1
|
Tan DCS, Jung S, Deng Y, Morey N, Chan G, Bongers A, Ke YD, Ittner LM, Delerue F. PLP1-Targeting Antisense Oligonucleotides Improve FOXG1 Syndrome Mice. Int J Mol Sci 2024; 25:10846. [PMID: 39409184 PMCID: PMC11477415 DOI: 10.3390/ijms251910846] [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: 09/18/2024] [Revised: 10/02/2024] [Accepted: 10/04/2024] [Indexed: 10/20/2024] Open
Abstract
FOXG1 syndrome is a rare neurodevelopmental disorder of the telencephalon, for which there is no cure. Underlying heterozygous pathogenic variants in the Forkhead Box G1 (FOXG1) gene with resulting impaired or loss of FOXG1 function lead to severe neurological impairments. Here, we report a patient with a de novo pathogenic single nucleotide deletion c.946del (p.Leu316Cysfs*10) of the FOXG1 gene that causes a premature protein truncation. To study this variant in vivo, we generated and characterized Foxg1 c946del mice that recapitulate hallmarks of the human disorder. Accordingly, heterozygous Foxg1 c946del mice display neurological symptoms with aberrant neuronal networks and increased seizure susceptibility. Gene expression profiling identified increased oligodendrocyte- and myelination-related gene clusters. Specifically, we showed that expression of the c946del mutant and of other pathogenic FOXG1 variants correlated with overexpression of proteolipid protein 1 (Plp1), a gene linked to white matter disorders. Postnatal administration of Plp1-targeting antisense oligonucleotides (ASOs) in Foxg1 c946del mice improved neurological deficits. Our data suggest Plp1 as a new target for therapeutic strategies mitigating disease phenotypes in FOXG1 syndrome patients.
Collapse
Affiliation(s)
- Daniel C. S. Tan
- Dementia Research Centre, Department of Biomedical Sciences, Faculty of Medicine Health and Human Sciences, Macquarie University, Sydney, NSW 2109, Australia; (D.C.S.T.); (S.J.); (Y.D.); (N.M.); (G.C.); (Y.D.K.)
| | - Seonghee Jung
- Dementia Research Centre, Department of Biomedical Sciences, Faculty of Medicine Health and Human Sciences, Macquarie University, Sydney, NSW 2109, Australia; (D.C.S.T.); (S.J.); (Y.D.); (N.M.); (G.C.); (Y.D.K.)
| | - Yuanyuan Deng
- Dementia Research Centre, Department of Biomedical Sciences, Faculty of Medicine Health and Human Sciences, Macquarie University, Sydney, NSW 2109, Australia; (D.C.S.T.); (S.J.); (Y.D.); (N.M.); (G.C.); (Y.D.K.)
| | - Nicolle Morey
- Dementia Research Centre, Department of Biomedical Sciences, Faculty of Medicine Health and Human Sciences, Macquarie University, Sydney, NSW 2109, Australia; (D.C.S.T.); (S.J.); (Y.D.); (N.M.); (G.C.); (Y.D.K.)
| | - Gabriella Chan
- Dementia Research Centre, Department of Biomedical Sciences, Faculty of Medicine Health and Human Sciences, Macquarie University, Sydney, NSW 2109, Australia; (D.C.S.T.); (S.J.); (Y.D.); (N.M.); (G.C.); (Y.D.K.)
| | - Andre Bongers
- Biological Resources Imaging Laboratory, University of New South Wales, Sydney, NSW 2052, Australia; (A.B.)
| | - Yazi D. Ke
- Dementia Research Centre, Department of Biomedical Sciences, Faculty of Medicine Health and Human Sciences, Macquarie University, Sydney, NSW 2109, Australia; (D.C.S.T.); (S.J.); (Y.D.); (N.M.); (G.C.); (Y.D.K.)
| | - Lars M. Ittner
- Dementia Research Centre, Department of Biomedical Sciences, Faculty of Medicine Health and Human Sciences, Macquarie University, Sydney, NSW 2109, Australia; (D.C.S.T.); (S.J.); (Y.D.); (N.M.); (G.C.); (Y.D.K.)
| | - Fabien Delerue
- Dementia Research Centre, Department of Biomedical Sciences, Faculty of Medicine Health and Human Sciences, Macquarie University, Sydney, NSW 2109, Australia; (D.C.S.T.); (S.J.); (Y.D.); (N.M.); (G.C.); (Y.D.K.)
| |
Collapse
|
2
|
Tsitkov S, Valentine K, Kozareva V, Donde A, Frank A, Lei S, E Van Eyk J, Finkbeiner S, Rothstein JD, Thompson LM, Sareen D, Svendsen CN, Fraenkel E. Disease related changes in ATAC-seq of iPSC-derived motor neuron lines from ALS patients and controls. Nat Commun 2024; 15:3606. [PMID: 38697975 PMCID: PMC11066062 DOI: 10.1038/s41467-024-47758-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Accepted: 04/09/2024] [Indexed: 05/05/2024] Open
Abstract
Amyotrophic Lateral Sclerosis (ALS), like many other neurodegenerative diseases, is highly heritable, but with only a small fraction of cases explained by monogenic disease alleles. To better understand sporadic ALS, we report epigenomic profiles, as measured by ATAC-seq, of motor neuron cultures derived from a diverse group of 380 ALS patients and 80 healthy controls. We find that chromatin accessibility is heavily influenced by sex, the iPSC cell type of origin, ancestry, and the inherent variance arising from sequencing. Once these covariates are corrected for, we are able to identify ALS-specific signals in the data. Additionally, we find that the ATAC-seq data is able to predict ALS disease progression rates with similar accuracy to methods based on biomarkers and clinical status. These results suggest that iPSC-derived motor neurons recapitulate important disease-relevant epigenomic changes.
Collapse
Affiliation(s)
- Stanislav Tsitkov
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Kelsey Valentine
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Velina Kozareva
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Aneesh Donde
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Aaron Frank
- Cedars-Sinai Biomanufacturing Center, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Susan Lei
- Cedars-Sinai Biomanufacturing Center, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Jennifer E Van Eyk
- Advanced Clinical Biosystems Research Institute, Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Steve Finkbeiner
- Center for Systems and Therapeutics, Gladstone Institutes, San Francisco, CA, USA
- Taube/Koret Center for Neurodegenerative Disease, Gladstone Institutes, San Francisco, CA, USA
- Departments of Neurology and Physiology, University of California, San Francisco, San Francisco, CA, USA
| | - Jeffrey D Rothstein
- Brain Science Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Leslie M Thompson
- Department of Neurobiology and Behavior, University of California, Irvine, CA, USA
- Department of Psychiatry and Human Behavior, University of California, Irvine, CA, USA
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, CA, USA
- Sue and Bill Gross Stem Cell Center, University of California, Irvine, CA, USA
| | - Dhruv Sareen
- Cedars-Sinai Biomanufacturing Center, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- The Board of Governors Regenerative Medicine Institute and Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Clive N Svendsen
- The Board of Governors Regenerative Medicine Institute and Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Ernest Fraenkel
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA.
| |
Collapse
|
3
|
Takahashi K, Rensing NR, Eultgen EM, Wang SH, Nelvagal HR, Le SQ, Roberts MS, Doray B, Han EB, Dickson PI, Wong M, Sands MS, Cooper JD. GABAergic interneurons contribute to the fatal seizure phenotype of CLN2 disease mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.29.587276. [PMID: 38585903 PMCID: PMC10996664 DOI: 10.1101/2024.03.29.587276] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/09/2024]
Abstract
GABAergic interneuron deficits have been implicated in the epileptogenesis of multiple neurological diseases. While epileptic seizures are a key clinical hallmark of CLN2 disease, a childhood-onset neurodegenerative lysosomal storage disorder caused by a deficiency of tripeptidyl peptidase 1 (TPP1), the etiology of these seizures remains elusive. Given that Cln2 R207X/R207X mice display fatal spontaneous seizures and an early loss of several cortical interneuron populations, we hypothesized that those two events might be causally related. To address this hypothesis, we first generated an inducible transgenic mouse expressing lysosomal membrane-tethered TPP1 (TPP1LAMP1) on the Cln2 R207X/R207X genetic background to study the cell-autonomous effects of cell-type-specific TPP1 deficiency. We crossed the TPP1LAMP1 mice with Vgat-Cre mice to introduce interneuron-specific TPP1 deficiency. Vgat-Cre ; TPP1LAMP1 mice displayed storage material accumulation in several interneuron populations both in cortex and striatum, and increased susceptibility to die after PTZ-induced seizures. Secondly, to test the role of GABAergic interneuron activity in seizure progression, we selectively activated these cells in Cln2 R207X/R207X mice using Designer Receptor Exclusively Activated by Designer Drugs (DREADDs) in in Vgat-Cre : Cln2 R207X/R207X mice. EEG monitoring revealed that DREADD-mediated activation of interneurons via chronic deschloroclozapine administration accelerated the onset of spontaneous seizures and seizure-associated death in Vgat-Cre : Cln2 R207X/R207X mice, suggesting that modulating interneuron activity can exert influence over epileptiform abnormalities in CLN2 disease. Taken together, these results provide new mechanistic insights into the underlying etiology of seizures and premature death that characterize CLN2 disease.
Collapse
|
4
|
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
|
5
|
Brimble E, Reyes KG, Kuhathaas K, Devinsky O, Ruzhnikov MRZ, Ortiz-Gonzalez XR, Scheffer I, Bahi-Buisson N, Olson H. Expanding genotype-phenotype correlations in FOXG1 syndrome: results from a patient registry. Orphanet J Rare Dis 2023; 18:149. [PMID: 37308910 PMCID: PMC10262363 DOI: 10.1186/s13023-023-02745-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Accepted: 05/18/2023] [Indexed: 06/14/2023] Open
Abstract
BACKGROUND We refine the clinical spectrum of FOXG1 syndrome and expand genotype-phenotype correlations through evaluation of 122 individuals enrolled in an international patient registry. METHODS The FOXG1 syndrome online patient registry allows for remote collection of caregiver-reported outcomes. Inclusion required documentation of a (likely) pathogenic variant in FOXG1. Caregivers were administered a questionnaire to evaluate clinical severity of core features of FOXG1 syndrome. Genotype-phenotype correlations were determined using nonparametric analyses. RESULTS We studied 122 registry participants with FOXG1 syndrome, aged < 12 months to 24 years. Caregivers described delayed or absent developmental milestone attainment, seizures (61%), and movement disorders (58%). Participants harbouring a missense variant had a milder phenotype. Compared to individuals with gene deletions (0%) or nonsense variants (20%), missense variants were associated with more frequent attainment of sitting (73%). Further, individuals with missense variants (41%) achieved independent walking more frequently than those with gene deletions (0%) or frameshift variants (6%). Presence of epilepsy also varied by genotype and was significantly more common in those with gene deletions (81%) compared to missense variants (47%). Individuals with gene deletions were more likely to have higher seizure burden than other genotypes with 53% reporting daily seizures, even at best control. We also observed that truncations preserving the forkhead DNA binding domain were associated with better developmental outcomes. CONCLUSION We refine the phenotypic spectrum of neurodevelopmental features associated with FOXG1 syndrome. We strengthen genotype-driven outcomes, where missense variants are associated with a milder clinical course.
Collapse
|
6
|
Dec K, Alsaqati M, Morgan J, Deshpande S, Wood J, Hall J, Harwood AJ. A high ratio of linoleic acid (n-6 PUFA) to alpha-linolenic acid (n-3 PUFA) adversely affects early stage of human neuronal differentiation and electrophysiological activity of glutamatergic neurons in vitro. Front Cell Dev Biol 2023; 11:1166808. [PMID: 37255597 PMCID: PMC10225581 DOI: 10.3389/fcell.2023.1166808] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Accepted: 04/27/2023] [Indexed: 06/01/2023] Open
Abstract
Introduction: There is a growing interest in the possibility of dietary supplementation with polyunsaturated fatty acids (PUFAs) for treatment and prevention of neurodevelopmental and neuropsychiatric disorders. Studies have suggested that of the two important classes of polyunsaturated fatty acids, omega-6 (n-6) and omega-3 (n-3), n-3 polyunsaturated fatty acids support brain development and function, and when used as a dietary supplement may have beneficial effects for maintenance of a healthy brain. However, to date epidemiological studies and clinical trials on children and adults have been inconclusive regarding treatment length, dosage and use of specific n-3 polyunsaturated fatty acids. The aim of this study is to generate a simplified in vitro cell-based model system to test how different n-6 to n-3 polyunsaturated fatty acids ratios affect human-derived neurons activity as a cellular correlate for brain function and to probe the mechanism of their action. Methods: All experiments were performed by use of human induced pluripotent stem cells (iPSCs). In this study, we examined the effect of different ratios of linoleic acid (n-6) to alpha-linolenic acid in cell growth medium on induced pluripotent stem cell proliferation, generation of neuronal precursors and electrophysiology of cortical glutamatergic neurons by multielectrode array (MEA) analysis. Results: This study shows that at a n-6:n-3 ratio of 5:1 polyunsaturated fatty acids induce stem cell proliferation, generating a large increase in number of cells after 72 h treatment; suppress generation of neuronal progenitor cells, as measured by decreased expression of FOXG1 and Nestin in neuronal precursor cells (NPC) after 20 days of development; and disrupt neuronal activity in vitro, increasing spontaneous neuronal firing, reducing synchronized bursting receptor subunits. We observed no significant differences for neuronal precursor cells treated with ratios 1:3 and 3:1, in comparison to 1:1 control ratio, but higher ratios of n-6 to n-3 polyunsaturated fatty acids adversely affect early stages of neuronal differentiation. Moreover, a 5:1 ratio in cortical glutamatergic neurons induce expression of GABA receptors which may explain the observed abnormal electrophysiological activity.
Collapse
Affiliation(s)
- Karolina Dec
- Neuroscience and Mental Health Innovation Institute, School of Medicine, Cardiff University, Cardiff, Wales, United Kingdom
- Division of Psychological Medicine and Clinical Neurosciences, School of Medicine, Cardiff University, Cardiff, Wales, United Kingdom
| | - Mouhamed Alsaqati
- Neuroscience and Mental Health Innovation Institute, School of Medicine, Cardiff University, Cardiff, Wales, United Kingdom
- School of Pharmacy, Newcastle University, Newcastle Upon Tyne, England, United Kingdom
| | - Joanne Morgan
- Division of Psychological Medicine and Clinical Neurosciences, School of Medicine, Cardiff University, Cardiff, Wales, United Kingdom
| | - Sumukh Deshpande
- School of Medicine, Cardiff University, Cardiff, Wales, United Kingdom
| | - Jamie Wood
- Neuroscience and Mental Health Innovation Institute, School of Medicine, Cardiff University, Cardiff, Wales, United Kingdom
- School of Biosciences, Cardiff University, Cardiff, Wales, United Kingdom
| | - Jeremy Hall
- Neuroscience and Mental Health Innovation Institute, School of Medicine, Cardiff University, Cardiff, Wales, United Kingdom
| | - Adrian J. Harwood
- Neuroscience and Mental Health Innovation Institute, School of Medicine, Cardiff University, Cardiff, Wales, United Kingdom
- School of Biosciences, Cardiff University, Cardiff, Wales, United Kingdom
| |
Collapse
|
7
|
Transition from Animal-Based to Human Induced Pluripotent Stem Cells (iPSCs)-Based Models of Neurodevelopmental Disorders: Opportunities and Challenges. Cells 2023; 12:cells12040538. [PMID: 36831205 PMCID: PMC9954744 DOI: 10.3390/cells12040538] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Revised: 01/25/2023] [Accepted: 02/02/2023] [Indexed: 02/11/2023] Open
Abstract
Neurodevelopmental disorders (NDDs) arise from the disruption of highly coordinated mechanisms underlying brain development, which results in impaired sensory, motor and/or cognitive functions. Although rodent models have offered very relevant insights to the field, the translation of findings to clinics, particularly regarding therapeutic approaches for these diseases, remains challenging. Part of the explanation for this failure may be the genetic differences-some targets not being conserved between species-and, most importantly, the differences in regulation of gene expression. This prompts the use of human-derived models to study NDDS. The generation of human induced pluripotent stem cells (hIPSCs) added a new suitable alternative to overcome species limitations, allowing for the study of human neuronal development while maintaining the genetic background of the donor patient. Several hIPSC models of NDDs already proved their worth by mimicking several pathological phenotypes found in humans. In this review, we highlight the utility of hIPSCs to pave new paths for NDD research and development of new therapeutic tools, summarize the challenges and advances of hIPSC-culture and neuronal differentiation protocols and discuss the best way to take advantage of these models, illustrating this with examples of success for some NDDs.
Collapse
|
8
|
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
|
9
|
Multimodal epigenetic changes and altered NEUROD1 chromatin binding in the mouse hippocampus underlie FOXG1 syndrome. Proc Natl Acad Sci U S A 2023; 120:e2122467120. [PMID: 36598943 PMCID: PMC9926245 DOI: 10.1073/pnas.2122467120] [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] [Indexed: 01/05/2023] Open
Abstract
Forkhead box G1 (FOXG1) has important functions in neuronal differentiation and balances excitatory/inhibitory network activity. Thus far, molecular processes underlying FOXG1 function are largely unexplored. Here, we present a multiomics data set exploring how FOXG1 impacts neuronal maturation at the chromatin level in the mouse hippocampus. At a genome-wide level, FOXG1 i) both represses and activates transcription, ii) binds mainly to enhancer regions, iii) reconfigures the epigenetic landscape through bidirectional alteration of H3K27ac, H3K4me3, and chromatin accessibility, and iv) operates synergistically with NEUROD1. Interestingly, we could not detect a clear hierarchy of FOXG1 and NEUROD1, but instead, provide the evidence that they act in a highly cooperative manner to control neuronal maturation. Genes affected by the chromatin alterations impact synaptogenesis and axonogenesis. Inhibition of histone deacetylases partially rescues transcriptional alterations upon FOXG1 reduction. This integrated multiomics view of changes upon FOXG1 reduction reveals an unprecedented multimodality of FOXG1 functions converging on neuronal maturation. It fuels therapeutic options based on epigenetic drugs to alleviate, at least in part, neuronal dysfunction.
Collapse
|
10
|
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: 1] [Impact Index Per Article: 1.0] [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
|
11
|
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: 12] [Impact Index Per Article: 6.0] [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
|
12
|
Seo Y, Bang S, Son J, Kim D, Jeong Y, Kim P, Yang J, Eom JH, Choi N, Kim HN. Brain physiome: A concept bridging in vitro 3D brain models and in silico models for predicting drug toxicity in the brain. Bioact Mater 2022; 13:135-148. [PMID: 35224297 PMCID: PMC8843968 DOI: 10.1016/j.bioactmat.2021.11.009] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 11/01/2021] [Accepted: 11/06/2021] [Indexed: 12/12/2022] Open
Abstract
In the last few decades, adverse reactions to pharmaceuticals have been evaluated using 2D in vitro models and animal models. However, with increasing computational power, and as the key drivers of cellular behavior have been identified, in silico models have emerged. These models are time-efficient and cost-effective, but the prediction of adverse reactions to unknown drugs using these models requires relevant experimental input. Accordingly, the physiome concept has emerged to bridge experimental datasets with in silico models. The brain physiome describes the systemic interactions of its components, which are organized into a multilevel hierarchy. Because of the limitations in obtaining experimental data corresponding to each physiome component from 2D in vitro models and animal models, 3D in vitro brain models, including brain organoids and brain-on-a-chip, have been developed. In this review, we present the concept of the brain physiome and its hierarchical organization, including cell- and tissue-level organizations. We also summarize recently developed 3D in vitro brain models and link them with the elements of the brain physiome as a guideline for dataset collection. The connection between in vitro 3D brain models and in silico modeling will lead to the establishment of cost-effective and time-efficient in silico models for the prediction of the safety of unknown drugs.
Collapse
Affiliation(s)
- Yoojin Seo
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
| | - Seokyoung Bang
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
| | - Jeongtae Son
- Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Republic of Korea
| | - Dongsup Kim
- Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Republic of Korea
| | - Yong Jeong
- Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Republic of Korea
| | - Pilnam Kim
- Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Republic of Korea
| | - Jihun Yang
- Next&Bio Inc., Seoul, 02841, Republic of Korea
| | - Joon-Ho Eom
- Medical Device Research Division, National Institute of Food and Drug Safety Evaluation, Cheongju, 28159, Republic of Korea
| | - Nakwon Choi
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
- Division of Bio-Medical Science & Technology, KIST School, Korea University of Science and Technology (UST), Seoul, 02792, Republic of Korea
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, 02841, Republic of Korea
| | - Hong Nam Kim
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
- Division of Bio-Medical Science & Technology, KIST School, Korea University of Science and Technology (UST), Seoul, 02792, Republic of Korea
- School of Mechanical Engineering, Yonsei University, Seoul, 03722, Republic of Korea
- Yonsei-KIST Convergence Research Institute, Yonsei University, Seoul, 03722, Republic of Korea
| |
Collapse
|
13
|
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: 7.0] [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
|
14
|
Hettige NC, Peng H, Wu H, Zhang X, Yerko V, Zhang Y, Jefri M, Soubannier V, Maussion G, Alsuwaidi S, Ni A, Rocha C, Krishnan J, McCarty V, Antonyan L, Schuppert A, Turecki G, Fon EA, Durcan TM, Ernst C. FOXG1 dose tunes cell proliferation dynamics in human forebrain progenitor cells. Stem Cell Reports 2022; 17:475-488. [PMID: 35148845 PMCID: PMC9040178 DOI: 10.1016/j.stemcr.2022.01.010] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Revised: 01/12/2022] [Accepted: 01/13/2022] [Indexed: 10/26/2022] Open
Abstract
Heterozygous loss-of-function mutations in Forkhead box G1 (FOXG1), a uniquely brain-expressed gene, cause microcephaly, seizures, and severe intellectual disability, whereas increased FOXG1 expression is frequently observed in glioblastoma. To investigate the role of FOXG1 in forebrain cell proliferation, we modeled FOXG1 syndrome using cells from three clinically diagnosed cases with two sex-matched healthy parents and one unrelated sex-matched control. Cells with heterozygous FOXG1 loss showed significant reduction in cell proliferation, increased ratio of cells in G0/G1 stage of the cell cycle, and increased frequency of primary cilia. Engineered loss of FOXG1 recapitulated this effect, while isogenic repair of a patient mutation reverted output markers to wild type. An engineered inducible FOXG1 cell line derived from a FOXG1 syndrome case demonstrated that FOXG1 dose-dependently affects all cell proliferation outputs measured. These findings provide strong support for the critical importance of FOXG1 levels in controlling human brain cell growth in health and disease.
Collapse
Affiliation(s)
- Nuwan C Hettige
- Department of Human Genetics, McGill University, Montreal, QC H3A 0C7, Canada; Psychiatric Genetics Group, Douglas Mental Health University Institute, 6875 Boulevard LaSalle, Montreal, QC H4H 1R3, Canada
| | - Huashan Peng
- Psychiatric Genetics Group, Douglas Mental Health University Institute, 6875 Boulevard LaSalle, Montreal, QC H4H 1R3, Canada
| | - Hanrong Wu
- Psychiatric Genetics Group, Douglas Mental Health University Institute, 6875 Boulevard LaSalle, Montreal, QC H4H 1R3, Canada
| | - Xin Zhang
- Psychiatric Genetics Group, Douglas Mental Health University Institute, 6875 Boulevard LaSalle, Montreal, QC H4H 1R3, Canada
| | - Volodymyr Yerko
- Department of Psychiatry, McGill University, Montreal, QC H3A 1A1, Canada
| | - Ying Zhang
- Psychiatric Genetics Group, Douglas Mental Health University Institute, 6875 Boulevard LaSalle, Montreal, QC H4H 1R3, Canada
| | - Malvin Jefri
- Psychiatric Genetics Group, Douglas Mental Health University Institute, 6875 Boulevard LaSalle, Montreal, QC H4H 1R3, Canada; Integrated Program in Neuroscience, McGill University, Montreal, QC H3A 2B4, Canada
| | - Vincent Soubannier
- McGill Parkinson Program and Neurodegenerative Diseases Group, Montreal Neurological Institute, Department of Neurology and Neurosurgery, Montreal, QC H3A 2B4, Canada; The Neuro's Early Drug Discovery Unit (EDDU), McGill University, 3801 University Street, Montreal, QC H3A 2B4, Canada
| | - Gilles Maussion
- McGill Parkinson Program and Neurodegenerative Diseases Group, Montreal Neurological Institute, Department of Neurology and Neurosurgery, Montreal, QC H3A 2B4, Canada; The Neuro's Early Drug Discovery Unit (EDDU), McGill University, 3801 University Street, Montreal, QC H3A 2B4, Canada
| | - Shaima Alsuwaidi
- Department of Human Genetics, McGill University, Montreal, QC H3A 0C7, Canada; Psychiatric Genetics Group, Douglas Mental Health University Institute, 6875 Boulevard LaSalle, Montreal, QC H4H 1R3, Canada
| | - Anjie Ni
- Department of Human Genetics, McGill University, Montreal, QC H3A 0C7, Canada; Psychiatric Genetics Group, Douglas Mental Health University Institute, 6875 Boulevard LaSalle, Montreal, QC H4H 1R3, Canada
| | - Cecilia Rocha
- The Neuro's Early Drug Discovery Unit (EDDU), McGill University, 3801 University Street, Montreal, QC H3A 2B4, Canada
| | - Jeyashree Krishnan
- Institute for Computational Biomedicine, Aachen University, Pauwelsstraße 19, 52074 Aachen, Germany
| | - Vincent McCarty
- Department of Human Genetics, McGill University, Montreal, QC H3A 0C7, Canada; Psychiatric Genetics Group, Douglas Mental Health University Institute, 6875 Boulevard LaSalle, Montreal, QC H4H 1R3, Canada
| | - Lilit Antonyan
- Department of Human Genetics, McGill University, Montreal, QC H3A 0C7, Canada; Psychiatric Genetics Group, Douglas Mental Health University Institute, 6875 Boulevard LaSalle, Montreal, QC H4H 1R3, Canada
| | - Andreas Schuppert
- Institute for Computational Biomedicine, Aachen University, Pauwelsstraße 19, 52074 Aachen, Germany
| | - Gustavo Turecki
- Department of Human Genetics, McGill University, Montreal, QC H3A 0C7, Canada; Department of Psychiatry, McGill University, Montreal, QC H3A 1A1, Canada; Integrated Program in Neuroscience, McGill University, Montreal, QC H3A 2B4, Canada
| | - Edward A Fon
- McGill Parkinson Program and Neurodegenerative Diseases Group, Montreal Neurological Institute, Department of Neurology and Neurosurgery, Montreal, QC H3A 2B4, Canada
| | - Thomas M Durcan
- McGill Parkinson Program and Neurodegenerative Diseases Group, Montreal Neurological Institute, Department of Neurology and Neurosurgery, Montreal, QC H3A 2B4, Canada; The Neuro's Early Drug Discovery Unit (EDDU), McGill University, 3801 University Street, Montreal, QC H3A 2B4, Canada
| | - Carl Ernst
- Department of Human Genetics, McGill University, Montreal, QC H3A 0C7, Canada; Psychiatric Genetics Group, Douglas Mental Health University Institute, 6875 Boulevard LaSalle, Montreal, QC H4H 1R3, Canada; Department of Psychiatry, McGill University, Montreal, QC H3A 1A1, Canada; Integrated Program in Neuroscience, McGill University, Montreal, QC H3A 2B4, Canada.
| |
Collapse
|
15
|
Akol I, Gather F, Vogel T. Paving Therapeutic Avenues for FOXG1 Syndrome: Untangling Genotypes and Phenotypes from a Molecular Perspective. Int J Mol Sci 2022; 23:ijms23020954. [PMID: 35055139 PMCID: PMC8780739 DOI: 10.3390/ijms23020954] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Revised: 12/23/2021] [Accepted: 01/13/2022] [Indexed: 02/01/2023] Open
Abstract
Development of the central nervous system (CNS) depends on accurate spatiotemporal control of signaling pathways and transcriptional programs. Forkhead Box G1 (FOXG1) is one of the master regulators that play fundamental roles in forebrain development; from the timing of neurogenesis, to the patterning of the cerebral cortex. Mutations in the FOXG1 gene cause a rare neurodevelopmental disorder called FOXG1 syndrome, also known as congenital form of Rett syndrome. Patients presenting with FOXG1 syndrome manifest a spectrum of phenotypes, ranging from severe cognitive dysfunction and microcephaly to social withdrawal and communication deficits, with varying severities. To develop and improve therapeutic interventions, there has been considerable progress towards unravelling the multi-faceted functions of FOXG1 in the neurodevelopment and pathogenesis of FOXG1 syndrome. Moreover, recent advances in genome editing and stem cell technologies, as well as the increased yield of information from high throughput omics, have opened promising and important new avenues in FOXG1 research. In this review, we provide a summary of the clinical features and emerging molecular mechanisms underlying FOXG1 syndrome, and explore disease-modelling approaches in animals and human-based systems, to highlight the prospects of research and possible clinical interventions.
Collapse
Affiliation(s)
- Ipek Akol
- Department of Molecular Embryology, Institute for Anatomy and Cell Biology, Medical Faculty, University of Freiburg, 79104 Freiburg, Germany; (I.A.); (F.G.)
- Faculty of Biology, University of Freiburg, 79104 Freiburg, Germany
- Center for Basics in NeuroModulation (NeuroModul Basics), Medical Faculty, Albert-Ludwigs-University Freiburg, 79104 Freiburg, Germany
| | - Fabian Gather
- Department of Molecular Embryology, Institute for Anatomy and Cell Biology, Medical Faculty, University of Freiburg, 79104 Freiburg, Germany; (I.A.); (F.G.)
| | - Tanja Vogel
- Department of Molecular Embryology, Institute for Anatomy and Cell Biology, Medical Faculty, University of Freiburg, 79104 Freiburg, Germany; (I.A.); (F.G.)
- Center for Basics in NeuroModulation (NeuroModul Basics), Medical Faculty, Albert-Ludwigs-University Freiburg, 79104 Freiburg, Germany
- Correspondence:
| |
Collapse
|
16
|
Lutz AK, Pérez Arévalo A, Ioannidis V, Stirmlinger N, Demestre M, Delorme R, Bourgeron T, Boeckers TM. SHANK2 Mutations Result in Dysregulation of the ERK1/2 Pathway in Human Induced Pluripotent Stem Cells-Derived Neurons and Shank2(-/-) Mice. Front Mol Neurosci 2021; 14:773571. [PMID: 34899182 PMCID: PMC8662699 DOI: 10.3389/fnmol.2021.773571] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Accepted: 10/07/2021] [Indexed: 12/28/2022] Open
Abstract
SHANK2 (ProSAP1) is a postsynaptic scaffolding protein of excitatory synapses in the central nervous system and implicated in the development of autism spectrum disorders (ASD). Patients with mutations in SHANK2 show autism-like behaviors, developmental delay, and intellectual disability. We generated human induced pluripotent stem cells (hiPSC) from a patient carrying a heterozygous deletion of SHANK2 and from the unaffected parents. In patient hiPSCs and derived neurons SHANK2 mRNA and protein expression was reduced. During neuronal maturation, a reduction in growth cone size and a transient increase in neuronal soma size were observed. Neuronal proliferation was increased, and apoptosis was decreased in young and mature neurons. Additionally, mature patient hiPSC-derived neurons showed dysregulated excitatory signaling and a decrease of a broad range of signaling molecules of the ERK-MAP kinase pathway. These findings could be confirmed in brain samples from Shank2(−/−) mice, which also showed decreased mGluR5 and phospho-ERK1/2 expression. Our study broadens the current knowledge of SHANK2-related ASD. We highlight the importance of excitatory-inhibitory balance and mGluR5 dysregulation with disturbed downstream ERK1/2 signaling in ASD, which provides possible future therapeutic strategies for SHANK2-related ASD.
Collapse
Affiliation(s)
- Anne-Kathrin Lutz
- Institute for Anatomy and Cell Biology, Ulm University, Ulm, Germany
| | | | | | | | - Maria Demestre
- Institute for Anatomy and Cell Biology, Ulm University, Ulm, Germany
| | | | - Thomas Bourgeron
- Génétique Humaine et Fonctions Cognitives, Institut Pasteur, Université Paris Diderot, Paris, France
| | - Tobias M Boeckers
- Institute for Anatomy and Cell Biology, Ulm University, Ulm, Germany.,Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), Ulm Site, Ulm, Germany
| |
Collapse
|
17
|
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
|
18
|
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: 1.3] [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
|
19
|
Sabitha KR, Shetty AK, Upadhya D. Patient-derived iPSC modeling of rare neurodevelopmental disorders: Molecular pathophysiology and prospective therapies. Neurosci Biobehav Rev 2020; 121:201-219. [PMID: 33370574 DOI: 10.1016/j.neubiorev.2020.12.025] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Revised: 12/18/2020] [Accepted: 12/19/2020] [Indexed: 12/12/2022]
Abstract
The pathological alterations that manifest during the early embryonic development due to inherited and acquired factors trigger various neurodevelopmental disorders (NDDs). Besides major NDDs, there are several rare NDDs, exhibiting specific characteristics and varying levels of severity triggered due to genetic and epigenetic anomalies. The rarity of subjects, paucity of neural tissues for detailed analysis, and the unavailability of disease-specific animal models have hampered detailed comprehension of rare NDDs, imposing heightened challenge to the medical and scientific community until a decade ago. The generation of functional neurons and glia through directed differentiation protocols for patient-derived iPSCs, CRISPR/Cas9 technology, and 3D brain organoid models have provided an excellent opportunity and vibrant resource for decoding the etiology of brain development for rare NDDs caused due to monogenic as well as polygenic disorders. The present review identifies cellular and molecular phenotypes demonstrated from patient-derived iPSCs and possible therapeutic opportunities identified for these disorders. New insights to reinforce the existing knowledge of the pathophysiology of these disorders and prospective therapeutic applications are discussed.
Collapse
Affiliation(s)
- K R Sabitha
- Centre for Molecular Neurosciences, Kasturba Medical College, Manipal Academy of Higher Education, Manipal, 576104, Karnataka, India
| | - Ashok K Shetty
- Institute for Regenerative Medicine, Department of Molecular and Cellular Medicine, Texas A&M University College of Medicine, College Station, TX, USA.
| | - Dinesh Upadhya
- Centre for Molecular Neurosciences, Kasturba Medical College, Manipal Academy of Higher Education, Manipal, 576104, Karnataka, India.
| |
Collapse
|
20
|
Ye XC, Roslin NM, Paterson AD, Lyons CJ, Pegado V, Richmond P, Shyr C, Fornes O, Han X, Higginson M, Ross CJ, Giaschi D, Gregory-Evans C, Patel MS, Wasserman WW. Linkage analysis identifies an isolated strabismus locus at 14q12 overlapping with FOXG1 syndrome region. J Med Genet 2020; 59:46-55. [PMID: 33257509 PMCID: PMC8685624 DOI: 10.1136/jmedgenet-2020-107226] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Revised: 09/05/2020] [Accepted: 09/29/2020] [Indexed: 11/21/2022]
Abstract
Strabismus is a common condition, affecting 1%–4% of individuals. Isolated strabismus has been studied in families with Mendelian inheritance patterns. Despite the identification of multiple loci via linkage analyses, no specific genes have been identified from these studies. The current study is based on a seven-generation family with isolated strabismus inherited in an autosomal dominant manner. A total of 13 individuals from a common ancestor have been included for linkage analysis. Among these, nine are affected and four are unaffected. A single linkage signal has been identified at an 8.5 Mb region of chromosome 14q12 with a multipoint LOD (logarithm of the odds) score of 4.69. Disruption of this locus is known to cause FOXG1 syndrome (or congenital Rett syndrome; OMIM #613454 and *164874), in which 84% of affected individuals present with strabismus. With the incorporation of next-generation sequencing and in-depth bioinformatic analyses, a 4 bp non-coding deletion was prioritised as the top candidate for the observed strabismus phenotype. The deletion is predicted to disrupt regulation of FOXG1, which encodes a transcription factor of the Forkhead family. Suggestive of an autoregulation effect, the disrupted sequence matches the consensus FOXG1 and Forkhead family transcription factor binding site and has been observed in previous ChIP-seq studies to be bound by Foxg1 in early mouse brain development. Future study of this specific deletion may shed light on the regulation of FOXG1 expression and may enhance our understanding of the mechanisms contributing to strabismus and FOXG1 syndrome.
Collapse
Affiliation(s)
- Xin Cynthia Ye
- Centre for Molecular Medicine and Therapeutics, The University of British Columbia, Vancouver, British Columbia, Canada.,Department of Medical Genetics, The University of British Columbia, Vancouver, British Columbia, Canada.,BC Children's Hospital Research Institute, The University of British Columbia, Vancouver, British Columbia, Canada
| | - Nicole M Roslin
- The Centre for Applied Genomics, Hospital for Sick Children Research Institute, University of Toronto, Toronto, Ontario, Canada
| | - Andrew D Paterson
- The Centre for Applied Genomics, Hospital for Sick Children Research Institute, University of Toronto, Toronto, Ontario, Canada.,Genetics and Genome Biology, Hospital for Sick Children, Toronto, Ontario, Canada.,Divisions of Epidemiology and Biostatistics, Dalla Lana School of Public Health, University of Toronto, Toronto, Ontario, Canada
| | - Christopher J Lyons
- BC Children's Hospital Research Institute, The University of British Columbia, Vancouver, British Columbia, Canada.,Department of Ophthalmology and Visual Sciences, The University of British Columbia, Vancouver, British Columbia, Canada
| | - Victor Pegado
- Department of Ophthalmology and Visual Sciences, The University of British Columbia, Vancouver, British Columbia, Canada
| | - Phillip Richmond
- Centre for Molecular Medicine and Therapeutics, The University of British Columbia, Vancouver, British Columbia, Canada.,BC Children's Hospital Research Institute, The University of British Columbia, Vancouver, British Columbia, Canada
| | - Casper Shyr
- Centre for Molecular Medicine and Therapeutics, The University of British Columbia, Vancouver, British Columbia, Canada.,BC Children's Hospital Research Institute, The University of British Columbia, Vancouver, British Columbia, Canada
| | - Oriol Fornes
- Centre for Molecular Medicine and Therapeutics, The University of British Columbia, Vancouver, British Columbia, Canada.,BC Children's Hospital Research Institute, The University of British Columbia, Vancouver, British Columbia, Canada
| | - XiaoHua Han
- Centre for Molecular Medicine and Therapeutics, The University of British Columbia, Vancouver, British Columbia, Canada
| | - Michelle Higginson
- Centre for Molecular Medicine and Therapeutics, The University of British Columbia, Vancouver, British Columbia, Canada
| | - Colin J Ross
- Faculty of Pharmaceutical Sciences, The University of British Columbia, Vancouver, British Columbia, Canada
| | - Deborah Giaschi
- BC Children's Hospital Research Institute, The University of British Columbia, Vancouver, British Columbia, Canada.,Department of Ophthalmology and Visual Sciences, The University of British Columbia, Vancouver, British Columbia, Canada
| | - Cheryl Gregory-Evans
- Department of Ophthalmology and Visual Sciences, The University of British Columbia, Vancouver, British Columbia, Canada
| | - Millan S Patel
- Department of Medical Genetics, The University of British Columbia, Vancouver, British Columbia, Canada .,BC Children's Hospital Research Institute, The University of British Columbia, Vancouver, British Columbia, Canada
| | - Wyeth W Wasserman
- Centre for Molecular Medicine and Therapeutics, The University of British Columbia, Vancouver, British Columbia, Canada .,Department of Medical Genetics, The University of British Columbia, Vancouver, British Columbia, Canada.,BC Children's Hospital Research Institute, The University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
21
|
Falcone C, Santo M, Liuzzi G, Cannizzaro N, Grudina C, Valencic E, Peruzzotti-Jametti L, Pluchino S, Mallamaci A. Foxg1 Antagonizes Neocortical Stem Cell Progression to Astrogenesis. Cereb Cortex 2020; 29:4903-4918. [PMID: 30821834 DOI: 10.1093/cercor/bhz031] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2018] [Revised: 01/06/2019] [Accepted: 02/09/2019] [Indexed: 12/12/2022] Open
Abstract
Neocortical astrogenesis follows neuronogenesis and precedes oligogenesis. Among key factors dictating its temporal articulation, there are progression rates of pallial stem cells (SCs) towards astroglial lineages as well as activation rates of astrocyte differentiation programs in response to extrinsic gliogenic cues. In this study, we showed that high Foxg1 SC expression antagonizes astrocyte generation, while stimulating SC self-renewal and committing SCs to neuronogenesis. We found that mechanisms underlying this activity are mainly cell autonomous and highly pleiotropic. They include a concerted downregulation of 4 key effectors channeling neural SCs to astroglial fates, as well as defective activation of core molecular machineries implementing astroglial differentiation programs. Next, we found that SC Foxg1 levels specifically decline during the neuronogenic-to-gliogenic transition, pointing to a pivotal Foxg1 role in temporal modulation of astrogenesis. Finally, we showed that Foxg1 inhibits astrogenesis from human neocortical precursors, suggesting that this is an evolutionarily ancient trait.
Collapse
Affiliation(s)
- Carmen Falcone
- Laboratory of Cerebral Cortex Development, Neuroscience Area, SISSA, Trieste, Italy
| | - Manuela Santo
- Laboratory of Cerebral Cortex Development, Neuroscience Area, SISSA, Trieste, Italy
| | - Gabriele Liuzzi
- Laboratory of Cerebral Cortex Development, Neuroscience Area, SISSA, Trieste, Italy
| | - Noemi Cannizzaro
- Laboratory of Cerebral Cortex Development, Neuroscience Area, SISSA, Trieste, Italy
| | - Clara Grudina
- Laboratory of Cerebral Cortex Development, Neuroscience Area, SISSA, Trieste, Italy
| | - Erica Valencic
- Department of Diagnostics, Institute for Maternal and Child Health, IRCCS Burlo Garofolo, Trieste, Italy
| | - Luca Peruzzotti-Jametti
- Dept of Clinical Neurosciences, University of Cambridge, Clifford Allbutt Building -- Cambridge Biosciences Campus, Hills Road, Cambridge, UK
| | - Stefano Pluchino
- Dept of Clinical Neurosciences, University of Cambridge, Clifford Allbutt Building -- Cambridge Biosciences Campus, Hills Road, Cambridge, UK
| | - Antonello Mallamaci
- Laboratory of Cerebral Cortex Development, Neuroscience Area, SISSA, Trieste, Italy
| |
Collapse
|
22
|
Hoover AH, Pavuluri R, Shelkar GP, Dravid SM, Smith Y, Villalba RM. Ultrastructural localization of glutamate delta 1 (GluD1) receptor immunoreactivity in the mouse and monkey striatum. J Comp Neurol 2020; 529:1703-1718. [PMID: 33084025 DOI: 10.1002/cne.25051] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Revised: 10/04/2020] [Accepted: 10/05/2020] [Indexed: 12/26/2022]
Abstract
The glutamate receptor delta 1 (GluD1) is strongly expressed in the striatum. Knockout of GluD1 expression in striatal neurons elicits cognitive deficits and disrupts the thalamostriatal system in mice. To understand the potential role of GluD1 in the primate striatum, we compared the cellular and subcellular localization of striatal GluD1 immunoreactivity (GluD1-IR) in mice and monkeys. In both species, striatal GluD1-IR displayed a patchy pattern of distribution in register with the striosome/matrix compartmentation, but in an opposite fashion. While GluD1 was more heavily expressed in the striosomes than the matrix in the monkey caudate nucleus, the opposite was found in the mouse striatum. At the electron microscopic level, GluD1-IR was preferentially expressed in dendritic shafts (47.9 ± 1.2%), followed by glia (37.7 ± 2.5%), and dendritic spines (14.3 ± 2.6%) in the matrix of the mouse striatum. This pattern was not statistically different from the labeling in the striosome and matrix compartments of the monkey caudate nucleus, with the exception of a small amount of GluD1-positive unmyelinated axons and axon terminals in the primate striatum. Immunogold staining revealed synaptic and perisynaptic GluD1 labeling at putative axo-dendritic and axo-spinous glutamatergic synapses, and intracellular labeling on the surface of mitochondria. Confocal microscopy showed that GluD1 is preferentially colocalized with thalamic over cortical terminals in both the striosome and matrix compartments. These data provide the anatomical substrate for a deeper understanding of GluD1 regulation of striatal glutamatergic synapses, but also suggest possible extrasynaptic, glial, and mitochondrial GluD1 functions.
Collapse
Affiliation(s)
- Andrew H Hoover
- Yerkes National Primate Research Center, Atlanta, Georgia, USA.,UDALL Center of Excellence for Parkinson's Disease, Atlanta, Georgia, USA
| | - Ratnamala Pavuluri
- Department of Pharmacology and Neuroscience, Creighton University School of Medicine, Omaha, Nebraska, USA
| | - Gajanan P Shelkar
- Department of Pharmacology and Neuroscience, Creighton University School of Medicine, Omaha, Nebraska, USA
| | - Shashank M Dravid
- Department of Pharmacology and Neuroscience, Creighton University School of Medicine, Omaha, Nebraska, USA
| | - Yoland Smith
- Yerkes National Primate Research Center, Atlanta, Georgia, USA.,UDALL Center of Excellence for Parkinson's Disease, Atlanta, Georgia, USA.,Department of Neurology, Emory University, Atlanta, Georgia, USA
| | - Rosa M Villalba
- Yerkes National Primate Research Center, Atlanta, Georgia, USA.,UDALL Center of Excellence for Parkinson's Disease, Atlanta, Georgia, USA
| |
Collapse
|
23
|
Hirose S, Tanaka Y, Shibata M, Kimura Y, Ishikawa M, Higurashi N, Yamamoto T, Ichise E, Chiyonobu T, Ishii A. Application of induced pluripotent stem cells in epilepsy. Mol Cell Neurosci 2020; 108:103535. [DOI: 10.1016/j.mcn.2020.103535] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Revised: 07/10/2020] [Accepted: 07/31/2020] [Indexed: 02/06/2023] Open
|
24
|
High rate of HDR in gene editing of p.(Thr158Met) MECP2 mutational hotspot. Eur J Hum Genet 2020; 28:1231-1242. [PMID: 32332872 PMCID: PMC7609331 DOI: 10.1038/s41431-020-0624-x] [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: 06/04/2019] [Revised: 03/16/2020] [Accepted: 03/24/2020] [Indexed: 02/06/2023] Open
Abstract
Rett syndrome is a progressive neurodevelopmental disorder which affects almost exclusively girls, caused by variants in MECP2 gene. Effective therapies for this devastating disorder are not yet available and the need for tight regulation of MECP2 expression for brain to properly function makes gene replacement therapy risky. For this reason, gene editing with CRISPR/Cas9 technology appears as a preferable option for the development of new therapies. To study the disease, we developed and characterized a human neuronal model obtained by genetic reprogramming of patient-derived primary fibroblasts into induced Pluripotent Stem Cells. This cellular model represents an important source for our studies, aiming to correct MECP2 variants in neurons which represent the primarily affected cell type. We engineered a gene editing toolkit composed by a two-plasmid system to correct a hotspot missense variant in MECP2, c.473 C > T (p.(Thr158Met)). The first construct expresses the variant-specific sgRNA and the Donor DNA along with a fluorescent reporter system. The second construct brings Cas9 and targets for auto-cleaving, to avoid long-term Cas9 expression. NGS analysis on sorted cells from four independent patients demonstrated an exceptionally high editing efficiency, with up to 80% of HDR and less than 1% of indels in all patients, outlining the relevant potentiality of the approach for Rett syndrome therapy.
Collapse
|
25
|
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: 1.5] [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
|
26
|
Tigani W, Rossi MP, Artimagnella O, Santo M, Rauti R, Sorbo T, Ulloa Severino FP, Provenzano G, Allegra M, Caleo M, Ballerini L, Bozzi Y, Mallamaci A. Foxg1 Upregulation Enhances Neocortical Activity. Cereb Cortex 2020; 30:5147-5165. [PMID: 32383447 DOI: 10.1093/cercor/bhaa107] [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: 02/20/2020] [Revised: 04/03/2020] [Accepted: 04/05/2020] [Indexed: 12/19/2022] Open
Abstract
Foxg1 is an ancient transcription factor gene orchestrating a number of neurodevelopmental processes taking place in the rostral brain. In this study, we investigated its impact on neocortical activity. We found that mice overexpressing Foxg1 in neocortical pyramidal cells displayed an electroencephalography (EEG) with increased spike frequency and were more prone to kainic acid (KA)-induced seizures. Consistently, primary cultures of neocortical neurons gain-of-function for Foxg1 were hyperactive and hypersynchronized. That reflected an unbalanced expression of key genes encoding for ion channels, gamma aminobutyric acid and glutamate receptors, and was likely exacerbated by a pronounced interneuron depletion. We also detected a transient Foxg1 upregulation ignited in turn by neuronal activity and mediated by immediate early genes. Based on this, we propose that even small changes of Foxg1 levels may result in a profound impact on pyramidal cell activity, an issue relevant to neuronal physiology and neurological aberrancies associated to FOXG1 copy number variations.
Collapse
Affiliation(s)
- Wendalina Tigani
- Laboratory of Cerebral Cortex Development, Neuroscience Area, SISSA, Trieste 34136, Italy
| | - Moira Pinzan Rossi
- Laboratory of Cerebral Cortex Development, Neuroscience Area, SISSA, Trieste 34136, Italy.,AgenTus Therapeutics, Inc., Cambridge CB4 OWG, United Kingdom
| | - Osvaldo Artimagnella
- Laboratory of Cerebral Cortex Development, Neuroscience Area, SISSA, Trieste 34136, Italy
| | - Manuela Santo
- Laboratory of Cerebral Cortex Development, Neuroscience Area, SISSA, Trieste 34136, Italy
| | - Rossana Rauti
- Laboratory of Neurons and Nanomaterials, Neuroscience Area, SISSA, Trieste 34136, Italy.,Dept. Biomedical Engineering, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Teresa Sorbo
- Laboratory of Neurons and Nanomaterials, Neuroscience Area, SISSA, Trieste 34136, Italy
| | - Francesco Paolo Ulloa Severino
- Laboratory of Bionanotechnologies, Neuroscience Area, SISSA, Trieste 34136, Italy.,Cell Biology Dept, Duke University Medical Center, Duke University, Durham NC-27710, USA
| | - Giovanni Provenzano
- Department of Cellular, Computational, and Integrative Biology (CIBIO), University of Trento, Trento 38123, Italy
| | - Manuela Allegra
- Neuroscience Institute, Neurophysiology Section, National Research Council (CNR), Pisa 56124, Italy.,Laboratory G5 Circuits Neuronaux, Institut Pasteur, Paris 75015, France
| | - Matteo Caleo
- Neuroscience Institute, Neurophysiology Section, National Research Council (CNR), Pisa 56124, Italy.,Department of Biomedical Sciences, University of Padua, Padua 35121, Italy
| | - Laura Ballerini
- Laboratory of Neurons and Nanomaterials, Neuroscience Area, SISSA, Trieste 34136, Italy
| | - Yuri Bozzi
- Neuroscience Institute, Neurophysiology Section, National Research Council (CNR), Pisa 56124, Italy.,Center for Mind/Brain Sciences, University of Trento, Trento 38068, Italy
| | - Antonello Mallamaci
- Laboratory of Cerebral Cortex Development, Neuroscience Area, SISSA, Trieste 34136, Italy
| |
Collapse
|
27
|
Hou PS, hAilín DÓ, Vogel T, Hanashima C. Transcription and Beyond: Delineating FOXG1 Function in Cortical Development and Disorders. Front Cell Neurosci 2020; 14:35. [PMID: 32158381 PMCID: PMC7052011 DOI: 10.3389/fncel.2020.00035] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2019] [Accepted: 02/04/2020] [Indexed: 11/13/2022] Open
Abstract
Forkhead Box G1 (FOXG1) is a member of the Forkhead family of genes with non-redundant roles in brain development, where alteration of this gene's expression significantly affects the formation and function of the mammalian cerebral cortex. FOXG1 haploinsufficiency in humans is associated with prominent differences in brain size and impaired intellectual development noticeable in early childhood, while homozygous mutations are typically fatal. As such, FOXG1 has been implicated in a wide spectrum of congenital brain disorders, including the congenital variant of Rett syndrome, infantile spasms, microcephaly, autism spectrum disorder (ASD) and schizophrenia. Recent technological advances have yielded greater insight into phenotypic variations observed in FOXG1 syndrome, molecular mechanisms underlying pathogenesis of the disease, and multifaceted roles of FOXG1 expression. In this review, we explore the emerging mechanisms of FOXG1 in a range of transcriptional to posttranscriptional events in order to evolve our current view of how a single transcription factor governs the assembly of an elaborate cortical circuit responsible for higher cognitive functions and neurological disorders.
Collapse
Affiliation(s)
- Pei-Shan Hou
- Laboratory for Developmental Biology, Department of Biology, Faculty of Education and Integrated Arts and Sciences, Waseda University, Tokyo, Japan.,Institute of Anatomy and Cell Biology, School of Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Darren Ó hAilín
- Department of Molecular Embryology, Institute of Anatomy and Cell Biology, Medical Faculty, University of Freiburg, Freiburg, Germany
| | - Tanja Vogel
- Department of Molecular Embryology, Institute of Anatomy and Cell Biology, Medical Faculty, University of Freiburg, Freiburg, Germany.,Center for Basics in NeuroModulation (NeuroModul Basics), Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Carina Hanashima
- Laboratory for Developmental Biology, Department of Biology, Faculty of Education and Integrated Arts and Sciences, Waseda University, Tokyo, Japan.,Department of Integrative Bioscience and Biomedical Engineering, Graduate School of Advanced Science and Engineering, Waseda University Center for Advanced Biomedical Sciences, Tokyo, Japan
| |
Collapse
|
28
|
Liu J, Shelkar GP, Gandhi PJ, Gawande DY, Hoover A, Villalba RM, Pavuluri R, Smith Y, Dravid SM. Striatal glutamate delta-1 receptor regulates behavioral flexibility and thalamostriatal connectivity. Neurobiol Dis 2020; 137:104746. [PMID: 31945419 PMCID: PMC7204410 DOI: 10.1016/j.nbd.2020.104746] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Revised: 12/19/2019] [Accepted: 01/12/2020] [Indexed: 10/31/2022] Open
Abstract
Impaired behavioral flexibility and repetitive behavior is a common phenotype in autism and other neuropsychiatric disorders, but the underlying synaptic mechanisms are poorly understood. The trans-synaptic glutamate delta (GluD)-Cerebellin 1-Neurexin complex, critical for synapse formation/maintenance, represents a vulnerable axis for neuropsychiatric diseases. We have previously found that GluD1 deletion results in reversal learning deficit and repetitive behavior. In this study, we show that selective ablation of GluD1 from the dorsal striatum impairs behavioral flexibility in a water T-maze task. We further found that striatal GluD1 is preferentially found in dendritic shafts, and more frequently associated with thalamic than cortical glutamatergic terminals suggesting localization to projections from the thalamic parafascicular nucleus (Pf). Conditional deletion of GluD1 from the striatum led to a selective loss of thalamic, but not cortical, terminals, and reduced glutamatergic neurotransmission. Optogenetic studies demonstrated functional changes at thalamostriatal synapses from the Pf, but no effect on the corticostriatal system, upon ablation of GluD1 in the dorsal striatum. These studies suggest a novel molecular mechanism by which genetic variations associated with neuropsychiatric disorders may impair behavioral flexibility, and reveal a unique principle by which GluD1 subunit regulates forebrain circuits.
Collapse
Affiliation(s)
- Jinxu Liu
- Department of Pharmacology and Neuroscience, Creighton University School of Medicine, Omaha, NE 68178, USA
| | - Gajanan P Shelkar
- Department of Pharmacology and Neuroscience, Creighton University School of Medicine, Omaha, NE 68178, USA
| | - Pauravi J Gandhi
- Department of Pharmacology and Neuroscience, Creighton University School of Medicine, Omaha, NE 68178, USA
| | - Dinesh Y Gawande
- Department of Pharmacology and Neuroscience, Creighton University School of Medicine, Omaha, NE 68178, USA
| | - Andrew Hoover
- Yerkes National Primate Research Center, Atlanta, GA 30329, USA; UDALL Center of Excellence for Parkinson's Disease, Atlanta, GA 30329, USA
| | - Rosa M Villalba
- Yerkes National Primate Research Center, Atlanta, GA 30329, USA; UDALL Center of Excellence for Parkinson's Disease, Atlanta, GA 30329, USA
| | - Ratnamala Pavuluri
- Department of Pharmacology and Neuroscience, Creighton University School of Medicine, Omaha, NE 68178, USA
| | - Yoland Smith
- Yerkes National Primate Research Center, Atlanta, GA 30329, USA; UDALL Center of Excellence for Parkinson's Disease, Atlanta, GA 30329, USA; Dept. Neurology, Emory University, Atlanta, GA 30329, USA
| | - Shashank M Dravid
- Department of Pharmacology and Neuroscience, Creighton University School of Medicine, Omaha, NE 68178, USA.
| |
Collapse
|
29
|
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: 2.0] [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
|
30
|
Brindisi M, Saraswati AP, Brogi S, Gemma S, Butini S, Campiani G. Old but Gold: Tracking the New Guise of Histone Deacetylase 6 (HDAC6) Enzyme as a Biomarker and Therapeutic Target in Rare Diseases. J Med Chem 2019; 63:23-39. [PMID: 31415174 DOI: 10.1021/acs.jmedchem.9b00924] [Citation(s) in RCA: 72] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Epigenetic regulation orchestrates many cellular processes and greatly influences key disease mechanisms. Histone deacetylase (HDAC) enzymes play a crucial role either as biomarkers or therapeutic targets owing to their involvement in specific pathophysiological pathways. Beyond their well-characterized role as histone modifiers, HDACs also interact with several nonhistone substrates and their increased expression has been highlighted in specific diseases. The HDAC6 isoform, due to its unique cytoplasmic localization, modulates the acetylation status of tubulin, HSP90, TGF-β, and peroxiredoxins. HDAC6 also exerts noncatalytic activities through its interaction with ubiquitin. Both catalytic and noncatalytic functions of HDACs are being actively studied in the field of specific rare disorders beyond the well-established role in carcinogenesis. This Perspective outlines the application of HDAC(6) inhibitors in rare diseases, such as Rett syndrome, inherited retinal disorders, idiopathic pulmonary fibrosis, and Charcot-Marie-Tooth disease, highlighting their therapeutic potential as innovative and targeted disease-modifying agents.
Collapse
Affiliation(s)
- Margherita Brindisi
- Department of Pharmacy, Department of Excellence 2018-2022 , University of Naples Federico II , Via D. Montesano 49 , I-80131 Naples , Italy
| | - A Prasanth Saraswati
- Department of Biotechnology, Chemistry and Pharmacy, Department of Excellence 2018-2022 , University of Siena , via Aldo Moro 2 , 53100 , Siena , Italy
| | - Simone Brogi
- Department of Pharmacy , University of Pisa , via Bonanno 6 , 56126 , Pisa , Italy
| | - Sandra Gemma
- Department of Biotechnology, Chemistry and Pharmacy, Department of Excellence 2018-2022 , University of Siena , via Aldo Moro 2 , 53100 , Siena , Italy
| | - Stefania Butini
- Department of Biotechnology, Chemistry and Pharmacy, Department of Excellence 2018-2022 , University of Siena , via Aldo Moro 2 , 53100 , Siena , Italy
| | - Giuseppe Campiani
- Department of Biotechnology, Chemistry and Pharmacy, Department of Excellence 2018-2022 , University of Siena , via Aldo Moro 2 , 53100 , Siena , Italy
| |
Collapse
|
31
|
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: 46] [Impact Index Per Article: 9.2] [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
|
32
|
Cortical Seizures in FoxG1+/- Mice are Accompanied by Akt/S6 Overactivation, Excitation/Inhibition Imbalance and Impaired Synaptic Transmission. Int J Mol Sci 2019; 20:ijms20174127. [PMID: 31450553 PMCID: PMC6747530 DOI: 10.3390/ijms20174127] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Accepted: 08/22/2019] [Indexed: 12/14/2022] Open
Abstract
The correct morphofunctional shaping of the cerebral cortex requires a continuous interaction between intrinsic (genes/molecules expressed within the tissue) and extrinsic (e.g., neural activity) factors at all developmental stages. Forkhead Box G1 (FOXG1) is an evolutionarily conserved transcription factor, essential for the cerebral cortex patterning and layering. FOXG1-related disorders, including the congenital form of Rett syndrome, can be caused by deletions, intragenic mutations or duplications. These genetic alterations are associated with a complex phenotypic spectrum, spanning from intellectual disability, microcephaly, to autistic features, and epilepsy. We investigated the functional correlates of dysregulated gene expression by performing electrophysiological assays on FoxG1+/- mice. Local Field Potential (LFP) recordings on freely moving animals detected cortical hyperexcitability. On the other hand, patch-clamp recordings showed a downregulation of spontaneous glutamatergic transmission. These findings were accompanied by overactivation of Akt/S6 signaling. Furthermore, the expression of vesicular glutamate transporter 2 (vGluT2) was increased, whereas the level of the potassium/chloride cotransporter KCC2 was reduced, thus indicating a higher excitation/inhibition ratio. Our findings provide evidence that altered expression of a key gene for cortical development can result in specific alterations in neural circuit function at the macro- and micro-scale, along with dysregulated intracellular signaling and expression of proteins controlling circuit excitability.
Collapse
|
33
|
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: 1.4] [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
|
34
|
Abstract
Brain development is a highly regulated process that involves the precise spatio-temporal activation of cell signaling cues. Transcription factors play an integral role in this process by relaying information from external signaling cues to the genome. The transcription factor Forkhead box G1 (FOXG1) is expressed in the developing nervous system with a critical role in forebrain development. Altered dosage of FOXG1 due to deletions, duplications, or functional gain- or loss-of-function mutations, leads to a complex array of cellular effects with important consequences for human disease including neurodevelopmental disorders. Here, we review studies in multiple species and cell models where FOXG1 dose is altered. We argue against a linear, symmetrical relationship between FOXG1 dosage states, although FOXG1 levels at the right time and place need to be carefully regulated. Neurodevelopmental disease states caused by mutations in FOXG1 may therefore be regulated through different mechanisms.
Collapse
Affiliation(s)
- Nuwan C Hettige
- Department of Human Genetics, McGill University, Montreal, QC, Canada.,Psychiatric Genetics Group, Douglas Mental Health University Institute, Montreal, QC, Canada
| | - Carl Ernst
- Department of Human Genetics, McGill University, Montreal, QC, Canada.,Psychiatric Genetics Group, Douglas Mental Health University Institute, Montreal, QC, Canada.,Department of Psychiatry, McGill University, Montreal, QC, Canada.,Integrated Program in Neuroscience, McGill University, Montreal, QC, Canada
| |
Collapse
|
35
|
Vitrac A, Cloëz-Tayarani I. Induced pluripotent stem cells as a tool to study brain circuits in autism-related disorders. Stem Cell Res Ther 2018; 9:226. [PMID: 30139379 PMCID: PMC6107940 DOI: 10.1186/s13287-018-0966-2] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
The mammalian brain is a very complex organ containing an estimated 200 billion cells in humans. Therefore, studying human brain development has become very challenging given all the data that are available from different approaches, notably genetic studies.Recent pluripotent stem cell methods have given rise to the possibility of modeling neurodevelopmental diseases associated with genetic defects. Fibroblasts from patients have been reprogrammed into pluripotent stem cells to derive appropriate neuronal lineages. They specifically include different subtypes of cortical neurons that are at the core of human-specific cognitive abilities. The use of neurons derived from induced pluripotent stem cells (iPSC) has led to deciphering convergent and pleiotropic neuronal synaptic phenotypes found in neurodevelopmental disorders such as autism spectrum disorders (ASD) and their associated syndromes. In addition to these initial studies, remarkable progress has been made in the field of stem cells, with the major objective of reproducing the in vivo maturation steps of human neurons. Recently, several studies have demonstrated the ability of human progenitors to respond to guidance cues and signals in vivo that can direct neurons to their appropriate sites of differentiation where they become fully mature neurons.We provide a brief overview on research using human iPSC in ASD and associated syndromes and on the current understanding of new theories using the re-implantation of neural precursors in mouse brain.
Collapse
Affiliation(s)
- Aline Vitrac
- Human Genetics and Cognitive Functions, Institut Pasteur, 25 rue du Docteur Roux, 75015 Paris, France
- CNRS UMR 3571, Institut Pasteur, 25 rue du Docteur Roux, Paris, France
- Université Paris Diderot, Sorbonne Paris Cité, Human Genetics and Cognitive Functions, 25 rue du Docteur Roux, Paris, France
| | - Isabelle Cloëz-Tayarani
- Human Genetics and Cognitive Functions, Institut Pasteur, 25 rue du Docteur Roux, 75015 Paris, France
- CNRS UMR 3571, Institut Pasteur, 25 rue du Docteur Roux, Paris, France
- Université Paris Diderot, Sorbonne Paris Cité, Human Genetics and Cognitive Functions, 25 rue du Docteur Roux, Paris, France
| |
Collapse
|
36
|
The chromatin basis of neurodevelopmental disorders: Rethinking dysfunction along the molecular and temporal axes. Prog Neuropsychopharmacol Biol Psychiatry 2018; 84:306-327. [PMID: 29309830 DOI: 10.1016/j.pnpbp.2017.12.013] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2017] [Revised: 12/19/2017] [Accepted: 12/24/2017] [Indexed: 12/13/2022]
Abstract
The complexity of the human brain emerges from a long and finely tuned developmental process orchestrated by the crosstalk between genome and environment. Vis à vis other species, the human brain displays unique functional and morphological features that result from this extensive developmental process that is, unsurprisingly, highly vulnerable to both genetically and environmentally induced alterations. One of the most striking outcomes of the recent surge of sequencing-based studies on neurodevelopmental disorders (NDDs) is the emergence of chromatin regulation as one of the two domains most affected by causative mutations or Copy Number Variations besides synaptic function, whose involvement had been largely predicted for obvious reasons. These observations place chromatin dysfunction at the top of the molecular pathways hierarchy that ushers in a sizeable proportion of NDDs and that manifest themselves through synaptic dysfunction and recurrent systemic clinical manifestation. Here we undertake a conceptual investigation of chromatin dysfunction in NDDs with the aim of systematizing the available evidence in a new framework: first, we tease out the developmental vulnerabilities in human corticogenesis as a structuring entry point into the causation of NDDs; second, we provide a much needed clarification of the multiple meanings and explanatory frameworks revolving around "epigenetics", highlighting those that are most relevant for the analysis of these disorders; finally we go in-depth into paradigmatic examples of NDD-causing chromatin dysregulation, with a special focus on human experimental models and datasets.
Collapse
|
37
|
Linda K, Fiuza C, Nadif Kasri N. The promise of induced pluripotent stem cells for neurodevelopmental disorders. Prog Neuropsychopharmacol Biol Psychiatry 2018; 84:382-391. [PMID: 29128445 DOI: 10.1016/j.pnpbp.2017.11.009] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2017] [Revised: 10/30/2017] [Accepted: 11/07/2017] [Indexed: 12/19/2022]
Abstract
A major challenge in clinical genetics and medicine is represented by genetically and phenotypically highly diverse neurodevelopmental disorders, like for example intellectual disability and autism. Intellectual disability is characterized by substantial limitations in cognitive function and adaptive behaviour. At the cellular level, this is reflected by deficits in synaptic structure and plasticity and therefore has been coined as a synaptic disorder or "synaptopathy". In this review, we summarize the findings from recent studies in which iPSCs have been used to model specific neurodevelopmental syndromes, including Fragile X syndrome, Rett syndrome, Williams-Beuren syndrome and Phelan-McDermid syndrome. We discuss what we have learned from these studies and what key issues need to be addressed to move the field forward.
Collapse
Affiliation(s)
- Katrin Linda
- Department of Human Genetics, Department of Cognitive Neuroscience, Radboudumc, 6500 HB, Nijmegen, The Netherlands; Donders Institute for Brain, Cognition, and Behaviour, Centre for Neuroscience, 6525 AJ, Nijmegen, The Netherlands
| | - Carol Fiuza
- Department of Human Genetics, Department of Cognitive Neuroscience, Radboudumc, 6500 HB, Nijmegen, The Netherlands; Donders Institute for Brain, Cognition, and Behaviour, Centre for Neuroscience, 6525 AJ, Nijmegen, The Netherlands
| | - Nael Nadif Kasri
- Department of Human Genetics, Department of Cognitive Neuroscience, Radboudumc, 6500 HB, Nijmegen, The Netherlands; Donders Institute for Brain, Cognition, and Behaviour, Centre for Neuroscience, 6525 AJ, Nijmegen, The Netherlands.
| |
Collapse
|
38
|
Landucci E, Brindisi M, Bianciardi L, Catania LM, Daga S, Croci S, Frullanti E, Fallerini C, Butini S, Brogi S, Furini S, Melani R, Molinaro A, Lorenzetti FC, Imperatore V, Amabile S, Mariani J, Mari F, Ariani F, Pizzorusso T, Pinto AM, Vaccarino FM, Renieri A, Campiani G, Meloni I. iPSC-derived neurons profiling reveals GABAergic circuit disruption and acetylated α-tubulin defect which improves after iHDAC6 treatment in Rett syndrome. Exp Cell Res 2018; 368:225-235. [PMID: 29730163 DOI: 10.1016/j.yexcr.2018.05.001] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2017] [Revised: 04/30/2018] [Accepted: 05/02/2018] [Indexed: 12/22/2022]
Abstract
Mutations in MECP2 gene have been identified in more than 95% of patients with classic Rett syndrome, one of the most common neurodevelopmental disorders in females. Taking advantage of the breakthrough technology of genetic reprogramming, we investigated transcriptome changes in neurons differentiated from induced Pluripotent Stem Cells (iPSCs) derived from patients with different mutations. Profiling by RNA-seq in terminally differentiated neurons revealed a prominent GABAergic circuit disruption along with a perturbation of cytoskeleton dynamics. In particular, in mutated neurons we identified a significant decrease of acetylated α-tubulin which can be reverted by treatment with selective inhibitors of HDAC6, the main α-tubulin deacetylase. These findings contribute to shed light on Rett pathogenic mechanisms and provide hints for the treatment of Rett-associated epileptic behavior as well as for the definition of new therapeutic strategies for Rett syndrome.
Collapse
Affiliation(s)
- Elisa Landucci
- Medical Genetics, University of Siena, Strada delle Scotte 4, 53100, Siena, Italy
| | - Margherita Brindisi
- NatSynDrugs, Department of Biotechnology, Chemistry and Pharmacy, DoE 2018-2022 University of Siena, via Aldo Moro 2, 53100 Siena, Italy
| | - Laura Bianciardi
- Medical Genetics, University of Siena, Strada delle Scotte 4, 53100, Siena, Italy
| | - Lorenza M Catania
- Medical Genetics, University of Siena, Strada delle Scotte 4, 53100, Siena, Italy
| | - Sergio Daga
- Medical Genetics, University of Siena, Strada delle Scotte 4, 53100, Siena, Italy
| | - Susanna Croci
- Medical Genetics, University of Siena, Strada delle Scotte 4, 53100, Siena, Italy
| | - Elisa Frullanti
- Medical Genetics, University of Siena, Strada delle Scotte 4, 53100, Siena, Italy
| | - Chiara Fallerini
- Medical Genetics, University of Siena, Strada delle Scotte 4, 53100, Siena, Italy
| | - Stefania Butini
- NatSynDrugs, Department of Biotechnology, Chemistry and Pharmacy, DoE 2018-2022 University of Siena, via Aldo Moro 2, 53100 Siena, Italy
| | - Simone Brogi
- NatSynDrugs, Department of Biotechnology, Chemistry and Pharmacy, DoE 2018-2022 University of Siena, via Aldo Moro 2, 53100 Siena, Italy
| | - Simone Furini
- Department of Medical Biotechnologies, University of Siena, Strada delle Scotte 4, 53100 Siena, Italy
| | - Riccardo Melani
- Institute of Neuroscience, National Research Council (CNR), Via Giuseppe Moruzzi, 1, 56124 Pisa, Italy
| | - Angelo Molinaro
- Institute of Neuroscience, National Research Council (CNR), Via Giuseppe Moruzzi, 1, 56124 Pisa, Italy; Department of Neuroscience, Psychology, Drug Research and Child Health NEUROFARBA, University of Florence, Viale Gaetano Pieraccini, 6, 50139 Florence, Italy
| | | | - Valentina Imperatore
- Medical Genetics, University of Siena, Strada delle Scotte 4, 53100, Siena, Italy
| | - Sonia Amabile
- Medical Genetics, University of Siena, Strada delle Scotte 4, 53100, Siena, Italy
| | - Jessica Mariani
- Yale University, Child Study Center, 230 South Frontage Rd, New Haven, CT 06520, United States
| | - Francesca Mari
- Medical Genetics, University of Siena, Strada delle Scotte 4, 53100, Siena, Italy; Genetica Medica, Azienda Ospedaliera Universitaria Senese, Viale Mario Bracci 2, 53100 Siena, Italy
| | - Francesca Ariani
- Medical Genetics, University of Siena, Strada delle Scotte 4, 53100, Siena, Italy; Genetica Medica, Azienda Ospedaliera Universitaria Senese, Viale Mario Bracci 2, 53100 Siena, Italy
| | - Tommaso Pizzorusso
- Institute of Neuroscience, National Research Council (CNR), Via Giuseppe Moruzzi, 1, 56124 Pisa, Italy; Department of Neuroscience, Psychology, Drug Research and Child Health NEUROFARBA, University of Florence, Viale Gaetano Pieraccini, 6, 50139 Florence, Italy; BIO@SNS lab, Scuola Normale Superiore, Piazza dei Cavalieri, 7, 56126 Pisa, Italy
| | - Anna Maria Pinto
- Medical Genetics, University of Siena, Strada delle Scotte 4, 53100, Siena, Italy; Genetica Medica, Azienda Ospedaliera Universitaria Senese, Viale Mario Bracci 2, 53100 Siena, Italy
| | - Flora M Vaccarino
- Yale University, Child Study Center, 230 South Frontage Rd, New Haven, CT 06520, United States
| | - Alessandra Renieri
- Medical Genetics, University of Siena, Strada delle Scotte 4, 53100, Siena, Italy; Genetica Medica, Azienda Ospedaliera Universitaria Senese, Viale Mario Bracci 2, 53100 Siena, Italy.
| | - Giuseppe Campiani
- NatSynDrugs, Department of Biotechnology, Chemistry and Pharmacy, DoE 2018-2022 University of Siena, via Aldo Moro 2, 53100 Siena, Italy.
| | - Ilaria Meloni
- Medical Genetics, University of Siena, Strada delle Scotte 4, 53100, Siena, Italy
| |
Collapse
|
39
|
Fink JJ, Levine ES. Uncovering True Cellular Phenotypes: Using Induced Pluripotent Stem Cell-Derived Neurons to Study Early Insults in Neurodevelopmental Disorders. Front Neurol 2018; 9:237. [PMID: 29713304 PMCID: PMC5911479 DOI: 10.3389/fneur.2018.00237] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2017] [Accepted: 03/26/2018] [Indexed: 01/07/2023] Open
Abstract
Animal models of neurodevelopmental disorders have provided invaluable insights into the molecular-, cellular-, and circuit-level defects associated with a plethora of genetic disruptions. In many cases, these deficits have been linked to changes in disease-relevant behaviors, but very few of these findings have been translated to treatments for human disease. This may be due to significant species differences and the difficulty in modeling disorders that involve deletion or duplication of multiple genes. The identification of primary underlying pathophysiology in these models is confounded by the accumulation of secondary disease phenotypes in the mature nervous system, as well as potential compensatory mechanisms. The discovery of induced pluripotent stem cell technology now provides a tool to accurately model complex genetic neurogenetic disorders. Using this technique, patient-specific cell lines can be generated and differentiated into specific subtypes of neurons that can be used to identify primary cellular and molecular phenotypes. It is clear that impairments in synaptic structure and function are a common pathophysiology across neurodevelopmental disorders, and electrophysiological analysis at the earliest stages of neuronal development is critical for identifying changes in activity and excitability that can contribute to synaptic dysfunction and identify targets for disease-modifying therapies.
Collapse
Affiliation(s)
- James J Fink
- Department of Neuroscience, University of Connecticut School of Medicine, Farmington, CT, United States
| | - Eric S Levine
- Department of Neuroscience, University of Connecticut School of Medicine, Farmington, CT, United States
| |
Collapse
|
40
|
Chiola S, Do MD, Centrone L, Mallamaci A. Foxg1 Overexpression in Neocortical Pyramids Stimulates Dendrite Elongation Via Hes1 and pCreb1 Upregulation. Cereb Cortex 2018; 29:1006-1019. [DOI: 10.1093/cercor/bhy007] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2017] [Accepted: 01/07/2018] [Indexed: 12/21/2022] Open
Affiliation(s)
- Simone Chiola
- Lab of Cerebral Cortex Development, Neuroscience Area, SISSA, via Bonomea Trieste, Italy
| | - Mihn Duc Do
- Lab of Cerebral Cortex Development, Neuroscience Area, SISSA, via Bonomea Trieste, Italy
| | - Lucy Centrone
- Lab of Cerebral Cortex Development, Neuroscience Area, SISSA, via Bonomea Trieste, Italy
| | - Antonello Mallamaci
- Lab of Cerebral Cortex Development, Neuroscience Area, SISSA, via Bonomea Trieste, Italy
| |
Collapse
|
41
|
Murine pluripotent stem cells with a homozygous knockout of Foxg1 show reduced differentiation towards cortical progenitors in vitro. Stem Cell Res 2017; 25:50-60. [PMID: 29080444 DOI: 10.1016/j.scr.2017.10.012] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2017] [Revised: 10/13/2017] [Accepted: 10/17/2017] [Indexed: 01/05/2023] Open
Abstract
Foxg1 is a transcription factor critical for the development of the mammalian telencephalon. Foxg1 controls the proliferation of dorsal telencephalon progenitors and the specification of the ventral telencephalon. Homozygous knockout of Foxg1 in mice leads to severe microcephaly, attributed to premature differentiation of telencephalic progenitors, mainly of cortical progenitors. Here, we analyzed the influence of a Foxg1 knockout on differentiation of murine pluripotent stem cells (mPSCs) in an in vitro model of neuronal development. Murine PSCs were prone to neuronal differentiation in embryoid body like culture with minimal medium conditions, based on the intrinsic default of PSCs to develop into cortical progenitors. Differences between Foxg1 wildtype (Foxg1WT) and knockout (Foxg1KO) mPSCs were analyzed. Several mPSC lines with homozygous mutations in Foxg1 were produced using the CRISPR/Cas9 system leading to loss of functional domains. Analysis of mRNA expression using quantitative Real-Time (q) PCR revealed that Foxg1KO mPSCs expressed significantly less mRNA of Foxg1, Emx1, and VGlut1 compared to Foxg1WT controls, indicating reduced differentiation towards dorsal telencephalic progenitors. However, the size of the derived EB-like structures did not differ between Foxg1WT and Foxg1KO mPSCs. These results show that loss of dorsal telencephalic progenitors can be detected using a simple and rapid differentiation protocol. This study is a first hint that this differentiation method can be used to analyze even extreme phenotypes that are lethal in vivo.
Collapse
|
42
|
Ardhanareeswaran K, Mariani J, Coppola G, Abyzov A, Vaccarino FM. Human induced pluripotent stem cells for modelling neurodevelopmental disorders. Nat Rev Neurol 2017; 13:265-278. [PMID: 28418023 DOI: 10.1038/nrneurol.2017.45] [Citation(s) in RCA: 106] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
We currently have a poor understanding of the pathogenesis of neurodevelopmental disorders, owing to the fact that postmortem and imaging studies can only measure the postnatal status quo and offer little insight into the processes that give rise to the observed outcomes. Human induced pluripotent stem cells (hiPSCs) should, in principle, prove powerful for elucidating the pathways that give rise to neurodevelopmental disorders. hiPSCs are embryonic-stem-cell-like cells that can be derived from somatic cells. They retain the unique genetic signature of the individual from whom they were derived, and thus enable researchers to recapitulate that individual's idiosyncratic neural development in a dish. In the case of individuals with disease, we can re-enact the disease-altered trajectory of brain development and examine how and why phenotypic and molecular abnormalities arise in these diseased brains. Here, we review hiPSC biology and possible experimental designs when using hiPSCs to model disease. We then discuss existing hiPSC models of neurodevelopmental disorders. Our hope is that, as some studies have already shown, hiPSCs will illuminate the pathophysiology of developmental disorders of the CNS and lead to therapeutic options for the millions that are affected by these conditions.
Collapse
Affiliation(s)
- Karthikeyan Ardhanareeswaran
- Child Study Center, Yale University School of Medicine, 230 South Frontage Road, New Haven, Connecticut 06520, USA
| | - Jessica Mariani
- Child Study Center, Yale University School of Medicine, 230 South Frontage Road, New Haven, Connecticut 06520, USA
| | - Gianfilippo Coppola
- Child Study Center, Yale University School of Medicine, 230 South Frontage Road, New Haven, Connecticut 06520, USA
| | - Alexej Abyzov
- Department of Health Sciences Research, Center for Individualized Medicine, 200 First Street SW, Rochester, Minnesota 55905, USA
| | - Flora M Vaccarino
- Child Study Center, Yale University School of Medicine, 230 South Frontage Road, New Haven, Connecticut 06520, USA.,Department of Neuroscience, Yale Kavli Institute for Neuroscience, Yale University School of Medicine, 200 South Frontage Road, New Haven, Connecticut 06510, USA
| |
Collapse
|
43
|
Gomathi M, Balachandar V. Novel therapeutic approaches: Rett syndrome and human induced pluripotent stem cell technology. Stem Cell Investig 2017; 4:20. [PMID: 28447035 DOI: 10.21037/sci.2017.02.11] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2017] [Accepted: 02/21/2017] [Indexed: 01/28/2023]
Abstract
Recent advances in induced pluripotent stem cell (iPSC) technology target screening and discovering of therapeutic agents for the possible cure of human diseases. Human induced pluripotent stem cells (hiPSC) are the right kind of platform for testing potency of specific active compounds. Ayurveda, the Indian traditional system of medicine developed between 2,500 and 500 BC, is a science involving the intelligent formulations of herbs and minerals. It can serve as a "goldmine" for novel neuroprotective agents used for centuries to treat neurological disorders. This review discusses limitations in screening drugs for neurological disorders and the advantages offered by hiPSC integrated with Indian traditional system of medicine. We begin by describing the current state of hiPSC technology in research on Rett syndrome (RTT) followed by the current controversies in RTT research combined with the emergence of patient-specific hiPSC that indicate an urgent need for researchers to understand the etiology and drug mechanism. We conclude by offering recommendations to reinforce the screening of active compounds present in the ayurvedic medicines using the human induced pluripotent neural model system for research involving drug discovery for RTT. This integrative approach will fill the current knowledge gap in the traditional medicines and drug discovery.
Collapse
Affiliation(s)
- Mohan Gomathi
- Human Molecular Genetics and Stem Cell Laboratory, Department of Human Genetics and Molecular Biology, Bharathiar University, Coimbatore-641 046, Tamil Nadu, India
| | - Vellingiri Balachandar
- Human Molecular Genetics and Stem Cell Laboratory, Department of Human Genetics and Molecular Biology, Bharathiar University, Coimbatore-641 046, Tamil Nadu, India
| |
Collapse
|
44
|
Pizzo R, Gurgone A, Castroflorio E, Amendola E, Gross C, Sassoè-Pognetto M, Giustetto M. Lack of Cdkl5 Disrupts the Organization of Excitatory and Inhibitory Synapses and Parvalbumin Interneurons in the Primary Visual Cortex. Front Cell Neurosci 2016; 10:261. [PMID: 27965538 PMCID: PMC5124713 DOI: 10.3389/fncel.2016.00261] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2016] [Accepted: 10/26/2016] [Indexed: 01/31/2023] Open
Abstract
Cyclin-dependent kinase-like 5 (CDKL5) mutations are found in severe neurodevelopmental disorders, including the Hanefeld variant of Rett syndrome (RTT; CDKL5 disorder). CDKL5 loss-of-function murine models recapitulate pathological signs of the human disease, such as visual attention deficits and reduced visual acuity. Here we investigated the cellular and synaptic substrates of visual defects by studying the organization of the primary visual cortex (V1) of Cdkl5−/y mice. We found a severe reduction of c-Fos expression in V1 of Cdkl5−/y mutants, suggesting circuit hypoactivity. Glutamatergic presynaptic structures were increased, but postsynaptic PSD-95 and Homer were significantly downregulated in CDKL5 mutants. Interneurons expressing parvalbumin, but not other types of interneuron, had a higher density in mutant V1, and were hyperconnected with pyramidal neurons. Finally, the developmental trajectory of pavalbumin-containing cells was also affected in Cdkl5−/y mice, as revealed by fainter appearance perineuronal nets at the closure of the critical period (CP). The present data reveal an overall disruption of V1 cellular and synaptic organization that may cause a shift in the excitation/inhibition balance likely to underlie the visual deficits characteristic of CDKL5 disorder. Moreover, ablation of CDKL5 is likely to tamper with the mechanisms underlying experience-dependent refinement of cortical circuits during the CP of development.
Collapse
Affiliation(s)
- Riccardo Pizzo
- Department of Neuroscience, University of Turin Turin, Italy
| | - Antonia Gurgone
- Department of Neuroscience, University of Turin Turin, Italy
| | | | - Elena Amendola
- Molecular Medicine and Medical Biotechnologies, University of Naples "Federico II" Napoli, Italy
| | - Cornelius Gross
- Mouse Biology Unit, European Molecular Biology Laboratory (EMBL) Monterotondo, Italy
| | - Marco Sassoè-Pognetto
- Department of Neuroscience, University of TurinTurin, Italy; National Institute of Neuroscience-ItalyTurin, Italy
| | - Maurizio Giustetto
- Department of Neuroscience, University of TurinTurin, Italy; National Institute of Neuroscience-ItalyTurin, Italy
| |
Collapse
|
45
|
Baldo C, Casareto L, Renieri A, Merla G, Garavaglia B, Goldwurm S, Pegoraro E, Moggio M, Mora M, Politano L, Sangiorgi L, Mazzotti R, Viotti V, Meloni I, Pellico MT, Barzaghi C, Wang CM, Monaco L, Filocamo M. The alliance between genetic biobanks and patient organisations: the experience of the telethon network of genetic biobanks. Orphanet J Rare Dis 2016; 11:142. [PMID: 27776540 PMCID: PMC5078978 DOI: 10.1186/s13023-016-0527-7] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2016] [Accepted: 10/18/2016] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Rare diseases (RDs) are often neglected because they affect a small percentage of the population (6-8 %), which makes research and development of new therapies challenging processes. Easy access to high-quality samples and associated clinical data is therefore a key prerequisite for biomedical research. In this context, Genetic Biobanks are critical to developing basic, translational and clinical research on RDs. The Telethon Network of Genetic Biobanks (TNGB) is aware of the importance of biobanking as a service for patients and has started a dialogue with RD-Patient Organisations via promotion of dedicated meetings and round-tables, as well as by including their representatives on the TNGB Advisory Board. This has enabled the active involvement of POs in drafting biobank policies and procedures, including those concerning ethical issues. Here, we report on our experience with RD-Patient Organisations who have requested the services of existing biobanks belonging to TNGB and describe how these relationships were established, formalised and maintained. RESULTS The process of patient engagement has proven to be successful both for lay members, who increased their understanding of the complex processes of biobanking, and for professionals, who gained awareness of the needs and expectations of the people involved. This collaboration has resulted in a real interest on the part of Patient Organisations in the biobanking service, which has led to 13 written agreements designed to formalise this process. These agreements enabled the centralisation of rare genetic disease biospecimens and their related data, thus making them available to the scientific community. CONCLUSIONS The TNGB experience has proven to be an example of good practice with regard to patient engagement in biobanking and may serve as a model of collaboration between disease-oriented Biobanks and Patient Organisations. Such collaboration serves to enhance awareness and trust and to encourage the scientific community to address research on RDs.
Collapse
Affiliation(s)
- Chiara Baldo
- S.C. Laboratorio di Genetica Umana, E.O. Ospedali Galliera, Genoa, Italy
| | - Lorena Casareto
- Ufficio Coordinamento Network, c/o U.O.S.D. Centro di Diagnostica Genetica e Biochimica delle Malattie Metaboliche, Istituto G. Gaslini, Genoa, Italy
| | - Alessandra Renieri
- Medical Genetics, University of Siena and Genetica Medica, Azienda Ospedaliera Universitaria Senese, Siena, Italy
| | - Giuseppe Merla
- Medical Genetics Unit, IRCCS Casa Sollievo della Sofferenza, S. Giovanni Rotondo, FG Italy
| | - Barbara Garavaglia
- U.O.C. Neurogenetica Molecolare, Fondazione I.R.C.C.S. Istituto Neurologico Carlo Besta, Milan, Italy
| | - Stefano Goldwurm
- Parkinson Institute, ASST Centro Specialistico Ortopedico Traumatologico G. Pini – CTO, Milan, Italy
| | - Elena Pegoraro
- Università di Padova, Azienda Ospedaliera Universitaria, Padova, Italy
| | - Maurizio Moggio
- Neuromuscular and Rare Diseases Unit, Dino Ferrari Centre, IRCCS Foundation Ca’ Granda Ospedale Maggiore Policlinico, University of Milan, Milan, Italy
| | - Marina Mora
- Laboratorio di Biologia Cellulare, UO Malattie Neuromuscolari e Neuroimmunologia, Fond. IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Luisa Politano
- Cardiomiologia e Genetica Medica, Dipartimento di Medicina Sperimentale, Seconda Università di Napoli e Azienda Ospedaliera Universitaria della SUN, Naples, Italy
| | - Luca Sangiorgi
- S.S.D. Genetica Medica e Malattie Rare Ortopediche Istituto Ortopedico Rizzoli, Bologna, Italy
| | - Raffaella Mazzotti
- U.O.S.D. Centro di Diagnostica Genetica e Biochimica delle Malattie Metaboliche, Istituto G. Gaslini, Via G. Gaslini 5, 16147 Genoa, Italy
| | - Valeria Viotti
- S.C. Laboratorio di Genetica Umana, E.O. Ospedali Galliera, Genoa, Italy
| | - Ilaria Meloni
- Medical Genetics, University of Siena and Genetica Medica, Azienda Ospedaliera Universitaria Senese, Siena, Italy
| | - Maria Teresa Pellico
- Medical Genetics Unit, IRCCS Casa Sollievo della Sofferenza, S. Giovanni Rotondo, FG Italy
| | - Chiara Barzaghi
- U.O.C. Neurogenetica Molecolare, Fondazione I.R.C.C.S. Istituto Neurologico Carlo Besta, Milan, Italy
| | | | | | - Mirella Filocamo
- U.O.S.D. Centro di Diagnostica Genetica e Biochimica delle Malattie Metaboliche, Istituto G. Gaslini, Via G. Gaslini 5, 16147 Genoa, Italy
| |
Collapse
|
46
|
Advancing drug discovery for neuropsychiatric disorders using patient-specific stem cell models. Mol Cell Neurosci 2016; 73:104-15. [PMID: 26826498 DOI: 10.1016/j.mcn.2016.01.011] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2015] [Revised: 01/22/2016] [Accepted: 01/25/2016] [Indexed: 12/17/2022] Open
Abstract
Compelling clinical, social, and economic reasons exist to innovate in the process of drug discovery for neuropsychiatric disorders. The use of patient-specific, induced pluripotent stem cells (iPSCs) now affords the ability to generate neuronal cell-based models that recapitulate key aspects of human disease. In the context of neuropsychiatric disorders, where access to physiologically active and relevant cell types of the central nervous system for research is extremely limiting, iPSC-derived in vitro culture of human neurons and glial cells is transformative. Potential applications relevant to early stage drug discovery, include support of quantitative biochemistry, functional genomics, proteomics, and perhaps most notably, high-throughput and high-content chemical screening. While many phenotypes in human iPSC-derived culture systems may prove adaptable to screening formats, addressing the question of which in vitro phenotypes are ultimately relevant to disease pathophysiology and therefore more likely to yield effective pharmacological agents that are disease-modifying treatments requires careful consideration. Here, we review recent examples of studies of neuropsychiatric disorders using human stem cell models where cellular phenotypes linked to disease and functional assays have been reported. We also highlight technical advances using genome-editing technologies in iPSCs to support drug discovery efforts, including the interpretation of the functional significance of rare genetic variants of unknown significance and for the purpose of creating cell type- and pathway-selective functional reporter assays. Additionally, we evaluate the potential of in vitro stem cell models to investigate early events of disease pathogenesis, in an effort to understand the underlying molecular mechanism, including the basis of selective cell-type vulnerability, and the potential to create new cell-based diagnostics to aid in the classification of patients and subsequent selection for clinical trials. A number of key challenges remain, including the scaling of iPSC models to larger cohorts and integration with rich clinicopathological information and translation of phenotypes. Still, the overall use of iPSC-based human cell models with functional cellular and biochemical assays holds promise for supporting the discovery of next-generation neuropharmacological agents for the treatment and ultimately prevention of a range of severe mental illnesses.
Collapse
|