1
|
Lee AS, Arefin TM, Gubanova A, Stephen DN, Liu Y, Lao Z, Krishnamurthy A, De Marco García NV, Heck DH, Zhang J, Rajadhyaksha AM, Joyner AL. Cerebellar output neurons can impair non-motor behaviors by altering development of extracerebellar connectivity. Nat Commun 2025; 16:1858. [PMID: 39984491 PMCID: PMC11845701 DOI: 10.1038/s41467-025-57080-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Accepted: 02/10/2025] [Indexed: 02/23/2025] Open
Abstract
The capacity of the brain to compensate for insults during development depends on the type of cell loss, whereas the consequences of genetic mutations in the same neurons are difficult to predict. We reveal powerful compensation from outside the mouse cerebellum when the excitatory cerebellar output neurons are ablated embryonically and demonstrate that the main requirement for these neurons is for motor coordination and not basic learning and social behaviors. In contrast, loss of the homeobox transcription factors Engrailed1/2 (EN1/2) in the cerebellar excitatory lineage leads to additional deficits in adult learning and spatial working memory, despite half of the excitatory output neurons being intact. Diffusion MRI indicates increased thalamo-cortico-striatal connectivity in En1/2 mutants, showing that the remaining excitatory neurons lacking En1/2 exert adverse effects on extracerebellar circuits regulating motor learning and select non-motor behaviors. Thus, an absence of cerebellar output neurons is less disruptive than having cerebellar genetic mutations.
Collapse
Affiliation(s)
- Andrew S Lee
- Developmental Biology Program, Sloan Kettering Institute, New York, NY, USA
- Neuroscience Program, Weill Cornell Graduate School of Medical Sciences, New York, NY, USA
| | - Tanzil M Arefin
- Bernard and Irene Schwartz Center for Biomedical Imaging, Department of Radiology, New York University Grossman School of Medicine, New York, NY, USA
- Department of Neuroscience, University of Rochester Medical Center, Rochester, NY, USA
| | - Alina Gubanova
- Developmental Biology Program, Sloan Kettering Institute, New York, NY, USA
| | - Daniel N Stephen
- Developmental Biology Program, Sloan Kettering Institute, New York, NY, USA
| | - Yu Liu
- Department of Biomedical Sciences, University of Minnesota Medical School, Duluth, MN, USA
- Center for Cerebellar Network Structure and Function in Health and Disease, University of Minnesota, Duluth, MN, USA
| | - Zhimin Lao
- Developmental Biology Program, Sloan Kettering Institute, New York, NY, USA
| | - Anjana Krishnamurthy
- Developmental Biology Program, Sloan Kettering Institute, New York, NY, USA
- Neuroscience Program, Weill Cornell Graduate School of Medical Sciences, New York, NY, USA
| | - Natalia V De Marco García
- Neuroscience Program, Weill Cornell Graduate School of Medical Sciences, New York, NY, USA
- Center for Neurogenetics, Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - Detlef H Heck
- Department of Biomedical Sciences, University of Minnesota Medical School, Duluth, MN, USA
- Center for Cerebellar Network Structure and Function in Health and Disease, University of Minnesota, Duluth, MN, USA
| | - Jiangyang Zhang
- Bernard and Irene Schwartz Center for Biomedical Imaging, Department of Radiology, New York University Grossman School of Medicine, New York, NY, USA
| | - Anjali M Rajadhyaksha
- Neuroscience Program, Weill Cornell Graduate School of Medical Sciences, New York, NY, USA
- Pediatric Neurology, Department of Pediatrics, Weill Cornell Medicine, New York, NY, USA
- Weill Cornell Autism Research Program, Weill Cornell Medicine, New York, NY, USA
- Center for Substance Abuse Research, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA
- Department of Neural Sciences, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA
| | - Alexandra L Joyner
- Developmental Biology Program, Sloan Kettering Institute, New York, NY, USA.
- Neuroscience Program, Weill Cornell Graduate School of Medical Sciences, New York, NY, USA.
- Biochemistry, Cell and Molecular Biology Program, Weill Cornell Graduate School of Medical Sciences, New York, NY, USA.
| |
Collapse
|
2
|
Krishnamurthy A, Lee AS, Bayin NS, Stephen DN, Nasef O, Lao Z, Joyner AL. Engrailed transcription factors direct excitatory cerebellar neuron diversity and survival. Development 2024; 151:dev202502. [PMID: 38912572 PMCID: PMC11369685 DOI: 10.1242/dev.202502] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Accepted: 06/17/2024] [Indexed: 06/25/2024]
Abstract
The neurons of the three cerebellar nuclei (CN) are the primary output neurons of the cerebellum. The excitatory neurons (e) of the medial (m) CN (eCNm) were recently divided into molecularly defined subdomains in the adult; however, how they are established during development is not known. We define molecular subdomains of the mouse embryonic eCNm using single-cell RNA-sequencing and spatial expression analysis, showing that they evolve during embryogenesis to prefigure the adult. Furthermore, eCNm are transcriptionally divergent from cells in the other nuclei by embryonic day 14.5. We previously showed that loss of the homeobox genes En1 and En2 leads to loss of approximately half of the embryonic eCNm. We demonstrate that mutation of En1/2 in the embryonic eCNm results in death of specific posterior eCNm molecular subdomains and downregulation of TBR2 (EOMES) in an anterior embryonic subdomain, as well as reduced synaptic gene expression. We further reveal a similar function for EN1/2 in mediating TBR2 expression, neuron differentiation and survival in the other excitatory neurons (granule and unipolar brush cells). Thus, our work defines embryonic eCNm molecular diversity and reveals conserved roles for EN1/2 in the cerebellar excitatory neuron lineage.
Collapse
Affiliation(s)
- Anjana Krishnamurthy
- Developmental Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10021, USA
- Neuroscience Program, Weill Cornell Graduate School of Medical Sciences, New York, NY 10065, USA
| | - Andrew S. Lee
- Developmental Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10021, USA
- Neuroscience Program, Weill Cornell Graduate School of Medical Sciences, New York, NY 10065, USA
| | - N. Sumru Bayin
- Developmental Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10021, USA
| | - Daniel N. Stephen
- Developmental Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10021, USA
| | - Olivia Nasef
- Developmental Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10021, USA
| | - Zhimin Lao
- Developmental Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10021, USA
| | - Alexandra L. Joyner
- Developmental Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10021, USA
- Neuroscience Program, Weill Cornell Graduate School of Medical Sciences, New York, NY 10065, USA
- Biochemistry, Cell and Molecular Biology Program, Weill Cornell Graduate School of Medical Sciences, New York, NY 10065, USA
| |
Collapse
|
3
|
Lee AS, Arefin TM, Gubanova A, Stephen DN, Liu Y, Lao Z, Krishnamurthy A, De Marco García NV, Heck DH, Zhang J, Rajadhyaksha AM, Joyner AL. Cerebellar output neurons impair non-motor behaviors by altering development of extracerebellar connectivity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.08.602496. [PMID: 39026865 PMCID: PMC11257463 DOI: 10.1101/2024.07.08.602496] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/20/2024]
Abstract
The capacity of the brain to compensate for insults during development depends on the type of cell loss, whereas the consequences of genetic mutations in the same neurons are difficult to predict. We reveal powerful compensation from outside the cerebellum when the excitatory cerebellar output neurons are ablated embryonically and demonstrate that the minimum requirement for these neurons is for motor coordination and not learning and social behaviors. In contrast, loss of the homeobox transcription factors Engrailed1/2 (EN1/2) in the cerebellar excitatory lineage leads to additional deficits in adult learning and spatial working memory, despite half of the excitatory output neurons being intact. Diffusion MRI indicates increased thalamo-cortico-striatal connectivity in En1/2 mutants, showing that the remaining excitatory neurons lacking En1/2 exert adverse effects on extracerebellar circuits regulating motor learning and select non-motor behaviors. Thus, an absence of cerebellar output neurons is less disruptive than having cerebellar genetic mutations.
Collapse
Affiliation(s)
- Andrew S. Lee
- Developmental Biology Program, Sloan Kettering Institute, New York 10065, NY, USA
- Neuroscience Program, Weill Cornell Graduate School of Medical Sciences, New York 10021, NY, USA
| | - Tanzil M. Arefin
- Bernard and Irene Schwartz Center for Biomedical Imaging, Department of Radiology, New York University Grossman School of Medicine, New York 10016, NY, USA
- Present Address: Center for Neurotechnology in Mental Health Research, Department of Biomedical Engineering, The Pennsylvania State University, University Park, PA 16801, USA
| | - Alina Gubanova
- Developmental Biology Program, Sloan Kettering Institute, New York 10065, NY, USA
| | - Daniel N. Stephen
- Developmental Biology Program, Sloan Kettering Institute, New York 10065, NY, USA
| | - Yu Liu
- Department of Biomedical Sciences, University of Minnesota Medical School, Duluth, MN 55812, USA
- Center for Cerebellar Network Structure and Function in Health and Disease, University of Minnesota, Duluth, MN 55812, USA
| | - Zhimin Lao
- Developmental Biology Program, Sloan Kettering Institute, New York 10065, NY, USA
| | - Anjana Krishnamurthy
- Developmental Biology Program, Sloan Kettering Institute, New York 10065, NY, USA
- Neuroscience Program, Weill Cornell Graduate School of Medical Sciences, New York 10021, NY, USA
| | - Natalia V. De Marco García
- Neuroscience Program, Weill Cornell Graduate School of Medical Sciences, New York 10021, NY, USA
- Center for Neurogenetics, Brain and Mind Research Institute, Weill Cornell Medicine, New York 10021, NY 10021, USA
| | - Detlef H. Heck
- Department of Biomedical Sciences, University of Minnesota Medical School, Duluth, MN 55812, USA
- Center for Cerebellar Network Structure and Function in Health and Disease, University of Minnesota, Duluth, MN 55812, USA
| | - Jiangyang Zhang
- Bernard and Irene Schwartz Center for Biomedical Imaging, Department of Radiology, New York University Grossman School of Medicine, New York 10016, NY, USA
| | - Anjali M. Rajadhyaksha
- Neuroscience Program, Weill Cornell Graduate School of Medical Sciences, New York 10021, NY, USA
- Pediatric Neurology, Department of Pediatrics, Weill Cornell Medicine, New York 10021, NY, USA
- Weill Cornell Autism Research Program, Weill Cornell Medicine, New York 10021, NY, USA
- Present address: Center for Substance Abuse Research, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA and Department of Neural Sciences, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA
| | - Alexandra L. Joyner
- Developmental Biology Program, Sloan Kettering Institute, New York 10065, NY, USA
- Neuroscience Program, Weill Cornell Graduate School of Medical Sciences, New York 10021, NY, USA
- Biochemistry, Cell and Molecular Biology Program, Weill Cornell Graduate School of Medical Sciences, New York 10021, NY, USA
| |
Collapse
|
4
|
Mattioni L, Barbieri A, Grigoli A, Balasco L, Bozzi Y, Provenzano G. Alterations of Perineuronal Net Expression and Abnormal Social Behavior and Whisker-dependent Texture Discrimination in Mice Lacking the Autism Candidate Gene Engrailed 2. Neuroscience 2024; 546:63-74. [PMID: 38537894 DOI: 10.1016/j.neuroscience.2024.03.023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Revised: 02/27/2024] [Accepted: 03/19/2024] [Indexed: 04/06/2024]
Abstract
GABAergic interneurons and perineuronal nets (PNNs) are important regulators of plasticity throughout life and their dysfunction has been implicated in the pathogenesis of several neuropsychiatric conditions, including autism spectrum disorders (ASD). PNNs are condensed portions of the extracellular matrix (ECM) that are crucial for neural development and proper formation of synaptic connections. We previously showed a reduced expression of GABAergic interneuron markers in the hippocampus and somatosensory cortex of adult mice lacking the Engrailed2 gene (En2-/- mice), a mouse model of ASD. Since alterations in PNNs have been proposed as a possible pathogenic mechanism in ASD, we hypothesized that the PNN dysfunction may contribute to the neural and behavioral abnormalities of En2-/- mice. Here, we show an increase in the PNN fluorescence intensity, evaluated by Wisteria floribunda agglutinin, in brain regions involved in social behavior and somatosensory processing. In addition, we found that En2-/- mice exhibit altered texture discrimination through whiskers and display a marked decrease in the preference for social novelty. Our results raise the possibility that altered expression of PNNs, together with defects of GABAergic interneurons, might contribute to the pathogenesis of social and sensory behavioral abnormalities.
Collapse
Affiliation(s)
- Lorenzo Mattioni
- Department of Cellular, Computational, and Integrative Biology (CIBIO), University of Trento, via Sommarive 9, 38123 Trento, Italy.
| | - Anna Barbieri
- Department of Cellular, Computational, and Integrative Biology (CIBIO), University of Trento, via Sommarive 9, 38123 Trento, Italy
| | - Andrea Grigoli
- Department of Cellular, Computational, and Integrative Biology (CIBIO), University of Trento, via Sommarive 9, 38123 Trento, Italy
| | - Luigi Balasco
- CIMeC - Center for Mind/Brain Sciences, University of Trento, Piazza della Manifattura 1, 38068 Rovereto, Trento, Italy
| | - Yuri Bozzi
- CIMeC - Center for Mind/Brain Sciences, University of Trento, Piazza della Manifattura 1, 38068 Rovereto, Trento, Italy; CNR Neuroscience Institute, via Moruzzi 1, 56124 Pisa, Italy
| | - Giovanni Provenzano
- Department of Cellular, Computational, and Integrative Biology (CIBIO), University of Trento, via Sommarive 9, 38123 Trento, Italy.
| |
Collapse
|
5
|
Senovilla-Ganzo R, García-Moreno F. The Phylotypic Brain of Vertebrates, from Neural Tube Closure to Brain Diversification. BRAIN, BEHAVIOR AND EVOLUTION 2024; 99:45-68. [PMID: 38342091 DOI: 10.1159/000537748] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Accepted: 02/04/2024] [Indexed: 02/13/2024]
Abstract
BACKGROUND The phylotypic or intermediate stages are thought to be the most evolutionary conserved stages throughout embryonic development. The contrast with divergent early and later stages derived from the concept of the evo-devo hourglass model. Nonetheless, this developmental constraint has been studied as a whole embryo process, not at organ level. In this review, we explore brain development to assess the existence of an equivalent brain developmental hourglass. In the specific case of vertebrates, we propose to split the brain developmental stages into: (1) Early: Neurulation, when the neural tube arises after gastrulation. (2) Intermediate: Brain patterning and segmentation, when the neuromere identities are established. (3) Late: Neurogenesis and maturation, the stages when the neurons acquire their functionality. Moreover, we extend this analysis to other chordates brain development to unravel the evolutionary origin of this evo-devo constraint. SUMMARY Based on the existing literature, we hypothesise that a major conservation of the phylotypic brain might be due to the pleiotropy of the inductive regulatory networks, which are predominantly expressed at this stage. In turn, earlier stages such as neurulation are rather mechanical processes, whose regulatory networks seem to adapt to environment or maternal geometries. The later stages are also controlled by inductive regulatory networks, but their effector genes are mostly tissue-specific and functional, allowing diverse developmental programs to generate current brain diversity. Nonetheless, all stages of the hourglass are highly interconnected: divergent neurulation must have a vertebrate shared end product to reproduce the vertebrate phylotypic brain, and the boundaries and transcription factor code established during the highly conserved patterning will set the bauplan for the specialised and diversified adult brain. KEY MESSAGES The vertebrate brain is conserved at phylotypic stages, but the highly conserved mechanisms that occur during these brain mid-development stages (Inducing Regulatory Networks) are also present during other stages. Oppositely, other processes as cell interactions and functional neuronal genes are more diverse and majoritarian in early and late stages of development, respectively. These phenomena create an hourglass of transcriptomic diversity during embryonic development and evolution, with a really conserved bottleneck that set the bauplan for the adult brain around the phylotypic stage.
Collapse
Affiliation(s)
- Rodrigo Senovilla-Ganzo
- Achucarro Basque Center for Neuroscience, Scientific Park of the University of the Basque Country (UPV/EHU), Leioa, Spain
- Department of Neuroscience, Faculty of Medicine and Odontology, UPV/EHU, Leioa, Spain
| | - Fernando García-Moreno
- Achucarro Basque Center for Neuroscience, Scientific Park of the University of the Basque Country (UPV/EHU), Leioa, Spain
- Department of Neuroscience, Faculty of Medicine and Odontology, UPV/EHU, Leioa, Spain
- IKERBASQUE Foundation, Bilbao, Spain
| |
Collapse
|
6
|
Krishnamurthy A, Lee AS, Bayin NS, Stephen DN, Nasef O, Lao Z, Joyner AL. Engrailed transcription factors direct excitatory cerebellar neuron diversity and survival. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.11.30.569445. [PMID: 38077070 PMCID: PMC10705369 DOI: 10.1101/2023.11.30.569445] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/09/2024]
Abstract
The excitatory neurons of the three cerebellar nuclei (eCN) form the primary output for the cerebellar circuit. The medial eCN (eCNm) were recently divided into molecularly defined subdomains in the adult, however how they are established during development is not known. We define molecular subdomains of the eCNm using scRNA-seq and spatial expression analysis and show they evolve during embryogenesis to resemble the adult. Furthermore, the eCNm is transcriptionally divergent from the rest of the eCN by E14.5. We previously showed that loss of the homeobox genes En1 and En2 leads to death of a subset of embryonic eCNm. We demonstrate that mutation of En1/2 in embryonic eCNm results in cell death of specific posterior eCNm molecular subdomains and loss of TBR2 (EOMES) expression in an anterior subdomain, as well as reduced synaptic gene expression. We further reveal a similar function for EN1/2 in mediating TBR2 expression, neuron differentiation and survival in the two other cerebellar excitatory neuron types. Thus, our work defines embryonic eCNm molecular diversity and reveals conserved roles for EN1/2 in the cerebellar excitatory neuron lineage.
Collapse
|
7
|
Cardon S, Hervis YP, Bolbach G, Lopin-Bon C, Jacquinet JC, Illien F, Walrant A, Ravault D, He B, Molina L, Burlina F, Lequin O, Joliot A, Carlier L, Sagan S. A cationic motif upstream Engrailed2 homeodomain controls cell internalization through selective interaction with heparan sulfates. Nat Commun 2023; 14:1998. [PMID: 37032404 PMCID: PMC10083169 DOI: 10.1038/s41467-023-37757-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Accepted: 03/29/2023] [Indexed: 04/11/2023] Open
Abstract
Engrailed2 (En2) is a transcription factor that transfers from cell to cell through unconventional pathways. The poorly understood internalization mechanism of this cationic protein is proposed to require an initial interaction with cell-surface glycosaminoglycans (GAGs). To decipher the role of GAGs in En2 internalization, we have quantified the entry of its homeodomain region in model cells that differ in their content in cell-surface GAGs. The binding specificity to GAGs and the influence of this interaction on the structure and dynamics of En2 was also investigated at the amino acid level. Our results show that a high-affinity GAG-binding sequence (RKPKKKNPNKEDKRPR), upstream of the homeodomain, controls En2 internalization through selective interactions with highly-sulfated heparan sulfate GAGs. Our data underline the functional importance of the intrinsically disordered basic region upstream of En2 internalization domain, and demonstrate the critical role of GAGs as an entry gate, finely tuning homeoprotein capacity to internalize into cells.
Collapse
Affiliation(s)
- Sébastien Cardon
- Sorbonne Université, École Normale Supérieure, PSL University, CNRS, Laboratoire des Biomolécules (LBM), 75005, Paris, France
| | - Yadira P Hervis
- Sorbonne Université, École Normale Supérieure, PSL University, CNRS, Laboratoire des Biomolécules (LBM), 75005, Paris, France
| | - Gérard Bolbach
- Sorbonne Université, École Normale Supérieure, PSL University, CNRS, Laboratoire des Biomolécules (LBM), 75005, Paris, France
- Sorbonne Université, Mass Spectrometry Sciences Sorbonne University, MS3U platform, 75005, Paris, France
| | | | | | - Françoise Illien
- Sorbonne Université, École Normale Supérieure, PSL University, CNRS, Laboratoire des Biomolécules (LBM), 75005, Paris, France
| | - Astrid Walrant
- Sorbonne Université, École Normale Supérieure, PSL University, CNRS, Laboratoire des Biomolécules (LBM), 75005, Paris, France
| | - Delphine Ravault
- Sorbonne Université, École Normale Supérieure, PSL University, CNRS, Laboratoire des Biomolécules (LBM), 75005, Paris, France
| | - Bingwei He
- Sorbonne Université, École Normale Supérieure, PSL University, CNRS, Laboratoire des Biomolécules (LBM), 75005, Paris, France
| | - Laura Molina
- Sorbonne Université, École Normale Supérieure, PSL University, CNRS, Laboratoire des Biomolécules (LBM), 75005, Paris, France
| | - Fabienne Burlina
- Sorbonne Université, École Normale Supérieure, PSL University, CNRS, Laboratoire des Biomolécules (LBM), 75005, Paris, France
| | - Olivier Lequin
- Sorbonne Université, École Normale Supérieure, PSL University, CNRS, Laboratoire des Biomolécules (LBM), 75005, Paris, France
| | - Alain Joliot
- INSERM U932, Institut Curie Centre de Recherche, PSL Research University, Paris, France
| | - Ludovic Carlier
- Sorbonne Université, École Normale Supérieure, PSL University, CNRS, Laboratoire des Biomolécules (LBM), 75005, Paris, France.
| | - Sandrine Sagan
- Sorbonne Université, École Normale Supérieure, PSL University, CNRS, Laboratoire des Biomolécules (LBM), 75005, Paris, France.
| |
Collapse
|
8
|
Lu T, Ang CE, Zhuang X. Spatially resolved epigenomic profiling of single cells in complex tissues. Cell 2022; 185:4448-4464.e17. [PMID: 36272405 PMCID: PMC9691621 DOI: 10.1016/j.cell.2022.09.035] [Citation(s) in RCA: 90] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 08/22/2022] [Accepted: 09/26/2022] [Indexed: 11/06/2022]
Abstract
The recent development of spatial omics methods has enabled single-cell profiling of the transcriptome and 3D genome organization with high spatial resolution. Expanding the repertoire of spatial omics tools, a spatially resolved single-cell epigenomics method will accelerate understanding of the spatial regulation of cell and tissue functions. Here, we report a method for spatially resolved epigenomic profiling of single cells using in situ tagmentation and transcription followed by multiplexed imaging. We demonstrated the ability to profile histone modifications marking active promoters, putative enhancers, and silent promoters in individual cells, and generated high-resolution spatial atlas of hundreds of active promoters and putative enhancers in embryonic and adult mouse brains. Our results suggested putative promoter-enhancer pairs and enhancer hubs regulating developmentally important genes. We envision this approach will be generally applicable to spatial profiling of epigenetic modifications and DNA-binding proteins, advancing our understanding of how gene expression is spatiotemporally regulated by the epigenome.
Collapse
Affiliation(s)
- Tian Lu
- Howard Hughes Medical Institute, Harvard University, Cambridge, MA 02138, USA; Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA 02138, USA; Department of Physics, Harvard University, Cambridge, MA 02138, USA
| | - Cheen Euong Ang
- Howard Hughes Medical Institute, Harvard University, Cambridge, MA 02138, USA; Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA 02138, USA; Department of Physics, Harvard University, Cambridge, MA 02138, USA
| | - Xiaowei Zhuang
- Howard Hughes Medical Institute, Harvard University, Cambridge, MA 02138, USA; Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA 02138, USA; Department of Physics, Harvard University, Cambridge, MA 02138, USA.
| |
Collapse
|
9
|
Nisar S, Bhat AA, Masoodi T, Hashem S, Akhtar S, Ali TA, Amjad S, Chawla S, Bagga P, Frenneaux MP, Reddy R, Fakhro K, Haris M. Genetics of glutamate and its receptors in autism spectrum disorder. Mol Psychiatry 2022; 27:2380-2392. [PMID: 35296811 PMCID: PMC9135628 DOI: 10.1038/s41380-022-01506-w] [Citation(s) in RCA: 71] [Impact Index Per Article: 23.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2021] [Revised: 02/11/2022] [Accepted: 02/22/2022] [Indexed: 12/11/2022]
Abstract
Autism spectrum disorder (ASD) is a neurodevelopmental impairment characterized by deficits in social interaction skills, impaired communication, and repetitive and restricted behaviors that are thought to be due to altered neurotransmission processes. The amino acid glutamate is an essential excitatory neurotransmitter in the human brain that regulates cognitive functions such as learning and memory, which are usually impaired in ASD. Over the last several years, increasing evidence from genetics, neuroimaging, protein expression, and animal model studies supporting the notion of altered glutamate metabolism has heightened the interest in evaluating glutamatergic dysfunction in ASD. Numerous pharmacological, behavioral, and imaging studies have demonstrated the imbalance in excitatory and inhibitory neurotransmitters, thus revealing the involvement of the glutamatergic system in ASD pathology. Here, we review the effects of genetic alterations on glutamate and its receptors in ASD and the role of non-invasive imaging modalities in detecting these changes. We also highlight the potential therapeutic targets associated with impaired glutamatergic pathways.
Collapse
Affiliation(s)
- Sabah Nisar
- Laboratory of Molecular and Metabolic Imaging, Sidra Medicine, P.O. Box 26999, Doha, Qatar
| | - Ajaz A Bhat
- Laboratory of Molecular and Metabolic Imaging, Sidra Medicine, P.O. Box 26999, Doha, Qatar
| | - Tariq Masoodi
- Laboratory of Molecular and Metabolic Imaging, Sidra Medicine, P.O. Box 26999, Doha, Qatar
| | - Sheema Hashem
- Laboratory of Molecular and Metabolic Imaging, Sidra Medicine, P.O. Box 26999, Doha, Qatar
| | - Sabah Akhtar
- Laboratory of Molecular and Metabolic Imaging, Sidra Medicine, P.O. Box 26999, Doha, Qatar
| | - Tayyiba Akbar Ali
- Laboratory of Molecular and Metabolic Imaging, Sidra Medicine, P.O. Box 26999, Doha, Qatar
| | - Sara Amjad
- Shibli National College, Azamgarh, Uttar Pradesh, 276001, India
| | - Sanjeev Chawla
- Department of Radiology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Puneet Bagga
- Department of Diagnostic Imaging, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Michael P Frenneaux
- Academic Health System, Hamad Medical Corporation, P.O. Box 3050, Doha, Qatar
| | - Ravinder Reddy
- Center for Advanced Metabolic Imaging in Precision Medicine, Department of Radiology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Khalid Fakhro
- Department of Human Genetics, Sidra Medicine, P.O. Box 26999, Doha, Qatar
- Department of Genetic Medicine, Weill Cornell Medical College, P.O. Box 24144, Doha, Qatar
| | - Mohammad Haris
- Laboratory of Molecular and Metabolic Imaging, Sidra Medicine, P.O. Box 26999, Doha, Qatar.
- Center for Advanced Metabolic Imaging in Precision Medicine, Department of Radiology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, 19104, USA.
- Laboratory of Animal Research, Qatar University, P.O. Box 2713, Doha, Qatar.
| |
Collapse
|
10
|
Silva TP, Sousa-Luís R, Fernandes TG, Bekman EP, Rodrigues CAV, Vaz SH, Moreira LM, Hashimura Y, Jung S, Lee B, Carmo-Fonseca M, Cabral JMS. Transcriptome profiling of human pluripotent stem cell-derived cerebellar organoids reveals faster commitment under dynamic conditions. Biotechnol Bioeng 2021; 118:2781-2803. [PMID: 33871054 DOI: 10.1002/bit.27797] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Revised: 03/30/2021] [Accepted: 04/14/2021] [Indexed: 12/14/2022]
Abstract
Human-induced pluripotent stem cells (iPSCs) have great potential for disease modeling. However, generating iPSC-derived models to study brain diseases remains a challenge. In particular, the ability to recapitulate cerebellar development in vitro is still limited. We presented a reproducible and scalable production of cerebellar organoids by using the novel single-use Vertical-Wheel bioreactors, in which functional cerebellar neurons were obtained. Here, we evaluate the global gene expression profiles by RNA sequencing (RNA-seq) across cerebellar differentiation, demonstrating a faster cerebellar commitment in this novel dynamic differentiation protocol. Furthermore, transcriptomic profiles suggest a significant enrichment of extracellular matrix (ECM) in dynamic-derived cerebellar organoids, which can better mimic the neural microenvironment and support a consistent neuronal network. Thus, an efficient generation of organoids with cerebellar identity was achieved for the first time in a continuous process using a dynamic system without the need of organoids encapsulation in ECM-based hydrogels, allowing the possibility of large-scale production and application in high-throughput processes. The presence of factors that favors angiogenesis onset was also detected in dynamic conditions, which can enhance functional maturation of cerebellar organoids. We anticipate that large-scale production of cerebellar organoids may help developing models for drug screening, toxicological tests, and studying pathological pathways involved in cerebellar degeneration.
Collapse
Affiliation(s)
- Teresa P Silva
- Department of Bioengineering and Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Lisboa, Portugal.,Associate Laboratory i4HB - Institute for Health and Bioeconomy, Instituto Superior Técnico, Universidade de Lisboa, Lisboa, Portugal.,Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - Rui Sousa-Luís
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - Tiago G Fernandes
- Department of Bioengineering and Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Lisboa, Portugal.,Associate Laboratory i4HB - Institute for Health and Bioeconomy, Instituto Superior Técnico, Universidade de Lisboa, Lisboa, Portugal
| | - Evguenia P Bekman
- Department of Bioengineering and Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Lisboa, Portugal.,Associate Laboratory i4HB - Institute for Health and Bioeconomy, Instituto Superior Técnico, Universidade de Lisboa, Lisboa, Portugal.,Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - Carlos A V Rodrigues
- Department of Bioengineering and Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Lisboa, Portugal.,Associate Laboratory i4HB - Institute for Health and Bioeconomy, Instituto Superior Técnico, Universidade de Lisboa, Lisboa, Portugal
| | - Sandra H Vaz
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal.,Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal, Portugal
| | - Leonilde M Moreira
- Department of Bioengineering and Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Lisboa, Portugal.,Associate Laboratory i4HB - Institute for Health and Bioeconomy, Instituto Superior Técnico, Universidade de Lisboa, Lisboa, Portugal
| | | | | | - Brian Lee
- PBS Biotech, Camarillo, California, USA
| | - Maria Carmo-Fonseca
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - Joaquim M S Cabral
- Department of Bioengineering and Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Lisboa, Portugal.,Associate Laboratory i4HB - Institute for Health and Bioeconomy, Instituto Superior Técnico, Universidade de Lisboa, Lisboa, Portugal
| |
Collapse
|
11
|
Niu Y, Moghimyfiroozabad S, Moghimyfiroozabad A, Tierney TS, Alavian KN. The factors for the early and late development of midbrain dopaminergic neurons segregate into two distinct evolutionary clusters. BRAIN DISORDERS 2021. [DOI: 10.1016/j.dscb.2021.100002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
|
12
|
Vijayalingam S, Ezekiel UR, Xu F, Subramanian T, Geerling E, Hoelscher B, San K, Ganapathy A, Pemberton K, Tycksen E, Pinto AK, Brien JD, Beck DB, Chung WK, Gurnett CA, Chinnadurai G. Human iPSC-Derived Neuronal Cells From CTBP1-Mutated Patients Reveal Altered Expression of Neurodevelopmental Gene Networks. Front Neurosci 2020; 14:562292. [PMID: 33192249 PMCID: PMC7653094 DOI: 10.3389/fnins.2020.562292] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Accepted: 10/01/2020] [Indexed: 11/17/2022] Open
Abstract
A recurrent de novo mutation in the transcriptional corepressor CTBP1 is associated with neurodevelopmental disabilities in children (Beck et al., 2016, 2019; Sommerville et al., 2017). All reported patients harbor a single recurrent de novo heterozygous missense mutation (p.R342W) within the cofactor recruitment domain of CtBP1. To investigate the transcriptional activity of the pathogenic CTBP1 mutant allele in physiologically relevant human cell models, we generated induced pluripotent stem cells (iPSC) from the dermal fibroblasts derived from patients and normal donors. The transcriptional profiles of the iPSC-derived “early” neurons were determined by RNA-sequencing. Comparison of the RNA-seq data of the neurons from patients and normal donors revealed down regulation of gene networks involved in neurodevelopment, synaptic adhesion and anti-viral (interferon) response. Consistent with the altered gene expression patterns, the patient-derived neurons exhibited morphological and electrophysiological abnormalities, and susceptibility to viral infection. Taken together, our studies using iPSC-derived neuron models provide novel insights into the pathological activities of the CTBP1 p.R342W allele.
Collapse
Affiliation(s)
- S Vijayalingam
- Department of Molecular Microbiology and Immunology, Saint Louis University School of Medicine, Edward A. Doisy Research Center, St. Louis, MO, United States
| | - Uthayashanker R Ezekiel
- Department of Clinical Health Sciences, Doisy College of Health Science, Saint Louis University School of Medicine, Saint Louis, MO, United States
| | - Fenglian Xu
- Department of Biology and Henry and Amelia Nasrallah Center for Neuroscience, Saint Louis University, St. Louis, MO, United States
| | - T Subramanian
- Department of Molecular Microbiology and Immunology, Saint Louis University School of Medicine, Edward A. Doisy Research Center, St. Louis, MO, United States
| | - Elizabeth Geerling
- Department of Molecular Microbiology and Immunology, Saint Louis University School of Medicine, Edward A. Doisy Research Center, St. Louis, MO, United States
| | - Brittany Hoelscher
- Department of Biology and Henry and Amelia Nasrallah Center for Neuroscience, Saint Louis University, St. Louis, MO, United States
| | - KayKay San
- Department of Clinical Health Sciences, Doisy College of Health Science, Saint Louis University School of Medicine, Saint Louis, MO, United States
| | - Aravinda Ganapathy
- Department of Clinical Health Sciences, Doisy College of Health Science, Saint Louis University School of Medicine, Saint Louis, MO, United States
| | - Kyle Pemberton
- Department of Biology and Henry and Amelia Nasrallah Center for Neuroscience, Saint Louis University, St. Louis, MO, United States
| | - Eric Tycksen
- McDonnell Genome Institute, Washington University School of Medicine, St. Louis, MO, United States
| | - Amelia K Pinto
- Department of Molecular Microbiology and Immunology, Saint Louis University School of Medicine, Edward A. Doisy Research Center, St. Louis, MO, United States
| | - James D Brien
- Department of Molecular Microbiology and Immunology, Saint Louis University School of Medicine, Edward A. Doisy Research Center, St. Louis, MO, United States
| | - David B Beck
- National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, United States
| | - Wendy K Chung
- Department of Pediatrics and Medicine, Columbia University Medical Center, New York, NY, United States
| | - Christina A Gurnett
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, United States
| | - G Chinnadurai
- Department of Molecular Microbiology and Immunology, Saint Louis University School of Medicine, Edward A. Doisy Research Center, St. Louis, MO, United States
| |
Collapse
|
13
|
Investigating developmental and disease mechanisms of the cerebellum with pluripotent stem cells. Mol Cell Neurosci 2020; 107:103530. [PMID: 32693017 DOI: 10.1016/j.mcn.2020.103530] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Revised: 04/15/2020] [Accepted: 07/15/2020] [Indexed: 01/02/2023] Open
Abstract
The cerebellum is a brain region located in the dorsal part of the anterior hindbrain, composed of a highly stereotyped neural circuit structure with small sets of neurons. The cerebellum is involved in a wide variety of functions such as motor control, learning, cognition and others. Damage to the cerebellum often leads to impairments in motor skills (cerebellar ataxia). Cerebellar ataxia can occur as a result of neurodegenerative diseases such as spinocerebellar ataxia. Recent advances in technologies related to pluripotent stem cells and their neural differentiation has enabled researchers to investigate the mechanisms of development and of disease in the human brain. Here, we review recent applications of leading-edge stem cell technologies to the mechanistic investigation of human cerebellar development and neurological diseases affecting the cerebellum.
Collapse
|
14
|
Pirone A, Viaggi C, Cantile C, Giannessi E, Pardini C, Vaglini F, Miragliotta V. Morphological alterations of the reticular thalamic nucleus in Engrailed-2 knockout mice. J Anat 2020; 236:883-890. [PMID: 31972897 DOI: 10.1111/joa.13150] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 12/10/2019] [Accepted: 12/10/2019] [Indexed: 11/27/2022] Open
Abstract
The reticular thalamic nucleus (Rt) is a sheet of neurons that surrounds the dorsal thalamus laterally, along its dorso-ventral and rostro-caudal axes. It consists of inhibitory neurons releasing gamma-aminobutyric acid (GABA). This nucleus participates in the circuitry between the thalamus and the cerebral cortex, and its impairment is associated with neuro-psychiatric disorders. In this study, we investigated the Rt anatomy of Engrailed-2 knockout mice (En2-/- ), a mouse model of autism spectrum disorder (ASD), using parvalbumin as an immunohistochemical marker. We compared 4- and 6-week-old wild type (WT) and En2-/- mice using various morphometric parameters: cell area, shape factor, circularity and cell density. Significant differences were present in 6-week-old male mice with different genetic background (WT vs. En2-/- ): the Rt neurons of En2-/- mice showed a bigger cell area, shape factor and circularity when compared with WT. Age (4 weeks vs. 6 weeks) influenced the shape factor of WT females, the circularity and cell density of En2-/- males, and the shape factor and circularity of En2-/- females. Gender affected cell density in 4-week-old WT mice, shape factor and cellularity of 6-week-old WT mice, and cell area, shape factor and cell density of En2-/- at 6 weeks. Intrasubject (left-right) asymmetry of Rt was never observed. These results show for the first time that sex- and age-related changes occur in the Rt GABAergic neurons of the En2-/- ASD mouse model.
Collapse
Affiliation(s)
- Andrea Pirone
- Department of Veterinary Sciences, University of Pisa, Pisa, Italy
| | - Cristina Viaggi
- Department of Translational Research and of New Surgical and Medical Technologies, University of Pisa, Pisa, Italy
| | - Carlo Cantile
- Department of Veterinary Sciences, University of Pisa, Pisa, Italy
| | | | - Carla Pardini
- Department of Translational Research and of New Surgical and Medical Technologies, University of Pisa, Pisa, Italy
| | - Francesca Vaglini
- Department of Translational Research and of New Surgical and Medical Technologies, University of Pisa, Pisa, Italy
| | | |
Collapse
|
15
|
Parker HJ, Krumlauf R. A Hox gene regulatory network for hindbrain segmentation. Curr Top Dev Biol 2020; 139:169-203. [DOI: 10.1016/bs.ctdb.2020.03.001] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
16
|
Zhang X, Piano I, Messina A, D'Antongiovanni V, Crò F, Provenzano G, Bozzi Y, Gargini C, Casarosa S. Retinal defects in mice lacking the autism-associated gene Engrailed-2. Neuroscience 2019; 408:177-190. [DOI: 10.1016/j.neuroscience.2019.03.061] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Revised: 03/26/2019] [Accepted: 03/31/2019] [Indexed: 10/27/2022]
|
17
|
Hall C, Rodriguez M, Garcia J, Posfai D, DuMez R, Wictor E, Quintero OA, Hill MS, Rivera AS, Hill AL. Secreted frizzled related protein is a target of PaxB and plays a role in aquiferous system development in the freshwater sponge, Ephydatia muelleri. PLoS One 2019; 14:e0212005. [PMID: 30794564 PMCID: PMC6386478 DOI: 10.1371/journal.pone.0212005] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2018] [Accepted: 01/25/2019] [Indexed: 12/19/2022] Open
Abstract
Canonical and non-canonical Wnt signaling, as well as the Pax/Six gene network, are involved in patterning the freshwater sponge aquiferous system. Using computational approaches to identify transcription factor binding motifs in a freshwater sponge genome, we located putative PaxB binding sites near a Secreted Frizzled Related Protein (SFRP) gene in Ephydatia muelleri. EmSFRP is expressed throughout development, but with highest levels in juvenile sponges. In situ hybridization and antibody staining show EmSFRP expression throughout the pinacoderm and choanoderm in a subpopulation of amoeboid cells that may be differentiating archeocytes. Knockdown of EmSFRP leads to ectopic oscula formation during development, suggesting that EmSFRP acts as an antagonist of Wnt signaling in E. muelleri. Our findings support a hypothesis that regulation of the Wnt pathway by the Pax/Six network as well as the role of Wnt signaling in body plan morphogenesis was established before sponges diverged from the rest of the metazoans.
Collapse
Affiliation(s)
- Chelsea Hall
- Department of Biology, University of Richmond, Richmond, Virginia, United States of America
| | - Melanie Rodriguez
- Department of Biology, University of Richmond, Richmond, Virginia, United States of America
| | - Josephine Garcia
- Department of Biology, University of Richmond, Richmond, Virginia, United States of America
| | - Dora Posfai
- Department of Biology, University of Richmond, Richmond, Virginia, United States of America
| | - Rachel DuMez
- Department of Biology, University of Richmond, Richmond, Virginia, United States of America
| | - Erik Wictor
- Department of Biological Sciences, University of the Pacific, Stockton, California, United States of America
| | - Omar A. Quintero
- Department of Biology, University of Richmond, Richmond, Virginia, United States of America
| | - Malcolm S. Hill
- Department of Biology, University of Richmond, Richmond, Virginia, United States of America
- Department of Biology, Bates College, Lewiston, Maine, United States of America
| | - Ajna S. Rivera
- Department of Biological Sciences, University of the Pacific, Stockton, California, United States of America
| | - April L. Hill
- Department of Biology, University of Richmond, Richmond, Virginia, United States of America
- Department of Biology, Bates College, Lewiston, Maine, United States of America
| |
Collapse
|
18
|
Di Nardo AA, Fuchs J, Joshi RL, Moya KL, Prochiantz A. The Physiology of Homeoprotein Transduction. Physiol Rev 2019; 98:1943-1982. [PMID: 30067157 DOI: 10.1152/physrev.00018.2017] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
The homeoprotein family comprises ~300 transcription factors and was long seen as primarily involved in developmental programs through cell autonomous regulation. However, recent evidence reveals that many of these factors are also expressed in the adult where they exert physiological functions not yet fully deciphered. Furthermore, the DNA-binding domain of most homeoproteins contains two signal sequences allowing their secretion and internalization, thus intercellular transfer. This review focuses on this new-found signaling in cell migration, axon guidance, and cerebral cortex physiological homeostasis and speculates on how it may play important roles in early arealization of the neuroepithelium. It also describes the use of homeoproteins as therapeutic proteins in mouse models of diseases affecting the central nervous system, in particular Parkinson disease and glaucoma.
Collapse
Affiliation(s)
- Ariel A Di Nardo
- Centre for Interdisciplinary Research in Biology (CIRB), Collège de France, CNRS UMR 7241, INSERM U1050, Labex MemoLife, PSL Research University , Paris , France
| | - Julia Fuchs
- Centre for Interdisciplinary Research in Biology (CIRB), Collège de France, CNRS UMR 7241, INSERM U1050, Labex MemoLife, PSL Research University , Paris , France
| | - Rajiv L Joshi
- Centre for Interdisciplinary Research in Biology (CIRB), Collège de France, CNRS UMR 7241, INSERM U1050, Labex MemoLife, PSL Research University , Paris , France
| | - Kenneth L Moya
- Centre for Interdisciplinary Research in Biology (CIRB), Collège de France, CNRS UMR 7241, INSERM U1050, Labex MemoLife, PSL Research University , Paris , France
| | - Alain Prochiantz
- Centre for Interdisciplinary Research in Biology (CIRB), Collège de France, CNRS UMR 7241, INSERM U1050, Labex MemoLife, PSL Research University , Paris , France
| |
Collapse
|
19
|
Impaired Neuronal Differentiation of Neural Stem Cells Lacking the Engrailed-2 Gene. Neuroscience 2018; 386:137-149. [DOI: 10.1016/j.neuroscience.2018.06.032] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2018] [Revised: 06/01/2018] [Accepted: 06/19/2018] [Indexed: 12/24/2022]
|
20
|
Cerebellar networks and neuropathology of cerebellar developmental disorders. HANDBOOK OF CLINICAL NEUROLOGY 2018; 154:109-128. [PMID: 29903435 DOI: 10.1016/b978-0-444-63956-1.00007-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The cerebellar system is a series of axonal projections and synaptic circuits as networks, similar to those of the limbic system and those subserving the propagation and spread of seizures. Three principal cerebellar networks are identified and cerebellar disease often affects components of the networks other than just the cerebellar cortex. Contemporary developmental neuropathology of the cerebellum is best considered in the context of alterations of developmental processes: embryonic segmentation and genetic gradients along the three axes of the neural tube, individual neuronal and glial cell differentiation, migration, synaptogenesis, and myelination. Precisely timed developmental processes may be delayed or precocious rhombencephalosynapsis and pontocerebellar hypoplasia exemplify opposite gradients in the horizontal axis. Chiari II malformation may be reconsidered as a disorder of segmentation rather than simply due to mechanical forces upon normally developing hindbrain structures. Cellular nodules in the roof of the fourth ventricle are heterotopia of histologically differentiated but architecturally disoriented and disorganized neurons and glial cells; they often are less mature immunocytochemically than similar cells in adjacent normal folia. Cell rests are nodules of undifferentiated neuroepithelial cells. Both are frequent in human fetuses and neonates. Axonal projections from heterotopia to adjacent cerebellar folia or nuclei are few or absent, hence these nodules are clinically silent despite neuronal differentiation.
Collapse
|
21
|
Holmes DB, Heine VM. Streamlined 3D Cerebellar Differentiation Protocol with Optional 2D Modification. J Vis Exp 2017. [PMID: 29286492 PMCID: PMC5755539 DOI: 10.3791/56888] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
Reducing the complexity and cost of differentiation protocols is important for researchers. This interest fits with concerns about possible unintended effects that extrinsic patterning factors might introduce into human pluripotent stem cell (hPSC) models of brain development or pathophysiology, such as masking disease phenotype. Here, we present two cerebellar differentiation protocols for hPSCs, designed with simpler startup method, fewer patterning factors, and less material requirements than previous protocols. Recently, we developed culture procedures, which generate free-floating 3-dimensional (3D) products consistent with other brain "organoid" protocols, including morphologies relevant to modeling brain development such as sub/ventricular zone- and rhombic lip-like structures. The second uses an adherent, 2D monolayer procedure to complete differentiation, which is shown capable of generating functional cerebellar neurons, as products are positive for cerebellar-associated markers, and exhibit neuron-like calcium influxes. Together, these protocols offer scientists a choice of options suited to different research purposes, as well as a basic model for testing other types of streamlined neural differentiations.
Collapse
Affiliation(s)
- Dwayne B Holmes
- Department of Pediatrics/Child Neurology, Amsterdam Neuroscience, VU University Medical Center
| | - Vivi M Heine
- Department of Pediatrics/Child Neurology, Amsterdam Neuroscience, VU University Medical Center; Department of Complex Trait Genetics, Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, Vrije Universiteit Amsterdam;
| |
Collapse
|
22
|
Soltani A, Lebrun S, Carpentier G, Zunino G, Chantepie S, Maïza A, Bozzi Y, Desnos C, Darchen F, Stettler O. Increased signaling by the autism-related Engrailed-2 protein enhances dendritic branching and spine density, alters synaptic structural matching, and exaggerates protein synthesis. PLoS One 2017; 12:e0181350. [PMID: 28809922 PMCID: PMC5557355 DOI: 10.1371/journal.pone.0181350] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2016] [Accepted: 06/29/2017] [Indexed: 12/13/2022] Open
Abstract
Engrailed 1 (En1) and 2 (En2) code for closely related homeoproteins acting as transcription factors and as signaling molecules that contribute to midbrain and hindbrain patterning, to development and maintenance of monoaminergic pathways, and to retinotectal wiring. En2 has been suggested to be an autism susceptibility gene and individuals with autism display an overexpression of this homeogene but the mechanisms remain unclear. We addressed in the present study the effect of exogenously added En2 on the morphology of hippocampal cells that normally express only low levels of Engrailed proteins. By means of RT-qPCR, we confirmed that En1 and En2 were expressed at low levels in hippocampus and hippocampal neurons, and observed a pronounced decrease in En2 expression at birth and during the first postnatal week, a period characterized by intense synaptogenesis. To address a putative effect of Engrailed in dendritogenesis or synaptogenesis, we added recombinant En1 or En2 proteins to hippocampal cell cultures. Both En1 and En2 treatment increased the complexity of the dendritic tree of glutamatergic neurons, but only En2 increased that of GABAergic cells. En1 increased the density of dendritic spines both in vitro and in vivo. En2 had similar but less pronounced effect on spine density. The number of mature synapses remained unchanged upon En1 treatment but was reduced by En2 treatment, as well as the area of post-synaptic densities. Finally, both En1 and En2 elevated mTORC1 activity and protein synthesis in hippocampal cells, suggesting that some effects of Engrailed proteins may require mRNA translation. Our results indicate that Engrailed proteins can play, even at low concentrations, an active role in the morphogenesis of hippocampal cells. Further, they emphasize the over-regulation of GABA cell morphology and the vulnerability of excitatory synapses in a pathological context of En2 overexpression.
Collapse
Affiliation(s)
- Asma Soltani
- UMR 8250, Centre National de la Recherche Scientifique, Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Solène Lebrun
- UMR 8250, Centre National de la Recherche Scientifique, Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Gilles Carpentier
- Laboratoire Croissance, Réparation et Régénération Tissulaires (CRRET), EA 4397 / ERL 9215, Centre National de la Recherche Scientifique, Université Paris Est Créteil, Créteil, France
| | - Giulia Zunino
- Centre for Integrative Biology, University of Trento, Trento, Italy
| | - Sandrine Chantepie
- Laboratoire Croissance, Réparation et Régénération Tissulaires (CRRET), EA 4397 / ERL 9215, Centre National de la Recherche Scientifique, Université Paris Est Créteil, Créteil, France
| | - Auriane Maïza
- Laboratoire Croissance, Réparation et Régénération Tissulaires (CRRET), EA 4397 / ERL 9215, Centre National de la Recherche Scientifique, Université Paris Est Créteil, Créteil, France
| | - Yuri Bozzi
- Centre for Integrative Biology, University of Trento, Trento, Italy
| | - Claire Desnos
- UMR 8250, Centre National de la Recherche Scientifique, Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - François Darchen
- UMR 8250, Centre National de la Recherche Scientifique, Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Olivier Stettler
- UMR 8250, Centre National de la Recherche Scientifique, Université Paris Descartes, Sorbonne Paris Cité, Paris, France
- Laboratoire Croissance, Réparation et Régénération Tissulaires (CRRET), EA 4397 / ERL 9215, Centre National de la Recherche Scientifique, Université Paris Est Créteil, Créteil, France
| |
Collapse
|
23
|
Knockdown of epigenetic transcriptional co-regulator Brd2a disrupts apoptosis and proper formation of hindbrain and midbrain-hindbrain boundary (MHB) region in zebrafish. Mech Dev 2017; 146:10-30. [PMID: 28549975 DOI: 10.1016/j.mod.2017.05.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2016] [Revised: 05/17/2017] [Accepted: 05/22/2017] [Indexed: 01/03/2023]
Abstract
Brd2 is a member of the bromodomain-extraterminal domain (BET) family of proteins and functions as an acetyl-histone-directed transcriptional co-regulator and recruitment scaffold in chromatin modification complexes affecting signal-dependent transcription. While Brd2 acts as a protooncogene in mammalian blood, developmental studies link it to regulation of neuronal apoptosis and epilepsy, and complete knockout of the gene is invariably embryonic lethal. In Drosophila, the Brd2 homolog acts as a maternal effect factor necessary for segment formation and identity and proper expression of homeotic loci, including Ultrabithorax and engrailed. To test the various roles attributed to Brd2 in a single developmental system representing a non-mammalian vertebrate, we conducted a phenotypic characterization of Brd2a deficient zebrafish embryos produced by morpholino knockdown and corroborated by Crispr-Cas9 disruption and small molecule inhibitor treatments. brd2aMO morphants exhibit reduced hindbrain with an ill-defined midbrain-hindbrain boundary (MHB) region; irregular notochord, neural tube, and somites; and abnormalities in ventral trunk and ventral nerve cord interneuron positioning. Using whole mount TUNEL and confocal microscopy, we uncover a significant decrease, then a dramatic increase, of p53-independent cell death at the start and end of segmentation, respectively. In contrast, using qualitative and quantitative analyses of BrdU incorporation, phosphohistone H3-tagging, and flow cytometry, we detect little effect of Brd2a knockdown on overall proliferation levels in embryos. RNA in situ hybridization shows reduced or absent expression of homeobox gene eng2a and paired box gene pax2a, in the hindbrain domain of the MHB region, and an overabundance of pax2a-positive kidney progenitors, in knockdowns. Together, these results suggest an evolutionarily conserved role for Brd2 in the proper formation and/or patterning of segmented tissues, including the vertebrate CNS, where it acts as a bi-modal regulator of apoptosis, and is necessary, directly or indirectly, for proper expression of genes that pattern the MHB and/or regulate differentiation in the anterior hindbrain.
Collapse
|
24
|
Abstract
Here, we report a 3D cerebellar differentiation protocol with quick startup method, defined medium and no special materials or handling requirements. Three fibroblast growth factors (FGF2, 4 and 8) were used for cerebellar patterning and smoothened agonist (SAG) for granule cell development. After 35 days, differentiation products exhibited similar structures and neuronal markers reported in prior ‘organoid’ and ‘spheroid’ protocols. This included cells positive for KIRREL2 (a marker of early cerebellar neuroepithelium) and ZIC1 (a marker for granule cells). Follow-up tests indicated that addition of FGFs, if helpful, was not required to generate observed structures and cell types. This suggests that intrinsic production of patterning factors by aggregates themselves may be adequate for region-specific 3D modeling. This protocol may be used as a quick, easy and cost-efficient method for 3D culture, whether to research development of the early cerebellar neuroepithelium, a base to generate mature cortical structures, or to optimize minimal-factor protocols for other brain regions. Summary: We present a simplified method to generate early neuroepithelium of the cerebellar cortex in a 3D culture using defined medium and minimal addition of growth factors.
Collapse
Affiliation(s)
- Dwayne B Holmes
- Department of Pediatrics/Child Neurology, Amsterdam Neuroscience, VU University Medical Center, Amsterdam 1081 HV, The Netherlands
| | - Vivi M Heine
- Department of Pediatrics/Child Neurology, Amsterdam Neuroscience, VU University Medical Center, Amsterdam 1081 HV, The Netherlands .,Department of Complex Trait Genetics, Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, VU University, Amsterdam 1081 HV, The Netherlands
| |
Collapse
|
25
|
Harada H, Sato T, Nakamura H. Fgf8 signaling for development of the midbrain and hindbrain. Dev Growth Differ 2016; 58:437-45. [PMID: 27273073 DOI: 10.1111/dgd.12293] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2016] [Revised: 04/22/2016] [Accepted: 04/22/2016] [Indexed: 01/31/2023]
Abstract
In this paper, we review how midbrain and hindbrain are specified. Otx2 and Gbx2 are expressed from the early phase of development, and their expression abuts at the midbrain hindbrain boundary (MHB), where Fgf8 expression is induced, and functions as an organizing molecule for the midbrain and hindbrain. Fgf8 induces En1 and Pax2 expression at the region where Otx2 is expressed to specify midbrain. Fgf8 activates Ras-ERK pathway to specify hindbrain. Downstream of ERK, Pea3 specifies isthmus (rhombomere 0, r0), and Irx2 may specify r1, where the cerebellum is formed.
Collapse
Affiliation(s)
- Hidekiyo Harada
- Genetics and Development Division, Toronto Krembil Research Institute, Toronto, Ontario, Canada.,Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Tatsuya Sato
- Department of Developmental Neuroscience, Graduate School of Medicine, Tohoku University, Sendai, 980-8575, Japan.,Frontier Research Institute for Interdisciplinary Science, Tohoku University, Sendai, 980-8578, Japan
| | - Harukazu Nakamura
- Frontier Research Institute for Interdisciplinary Science, Tohoku University, Sendai, 980-8578, Japan
| |
Collapse
|
26
|
Verpeut JL, DiCicco-Bloom E, Bello NT. Ketogenic diet exposure during the juvenile period increases social behaviors and forebrain neural activation in adult Engrailed 2 null mice. Physiol Behav 2016; 161:90-98. [PMID: 27080080 DOI: 10.1016/j.physbeh.2016.04.001] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2016] [Revised: 03/16/2016] [Accepted: 04/02/2016] [Indexed: 11/16/2022]
Abstract
Prolonged consumption of ketogenic diets (KD) has reported neuroprotective benefits. Several studies suggest KD interventions could be useful in the management of neurological and developmental disorders. Alterations in the Engrailed (En) genes, specifically Engrailed 2 (En2), have neurodevelopmental consequences and produce autism-related behaviors. The following studies used En2 knockout (KO; En2(-/-)), and wild-type (WT; En2(+/+)), male mice fed either KD (80% fat, 0.1% carbohydrates) or control diet (CD; 10% fat, 70% carbohydrates). The objective was to determine whether a KD fed from weaning at postnatal day (PND) 21 to adulthood (PND 60) would alter brain monoamines concentrations, previously found dysregulated, and improve social outcomes. In WT animals, there was an increase in hypothalamic norepinephrine content in the KD-fed group. However, regional monoamines were not altered in KO mice in KD-fed compared with CD-fed group. In order to determine the effects of juvenile exposure to KD in mice with normal blood ketone levels, separate experiments were conducted in mice removed from the KD or CD and fed standard chow for 2days (PND 62). In a three-chamber social test with a novel mouse, KO mice previously exposed to the KD displayed similar social and self-grooming behaviors compared with the WT group. Groups previously exposed to a KD, regardless of genotype, had more c-Fos-positive cells in the cingulate cortex, lateral septal nuclei, and anterior bed nucleus of the stria terminalis. In the novel object condition, KO mice previously exposed to KD had similar behavioral responses and pattern of c-Fos immunoreactivity compared with the WT group. Thus, juvenile exposure to KD resulted in short-term consequences of improving social interactions and appropriate exploratory behaviors in a mouse model that displays autism-related behaviors. Such findings further our understanding of metabolic-based therapies for neurological and developmental disorders.
Collapse
Affiliation(s)
- Jessica L Verpeut
- Department of Animal Sciences, Graduate Program in Endocrinology and Animal Biosciences, School of Environmental and Biological Sciences, Rutgers, The State University of New Jersey, New Brunswick, NJ, USA
| | - Emanuel DiCicco-Bloom
- Department of Neuroscience and Cell Biology/Pediatrics, Rutgers Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, Piscataway, NJ, USA
| | - Nicholas T Bello
- Department of Animal Sciences, Graduate Program in Endocrinology and Animal Biosciences, School of Environmental and Biological Sciences, Rutgers, The State University of New Jersey, New Brunswick, NJ, USA.
| |
Collapse
|
27
|
Rapacioli M, Palma V, Flores V. Morphogenetic and Histogenetic Roles of the Temporal-Spatial Organization of Cell Proliferation in the Vertebrate Corticogenesis as Revealed by Inter-specific Analyses of the Optic Tectum Cortex Development. Front Cell Neurosci 2016; 10:67. [PMID: 27013978 PMCID: PMC4794495 DOI: 10.3389/fncel.2016.00067] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2015] [Accepted: 03/01/2016] [Indexed: 12/11/2022] Open
Abstract
The central nervous system areas displaying the highest structural and functional complexity correspond to the so called cortices, i.e., concentric alternating neuronal and fibrous layers. Corticogenesis, i.e., the development of the cortical organization, depends on the temporal-spatial organization of several developmental events: (a) the duration of the proliferative phase of the neuroepithelium, (b) the relative duration of symmetric (expansive) versus asymmetric (neuronogenic) sub phases, (c) the spatial organization of each kind of cell division, (e) the time of determination and cell cycle exit and (f) the time of onset of the post-mitotic neuronal migration and (g) the time of onset of the neuronal structural and functional differentiation. The first five events depend on molecular mechanisms that perform a fine tuning of the proliferative activity. Changes in any of them significantly influence the cortical size or volume (tangential expansion and radial thickness), morphology, architecture and also impact on neuritogenesis and synaptogenesis affecting the cortical wiring. This paper integrates information, obtained in several species, on the developmental roles of cell proliferation in the development of the optic tectum (OT) cortex, a multilayered associative area of the dorsal (alar) midbrain. The present review (1) compiles relevant information on the temporal and spatial organization of cell proliferation in different species (fish, amphibians, birds, and mammals), (2) revises the main molecular events involved in the isthmic organizer (IsO) determination and localization, (3) describes how the patterning installed by IsO is translated into spatially organized neural stem cell proliferation (i.e., by means of growth factors, receptors, transcription factors, signaling pathways, etc.) and (4) describes the morpho- and histogenetic effect of a spatially organized cell proliferation in the above mentioned species. A brief section on the OT evolution is also included. This section considers how the differential operation of cell proliferation could explain differences among species.
Collapse
Affiliation(s)
- Melina Rapacioli
- Interdisciplinary Group in Theoretical Biology, Department of Biostructural Sciences, Favaloro UniversityBuenos Aires, Argentina
| | - Verónica Palma
- Laboratory of Stem Cell and Developmental Biology, Faculty of Science, University of ChileSantiago, Chile
| | - Vladimir Flores
- Interdisciplinary Group in Theoretical Biology, Department of Biostructural Sciences, Favaloro UniversityBuenos Aires, Argentina
| |
Collapse
|
28
|
Gonsar N, Coughlin A, Clay-Wright JA, Borg BR, Kindt LM, Liang JO. Temporal and spatial requirements for Nodal-induced anterior mesendoderm and mesoderm in anterior neurulation. Genesis 2016; 54:3-18. [PMID: 26528772 DOI: 10.1002/dvg.22908] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2015] [Revised: 10/29/2015] [Accepted: 10/30/2015] [Indexed: 01/28/2023]
Abstract
Zebrafish with defective Nodal signaling have a phenotype analogous to the fatal human birth defect anencephaly, which is caused by an open anterior neural tube. Previous work in our laboratory found that anterior open neural tube phenotypes in Nodal signaling mutants were caused by lack of mesendodermal/mesodermal tissues. Defects in these mutants are already apparent at neural plate stage, before the neuroepithelium starts to fold into a tube. Consistent with this, we found that the requirement for Nodal signaling maps to mid-late blastula stages. This timing correlates with the timing of prechordal plate mesendoderm and anterior mesoderm induction, suggesting these tissues act to promote neurulation. To further identify tissues important for neurulation, we took advantage of the variable phenotypes in Nodal signaling-deficient sqt mutant and Lefty1-overexpressing embryos. Statistical analysis indicated a strong, positive correlation between a closed neural tube and presence of several mesendoderm/mesoderm-derived tissues (hatching glands, cephalic paraxial mesoderm, notochord, and head muscles). However, the neural tube was closed in a subset of embryos that lacked any one of these tissues. This suggests that several types of Nodal-induced mesendodermal/mesodermal precursors are competent to promote neurulation.
Collapse
Affiliation(s)
- Ngawang Gonsar
- Department of Biology, University of Minnesota Duluth, Duluth, MN.,Integrated Biosciences Graduate Program, University of Minnesota, Duluth, MN
| | - Alicia Coughlin
- Department of Biology, University of Minnesota Duluth, Duluth, MN.,Integrated Biosciences Graduate Program, University of Minnesota, Duluth, MN
| | | | - Bethanie R Borg
- Department of Biology, University of Minnesota Duluth, Duluth, MN
| | - Lexy M Kindt
- Department of Biology, University of Minnesota Duluth, Duluth, MN.,Integrated Biosciences Graduate Program, University of Minnesota, Duluth, MN
| | - Jennifer O Liang
- Department of Biology, University of Minnesota Duluth, Duluth, MN.,Integrated Biosciences Graduate Program, University of Minnesota, Duluth, MN
| |
Collapse
|
29
|
Abstract
The difficulty to understand, diagnose, and treat neurological disorders stems from the great complexity of the central nervous system on different levels of physiological granularity. The individual components, their interactions, and dynamics involved in brain development and function can be represented as molecular, cellular, or functional networks, where diseases are perturbations of networks. These networks can become a useful research tool in investigating neurological disorders if they are properly tailored to reflect corresponding mechanisms. Here, we review approaches to construct networks specific for neurological disorders describing disease-related pathology on different scales: the molecular, cellular, and brain level. We also briefly discuss cross-scale network analysis as a necessary integrator of these scales.
Collapse
|
30
|
|
31
|
Sundberg M, Sahin M. Cerebellar Development and Autism Spectrum Disorder in Tuberous Sclerosis Complex. J Child Neurol 2015; 30:1954-62. [PMID: 26303409 PMCID: PMC4644486 DOI: 10.1177/0883073815600870] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2015] [Accepted: 07/15/2015] [Indexed: 01/08/2023]
Abstract
Approximately 50% of patients with the genetic disease tuberous sclerosis complex present with autism spectrum disorder. Although a number of studies have investigated the link between autism and tuberous sclerosis complex, the etiology of autism spectrum disorder in these patients remains unclear. Abnormal cerebellar function during critical phases of development could disrupt functional processes in the brain, leading to development of autistic features. Accordingly, the authors review the potential role of cerebellar dysfunction in the pathogenesis of autism spectrum disorder in tuberous sclerosis complex. The authors also introduce conditional knockout mouse models of Tsc1 and Tsc2 that link cerebellar circuitry to the development of autistic-like features. Taken together, these preclinical and clinical investigations indicate the cerebellum has a profound regulatory role during development of social communication and repetitive behaviors.
Collapse
Affiliation(s)
- Maria Sundberg
- The F.M. Kirby Neurobiology Center, Department of Neurology, Children’s Hospital Boston, Harvard Medical School, Boston, MA, USA
| | - Mustafa Sahin
- F.M. Kirby Neurobiology Center, Department of Neurology, Children's Hospital Boston, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
32
|
Cheah PS, Thomas PQ. SOX3 expression in the glial system of the developing and adult mouse cerebellum. SPRINGERPLUS 2015; 4:400. [PMID: 26261758 PMCID: PMC4527974 DOI: 10.1186/s40064-015-1194-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/22/2015] [Accepted: 07/29/2015] [Indexed: 11/17/2022]
Abstract
Background The cerebellum plays a vital role in equilibrium, motor control, and motor learning. The discrete neural and glial fates of cerebellar cells are determined by the molecular specifications (e.g. transcription factors) of neuroprogenitor cells that are influenced by local microenvironment signals. In this study, we evaluated the expression and function of Sox3, a single-exon gene located on the X chromosome, in the developing cerebellum. Result In the embryonic and early postnatal cerebellum, SOX3-positive-cells were detected in the ventricular zone, indicating that SOX3 expression is present in a subset of the cerebellar precursor cell population. In the young adult cerebellum, this expression was diminished in cerebellar cells, suggesting its limited role in cerebellar progenitors. SOX3-positive-cells were also found in the cerebellar mantle zone. Further immunohistochemistry analyses revealed that SOX3 was not expressed in Purkinje neurons. Using glial markers in the early postnatal cerebellum, we found that virtually all of the SOX3-positive-cells were glial cells, although not all glial cells were SOX3-positive-cells. We also determined the impact of transgenic expression using a loss-of-function (Sox3 null) model. We did not observe any developmental defects in the cerebellum of the Sox3 null mice. Conclusions Our results indicate that the SOX3 protein is not expressed in cerebellar neurons and is instead expressed exclusively in the cerebellar glial system in a subset of mature glial cells. Although the expression of Sox3 cerebellar glial development is lineage-restricted, it appears that the absence of Sox3 in the ventricular germinal epithelium and migrating glia does not affect cerebellar development, suggesting functional redundancy with other SoxB1 subgroup genes. Electronic supplementary material The online version of this article (doi:10.1186/s40064-015-1194-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Pike-See Cheah
- Department of Human Anatomy, Faculty of Medicine, Health Sciences, University Putra Malaysia, 43400 Serdang, Selangor Malaysia ; Neurobiology and Genetics Group, Genetics and Regenerative Medicine Research Center, Faculty of Medicine and Health Sciences, University Putra Malaysia, 43400 Serdang, Malaysia
| | - Paul Q Thomas
- Discipline of Biochemistry, School of Molecular and Biomedical Sciences, University of Adelaide, Adelaide, Australia
| |
Collapse
|
33
|
Serotonin abnormalities in Engrailed-2 knockout mice: New insight relevant for a model of Autism Spectrum Disorder. Neurochem Int 2015; 87:34-42. [DOI: 10.1016/j.neuint.2015.05.004] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2014] [Revised: 04/02/2015] [Accepted: 05/14/2015] [Indexed: 12/14/2022]
|
34
|
Genestine M, Lin L, Durens M, Yan Y, Jiang Y, Prem S, Bailoor K, Kelly B, Sonsalla PK, Matteson PG, Silverman J, Crawley JN, Millonig JH, DiCicco-Bloom E. Engrailed-2 (En2) deletion produces multiple neurodevelopmental defects in monoamine systems, forebrain structures and neurogenesis and behavior. Hum Mol Genet 2015. [PMID: 26220976 DOI: 10.1093/hmg/ddv301] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Many genes involved in brain development have been associated with human neurodevelopmental disorders, but underlying pathophysiological mechanisms remain undefined. Human genetic and mouse behavioral analyses suggest that ENGRAILED-2 (EN2) contributes to neurodevelopmental disorders, especially autism spectrum disorder. In mouse, En2 exhibits dynamic spatiotemporal expression in embryonic mid-hindbrain regions where monoamine neurons emerge. Considering their importance in neuropsychiatric disorders, we characterized monoamine systems in relation to forebrain neurogenesis in En2-knockout (En2-KO) mice. Transmitter levels of serotonin, dopamine and norepinephrine (NE) were dysregulated from Postnatal day 7 (P7) to P21 in En2-KO, though NE exhibited the greatest abnormalities. While NE levels were reduced ∼35% in forebrain, they were increased 40 -: 75% in hindbrain and cerebellum, and these patterns paralleled changes in locus coeruleus (LC) fiber innervation, respectively. Although En2 promoter was active in Embryonic day 14.5 -: 15.5 LC neurons, expression diminished thereafter and gene deletion did not alter brainstem NE neuron numbers. Significantly, in parallel with reduced NE levels, En2-KO forebrain regions exhibited reduced growth, particularly hippocampus, where P21 dentate gyrus granule neurons were decreased 16%, suggesting abnormal neurogenesis. Indeed, hippocampal neurogenic regions showed increased cell death (+77%) and unexpectedly, increased proliferation. Excess proliferation was restricted to early Sox2/Tbr2 progenitors whereas increased apoptosis occurred in differentiating (Dcx) neuroblasts, accompanied by reduced newborn neuron survival. Abnormal neurogenesis may reflect NE deficits because intra-hippocampal injections of β-adrenergic agonists reversed cell death. These studies suggest that disruption of hindbrain patterning genes can alter monoamine system development and thereby produce forebrain defects that are relevant to human neurodevelopmental disorders.
Collapse
Affiliation(s)
- Matthieu Genestine
- Department of Neuroscience and Cell Biology, Rutgers Robert Wood Johnson Medical School, Rutgers
| | - Lulu Lin
- Department of Neuroscience and Cell Biology, Rutgers Robert Wood Johnson Medical School, Rutgers, Graduate School of Biological Sciences, Rutgers
| | - Madel Durens
- Department of Neuroscience and Cell Biology, Rutgers Robert Wood Johnson Medical School, Rutgers, Graduate School of Biological Sciences, Rutgers
| | - Yan Yan
- Department of Neuroscience and Cell Biology, Rutgers Robert Wood Johnson Medical School, Rutgers, Graduate School of Biological Sciences, Rutgers
| | - Yiqin Jiang
- Department of Neuroscience and Cell Biology, Rutgers Robert Wood Johnson Medical School, Rutgers
| | - Smrithi Prem
- Department of Neuroscience and Cell Biology, Rutgers Robert Wood Johnson Medical School, Rutgers
| | - Kunal Bailoor
- Rutgers, The State University of New Jersey, Piscataway, NJ, USA
| | - Brian Kelly
- Rutgers, The State University of New Jersey, Piscataway, NJ, USA
| | - Patricia K Sonsalla
- Department of Neurology, Rutgers Robert Wood Johnson Medical School, Rutgers
| | - Paul G Matteson
- Center for Advanced Biotechnology and Medicine, Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, Piscataway, NJ, USA
| | - Jill Silverman
- MIND Institute, University of California Davis School of Medicine, Sacramento, CA, USA
| | - Jacqueline N Crawley
- MIND Institute, University of California Davis School of Medicine, Sacramento, CA, USA
| | - James H Millonig
- Department of Neuroscience and Cell Biology, Rutgers Robert Wood Johnson Medical School, Rutgers, Center for Advanced Biotechnology and Medicine, Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, Piscataway, NJ, USA
| | - Emanuel DiCicco-Bloom
- Department of Neuroscience and Cell Biology, Rutgers Robert Wood Johnson Medical School, Rutgers, Graduate School of Biological Sciences, Rutgers, Department of Pediatrics, Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, New Brunswick, NJ, USA and
| |
Collapse
|
35
|
Rampon C, Gauron C, Lin T, Meda F, Dupont E, Cosson A, Ipendey E, Frerot A, Aujard I, Le Saux T, Bensimon D, Jullien L, Volovitch M, Vriz S, Joliot A. Control of brain patterning by Engrailed paracrine transfer: a new function of the Pbx interaction domain. Development 2015; 142:1840-9. [PMID: 25926358 PMCID: PMC4440920 DOI: 10.1242/dev.114181] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2014] [Accepted: 03/02/2015] [Indexed: 12/28/2022]
Abstract
Homeoproteins of the Engrailed family are involved in the patterning of mesencephalic boundaries through a mechanism classically ascribed to their transcriptional functions. In light of recent reports on the paracrine activity of homeoproteins, including Engrailed, we asked whether Engrailed intercellular transfer was also involved in brain patterning and boundary formation. Using time-controlled activation of Engrailed combined with tools that block its transfer, we show that the positioning of the diencephalic-mesencephalic boundary (DMB) requires Engrailed paracrine activity. Both zebrafish Eng2a and Eng2b are competent for intercellular transfer in vivo, but only extracellular endogenous Eng2b, and not Eng2a, participates in DMB positioning. In addition, disruption of the Pbx-interacting motif in Engrailed, known to strongly reduce the gain-of-function phenotype, also downregulates Engrailed transfer, thus revealing an unsuspected participation of the Pbx interaction domain in this pathway.
Collapse
Affiliation(s)
- Christine Rampon
- Université Paris Diderot, Sorbonne Paris Cité, Paris 75205, Cedex 13, France Center for Interdisciplinary Research in Biology (CIRB) - CNRS UMR 7241, INSERM U1050, Labex MemoLife, PSL Research University, Collège de France, Paris F-75005, France
| | - Carole Gauron
- Center for Interdisciplinary Research in Biology (CIRB) - CNRS UMR 7241, INSERM U1050, Labex MemoLife, PSL Research University, Collège de France, Paris F-75005, France
| | - Thibault Lin
- Center for Interdisciplinary Research in Biology (CIRB) - CNRS UMR 7241, INSERM U1050, Labex MemoLife, PSL Research University, Collège de France, Paris F-75005, France
| | - Francesca Meda
- Center for Interdisciplinary Research in Biology (CIRB) - CNRS UMR 7241, INSERM U1050, Labex MemoLife, PSL Research University, Collège de France, Paris F-75005, France École Normale Supérieure, Institute of Biology at the Ecole Normale Supérieure (IBENS), CNRS UMR8197, INSERM U1024, PSL Research University, Paris F-75005, France
| | - Edmond Dupont
- Center for Interdisciplinary Research in Biology (CIRB) - CNRS UMR 7241, INSERM U1050, Labex MemoLife, PSL Research University, Collège de France, Paris F-75005, France
| | - Adrien Cosson
- Center for Interdisciplinary Research in Biology (CIRB) - CNRS UMR 7241, INSERM U1050, Labex MemoLife, PSL Research University, Collège de France, Paris F-75005, France
| | - Eliane Ipendey
- Center for Interdisciplinary Research in Biology (CIRB) - CNRS UMR 7241, INSERM U1050, Labex MemoLife, PSL Research University, Collège de France, Paris F-75005, France École Normale Supérieure, Institute of Biology at the Ecole Normale Supérieure (IBENS), CNRS UMR8197, INSERM U1024, PSL Research University, Paris F-75005, France
| | - Alice Frerot
- Center for Interdisciplinary Research in Biology (CIRB) - CNRS UMR 7241, INSERM U1050, Labex MemoLife, PSL Research University, Collège de France, Paris F-75005, France
| | - Isabelle Aujard
- Ecole Normale Supérieure-PSL Research University, Département de Chimie, UMR 8640 CNRS-ENS-UPMC PASTEUR, 24, rue Lhomond, Paris 75005, France
| | - Thomas Le Saux
- Ecole Normale Supérieure-PSL Research University, Département de Chimie, UMR 8640 CNRS-ENS-UPMC PASTEUR, 24, rue Lhomond, Paris 75005, France
| | - David Bensimon
- École Normale Supérieure, Institute of Biology at the Ecole Normale Supérieure (IBENS), CNRS UMR8197, INSERM U1024, PSL Research University, Paris F-75005, France Laboratoire de Physique Statistique, UMR CNRS-ENS 8550, Paris F-75005, France Department of Chemistry and Biochemistry, UCLA, Los Angeles, CA 90095-1569, USA
| | - Ludovic Jullien
- Ecole Normale Supérieure-PSL Research University, Département de Chimie, UMR 8640 CNRS-ENS-UPMC PASTEUR, 24, rue Lhomond, Paris 75005, France
| | - Michel Volovitch
- Center for Interdisciplinary Research in Biology (CIRB) - CNRS UMR 7241, INSERM U1050, Labex MemoLife, PSL Research University, Collège de France, Paris F-75005, France École Normale Supérieure, Institute of Biology at the Ecole Normale Supérieure (IBENS), CNRS UMR8197, INSERM U1024, PSL Research University, Paris F-75005, France
| | - Sophie Vriz
- Université Paris Diderot, Sorbonne Paris Cité, Paris 75205, Cedex 13, France Center for Interdisciplinary Research in Biology (CIRB) - CNRS UMR 7241, INSERM U1050, Labex MemoLife, PSL Research University, Collège de France, Paris F-75005, France
| | - Alain Joliot
- Center for Interdisciplinary Research in Biology (CIRB) - CNRS UMR 7241, INSERM U1050, Labex MemoLife, PSL Research University, Collège de France, Paris F-75005, France
| |
Collapse
|
36
|
Wang S, Wang B, Pan N, Fu L, Wang C, Song G, An J, Liu Z, Zhu W, Guan Y, Xu ZQD, Chan P, Chen Z, Zhang YA. Differentiation of human induced pluripotent stem cells to mature functional Purkinje neurons. Sci Rep 2015; 5:9232. [PMID: 25782665 PMCID: PMC4363833 DOI: 10.1038/srep09232] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2014] [Accepted: 02/24/2015] [Indexed: 12/20/2022] Open
Abstract
It remains a challenge to differentiate human induced pluripotent stem cells (iPSCs) or embryonic stem (ES) cells to Purkinje cells. In this study, we derived iPSCs from human fibroblasts and directed the specification of iPSCs first to Purkinje progenitors, by adding Fgf2 and insulin to the embryoid bodies (EBs) in a time-sensitive manner, which activates the endogenous production of Wnt1 and Fgf8 from EBs that further patterned the cells towards a midbrain-hindbrain-boundary tissue identity. Neph3-positive human Purkinje progenitors were sorted out by using flow cytometry and cultured either alone or with granule cell precursors, in a 2-dimensional or 3-dimensional environment. However, Purkinje progenitors failed to mature further under above conditions. By co-culturing human Purkinje progenitors with rat cerebellar slices, we observed mature Purkinje-like cells with right morphology and marker expression patterns, which yet showed no appropriate membrane properties. Co-culture with human fetal cerebellar slices drove the progenitors to not only morphologically correct but also electrophysiologically functional Purkinje neurons. Neph3-posotive human cells could also survive transplantation into the cerebellum of newborn immunodeficient mice and differentiate to L7- and Calbindin-positive neurons. Obtaining mature human Purkinje cells in vitro has significant implications in studying the mechanisms of spinocerebellar ataxias and other cerebellar diseases.
Collapse
Affiliation(s)
- Shuyan Wang
- 1] Sanofi-Xuanwu Joint Lab for Regenerative Medicine, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China [2] Center of Neural Injury and Repair, Beijing Institute for Brain Disorders, Beijing, China [3] Center of Parkinson's Disease, Beijing Institute for Brain Disorders, Beijing, China
| | - Bin Wang
- Sanofi-Xuanwu Joint Lab for Regenerative Medicine, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China
| | - Na Pan
- Department of Neurobiology, Beijing Key Laboratory of Major Brain Disorders, Beijing Institute of Brain Disorders, Capital Medical University, Beijing, 100069, China
| | - Linlin Fu
- Sanofi-Xuanwu Joint Lab for Regenerative Medicine, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China
| | - Chaodong Wang
- Department of Neurobiology, Xuanwu Hosptial, Capital Medical University, Beijing, 100053, China
| | - Gongru Song
- Sanofi-Xuanwu Joint Lab for Regenerative Medicine, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China
| | - Jing An
- Sanofi-Xuanwu Joint Lab for Regenerative Medicine, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China
| | - Zhongfeng Liu
- Sanofi-Xuanwu Joint Lab for Regenerative Medicine, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China
| | - Wanwan Zhu
- Sanofi-Xuanwu Joint Lab for Regenerative Medicine, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China
| | - Yunqian Guan
- Sanofi-Xuanwu Joint Lab for Regenerative Medicine, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China
| | - Zhi-Qing David Xu
- Department of Neurobiology, Beijing Key Laboratory of Major Brain Disorders, Beijing Institute of Brain Disorders, Capital Medical University, Beijing, 100069, China
| | - Piu Chan
- 1] Center of Parkinson's Disease, Beijing Institute for Brain Disorders, Beijing, China [2] Department of Neurobiology, Xuanwu Hosptial, Capital Medical University, Beijing, 100053, China
| | - Zhiguo Chen
- 1] Sanofi-Xuanwu Joint Lab for Regenerative Medicine, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China [2] Center of Neural Injury and Repair, Beijing Institute for Brain Disorders, Beijing, China [3] Center of Parkinson's Disease, Beijing Institute for Brain Disorders, Beijing, China
| | - Y Alex Zhang
- 1] Sanofi-Xuanwu Joint Lab for Regenerative Medicine, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China [2] China R&D Center, Sanofi, Beijing 100022, China
| |
Collapse
|
37
|
Hattapoğlu S, Hamidi C, Göya C, Çetinçakmak MG, Teke M, Ekici F. A Surprising Case: A Supernumerary Heterotopic Hemicerebellum. Clin Neuroradiol 2015; 25:431-4. [PMID: 25622771 DOI: 10.1007/s00062-015-0371-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2014] [Accepted: 01/08/2015] [Indexed: 11/24/2022]
Affiliation(s)
- S Hattapoğlu
- Department of Radiology, Medical School, Dicle University, 21280, Sur/Diyarbakir, Turkey.
| | - C Hamidi
- Department of Radiology, Medical School, Dicle University, 21280, Sur/Diyarbakir, Turkey
| | - C Göya
- Department of Radiology, Medical School, Dicle University, 21280, Sur/Diyarbakir, Turkey
| | - M G Çetinçakmak
- Department of Radiology, Medical School, Dicle University, 21280, Sur/Diyarbakir, Turkey
| | - M Teke
- Department of Radiology, Medical School, Dicle University, 21280, Sur/Diyarbakir, Turkey
| | - F Ekici
- Department of Radiology, Medical School, Dicle University, 21280, Sur/Diyarbakir, Turkey
| |
Collapse
|
38
|
Hippocampal dysregulation of neurofibromin-dependent pathways is associated with impaired spatial learning in engrailed 2 knock-out mice. J Neurosci 2015; 34:13281-8. [PMID: 25274808 DOI: 10.1523/jneurosci.2894-13.2014] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Genome-wide association studies indicated the homeobox-containing transcription factor Engrailed-2 (En2) as a candidate gene for autism spectrum disorders (ASD). Accordingly, En2 knock-out (En2(-/-)) mice show anatomical and behavioral "ASD-like" features, including decreased sociability and learning deficits. The molecular pathways underlying these deficits in En2(-/-) mice are not known. Deficits in signaling pathways involving neurofibromin and extracellular-regulated kinase (ERK) have been associated with impaired learning. Here we investigated the neurofibromin-ERK cascade in the hippocampus of wild-type (WT) and En2(-/-) mice before and after spatial learning testing. When compared with WT littermates, En2(-/-) mice showed impaired performance in the Morris water maze (MWM), which was accompanied by lower expression of the activity-dependent gene Arc. Quantitative RT-PCR, immunoblotting, and immunohistochemistry experiments showed a marked downregulation of neurofibromin expression in the dentate gyrus of both naive and MWM-treated En2(-/-) mice. ERK phosphorylation, known to be induced in the presence of neurofibromin deficiency, was increased in the dentate gyrus of En2(-/-) mice after MWM. Treatment of En2(-/-) mice with lovastatin, an indirect inhibitor of ERK phosphorylation, markedly reduced ERK phosphorylation in the dentate gyrus, but was unable to rescue learning deficits in MWM-trained mutant mice. Further investigation is needed to unravel the complex molecular mechanisms linking dysregulation of neurofibromin-dependent pathways to spatial learning deficits in the En2 mouse model of ASD.
Collapse
|
39
|
Engrailed homeoproteins in visual system development. Cell Mol Life Sci 2014; 72:1433-45. [PMID: 25432704 PMCID: PMC4366559 DOI: 10.1007/s00018-014-1776-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2014] [Revised: 10/31/2014] [Accepted: 11/06/2014] [Indexed: 12/28/2022]
Abstract
Engrailed is a homeoprotein transcription factor. This family of transcription factors is characterized by their DNA-binding homeodomain and some members, including Engrailed, can transfer between cells and regulate protein translation in addition to gene transcription. Engrailed is intimately involved in the development of the vertebrate visual system. Early expression of Engrailed in dorsal mesencephalon contributes to the development and organization of a visual structure, the optic tectum/superior colliculus. This structure is an important target for retinal ganglion cell axons that carry visual information from the retina. Engrailed regulates the expression of Ephrin axon guidance cues in the tectum/superior colliculus. More recently it has been reported that Engrailed itself acts as an axon guidance cue in synergy with the Ephrin system and is proposed to enhance retinal topographic precision.
Collapse
|
40
|
Reduced phosphorylation of synapsin I in the hippocampus of Engrailed-2 knockout mice, a model for autism spectrum disorders. Neuroscience 2014; 286:122-30. [PMID: 25463523 DOI: 10.1016/j.neuroscience.2014.11.041] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2014] [Revised: 11/11/2014] [Accepted: 11/13/2014] [Indexed: 12/13/2022]
Abstract
Mice lacking the homeodomain transcription factor Engrailed-2 (En2(-/-) mice) are a well-characterized model for autism spectrum disorders (ASD). En2(-/-) mice present molecular, neuropathological and behavioral deficits related to ASD, including down-regulation of ASD-associated genes, cerebellar hypoplasia, interneuron loss, enhanced seizure susceptibility, decreased sociability and impaired cognition. Specifically, impaired spatial learning in the Morris water maze (MWM) is associated with reduced expression of neurofibromin and increased phosphorylation of extracellular-regulated kinase (ERK) in the hippocampus of En2(-/-) adult mice. In the attempt to better understand the molecular cascades underlying neurofibromin-dependent cognitive deficits in En2 mutant mice, we investigated the expression and phosphorylation of synapsin I (SynI; a major target of neurofibromin-dependent signaling) in the hippocampus of wild-type (WT) and En2(-/-) mice before and after MWM. Here we show that SynI mRNA and protein levels are down-regulated in the hippocampus of naïve and MWM-treated En2(-/-) mice, as compared to WT controls. This down-regulation is paralleled by reduced levels of SynI phosphorylation at Ser549 and Ser553 residues in the hilus of mutant mice, before and after MWM. These data indicate that in En2(-/-) hippocampus, neurofibromin-dependent pathways converging on SynI phosphorylation might underlie hippocampal-dependent learning deficits observed in En2(-/-) mice.
Collapse
|
41
|
Stettler O, Moya KL. Distinct roles of homeoproteins in brain topographic mapping and in neural circuit formation. Semin Cell Dev Biol 2014; 35:165-72. [PMID: 25042849 DOI: 10.1016/j.semcdb.2014.07.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2014] [Accepted: 07/07/2014] [Indexed: 01/02/2023]
Abstract
The construction of the brain is a highly regulated process, requiring coordination of various cellular and molecular mechanisms that together ensure the stability of the cerebrum architecture and functions. The mature brain is an organ that performs complex computational operations using specific sensory information from the outside world and this requires precise organization within sensory networks and a separation of sensory modalities during development. We review here the role of homeoproteins in the arealization of the brain according to sensorimotor functions, the micropartition of its cytoarchitecture, and the maturation of its sensory circuitry. One of the most interesting observation about homeoproteins in recent years concerns their ability to act both in a cell-autonomous and non-cell-autonomous manner. The highlights in the present review collectively show how these two modes of action of homeoproteins confer various functions in shaping cortical maps.
Collapse
Affiliation(s)
- Olivier Stettler
- Laboratoire CRRET EAC 7149, Université Paris-Est Créteil, 61, Av. du Général de Gaulle, 94010 Créteil Cedex, France.
| | - Kenneth L Moya
- Collège de France, Center for Interdisciplinary Research in Biology, UMR CNRS 7241/INSERM U1050, 11 place Marcelin Berthelot, 75005 Paris, France; Labex Memolife, PSL Research University, France
| |
Collapse
|
42
|
Green MJ, Myat AM, Emmenegger BA, Wechsler-Reya RJ, Wilson LJ, Wingate RJT. Independently specified Atoh1 domains define novel developmental compartments in rhombomere 1. Development 2014; 141:389-98. [PMID: 24381197 PMCID: PMC3879817 DOI: 10.1242/dev.099119] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
The rhombic lip gives rise to neuronal populations that contribute to cerebellar, proprioceptive and interoceptive networks. Cell production depends on the expression of the basic helix-loop-helix (bHLH) transcription factor Atoh1. In rhombomere 1, Atoh1-positive cells give rise to both cerebellar neurons and extra-cerebellar nuclei in ventral hindbrain. The origin of this cellular diversity has previously been attributed to temporal signals rather than spatial patterning. Here, we show that in both chick and mouse the cerebellar Atoh1 precursor pool is partitioned into initially cryptic spatial domains that reflect the activity of two different organisers: an isthmic Atoh1 domain, which gives rise to isthmic nuclei, and the rhombic lip, which generates deep cerebellar nuclei and granule cells. We use a combination of in vitro explant culture, genetic fate mapping and gene overexpression and knockdown to explore the role of isthmic signalling in patterning these domains. We show that an FGF-dependent isthmic Atoh1 domain is the origin of distinct populations of Lhx9-positive neurons in the extra-cerebellar isthmic nuclei. In the cerebellum, ectopic FGF induces proliferation while blockade reduces the length of the cerebellar rhombic lip. FGF signalling is not required for the specification of cerebellar cell types from the rhombic lip and its upregulation inhibits their production. This suggests that although the isthmus regulates the size of the cerebellar anlage, the downregulation of isthmic FGF signals is required for induction of rhombic lip-derived cerebellar neurons.
Collapse
Affiliation(s)
- Mary J Green
- MRC Centre for Developmental Neurobiology, King's College London, 4th floor New Hunt's House, London SE1 1UL, UK
| | | | | | | | | | | |
Collapse
|
43
|
Rossman IT, Lin L, Morgan KM, Digiovine M, Van Buskirk EK, Kamdar S, Millonig JH, Dicicco-Bloom E. Engrailed2 modulates cerebellar granule neuron precursor proliferation, differentiation and insulin-like growth factor 1 signaling during postnatal development. Mol Autism 2014; 5:9. [PMID: 24507165 PMCID: PMC3932947 DOI: 10.1186/2040-2392-5-9] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2013] [Accepted: 01/14/2014] [Indexed: 01/16/2023] Open
Abstract
Background The homeobox transcription factor Engrailed2 (En2) has been studied extensively in neurodevelopment, particularly in the midbrain/hindbrain region and cerebellum, where it exhibits dynamic patterns of expression and regulates cell patterning and morphogenesis. Because of its roles in regulating cerebellar development and evidence of cerebellar pathology in autism spectrum disorder (ASD), we previously examined an ENGRAILED2 association and found evidence to support EN2 as a susceptibility gene, a finding replicated by several other investigators. However, its functions at the cell biological level remain undefined. In the mouse, En2 gene is expressed in granule neuron precursors (GNPs) just as they exit the cell cycle and begin to differentiate, raising the possibility that En2 may modulate these developmental processes. Methods To define En2 functions, we examined proliferation, differentiation and signaling pathway activation in En2 knockout (KO) and wild-type (WT) GNPs in response to a variety of extracellular growth factors and following En2 cDNA overexpression in cell culture. In vivo analyses of cerebellar GNP proliferation as well as responses to insulin-like growth factor-1 (IGF1) treatment were also conducted. Results Proliferation markers were increased in KO GNPs in vivo and in 24-h cultures, suggesting En2 normally serves to promote cell cycle exit. Significantly, IGF1 stimulated greater DNA synthesis in KO than WT cells in culture, a finding associated with markedly increased phospho-S6 kinase activation. Similarly, there was three-fold greater DNA synthesis in the KO cerebellum in response to IGF1 in vivo. On the other hand, KO GNPs exhibited reduced neurite outgrowth and differentiation. Conversely, En2 overexpression increased cell cycle exit and promoted neuronal differentiation. Conclusions In aggregate, our observations suggest that the ASD-associated gene En2 promotes GNP cell cycle exit and differentiation, and modulates IGF1 activity during postnatal cerebellar development. Thus, genetic/epigenetic alterations of EN2 expression may impact proliferation, differentiation and IGF1 signaling as possible mechanisms that may contribute to ASD pathogenesis.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Emanuel Dicicco-Bloom
- Department of Neuroscience & Cell Biology, Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, 675 Hoes, Lane, Piscataway, NJ 08854, USA.
| |
Collapse
|
44
|
Brielmaier J, Senerth JM, Silverman JL, Matteson PG, Millonig JH, DiCicco-Bloom E, Crawley JN. Chronic desipramine treatment rescues depression-related, social and cognitive deficits inEngrailed-2knockout mice. GENES BRAIN AND BEHAVIOR 2014; 13:286-298. [DOI: 10.1111/gbb.12115] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Affiliation(s)
| | | | | | | | | | - E. DiCicco-Bloom
- Department of Pediatrics; University of Medicine and Dentistry of New Jersey (UMDNJ)-Robert Wood Johnson Medical School; New Brunswick NJ USA
| | | |
Collapse
|
45
|
Sung PL, Cheng EE, Chen YJ, Chern SR, Shih CY, Chang CM, Wang PH, Yen MS, Huang CYF, Chen CP. Prenatal diagnosis of de novo monosomy 7q33-qter associated with hydrops fetalis, semilobar holoprosencephaly, and premaxillary dysgenesis. Taiwan J Obstet Gynecol 2014; 52:602-6. [PMID: 24411055 DOI: 10.1016/j.tjog.2013.10.030] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2013] [Accepted: 05/04/2013] [Indexed: 11/16/2022] Open
Affiliation(s)
- Pi-Lin Sung
- Department of Obstetrics and Gynecology, Taipei Veterans General Hospital, Taipei, Taiwan; Department of Obstetrics and Gynecology, School of Medicine, National Yang-Ming University School of Medicine, Taipei, Taiwan; Institute of Clinical Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Eong-Eong Cheng
- Department of Obstetrics and Gynecology, Taipei Veterans General Hospital, Taipei, Taiwan; Department of Obstetrics and Gynecology, School of Medicine, National Yang-Ming University School of Medicine, Taipei, Taiwan
| | - Yann-Jang Chen
- Department of Life Sciences and Institute of Genome Sciences, National Yang-Ming University, Taipei, Taiwan
| | - Schu-Rern Chern
- Department of Medical Research, Mackay Memorial Hospital, Taipei, Taiwan
| | - Chung-Yu Shih
- Department of Obstetrics and Gynecology, Taipei Veterans General Hospital, Taipei, Taiwan; Department of Obstetrics and Gynecology, School of Medicine, National Yang-Ming University School of Medicine, Taipei, Taiwan
| | - Chia-Ming Chang
- Department of Obstetrics and Gynecology, Taipei Veterans General Hospital, Taipei, Taiwan; Department of Obstetrics and Gynecology, School of Medicine, National Yang-Ming University School of Medicine, Taipei, Taiwan
| | - Peng-Hui Wang
- Department of Obstetrics and Gynecology, Taipei Veterans General Hospital, Taipei, Taiwan; Department of Obstetrics and Gynecology, School of Medicine, National Yang-Ming University School of Medicine, Taipei, Taiwan
| | - Ming-Shyen Yen
- Department of Obstetrics and Gynecology, Taipei Veterans General Hospital, Taipei, Taiwan; Department of Obstetrics and Gynecology, School of Medicine, National Yang-Ming University School of Medicine, Taipei, Taiwan
| | - Chi-Ying F Huang
- Institute of Clinical Medicine, National Yang-Ming University, Taipei, Taiwan; Institute of Biopharmaceutical Sciences, National Yang-Ming University, Taipei, Taiwan
| | - Chih-Ping Chen
- Department of Obstetrics and Gynecology, School of Medicine, National Yang-Ming University School of Medicine, Taipei, Taiwan; Department of Medical Research, Mackay Memorial Hospital, Taipei, Taiwan; Department of Biotechnology, Asia University, Taipei, Taiwan; School of Chinese Medicine, College of Chinese Medicine, China Medical University, Taichung, Taiwan; Department of Obstetrics and Gynecology, Mackay Memorial Hospital, Taipei, Taiwan; Institute of Clinical and Community Health Nursing, National Yang-Ming University, Taipei, Taiwan.
| |
Collapse
|
46
|
Sgadò P, Provenzano G, Dassi E, Adami V, Zunino G, Genovesi S, Casarosa S, Bozzi Y. Transcriptome profiling in engrailed-2 mutant mice reveals common molecular pathways associated with autism spectrum disorders. Mol Autism 2013; 4:51. [PMID: 24355397 PMCID: PMC3896729 DOI: 10.1186/2040-2392-4-51] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2013] [Accepted: 11/27/2013] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND Transcriptome analysis has been used in autism spectrum disorder (ASD) to unravel common pathogenic pathways based on the assumption that distinct rare genetic variants or epigenetic modifications affect common biological pathways. To unravel recurrent ASD-related neuropathological mechanisms, we took advantage of the En2-/- mouse model and performed transcriptome profiling on cerebellar and hippocampal adult tissues. METHODS Cerebellar and hippocampal tissue samples from three En2-/- and wild type (WT) littermate mice were assessed for differential gene expression using microarray hybridization followed by RankProd analysis. To identify functional categories overrepresented in the differentially expressed genes, we used integrated gene-network analysis, gene ontology enrichment and mouse phenotype ontology analysis. Furthermore, we performed direct enrichment analysis of ASD-associated genes from the SFARI repository in our differentially expressed genes. RESULTS Given the limited number of animals used in the study, we used permissive criteria and identified 842 differentially expressed genes in En2-/- cerebellum and 862 in the En2-/- hippocampus. Our functional analysis revealed that the molecular signature of En2-/- cerebellum and hippocampus shares convergent pathological pathways with ASD, including abnormal synaptic transmission, altered developmental processes and increased immune response. Furthermore, when directly compared to the repository of the SFARI database, our differentially expressed genes in the hippocampus showed enrichment of ASD-associated genes significantly higher than previously reported. qPCR was performed for representative genes to confirm relative transcript levels compared to those detected in microarrays. CONCLUSIONS Despite the limited number of animals used in the study, our bioinformatic analysis indicates the En2-/- mouse is a valuable tool for investigating molecular alterations related to ASD.
Collapse
Affiliation(s)
- Paola Sgadò
- Laboratory of Molecular Neuropathology, Centre for Integrative Biology (CIBIO), University of Trento, Via delle Regole 101, 38123 Trento, Italy
| | - Giovanni Provenzano
- Laboratory of Molecular Neuropathology, Centre for Integrative Biology (CIBIO), University of Trento, Via delle Regole 101, 38123 Trento, Italy
| | - Erik Dassi
- Laboratory of Translational Genomics, Centre for Integrative Biology (CIBIO), University of Trento, Via delle Regole 101, 38123 Trento, Italy
| | - Valentina Adami
- High Throughput Screening Core Facility, Centre for Integrative Biology (CIBIO), University of Trento, Via delle Regole 101, 38123 Trento, Italy
| | - Giulia Zunino
- Laboratory of Molecular Neuropathology, Centre for Integrative Biology (CIBIO), University of Trento, Via delle Regole 101, 38123 Trento, Italy
| | - Sacha Genovesi
- Laboratory of Molecular Neuropathology, Centre for Integrative Biology (CIBIO), University of Trento, Via delle Regole 101, 38123 Trento, Italy
| | - Simona Casarosa
- Laboratory of Developmental Neurobiology, Centre for Integrative Biology (CIBIO), University of Trento, Via delle Regole 101, 38123 Trento, Italy.,C.N.R. Neuroscience Institute, via G. Moruzzi 1, 56124 Pisa, Italy
| | - Yuri Bozzi
- Laboratory of Molecular Neuropathology, Centre for Integrative Biology (CIBIO), University of Trento, Via delle Regole 101, 38123 Trento, Italy.,C.N.R. Neuroscience Institute, via G. Moruzzi 1, 56124 Pisa, Italy
| |
Collapse
|
47
|
Affiliation(s)
| | - Richard Hawkes
- Department of Cell Biology and Anatomy, Genes and Development Research Group and Hotchkiss Brain Institute, Faculty of Medicine, University of Calgary
| |
Collapse
|
48
|
Won H, Mah W, Kim E. Autism spectrum disorder causes, mechanisms, and treatments: focus on neuronal synapses. Front Mol Neurosci 2013; 6:19. [PMID: 23935565 PMCID: PMC3733014 DOI: 10.3389/fnmol.2013.00019] [Citation(s) in RCA: 129] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2013] [Accepted: 07/16/2013] [Indexed: 12/24/2022] Open
Abstract
Autism spectrum disorder (ASD) is a group of developmental disabilities characterized by impairments in social interaction and communication and restricted and repetitive interests/behaviors. Advances in human genomics have identified a large number of genetic variations associated with ASD. These associations are being rapidly verified by a growing number of studies using a variety of approaches, including mouse genetics. These studies have also identified key mechanisms underlying the pathogenesis of ASD, many of which involve synaptic dysfunctions, and have investigated novel, mechanism-based therapeutic strategies. This review will try to integrate these three key aspects of ASD research: human genetics, animal models, and potential treatments. Continued efforts in this direction should ultimately reveal core mechanisms that account for a larger fraction of ASD cases and identify neural mechanisms associated with specific ASD symptoms, providing important clues to efficient ASD treatment.
Collapse
Affiliation(s)
- Hyejung Won
- Department of Biological Sciences, Korea Advanced Institute of Science and TechnologyDaejeon, South Korea
| | - Won Mah
- Department of Biological Sciences, Korea Advanced Institute of Science and TechnologyDaejeon, South Korea
- Center for Synaptic Brain Dysfunctions, Institute for Basic ScienceDaejeon, South Korea
| | - Eunjoon Kim
- Department of Biological Sciences, Korea Advanced Institute of Science and TechnologyDaejeon, South Korea
- Center for Synaptic Brain Dysfunctions, Institute for Basic ScienceDaejeon, South Korea
| |
Collapse
|
49
|
Martinez S, Andreu A, Mecklenburg N, Echevarria D. Cellular and molecular basis of cerebellar development. Front Neuroanat 2013; 7:18. [PMID: 23805080 PMCID: PMC3693072 DOI: 10.3389/fnana.2013.00018] [Citation(s) in RCA: 79] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2013] [Accepted: 06/03/2013] [Indexed: 01/14/2023] Open
Abstract
Historically, the molecular and cellular mechanisms of cerebellar development were investigated through structural descriptions and studying spontaneous mutations in animal models and humans. Advances in experimental embryology, genetic engineering, and neuroimaging techniques render today the possibility to approach the analysis of molecular mechanisms underlying histogenesis and morphogenesis of the cerebellum by experimental designs. Several genes and molecules were identified to be involved in the cerebellar plate regionalization, specification, and differentiation of cerebellar neurons, as well as the establishment of cellular migratory routes and the subsequent neuronal connectivity. Indeed, pattern formation of the cerebellum requires the adequate orchestration of both key morphogenetic signals, arising from distinct brain regions, and local expression of specific transcription factors. Thus, the present review wants to revisit and discuss these morphogenetic and molecular mechanisms taking place during cerebellar development in order to understand causal processes regulating cerebellar cytoarchitecture, its highly topographically ordered circuitry and its role in brain function.
Collapse
Affiliation(s)
- Salvador Martinez
- Experimental Embryology Lab, Consejo Superior de Investigaciones Científicas, Instituto de Neurociencias de Alicante, Universidad Miguel Hernandez Alicante, Spain
| | | | | | | |
Collapse
|
50
|
Rogers TD, McKimm E, Dickson PE, Goldowitz D, Blaha CD, Mittleman G. Is autism a disease of the cerebellum? An integration of clinical and pre-clinical research. Front Syst Neurosci 2013; 7:15. [PMID: 23717269 PMCID: PMC3650713 DOI: 10.3389/fnsys.2013.00015] [Citation(s) in RCA: 99] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2012] [Accepted: 04/23/2013] [Indexed: 01/07/2023] Open
Abstract
Autism spectrum disorders are a group of neurodevelopmental disorders characterized by deficits in social skills and communication, stereotyped and repetitive behavior, and a range of deficits in cognitive function. While the etiology of autism is unknown, current research indicates that abnormalities of the cerebellum, now believed to be involved in cognitive function and the prefrontal cortex (PFC), are associated with autism. The current paper proposes that impaired cerebello-cortical circuitry could, at least in part, underlie autistic symptoms. The use of animal models that allow for manipulation of genetic and environmental influences are an effective means of elucidating both distal and proximal etiological factors in autism and their potential impact on cerebello-cortical circuitry. Some existing rodent models of autism, as well as some models not previously applied to the study of the disorder, display cerebellar and behavioral abnormalities that parallel those commonly seen in autistic patients. The novel findings produced from research utilizing rodent models could provide a better understanding of the neurochemical and behavioral impact of changes in cerebello-cortical circuitry in autism.
Collapse
Affiliation(s)
- Tiffany D Rogers
- Department of Psychology, The University of Memphis Memphis, TN, USA
| | | | | | | | | | | |
Collapse
|