1
|
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
|
2
|
Liuzzi G, Artimagnella O, Frisari S, Mallamaci A. Foxg1 bimodally tunes L1-mRNA and -DNA dynamics in the developing murine neocortex. Development 2024; 151:dev202292. [PMID: 38655654 PMCID: PMC11190451 DOI: 10.1242/dev.202292] [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: 08/23/2023] [Accepted: 04/16/2024] [Indexed: 04/26/2024]
Abstract
Foxg1 masters telencephalic development via a pleiotropic control over its progression. Expressed within the central nervous system (CNS), L1 retrotransposons are implicated in progression of its histogenesis and tuning of its genomic plasticity. Foxg1 represses gene transcription, and L1 elements share putative Foxg1-binding motifs, suggesting the former might limit telencephalic expression (and activity) of the latter. We tested such a prediction, in vivo as well as in engineered primary neural cultures, using loss- and gain-of-function approaches. We found that Foxg1-dependent, transcriptional L1 repression specifically occurs in neopallial neuronogenic progenitors and post-mitotic neurons, where it is supported by specific changes in the L1 epigenetic landscape. Unexpectedly, we discovered that Foxg1 physically interacts with L1-mRNA and positively regulates neonatal neopallium L1-DNA content, antagonizing the retrotranscription-suppressing activity exerted by Mov10 and Ddx39a helicases. To the best of our knowledge, Foxg1 represents the first CNS patterning gene acting as a bimodal retrotransposon modulator, limiting transcription of L1 elements and promoting their amplification, within a specific domain of the developing mouse brain.
Collapse
Affiliation(s)
- Gabriele Liuzzi
- Laboratory of Cerebral Cortex Development, SISSA, Trieste 34136, Italy
| | | | - Simone Frisari
- Laboratory of Cerebral Cortex Development, SISSA, Trieste 34136, Italy
| | | |
Collapse
|
3
|
Costa M, Vannini E. Cellular and Molecular Mechanisms in Neurodevelopmental Disorders and Brain Tumors. Int J Mol Sci 2023; 24:ijms24119469. [PMID: 37298419 DOI: 10.3390/ijms24119469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Revised: 05/18/2023] [Accepted: 05/19/2023] [Indexed: 06/12/2023] Open
Abstract
The normal growth and operation of the central nervous system (CNS) at all stages of development, including adulthood, depend on the interaction between intrinsic and extrinsic factors [...].
Collapse
Affiliation(s)
- Mario Costa
- Neuroscience Institute, National Research Council (CNR), Via G. Moruzzi 1, 56124 Pisa, Italy
- Centro Pisano Ricerca e Implementazione Clinica Flash Radiotherapy "CPFR@CISUP", "S. Chiara" Hospital, 56124 Pisa, Italy
- Laboratory of Biology BIO@SNS, Scuola Normale Superiore, Piazza di Cavalieri 7, 56124 Pisa, Italy
| | - Eleonora Vannini
- Neuroscience Institute, National Research Council (CNR), Via G. Moruzzi 1, 56124 Pisa, Italy
- Centro Pisano Ricerca e Implementazione Clinica Flash Radiotherapy "CPFR@CISUP", "S. Chiara" Hospital, 56124 Pisa, Italy
| |
Collapse
|
4
|
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
|
5
|
Xi H, He Z, Lv C. FOXG1 improves mitochondrial function and promotes the progression of nasopharyngeal carcinoma. Mol Med Rep 2021; 24:651. [PMID: 34278485 PMCID: PMC8299199 DOI: 10.3892/mmr.2021.12290] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2020] [Accepted: 05/14/2021] [Indexed: 12/28/2022] Open
Abstract
Forkhead‑box gene 1 (FOXG1) has been reported to serve an important role in various malignancies, but its effects on nasopharyngeal cancer (NPC) remain unknown. Thus, the present study aimed to investigate the specific regulatory relationship between FOXG1 and NPC progression. Tumor tissues and matching para‑carcinoma tissues were obtained from patients with NPC. Small interfering (si)RNA‑FOXG1 and pcDNA3.1‑FOXG1 were transfected into SUNE‑1 and C666‑1 cells to knockdown and overexpress FOXG1 expression, respectively. FOXG1 expression was detected using reverse transcription‑quantitative PCR and immunohistochemistry. Cell proliferation was detected using MTT and 5‑ethynyl‑20‑deoxyuridine assays. Transwell invasion assay, wound healing assay and flow cytometry were used to detect cell invasion, migration and apoptosis, respectively. Western blotting was conducted to detect the expression levels of mitochondrial markers (succinate dehydrogenase complex flavoprotein subunit A, heat shock protein 60 and pyruvate dehydrogenase), epithelial‑mesenchymal transition (EMT) related proteins (N‑cadherin, Snail and E‑cadherin) and apoptosis‑related proteins [Bax, Bcl‑2, poly(ADP‑ribose) polymerase 1 (PARP), cleaved PARP, cleaved caspase‑3, cleaved caspase‑8, cleaved caspase‑9, caspase‑3, caspase‑8 and caspase‑9]. The mitochondrial membrane potential was detected via flow cytometry, while the ATP/ADP ratio was determined using the ADP/ATP ratio assay kit. The present results demonstrated that FOXG1 expression was upregulated in NPC tissues and cells, and was associated with distant metastasis and TNM stage. Moreover, knockdown of FOXG1 inhibited the proliferation, migration, invasion, EMT and mitochondrial function of SUNE‑1 cells, as well as promoted cell apoptosis, while the opposite results were observed in C666‑1 cells. In conclusion, FOXG1 enhanced proliferation, migration and invasion, induced EMT and improved mitochondrial function in NPC cells. The current findings provide an adequate theoretical basis for the treatment of NPC.
Collapse
Affiliation(s)
- Huajun Xi
- Department of Otolaryngology and Stomatology, Shouguang People's Hospital, Shouguang, Shandong 262700, P.R. China
| | - Zhengxiang He
- Department of Otolaryngology and Maxillofacial Surgery, Wuwei People's Hospital, Wuwei, Gansu 733000, P.R. China
| | - Cao Lv
- Department of Otolaryngology, Head and Neck Surgery, The Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650101, P.R. China
| |
Collapse
|
6
|
De Masi C, Spitalieri P, Murdocca M, Novelli G, Sangiuolo F. Application of CRISPR/Cas9 to human-induced pluripotent stem cells: from gene editing to drug discovery. Hum Genomics 2020; 14:25. [PMID: 32591003 PMCID: PMC7318728 DOI: 10.1186/s40246-020-00276-2] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Accepted: 06/17/2020] [Indexed: 12/12/2022] Open
Abstract
Human-induced pluripotent stem cells (hiPSCs) and CRISPR/Cas9 gene editing system represent two instruments of basic and translational research, which both allow to acquire deep insight about the molecular bases of many diseases but also to develop pharmacological research.This review is focused to draw up the latest technique of gene editing applied on hiPSCs, exploiting some of the genetic manipulation directed to the discovery of innovative therapeutic strategies. There are many expediencies provided by the use of hiPSCs, which can represent a disease model clinically relevant and predictive, with a great potential if associated to CRISPR/Cas9 technology, a gene editing tool powered by ease and precision never seen before.Here, we describe the possible applications of CRISPR/Cas9 to hiPSCs: from drug development to drug screening and from gene therapy to the induction of the immunological response to specific virus infection, such as HIV and SARS-Cov-2.
Collapse
Affiliation(s)
- Claudia De Masi
- Department of Biomedicine and Prevention, University of Rome "Tor Vergata", Rome, Italy
| | - Paola Spitalieri
- Department of Biomedicine and Prevention, University of Rome "Tor Vergata", Rome, Italy
| | - Michela Murdocca
- Department of Biomedicine and Prevention, University of Rome "Tor Vergata", Rome, Italy
| | - Giuseppe Novelli
- Department of Biomedicine and Prevention, University of Rome "Tor Vergata", Rome, Italy
| | - Federica Sangiuolo
- Department of Biomedicine and Prevention, University of Rome "Tor Vergata", Rome, Italy.
| |
Collapse
|
7
|
Croci S, Carriero ML, Capitani K, Daga S, Donati F, Papa FT, Frullanti E, Lopergolo D, Lamacchia V, Tita R, Giliberti A, Benetti E, Niccheri F, Furini S, Lo Rizzo C, Conticello SG, Renieri A, Meloni I. AAV-mediated FOXG1 gene editing in human Rett primary cells. Eur J Hum Genet 2020; 28:1446-1458. [PMID: 32541681 PMCID: PMC7608362 DOI: 10.1038/s41431-020-0652-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2019] [Revised: 04/16/2020] [Accepted: 04/24/2020] [Indexed: 12/21/2022] Open
Abstract
Variations in the Forkhead Box G1 (FOXG1) gene cause FOXG1 syndrome spectrum, including the congenital variant of Rett syndrome, characterized by early onset of regression, Rett-like and jerky movements, and cortical visual impairment. Due to the largely unknown pathophysiological mechanisms downstream the impairment of this transcriptional regulator, a specific treatment is not yet available. Since both haploinsufficiency and hyper-expression of FOXG1 cause diseases in humans, we reasoned that adding a gene under nonnative regulatory sequences would be a risky strategy as opposed to a genome editing approach where the mutated gene is reversed into wild-type. Here, we demonstrate that an adeno-associated viruses (AAVs)-coupled CRISPR/Cas9 system is able to target and correct FOXG1 variants in patient-derived fibroblasts, induced Pluripotent Stem Cells (iPSCs) and iPSC-derived neurons. Variant-specific single-guide RNAs (sgRNAs) and donor DNAs have been selected and cloned together with a mCherry/EGFP reporter system. Specific sgRNA recognition sequences were inserted upstream and downstream Cas9 CDS to allow self-cleavage and inactivation. We demonstrated that AAV serotypes vary in transduction efficiency depending on the target cell type, the best being AAV9 in fibroblasts and iPSC-derived neurons, and AAV2 in iPSCs. Next-generation sequencing (NGS) of mCherry+/EGFP+ transfected cells demonstrated that the mutated alleles were repaired with high efficiency (20–35% reversion) and precision both in terms of allelic discrimination and off-target activity. The genome editing strategy tested in this study has proven to precisely repair FOXG1 and delivery through an AAV9-based system represents a step forward toward the development of a therapy for Rett syndrome.
Collapse
Affiliation(s)
| | | | - Katia Capitani
- Medical Genetics, University of Siena, Siena, Italy.,Molecular Mechanisms of Oncogenesis, ISPRO Core Research Laboratory (CRL), Firenze, Italy
| | - Sergio Daga
- Medical Genetics, University of Siena, Siena, Italy
| | - Francesco Donati
- Medical Genetics, University of Siena, Siena, Italy.,Molecular Mechanisms of Oncogenesis, ISPRO Core Research Laboratory (CRL), Firenze, Italy
| | | | | | - Diego Lopergolo
- Medical Genetics, University of Siena, Siena, Italy.,Genetica Medica, Azienda Ospedaliera Universitaria Senese, Siena, Italy
| | - Vittoria Lamacchia
- Medical Genetics, University of Siena, Siena, Italy.,Genetica Medica, Azienda Ospedaliera Universitaria Senese, Siena, Italy
| | - Rossella Tita
- Genetica Medica, Azienda Ospedaliera Universitaria Senese, Siena, Italy
| | | | - Elisa Benetti
- Medical Genetics, University of Siena, Siena, Italy.,Department of Medical Biotechnologies, University of Siena, Siena, Italy
| | - Francesca Niccheri
- Molecular Mechanisms of Oncogenesis, ISPRO Core Research Laboratory (CRL), Firenze, Italy
| | - Simone Furini
- Department of Medical Biotechnologies, University of Siena, Siena, Italy
| | - Caterina Lo Rizzo
- Genetica Medica, Azienda Ospedaliera Universitaria Senese, Siena, Italy
| | | | - Alessandra Renieri
- Medical Genetics, University of Siena, Siena, Italy. .,Genetica Medica, Azienda Ospedaliera Universitaria Senese, Siena, Italy.
| | | |
Collapse
|
8
|
FOXG1-Related Syndrome: From Clinical to Molecular Genetics and Pathogenic Mechanisms. Int J Mol Sci 2019; 20:ijms20174176. [PMID: 31454984 PMCID: PMC6747066 DOI: 10.3390/ijms20174176] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Revised: 08/23/2019] [Accepted: 08/25/2019] [Indexed: 12/29/2022] Open
Abstract
Individuals with mutations in forkhead box G1 (FOXG1) belong to a distinct clinical entity, termed “FOXG1-related encephalopathy”. There are two clinical phenotypes/syndromes identified in FOXG1-related encephalopathy, duplications and deletions/intragenic mutations. In children with deletions or intragenic mutations of FOXG1, the recognized clinical features include microcephaly, developmental delay, severe cognitive disabilities, early-onset dyskinesia and hyperkinetic movements, stereotypies, epilepsy, and cerebral malformation. In contrast, children with duplications of FOXG1 are typically normocephalic and have normal brain magnetic resonance imaging. They also have different clinical characteristics in terms of epilepsy, movement disorders, and neurodevelopment compared with children with deletions or intragenic mutations. FOXG1 is a transcriptional factor. It is expressed mainly in the telencephalon and plays a pleiotropic role in the development of the brain. It is a key player in development and territorial specification of the anterior brain. In addition, it maintains the expansion of the neural proliferating pool, and also regulates the pace of neocortical neuronogenic progression. It also facilitates cortical layer and corpus callosum formation. Furthermore, it promotes dendrite elongation and maintains neural plasticity, including dendritic arborization and spine densities in mature neurons. In this review, we summarize the clinical features, molecular genetics, and possible pathogenesis of FOXG1-related syndrome.
Collapse
|
9
|
Chen D, Wang C, Li M, She X, Yuan Y, Chen H, Zhang W, Zhao C. Loss of Foxg1 Impairs the Development of Cortical SST-Interneurons Leading to Abnormal Emotional and Social Behaviors. Cereb Cortex 2019; 29:3666-3682. [DOI: 10.1093/cercor/bhz114] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Revised: 04/10/2019] [Accepted: 05/05/2019] [Indexed: 12/19/2022] Open
Abstract
Abstract
FOXG1 syndrome is a severe encephalopathy that exhibit intellectual disability, emotional disorder, and limited social communication. To elucidate the contribution of somatostatin-expressing interneurons (SST-INs) to the cellular basis underlying FOXG1 syndrome, here, by crossing SST-cre with a Foxg1fl/fl line, we selectively ablated Foxg1. Loss of Foxg1 resulted in an obvious reduction in the number of SST-INs, accompanied by an altered ratio of subtypes. Foxg1-deficient SST-INs exhibited decreased membrane excitability and a changed ratio of electrophysiological firing patterns, which subsequently led to an excitatory/inhibitory imbalance. Moreover, cognitive defects, limited social interactions, and depression-like behaviors were detected in Foxg1 cKO mice. Treatment with low-dose of clonazepam effectively alleviated the defects. These results identify a link of SST-IN development to the aberrant emotion, cognition, and social capacities in patients. Our findings identify a novel role of Foxg1 in SST-IN development and put new insights into the cellular basis of FOXG1 syndrome.
Collapse
Affiliation(s)
- Dongsheng Chen
- Key Laboratory of Developmental Genes and Human Diseases, MOE, School of Medicine, Southeast University, Nanjing, Jiangsu Province, China
| | - Chunlian Wang
- Key Lab of Cognition and Personality, MOE, School of Psychology, Southwest University, Chongqing, China
| | - Meiyi Li
- Key Lab of Cognition and Personality, MOE, School of Psychology, Southwest University, Chongqing, China
| | - Xinyu She
- Key Laboratory of Developmental Genes and Human Diseases, MOE, School of Medicine, Southeast University, Nanjing, Jiangsu Province, China
| | - Yonggui Yuan
- Department of Psychosomatics and Psychiatry, ZhongDa Hospital, Medical School of Southeast University, Nanjing, Jiangsu Province, China
| | - Huanxin Chen
- Key Lab of Cognition and Personality, MOE, School of Psychology, Southwest University, Chongqing, China
| | - Weining Zhang
- School of Medicine, Jiangsu University, ZhenJiang, Jiangsu Province, China
| | - Chunjie Zhao
- Key Laboratory of Developmental Genes and Human Diseases, MOE, School of Medicine, Southeast University, Nanjing, Jiangsu Province, China
| |
Collapse
|
10
|
Testa G, Mainardi M, Olimpico F, Pancrazi L, Cattaneo A, Caleo M, Costa M. A triheptanoin-supplemented diet rescues hippocampal hyperexcitability and seizure susceptibility in FoxG1 +/- mice. Neuropharmacology 2019; 148:305-310. [PMID: 30639390 DOI: 10.1016/j.neuropharm.2019.01.005] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2018] [Revised: 12/11/2018] [Accepted: 01/08/2019] [Indexed: 12/27/2022]
Abstract
The Forkhead Box G1 (FOXG1) gene encodes a transcription factor with an essential role in mammalian telencephalon development. FOXG1-related disorders, caused by deletions, intragenic mutations or duplications, are usually associated with severe intellectual disability, autistic features, and, in 87% of subjects, epileptiform manifestations. In a subset of patients with FoxG1 mutations, seizures remain intractable, prompting the need for novel therapeutic options. To address this issue, we took advantage of a haploinsufficient animal model, the FoxG1+/- mouse. In vivo electrophysiological analyses of FoxG1+/- mice detected hippocampal hyperexcitability, which turned into overt seizures upon delivery of the proconvulsant kainic acid, as confirmed by behavioral observations. These alterations were associated with decreased expression of the chloride transporter KCC2. Next, we tested whether a triheptanoin-based anaplerotic diet could have an impact on the pathological phenotype of FoxG1+/- mice. This manipulation abated altered neural activity and normalized enhanced susceptibility to proconvulsant-induced seizures, in addition to rescuing altered expression of KCC2 and increasing the levels of the GABA transporter vGAT. In conclusion, our data show that FoxG1 haploinsufficiency causes dysfunction of hippocampal circuits and increases the susceptibility to a proconvulsant insult, and that these alterations are rescued by triheptanoin dietary treatment.
Collapse
Affiliation(s)
- Giovanna Testa
- Laboratory of Biology "Bio@SNS", Scuola Normale Superiore, Piazza dei Cavalieri, 7, 56124, Pisa, Italy
| | - Marco Mainardi
- Laboratory of Biology "Bio@SNS", Scuola Normale Superiore, Piazza dei Cavalieri, 7, 56124, Pisa, Italy; Institute of Neuroscience, Italian National Research Council (CNR), Via Moruzzi, 1, 56124, Pisa, Italy.
| | - Francesco Olimpico
- Laboratory of Biology "Bio@SNS", Scuola Normale Superiore, Piazza dei Cavalieri, 7, 56124, Pisa, Italy
| | - Laura Pancrazi
- Institute of Neuroscience, Italian National Research Council (CNR), Via Moruzzi, 1, 56124, Pisa, Italy
| | - Antonino Cattaneo
- Laboratory of Biology "Bio@SNS", Scuola Normale Superiore, Piazza dei Cavalieri, 7, 56124, Pisa, Italy
| | - Matteo Caleo
- Laboratory of Biology "Bio@SNS", Scuola Normale Superiore, Piazza dei Cavalieri, 7, 56124, Pisa, Italy; Institute of Neuroscience, Italian National Research Council (CNR), Via Moruzzi, 1, 56124, Pisa, Italy
| | - Mario Costa
- Laboratory of Biology "Bio@SNS", Scuola Normale Superiore, Piazza dei Cavalieri, 7, 56124, Pisa, Italy; Institute of Neuroscience, Italian National Research Council (CNR), Via Moruzzi, 1, 56124, Pisa, Italy.
| |
Collapse
|
11
|
Pringsheim M, Mitter D, Schröder S, Warthemann R, Plümacher K, Kluger G, Baethmann M, Bast T, Braun S, Büttel HM, Conover E, Courage C, Datta AN, Eger A, Grebe TA, Hasse-Wittmer A, Heruth M, Höft K, Kaindl AM, Karch S, Kautzky T, Korenke GC, Kruse B, Lutz RE, Omran H, Patzer S, Philippi H, Ramsey K, Rating T, Rieß A, Schimmel M, Westman R, Zech FM, Zirn B, Ulmke PA, Sokpor G, Tuoc T, Leha A, Staudt M, Brockmann K. Structural brain anomalies in patients with FOXG1 syndrome and in Foxg1+/- mice. Ann Clin Transl Neurol 2019; 6:655-668. [PMID: 31019990 PMCID: PMC6469254 DOI: 10.1002/acn3.735] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2018] [Accepted: 01/22/2019] [Indexed: 01/11/2023] Open
Abstract
Objective FOXG1 syndrome is a rare neurodevelopmental disorder associated with heterozygous FOXG1 variants or chromosomal microaberrations in 14q12. The study aimed at assessing the scope of structural cerebral anomalies revealed by neuroimaging to delineate the genotype and neuroimaging phenotype associations. Methods We compiled 34 patients with a heterozygous (likely) pathogenic FOXG1 variant. Qualitative assessment of cerebral anomalies was performed by standardized re-analysis of all 34 MRI data sets. Statistical analysis of genetic, clinical and neuroimaging data were performed. We quantified clinical and neuroimaging phenotypes using severity scores. Telencephalic phenotypes of adult Foxg1+/- mice were examined using immunohistological stainings followed by quantitative evaluation of structural anomalies. Results Characteristic neuroimaging features included corpus callosum anomalies (82%), thickening of the fornix (74%), simplified gyral pattern (56%), enlargement of inner CSF spaces (44%), hypoplasia of basal ganglia (38%), and hypoplasia of frontal lobes (29%). We observed a marked, filiform thinning of the rostrum as recurrent highly typical pattern of corpus callosum anomaly in combination with distinct thickening of the fornix as a characteristic feature. Thickening of the fornices was not reported previously in FOXG1 syndrome. Simplified gyral pattern occurred significantly more frequently in patients with early truncating variants. Higher clinical severity scores were significantly associated with higher neuroimaging severity scores. Modeling of Foxg1 heterozygosity in mouse brain recapitulated the associated abnormal cerebral morphology phenotypes, including the striking enlargement of the fornix. Interpretation Combination of specific corpus callosum anomalies with simplified gyral pattern and hyperplasia of the fornices is highly characteristic for FOXG1 syndrome.
Collapse
Affiliation(s)
- Milka Pringsheim
- Klinik für Neuropädiatrie und Neurologische Rehabilitation Epilepsiezentrum für Kinder und Jugendliche Schön Klinik Vogtareuth Vogtareuth Germany.,Research Institute "Rehabilitation, Transition, Rehabilitation" Paracelsus Medical University Salzburg Austria
| | - Diana Mitter
- Institute of Human Genetics University of Leipzig Medical Center Leipzig Germany
| | - Simone Schröder
- Interdisciplinary Pediatric Center for Children with Developmental Disabilities and Severe Chronic Disorders University Medical Center Göttingen Göttingen Germany
| | - Rita Warthemann
- Interdisciplinary Pediatric Center for Children with Developmental Disabilities and Severe Chronic Disorders University Medical Center Göttingen Göttingen Germany
| | - Kim Plümacher
- Interdisciplinary Pediatric Center for Children with Developmental Disabilities and Severe Chronic Disorders University Medical Center Göttingen Göttingen Germany
| | - Gerhard Kluger
- Klinik für Neuropädiatrie und Neurologische Rehabilitation Epilepsiezentrum für Kinder und Jugendliche Schön Klinik Vogtareuth Vogtareuth Germany.,Research Institute "Rehabilitation, Transition, Rehabilitation" Paracelsus Medical University Salzburg Austria
| | | | - Thomas Bast
- Epilepsiezentrum Kork Kehl-Kork Germany.,Medical Faculty University of Freiburg Freiburg Germany
| | - Sarah Braun
- Asklepios Children's Hospital St. Augustin Germany
| | | | - Elizabeth Conover
- Department of Genetic Medicine Munroe Meyer Institute University of Nebraska Medical Center Omaha Omaha Nebraska USA
| | - Carolina Courage
- Division of Human Genetics Department of Pediatrics, Inselspital University of Bern Bern Switzerland.,The Folkhälsan Institute of Genetics University of Helsinki Helsinki Finland
| | - Alexandre N Datta
- Department of Pediatric Neurology and Developmental Medicine University of Basel Children's Hospital Basel Switzerland
| | - Angelika Eger
- Sozialpädiatrisches Zentrum Leipzig (Frühe Hilfe Leipzig) Leipzig Germany
| | - Theresa A Grebe
- Division of Genetics and Metabolism Phoenix Children's Hospital Phoenix Arizona USA
| | | | - Marion Heruth
- Klinik für Kinder- und Jugendmedizin Sana Kliniken Leipziger Land Borna Germany
| | - Karen Höft
- Klinik für Kinder- und Jugendmedizin Klinikum Magdeburg gGmbH Magdeburg Germany
| | - Angela M Kaindl
- Klinik für Pädiatrie m.S. Neurologie Sozialpädiatrisches Zentrum Institut für Zell- und Neurobiologie Charité-Universitätsmedizin Berlin Berlin Germany
| | - Stephanie Karch
- Klinik für Kinder- und Jugendmedizin Sozialpädiatrisches Zentrum Universitätsklinikum Heidelberg Heidelberg Germany
| | | | - Georg C Korenke
- Klinik für Neuropädiatrie und angeborene Stoffwechselerkrankungen Elisabeth Kinderkrankenhaus Klinikum Oldenburg Germany
| | - Bernd Kruse
- Neuropediatric Department Helios-Klinikum Hildesheim Hildesheim Germany
| | - Richard E Lutz
- Department of Genetic Medicine Munroe Meyer Institute University of Nebraska Medical Center Omaha Omaha Nebraska USA
| | - Heymut Omran
- Department of General Pediatrics University Children's Hospital Muenster Muenster Germany
| | - Steffi Patzer
- Klinik für Kinder- und Jugendmedizin Krankenhaus St. Elisabeth und St. Barbara Halle/Saale Germany
| | - Heike Philippi
- Sozialpädiatrisches Zentrum Frankfurt Mitte Frankfurt am Main Germany
| | - Keri Ramsey
- Center for Rare Childhood Disorders Translational Genomics Research Institute Phoenix Arizona USA
| | - Tina Rating
- Sozialpädiatrisches Institut Klinikum Bremen-Mitte Bremen Germany
| | - Angelika Rieß
- Institut für Medizinische Genetik und angewandte Genomik Universitätsklinikum Tübingen Tübingen Germany
| | - Mareike Schimmel
- Children's Hospital Section of Neuropaediatrics Klinikum Augsburg Augsburg Germany
| | - Rachel Westman
- Children's Specialty Center St. Luke's Children's Hospital Boise Idaho USA
| | - Frank-Martin Zech
- Klinik für Kinder- und Jugendmedizin St. Vincenz-Krankenhaus Paderborn Paderborn Germany
| | - Birgit Zirn
- Genetic Counselling and Diagnostic, genetikum Stuttgart Stuttgart Germany
| | - Pauline A Ulmke
- Institute of Neuroanatomy University Medical Center Georg August University Göttingen Germany
| | - Godwin Sokpor
- Institute of Neuroanatomy University Medical Center Georg August University Göttingen Germany
| | - Tran Tuoc
- Institute of Neuroanatomy University Medical Center Georg August University Göttingen Germany
| | - Andreas Leha
- 'Core Facility Medical Biometry and Statistical Bioinformatics' Department of Medical Statistics University Medical Center Göttingen Göttingen Germany
| | - Martin Staudt
- Klinik für Neuropädiatrie und Neurologische Rehabilitation Epilepsiezentrum für Kinder und Jugendliche Schön Klinik Vogtareuth Vogtareuth Germany
| | - Knut Brockmann
- Interdisciplinary Pediatric Center for Children with Developmental Disabilities and Severe Chronic Disorders University Medical Center Göttingen Göttingen Germany
| |
Collapse
|
12
|
Zhu W, Zhang B, Li M, Mo F, Mi T, Wu Y, Teng Z, Zhou Q, Li W, Hu B. Precisely controlling endogenous protein dosage in hPSCs and derivatives to model FOXG1 syndrome. Nat Commun 2019; 10:928. [PMID: 30804331 PMCID: PMC6389984 DOI: 10.1038/s41467-019-08841-7] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2018] [Accepted: 01/23/2019] [Indexed: 01/25/2023] Open
Abstract
Dosage of key regulators impinge on developmental disorders such as FOXG1 syndrome. Since neither knock-out nor knock-down strategy assures flexible and precise protein abundance control, to study hypomorphic or haploinsufficiency expression remains challenging. We develop a system in human pluripotent stem cells (hPSCs) using CRISPR/Cas9 and SMASh technology, with which we can target endogenous proteins for precise dosage control in hPSCs and at multiple stages of neural differentiation. We also reveal FOXG1 dose-dependently affect the cellular constitution of human brain, with 60% mildly affect GABAergic interneuron development while 30% thresholds the production of MGE derived neurons. Abnormal interneuron differentiation accounts for various neurological defects such as epilepsy or seizures, which stimulates future innovative cures of FOXG1 syndrome. By means of its robustness and easiness, dosage-control of proteins in hPSCs and their derivatives will update the understanding and treatment of additional diseases caused by abnormal protein dosage. Altered dosage of developmental regulators such as transcription factors can result in disorders, such as FOXG1 syndrome. Here, the authors demonstrate the utility of SMASh technology for modulating protein dosage by modeling FOXG1 syndrome using human pluripotent stem cell-derived neurons and neural organoids.
Collapse
Affiliation(s)
- Wenliang Zhu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China.,Institute for Stem cell and Regeneration, Chinese Academy of Sciences, Beijing, China
| | - Boya Zhang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,Institute for Stem cell and Regeneration, Chinese Academy of Sciences, Beijing, China
| | - Mengqi Li
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,Institute for Stem cell and Regeneration, Chinese Academy of Sciences, Beijing, China
| | - Fan Mo
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China.,Institute for Stem cell and Regeneration, Chinese Academy of Sciences, Beijing, China
| | - Tingwei Mi
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Yihui Wu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China.,Institute for Stem cell and Regeneration, Chinese Academy of Sciences, Beijing, China
| | - Zhaoqian Teng
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China.,Institute for Stem cell and Regeneration, Chinese Academy of Sciences, Beijing, China
| | - Qi Zhou
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China. .,University of Chinese Academy of Sciences, Beijing, China. .,Institute for Stem cell and Regeneration, Chinese Academy of Sciences, Beijing, China.
| | - Wei Li
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China. .,University of Chinese Academy of Sciences, Beijing, China. .,Institute for Stem cell and Regeneration, Chinese Academy of Sciences, Beijing, China.
| | - Baoyang Hu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China. .,University of Chinese Academy of Sciences, Beijing, China. .,Institute for Stem cell and Regeneration, Chinese Academy of Sciences, Beijing, China.
| |
Collapse
|
13
|
Vegas N, Cavallin M, Maillard C, Boddaert N, Toulouse J, Schaefer E, Lerman-Sagie T, Lev D, Magalie B, Moutton S, Haan E, Isidor B, Heron D, Milh M, Rondeau S, Michot C, Valence S, Wagner S, Hully M, Mignot C, Masurel A, Datta A, Odent S, Nizon M, Lazaro L, Vincent M, Cogné B, Guerrot AM, Arpin S, Pedespan JM, Caubel I, Pontier B, Troude B, Rivier F, Philippe C, Bienvenu T, Spitz MA, Bery A, Bahi-Buisson N. Delineating FOXG1 syndrome: From congenital microcephaly to hyperkinetic encephalopathy. NEUROLOGY-GENETICS 2018; 4:e281. [PMID: 30533527 PMCID: PMC6244024 DOI: 10.1212/nxg.0000000000000281] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/24/2018] [Accepted: 07/12/2018] [Indexed: 12/24/2022]
Abstract
Objective To provide new insights into the FOXG1-related clinical and imaging phenotypes and refine the phenotype-genotype correlation in FOXG1 syndrome. Methods We analyzed the clinical and imaging phenotypes of a cohort of 45 patients with a pathogenic or likely pathogenic FOXG1 variant and performed phenotype-genotype correlations. Results A total of 37 FOXG1 different heterozygous mutations were identified, of which 18 are novel. We described a broad spectrum of neurodevelopmental phenotypes, characterized by severe postnatal microcephaly and developmental delay accompanied by a hyperkinetic movement disorder, stereotypes and sleep disorders, and epileptic seizures. Our data highlighted 3 patterns of gyration, including frontal pachygyria in younger patients (26.7%), moderate simplified gyration (24.4%) and mildly simplified or normal gyration (48.9%), corpus callosum hypogenesis mostly in its frontal part, combined with moderate-to-severe myelination delay that improved and normalized with age. Frameshift and nonsense mutations in the N-terminus of FOXG1, which are the most common mutation types, show the most severe clinical features and MRI anomalies. However, patients with recurrent frameshift mutations c.460dupG and c.256dupC had variable clinical and imaging presentations. Conclusions These findings have implications for genetic counseling, providing evidence that N-terminal mutations and large deletions lead to more severe FOXG1 syndrome, although genotype-phenotype correlations are not necessarily straightforward in recurrent mutations. Together, these analyses support the view that FOXG1 syndrome is a specific disorder characterized by frontal pachygyria and delayed myelination in its most severe form and hypogenetic corpus callosum in its milder form.
Collapse
|
14
|
Stefano GB, Pilonis N, Ptacek R, Raboch J, Vnukova M, Kream RM. Gut, Microbiome, and Brain Regulatory Axis: Relevance to Neurodegenerative and Psychiatric Disorders. Cell Mol Neurobiol 2018; 38:1197-1206. [PMID: 29802603 PMCID: PMC6061125 DOI: 10.1007/s10571-018-0589-2] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2018] [Accepted: 05/07/2018] [Indexed: 12/23/2022]
Abstract
It has become apparent that the molecular and biochemical integrity of interactive families, genera, and species of human gut microflora is critically linked to maintaining complex metabolic and behavioral processes mediated by peripheral organ systems and central nervous system neuronal groupings. Relatively recent studies have established intrinsic ratios of enterotypes contained within the human microbiome across demographic subpopulations and have empirically linked significant alterations in the expression of bacterial enterotypes with the initiation and persistence of several major metabolic and psychiatric disorders. Accordingly, the goal of our review is to highlight potential thematic/functional linkages of pathophysiological alterations in gut microbiota and bidirectional gut-brain signaling pathways with special emphasis on the potential roles of gut dysbiosis on the pathophysiology of psychiatric illnesses. We provide critical discussion of putative thematic linkages of Parkinson's disease (PD) data sets to similar pathophysiological events as potential causative factors in the development and persistence of diverse psychiatric illnesses. Finally, we include a concise review of preclinical paradigms that involve immunologically-induced GI deficits and dysbiosis of maternal microflora that are functionally linked to impaired neurodevelopmental processes leading to affective behavioral syndromes in the offspring.
Collapse
Affiliation(s)
- G B Stefano
- Department of Psychiatry, First Faculty of Medicine Charles University in Prague and General University Hospital in Prague, Center for Cognitive and Molecular Neuroscience, Ke Karlovu 11, 120 00, Prague 2, Czech Republic.
| | - N Pilonis
- Warsaw Medical University, Public Central Teaching Hospital, Warsaw, Poland
| | - R Ptacek
- Department of Psychiatry, First Faculty of Medicine Charles University in Prague and General University Hospital in Prague, Center for Cognitive and Molecular Neuroscience, Ke Karlovu 11, 120 00, Prague 2, Czech Republic
| | - J Raboch
- Department of Psychiatry, First Faculty of Medicine Charles University in Prague and General University Hospital in Prague, Center for Cognitive and Molecular Neuroscience, Ke Karlovu 11, 120 00, Prague 2, Czech Republic
| | - M Vnukova
- Department of Psychiatry, First Faculty of Medicine Charles University in Prague and General University Hospital in Prague, Center for Cognitive and Molecular Neuroscience, Ke Karlovu 11, 120 00, Prague 2, Czech Republic
| | - R M Kream
- Department of Psychiatry, First Faculty of Medicine Charles University in Prague and General University Hospital in Prague, Center for Cognitive and Molecular Neuroscience, Ke Karlovu 11, 120 00, Prague 2, Czech Republic
| |
Collapse
|
15
|
Vezzali R, Weise SC, Hellbach N, Machado V, Heidrich S, Vogel T. The FOXG1/FOXO/SMAD network balances proliferation and differentiation of cortical progenitors and activates Kcnh3 expression in mature neurons. Oncotarget 2018; 7:37436-37455. [PMID: 27224923 PMCID: PMC5122323 DOI: 10.18632/oncotarget.9545] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2015] [Accepted: 05/11/2016] [Indexed: 12/02/2022] Open
Abstract
Transforming growth factor β (TGFβ)-mediated anti-proliferative and differentiating effects promote neuronal differentiation during embryonic central nervous system development. TGFβ downstream signals, composed of activated SMAD2/3, SMAD4 and a FOXO family member, promote the expression of cyclin-dependent kinase inhibitor Cdkn1a. In early CNS development, IGF1/PI3K signaling and the transcription factor FOXG1 inhibit FOXO- and TGFβ-mediated Cdkn1a transcription. FOXG1 prevents cell cycle exit by binding to the SMAD/FOXO-protein complex. In this study we provide further details on the FOXG1/FOXO/SMAD transcription factor network. We identified ligands of the TGFβ- and IGF-family, Foxo1, Foxo3 and Kcnh3 as novel FOXG1-target genes during telencephalic development and showed that FOXG1 interferes with Foxo1 and Tgfβ transcription. Our data specify that FOXO1 activates Cdkn1a transcription. This process is under control of the IGF1-pathway, as Cdkn1a transcription increases when IGF1-signaling is pharmacologically inhibited. However, overexpression of CDKN1A and knockdown of Foxo1 and Foxo3 is not sufficient for neuronal differentiation, which is probably instructed by TGFβ-signaling. In mature neurons, FOXG1 activates transcription of the seizure-related Kcnh3, which might be a FOXG1-target gene involved in the FOXG1 syndrome pathology.
Collapse
Affiliation(s)
- Riccardo Vezzali
- Department of Molecular Embryology, Institute of Anatomy and Cell Biology, Faculty of Medicine, University of Freiburg, Freiburg, Germany.,Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Stefan Christopher Weise
- Department of Molecular Embryology, Institute of Anatomy and Cell Biology, Faculty of Medicine, University of Freiburg, Freiburg, Germany.,Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Nicole Hellbach
- Department of Molecular Embryology, Institute of Anatomy and Cell Biology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Venissa Machado
- Department of Molecular Embryology, Institute of Anatomy and Cell Biology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Stefanie Heidrich
- Department of Molecular Embryology, Institute of Anatomy and Cell Biology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Tanja Vogel
- Department of Molecular Embryology, Institute of Anatomy and Cell Biology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| |
Collapse
|
16
|
Harada K, Yamamoto M, Konishi Y, Koyano K, Takahashi S, Namba M, Kusaka T. Hypoplastic hippocampus in atypical Rett syndrome with a novel FOXG1 mutation. Brain Dev 2018; 40:49-52. [PMID: 28781028 DOI: 10.1016/j.braindev.2017.07.007] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2017] [Revised: 06/24/2017] [Accepted: 07/17/2017] [Indexed: 11/28/2022]
Abstract
The forkhead box G1 (FOXG1) gene encodes a brain-specific transcription factor and is associated with a congenital variant of atypical Rett syndrome (RTT); several FOXG1 mutations have been identified. The congenital variant of RTT shows a hypoplastic corpus callosum, delayed myelination, and frontal and temporal atrophy. Although no report has described a hippocampal abnormality in humans, the current study suggests that FOXG1 also regulates neurogenesis in the postnatal hippocampus. In the present case, severe developmental delay was observed in a patient with a congenital variant of RTT from about 4months, in conjunction with acquired microcephaly, hypotonia, limited motor function, absent purposeful hand use, and repetitive jerky movements of the upper limbs. A novel missense mutation was identified in FOXG1 on gene analysis (c. 569T>A, p. Ile190Asn). The patient showed not only the typical cerebral abnormalities of a congenital variant of RTT, but also a hypoplastic hippocampus. This novel mutation and cerebral findings may provide new insights into the pathophysiology of the congenital variant of RTT.
Collapse
Affiliation(s)
- Kotoha Harada
- Department of Pediatrics, Shodoshima Central Hospital, Japan.
| | - Mayumi Yamamoto
- Department of Pediatrics, Shodoshima Central Hospital, Japan
| | - Yukihiko Konishi
- Department of Pediatrics, Faculty of Medicine, Kagawa University, Japan
| | - Kaori Koyano
- Department of Pediatrics, Faculty of Medicine, Kagawa University, Japan
| | | | - Masanori Namba
- Department of Pediatrics, Kagawa Rehabilitation Center, Japan
| | - Takashi Kusaka
- Department of Pediatrics, Faculty of Medicine, Kagawa University, Japan
| |
Collapse
|
17
|
FOXG1 syndrome: genotype-phenotype association in 83 patients with FOXG1 variants. Genet Med 2017; 20:98-108. [PMID: 28661489 DOI: 10.1038/gim.2017.75] [Citation(s) in RCA: 65] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2016] [Accepted: 03/16/2017] [Indexed: 12/31/2022] Open
Abstract
PurposeThe study aimed at widening the clinical and genetic spectrum and assessing genotype-phenotype associations in FOXG1 syndrome due to FOXG1 variants.MethodsWe compiled 30 new and 53 reported patients with a heterozygous pathogenic or likely pathogenic variant in FOXG1. We grouped patients according to type and location of the variant. Statistical analysis of molecular and clinical data was performed using Fisher's exact test and a nonparametric multivariate test.ResultsAmong the 30 new patients, we identified 19 novel FOXG1 variants. Among the total group of 83 patients, there were 54 variants: 20 frameshift (37%), 17 missense (31%), 15 nonsense (28%), and 2 in-frame variants (4%). Frameshift and nonsense variants are distributed over all FOXG1 protein domains; missense variants cluster within the conserved forkhead domain. We found a higher phenotypic variability than previously described. Genotype-phenotype association revealed significant differences in psychomotor development and neurological features between FOXG1 genotype groups. More severe phenotypes were associated with truncating FOXG1 variants in the N-terminal domain and the forkhead domain (except conserved site 1) and milder phenotypes with missense variants in the forkhead conserved site 1.ConclusionsThese data may serve for improved interpretation of new FOXG1 sequence variants and well-founded genetic counseling.
Collapse
|
18
|
Boggio E, Pancrazi L, Gennaro M, Lo Rizzo C, Mari F, Meloni I, Ariani F, Panighini A, Novelli E, Biagioni M, Strettoi E, Hayek J, Rufa A, Pizzorusso T, Renieri A, Costa M. Visual impairment in FOXG1-mutated individuals and mice. Neuroscience 2016; 324:496-508. [DOI: 10.1016/j.neuroscience.2016.03.027] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2015] [Revised: 03/01/2016] [Accepted: 03/08/2016] [Indexed: 01/01/2023]
|
19
|
Pancrazi L, Di Benedetto G, Colombaioni L, Della Sala G, Testa G, Olimpico F, Reyes A, Zeviani M, Pozzan T, Costa M. Foxg1 localizes to mitochondria and coordinates cell differentiation and bioenergetics. Proc Natl Acad Sci U S A 2015; 112:13910-5. [PMID: 26508630 PMCID: PMC4653140 DOI: 10.1073/pnas.1515190112] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Forkhead box g1 (Foxg1) is a nuclear-cytosolic transcription factor essential for the forebrain development and involved in neurodevelopmental and cancer pathologies. Despite the importance of this protein, little is known about the modalities by which it exerts such a large number of cellular functions. Here we show that a fraction of Foxg1 is localized within the mitochondria in cell lines, primary neuronal or glial cell cultures, and in the mouse cortex. Import of Foxg1 in isolated mitochondria appears to be membrane potential-dependent. Amino acids (aa) 277-302 were identified as critical for mitochondrial localization. Overexpression of full-length Foxg1 enhanced mitochondrial membrane potential (ΔΨm) and promoted mitochondrial fission and mitosis. Conversely, overexpression of the C-term Foxg1 (aa 272-481), which is selectively localized in the mitochondrial matrix, enhanced organelle fusion and promoted the early phase of neuronal differentiation. These findings suggest that the different subcellular localizations of Foxg1 control the machinery that brings about cell differentiation, replication, and bioenergetics, possibly linking mitochondrial functions to embryonic development and pathological conditions.
Collapse
Affiliation(s)
| | - Giulietta Di Benedetto
- Institute of Neuroscience, Italian National Research Council, 35121 Padova, Italy; Venetian Institute of Molecular Medicine, 35129 Padova, Italy
| | - Laura Colombaioni
- Institute of Neuroscience, Italian National Research Council, 56124 Pisa, Italy
| | - Grazia Della Sala
- Institute of Neuroscience, Italian National Research Council, 56124 Pisa, Italy; Department of Neuroscience, Psychology, Drug Research and Child Health, University of Florence, 50139 Florence, Italy
| | | | - Francesco Olimpico
- Institute of Neuroscience, Italian National Research Council, 56124 Pisa, Italy
| | - Aurelio Reyes
- Mitochondrial Biology Unit, Medical Research Council, Cambridge CB20XY, United Kingdom
| | - Massimo Zeviani
- Mitochondrial Biology Unit, Medical Research Council, Cambridge CB20XY, United Kingdom
| | - Tullio Pozzan
- Institute of Neuroscience, Italian National Research Council, 35121 Padova, Italy; Venetian Institute of Molecular Medicine, 35129 Padova, Italy; Department Biomedical Sciences, University of Padova, 35121 Padova, Italy
| | - Mario Costa
- Scuola Normale Superiore, 56126 Pisa, Italy; Institute of Neuroscience, Italian National Research Council, 56124 Pisa, Italy;
| |
Collapse
|
20
|
A haploinsufficiency of FOXG1 identified in a boy with congenital variant of Rett syndrome. Brain Dev 2014; 36:725-9. [PMID: 24139857 DOI: 10.1016/j.braindev.2013.09.006] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2012] [Revised: 09/01/2013] [Accepted: 09/13/2013] [Indexed: 11/23/2022]
Abstract
BACKGROUND Forkhead box G1 gene (FOXG1) mutations and deletions are associated with a congenital variant of Rett syndrome (RTT). Nucleotide alterations of the coding region of FOXG1 have never caused dysmorphic features. PATIENT An 8-year-old boy with the congenital variant of RTT who showed severe psychomotor deterioration, epilepsy, acquired microcephaly, and involuntary movements including jerky movements of the upper limbs and tongue protrusion. He showed dysmorphic features including round face, anteverted nostrils, and tented upper lips. Brain magnetic resonance imaging showed hypoplasia of the frontal lobes and the rostral part of the corpus callosum. The molecular cytogenetic analysis confirmed a de novo deletion of 14q12 including FOXG1 in this patient. CONCLUSION We identified the smallest deletion of 14q12 involving FOXG1 among those previously reported. Dysmorphic facial features are a characteristic for the patients with chromosomal deletion including FOXG1. In our patient, C14orf23 is the only transcript other than FOXG1. Therefore, C14orf23 might be responsible for facial dysmorphism.
Collapse
|
21
|
Maortua H, Martínez-Bouzas C, García-Ribes A, Martínez MJ, Guillen E, Domingo MR, Calvo MT, Guitart M, Gabau E, Botella MP, Gener B, Rubio I, López-Aríztegui MA, Tejada MI. MECP2 gene study in a large cohort: testing of 240 female patients and 861 healthy controls (519 females and 342 males). J Mol Diagn 2013; 15:723-9. [PMID: 23810759 DOI: 10.1016/j.jmoldx.2013.05.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2012] [Revised: 05/08/2013] [Accepted: 05/15/2013] [Indexed: 11/24/2022] Open
Abstract
The MECP2 gene located on Xq28 is one of the most important genes contributing to the spectrum of neurodevelopmental disorders. Therefore, we present our experience in the molecular study of this gene. MECP2 was thoroughly tested for the presence of mutations (sequencing of four exons and rearrangements) in 120 female patients: 28 with classic Rett syndrome, five with atypical Rett syndrome, and 87 with heterogeneous phenotypes with some Rett-like features. Another 120 female patients with intellectual disability of unknown origin were also studied, but in these cases we only tested exons 3 and 4. Finally, 861 healthy controls (519 females and 342 males) were also studied for exon 3 and 4. Eighteen different pathological mutations were found, five of them previously undescribed, and four large deletions detected by multiplex ligation-dependent probe amplification. All were de novo mutations not present in the parents. In conclusion, i) MECP2 is one of the most important genes in the diagnosis of genetic intellectual disability in females; ii) MECP2 must be studied not only in patients with classical/atypical Rett syndrome but also in patients with other phenotypes related to Rett syndrome; and iii) for the new variants, it is important to perform complementary studies, including the analysis of large populations of healthy individuals and the use of in silico programs.
Collapse
Affiliation(s)
- Hiart Maortua
- Molecular Genetics Laboratory, Genetics Service, BioCruces Health Research Institute, Cruces University Hospital, Barakaldo-Bizkaia, Spain
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
14q12 and severe Rett-like phenotypes: new clinical insights and physical mapping of FOXG1-regulatory elements. Eur J Hum Genet 2012; 20:1216-23. [PMID: 22739344 DOI: 10.1038/ejhg.2012.127] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
The Forkhead box G1 (FOXG1) gene has been implicated in severe Rett-like phenotypes. It encodes the Forkhead box protein G1, a winged-helix transcriptional repressor critical for forebrain development. Recently, the core FOXG1 syndrome was defined as postnatal microcephaly, severe mental retardation, absent language, dyskinesia, and dysgenesis of the corpus callosum. We present seven additional patients with a severe Rett-like neurodevelopment disorder associated with de novo FOXG1 point mutations (two cases) or 14q12 deletions (five cases). We expand the mutational spectrum in patients with FOXG1-related encephalopathies and precise the core FOXG1 syndrome phenotype. Dysgenesis of the corpus callosum and dyskinesia are not always present in FOXG1-mutated patients. We believe that the FOXG1 gene should be considered in severely mentally retarded patients (no speech-language) with severe acquired microcephaly (-4 to-6 SD) and few clinical features suggestive of Rett syndrome. Interestingly enough, three 14q12 deletions that do not include the FOXG1 gene are associated with phenotypes very reminiscent to that of FOXG1-mutation-positive patients. We physically mapped a putative long-range FOXG1-regulatory element in a 0.43 Mb DNA segment encompassing the PRKD1 locus. In fibroblast cells, a cis-acting regulatory sequence located more than 0.6 Mb away from FOXG1 acts as a silencer at the transcriptional level. These data are important for clinicians and for molecular biologists involved in the management of patients with severe encephalopathies compatible with a FOXG1-related phenotype.
Collapse
|