1
|
Wang L, Hu Z, Bai H, Chang L, Chen C, Li W. miRNA-105-5p regulates the histone deacetylase HDAC2 through FOXG1 to affect the malignant biological behavior of triple-negative breast cancer cells. Am J Med Sci 2025; 369:513-523. [PMID: 39313115 DOI: 10.1016/j.amjms.2024.09.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Revised: 09/19/2024] [Accepted: 09/20/2024] [Indexed: 09/25/2024]
Abstract
BACKGROUND Triple-negative breast cancer (TNBC) is a specific subtype of breast cancer (BC). Some potential molecular targets have been identified, and miR-105-5p was found to be abnormally expressed in TNBC tissues. OBJECTIVE The objective of this study was to probe the effect of miR-105-5p on TNBC via FOXG1/HDAC2-mediated acetylation. METHODS An animal model of TNBC was established by injecting BC cells into the axillary area of nude mice. The levels of miR-105-5p, FOXG1, HDAC2, Bcl-2, Bax, and Ki67 were detected via RT‒qPCR, Western blotting and immunohistochemistry. Flow cytometry, CCK-8, Transwell and colony formation assays were used to measure apoptosis, proliferation and migration, respectively. Total histone acetylation levels were measured by ELISA. The binding of FOXG1 to HDAC2 was detected by co-immunoprecipitation. The binding relationship between miR-105-5p and FOXG1 was verified using a dual-luciferase reporter gene assay. RESULTS In this study, miR-105-5p and HDAC2 were highly expressed in the MDA-MB-231 and BT-549 BC cell lines, whereas FOXG1 was expressed at low levels. The inhibition of miR-105-5p inhibited the proliferation and migration of MDA-MB-231 and BT-549 cells and promoted their apoptosis. Bioinformatics analysis revealed that miR-105-5p and FOXG1 had a negative targeting regulatory relationship. FOXG1 overexpression had a similar effect on cancer cells as the inhibition of miR-105-5p. Moreover, experiments revealed that FOXG1 and HDAC2 could bind to each other and that HDAC2 overexpression or treatment with the histone acetyltransferase inhibitor Garcinol weakened the effect of FOXG1 overexpression. In addition, FOXG1 knockdown inhibited the effect of the miR-105-5p inhibitor, while Garcinol treatment further enhanced the effect of FOXG1 knockdown, inhibited histone acetylation, promoted the proliferation and migration of cancer cells, and inhibited apoptosis. Moreover, the in vivo results confirmed the in vitro results. CONCLUSION miR-105-5p promotes HDAC2 expression by reducing FOXG1, inhibits histone acetylation, and aggravates the malignant biological behavior of TNBC cells.
Collapse
Affiliation(s)
- Li Wang
- Department of Radiotherapy, Third Affiliated Hospital of Kunming Medical University (Yunnan Cancer Hospital, Yunnan Cancer Center), Kunming, 650000, Yunnan, China
| | - Zaoxiu Hu
- Department of Pathology, Third Affiliated Hospital of Kunming Medical University (Yunnan Cancer Hospital, Yunnan Cancer Center), Kunming, 650000, Yunnan, China
| | - Han Bai
- Department of Radiotherapy, Third Affiliated Hospital of Kunming Medical University (Yunnan Cancer Hospital, Yunnan Cancer Center), Kunming, 650000, Yunnan, China
| | - Li Chang
- Department of Radiotherapy, Third Affiliated Hospital of Kunming Medical University (Yunnan Cancer Hospital, Yunnan Cancer Center), Kunming, 650000, Yunnan, China
| | - Ceshi Chen
- Third Affiliated Hospital of Kunming Medical University (Yunnan Cancer Hospital, Yunnan Cancer Center), Kunming, 650000, Yunnan, China.
| | - Wenhui Li
- Department of Radiotherapy, Third Affiliated Hospital of Kunming Medical University (Yunnan Cancer Hospital, Yunnan Cancer Center), Kunming, 650000, Yunnan, China.
| |
Collapse
|
2
|
Jeon S, Park J, Moon JH, Shin D, Li L, O'Shea H, Hwang SU, Lee HJ, Brimble E, Lee JW, Clark S, Lee SK. The patient-specific mouse model with Foxg1 frameshift mutation provides insights into the pathophysiology of FOXG1 syndrome. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.21.634140. [PMID: 39896554 PMCID: PMC11785084 DOI: 10.1101/2025.01.21.634140] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/04/2025]
Abstract
Single allelic mutations in the forebrain-specific transcription factor gene FOXG1 lead to FOXG1 syndrome (FS). To decipher the disease mechanisms of FS, which vary depending on FOXG1 mutation types, patient-specific animal models are critical. Here, we report the first patient-specific FS mouse model, Q84Pfs heterozygous (Q84Pfs-Het) mice, which emulates one of the most predominant FS variants. Remarkably, Q84Pfs-Het mice recapitulate various human FS phenotypes across cellular, brain structural, and behavioral levels, such as microcephaly, corpus callosum agenesis, movement disorders, repetitive behaviors, and anxiety. Q84Pfs-Het cortex showed dysregulations of genes controlling cell proliferation, neuronal projection and migration, synaptic assembly, and synaptic vesicle transport. Interestingly, the FS-causing Q84Pfs allele produced the N-terminal fragment of FOXG1, denoted as Q84Pfs protein, in Q84Pfs-Het mouse brains. Q84Pfs fragment forms intracellular speckles, interacts with FOXG1 full-length protein, and triggers the sequestration of FOXG1 to distinct subcellular domains. Q84Pfs protein also promotes the radial glial cell identity and suppresses neuronal migration in the cortex. Together, our study uncovered the role of the FOXG1 fragment derived from FS-causing FOXG1 variants and identified the genes involved in FS-like cellular and behavioral phenotypes, providing essential insights into the pathophysiology of FS.
Collapse
|
3
|
Narita A, Asano H, Kudo H, Miyata S, Shutoh F, Miyoshi G. A novel quadrant spatial assay reveals environmental preference in mouse spontaneous and parental behaviors. Neurosci Res 2024; 209:18-27. [PMID: 39134225 DOI: 10.1016/j.neures.2024.08.002] [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: 08/01/2024] [Accepted: 08/05/2024] [Indexed: 08/27/2024]
Abstract
Environmental factors have well-documented impacts on brain development and mental health. Therefore, it is crucial to employ a reliable assay system to assess the spatial preference of model animals. In this study, we introduced an unbiased quadrant chamber assay system and discovered that parental pup-gathering behavior takes place in a very efficient manner. Furthermore, we found that test mice exhibited preferences for specific environments in both spontaneous and parental pup-gathering behavior contexts. Notably, the spatial preferences of autism spectrum disorder model animals were initially suppressed but later equalized during the spontaneous behavior assay, accompanied by increased time spent in the preferred chamber. In conclusion, our novel quadrant chamber assay system provides an ideal platform for investigating the spatial preference of mice, offering potential applications in studying environmental impacts and exploring neurodevelopmental and psychiatric disorder models.
Collapse
Affiliation(s)
- Aito Narita
- Department of Developmental Genetics and Behavioral Neuroscience, Gunma University Graduate School of Medicine, 3-39-22 Showa-machi, Maebashi city, Gunma 371-8511, Japan
| | - Hirofumi Asano
- Department of Developmental Genetics and Behavioral Neuroscience, Gunma University Graduate School of Medicine, 3-39-22 Showa-machi, Maebashi city, Gunma 371-8511, Japan
| | - Hayato Kudo
- Department of Developmental Genetics and Behavioral Neuroscience, Gunma University Graduate School of Medicine, 3-39-22 Showa-machi, Maebashi city, Gunma 371-8511, Japan
| | - Shigeo Miyata
- Department of Developmental Genetics and Behavioral Neuroscience, Gunma University Graduate School of Medicine, 3-39-22 Showa-machi, Maebashi city, Gunma 371-8511, Japan
| | - Fumihiro Shutoh
- Division of Informatics, Bioengineering and Bioscience, Maebashi Institute of Technology, 460-1 Kamisadori-machi, Maebashi city, Gunma 371-0816, Japan
| | - Goichi Miyoshi
- Department of Developmental Genetics and Behavioral Neuroscience, Gunma University Graduate School of Medicine, 3-39-22 Showa-machi, Maebashi city, Gunma 371-8511, Japan.
| |
Collapse
|
4
|
Taufer NP, Santos-Souza C, Larentis LT, Santos CND, Creuzet SE, Garcez RC. Integrative analysis of molecular pathways and morphological anomalies associated with congenital Zika syndrome. J Neurol Sci 2024; 465:123190. [PMID: 39182423 DOI: 10.1016/j.jns.2024.123190] [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: 06/05/2024] [Revised: 08/09/2024] [Accepted: 08/18/2024] [Indexed: 08/27/2024]
Abstract
Congenital Zika syndrome (CZS) comprises a set of clinical manifestations that can be presented by neonates born to mothers infected by the Zika virus (ZIKV). CZS-associated phenotypes include neurological, skeletal, and systemic alterations and long-term developmental sequelae. One of the most frequently reported clinical conditions is microcephaly characterized by a reduction in head circumference and cognitive complications. Nevertheless, the associations among the diverse signaling pathways underlying CZS phenotypes remain to be elucidated. To shed light on CZS, we have extensively reviewed the morphological anomalies resulting from ZIKV infection, as well as genes and proteins of interest obtained from the published literature. With this list of genes or proteins, we performed computational analyses to explore the cellular processes, molecular mechanisms, and molecular pathways related to ZIKV infection. Therefore, in this review, we comprehensively describe the morphological abnormalities caused by congenital ZIKV infection and, through the analysis noted above, propose common molecular pathways altered by ZIKV that could explain both central nervous system and craniofacial skeletal alterations.
Collapse
Affiliation(s)
- Nathali Parise Taufer
- Graduate Program in Cell and Developmental Biology, Universidade Federal de Santa Catarina, Florianópolis, SC, Brazil; Laboratório de Células-Tronco e Regeneração Tecidual (LACERT), Department of Cell Biology, Embryology, and Genetics, Universidade Federal de Santa Catarina, Florianópolis, SC, Brazil
| | - Camila Santos-Souza
- Graduate Program in Cell and Developmental Biology, Universidade Federal de Santa Catarina, Florianópolis, SC, Brazil; Laboratório de Células-Tronco e Regeneração Tecidual (LACERT), Department of Cell Biology, Embryology, and Genetics, Universidade Federal de Santa Catarina, Florianópolis, SC, Brazil
| | - Lucas Trentin Larentis
- Graduate Program in Cell and Developmental Biology, Universidade Federal de Santa Catarina, Florianópolis, SC, Brazil; Laboratório de Células-Tronco e Regeneração Tecidual (LACERT), Department of Cell Biology, Embryology, and Genetics, Universidade Federal de Santa Catarina, Florianópolis, SC, Brazil
| | | | - Sophie Emmanuelle Creuzet
- Institut des Neurosciences Paris-Saclay (NeuroPSI), Paris-Saclay University, Centre National de la Recherche Scientifique UMR 9197, Saclay, France.
| | - Ricardo Castilho Garcez
- Graduate Program in Cell and Developmental Biology, Universidade Federal de Santa Catarina, Florianópolis, SC, Brazil; Laboratório de Células-Tronco e Regeneração Tecidual (LACERT), Department of Cell Biology, Embryology, and Genetics, Universidade Federal de Santa Catarina, Florianópolis, SC, Brazil.
| |
Collapse
|
5
|
Bell JM, Turner EM, Biesemeyer C, Vanderbeck MM, Hendricks R, McGraw HF. foxg1a is required for hair cell development and regeneration in the zebrafish lateral line. Biol Open 2024; 13:bio060580. [PMID: 39301848 PMCID: PMC11423914 DOI: 10.1242/bio.060580] [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: 08/19/2024] [Accepted: 08/21/2024] [Indexed: 09/22/2024] Open
Abstract
Mechanosensory hair cells located in the inner ear mediate the sensations of hearing and balance. If damaged, mammalian inner ear hair cells are unable to regenerate, resulting in permanent sensory deficits. Aquatic vertebrates like zebrafish (Danio rerio) have a specialized class of mechanosensory hair cells found in the lateral line system, allowing them to sense changes in water current. Unlike mammalian inner ear hair cells, lateral line hair cells can robustly regenerate following damage. In mammals, the transcription factor Foxg1 functions to promote normal development of the inner ear. Foxg1a is expressed in lateral line sensory organs in zebrafish larvae, but its function during lateral line development and regeneration has not been investigated. Our study demonstrates that mutation of foxg1a results in slower posterior lateral line primordium migration and delayed neuromast formation. In developing and regenerating neuromasts, we find that loss of Foxg1a function results in reduced hair cell numbers, as well as decreased proliferation of neuromast cells. Foxg1a specifically regulates the development and regeneration of Islet1-labeled hair cells. These data suggest that Foxg1 may be a valuable target for investigation of clinical hair cell regeneration.
Collapse
Affiliation(s)
- Jon M. Bell
- University of Missouri Kansas City, School of Science and Engineering, Division of Biological and Biomedical Systems, Kansas City, MO 64110, USA
| | - Emily M. Turner
- University of Missouri Kansas City, School of Science and Engineering, Division of Biological and Biomedical Systems, Kansas City, MO 64110, USA
| | - Cole Biesemeyer
- University of Missouri Kansas City, School of Science and Engineering, Division of Biological and Biomedical Systems, Kansas City, MO 64110, USA
- Research Organisms, Stowers Institute for Medical Research, Kansas City, MO 64110, USA
| | - Madison M. Vanderbeck
- University of Missouri Kansas City, School of Science and Engineering, Division of Biological and Biomedical Systems, Kansas City, MO 64110, USA
| | - Roe Hendricks
- University of Missouri Kansas City, School of Science and Engineering, Division of Biological and Biomedical Systems, Kansas City, MO 64110, USA
| | - Hillary F. McGraw
- University of Missouri Kansas City, School of Science and Engineering, Division of Biological and Biomedical Systems, Kansas City, MO 64110, USA
| |
Collapse
|
6
|
Mazel B, Delanne J, Garde A, Racine C, Bruel AL, Duffourd Y, Lopergolo D, Santorelli FM, Marchi V, Pinto AM, Mencarelli MA, Canitano R, Valentino F, Papa FT, Fallerini C, Mari F, Renieri A, Munnich A, Niclass T, Le Guyader G, Thauvin-Robinet C, Philippe C, Faivre L. FOXG1 variants can be associated with milder phenotypes than congenital Rett syndrome with unassisted walking and language development. Am J Med Genet B Neuropsychiatr Genet 2024; 195:e32970. [PMID: 38459409 DOI: 10.1002/ajmg.b.32970] [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: 09/07/2023] [Revised: 01/22/2024] [Accepted: 01/30/2024] [Indexed: 03/10/2024]
Abstract
Since 2008, FOXG1 haploinsufficiency has been linked to a severe neurodevelopmental phenotype resembling Rett syndrome but with earlier onset. Most patients are unable to sit, walk, or speak. For years, FOXG1 sequencing was only prescribed in such severe cases, limiting insight into the full clinical spectrum associated with this gene. Next-generation sequencing (NGS) now enables unbiased diagnostics. Through the European Reference Network for Rare Malformation Syndromes, Intellectual and Other Neurodevelopmental Disorders, we gathered data from patients with heterozygous FOXG1 variants presenting a mild phenotype, defined as able to speak and walk independently. We also reviewed data from three previously reported patients meeting our criteria. We identified five new patients with pathogenic FOXG1 missense variants, primarily in the forkhead domain, showing varying nonspecific intellectual disability and developmental delay. These features are not typical of congenital Rett syndrome and were rarely associated with microcephaly and epilepsy. Our findings are consistent with a previous genotype-phenotype analysis by Mitter et al. suggesting the delineation of five different FOXG1 genotype groups. Milder phenotypes were associated with missense variants in the forkhead domain. This information may facilitate prognostic assessments in children carrying a FOXG1 variant and improve the interpretation of new variants identified with genomic sequencing.
Collapse
Affiliation(s)
- Benoit Mazel
- Centre de Référence Anomalies du Développement et Syndromes Malformatifs, Centre de Génétique, FHU TRANSLAD - CHU Dijon Bourgogne, Dijon, France
- Inserm UMR1231 GAD, Génétique des Anomalies du Développement, Université de Bourgogne, Dijon, France
| | - Julian Delanne
- Centre de Référence Anomalies du Développement et Syndromes Malformatifs, Centre de Génétique, FHU TRANSLAD - CHU Dijon Bourgogne, Dijon, France
- Centre de référence Déficiences Intellectuelles de Causes Rares, CHU Dijon Bourgogne, Dijon, France
| | - Aurore Garde
- Centre de Référence Anomalies du Développement et Syndromes Malformatifs, Centre de Génétique, FHU TRANSLAD - CHU Dijon Bourgogne, Dijon, France
- Inserm UMR1231 GAD, Génétique des Anomalies du Développement, Université de Bourgogne, Dijon, France
| | - Caroline Racine
- Centre de Référence Anomalies du Développement et Syndromes Malformatifs, Centre de Génétique, FHU TRANSLAD - CHU Dijon Bourgogne, Dijon, France
- Inserm UMR1231 GAD, Génétique des Anomalies du Développement, Université de Bourgogne, Dijon, France
| | - Ange-Line Bruel
- Inserm UMR1231 GAD, Génétique des Anomalies du Développement, Université de Bourgogne, Dijon, France
- Laboratoire de Génomique Médicale, Unité Fonctionnelle Innovation en diagnostic génomique, Unité fonctionnelle innovation en diagnostic génomique des maladies rares, CHU Dijon Bourgogne, Dijon, France
| | - Yannis Duffourd
- Inserm UMR1231 GAD, Génétique des Anomalies du Développement, Université de Bourgogne, Dijon, France
- Laboratoire de Génomique Médicale, Unité Fonctionnelle Innovation en diagnostic génomique, Unité fonctionnelle innovation en diagnostic génomique des maladies rares, CHU Dijon Bourgogne, Dijon, France
| | - Diego Lopergolo
- Molecular Medicine for Neurodegenerative and Neuromuscular Diseases Unit, IRCCS Stella Maris Foudation, Pisa, Italy
| | - Filippo Maria Santorelli
- Molecular Medicine for Neurodegenerative and Neuromuscular Diseases Unit, IRCCS Stella Maris Foudation, Pisa, Italy
| | - Viviana Marchi
- Department of Developmental Neuroscience, Stella Maris Scientific Institute, IRCCS Fondazione Stella Maris Foundation, Pisa, Italy
| | - Anna Maria Pinto
- Genetica Medica, Azienda Ospedaliera Universitaria Senese, Siena, Italy
| | | | - Roberto Canitano
- Division of Child and Adolescent Neuropsychiatry, University Hospital of Siena, Siena, Italy
| | - Floriana Valentino
- Medical Genetics Unit, University of Siena, Policlinico Le Scotte, Siena, Italy
| | | | - Chiara Fallerini
- Medical Genetics Unit, University of Siena, Policlinico Le Scotte, Siena, Italy
- Department of Medical Biotechnologies, Med Biotech Hub and Competence Center, University of Siena, Siena, Italy
| | - Francesca Mari
- Genetica Medica, Azienda Ospedaliera Universitaria Senese, Siena, Italy
- Medical Genetics Unit, University of Siena, Policlinico Le Scotte, Siena, Italy
| | - Alessandra Renieri
- Genetica Medica, Azienda Ospedaliera Universitaria Senese, Siena, Italy
- Medical Genetics Unit, University of Siena, Policlinico Le Scotte, Siena, Italy
- Department of Medical Biotechnologies, Med Biotech Hub and Competence Center, University of Siena, Siena, Italy
| | - Arnold Munnich
- Service de Génétique Médicale et Clinique, Hôpital Necker Enfants Malades, Paris, France
| | - Tanguy Niclass
- Service de Génétique Clinique, CHU de Poitiers, Poitiers, France
| | | | - Christel Thauvin-Robinet
- Centre de Référence Anomalies du Développement et Syndromes Malformatifs, Centre de Génétique, FHU TRANSLAD - CHU Dijon Bourgogne, Dijon, France
- Inserm UMR1231 GAD, Génétique des Anomalies du Développement, Université de Bourgogne, Dijon, France
- Centre de référence Déficiences Intellectuelles de Causes Rares, CHU Dijon Bourgogne, Dijon, France
- Laboratoire de Génomique Médicale, Unité Fonctionnelle Innovation en diagnostic génomique, Unité fonctionnelle innovation en diagnostic génomique des maladies rares, CHU Dijon Bourgogne, Dijon, France
| | - Christophe Philippe
- Inserm UMR1231 GAD, Génétique des Anomalies du Développement, Université de Bourgogne, Dijon, France
- Laboratoire de Génomique Médicale, Unité Fonctionnelle Innovation en diagnostic génomique, Unité fonctionnelle innovation en diagnostic génomique des maladies rares, CHU Dijon Bourgogne, Dijon, France
| | - Laurence Faivre
- Centre de Référence Anomalies du Développement et Syndromes Malformatifs, Centre de Génétique, FHU TRANSLAD - CHU Dijon Bourgogne, Dijon, France
- Inserm UMR1231 GAD, Génétique des Anomalies du Développement, Université de Bourgogne, Dijon, France
| |
Collapse
|
7
|
Artimagnella O, Maftei ES, Esposito M, Sanges R, Mallamaci A. Foxg1 regulates translation of neocortical neuronal genes, including the main NMDA receptor subunit gene, Grin1. BMC Biol 2024; 22:180. [PMID: 39183266 PMCID: PMC11346056 DOI: 10.1186/s12915-024-01979-x] [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: 08/04/2023] [Accepted: 08/12/2024] [Indexed: 08/27/2024] Open
Abstract
BACKGROUND Mainly known as a transcription factor patterning the rostral brain and governing its histogenesis, FOXG1 has been also detected outside the nucleus; however, biological meaning of that has been only partially clarified. RESULTS Prompted by FOXG1 expression in cytoplasm of pallial neurons, we investigated its implication in translational control. We documented the impact of FOXG1 on ribosomal recruitment of Grin1-mRNA, encoding for the main subunit of NMDA receptor. Next, we showed that FOXG1 increases GRIN1 protein level by enhancing the translation of its mRNA, while not increasing its stability. Molecular mechanisms underlying this activity included FOXG1 interaction with EIF4E and, possibly, Grin1-mRNA. Besides, we found that, within murine neocortical cultures, de novo synthesis of GRIN1 undergoes a prominent and reversible, homeostatic regulation and FOXG1 is instrumental to that. Finally, by integrated analysis of multiple omic data, we inferred that FOXG1 is implicated in translational control of hundreds of neuronal genes, modulating ribosome engagement and progression. In a few selected cases, we experimentally verified such inference. CONCLUSIONS These findings point to FOXG1 as a key effector, potentially crucial to multi-scale temporal tuning of neocortical pyramid activity, an issue with profound physiological and neuropathological implications.
Collapse
Affiliation(s)
- Osvaldo Artimagnella
- Laboratory of Cerebral Cortex Development, SISSA, Via Bonomea 265, 34136, Trieste, Italy
- Present Address: IRCCS Centro Neurolesi "Bonino-Pulejo", Messina, Italy
| | - Elena Sabina Maftei
- Laboratory of Cerebral Cortex Development, SISSA, Via Bonomea 265, 34136, Trieste, Italy
| | - Mauro Esposito
- Laboratory of Computational Genomics, SISSA, via Bonomea 265, 34136, Trieste, Italy
- Present Address: Neomatrix SRL, Rome, Italy
| | - Remo Sanges
- Laboratory of Computational Genomics, SISSA, via Bonomea 265, 34136, Trieste, Italy
| | - Antonello Mallamaci
- Laboratory of Cerebral Cortex Development, SISSA, Via Bonomea 265, 34136, Trieste, Italy.
| |
Collapse
|
8
|
Fuchs C, ‘t Hoen PAC, Müller AR, Ehrhart F, Van Karnebeek CDM. Drug repurposing in Rett and Rett-like syndromes: a promising yet underrated opportunity? Front Med (Lausanne) 2024; 11:1425038. [PMID: 39135718 PMCID: PMC11317438 DOI: 10.3389/fmed.2024.1425038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Accepted: 07/12/2024] [Indexed: 08/15/2024] Open
Abstract
Rett syndrome (RTT) and Rett-like syndromes [i.e., CDKL5 deficiency disorder (CDD) and FOXG1-syndrome] represent rare yet profoundly impactful neurodevelopmental disorders (NDDs). The severity and complexity of symptoms associated with these disorders, including cognitive impairment, motor dysfunction, seizures and other neurological features significantly affect the quality of life of patients and families. Despite ongoing research efforts to identify potential therapeutic targets and develop novel treatments, current therapeutic options remain limited. Here the potential of drug repurposing (DR) as a promising avenue for addressing the unmet medical needs of individuals with RTT and related disorders is explored. Leveraging existing drugs for new therapeutic purposes, DR presents an attractive strategy, particularly suited for neurological disorders given the complexities of the central nervous system (CNS) and the challenges in blood-brain barrier penetration. The current landscape of DR efforts in these syndromes is thoroughly examined, with partiuclar focus on shared molecular pathways and potential common drug targets across these conditions.
Collapse
Affiliation(s)
| | - Peter A. C. ‘t Hoen
- Center for Molecular and Biomolecular Informatics, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, Netherlands
| | - Annelieke R. Müller
- Department of Pediatrics, Emma Children’s Hospital, Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam UMC Location University of Amsterdam, Amsterdam, Netherlands
- Amsterdam UMC, Emma Center for Personalized Medicine, Amsterdam, Netherlands
- Department of Human Genetics, Amsterdam Reproduction and Development, Amsterdam UMC Location University of Amsterdam, Amsterdam, Netherlands
| | - Friederike Ehrhart
- Department of Bioinformatics – BiGCaT, Research Institute of Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University, Maastricht, Netherlands
| | - Clara D. M. Van Karnebeek
- Department of Pediatrics, Emma Children’s Hospital, Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam UMC Location University of Amsterdam, Amsterdam, Netherlands
- Amsterdam UMC, Emma Center for Personalized Medicine, Amsterdam, Netherlands
- Department of Human Genetics, Amsterdam Reproduction and Development, Amsterdam UMC Location University of Amsterdam, Amsterdam, Netherlands
| |
Collapse
|
9
|
Aji A, Zhang C, Liu W, Chen T, Liu Z, Zuo J, Li H, Mi W, Mao-Ying QL, Wang Y, Zhao Q, Chu YX. Foxg1 Modulation of the Prkcd Gene in the Lateral Habenula Mediates Trigeminal Neuralgia-Associated Anxiety-Like Behaviors in Mice. Mol Neurobiol 2024; 61:4335-4351. [PMID: 38085455 DOI: 10.1007/s12035-023-03856-0] [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] [Received: 04/20/2023] [Accepted: 11/28/2023] [Indexed: 07/11/2024]
Abstract
Trigeminal Neuralgia (TN) is a debilitating disorder frequently accompanied by mood complications such as depression and anxiety. The current study sought to elucidate the molecular underpinnings that contribute to the pathogenesis of TN and its associated anxiety. Employing a partial transection of the infraorbital nerve (pT-ION) in a murine model, we successfully induced sustained primary and secondary orofacial allodynia alongside anxiety-like behavioral manifestations. Transcriptome-wide gene microarray analyses revealed a marked upregulation of Foxg1 subsequent to pT-ION. Targeted knockdown of Foxg1, achieved through bilateral microinjection of adeno-associated virus harboring Foxg1-specific shRNA into the lateral habenula (LHb), resulted in a significant attenuation of both orofacial pain and anxiety-like behaviors. Subsequent RNA sequencing implicated Prkcd as a downstream effector gene modulated by Foxg1. Pharmacological inhibition of protein kinase C delta, encoded by Prkcd, within the LHb markedly ameliorated pT-ION-induced symptomatology. The dual luciferase assay revealed that Foxg1 substantially enhances the transcriptional activity of the Prkcd gene. Collectively, these findings indicate that trigeminal nerve injury leads to Foxg1 upregulation in the LHb, which in turn elevates the expression of Prkcd, culminating in the manifestation of orofacial pain and anxiety-like behaviors. This work offers promising therapeutic targets and a conceptual framework for the clinical management of TN and its psychological comorbidities.
Collapse
Affiliation(s)
- Abudula Aji
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Sciences, Shanghai Medical College, Institutes of Integrative Medicine, Shanghai Key Laboratory of Acupuncture Mechanism and Acupoint Function, Institute of Acupuncture Research, Fudan University, Shanghai, China
| | - Chen Zhang
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Sciences, Shanghai Medical College, Institutes of Integrative Medicine, Shanghai Key Laboratory of Acupuncture Mechanism and Acupoint Function, Institute of Acupuncture Research, Fudan University, Shanghai, China
| | - Wenbo Liu
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Sciences, Shanghai Medical College, Institutes of Integrative Medicine, Shanghai Key Laboratory of Acupuncture Mechanism and Acupoint Function, Institute of Acupuncture Research, Fudan University, Shanghai, China
| | - Teng Chen
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Sciences, Shanghai Medical College, Institutes of Integrative Medicine, Shanghai Key Laboratory of Acupuncture Mechanism and Acupoint Function, Institute of Acupuncture Research, Fudan University, Shanghai, China
| | - Zhechen Liu
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Sciences, Shanghai Medical College, Institutes of Integrative Medicine, Shanghai Key Laboratory of Acupuncture Mechanism and Acupoint Function, Institute of Acupuncture Research, Fudan University, Shanghai, China
| | - Jiaxin Zuo
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Sciences, Shanghai Medical College, Institutes of Integrative Medicine, Shanghai Key Laboratory of Acupuncture Mechanism and Acupoint Function, Institute of Acupuncture Research, Fudan University, Shanghai, China
| | - Haojun Li
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Sciences, Shanghai Medical College, Institutes of Integrative Medicine, Shanghai Key Laboratory of Acupuncture Mechanism and Acupoint Function, Institute of Acupuncture Research, Fudan University, Shanghai, China
| | - Wenli Mi
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Sciences, Shanghai Medical College, Institutes of Integrative Medicine, Shanghai Key Laboratory of Acupuncture Mechanism and Acupoint Function, Institute of Acupuncture Research, Fudan University, Shanghai, China
| | - Qi-Liang Mao-Ying
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Sciences, Shanghai Medical College, Institutes of Integrative Medicine, Shanghai Key Laboratory of Acupuncture Mechanism and Acupoint Function, Institute of Acupuncture Research, Fudan University, Shanghai, China
| | - Yanqing Wang
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Sciences, Shanghai Medical College, Institutes of Integrative Medicine, Shanghai Key Laboratory of Acupuncture Mechanism and Acupoint Function, Institute of Acupuncture Research, Fudan University, Shanghai, China
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, China
| | - Qing Zhao
- Shanghai Sunshine Rehabilitation Center, Shanghai Yangzhi Rehabilitation Hospital, Tongji University School of Medicine, Shanghai, China.
| | - Yu-Xia Chu
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Sciences, Shanghai Medical College, Institutes of Integrative Medicine, Shanghai Key Laboratory of Acupuncture Mechanism and Acupoint Function, Institute of Acupuncture Research, Fudan University, Shanghai, China.
| |
Collapse
|
10
|
Liu H, Zhang Y, Yip M, Ren L, Liang J, Chen X, Liu N, Du A, Wang J, Chang H, Oh H, Zhou C, Xing R, Xu M, Guo P, Gessler D, Xie J, Tai PW, Gao G, Wang D. Producing high-quantity and high-quality recombinant adeno-associated virus by low-cis triple transfection. Mol Ther Methods Clin Dev 2024; 32:101230. [PMID: 38558570 PMCID: PMC10979107 DOI: 10.1016/j.omtm.2024.101230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Accepted: 03/08/2024] [Indexed: 04/04/2024]
Abstract
Recombinant adeno-associated virus (rAAV)-based gene therapy is entering clinical and commercial stages at an unprecedented pace. Triple transfection of HEK293 cells is currently the most widely used platform for rAAV manufacturing. Here, we develop low-cis triple transfection that decreases transgene plasmid use by 10- to 100-fold and overcomes several major limitations associated with standard triple transfection. This new method improves packaging of yield-inhibiting transgenes by up to 10-fold, and generates rAAV batches with reduced plasmid backbone contamination that otherwise cannot be eliminated in downstream processing. When tested in mice and compared with rAAV produced by standard triple transfection, low-cis rAAV shows comparable or superior potency and results in diminished plasmid backbone DNA and RNA persistence in tissue. Mechanistically, low-cis triple transfection relies on the extensive replication of transgene cassette (i.e., inverted terminal repeat-flanked vector DNA) in HEK293 cells during production phase. This cost-effective method can be easily implemented and is widely applicable to producing rAAV of high quantity, purity, and potency.
Collapse
Affiliation(s)
- Hao Liu
- Horae Gene Therapy Center, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Yue Zhang
- Horae Gene Therapy Center, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Mitchell Yip
- Horae Gene Therapy Center, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Lingzhi Ren
- Horae Gene Therapy Center, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Jialing Liang
- Horae Gene Therapy Center, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Xiupeng Chen
- Horae Gene Therapy Center, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Nan Liu
- Horae Gene Therapy Center, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Ailing Du
- Horae Gene Therapy Center, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Jiaming Wang
- Horae Gene Therapy Center, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Hao Chang
- Horae Gene Therapy Center, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Hyejin Oh
- Horae Gene Therapy Center, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Chen Zhou
- Horae Gene Therapy Center, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Ruxiao Xing
- Horae Gene Therapy Center, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Mengyao Xu
- Horae Gene Therapy Center, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Peiyi Guo
- Horae Gene Therapy Center, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Dominic Gessler
- Horae Gene Therapy Center, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Jun Xie
- Horae Gene Therapy Center, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Phillip W.L. Tai
- Horae Gene Therapy Center, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Guangping Gao
- Horae Gene Therapy Center, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
- Department of Microbiology and Physiological Systems, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Dan Wang
- Horae Gene Therapy Center, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
- RNA Therapeutics Institute, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| |
Collapse
|
11
|
Zhuang K, Leng L, Su X, Wang S, Su Y, Chen Y, Yuan Z, Zi L, Li J, Xie W, Yan S, Xia Y, Wang H, Li H, Chen Z, Yuan T, Zhang J. Menin Deficiency Induces Autism-Like Behaviors by Regulating Foxg1 Transcription and Participates in Foxg1-Related Encephalopathy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2307953. [PMID: 38582517 PMCID: PMC11200012 DOI: 10.1002/advs.202307953] [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: 10/23/2023] [Revised: 03/18/2024] [Indexed: 04/08/2024]
Abstract
FOXG1 syndrome is a developmental encephalopathy caused by FOXG1 (Forkhead box G1) mutations, resulting in high phenotypic variability. However, the upstream transcriptional regulation of Foxg1 expression remains unclear. This report demonstrates that both deficiency and overexpression of Men1 (protein: menin, a pathogenic gene of MEN1 syndrome known as multiple endocrine neoplasia type 1) lead to autism-like behaviors, such as social defects, increased repetitive behaviors, and cognitive impairments. Multifaceted transcriptome analyses revealed that Foxg1 signaling is predominantly altered in Men1 deficiency mice, through its regulation of the Alpha Thalassemia/Mental Retardation Syndrome X-Linked (Atrx) factor. Atrx recruits menin to bind to the transcriptional start region of Foxg1 and mediates the regulation of Foxg1 expression by H3K4me3 (Trimethylation of histone H3 lysine 4) modification. The deficits observed in menin deficient mice are rescued by the over-expression of Foxg1, leading to normalized spine growth and restoration of hippocampal synaptic plasticity. These findings suggest that menin may have a putative role in the maintenance of Foxg1 expression, highlighting menin signaling as a potential therapeutic target for Foxg1-related encephalopathy.
Collapse
Affiliation(s)
- Kai Zhuang
- Institute of NeuroscienceCollege of MedicineXiamen UniversityXiamenFujian361105China
| | - Lige Leng
- Institute of NeuroscienceCollege of MedicineXiamen UniversityXiamenFujian361105China
| | - Xiao Su
- Institute of NeuroscienceCollege of MedicineXiamen UniversityXiamenFujian361105China
| | - Shuzhong Wang
- Institute of NeuroscienceCollege of MedicineXiamen UniversityXiamenFujian361105China
| | - Yuemin Su
- Institute of NeuroscienceCollege of MedicineXiamen UniversityXiamenFujian361105China
| | - Yanbing Chen
- Institute of NeuroscienceCollege of MedicineXiamen UniversityXiamenFujian361105China
| | - Ziqi Yuan
- Institute of NeuroscienceCollege of MedicineXiamen UniversityXiamenFujian361105China
| | - Liu Zi
- Institute of NeuroscienceCollege of MedicineXiamen UniversityXiamenFujian361105China
| | - Jieyin Li
- Institute of NeuroscienceCollege of MedicineXiamen UniversityXiamenFujian361105China
| | - Wenting Xie
- Institute of NeuroscienceCollege of MedicineXiamen UniversityXiamenFujian361105China
| | - Sihan Yan
- Institute of NeuroscienceCollege of MedicineXiamen UniversityXiamenFujian361105China
| | - Yujun Xia
- Institute of NeuroscienceCollege of MedicineXiamen UniversityXiamenFujian361105China
| | - Han Wang
- Institute of NeuroscienceCollege of MedicineXiamen UniversityXiamenFujian361105China
| | - Huifang Li
- Institute of NeuroscienceCollege of MedicineXiamen UniversityXiamenFujian361105China
| | - Zhenyi Chen
- Department of AnesthesiologyFirst Affiliated HospitalCollege of MedicineXiamen UniversityXiamenFujian361105China
| | - Tifei Yuan
- Shanghai Mental Health CenterShanghai Jiaotong University School of MedicineShanghai200030China
| | - Jie Zhang
- Institute of NeuroscienceCollege of MedicineXiamen UniversityXiamenFujian361105China
- Department of AnesthesiologyFirst Affiliated HospitalCollege of MedicineXiamen UniversityXiamenFujian361105China
- The Key Laboratory of Neural and Vascular BiologyMinistry of EducationCollege of Basic MedicineHebei Medical UniversityShijiazhuang050017China
| |
Collapse
|
12
|
Bell JM, Biesemeyer C, Turner EM, Vanderbeck MM, McGraw HF. foxg1a is required for hair cell development and regeneration in the zebrafish lateral line. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.12.589268. [PMID: 38659824 PMCID: PMC11042177 DOI: 10.1101/2024.04.12.589268] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/26/2024]
Abstract
Mechanosensory hair cells located in the inner ear mediate the sensations of hearing and balance. If damaged, mammalian inner ear hair cells are unable to regenerate, resulting in permanent sensory deficits. Aquatic vertebrates like zebrafish (Danio rerio) have a specialized class of mechanosensory hair cells found in the lateral line system, allowing them to sense changes in water current. Unlike mammalian inner ear hair cells, lateral line hair cells can robustly regenerate following damage. In mammalian models, the transcription factor Foxg1 functions to promote normal development of the inner ear. Foxg1a is expressed in lateral line sensory organs in zebrafish larvae, but its function during lateral line development and regeneration has not been investigated. We find that loss of Foxg1a function results in reduced hair cell development and regeneration, as well as decreased cellular proliferation in the lateral line system. These data suggest that Foxg1 may be a valuable target for investigation of clinical hair cell regeneration. Summary statement Our work demonstrates a role for Foxg1a in developing and regenerating new sensory cells through proliferation.
Collapse
|
13
|
Ye G, Pan G, Zhu X, Li N, Liu H, Geng G, Jiang J. An integrated analysis of the anticarcinogenic role of forkhead box protein 1 in oesophageal squamous cell carcinoma. J Cell Mol Med 2024; 28:e18294. [PMID: 38652109 PMCID: PMC11037412 DOI: 10.1111/jcmm.18294] [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: 12/12/2023] [Revised: 03/11/2024] [Accepted: 03/25/2024] [Indexed: 04/25/2024] Open
Abstract
Forkhead box protein 1 (FOXP1) serves as a tumour promoter or suppressor depending on different cancers, but its effect in oesophageal squamous cell carcinoma has not been fully elucidated. This study investigated the role of FOXP1 in oesophageal squamous cell carcinoma through bioinformatics analysis and experimental verification. We determined through public databases that FOXP1 expresses low in oesophageal squamous cell carcinoma compared with normal tissues, while high expression of FOXP1 indicates a better prognosis. We identified potential target genes regulated by FOXP1, and explored the potential biological processes and signalling pathways involved in FOXP1 in oesophageal squamous cell carcinoma through GO and KEGG enrichment, gene co-expression analysis, and protein interaction network construction. We also analysed the correlation between FOXP1 and tumour immune infiltration levels. We further validated the inhibitory effect of FOXP1 on the proliferation of oesophageal squamous cell carcinoma cells through CCK-8, colony formation and subcutaneous tumour formation assays. This study revealed the anticarcinogenic effect of FOXP1 in oesophageal squamous cell carcinoma, which may serve as a novel biological target for the treatment of tumour.
Collapse
Affiliation(s)
- Guanzhi Ye
- Department of Thoracic SurgeryThe First Affiliated Hospital of Xiamen UniversityXiamenFujianChina
| | - Gaojian Pan
- Department of Thoracic SurgeryThe First Affiliated Hospital of Xiamen UniversityXiamenFujianChina
| | - Xiaolei Zhu
- Department of Thoracic SurgeryThe First Affiliated Hospital of Xiamen UniversityXiamenFujianChina
| | - Ning Li
- Department of Thoracic SurgeryThe First Affiliated Hospital of Xiamen UniversityXiamenFujianChina
| | - Hongming Liu
- Department of Thoracic SurgeryThe First Affiliated Hospital of Xiamen UniversityXiamenFujianChina
| | - Guojun Geng
- Department of Thoracic SurgeryThe First Affiliated Hospital of Xiamen UniversityXiamenFujianChina
| | - Jie Jiang
- Department of Thoracic SurgeryThe First Affiliated Hospital of Xiamen UniversityXiamenFujianChina
| |
Collapse
|
14
|
Benedetti MC, D'andrea T, Colantoni A, Silachev D, de Turris V, Boussadia Z, Babenko VA, Volovikov EA, Belikova L, Bogomazova AN, Pepponi R, Whye D, Buttermore ED, Tartaglia GG, Lagarkova MA, Katanaev VL, Musayev I, Martinelli S, Fucile S, Rosa A. Cortical neurons obtained from patient-derived iPSCs with GNAO1 p.G203R variant show altered differentiation and functional properties. Heliyon 2024; 10:e26656. [PMID: 38434323 PMCID: PMC10907651 DOI: 10.1016/j.heliyon.2024.e26656] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Revised: 01/24/2024] [Accepted: 02/16/2024] [Indexed: 03/05/2024] Open
Abstract
Pathogenic variants in the GNAO1 gene, encoding the alpha subunit of an inhibitory heterotrimeric guanine nucleotide-binding protein (Go) highly expressed in the mammalian brain, have been linked to encephalopathy characterized by different combinations of neurological symptoms, including developmental delay, hypotonia, epilepsy and hyperkinetic movement disorder with life-threatening paroxysmal exacerbations. Currently, there are only symptomatic treatments, and little is known about the pathophysiology of GNAO1-related disorders. Here, we report the characterization of a new in vitro model system based on patient-derived induced pluripotent stem cells (hiPSCs) carrying the recurrent p.G203R amino acid substitution in Gαo, and a CRISPR-Cas9-genetically corrected isogenic control line. RNA-Seq analysis highlighted aberrant cell fate commitment in neuronal progenitor cells carrying the p.G203R pathogenic variant. Upon differentiation into cortical neurons, patients' cells showed reduced expression of early neural genes and increased expression of astrocyte markers, as well as premature and defective differentiation processes leading to aberrant formation of neuronal rosettes. Of note, comparable defects in gene expression and in the morphology of neural rosettes were observed in hiPSCs from an unrelated individual harboring the same GNAO1 variant. Functional characterization showed lower basal intracellular free calcium concentration ([Ca2+]i), reduced frequency of spontaneous activity, and a smaller response to several neurotransmitters in 40- and 50-days differentiated p.G203R neurons compared to control cells. These findings suggest that the GNAO1 pathogenic variant causes a neurodevelopmental phenotype characterized by aberrant differentiation of both neuronal and glial populations leading to a significant alteration of neuronal communication and signal transduction.
Collapse
Affiliation(s)
- Maria Cristina Benedetti
- Department of Biology and Biotechnologies “Charles Darwin”, Sapienza University of Rome, Rome, Italy
- Center for Life Nano- & Neuro-Science, Fondazione Istituto Italiano di Tecnologia (IIT), Rome, Italy
| | - Tiziano D'andrea
- Department of Physiology and Pharmacology “V. Erspamer”, Sapienza University of Rome, Rome, Italy
| | - Alessio Colantoni
- Department of Biology and Biotechnologies “Charles Darwin”, Sapienza University of Rome, Rome, Italy
- Center for Life Nano- & Neuro-Science, Fondazione Istituto Italiano di Tecnologia (IIT), Rome, Italy
| | - Denis Silachev
- School of Medicine and Life Sciences, Far Eastern Federal University, 690090, Vladivostok, Russia
- A.N. Belozersky Research Institute of Physico-Chemical Biology, Moscow State University, 119992, Moscow, Russia
- Department of Cell Physiology and Metabolism, Faculty of Medicine, Translational Research Center in Oncohaematology, University of Geneva, 1211, Geneva, Switzerland
| | - Valeria de Turris
- Center for Life Nano- & Neuro-Science, Fondazione Istituto Italiano di Tecnologia (IIT), Rome, Italy
| | - Zaira Boussadia
- National Center for Drug Research and Evaluation, Istituto Superiore di Sanità, Rome, Italy
| | - Valentina A. Babenko
- A.N. Belozersky Research Institute of Physico-Chemical Biology, Moscow State University, 119992, Moscow, Russia
- Department of Cell Physiology and Metabolism, Faculty of Medicine, Translational Research Center in Oncohaematology, University of Geneva, 1211, Geneva, Switzerland
| | - Egor A. Volovikov
- Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, 119435, Moscow, Russia
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, 119435, Moscow, Russia
| | - Lilia Belikova
- Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, 119435, Moscow, Russia
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, 119435, Moscow, Russia
| | - Alexandra N. Bogomazova
- Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, 119435, Moscow, Russia
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, 119435, Moscow, Russia
| | - Rita Pepponi
- National Center for Drug Research and Evaluation, Istituto Superiore di Sanità, Rome, Italy
| | - Dosh Whye
- Human Neuron Core, Rosamund Stone Zander Translational Neuroscience Center and F.M. Kirby Neurobiology Department, Boston Children's Hospital, Boston, MA, USA
| | - Elizabeth D. Buttermore
- Human Neuron Core, Rosamund Stone Zander Translational Neuroscience Center and F.M. Kirby Neurobiology Department, Boston Children's Hospital, Boston, MA, USA
| | - Gian Gaetano Tartaglia
- Center for Human Technologies (CHT), Istituto Italiano di Tecnologia (IIT), Genova, Italy
| | - Maria A. Lagarkova
- Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, 119435, Moscow, Russia
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, 119435, Moscow, Russia
| | - Vladimir L. Katanaev
- School of Medicine and Life Sciences, Far Eastern Federal University, 690090, Vladivostok, Russia
- Department of Cell Physiology and Metabolism, Faculty of Medicine, Translational Research Center in Oncohaematology, University of Geneva, 1211, Geneva, Switzerland
| | | | - Simone Martinelli
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Sergio Fucile
- Department of Physiology and Pharmacology “V. Erspamer”, Sapienza University of Rome, Rome, Italy
- IRCCS Neuromed, Pozzilli, Italy
| | - Alessandro Rosa
- Department of Biology and Biotechnologies “Charles Darwin”, Sapienza University of Rome, Rome, Italy
- Center for Life Nano- & Neuro-Science, Fondazione Istituto Italiano di Tecnologia (IIT), Rome, Italy
| |
Collapse
|
15
|
Pramanik S, Bala A, Pradhan A. Zebrafish in understanding molecular pathophysiology, disease modeling, and developing effective treatments for Rett syndrome. J Gene Med 2024; 26:e3677. [PMID: 38380785 DOI: 10.1002/jgm.3677] [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: 11/14/2023] [Revised: 01/04/2024] [Accepted: 01/28/2024] [Indexed: 02/22/2024] Open
Abstract
Rett syndrome (RTT) is a rare but dreadful X-linked genetic disease that mainly affects young girls. It is a neurological disease that affects nerve cell development and function, resulting in severe motor and intellectual disabilities. To date, no cure is available for treating this disease. In 90% of the cases, RTT is caused by a mutation in methyl-CpG-binding protein 2 (MECP2), a transcription factor involved in the repression and activation of transcription. MECP2 is known to regulate several target genes and is involved in different physiological functions. Mouse models exhibit a broad range of phenotypes in recapitulating human RTT symptoms; however, understanding the disease mechanisms remains incomplete, and many potential RTT treatments developed in mouse models have not shown translational effectiveness in human trials. Recent data hint that the zebrafish model emulates similar disrupted neurological functions following mutation of the mecp2 gene. This suggests that zebrafish can be used to understand the onset and progression of RTT pathophysiology and develop a possible cure. In this review, we elaborate on the molecular basis of RTT pathophysiology in humans and model organisms, including rodents and zebrafish, focusing on the zebrafish model to understand the molecular pathophysiology and the development of therapeutic strategies for RTT. Finally, we propose a rational treatment strategy, including antisense oligonucleotides, small interfering RNA technology and induced pluripotent stem cell-derived cell therapy.
Collapse
Affiliation(s)
- Subrata Pramanik
- Jyoti and Bhupat Mehta School of Health Sciences and Technology, Indian Institute of Technology Guwahati, Guwahati, Assam, India
- Centre for Nanotechnology, Indian Institute of Technology Guwahati, Guwahati, Assam, India
| | - Asis Bala
- Pharmacology and Drug Discovery Research Laboratory, Division of Life Sciences; Institute of Advanced Study in Science and Technology (IASST), An Autonomous Institute Under - Department of Science & Technology (Govt. of India) Vigyan Path, Guwahati, Assam, India
| | - Ajay Pradhan
- Biology, The Life Science Center, School of Science and Technology, Örebro University, Örebro, Sweden
| |
Collapse
|
16
|
Josephs KA, Josephs KA. Prosopagnosia: face blindness and its association with neurological disorders. Brain Commun 2024; 6:fcae002. [PMID: 38419734 PMCID: PMC10901275 DOI: 10.1093/braincomms/fcae002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Revised: 11/25/2023] [Accepted: 01/04/2024] [Indexed: 03/02/2024] Open
Abstract
Loss of facial recognition or prosopagnosia has been well-recognized for over a century. It has been categorized as developmental or acquired depending on whether the onset is in early childhood or beyond, and acquired cases can have degenerative or non-degenerative aetiologies. Prosopagnosia has been linked to involvement of the fusiform gyri, mainly in the right hemisphere. The literature on prosopagnosia comprises case reports and small case series. We aim to assess demographic, clinical and imaging characteristics and neurological and neuropathological disorders associated with a diagnosis of prosopagnosia in a large cohort. Patients were categorized as developmental versus acquired; those with acquired prosopagnosia were further subdivided into degenerative versus non-degenerative, based on neurological aetiology. We assessed regional involvement on [18F] fluorodeoxyglucose-PET and MRI of the right and left frontal, temporal, parietal and occipital lobes. The Intake and Referral Center at the Mayo Clinic identified 487 patients with possible prosopagnosia, of which 336 met study criteria for probable or definite prosopagnosia. Ten patients, 80.0% male, had developmental prosopagnosia including one with Niemann-Pick type C and another with a forkhead box G1 gene mutation. Of the 326 with acquired prosopagnosia, 235 (72.1%) were categorized as degenerative, 91 (27.9%) as non-degenerative. The most common degenerative diagnoses were posterior cortical atrophy, primary prosopagnosia syndrome, Alzheimer's disease dementia and semantic dementia, with each diagnosis accounting for >10% of this group. The most common non-degenerative diagnoses were infarcts (ischaemic and haemorrhagic), epilepsy-related and primary brain tumours, each accounting for >10%. We identified a group of patients with non-degenerative transient prosopagnosia in which facial recognition loss improved or resolved over time. These patients had migraine-related prosopagnosia, posterior reversible encephalopathy syndrome, delirium, hypoxic encephalopathy and ischaemic infarcts. On [18F] fluorodeoxyglucose-PET, the temporal lobes proved to be the most frequently affected regions in 117 patients with degenerative prosopagnosia, while in 82 patients with non-degenerative prosopagnosia, MRI revealed the right temporal and right occipital lobes as most affected by a focal lesion. The most common pathological findings in those with degenerative prosopagnosia were frontotemporal lobar degeneration with hippocampal sclerosis and mixed Alzheimer's and Lewy body disease pathology. In this large case series of patients diagnosed with prosopagnosia, we observed that facial recognition loss occurs across a wide range of acquired degenerative and non-degenerative neurological disorders, most commonly in males with developmental prosopagnosia. The right temporal and occipital lobes, and connecting fusiform gyrus, are key areas. Multiple different pathologies cause degenerative prosopagnosia.
Collapse
Affiliation(s)
| | - Keith A Josephs
- Department of Neurology, Mayo Clinic, Rochester, MN 55905, USA
| |
Collapse
|
17
|
Ermakova GV, Kucheryavyy AV, Mugue NS, Mischenko AV, Zaraisky AG, Bayramov AV. Three foxg1 paralogues in lampreys and gnathostomes-brothers or cousins? Front Cell Dev Biol 2024; 11:1321317. [PMID: 38229883 PMCID: PMC10789856 DOI: 10.3389/fcell.2023.1321317] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Accepted: 12/05/2023] [Indexed: 01/18/2024] Open
Abstract
Foxg1 is a key regulator of the early development of the vertebrate forebrain and sensory organs. In this study, we describe for the first time three foxg1 paralogues in lamprey, representative of one of two basally diverged lineages of vertebrates-the agnathans. We also first describe three foxg1 genes in sterlet-representative of one of the evolutionarily ancient clades of gnathostomes. According to the analysis of local genomic synteny, three foxg1 genes of agnathans and gnathostomes have a common origin as a result of two rounds of genomic duplications in the early evolution of vertebrates. At the same time, it is difficult to reliably establish pairwise orthology between foxg1 genes of agnathans and gnathostomes based on the analysis of phylogeny and local genomic synteny, as well as our studies of the spatiotemporal expression of foxg1 genes in the river lamprey Lampetra fluviatilis and the sterlet Acipenser ruthenus. Thus, the appearance of three foxg1 paralogues in agnathans and gnathostomes could have occurred either as a result of two rounds of duplication of the vertebrate common ancestor genome (2R hypothesis) or as a result of the first common round followed by subsequent independent polyploidizations in two evolutionary lineages (1R hypothesis).
Collapse
Affiliation(s)
- Galina V. Ermakova
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
| | | | - Nikolay S. Mugue
- Russian Federal Research Institute of Fisheries and Oceanography (VNIRO), Moscow, Russia
- Koltzov Institute of Developmental Biology of Russian Academy of Sciences, Moscow, Russia
| | - Aleksandr V. Mischenko
- Branch for the Freshwater Fisheries of the Russian Federal Research Institute of Fisheries and Oceanography, Moscow, Russia
| | - Andrey G. Zaraisky
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
- Pirogov Russian National Research Medical University, Moscow, Russia
| | - Andrey V. Bayramov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
| |
Collapse
|
18
|
Abstract
Rett syndrome is a neurodevelopmental disorder caused by loss-of-function mutations in the methyl-CpG binding protein-2 (MeCP2) gene that is characterized by epilepsy, intellectual disability, autistic features, speech deficits, and sleep and breathing abnormalities. Neurologically, patients with all three disorders display microcephaly, aberrant dendritic morphology, reduced spine density, and an imbalance of excitatory/inhibitory signaling. Loss-of-function mutations in the cyclin-dependent kinase-like 5 (CDKL5) and FOXG1 genes also cause similar behavioral and neurobiological defects and were referred to as congenital or variant Rett syndrome. The relatively recent realization that CDKL5 deficiency disorder (CDD), FOXG1 syndrome, and Rett syndrome are distinct neurodevelopmental disorders with some distinctive features have resulted in separate focus being placed on each disorder with the assumption that distinct molecular mechanisms underlie their pathogenesis. However, given that many of the core symptoms and neurological features are shared, it is likely that the disorders share some critical molecular underpinnings. This review discusses the possibility that deregulation of common molecules in neurons and astrocytes plays a central role in key behavioral and neurological abnormalities in all three disorders. These include KCC2, a chloride transporter, vGlut1, a vesicular glutamate transporter, GluD1, an orphan-glutamate receptor subunit, and PSD-95, a postsynaptic scaffolding protein. We propose that reduced expression or activity of KCC2, vGlut1, PSD-95, and AKT, along with increased expression of GluD1, is involved in the excitatory/inhibitory that represents a key aspect in all three disorders. In addition, astrocyte-derived brain-derived neurotrophic factor (BDNF), insulin-like growth factor 1 (IGF-1), and inflammatory cytokines likely affect the expression and functioning of these molecules resulting in disease-associated abnormalities.
Collapse
Affiliation(s)
- Santosh R D’Mello
- Department of Biological Sciences, Louisiana State University Shreveport, Shreveport, LA 71104, USA
| |
Collapse
|
19
|
Toudji I, Toumi A, Chamberland É, Rossignol E. Interneuron odyssey: molecular mechanisms of tangential migration. Front Neural Circuits 2023; 17:1256455. [PMID: 37779671 PMCID: PMC10538647 DOI: 10.3389/fncir.2023.1256455] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Accepted: 08/21/2023] [Indexed: 10/03/2023] Open
Abstract
Cortical GABAergic interneurons are critical components of neural networks. They provide local and long-range inhibition and help coordinate network activities involved in various brain functions, including signal processing, learning, memory and adaptative responses. Disruption of cortical GABAergic interneuron migration thus induces profound deficits in neural network organization and function, and results in a variety of neurodevelopmental and neuropsychiatric disorders including epilepsy, intellectual disability, autism spectrum disorders and schizophrenia. It is thus of paramount importance to elucidate the specific mechanisms that govern the migration of interneurons to clarify some of the underlying disease mechanisms. GABAergic interneurons destined to populate the cortex arise from multipotent ventral progenitor cells located in the ganglionic eminences and pre-optic area. Post-mitotic interneurons exit their place of origin in the ventral forebrain and migrate dorsally using defined migratory streams to reach the cortical plate, which they enter through radial migration before dispersing to settle in their final laminar allocation. While migrating, cortical interneurons constantly change their morphology through the dynamic remodeling of actomyosin and microtubule cytoskeleton as they detect and integrate extracellular guidance cues generated by neuronal and non-neuronal sources distributed along their migratory routes. These processes ensure proper distribution of GABAergic interneurons across cortical areas and lamina, supporting the development of adequate network connectivity and brain function. This short review summarizes current knowledge on the cellular and molecular mechanisms controlling cortical GABAergic interneuron migration, with a focus on tangential migration, and addresses potential avenues for cell-based interneuron progenitor transplants in the treatment of neurodevelopmental disorders and epilepsy.
Collapse
Affiliation(s)
- Ikram Toudji
- Centre Hospitalier Universitaire (CHU) Sainte-Justine Research Center, Montréal, QC, Canada
- Department of Neurosciences, Université de Montréal, Montréal, QC, Canada
| | - Asmaa Toumi
- Centre Hospitalier Universitaire (CHU) Sainte-Justine Research Center, Montréal, QC, Canada
- Department of Biochemistry and Molecular Medicine, Université de Montréal, Montréal, QC, Canada
| | - Émile Chamberland
- Centre Hospitalier Universitaire (CHU) Sainte-Justine Research Center, Montréal, QC, Canada
- Department of Neurosciences, Université de Montréal, Montréal, QC, Canada
| | - Elsa Rossignol
- Centre Hospitalier Universitaire (CHU) Sainte-Justine Research Center, Montréal, QC, Canada
- Department of Neurosciences, Université de Montréal, Montréal, QC, Canada
- Department of Pediatrics, Université de Montréal, Montréal, QC, Canada
| |
Collapse
|
20
|
Cao G, Sun C, Shen H, Qu D, Shen C, Lu H. Conditional Deletion of Foxg1 Delayed Myelination during Early Postnatal Brain Development. Int J Mol Sci 2023; 24:13921. [PMID: 37762220 PMCID: PMC10530892 DOI: 10.3390/ijms241813921] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2023] [Revised: 09/06/2023] [Accepted: 09/08/2023] [Indexed: 09/29/2023] Open
Abstract
FOXG1 (forkhead box G1) syndrome is a neurodevelopmental disorder caused by variants in the Foxg1 gene that affect brain structure and function. Individuals affected by FOXG1 syndrome frequently exhibit delayed myelination in neuroimaging studies, which may impair the rapid conduction of nerve impulses. To date, the specific effects of FOXG1 on oligodendrocyte lineage progression and myelination during early postnatal development remain unclear. Here, we investigated the effects of Foxg1 deficiency on myelin development in the mouse brain by conditional deletion of Foxg1 in neural progenitors using NestinCreER;Foxg1fl/fl mice and tamoxifen induction at postnatal day 0 (P0). We found that Foxg1 deficiency resulted in a transient delay in myelination, evidenced by decreased myelin formation within the first two weeks after birth, but ultimately recovered to the control levels by P30. We also found that Foxg1 deletion prevented the timely attenuation of platelet-derived growth factor receptor alpha (PDGFRα) signaling and reduced the cell cycle exit of oligodendrocyte precursor cells (OPCs), leading to their excessive proliferation and delayed maturation. Additionally, Foxg1 deletion increased the expression of Hes5, a myelin formation inhibitor, as well as Olig2 and Sox10, two promoters of OPC differentiation. Our results reveal the important role of Foxg1 in myelin development and provide new clues for further exploring the pathological mechanisms of FOXG1 syndrome.
Collapse
Affiliation(s)
- Guangliang Cao
- Department of Human Anatomy, School of Medicine, Southeast University, Nanjing 210009, China; (G.C.); (H.S.); (D.Q.)
| | - Congli Sun
- Department of Physiology, School of Medicine, Southeast University, Nanjing 210009, China;
| | - Hualin Shen
- Department of Human Anatomy, School of Medicine, Southeast University, Nanjing 210009, China; (G.C.); (H.S.); (D.Q.)
| | - Dewei Qu
- Department of Human Anatomy, School of Medicine, Southeast University, Nanjing 210009, China; (G.C.); (H.S.); (D.Q.)
| | - Chuanlu Shen
- Department of Pathophysiology, School of Medicine, Southeast University, Nanjing 210009, China;
| | - Haiqin Lu
- Department of Human Anatomy, School of Medicine, Southeast University, Nanjing 210009, China; (G.C.); (H.S.); (D.Q.)
| |
Collapse
|
21
|
Oluigbo DC. Rett Syndrome: A Tale of Altered Genetics, Synaptic Plasticity, and Neurodevelopmental Dynamics. Cureus 2023; 15:e41555. [PMID: 37554594 PMCID: PMC10405636 DOI: 10.7759/cureus.41555] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/07/2023] [Indexed: 08/10/2023] Open
Abstract
Rett syndrome (RTT) is a neurodevelopmental disorder that is a leading cause of severe cognitive and physical impairment. RTT typically occurs in females, although rare cases of males with the disease exist. Its genetic cause, symptoms, and clinical progression timeline have also become well-documented since its initial discovery. However, a relatively late diagnosis and lack of an available cure signify that our understanding of the disease is incomplete. Innovative research methods and tools are thereby helping to fill gaps in our knowledge of RTT. Specifically, mouse models of RTT, video analysis, and retrospective parental analysis are well-established tools that provide valuable insights into RTT. Moreover, current and anticipated treatment options are improving the quality of life of the RTT patient population. Collectively, these developments are creating optimistic future perspectives for RTT.
Collapse
Affiliation(s)
- David C Oluigbo
- Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, USA
| |
Collapse
|
22
|
Park J, Moon JH, O'Shea H, Shin D, Hwang SU, Li L, Lee H, Brimble E, Lee J, Clark S, Lee SK, Jeon S. The patient-specific mouse model with Foxg1 frameshift mutation uncovers the pathophysiology of FOXG1 syndrome. RESEARCH SQUARE 2023:rs.3.rs-2953760. [PMID: 37398410 PMCID: PMC10312924 DOI: 10.21203/rs.3.rs-2953760/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/04/2023]
Abstract
Single allelic mutations in the gene encoding the forebrain-specific transcription factor FOXG1 lead to FOXG1 syndrome (FS). Patient-specific animal models are needed to understand the etiology of FS, as FS patients show a wide spectrum of symptoms correlated with location and mutation type in the FOXG1 gene. Here we report the first patient-specific FS mouse model, Q84Pfs heterozygous (Q84Pfs-Het) mice, mimicking one of the most predominant single nucleotide variants in FS. Intriguingly, we found that Q84Pfs-Het mice faithfully recapitulate human FS phenotypes at the cellular, brain structural, and behavioral levels. Importantly, Q84Pfs-Het mice exhibited myelination deficits like FS patients. Further, our transcriptome analysis of Q84Pfs-Het cortex revealed a new role for FOXG1 in synapse and oligodendrocyte development. The dysregulated genes in Q84Pfs-Het brains also predicted motor dysfunction and autism-like phenotypes. Correspondingly, Q84Pfs-Het mice showed movement deficits, repetitive behaviors, increased anxiety, and prolonged behavior arrest. Together, our study revealed the crucial postnatal role of FOXG1 in neuronal maturation and myelination and elucidated the essential pathophysiology mechanisms of FS.
Collapse
|
23
|
Wong LC, Huang CH, Chou WY, Hsu CJ, Tsai WC, Lee WT. The clinical and sleep manifestations in children with FOXG1 syndrome. Autism Res 2023; 16:953-966. [PMID: 36942618 DOI: 10.1002/aur.2916] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2022] [Accepted: 02/25/2023] [Indexed: 03/23/2023]
Abstract
FOXG1 syndrome is a rare neurodevelopmental disorder associated with severe cognitive dysfunction, autistic behavior, and early-onset hyperkinetic movement disorders. Patients have also been reported to experience sleep disturbances. However, these findings are mainly based on subjective caregivers' reports, and limited by small case numbers. Moreover, no studies using objective evaluation tools, such as actigraphy, have been reported. We analyzed the clinical and sleep manifestations of children with FOXG1 syndrome registered in the International FOXG1 Research Foundation registry database. A total of 258 individuals with FOXG1 syndrome were included in this research. 132 (51.16%) had sleep disturbances. The more impaired of language acquisitions (absence of speech, OR: 3.99, 95%CI = 1.69-9.42, p = 0.002), hyperkinetic movement disorders (OR: 2.64, 95%CI = 1.34-5.20 p = 0.005) and feeding difficulties (OR: 2.81, 95% CI = 1.52-5.19, p = 0.001) were significantly associated with an increase in odds of sleep disturbance after adjusting for age, sex, and antiepileptic drugs. We also performed sleep studies on six individuals with FOXG1 syndrome using The Children's Sleep Habits Questionnaire (CSHQ), the Sleep Disturbance Scale for Children (SDSC), and 7-day data from Actiwatch. The Pittsburgh Sleep Quality Index (PSQI) and 7-day data from Actiwatch were also used to evaluate the sleep condition of their parents. The CSHQ scores revealed bedtime resistance, sleep onset delay, sleep duration, sleep anxiety, night-waking, and parasomnia. Sleep-wake transition disorders and disorders of initiating and maintaining sleep were also suggested by the SDSC scores. The children's actigraphy revealed short sleep durations, impaired sleep efficiency, longer wake after sleep onset, and frequent night-waking. All caregivers reported significantly higher PSQI scores, mildly declined sleep efficiency, and shorter total sleep duration. Sleep disturbances, especially in initiating and maintaining sleep, are common in individuals with FOXG1 syndrome and their caregivers. Sleep disorders in patients with FOXG1 syndrome and their caregivers should be investigated.
Collapse
Affiliation(s)
- Lee-Chin Wong
- Department of Pediatrics, National Taiwan University Hospital, Taipei City, Taiwan
- Graduate Institute of Clinical Medicine, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Cheng-Hsien Huang
- Department of Pediatrics, Sleep center, Yang-Ming Branch, Taipei City Hospital, Taipei City, Taiwan
- University of Taipei, Taipei City, Taiwan
| | - Wan-Yun Chou
- Department of Medical Research, Cathay General Hospital, Taipei City, Taiwan
| | - Chia-Jui Hsu
- Department of Pediatrics, Hsin-Chu Branch, National Taiwan University Hospital, Hsinchu City, Taiwan
| | - Wen-Che Tsai
- Department of Psychiatry, National Taiwan University Hospital, Taipei City, Taiwan
| | - Wang-Tso Lee
- Department of Pediatrics, National Taiwan University Hospital, Taipei City, Taiwan
- Graduate Institute of Brain and Mind Sciences, National Taiwan University College of Medicine, Taipei City, Taiwan
| |
Collapse
|
24
|
Spagnoli C, Fusco C, Pisani F. Pediatric-Onset Epilepsy and Developmental Epileptic Encephalopathies Followed by Early-Onset Parkinsonism. Int J Mol Sci 2023; 24:ijms24043796. [PMID: 36835207 PMCID: PMC9965035 DOI: 10.3390/ijms24043796] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Revised: 02/09/2023] [Accepted: 02/10/2023] [Indexed: 02/16/2023] Open
Abstract
Genetic early-onset Parkinsonism is unique due to frequent co-occurrence of hyperkinetic movement disorder(s) (MD), or additional neurological of systemic findings, including epilepsy in up to 10-15% of cases. Based on both the classification of Parkinsonism in children proposed by Leuzzi and coworkers and the 2017 ILAE epilepsies classification, we performed a literature review in PubMed. A few discrete presentations can be identified: Parkinsonism as a late manifestation of complex neurodevelopmental disorders, characterized by developmental and epileptic encephalopathies (DE-EE), with multiple, refractory seizure types and severely abnormal EEG characteristics, with or without preceding hyperkinetic MD; Parkinsonism in the context of syndromic conditions with unspecific reduced seizure threshold in infancy and childhood; neurodegenerative conditions with brain iron accumulation, in which childhood DE-EE is followed by neurodegeneration; and finally, monogenic juvenile Parkinsonism, in which a subset of patients with intellectual disability or developmental delay (ID/DD) develop hypokinetic MD between 10 and 30 years of age, following unspecific, usually well-controlled, childhood epilepsy. This emerging group of genetic conditions leading to epilepsy or DE-EE in childhood followed by juvenile Parkinsonism highlights the need for careful long-term follow-up, especially in the context of ID/DD, in order to readily identify individuals at increased risk of later Parkinsonism.
Collapse
Affiliation(s)
- Carlotta Spagnoli
- Child Neurology and Psychiatry Unit, Department of Pediatrics, Presidio Ospedaliero Santa Maria Nuova, AUSL-IRCCS di Reggio Emilia, 42122 Reggio Emilia, Italy
- Correspondence: ; Tel.: +39-0522-296033
| | - Carlo Fusco
- Child Neurology and Psychiatry Unit, Department of Pediatrics, Presidio Ospedaliero Santa Maria Nuova, AUSL-IRCCS di Reggio Emilia, 42122 Reggio Emilia, Italy
| | - Francesco Pisani
- Human Neurosciences Department, Sapienza University of Rome, 00185 Rome, Italy
| |
Collapse
|
25
|
Involvement of Mitochondrial Dysfunction in FOXG1 Syndrome. Genes (Basel) 2023; 14:genes14020246. [PMID: 36833172 PMCID: PMC9957531 DOI: 10.3390/genes14020246] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 01/12/2023] [Accepted: 01/16/2023] [Indexed: 01/19/2023] Open
Abstract
FOXG1 (Forkhead box g1) syndrome is a neurodevelopmental disorder caused by a defective transcription factor, FOXG1, important for normal brain development and function. As FOXG1 syndrome and mitochondrial disorders have shared symptoms and FOXG1 regulates mitochondrial function, we investigated whether defective FOXG1 leads to mitochondrial dysfunction in five individuals with FOXG1 variants compared to controls (n = 6). We observed a significant decrease in mitochondrial content and adenosine triphosphate (ATP) levels and morphological changes in mitochondrial network in the fibroblasts of affected individuals, indicating involvement of mitochondrial dysfunction in FOXG1 syndrome pathogenesis. Further investigations are warranted to elucidate how FOXG1 deficiency impairs mitochondrial homeostasis.
Collapse
|
26
|
Differential vulnerability of adult neurogenic niches to dosage of the neurodevelopmental-disorder linked gene Foxg1. Mol Psychiatry 2023; 28:497-514. [PMID: 35318461 PMCID: PMC9812795 DOI: 10.1038/s41380-022-01497-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Revised: 02/14/2022] [Accepted: 02/22/2022] [Indexed: 01/13/2023]
Abstract
The transcription factor FOXG1 serves pleiotropic functions in brain development ranging from the regulation of precursor proliferation to the control of cortical circuit formation. Loss-of-function mutations and duplications of FOXG1 are associated with neurodevelopmental disorders in humans illustrating the importance of FOXG1 dosage for brain development. Aberrant FOXG1 dosage has been found to disrupt the balanced activity of glutamatergic and GABAergic neurons, but the underlying mechanisms are not fully understood. We report that FOXG1 is expressed in the main adult neurogenic niches in mice, i.e. the hippocampal dentate gyrus and the subependymal zone/olfactory bulb system, where neurogenesis of glutamatergic and GABAergic neurons persists into adulthood. These niches displayed differential vulnerability to increased FOXG1 dosage: high FOXG1 levels severely compromised survival and glutamatergic dentate granule neuron fate acquisition in the hippocampal neurogenic niche, but left neurogenesis of GABAergic neurons in the subependymal zone/olfactory bulb system unaffected. Comparative transcriptomic analyses revealed a significantly higher expression of the apoptosis-linked nuclear receptor Nr4a1 in FOXG1-overexpressing hippocampal neural precursors. Strikingly, pharmacological interference with NR4A1 function rescued FOXG1-dependent death of hippocampal progenitors. Our results reveal differential vulnerability of neuronal subtypes to increased FOXG1 dosage and suggest that activity of a FOXG1/NR4A1 axis contributes to such subtype-specific response.
Collapse
|
27
|
Lu G, Zhang Y, Xia H, He X, Xu P, Wu L, Li D, Ma L, Wu J, Peng Q. Identification of a de novo mutation of the FOXG1 gene and comprehensive analysis for molecular factors in Chinese FOXG1-related encephalopathies. Front Mol Neurosci 2022; 15:1039990. [PMID: 36568277 PMCID: PMC9768341 DOI: 10.3389/fnmol.2022.1039990] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Accepted: 11/11/2022] [Indexed: 12/12/2022] Open
Abstract
Background FOXG1-related encephalopathy, also known as FOXG1 syndrome or FOXG1-related disorder, affects most aspects of development and causes microcephaly and brain malformations. This syndrome was previously considered to be the congenital variant of Rett syndrome. The abnormal function or expression of FOXG1, caused by intragenic mutations, microdeletions or microduplications, was considered to be crucial pathological factor for this disorder. Currently, most of the FOXG1-related encephalopathies have been identified in Europeans and North Americans, and relatively few Chinese cases were reported. Methods Array-Comparative Genomic Hybridization (Array-CGH) and whole-exome sequencing (WES) were carried out for the proband and her parent to detect pathogenic variants. Results A de novo nonsense mutation (c.385G>T, p.Glu129Ter) of FOXG1 was identified in a female child in a cohort of 73 Chinese children with neurodevelopmental disorders/intellectual disorders (NDDs/IDs). In order to have a comprehensive view of FOXG1-related encephalopathy in China, relevant published reports were browsed and twelve cases with mutations in FOXG1 or copy number variants (CNVs) involving FOXG1 gene were involved in the analysis eventually. Feeding difficulties, seizures, delayed speech, corpus callosum hypoplasia and underdevelopment of frontal and temporal lobes occurred in almost all cases. Out of the 12 cases, eight patients (66.67%) had single-nucleotide mutations of FOXG1 gene and four patients (33.33%) had CNVs involving FOXG1 (3 microdeletions and 1 microduplication). The expression of FOXG1 could also be potentially disturbed by deletions of several brain-active regulatory elements located in intergenic FOXG1-PRKD1 region. Further analysis indicated that PRKD1 might be a cooperating factor to regulate the expression of FOXG1, MECP2 and CDKL5 to contribute the RTT/RTT-like disorders. Discussion This re-analysis would broaden the existed knowledge about the molecular etiology and be helpful for diagnosis, treatment, and gene therapy of FOXG1-related disorders in the future.
Collapse
Affiliation(s)
- Guanting Lu
- Laboratory of Translational Medicine Research, Department of Pathology, Deyang People's Hospital, Deyang, China
- Key Laboratory of Tumor Molecular Research of Deyang, Deyang, China
| | - Yan Zhang
- Department of Obstetrics and Gynecology, Strategic Support Force Medical Center, Beijing, China
| | - Huiyun Xia
- Department of Child Healthcare, Shenzhen Baoan Women's and Children's Hospital, Jinan University, Shenzhen, China
| | - Xiaoyan He
- Laboratory of Translational Medicine Research, Department of Pathology, Deyang People's Hospital, Deyang, China
- Key Laboratory of Tumor Molecular Research of Deyang, Deyang, China
| | - Pei Xu
- Laboratory of Translational Medicine Research, Department of Pathology, Deyang People's Hospital, Deyang, China
- Key Laboratory of Tumor Molecular Research of Deyang, Deyang, China
| | - Lianying Wu
- Laboratory of Translational Medicine Research, Department of Pathology, Deyang People's Hospital, Deyang, China
- Key Laboratory of Tumor Molecular Research of Deyang, Deyang, China
| | - Ding Li
- Key Laboratory of Tumor Molecular Research of Deyang, Deyang, China
| | - Liya Ma
- Department of Child Healthcare, Shenzhen Baoan Women's and Children's Hospital, Jinan University, Shenzhen, China
| | - Jin Wu
- Laboratory of Translational Medicine Research, Department of Pathology, Deyang People's Hospital, Deyang, China
- Key Laboratory of Tumor Molecular Research of Deyang, Deyang, China
| | - Qiongling Peng
- Department of Child Healthcare, Shenzhen Baoan Women's and Children's Hospital, Jinan University, Shenzhen, China
| |
Collapse
|
28
|
Epigenetic genes and epilepsy - emerging mechanisms and clinical applications. Nat Rev Neurol 2022; 18:530-543. [PMID: 35859062 DOI: 10.1038/s41582-022-00693-y] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/16/2022] [Indexed: 12/21/2022]
Abstract
An increasing number of epilepsies are being attributed to variants in genes with epigenetic functions. The products of these genes include factors that regulate the structure and function of chromatin and the placing, reading and removal of epigenetic marks, as well as other epigenetic processes. In this Review, we provide an overview of the various epigenetic processes, structuring our discussion around five function-based categories: DNA methylation, histone modifications, histone-DNA crosstalk, non-coding RNAs and chromatin remodelling. We provide background information on each category, describing the general mechanism by which each process leads to altered gene expression. We also highlight key clinical and mechanistic aspects, providing examples of genes that strongly associate with epilepsy within each class. We consider the practical applications of these findings, including tissue-based and biofluid-based diagnostics and precision medicine-based treatments. We conclude that variants in epigenetic genes are increasingly found to be causally involved in the epilepsies, with implications for disease mechanisms, treatments and diagnostics.
Collapse
|
29
|
Yang H, Ryu J, Lim C, Choi JW, Park YJ, Jang SW, Shim S. SOXE group transcription factors regulates the expression of FoxG1 during inner ear development. Biochem Biophys Res Commun 2022; 623:96-103. [PMID: 35878429 DOI: 10.1016/j.bbrc.2022.07.048] [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: 07/06/2022] [Accepted: 07/13/2022] [Indexed: 11/24/2022]
Abstract
The transcription factor FOXG1 plays an important role in inner ear development; however, the cis-regulatory mechanisms controlling the inner-ear-specific expression of FOXG1 are poorly understood. In this study, we aimed to identify the element that specifically regulates FoxG1 expression in the otic vesicle, which develops into the inner ear, through comparative genome analysis between vertebrate species and chromatin immunoprecipitation. The cis-regulatory element (E2) identified showed high evolutionary conservation among vertebrates in the genomic DNA of FoxG1 spanning approximately 3 Mbp. We identified core sequences important for the activity of the otic-vesicle-specific enhancer through in vitro and in vivo reporter assays for various E2 enhancer mutants and determined the consensus sequence for SOX DNA binding. In addition, SoxE, a subfamily of the Sox family, was simultaneously expressed in the otic vesicles of developing embryos and showed a similar protein expression pattern as that of FoxG1. Furthermore, SOXE transcription factors induced specific transcriptional activity through the FoxG1 Otic enhancer (E2b). These findings suggest that the interaction between the otic enhancer of FoxG1 and SOXE transcription factor, in which the otic expression of FoxG1 is evolutionarily well-conserved, is important during early development of the inner ear, a sensory organ important for survival in nature.
Collapse
Affiliation(s)
- Hayoung Yang
- Department of Biochemistry, Chungbuk National University, Cheongju, 28644, Republic of Korea
| | - Jiho Ryu
- Department of Biochemistry, Chungbuk National University, Cheongju, 28644, Republic of Korea
| | - Chungun Lim
- Department of Biomedical Sciences, University of Ulsan College of Medicine, Seoul, 05505, Republic of Korea
| | - Jae-Won Choi
- Division of BT Convergence, Cheongju University, Cheongju, 28503, Republic of Korea
| | - Young-Jun Park
- Immunotherapy Research Center, Korea Research Institute of Bioscience and Biotechnology, Yuseong-gu, Daejeon, Republic of Korea
| | - Sung-Wuk Jang
- Department of Biomedical Sciences, University of Ulsan College of Medicine, Seoul, 05505, Republic of Korea; Asan Medical Institute of Convergence Science and Technology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, 138-736, Republic of Korea.
| | - Sungbo Shim
- Department of Biochemistry, Chungbuk National University, Cheongju, 28644, Republic of Korea.
| |
Collapse
|
30
|
Lottini G, Baggiani M, Chesi G, D'Orsi B, Quaranta P, Lai M, Pancrazi L, Onorati M, Pistello M, Freer G, Costa M. Zika virus induces FOXG1 nuclear displacement and downregulation in human neural progenitors. Stem Cell Reports 2022; 17:1683-1698. [PMID: 35714598 PMCID: PMC9287670 DOI: 10.1016/j.stemcr.2022.05.008] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Revised: 05/13/2022] [Accepted: 05/17/2022] [Indexed: 11/25/2022] Open
Abstract
Congenital alterations in the levels of the transcription factor Forkhead box g1 (FOXG1) coding gene trigger "FOXG1 syndrome," a spectrum that recapitulates birth defects found in the "congenital Zika syndrome," such as microcephaly and other neurodevelopmental conditions. Here, we report that Zika virus (ZIKV) infection alters FOXG1 nuclear localization and causes its downregulation, thus impairing expression of genes involved in cell replication and apoptosis in several cell models, including human neural progenitor cells. Growth factors, such as EGF and FGF2, and Thr271 residue located in FOXG1 AKT domain, take part in the nuclear displacement and apoptosis protection, respectively. Finally, by progressive deletion of FOXG1 sequence, we identify the C-terminus and the residues 428-481 as critical domains. Collectively, our data suggest a causal mechanism by which ZIKV affects FOXG1, its target genes, cell cycle progression, and survival of human neural progenitors, thus contributing to microcephaly.
Collapse
Affiliation(s)
- Giulia Lottini
- Centro Retrovirus, Department of Translational Research, University of Pisa, Pisa 56127, Italy; Department of Medical Biotechnologies, University of Siena, Siena 53100, Italy
| | - Matteo Baggiani
- Unit of Cell and Developmental Biology, Department of Biology, University of Pisa, Pisa 56127, Italy
| | - Giulia Chesi
- Centro Retrovirus, Department of Translational Research, University of Pisa, Pisa 56127, Italy
| | - Beatrice D'Orsi
- Institute of Neuroscience, Italian National Research Council (CNR), Via Moruzzi, 1, Pisa 56124, Italy; Centro Pisano ricerca e implementazione clinica Flash Radiotherapy (CPFR@CISUP), Presidio S. Chiara, ed.18 via Roma, 67, Pisa 56126, Italy
| | - Paola Quaranta
- Centro Retrovirus, Department of Translational Research, University of Pisa, Pisa 56127, Italy
| | - Michele Lai
- Centro Retrovirus, Department of Translational Research, University of Pisa, Pisa 56127, Italy
| | - Laura Pancrazi
- Institute of Neuroscience, Italian National Research Council (CNR), Via Moruzzi, 1, Pisa 56124, Italy
| | - Marco Onorati
- Unit of Cell and Developmental Biology, Department of Biology, University of Pisa, Pisa 56127, Italy
| | - Mauro Pistello
- Centro Retrovirus, Department of Translational Research, University of Pisa, Pisa 56127, Italy
| | - Giulia Freer
- Centro Retrovirus, Department of Translational Research, University of Pisa, Pisa 56127, Italy
| | - Mario Costa
- Institute of Neuroscience, Italian National Research Council (CNR), Via Moruzzi, 1, Pisa 56124, Italy; Centro Pisano ricerca e implementazione clinica Flash Radiotherapy (CPFR@CISUP), Presidio S. Chiara, ed.18 via Roma, 67, Pisa 56126, Italy; Laboratory of Biology "Bio@SNS", Scuola Normale Superiore, Piazza dei Cavalieri, Pisa 56124, Italy.
| |
Collapse
|
31
|
Tascini G, Dell'Isola GB, Mencaroni E, Di Cara G, Striano P, Verrotti A. Sleep Disorders in Rett Syndrome and Rett-Related Disorders: A Narrative Review. Front Neurol 2022; 13:817195. [PMID: 35299616 PMCID: PMC8923297 DOI: 10.3389/fneur.2022.817195] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Accepted: 02/02/2022] [Indexed: 11/13/2022] Open
Abstract
Rett Syndrome (RTT) is a rare and severe X-linked developmental brain disorder that occurs primarily in females, with a ratio of 1:10.000. De novo mutations in the Methyl-CpG Binding protein 2 (MECP2) gene on the long arm of X chromosome are responsible for more than 95% cases of classical Rett. In the remaining cases (atypical Rett), other genes are involved such as the cyclin-dependent kinase-like 5 (CDKL5) and the forkhead box G1 (FOXG1). Duplications of the MECP2 locus cause MECP2 duplication syndrome (MDS) which concerns about 1% of male patients with intellectual disability. Sleep disorders are common in individuals with intellectual disability, while the prevalence in children is between 16 and 42%. Over 80% of individuals affected by RTT show sleep problems, with a higher prevalence in the first 7 years of life and some degree of variability in correlation to age and genotype. Abnormalities in circadian rhythm and loss of glutamate homeostasis play a key role in the development of these disorders. Sleep disorders, epilepsy, gastrointestinal problems characterize CDKL5 Deficiency Disorder (CDD). Sleep impairment is an area of overlap between RTT and MECP2 duplication syndrome along with epilepsy, regression and others. Sleep dysfunction and epilepsy are deeply linked. Sleep deprivation could be an aggravating factor of epilepsy and anti-comitial therapy could interfere in sleep structure. Epilepsy prevalence in atypical Rett syndrome with severe clinical phenotype is higher than in classical Rett syndrome. However, RTT present a significant lifetime risk of epilepsy too. Sleep disturbances impact on child's development and patients' families and the evidence for its management is still limited. The aim of this review is to analyze pathophysiology, clinical features, the impact on other comorbidities and the management of sleep disorders in Rett syndrome and Rett-related syndrome.
Collapse
Affiliation(s)
- Giorgia Tascini
- Department of Pediatrics, University of Perugia, Perugia, Italy
| | | | | | | | - Pasquale Striano
- Pediatric Neurology and Muscular Diseases Unit, IRCCS "G. Gaslini" Institute, Genoa, Italy.,Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, University of Genoa, Genoa, Italy
| | | |
Collapse
|
32
|
Jang HN, Kim T, Jung AY, Lee BH, Yum MS, Ko TS. Identification of FOXG1 mutations in infantile hypotonia and postnatal microcephaly. Medicine (Baltimore) 2021; 100:e27949. [PMID: 34964776 PMCID: PMC8615421 DOI: 10.1097/md.0000000000027949] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Revised: 10/05/2021] [Accepted: 11/01/2021] [Indexed: 01/05/2023] Open
Abstract
ABSTRACT FOXG1, located at chromosome 14q12, is critical for brain development, and patients with FOXG1 mutation exhibit developmental encephalopathy with high phenotypic variability, known as FOXG1 syndrome. Here, we report 3 cases of FOXG1 syndrome that presented with infantile hypotonia and microcephaly.A total of 145 children with developmental delay and/or hypotonia were evaluated by whole-exome sequencing (WES) in the pediatric neurology clinic and medical genetics center at Asan Medical Center Children's Hospital, from 2017 to 2019. Each FOXG1 mutation was confirmed by Sanger sequencing. The clinical findings of each patient with FOXG1 mutation were reviewed.WES identified de-novo, pathogenic, and heterozygous FOXG1 mutations in 3 of 145 patients in our patient cohort with developmental delay and/or hypotonia. The characteristics of brain magnetic resonance imaging (MRI) were reported as callosal anomaly, decrease in frontal volume, fornix thickening, and hypoplastic olfactory bulbs. A phenotype-genotype correlation was demonstrated as a patient with a novel missense mutation, c.761A > C (p.Tyr254Ser), in the forkhead domain had better outcome and milder brain abnormalities than the other 2 patients with truncating mutation in the Groucho binding domain site, c.958delC (p.Arg320Alafs), or N-terminal domain, c.506dup (p.Lys170GlnfsThe). Importantly, all 3 patients had hypoplastic olfactory bulbs on their brain MRI, which is a distinct and previously unrecognized feature of FOXG1 syndrome.This is the first report of FOXG1 syndrome in a Korean population; this condition accounts for 2% (3 of 145 patients) of our patient cohort with developmental delays and/or hypotonia. Our report contributes to understanding this extremely rare genetic condition in the clinical and genetic perspectives.
Collapse
Affiliation(s)
- Han Na Jang
- Department of Pediatrics, Asan Medical Center Children's Hospital, University of Ulsan College of Medicine, Seoul, Korea
| | - Taeho Kim
- Biomedical Research Center, ASAN Institute for Life Sciences, Asan Medical Center, Seoul, South Korea
| | - Ah Young Jung
- Department of Radiology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Beom Hee Lee
- Department of Pediatrics, Asan Medical Center Children's Hospital, University of Ulsan College of Medicine, Seoul, Korea
| | - Mi-Sun Yum
- Department of Pediatrics, Asan Medical Center Children's Hospital, University of Ulsan College of Medicine, Seoul, Korea
| | - Tae-Sung Ko
- Department of Pediatrics, Asan Medical Center Children's Hospital, University of Ulsan College of Medicine, Seoul, Korea
| |
Collapse
|
33
|
Sharma A, Madaan P, Vyas S, Sankhyan N. FOXG1 variant presenting as unexplained irritability and peculiar crying spells. Seizure 2021; 93:32-33. [PMID: 34656939 DOI: 10.1016/j.seizure.2021.10.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Revised: 09/28/2021] [Accepted: 10/06/2021] [Indexed: 10/20/2022] Open
Affiliation(s)
- Ajay Sharma
- Pediatric Neurology Unit, Department of Pediatrics, Postgraduate Institute of Medical Education and Research (PGIMER), Chandigarh, India
| | - Priyanka Madaan
- Pediatric Neurology Unit, Department of Pediatrics, Postgraduate Institute of Medical Education and Research (PGIMER), Chandigarh, India.
| | - Sameer Vyas
- Department of Radiodiagnosis and Imaging (section of neuroimaging and interventional radiology), PGIMER, Chandigarh, India
| | - Naveen Sankhyan
- Pediatric Neurology Unit, Department of Pediatrics, Postgraduate Institute of Medical Education and Research (PGIMER), Chandigarh, India
| |
Collapse
|
34
|
Technological Improvements in the Genetic Diagnosis of Rett Syndrome Spectrum Disorders. Int J Mol Sci 2021; 22:ijms221910375. [PMID: 34638716 PMCID: PMC8508637 DOI: 10.3390/ijms221910375] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 09/17/2021] [Accepted: 09/22/2021] [Indexed: 11/17/2022] Open
Abstract
Rett syndrome (RTT) is a severe neurodevelopmental disorder that constitutes the second most common cause of intellectual disability in females worldwide. In the past few years, the advancements in genetic diagnosis brought by next generation sequencing (NGS), have made it possible to identify more than 90 causative genes for RTT and significantly overlapping phenotypes (RTT spectrum disorders). Therefore, the clinical entity known as RTT is evolving towards a spectrum of overlapping phenotypes with great genetic heterogeneity. Hence, simultaneous multiple gene testing and thorough phenotypic characterization are mandatory to achieve a fast and accurate genetic diagnosis. In this review, we revise the evolution of the diagnostic process of RTT spectrum disorders in the past decades, and we discuss the effectiveness of state-of-the-art genetic testing options, such as clinical exome sequencing and whole exome sequencing. Moreover, we introduce recent technological advancements that will very soon contribute to the increase in diagnostic yield in patients with RTT spectrum disorders. Techniques such as whole genome sequencing, integration of data from several “omics”, and mosaicism assessment will provide the tools for the detection and interpretation of genomic variants that will not only increase the diagnostic yield but also widen knowledge about the pathophysiology of these disorders.
Collapse
|
35
|
Li H, Li QQ, Hong Y. Global gene expression signatures in response to citrate-coated silver nanoparticles exposure. Toxicology 2021; 461:152898. [PMID: 34403730 DOI: 10.1016/j.tox.2021.152898] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2021] [Revised: 08/04/2021] [Accepted: 08/12/2021] [Indexed: 10/20/2022]
Abstract
Silver nanoparticles (AgNPs) are widely used in medical and commercial products for their unique antibacterial functions. However, the impact of AgNPs on human neural development is not well understood. To investigate the effect of AgNPs on human neural development, various doses of 20 nm citrate-coated AgNP (AgSC) were administered to human embryonic stem cell derived neural progenitors during the neuronal differentiation. Immunofluorescence staining with neuronal progenitor markers SOX2 (sex determining region Y-box 2) and Nestin (VI intermediate filament protein) showed that AgSC inhibited rosette formation, neuronal progenitor proliferation, and neurite outgrowth. Furthermore, AgSC promoted astrocyte activation and neuronal apoptosis. These adverse effects can be partially recovered with ascorbic acid. A genome-wide transcriptome analysis of both AgSC treated and untreated samples indicated that the most up-graduated genes were a group of Metallothionein (1F, 1E, 2A) proteins, a metal-binding protein that plays an essential role in metal homeostasis, heavy metal detoxification, and cellular anti-oxidative defence. The most significantly down-regulated genes were neuronal differentiation 6 (NEUROD6) and fork head box G1 (FOXG1). GO analyse indicated that the regulation of cholesterol biosynthetic process, neuron differentiation, synapse organization and pattern specification, oliogenesis, and neuronal apoptosis were the most impacted biological processes. KEGG pathway analyse showed that the most significantly impacted pathways were C5 isoprenoid, axon guidance, Notch, WNT, RAS-MAPK signalling pathways, lysosome, and apoptosis. Our data suggests that AgSCs interfered with metal homeostasis and cholesterol biosynthesis which induced oxidative stress, inhibited neurogenesis, axon guidance, and promoted apoptosis. Supplementation with ascorbic acid could act as an antioxidant to prevent AgSC-mediated neurotoxicity.
Collapse
Affiliation(s)
- Hao Li
- Key Laboratory of the Ministry of Education for Coastal and Wetland Ecosystem, College of the Environment and Ecology, Xiamen University, Xiamen, Fujian, 361102, China; Graduate College of Biomedical Sciences, Western University of Health Sciences, Pomona, CA, 91766-1854, USA
| | - Qingshun Q Li
- Key Laboratory of the Ministry of Education for Coastal and Wetland Ecosystem, College of the Environment and Ecology, Xiamen University, Xiamen, Fujian, 361102, China; Graduate College of Biomedical Sciences, Western University of Health Sciences, Pomona, CA, 91766-1854, USA.
| | - Yiling Hong
- College of Veterinary Medicine, Western University of Health Sciences, Pomona, CA, 91766-1854, USA.
| |
Collapse
|
36
|
Human neuropathology confirms projection neuron and interneuron defects and delayed oligodendrocyte production and maturation in FOXG1 syndrome. Eur J Med Genet 2021; 64:104282. [PMID: 34284163 DOI: 10.1016/j.ejmg.2021.104282] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 07/02/2021] [Accepted: 07/03/2021] [Indexed: 02/06/2023]
Abstract
The Forkhead transcription factor FOXG1 is a prerequisite for telencephalon development in mammals and is an essential factor controlling expansion of the dorsal telencephalon by promoting neuron and interneuron production. Heterozygous FOXG1 gene mutations cause FOXG1 syndrome characterized by severe intellectual disability, motor delay, dyskinetic movements and epilepsy. Neuroimaging studies in patients disclose constant features including microcephaly, corpus callosum dysgenesis and delayed myelination. Currently, investigative research on the underlying pathophysiology relies on mouse models only and indicates that de-repression of FOXG1 target genes may cause premature neuronal differentiation at the expense of the progenitor pool, patterning and migration defects with impaired formation of cortico-cortical projections. It remains an open question to which extent this recapitulates the neurodevelopmental pathophysiology in FOXG1-haploinsufficient patients. To close this gap, we performed neuropathological analyses in two foetal cases with FOXG1 premature stop codon mutations interrupted during the third trimester of the pregnancy for microcephaly and corpus callosum dysgenesis. In these foetuses, we observed cortical lamination defects and decreased neuronal density mainly affecting layers II, III and V that normally give rise to cortico-cortical and inter-hemispheric axonal projections. GABAergic interneurons were also reduced in number in the cortical plate and persisting germinative zones. Additionally, we observed more numerous PDGFRα-positive oligodendrocyte precursor cells and fewer Olig2-positive pre-oligodendrocytes compared to age-matched control brains, arguing for delayed production and differentiation of oligodendrocyte lineage leading to delayed myelination. These findings provide key insights into the human pathophysiology of FOXG1 syndrome.
Collapse
|
37
|
Santos-Terra J, Deckmann I, Fontes-Dutra M, Schwingel GB, Bambini-Junior V, Gottfried C. Transcription factors in neurodevelopmental and associated psychiatric disorders: A potential convergence for genetic and environmental risk factors. Int J Dev Neurosci 2021; 81:545-578. [PMID: 34240460 DOI: 10.1002/jdn.10141] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Revised: 06/23/2021] [Accepted: 07/02/2021] [Indexed: 12/16/2022] Open
Abstract
Neurodevelopmental disorders (NDDs) are a heterogeneous and highly prevalent group of psychiatric conditions marked by impairments in the nervous system. Their onset occurs during gestation, and the alterations are observed throughout the postnatal life. Although many genetic and environmental risk factors have been described in this context, the interactions between them challenge the understanding of the pathways associated with NDDs. Transcription factors (TFs)-a group of over 1,600 proteins that can interact with DNA, regulating gene expression through modulation of RNA synthesis-represent a point of convergence for different risk factors. In addition, TFs organize critical processes like angiogenesis, blood-brain barrier formation, myelination, neuronal migration, immune activation, and many others in a time and location-dependent way. In this review, we summarize important TF alterations in NDD and associated disorders, along with specific impairments observed in animal models, and, finally, establish hypotheses to explain how these proteins may be critical mediators in the context of genome-environment interactions.
Collapse
Affiliation(s)
- Júlio Santos-Terra
- Translational Research Group in Autism Spectrum Disorders (GETTEA), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil.,Department of Biochemistry, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil.,National Institute of Science and Technology on Neuroimmunomodulation (INCT-NIM), Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil.,School of Pharmacology and Biomedical Sciences, University of Central Lancashire, Autism Wellbeing And Research Development (AWARD) Institute, BR-UK-CA, Preston, UK
| | - Iohanna Deckmann
- Translational Research Group in Autism Spectrum Disorders (GETTEA), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil.,Department of Biochemistry, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil.,National Institute of Science and Technology on Neuroimmunomodulation (INCT-NIM), Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil.,School of Pharmacology and Biomedical Sciences, University of Central Lancashire, Autism Wellbeing And Research Development (AWARD) Institute, BR-UK-CA, Preston, UK
| | - Mellanie Fontes-Dutra
- Translational Research Group in Autism Spectrum Disorders (GETTEA), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil.,Department of Biochemistry, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil.,National Institute of Science and Technology on Neuroimmunomodulation (INCT-NIM), Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil.,School of Pharmacology and Biomedical Sciences, University of Central Lancashire, Autism Wellbeing And Research Development (AWARD) Institute, BR-UK-CA, Preston, UK
| | - Gustavo Brum Schwingel
- Translational Research Group in Autism Spectrum Disorders (GETTEA), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil.,Department of Biochemistry, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil.,National Institute of Science and Technology on Neuroimmunomodulation (INCT-NIM), Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil.,School of Pharmacology and Biomedical Sciences, University of Central Lancashire, Autism Wellbeing And Research Development (AWARD) Institute, BR-UK-CA, Preston, UK
| | - Victorio Bambini-Junior
- Translational Research Group in Autism Spectrum Disorders (GETTEA), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil.,National Institute of Science and Technology on Neuroimmunomodulation (INCT-NIM), Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil.,School of Pharmacology and Biomedical Sciences, University of Central Lancashire, Autism Wellbeing And Research Development (AWARD) Institute, BR-UK-CA, Preston, UK.,School of Pharmacology and Biomedical Sciences, University of Central Lancashire, Preston, UK
| | - Carmem Gottfried
- Translational Research Group in Autism Spectrum Disorders (GETTEA), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil.,Department of Biochemistry, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil.,National Institute of Science and Technology on Neuroimmunomodulation (INCT-NIM), Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil.,School of Pharmacology and Biomedical Sciences, University of Central Lancashire, Autism Wellbeing And Research Development (AWARD) Institute, BR-UK-CA, Preston, UK
| |
Collapse
|
38
|
Miyoshi G, Ueta Y, Natsubori A, Hiraga K, Osaki H, Yagasaki Y, Kishi Y, Yanagawa Y, Fishell G, Machold RP, Miyata M. FoxG1 regulates the formation of cortical GABAergic circuit during an early postnatal critical period resulting in autism spectrum disorder-like phenotypes. Nat Commun 2021; 12:3773. [PMID: 34145239 PMCID: PMC8213811 DOI: 10.1038/s41467-021-23987-z] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2020] [Accepted: 05/27/2021] [Indexed: 01/02/2023] Open
Abstract
Abnormalities in GABAergic inhibitory circuits have been implicated in the aetiology of autism spectrum disorder (ASD). ASD is caused by genetic and environmental factors. Several genes have been associated with syndromic forms of ASD, including FOXG1. However, when and how dysregulation of FOXG1 can result in defects in inhibitory circuit development and ASD-like social impairments is unclear. Here, we show that increased or decreased FoxG1 expression in both excitatory and inhibitory neurons results in ASD-related circuit and social behavior deficits in our mouse models. We observe that the second postnatal week is the critical period when regulation of FoxG1 expression is required to prevent subsequent ASD-like social impairments. Transplantation of GABAergic precursor cells prior to this critical period and reduction in GABAergic tone via Gad2 mutation ameliorates and exacerbates circuit functionality and social behavioral defects, respectively. Our results provide mechanistic insight into the developmental timing of inhibitory circuit formation underlying ASD-like phenotypes in mouse models.
Collapse
Affiliation(s)
- Goichi Miyoshi
- Department of Neurophysiology, Tokyo Women's Medical University, Shinjuku, Tokyo, Japan.
| | - Yoshifumi Ueta
- Department of Neurophysiology, Tokyo Women's Medical University, Shinjuku, Tokyo, Japan
| | - Akiyo Natsubori
- Sleep Disorders Project, Tokyo Metropolitan Institute of Medical Science, Setagaya, Tokyo, Japan
| | - Kou Hiraga
- Department of Neurophysiology, Tokyo Women's Medical University, Shinjuku, Tokyo, Japan
| | - Hironobu Osaki
- Department of Neurophysiology, Tokyo Women's Medical University, Shinjuku, Tokyo, Japan
| | - Yuki Yagasaki
- Department of Neurophysiology, Tokyo Women's Medical University, Shinjuku, Tokyo, Japan
| | - Yusuke Kishi
- Graduate School of Pharmaceutical Sciences, University of Tokyo, Tokyo, Japan
| | - Yuchio Yanagawa
- Department of Genetic and Behavioral Neuroscience, Gunma University Graduate School of Medicine, Maebashi, Japan
| | - Gord Fishell
- NYU Neuroscience Institute, Smilow Research Center, New York University School of Medicine, New York, NY, USA
- Department of Neurobiology, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
- Stanley Center at the Broad Institute, Cambridge, MA, USA
| | - Robert P Machold
- NYU Neuroscience Institute, Smilow Research Center, New York University School of Medicine, New York, NY, USA
| | - Mariko Miyata
- Department of Neurophysiology, Tokyo Women's Medical University, Shinjuku, Tokyo, Japan
| |
Collapse
|
39
|
Pecora G, Sortino V, Brafa Musicoro V, Salomone G, Pizzo F, Costanza G, Falsaperla R, Zanghì A, Praticò AD. FOXG1 Gene and Its Related Phenotypes. JOURNAL OF PEDIATRIC NEUROLOGY 2021. [DOI: 10.1055/s-0041-1727270] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
AbstractFOXG1 is an important transcriptional repressor found in cell precursor of the ventricular region and in neurons in the early stage of differentiation during the development of the nervous epithelium in the cerebrum and optical formation. Mutations involving FOXG1 gene have been described first in subjects with congenital Rett syndrome. They can cause seizure, delayed psychomotor development, language disorders, and autism. FOXG1 deletions or intragenic mutations also determinate reduction in head circumference, structural defects in the corpus callosum, abnormal movements, especially choreiform, and intellectual retardation with no speech. Patients with duplications of 14q12 present infantile spasms and have subsequent intellectual disability with autistic features, head circumference in the normal range, and regular aspect of corpus callosum. Clinical characteristics of patients with FOXG1 variants include growth deficit after birth associated with microcephaly, facial dysmorphisms, important delay with no language, deficit in social interaction like autism, sleep disorders, stereotypes, including dyskinesia, and seizures. In these patients, it is not characteristic a history of loss of acquired skills.
Collapse
Affiliation(s)
- Giulia Pecora
- Pediatric Postgraduate Residency Program, Section of Pediatrics and Child Neuropsychiatry, Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | - Vincenzo Sortino
- Pediatric Postgraduate Residency Program, Section of Pediatrics and Child Neuropsychiatry, Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | - Viviana Brafa Musicoro
- Pediatric Postgraduate Residency Program, Section of Pediatrics and Child Neuropsychiatry, Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | - Giulia Salomone
- Pediatric Postgraduate Residency Program, Section of Pediatrics and Child Neuropsychiatry, Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | - Francesco Pizzo
- Pediatric Postgraduate Residency Program, Section of Pediatrics and Child Neuropsychiatry, Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | - Giuseppe Costanza
- Pediatric Postgraduate Residency Program, Section of Pediatrics and Child Neuropsychiatry, Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | - Raffaele Falsaperla
- Unit of Pediatrics and Pediatric Emergency, University Hospital “Policlinico Rodolico-San Marco,” Catania, Italy
- Unit of Neonatal Intensive Care and Neonatology, University Hospital “Policlinico Rodolico-San Marco,” Catania, Italy
| | - Antonio Zanghì
- Department of Medical and Surgical Sciences and Advanced Technology “G.F. Ingrassia,” University of Catania, Catania, Italy
| | - Andrea D. Praticò
- Unit of Rare Diseases of the Nervous System in Childhood, Department of Clinical and Experimental Medicine, Section of Pediatrics and Child Neuropsychiatry, University of Catania, Catania, Italy
| |
Collapse
|
40
|
Li X, Kong Y, Li H, Xu M, Jiang M, Sun W, Xu S. CircRNA circ_0067772 aggravates the malignant progression of cutaneous squamous cell carcinoma by regulating miR-1238-3p/FOXG1 axis. Genes Genomics 2021; 43:491-501. [PMID: 33709381 DOI: 10.1007/s13258-021-01074-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2020] [Accepted: 03/01/2021] [Indexed: 02/07/2023]
Abstract
BACKGROUND Cutaneous squamous cell carcinoma (CSCC) is a severe malignancy derived from skin. Dysregulated circular RNAs (circRNAs) might play vital roles in tumor development. OBJECTIVE Here, we aimed to explore the function of a novel circRNA circ_0067772 in CSCC. METHODS Quantitative real-time PCR (qRT-PCR) or Western blot assay was performed to determine the expression of circ_0067772, microRNA (miR)-1238-3p and forkhead box protein G1 (FOXG1). Cell proliferation was assessed by Cell Counting Kit-8 (CCK-8) assay and colony formation assay. Transwell assay and wound healing assay were employed to examine cell metastasis. Flow cytometry was employed to monitor cell cycle and apoptosis. The target association between miR-1238-3p and circ_0067772 or FOXG1 was validated by dual-luciferase reporter assay. Moreover, role of circ_0067772 in vivo was investigated via xenograft model in nude mice. RESULTS Circ_0067772 and FOXG1 were upregulated, while miR-1238-3p was downregulated in CSCC tissues and cells. Circ_0067772 knockdown conferred inhibitory effects on cell proliferation, migration and invasion of CSCC cells. MiR-1238-3p served as a target of circ_0067772, whose silencing could reverse circ_0067772 knockdown-induced inhibitory impact on the malignant cellular behaviors. Circ_0067772 positively regulated FOXG1 expression by antagonizing miR-1238-3p. Additionally, miR-1238-3p could repress CSCC cell proliferation, migration and invasion by targeting FOXG1. Also, circ_0067772 knockdown hindered CSCC tumor growth in vivo. CONCLUSION Our study identified a novel oncogenic circRNA and the involvement of circ_0067772/miR-1238-3p/FOXG1 axis in CSCC development, providing a target for CSCC therapy.
Collapse
Affiliation(s)
- Xiaoqing Li
- Department of Dermatology, The Affiliated Huaian NO.1 People's Hospital of Nanjing Medical University, No. 6, West Beijing Road, Huaiyin District, Huaian, 223300, Jiangsu, China.
| | - Yinghui Kong
- Department of Dermatology, The Affiliated Huaian NO.1 People's Hospital of Nanjing Medical University, No. 6, West Beijing Road, Huaiyin District, Huaian, 223300, Jiangsu, China
| | - He Li
- Department of Dermatology, The Affiliated Huaian NO.1 People's Hospital of Nanjing Medical University, No. 6, West Beijing Road, Huaiyin District, Huaian, 223300, Jiangsu, China
| | - Manyuan Xu
- Department of Dermatology, The Affiliated Huaian NO.1 People's Hospital of Nanjing Medical University, No. 6, West Beijing Road, Huaiyin District, Huaian, 223300, Jiangsu, China
| | - Ming Jiang
- Department of Dermatology, The Affiliated Huaian NO.1 People's Hospital of Nanjing Medical University, No. 6, West Beijing Road, Huaiyin District, Huaian, 223300, Jiangsu, China
| | - Weiguo Sun
- Department of Dermatology, The Affiliated Huaian NO.1 People's Hospital of Nanjing Medical University, No. 6, West Beijing Road, Huaiyin District, Huaian, 223300, Jiangsu, China
| | - Suping Xu
- Department of Dermatology, The Affiliated Huaian NO.1 People's Hospital of Nanjing Medical University, No. 6, West Beijing Road, Huaiyin District, Huaian, 223300, Jiangsu, China
| |
Collapse
|
41
|
Akaba Y, Takahashi S, Takeguchi R, Tanaka R, Nabatame S, Saitsu H, Matsumoto N. Phenotypic overlap between pyruvate dehydrogenase complex deficiency and FOXG1 syndrome. Clin Case Rep 2021; 9:1711-1715. [PMID: 33768920 PMCID: PMC7981633 DOI: 10.1002/ccr3.3883] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2020] [Revised: 01/12/2021] [Accepted: 01/20/2021] [Indexed: 11/12/2022] Open
Abstract
Pyruvate dehydrogenase complex (PDHC) deficiency is a mitochondrial disorder. We report two cases of PDHC deficiency with clinical symptoms and brain imaging findings reminiscent of FOXG1 syndrome, suggesting a phenotypic overlap of these disorders.
Collapse
Affiliation(s)
- Yuichi Akaba
- Department of PediatricsAsahikawa Medical UniversityAsahikawaJapan
| | - Satoru Takahashi
- Department of PediatricsAsahikawa Medical UniversityAsahikawaJapan
| | - Ryo Takeguchi
- Department of PediatricsAsahikawa Medical UniversityAsahikawaJapan
| | - Ryosuke Tanaka
- Department of PediatricsAsahikawa Medical UniversityAsahikawaJapan
| | - Shin Nabatame
- Department of PediatricsGraduate School of MedicineOsaka UniversityOsakaJapan
| | - Hirotomo Saitsu
- Department of BiochemistryHamamatsu University School of MedicineHamamatsuJapan
| | - Naomichi Matsumoto
- Department of Human GeneticsGraduate School of MedicineYokohama City UniversityYokohamaJapan
| |
Collapse
|
42
|
D'Mello SR. MECP2 and the Biology of MECP2 Duplication Syndrome. J Neurochem 2021; 159:29-60. [PMID: 33638179 DOI: 10.1111/jnc.15331] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 01/21/2021] [Accepted: 02/18/2021] [Indexed: 11/27/2022]
Abstract
MECP2 duplication syndrome (MDS), a rare X-linked genomic disorder affecting predominantly males, is caused by duplication of the chromosomal region containing the methyl CpG binding protein-2 (MECP2) gene, which encodes methyl-CpG-binding protein 2 (MECP2), a multi-functional protein required for proper brain development and maintenance of brain function during adulthood. Disease symptoms include severe motor and cognitive impairment, delayed or absent speech development, autistic features, seizures, ataxia, recurrent respiratory infections and shortened lifespan. The cellular and molecular mechanisms by which a relatively modest increase in MECP2 protein causes such severe disease symptoms are poorly understood and consequently there are no treatments available for this fatal disorder. This review summarizes what is known to date about the structure and complex regulation of MECP2 and its many functions in the developing and adult brain. Additionally, recent experimental findings on the cellular and molecular underpinnings of MDS based on cell culture and mouse models of the disorder are reviewed. The emerging picture from these studies is that MDS is a neurodegenerative disorder in which neurons die in specific parts of the central nervous system, including the cortex, hippocampus, cerebellum and spinal cord. Neuronal death likely results from astrocytic dysfunction, including a breakdown of glutamate homeostatic mechanisms. The role of elevations in the expression of glial acidic fibrillary protein (GFAP) in astrocytes and the microtubule-associated protein, Tau, in neurons to the pathogenesis of MDS is discussed. Lastly, potential therapeutic strategies to potentially treat MDS are discussed.
Collapse
|
43
|
Fardghassemi Y, Parker JA. Overexpression of FKH-2/FOXG1 is neuroprotective in a C. elegans model of Machado-Joseph disease. Exp Neurol 2020; 337:113544. [PMID: 33290777 DOI: 10.1016/j.expneurol.2020.113544] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2020] [Revised: 11/27/2020] [Accepted: 12/02/2020] [Indexed: 12/12/2022]
Abstract
Machado-Joseph disease (MJD), also known as spinocerebellar ataxia type 3 (SCA3), is the most common form of dominantly inherited ataxia worldwide. This disease is caused by an expanded CAG repeat in the coding region of ATXN3. Due to our incomplete understanding of mechanisms and molecular pathways related to this disease, there are no therapies that successfully treat core MJD patients. Therefore, the identification of new candidate targets related to this disease is needed. In this study, we performed a large-scale RNA interference (RNAi) screen of 387 transcription factor genes leading to the identification of several modifiers (suppressors and enhancers) of impaired motility phenotypes in a mutant ATXN3 transgenic C. elegans model. We showed that inactivation of one particular gene, fkh-2/FOXG1, enhanced the motility defect, neurodegeneration and reduced longevity in our MJD models. Opposite to genetic inactivation, the overexpression of fkh-2 rescued the impaired motility, shortened-lifespan, and neurodegeneration phenotypes of mutant ATXN3 transgenics. We found that overexpression of FKH-2/FOXG1 in ATXN3 mutant worms is neuroprotective. Using our transgenic ATXN3 C. elegans models and the screening of an RNAi library, we gained insights into the pathways contributing to neurodegeneration, and found that FKH-2/FOXG1 has neuroprotective activity. These findings may aid the development of novel therapeutic interventions for MJD.
Collapse
Affiliation(s)
- Yasmin Fardghassemi
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), 900 St-Denis Street, Montreal, Quebec H2X 0A9, Canada; Department of Biochemistry, University of Montreal, 2900 Edouard Montpetit Blvd, Montreal, Quebec H3T 1J4, Canada
| | - J Alex Parker
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), 900 St-Denis Street, Montreal, Quebec H2X 0A9, Canada; Department of Biochemistry, University of Montreal, 2900 Edouard Montpetit Blvd, Montreal, Quebec H3T 1J4, Canada; Department of Neuroscience, University of Montreal, 2900 Edouard Montpetit Blvd, Montreal, Quebec H3T 1J4, Canada.
| |
Collapse
|
44
|
Waite AJ, Millar D, Clarke A. The generation of an induced pluripotent stem cell line (DCGi001-A) from an individual with FOXG1 syndrome carrying the c.460dupG (p.Glu154fs) variation in the FOXG1 gene. Stem Cell Res 2020; 49:102018. [PMID: 33096386 DOI: 10.1016/j.scr.2020.102018] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Revised: 09/21/2020] [Accepted: 09/27/2020] [Indexed: 11/25/2022] Open
Abstract
FOXG1 syndrome is a neurodevelopmental disorder caused by mutations in the FOXG1 gene. Here, an induced pluripotent stem cell (iPSC) line was generated from human dermal fibroblasts of an individual with the c.490dupG (p.Glu154fs) mutation in the FOXG1 gene. Fibroblasts were reprogrammed using non-integrating episomal plasmids and pluripotency marker expression was confirmed by both immunocytochemistry and quantitative PCR in the resultant iPSC line. There were no karyotypic abnormalities and the cell line successfully differentiated into all three germ layers. This cell line may prove useful in the study of the pathogenic mechanisms that underpin FOXG1 syndrome.
Collapse
Affiliation(s)
- Adrian J Waite
- Division of Cancer and Genetics, Cardiff University School of Medicine, Institute of Medical Genetics Building, Heath Park, Cardiff, Wales CF14 4XN, United Kingdom
| | - David Millar
- Division of Cancer and Genetics, Cardiff University School of Medicine, Institute of Medical Genetics Building, Heath Park, Cardiff, Wales CF14 4XN, United Kingdom
| | - Angus Clarke
- Division of Cancer and Genetics, Cardiff University School of Medicine, Institute of Medical Genetics Building, Heath Park, Cardiff, Wales CF14 4XN, United Kingdom.
| |
Collapse
|
45
|
Dai S, Li J, Zhang H, Chen X, Guo M, Chen Z, Chen Y. Structural Basis for DNA Recognition by FOXG1 and the Characterization of Disease-causing FOXG1 Mutations. J Mol Biol 2020; 432:6146-6156. [PMID: 33058871 DOI: 10.1016/j.jmb.2020.10.007] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 09/22/2020] [Accepted: 10/07/2020] [Indexed: 12/22/2022]
Abstract
Forkhead box G1 (FOXG1) is a transcription factor mainly expressed in the brain that plays a critical role in the development and regionalization of the forebrain. Aberrant expression of FOXG1 has implications in FOXG1 syndrome, a serious neurodevelopmental disorder. Here, we report the crystal structure of the FOXG1 DNA-binding domain (DBD) in complex with the forkhead consensus DNA site DBE2 at the resolution of 1.6 Å. FOXG1-DBD adopts a typical winged helix fold. Compared to those of other FOX-DBD/DBE2 structures, the N terminus, H3 helix and wing2 region of FOXG1-DBD exhibit differences in DNA recognition. The FOXG1-DBD wing2 region adopts a unique architecture composed of two β-strands that differs from all other known FOX-DBD wing2 folds. Mutation assays revealed that the disease-causing mutations within the FOXG1-DBD affect DNA binding, protein thermal stability, or both. Our report provides initial insight into how FOXG1 binds DNA and sheds light on how disease-causing mutations in FOXG1-DBD affect its DNA-binding ability.
Collapse
Affiliation(s)
- Shuyan Dai
- Department of Oncology, NHC Key Laboratory of Cancer Proteomics, Laboratory of Structural Biology, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Jun Li
- Department of Oncology, NHC Key Laboratory of Cancer Proteomics, Laboratory of Structural Biology, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China.
| | - Huajun Zhang
- Department of Oncology, NHC Key Laboratory of Cancer Proteomics, Laboratory of Structural Biology, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Xiaojuan Chen
- Department of Oncology, NHC Key Laboratory of Cancer Proteomics, Laboratory of Structural Biology, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Ming Guo
- Department of Oncology, NHC Key Laboratory of Cancer Proteomics, Laboratory of Structural Biology, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Zhuchu Chen
- Department of Oncology, NHC Key Laboratory of Cancer Proteomics, Laboratory of Structural Biology, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Yongheng Chen
- Department of Oncology, NHC Key Laboratory of Cancer Proteomics, Laboratory of Structural Biology, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China.
| |
Collapse
|
46
|
The FOXG1-related syndrome with two novel mutations in the FOXG1 gene. GENE REPORTS 2020. [DOI: 10.1016/j.genrep.2020.100723] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
47
|
Fahmi M, Yasui G, Seki K, Katayama S, Kaneko-Kawano T, Inazu T, Kubota Y, Ito M. In Silico Study of Rett Syndrome Treatment-Related Genes, MECP2, CDKL5, and FOXG1, by Evolutionary Classification and Disordered Region Assessment. Int J Mol Sci 2019; 20:ijms20225593. [PMID: 31717404 PMCID: PMC6888432 DOI: 10.3390/ijms20225593] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 10/31/2019] [Accepted: 11/05/2019] [Indexed: 12/28/2022] Open
Abstract
Rett syndrome (RTT), a neurodevelopmental disorder, is mainly caused by mutations in methyl CpG-binding protein 2 (MECP2), which has multiple functions such as binding to methylated DNA or interacting with a transcriptional co-repressor complex. It has been established that alterations in cyclin-dependent kinase-like 5 (CDKL5) or forkhead box protein G1 (FOXG1) correspond to distinct neurodevelopmental disorders, given that a series of studies have indicated that RTT is also caused by alterations in either one of these genes. We investigated the evolution and molecular features of MeCP2, CDKL5, and FOXG1 and their binding partners using phylogenetic profiling to gain a better understanding of their similarities. We also predicted the structural order-disorder propensity and assessed the evolutionary rates per site of MeCP2, CDKL5, and FOXG1 to investigate the relationships between disordered structure and other related properties with RTT. Here, we provide insight to the structural characteristics, evolution and interaction landscapes of those three proteins. We also uncovered the disordered structure properties and evolution of those proteins which may provide valuable information for the development of therapeutic strategies of RTT.
Collapse
Affiliation(s)
- Muhamad Fahmi
- Advanced Life Sciences Program, Graduate School of Life Sciences, Ritsumeikan University, Kusatsu, Shiga 525-8577, Japan; (M.F.); (G.Y.); (K.S.)
| | - Gen Yasui
- Advanced Life Sciences Program, Graduate School of Life Sciences, Ritsumeikan University, Kusatsu, Shiga 525-8577, Japan; (M.F.); (G.Y.); (K.S.)
| | - Kaito Seki
- Advanced Life Sciences Program, Graduate School of Life Sciences, Ritsumeikan University, Kusatsu, Shiga 525-8577, Japan; (M.F.); (G.Y.); (K.S.)
| | - Syouichi Katayama
- Department of Pharmacy, College of Pharmaceutical Sciences, Ritsumeikan University, Kusatsu, Shiga 525-8577, Japan; (S.K.); (T.K.-K.); (T.I.)
| | - Takako Kaneko-Kawano
- Department of Pharmacy, College of Pharmaceutical Sciences, Ritsumeikan University, Kusatsu, Shiga 525-8577, Japan; (S.K.); (T.K.-K.); (T.I.)
| | - Tetsuya Inazu
- Department of Pharmacy, College of Pharmaceutical Sciences, Ritsumeikan University, Kusatsu, Shiga 525-8577, Japan; (S.K.); (T.K.-K.); (T.I.)
| | - Yukihiko Kubota
- Department of Bioinformatics, College of Life Sciences, Ritsumeikan University, Kusatsu, Shiga 525-8577, Japan;
| | - Masahiro Ito
- Advanced Life Sciences Program, Graduate School of Life Sciences, Ritsumeikan University, Kusatsu, Shiga 525-8577, Japan; (M.F.); (G.Y.); (K.S.)
- Department of Bioinformatics, College of Life Sciences, Ritsumeikan University, Kusatsu, Shiga 525-8577, Japan;
- Correspondence:
| |
Collapse
|