse targets of ampakines along the respiratory neuraxis, survey the spectrum of respiratory disorders in which ampakines have been tested, and culminate with an examination of how ampakines modulate respiratory function after spinal cord injury. Collectively, the studies reviewed here indicate that ampakines may be a useful adjunctive strategy to pair with conventional respiratory rehabilitation approaches in conditions with impaired neural activation of the respiratory muscles.
Collapse
Affiliation(s)
- Sabhya Rana
- Department of Physical Therapy, University of Florida, Gainesville, FL 32610, United States of America; McKnight Brain Institute, University of Florida, Gainesville, FL 32610, United States of America; Center for Respiratory Research and Rehabilitation, University of Florida, Gainesville, FL 32610, United States of America.
| | - Anna F Fusco
- Department of Physical Therapy, University of Florida, Gainesville, FL 32610, United States of America; McKnight Brain Institute, University of Florida, Gainesville, FL 32610, United States of America; Center for Respiratory Research and Rehabilitation, University of Florida, Gainesville, FL 32610, United States of America
| | - Jeffrey M Witkin
- Laboratory of Antiepileptic Drug Discovery, St. Vincent's Hospital, Indianapolis, IN, United States of America; Departments of Neuroscience and Trauma Research, Ascension St. Vincent Hospital, Indianapolis, IN, United States of America; RespireRx Pharmaceuticals Inc, Glen Rock, NJ, United States of America
| | - Daniel P Radin
- RespireRx Pharmaceuticals Inc, Glen Rock, NJ, United States of America
| | - Rok Cerne
- Laboratory of Antiepileptic Drug Discovery, St. Vincent's Hospital, Indianapolis, IN, United States of America; RespireRx Pharmaceuticals Inc, Glen Rock, NJ, United States of America; Faculty of Medicine, University of Ljubljana, Zaloška cesta 4, Ljubljana, Slovenia
| | - Arnold Lippa
- RespireRx Pharmaceuticals Inc, Glen Rock, NJ, United States of America
| | - David D Fuller
- Department of Physical Therapy, University of Florida, Gainesville, FL 32610, United States of America; McKnight Brain Institute, University of Florida, Gainesville, FL 32610, United States of America; Center for Respiratory Research and Rehabilitation, University of Florida, Gainesville, FL 32610, United States of America
| |
Collapse
|
38
|
Anabella LYX, Rong YJ, Nagadia RH. Two Head and Neck Cancers in an Adult With Intellectual Disability and Rett Syndrome: A Case Report. SPECIAL CARE IN DENTISTRY 2025; 45:1-6. [PMID: 39817614 DOI: 10.1111/scd.13107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 12/26/2024] [Accepted: 12/28/2024] [Indexed: 01/18/2025]
Abstract
The cases of head and neck cancer among persons with intellectual disability (PWID) are infrequently reported and therefore poorly understood. PWID often face increased barriers of access to healthcare, which can be further compounded when faced with a cancer diagnosis. This report presents the case of a 34-year-old Chinese female patient with Rett syndrome and intellectual disability, presenting with two primary cancers of the tongue and the trachea. The case highlights the importance of regular dental review in cancer surveillance, post-operative supportive dental care, and challenges in behavioral management in cancer diagnosis and treatment.
Collapse
Affiliation(s)
- Low Yu Xin Anabella
- Department of Restorative Dentistry, National Dental Centre, Singapore, Republic of Singapore
| | - Yang Jing Rong
- Department of Restorative Dentistry, National Dental Centre, Singapore, Republic of Singapore
| | - Rahul Harshad Nagadia
- Department of Head & Neck Surgery, Singapore General Hospital, Singapore, Republic of Singapore
- Department of Oral & Maxillofacial Surgery, National Dental Centre, Singapore, Republic of Singapore
| |
Collapse
|
39
|
Sun C, Zhao Y, Guo L, Qiu J, Peng Q. The interplay between histone modifications and nuclear lamina in genome regulation. J Genet Genomics 2025; 52:24-38. [PMID: 39426590 DOI: 10.1016/j.jgg.2024.10.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Revised: 10/08/2024] [Accepted: 10/09/2024] [Indexed: 10/21/2024]
Abstract
Gene expression is regulated by chromatin architecture and epigenetic remodeling in cell homeostasis and pathologies. Histone modifications act as the key factors to modulate the chromatin accessibility. Different histone modifications are strongly associated with the localization of chromatin. Heterochromatin primarily localizes at the nuclear periphery, where it interacts with lamina proteins to suppress gene expression. In this review, we summarize the potential bridges that have regulatory functions of histone modifications in chromatin organization and transcriptional regulation at the nuclear periphery. We use lamina-associated domains (LADs) as examples to elucidate the biological roles of the interactions between histone modifications and nuclear lamina in cell differentiation and development. In the end, we highlight the technologies that are currently used to identify and visualize histone modifications and LADs, which could provide spatiotemporal information for understanding their regulatory functions in gene expression and discovering new targets for diseases.
Collapse
Affiliation(s)
- Chang Sun
- Institute of Systems and Physical Biology, Shenzhen Bay Laboratory, Shenzhen 518132, China; Faculty of Medicine and Health Sciences, Barcelona University, Barcelona, Spain
| | - Yanjing Zhao
- Institute of Systems and Physical Biology, Shenzhen Bay Laboratory, Shenzhen 518132, China; Division of Life Science, Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China
| | - Liping Guo
- Institute of Systems and Physical Biology, Shenzhen Bay Laboratory, Shenzhen 518132, China; School of Biology and Biological Engineering, South China University of Technology, Guangzhou, Guangdong 510006, China
| | - Juhui Qiu
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, College of Bioengineering, Chongqing University, Chongqing 400030, China.
| | - Qin Peng
- Institute of Systems and Physical Biology, Shenzhen Bay Laboratory, Shenzhen 518132, China.
| |
Collapse
|
40
|
Sonn JY, Zoghbi HY. MeCP2 goes into unmethylated territories. Nat Neurosci 2025; 28:4-5. [PMID: 39690321 DOI: 10.1038/s41593-024-01846-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2024]
Affiliation(s)
- Jun Young Sonn
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX, USA
| | - Huda Y Zoghbi
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA.
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX, USA.
- Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA.
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA.
- Department of Neurology, Baylor College of Medicine, Houston, TX, USA.
- Howard Hughes Medical Institute, Baylor College of Medicine, Houston, TX, USA.
| |
Collapse
|
41
|
Mishra GP, Sun EX, Chin T, Eckhardt M, Greenberg ME, Stroud H. Interaction of methyl-CpG-binding protein 2 (MeCP2) with distinct enhancers in the mouse cortex. Nat Neurosci 2025; 28:62-71. [PMID: 39578572 DOI: 10.1038/s41593-024-01808-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Accepted: 09/25/2024] [Indexed: 11/24/2024]
Abstract
Mutations in methyl-CpG-binding protein 2 (MeCP2) cause Rett syndrome. MeCP2 is thought to regulate gene transcription by binding to methylated DNA broadly across the genome. Here, using cleavage under target and release under nuclease (CUT&RUN) assays in the adult mouse cortex, we show that MeCP2 strongly binds to specific gene enhancers that we call MeCP2-binding hotspots (MBHs). Unexpectedly, we find that MeCP2 binding to MBHs occurs in a DNA methylation-independent manner at MBHs. Multiple MBH sites surrounding genes mediate the transcriptional repression of genes enriched for neuronal functions. We show that MBHs regulate genes irrespective of genic methylation levels, suggesting that MeCP2 controls transcription via an intragenic methylation-independent mechanism. Hence, disruption of intragenic methylation-independent gene regulation by MeCP2 may in part underlie Rett syndrome.
Collapse
Affiliation(s)
- Gyan Prakash Mishra
- Department of Neuroscience, Peter O'Donnell Jr. Brain Institute, UT Southwestern Medical Center, Dallas, TX, USA
| | - Eric X Sun
- Department of Neuroscience, Peter O'Donnell Jr. Brain Institute, UT Southwestern Medical Center, Dallas, TX, USA
| | - Tiffany Chin
- Department of Neuroscience, Peter O'Donnell Jr. Brain Institute, UT Southwestern Medical Center, Dallas, TX, USA
| | - Mandy Eckhardt
- Department of Neuroscience, Peter O'Donnell Jr. Brain Institute, UT Southwestern Medical Center, Dallas, TX, USA
| | | | - Hume Stroud
- Department of Neuroscience, Peter O'Donnell Jr. Brain Institute, UT Southwestern Medical Center, Dallas, TX, USA.
| |
Collapse
|
42
|
Byiers BJ, Merbler AM, Burkitt CC, Symons FJ. Challenges in Using Parent-Reported Bed and Wake Times for Actigraphy Scoring in Rett-Related Syndromes. AMERICAN JOURNAL ON INTELLECTUAL AND DEVELOPMENTAL DISABILITIES 2025; 130:1-12. [PMID: 39709989 DOI: 10.1352/1944-7558-130.1.1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Accepted: 06/18/2024] [Indexed: 12/24/2024]
Abstract
Sleep problems are common in Rett syndrome and other neurogenetic syndromes. Actigraphy is a cost-effective, objective method for measuring sleep. Current guidelines require caregiver-reported bed and wake times to facilitate actigraphy data scoring. The current study examined missingness and consistency of caregiver-reported bed and wake times from paper sleep diaries and actigraphy event mark button presses in a sample of 38 individuals with Rett and related syndromes (aged 2-36 years, mean = 13.1) across two 14-day collection time points. Rates of missingness and discrepancy between the 2 sources were relatively high and correlated with clinical severity and quality of life. Overall, the results suggest a need for alternative actigraphy scoring methods that do not rely on caregiver report in this population.
Collapse
Affiliation(s)
- Breanne J Byiers
- Breanne J. Byiers and Alyssa M. Merbler, University of Minnesota, Chantel C. Burkitt, Gillette Children's Specialty Healthcare, St. Paul, MN, and Frank J. Symons, University of Minnesota
| | - Alyssa M Merbler
- Breanne J. Byiers and Alyssa M. Merbler, University of Minnesota, Chantel C. Burkitt, Gillette Children's Specialty Healthcare, St. Paul, MN, and Frank J. Symons, University of Minnesota
| | - Chantel C Burkitt
- Breanne J. Byiers and Alyssa M. Merbler, University of Minnesota, Chantel C. Burkitt, Gillette Children's Specialty Healthcare, St. Paul, MN, and Frank J. Symons, University of Minnesota
| | - Frank J Symons
- Breanne J. Byiers and Alyssa M. Merbler, University of Minnesota, Chantel C. Burkitt, Gillette Children's Specialty Healthcare, St. Paul, MN, and Frank J. Symons, University of Minnesota
| |
Collapse
|
43
|
Torres RF, Llontop N, Espinoza CS, Kerr B. Environmental Enrichment and Epigenetic Changes in the Brain: From the Outside to the Deep Inside. Subcell Biochem 2025; 108:217-230. [PMID: 39820864 DOI: 10.1007/978-3-031-75980-2_6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2025]
Abstract
The brain plays a vital role in maintaining homeostasis and effective interaction with the environment, shaped by genetic and environmental factors throughout neurodevelopment and maturity. While genetic components dictate initial neurodevelopment stages, epigenetics-specifically neuroepigenetics-modulates gene expression in response to environmental influences, allowing for brain adaptability and plasticity. This interplay is particularly evident in neuropathologies like Rett syndrome and CDKL5 deficiency syndrome, where disruptions in neuroepigenetic processes underline significant cognitive and motor impairments. The environmental enrichment paradigm, introduced by Donald Hebb in the late 1940s, demonstrates how enriching stimuli-such as complex sensory, social, and cognitive inputs-affect brain structure and function. Despite methodological variability, evidence reveals that enriched environments catalyze beneficial changes in behavior and neuroanatomy, including increased synaptic plasticity, enhanced motor coordination, and improved cognitive performance in rodent models. Additionally, environmental enrichment induces epigenetic modifications that facilitate these outcomes, highlighting the necessity of understanding the mechanisms driving gene expression changes within the context of enriched experiences. Ultimately, this manifold relationship between environment, neuroepigenetic modulation, and brain function highlights the brain's capacity for change, reinforcing the importance of considering environmental factors in studies of neurodevelopment and therapy for neurological disorders.
Collapse
Affiliation(s)
- Rodrigo F Torres
- Facultad de Medicina y Ciencia, Universidad San Sebastián, Santiago, Chile
- Facultad de Medicina y Ciencia, Universidad San Sebastián, Puerto Montt, Chile
| | - Nuria Llontop
- Facultad de Medicina y Ciencia, Universidad San Sebastián, Santiago, Chile
| | - C Sofía Espinoza
- Facultad de Medicina y Ciencia, Universidad San Sebastián, Santiago, Chile
| | - Bredford Kerr
- Facultad de Medicina y Ciencia, Universidad San Sebastián, Santiago, Chile.
| |
Collapse
|
44
|
Vega-Hanna L, Casas-Alba D, Balsells S, Bolasell M, Rubio P, García-García A, García-García O, O’Callaghan M, Pascual-Alonso A, Armstrong J, Martinez-Monseny AF. MECP2 Duplication Syndrome: AI-Based Diagnosis, Severity Scale Development and Correlation with Clinical and Molecular Variables. Diagnostics (Basel) 2024; 15:10. [PMID: 39795538 PMCID: PMC11720083 DOI: 10.3390/diagnostics15010010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2024] [Revised: 12/13/2024] [Accepted: 12/20/2024] [Indexed: 01/13/2025] Open
Abstract
Background: MECP2 duplication syndrome (MDS) (MIM#300260) is a rare X-linked neurodevelopmental disorder. This study aims to (1) develop a specific clinical severity scale, (2) explore its correlation with clinical and molecular variables, and (3) automate diagnosis using the Face2gene platform. Methods: A retrospective study was conducted on genetically confirmed MDS patients who were evaluated at a pediatric hospital between 2012 and 2024. Epidemiological, clinical, and molecular data were collected. A standardized clinical questionnaire was collaboratively developed with input from physicians and parents. Patient photographs were used to train Face2Gene. Results: Thirty-five patients (0-24 years, 30 males) were included. Key features in males comprised intellectual disability (100%), hypotonia (93%), autism spectrum disorder (77%) and developmental regression (52%). Recurrent respiratory infections (79%), dysphagia (73%), constipation (73%) and gastroesophageal reflux (57%) were common. Seizures occurred in 53%, with 33% being treatment-refractory. The Face2Gene algorithm was successfully trained to identify MDS. A specific clinical severity scale (MECPDup) was developed and validated, correlating with the MBA (a scale developed for Rett syndrome). The MECPDup score was significantly higher in males (p < 0.001) and those with early death (p = 0.003). It showed significant positive correlations with age (p < 0.001) and duplication size (p = 0.044). Conclusions: This study expands the understanding of MDS through comprehensive clinical and molecular insights. The integration of AI-based facial recognition technology and the development of the MECPDup severity scale hold promise for enhancing diagnostic accuracy, monitoring disease progression, and evaluating treatment responses in individuals affected by MDS.
Collapse
Affiliation(s)
- Lourdes Vega-Hanna
- Genetics Department, Hospital Sant Joan de Déu, Member of ERN-ITHACA, 08950 Esplugues de Llobregat, Spain
| | - Dídac Casas-Alba
- Genetics Department, Hospital Sant Joan de Déu, Member of ERN-ITHACA, 08950 Esplugues de Llobregat, Spain
| | - Sol Balsells
- Statistics Department, Fundació de Recerca Sant Joan de Déu, 08950 Esplugues de Llobregat, Spain
| | - Mercè Bolasell
- Genetics Department, Hospital Sant Joan de Déu, Member of ERN-ITHACA, 08950 Esplugues de Llobregat, Spain
| | - Patricia Rubio
- Genetics Department, Hospital Sant Joan de Déu, Member of ERN-ITHACA, 08950 Esplugues de Llobregat, Spain
| | - Ana García-García
- Clinical Immunology Unit, Hospital Sant Joan de Déu-Hospital Clínic, 08950 Barcelona, Spain;
| | - Oscar García-García
- Servei d’Atenció Ambulatòria, Fundació Aspace Catalunya, 08038 Barcelona, Spain;
| | - Mar O’Callaghan
- Pediatric Neurology Department, Hospital Sant Joan de Déu, 08950 Esplugues de Llobregat, Spain;
| | | | - Judith Armstrong
- Genetics Department, Hospital Sant Joan de Déu, Member of ERN-ITHACA, 08950 Esplugues de Llobregat, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | | | | |
Collapse
|
45
|
Dominguez G, Wu Y, Zhou J. Epigenetic Regulation and Neurodevelopmental Disorders: From MeCP2 to the TCF20/PHF14 Complex. Genes (Basel) 2024; 15:1653. [PMID: 39766920 PMCID: PMC11728296 DOI: 10.3390/genes15121653] [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: 12/01/2024] [Revised: 12/17/2024] [Accepted: 12/17/2024] [Indexed: 01/12/2025] Open
Abstract
BACKGROUND Neurodevelopmental disorders (NDDs) affect approximately 15% of children and adolescents worldwide. This group of disorders is often polygenic with varying risk factors, with many associated genes converging on shared molecular pathways, including chromatin regulation and transcriptional control. Understanding how NDD-associated chromatin regulators and protein complexes orchestrate these regulatory pathways is crucial for elucidating NDD pathogenesis and developing targeted therapeutic strategies. Recently, the TCF20/PHF14 chromatin complex was identified in the mammalian brain, expanding the list of chromatin regulatory remodelers implicated in NDDs. This complex-which includes MeCP2, RAI1, TCF20, PHF14, and HMG20A-plays a vital role in epigenetic and transcriptional regulation. METHODS We review and summarize current research and clinical reports pertaining to the different components of the MeCP2-interacting TCF20/PHF14 complex. We examine the NDDs associated with the TCF20/PHF14 complex, explore the molecular and neuronal functions of its components, and discuss emerging therapeutic strategies targeting this complex to mitigate symptoms, with broader applicability to other NDDs. RESULTS Mutations in the genes encoding the components of the MeCP2-interacting TCF20/PHF14 complex have been linked to various NDDs, underscoring its critical contribution to brain development and NDD pathogenesis. CONCLUSIONS The MeCP2-interacting TCF20/PHF14 complex and its associated NDDs could serve as a model system to provide insight into the interplay between epigenetic regulation and NDD pathogenesis.
Collapse
Affiliation(s)
- Gaea Dominguez
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA 30322, USA; (G.D.); (Y.W.)
| | - Yongji Wu
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA 30322, USA; (G.D.); (Y.W.)
| | - Jian Zhou
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA 30322, USA; (G.D.); (Y.W.)
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA 30322, USA
| |
Collapse
|
46
|
Chi J, Song X, Liu J, Oh EG, Zhang Z, Xu Z, Yang H, Yuan H, Zhang Y. Scoliosis in Rett syndrome: a comparative analysis of postoperative complications. J Pediatr Orthop B 2024:01202412-990000000-00226. [PMID: 39718249 DOI: 10.1097/bpb.0000000000001223] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2024]
Abstract
Rett syndrome, a neurodevelopmental disorder primarily affecting females, presents unique challenges in managing associated scoliosis. This study aims to evaluate the efficacy and challenges of posterior spinal fusion (PSF) in Rett syndrome patients by analyzing postoperative complications. A retrospective cohort study was conducted using a large national database. We included Rett syndrome patients aged 10-18 years who underwent PSF between 2010 and 2020. Outcomes such as medical and surgical complications, emergency department visits, readmissions, mortality, and reoperation rates up to 5 years were compared with a matched neuromuscular scoliosis (NMS) group. The study identified 195 Rett syndrome patients and 973 NMS patients. Post-surgery, Rett syndrome patients showed a significantly higher incidence of pneumothorax (56.9%, P < 0.001), respiratory failure (24.6%, P = 0.013), and pneumonia (26.2%, P < 0.001). Additionally, ileus (7.2%, P = 0.041), acute kidney injury (14.9%, P = 0.029), and urinary tract infections (14.9%, P < 0.001) were also significantly more frequent in the Rett syndrome group. Rett syndrome group also had higher rates of transfusion (11.3%, P = 0.004). Interestingly, the incidence of pseudarthrosis, implant complications, junctional failures, and the necessity for reoperation did not significantly differ at postoperative year 2. Mid-term follow-up showed that the reoperation rates over a 5-year period did not significantly differ between the Rett syndrome and NMS groups. Rett syndrome is associated with increased immediate postoperative complications, necessitating tailored preoperative planning, and intensive postoperative care. Despite these challenges, the mid-term surgical outcomes are comparable to those in NMS patients.
Collapse
Affiliation(s)
- Jialun Chi
- Department of Orthopaedic Surgery, University of Virginia, Charlottesville, Virginia, USA
| | - Xiangwei Song
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Xinxiang Medical University, Weihui, Henan, China
| | - Ju Liu
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Xinxiang Medical University, Weihui, Henan, China
| | - Eunha G Oh
- Department of Orthopaedic Surgery, University of Virginia, Charlottesville, Virginia, USA
| | - Zhichang Zhang
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Xinxiang Medical University, Weihui, Henan, China
| | - Zhiwen Xu
- Department of Orthopaedic Surgery, University of Virginia, Charlottesville, Virginia, USA
| | - Hanzhi Yang
- Department of Orthopaedic Surgery, University of Virginia, Charlottesville, Virginia, USA
| | - Hui Yuan
- Department of Anesthesiology, University of California San Francisco, San Francisco, California, USA
| | - Yi Zhang
- Department of Spine Surgery, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| |
Collapse
|
47
|
Downs J, Pichard DC, Kaufmann WE, Horrigan JP, Raspa M, Townend G, Marsh ED, Leonard H, Motil K, Dietz AC, Garg N, Ananth A, Byiers B, Peters S, Beatty C, Symons F, Jacobs A, Youakim J, Suter B, Santosh P, Neul JL, Benke TA. International workshop: what is needed to ensure outcome measures for Rett syndrome are fit-for-purpose for clinical trials? June 7, 2023, Nashville, USA. Trials 2024; 25:845. [PMID: 39709426 PMCID: PMC11663341 DOI: 10.1186/s13063-024-08678-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Accepted: 12/02/2024] [Indexed: 12/23/2024] Open
Abstract
INTRODUCTION The clinical, research and advocacy communities for Rett syndrome are striving to achieve clinical trial readiness, including having fit-for-purpose clinical outcome assessments. This study aimed to (1) describe psychometric properties of clinical outcome assessment for Rett syndrome and (2) identify what is needed to ensure that fit-for-purpose clinical outcome assessments are available for clinical trials. METHODS Clinical outcome assessments for the top 10 priority domains identified in the Voice of the Patient Report for Rett syndrome were compiled and available psychometric data were extracted. The clinical outcome assessments measured clinical severity, functional abilities, comorbidities and quality of life, and electrophysiological biomarkers. An international and multidisciplinary panel of 29 experts with clinical, research, psychometric, biostatistical, industry and lived experience was identified through International Rett Syndrome Foundation networks, to discuss validation of the clinical outcome assessments, gaps and next steps, during a workshop and in a follow-up questionnaire. The identified gaps and limitations were coded using inductive content analysis. RESULTS Variable validation profiles across 26 clinical outcome assessments of clinical severity, functional abilities, and comorbidities were discussed. Reliability, validity, and responsiveness profiles were mostly incomplete; there were limited content validation data, particularly parent-informed relevance, comprehensiveness and comprehensibility of items; and no data on meaningful change or cross-cultural validity. The panel identified needs for standardised administration protocols and systematic validation programmes. CONCLUSION A pipeline of collaborative clinical outcome assessment development and validation research in Rett syndrome can now be designed, aiming to have fit-for-purpose measures that can evaluate meaningful change, to serve future clinical trials and clinical practice.
Collapse
Affiliation(s)
- Jenny Downs
- The Kids Research Institute Australia, Centre for Child Health Research, University of Western Australia, 15 Hospital Avenue, Nedlands, Perth, WA, 6009, Australia.
- Curtin School of Allied Health, Curtin University, GPO Box U1987, Perth, WA, 6845, Australia.
| | - Dominique C Pichard
- International Rett Syndrome Foundation, 4500 Cooper Road, Suite 204, Cincinnati, OH, 45242, USA
| | - Walter E Kaufmann
- Department of Human Genetics, Emory University School of Medicine, 615 Michael St, Atlanta, GA, 30322, USA
- Department of Neurology, Boston Children's Hospital, 300 Longwood Ave, Boston, MA, 02115, USA
| | - Joseph P Horrigan
- Duke Center for Autism and Brain Development, Duke University, 2608 Erwin Road, Suite 300, Durham, NC, 27705, USA
| | - Melissa Raspa
- RTI International, 3040 East Cornwallis Road, Research Triangle Park, Durham, NC, 27607, USA
| | - Gillian Townend
- School of Psychology and Clinical Language Sciences, University of Reading, Whiteknights Campus, Reading, RG6 6ES, UK
| | - Eric D Marsh
- Division of Child Neurology and University of Pennsylvania Perelman School of Medicine, Departments of Neurology and Pediatrics, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Helen Leonard
- The Kids Research Institute Australia, Centre for Child Health Research, University of Western Australia, 15 Hospital Avenue, Nedlands, Perth, WA, 6009, Australia
| | - Kathleen Motil
- USDA/ARS Children's Nutrition Research Center, Baylor College of Medicine, Houston, TX, 77030, USA
| | | | - Nupur Garg
- International Rett Syndrome Foundation, 4500 Cooper Road, Suite 204, Cincinnati, OH, 45242, USA
| | - Amitha Ananth
- University of Alabama at Birmingham, Birmingham, AL, USA
| | - Breanne Byiers
- Department of Educational Psychology, University of Minnesota, 56 E River Rd, Room 250, Minneapolis, MN, 55455, USA
| | - Sarika Peters
- Vanderbilt Kennedy Center, Vanderbilt University Medical Center, 230 Appleton Place, Nashville, TN, PMB4037204, USA
| | - Christopher Beatty
- Department of Pediatrics, Division of Neurology, Nationwide Children's Hospital and, The Ohio State University College of Medicine, 700 Children's Drive, Columbus, OH, 43205, USA
| | - Frank Symons
- Department of Educational Psychology, University of Minnesota, 56 E River Rd, Room 250, Minneapolis, MN, 55455, USA
| | - Aleksandra Jacobs
- Isabelle Rapin Division of Child Neurology, Montefiore Medical Center, Albert Einstein College of Medicine, New York, USA
| | - James Youakim
- Acadia Pharmaceuticals Inc., 502 Carnegie Center, Suite 300, Princeton, NJ, 08540, USA
| | - Bernhard Suter
- Department of Pediatrics & Neurology, Baylor College of Medicine, Houston, TX, USA
| | - Paramola Santosh
- Department of Child and Adolescent Psychiatry, Developmental Neuropsychiatry & Psychopharmacology, King's College, London, UK
- Centre for Interventional Paediatric Psychopharmacology and Rare Diseases (CIPPRD) & CIPP Rett Centre, Maudsley Hospital, London, UK
- HealthTracker Ltd, Gillingham, UK
| | - Jeffrey L Neul
- Department of Educational Psychology, University of Minnesota, 56 E River Rd, Room 250, Minneapolis, MN, 55455, USA
| | - Tim A Benke
- School of Medicine Depts of Pediatrics, Neurology and Pharmacology, Children's Hospital Colorado/University of Colorado, 12800 E 19th, MS8102, Aurora, CO, 80045, USA
| |
Collapse
|
48
|
Sloper E, Hunt M, Clarke AJ. Review of a specialist Rett syndrome clinic from 2003 to the COVID pandemic: clinic experience and carer perspectives. Orphanet J Rare Dis 2024; 19:477. [PMID: 39707431 PMCID: PMC11662725 DOI: 10.1186/s13023-024-03483-5] [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: 05/09/2024] [Accepted: 11/27/2024] [Indexed: 12/23/2024] Open
Abstract
BACKGROUND We have held a 'trouble-shooting' clinic for Rett syndrome patients from 2003 until the COVID pandemic in 2020. The clinic was multidisciplinary, including clinical genetics, paediatric neurology, adult learning disability psychiatry and physiotherapy. Access to specialist communication support and eye-gaze equipment was also often available. We have reviewed the files of patients seen in the clinic and conducted a survey of parents' and carers' satisfaction with the clinic and their experiences during COVID. RESULTS Of the 117 patients seen in the clinic, records were reviewed of 103 (97 female, six male) who attended a total of 123 appointments. The records were unavailable for 14 patients. The most common reasons for referral were assessment of 'episodes' of uncertain nature (possibly epileptic, possibly autonomic), the wish for a general review by an experienced team, and questions about the diagnosis. We discuss the nature of the advice we were able to provide and offer some brief case vignettes. We wrote to the parents or carers of all patients seen and 63 respondents were willing to be interviewed about the clinic and their experiences during COVID. Respondents were generally complimentary about the clinic team, emphasising the value of a specialist clinic for those affected by a rare condition. Respondents gave insight into the range of problems experienced during COVID, especially the isolation resulting from the withdrawal of services, demonstrating the value of community support. Some respondents mentioned the shift to remote consultations, which they hoped would continue after COVID for its convenience. However, others talked about how difficult it is in a remote consultation to explain the problems of the affected family member to professionals who do not know the patient or know about Rett syndrome. CONCLUSIONS Our findings demonstrate the value of a disease-specific clinic provided by staff experienced with the particular rare condition. Meeting the needs of patients with ultra-rare conditions presents additional challenges. We have also found that the shift to holding a virtual clinic during COVID brought the benefit of convenience but was unsatisfactory in other ways, as it makes clinical assessment more difficult and fails to overcome the sense of isolation during a pandemic.
Collapse
Affiliation(s)
- Emily Sloper
- All Wales Medical Genomics Service, Wales Genomic Health Centre, Cardiff Edge Business Park, Longwood Drive, Whitchurch, Cardiff, CF14 7YU, Wales, UK
| | - Megan Hunt
- Cardiff University School of Medicine, Institute of Medical Genetics, University Hospital of Wales, Heath Park, Cardiff, CF14 4XN, Wales, UK
| | - Angus John Clarke
- All Wales Medical Genomics Service, Wales Genomic Health Centre, Cardiff Edge Business Park, Longwood Drive, Whitchurch, Cardiff, CF14 7YU, Wales, UK.
- Cardiff University School of Medicine, Institute of Medical Genetics, University Hospital of Wales, Heath Park, Cardiff, CF14 4XN, Wales, UK.
| |
Collapse
|
49
|
Sun Z, Kou C, Gao Z, Guo X, Han B, Feng Y, Ding Q, Bai W. Association between the copy number variations of Methyl-CpG binding domain family and schizophrenia. Gene 2024; 930:148836. [PMID: 39127413 DOI: 10.1016/j.gene.2024.148836] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Revised: 07/17/2024] [Accepted: 08/07/2024] [Indexed: 08/12/2024]
Abstract
Schizophrenia is recognized as one of the most severe psychiatric disorders, with its pathogenesis likely involving genetic, epigenetic, developmental, and environmental factors. Members of the Methyl-CpG Binding Domain (MBD) Family play a crucial role in the regulation of genomic DNA methylation, and studies have implicated the association between MBD family and neurodevelopmental disorders. Copy number variations (CNVs) are a significant genetic basis for human genomic variation, also playing a critical role in the genetic processes of schizophrenia. Therefore, we aimed to evaluate the susceptibility of MBD family CNVs to schizophrenia by exploring and validating them in two separate populations using CNVplex™ and qPCR methods, and to explore the relationship between MBD family CNVs and clinical phenotypes in the overall population using chi-square tests and Fisher's exact tests. Results suggest that an increase in MBD1 gene copy number and a deficiency in MBD2 gene copy number may be associated with the risk of schizophrenia. The deficiency in MBD2 gene copy number may increase the risk of delusion of reference and delusion of persecutory in the overall sample, as well as in males. This research provides preliminary evidence supporting the association between MBD family CNVs and schizophrenia, highlighting the potential role of the MBD family in the pathogenesis of schizophrenia.
Collapse
Affiliation(s)
- Zhouyang Sun
- Department of Epidemiology and Biostatistics, School of Public Health, Jilin University, 1163 Xinmin Street, Changchun, Jilin Province, 130021, China
| | - Changgui Kou
- Department of Epidemiology and Biostatistics, School of Public Health, Jilin University, 1163 Xinmin Street, Changchun, Jilin Province, 130021, China
| | - Zibo Gao
- Department of Epidemiology and Biostatistics, School of Public Health, Jilin University, 1163 Xinmin Street, Changchun, Jilin Province, 130021, China
| | - Xinru Guo
- Department of Epidemiology and Biostatistics, School of Public Health, Jilin University, 1163 Xinmin Street, Changchun, Jilin Province, 130021, China
| | - Beibei Han
- Department of Epidemiology and Biostatistics, School of Public Health, Jilin University, 1163 Xinmin Street, Changchun, Jilin Province, 130021, China
| | - Yuan Feng
- Department of Epidemiology and Biostatistics, School of Public Health, Jilin University, 1163 Xinmin Street, Changchun, Jilin Province, 130021, China
| | - Qianlu Ding
- Department of Epidemiology and Biostatistics, School of Public Health, Jilin University, 1163 Xinmin Street, Changchun, Jilin Province, 130021, China
| | - Wei Bai
- Department of Epidemiology and Biostatistics, School of Public Health, Jilin University, 1163 Xinmin Street, Changchun, Jilin Province, 130021, China.
| |
Collapse
|
50
|
Pehlivan D, Bengtsson JD, Bajikar SS, Grochowski CM, Lun MY, Gandhi M, Jolly A, Trostle AJ, Harris HK, Suter B, Aras S, Ramocki MB, Du H, Mehaffey MG, Park K, Wilkey E, Karakas C, Eisfeldt JJ, Pettersson M, Liu L, Shinawi MS, Kimonis VE, Wiszniewski W, Mckenzie K, Roser T, Vianna-Morgante AM, Cornier AS, Abdelmoity A, Hwang JP, Jhangiani SN, Muzny DM, Mitani T, Muramatsu K, Nabatame S, Glaze DG, Fatih JM, Gibbs RA, Liu Z, Lindstrand A, Sedlazeck FJ, Lupski JR, Zoghbi HY, Carvalho CMB. Structural variant allelic heterogeneity in MECP2 duplication syndrome provides insight into clinical severity and variability of disease expression. Genome Med 2024; 16:146. [PMID: 39696717 PMCID: PMC11658439 DOI: 10.1186/s13073-024-01411-7] [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: 02/06/2024] [Accepted: 11/08/2024] [Indexed: 12/20/2024] Open
Abstract
BACKGROUND MECP2 Duplication Syndrome, also known as X-linked intellectual developmental disorder Lubs type (MRXSL; MIM: 300260), is a neurodevelopmental disorder caused by copy number gains spanning MECP2. Despite varying genomic rearrangement structures, including duplications and triplications, and a wide range of duplication sizes, no clear correlation exists between DNA rearrangement and clinical features. We had previously demonstrated that up to 38% of MRXSL families are characterized by complex genomic rearrangements (CGRs) of intermediate complexity (2 ≤ copy number variant breakpoints < 5), yet the impact of these genomic structures on regulation of gene expression and phenotypic manifestations have not been investigated. METHODS To study the role of the genomic rearrangement structures on an individual's clinical phenotypic variability, we employed a comprehensive genomics, transcriptomics, and deep phenotyping analysis approach on 137 individuals affected by MRXSL. Genomic structural information was correlated with transcriptomic and quantitative phenotypic analysis using Human Phenotype Ontology (HPO) semantic similarity scores. RESULTS Duplication sizes in the cohort ranging from 64.6 kb to 16.5 Mb were classified into four categories comprising of tandem duplications (48%), terminal duplications (22%), inverted triplications (20%), and other CGRs (10%). Most of the terminal duplication structures consist of translocations (65%) followed by recombinant chromosomes (23%). Notably, 65% of de novo events occurred in the Terminal duplication group in contrast with 17% observed in Tandem duplications. RNA-seq data from lymphoblastoid cell lines indicated that the MECP2 transcript quantity in MECP2 triplications is statistically different from all duplications, but not between other classes of genomic structures. We also observed a significant (p < 0.05) correlation (Pearson R = 0.6, Spearman p = 0.63) between the log-transformed MECP2 RNA levels and MECP2 protein levels, demonstrating that genomic aberrations spanning MECP2 lead to altered MECP2 RNA and MECP2 protein levels. Genotype-phenotype analyses indicated a gradual worsening of phenotypic features, including overall survival, developmental levels, microcephaly, epilepsy, and genitourinary/eye abnormalities in the following order: Tandem duplications, Other complex duplications, Terminal duplications/Translocations, and Triplications encompassing MECP2. CONCLUSION In aggregate, this combined analysis uncovers an interplay between MECP2 dosage, genomic rearrangement structure and phenotypic traits. Whereas the level of MECP2 is a key determinant of the phenotype, the DNA rearrangement structure can contribute to clinical severity and disease expression variability. Employing this type of analytical approach will advance our understanding of the impact of genomic rearrangements on genomic disorders and may help guide more targeted therapeutic approaches.
Collapse
Affiliation(s)
- Davut Pehlivan
- Department of Pediatrics, Section of Neurology and Developmental Neuroscience, Baylor College of Medicine, Houston, TX, 77030, USA.
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, 77030, USA.
- Department of Pediatrics, Baylor College of Medicine, Houston, TX, 77030, USA.
- Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital, Houston, TX, 77030, USA.
| | | | - Sameer S Bajikar
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, 77030, USA
- Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital, Houston, TX, 77030, USA
| | - Christopher M Grochowski
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, 77030, USA
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Ming Yin Lun
- Pacific Northwest Research Institute, Seattle, WA, 98122, USA
| | - Mira Gandhi
- Pacific Northwest Research Institute, Seattle, WA, 98122, USA
| | - Angad Jolly
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Alexander J Trostle
- Department of Pediatrics, Baylor College of Medicine, Houston, TX, 77030, USA
- Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital, Houston, TX, 77030, USA
| | - Holly K Harris
- Department of Pediatrics, Baylor College of Medicine, Houston, TX, 77030, USA
- The Meyer Center for Developmental Pediatrics and Autism, 8080 North Stadium Drive, Houston, TX, 77054, USA
| | - Bernhard Suter
- Department of Pediatrics, Section of Neurology and Developmental Neuroscience, Baylor College of Medicine, Houston, TX, 77030, USA
- Department of Pediatrics, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Sukru Aras
- Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital, Houston, TX, 77030, USA
| | - Melissa B Ramocki
- Department of Pediatrics, Section of Neurology and Developmental Neuroscience, Baylor College of Medicine, Houston, TX, 77030, USA
- University Otolaryngology, East Greenwich, RI, 02818, USA
| | - Haowei Du
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, 77030, USA
| | | | - KyungHee Park
- Pacific Northwest Research Institute, Seattle, WA, 98122, USA
| | - Ellen Wilkey
- Pacific Northwest Research Institute, Seattle, WA, 98122, USA
| | - Cemal Karakas
- Department of Pediatrics, Division of Neurology, University of Louisville, Louisville, KY, 40202, USA
| | - Jesper J Eisfeldt
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
| | - Maria Pettersson
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
| | - Lynn Liu
- Department of Neurology, Division of Epilepsy, University of North Carolina, Chapel Hill, NC, 27599, USA
| | - Marwan S Shinawi
- Department of Pediatrics, Division of Genetics and Genomic Medicine, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Virginia E Kimonis
- Department of Pediatrics, Division of Genetics and Genomic Medicine, University of California, Irvine, CA, 92697, USA
| | - Wojciech Wiszniewski
- Department of Molecular and Medical Genetics, Oregon Health and Science University, Portland, OR, 97239, USA
| | - Kyle Mckenzie
- Department of Pediatrics, Division of General and Community Pediatrics, University of Alberta, Edmonton, AB, T6G 2R7, Canada
| | - Timo Roser
- Department of Pediatrics, Division of Pediatric Neurology, Developmental Medicine and Social Pediatrics, Dr. Von Haunersches Children's Hospital, Ludwig Maximilian University of Munich, Munich, 80337, Germany
| | - Angela M Vianna-Morgante
- Department of Genetics and Evolutionary Biology, Institute of Biosciences, São Paulo - SP, 05508-090, Brazil
| | - Alberto S Cornier
- Department of Genetics, San Jorge Children's Hospital, San Juan, 00771, Puerto Rico
| | - Ahmed Abdelmoity
- Division of Neurology, Department of Pediatrics, Children's Mercy Kansas City, Kansas City, MO, 64108, USA
| | - James P Hwang
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Shalini N Jhangiani
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Donna M Muzny
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Tadahiro Mitani
- Department of Pediatrics, Jichi Medical University, Shimotsuke-City, Tochigi, 329-0498, Japan
| | - Kazuhiro Muramatsu
- Department of Pediatrics, Jichi Medical University, Shimotsuke-City, Tochigi, 329-0498, Japan
| | - Shin Nabatame
- Department of Pediatrics, Osaka University Graduate School of Medicine, Osaka, 565-0871, Japan
| | - Daniel G Glaze
- Department of Pediatrics, Section of Neurology and Developmental Neuroscience, Baylor College of Medicine, Houston, TX, 77030, USA
- Department of Pediatrics, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Jawid M Fatih
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Richard A Gibbs
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, 77030, USA
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Zhandong Liu
- Department of Pediatrics, Baylor College of Medicine, Houston, TX, 77030, USA
- Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital, Houston, TX, 77030, USA
| | - Anna Lindstrand
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
| | - Fritz J Sedlazeck
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX, 77030, USA
| | - James R Lupski
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, 77030, USA
- Department of Pediatrics, Baylor College of Medicine, Houston, TX, 77030, USA
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX, 77030, USA
- Texas Children's Hospital, Houston, TX, 77030, USA
| | - Huda Y Zoghbi
- Department of Pediatrics, Section of Neurology and Developmental Neuroscience, Baylor College of Medicine, Houston, TX, 77030, USA.
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, 77030, USA.
- Department of Pediatrics, Baylor College of Medicine, Houston, TX, 77030, USA.
- Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital, Houston, TX, 77030, USA.
- Howard Hughes Medical Institute and Jan and Dan Duncan Neurological Research Institute, Houston, TX, 77030, USA.
| | - Claudia M B Carvalho
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, 77030, USA.
- Pacific Northwest Research Institute, Seattle, WA, 98122, USA.
| |
Collapse
|