1
|
Saad HA, Marzouk M, Abdelrahman H, Moradikor N. Mechanisms underlying stress effects on the brain: Basic concepts and clinical implications. PROGRESS IN BRAIN RESEARCH 2025; 291:21-47. [PMID: 40222781 DOI: 10.1016/bs.pbr.2025.01.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/15/2025]
Abstract
Chronic stress impacts the brain through complex physiological, neurological, and immunological responses. The stress response involves the activation of the sympathetic-adrenal-medullary (SAM) system and the hypothalamic-pituitary-adrenal (HPA) axis, releasing stress hormones like norepinephrine and cortisol. While these responses are adaptive short-term, chronic stress disrupts homeostasis, increasing the risk of cardiovascular diseases, neurodegenerative disorders, and psychiatric conditions such as depression. This dysregulation is linked to persistent neuroinflammation, oxidative stress, and neurotransmitter imbalances involving dopamine and serotonin, impairing neuroplasticity and leading to structural changes in critical brain areas, such as the hippocampus and prefrontal cortex. Moreover, stress affects gene expression, particularly neuroinflammatory pathways, contributing to long-term cognitive function and emotional regulation alterations. Advancements in neuroimaging and molecular techniques, including MRI, PET, and SPECT, hold promise for identifying biomarkers and better understanding stress-induced brain changes. These insights are critical for developing targeted interventions to mitigate the adverse effects of chronic stress on brain health.
Collapse
Affiliation(s)
- Hager Adel Saad
- Faculty of Pharmacy and Biotechnology, German University in Cairo, (GUC), New Cairo, Cairo, Egypt.
| | - Mahmoud Marzouk
- Faculty of Pharmacy and Biotechnology, German University in Cairo, (GUC), New Cairo, Cairo, Egypt
| | - Hla Abdelrahman
- Faculty of Pharmacy and Biotechnology, German University in Cairo, (GUC), New Cairo, Cairo, Egypt
| | - Nasrollah Moradikor
- International Center for Neuroscience Research, Institute for Intelligent Research, Tbilisi, Georgia
| |
Collapse
|
2
|
Paşcalău R, Badea TC. Signaling - transcription interactions in mouse retinal ganglion cells early axon pathfinding -a literature review. FRONTIERS IN OPHTHALMOLOGY 2023; 3:1180142. [PMID: 38983012 PMCID: PMC11182120 DOI: 10.3389/fopht.2023.1180142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/05/2023] [Accepted: 04/21/2023] [Indexed: 07/11/2024]
Abstract
Sending an axon out of the eye and into the target brain nuclei is the defining feature of retinal ganglion cells (RGCs). The literature on RGC axon pathfinding is vast, but it focuses mostly on decision making events such as midline crossing at the optic chiasm or retinotopic mapping at the target nuclei. In comparison, the exit of RGC axons out of the eye is much less explored. The first checkpoint on the RGC axons' path is the optic cup - optic stalk junction (OC-OS). OC-OS development and the exit of the RGC pioneer axons out of the eye are coordinated spatially and temporally. By the time the optic nerve head domain is specified, the optic fissure margins are in contact and the fusion process is ongoing, the first RGCs are born in its proximity and send pioneer axons in the optic stalk. RGC differentiation continues in centrifugal waves. Later born RGC axons fasciculate with the more mature axons. Growth cones at the end of the axons respond to guidance cues to adopt a centripetal direction, maintain nerve fiber layer restriction and to leave the optic cup. Although there is extensive information on OC-OS development, we still have important unanswered questions regarding its contribution to the exit of the RGC axons out of the eye. We are still to distinguish the morphogens of the OC-OS from the axon guidance molecules which are expressed in the same place at the same time. The early RGC transcription programs responsible for axon emergence and pathfinding are also unknown. This review summarizes the molecular mechanisms for early RGC axon guidance by contextualizing mouse knock-out studies on OC-OS development with the recent transcriptomic studies on developing RGCs in an attempt to contribute to the understanding of human optic nerve developmental anomalies. The published data summarized here suggests that the developing optic nerve head provides a physical channel (the closing optic fissure) as well as molecular guidance cues for the pioneer RGC axons to exit the eye.
Collapse
Affiliation(s)
- Raluca Paşcalău
- Research and Development Institute, Transilvania University of Braşov, Braşov, Romania
- Ophthalmology Clinic, Cluj County Emergency Hospital, Cluj-Napoca, Romania
| | - Tudor Constantin Badea
- Research and Development Institute, Transilvania University of Braşov, Braşov, Romania
- National Center for Brain Research, Institutul de Cercetări pentru Inteligență Artificială, Romanian Academy, Bucharest, Romania
| |
Collapse
|
3
|
Gotoh H, Maruyama K, Yoshii K, Yamauchi N, Nomura T, Ohtsuka S, Shirasaki R, Takebayashi H, Ono K. Disruption of the anterior commissure in Olig2 deficient mice. Eur J Neurosci 2023; 57:5-16. [PMID: 36370145 DOI: 10.1111/ejn.15861] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 10/25/2022] [Accepted: 10/28/2022] [Indexed: 11/15/2022]
Abstract
In the present study, we examined neural circuit formation in the forebrain of the Olig2 knockout (Olig2-KO) mouse model and found disruption of the anterior commissure at the late foetal stage. Axon bundles of the anterior commissure encountered the wall of the third ventricle and ceased axonal extension. L1-CAM immunohistochemistry showed that Olig2-KO mice lose decussation formation in the basal forebrain. DiI tracing revealed that the thin bundles of the anterior commissure axons crossed the midline but ceased further extension into the deep part of the contralateral side. Furthermore, some fractions of DiI-labelled axons were oriented dorsolaterally, which was not observed in the control mouse forebrain. The rostral part of the third ventricle was much wider in the Olig2-KO mice than in wild-type mice, which likely resulted in the delay of midline fusion and subsequent delay and malformation of the anterior commissure. We analysed gene expression alterations in the Olig2-KO mice using a public database and found multiple genes, which are related to axon guidance and epithelial-mesenchymal transition, showing subtle expression changes. These results suggest that Olig2 is essential for anterior commissure formation, likely by regulating multiple biological processes.
Collapse
Affiliation(s)
- Hitoshi Gotoh
- Developmental Neurobiology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Kohei Maruyama
- Developmental Neurobiology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Kengo Yoshii
- Mathematics and Statistics in Medical Sciences, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Nao Yamauchi
- Developmental Neurobiology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan.,Okayama University Medical School, Okayama, Japan
| | - Tadashi Nomura
- Developmental Neurobiology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Satoshi Ohtsuka
- Laboratory for Experimental Animals, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Ryuichi Shirasaki
- Developmental Neurobiology Group, Graduate School of Frontier Biosciences Osaka University, Osaka, Japan
| | - Hirohide Takebayashi
- Division of Neurobiology and Anatomy, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
| | - Katsuhiko Ono
- Developmental Neurobiology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| |
Collapse
|
4
|
Medina-Cano D, Corrigan EK, Glenn RA, Islam MT, Lin Y, Kim J, Cho H, Vierbuchen T. Rapid and robust directed differentiation of mouse epiblast stem cells into definitive endoderm and forebrain organoids. Development 2022; 149:dev200561. [PMID: 35899604 PMCID: PMC10655922 DOI: 10.1242/dev.200561] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Accepted: 07/04/2022] [Indexed: 11/20/2022]
Abstract
Directed differentiation of pluripotent stem cells (PSCs) is a powerful model system for deconstructing embryonic development. Although mice are the most advanced mammalian model system for genetic studies of embryonic development, state-of-the-art protocols for directed differentiation of mouse PSCs into defined lineages require additional steps and generates target cell types with lower purity than analogous protocols for human PSCs, limiting their application as models for mechanistic studies of development. Here, we examine the potential of mouse epiblast stem cells cultured in media containing Wnt pathway inhibitors as a starting point for directed differentiation. As a proof of concept, we focused our efforts on two specific cell/tissue types that have proven difficult to generate efficiently and reproducibly from mouse embryonic stem cells: definitive endoderm and neural organoids. We present new protocols for rapid generation of nearly pure definitive endoderm and forebrain-patterned neural organoids that model the development of prethalamic and hippocampal neurons. These differentiation models present new possibilities for combining mouse genetic tools with in vitro differentiation to characterize molecular and cellular mechanisms of embryonic development.
Collapse
Affiliation(s)
- Daniel Medina-Cano
- Developmental Biology Program, Sloan Kettering Institute for Cancer Research, New York, NY 10065, USA
- Center for Stem Cell Biology, Sloan Kettering Institute for Cancer Research, New York, NY 10065, USA
| | - Emily K. Corrigan
- Developmental Biology Program, Sloan Kettering Institute for Cancer Research, New York, NY 10065, USA
- Center for Stem Cell Biology, Sloan Kettering Institute for Cancer Research, New York, NY 10065, USA
| | - Rachel A. Glenn
- Developmental Biology Program, Sloan Kettering Institute for Cancer Research, New York, NY 10065, USA
- Center for Stem Cell Biology, Sloan Kettering Institute for Cancer Research, New York, NY 10065, USA
- Cell and Developmental Biology Program, Weill Cornell Graduate School of Medical Sciences, Cornell University, New York, NY 10065, USA
| | - Mohammed T. Islam
- Developmental Biology Program, Sloan Kettering Institute for Cancer Research, New York, NY 10065, USA
- Center for Stem Cell Biology, Sloan Kettering Institute for Cancer Research, New York, NY 10065, USA
| | - Yuan Lin
- Developmental Biology Program, Sloan Kettering Institute for Cancer Research, New York, NY 10065, USA
- Center for Stem Cell Biology, Sloan Kettering Institute for Cancer Research, New York, NY 10065, USA
| | - Juliet Kim
- Developmental Biology Program, Sloan Kettering Institute for Cancer Research, New York, NY 10065, USA
- Center for Stem Cell Biology, Sloan Kettering Institute for Cancer Research, New York, NY 10065, USA
| | - Hyunwoo Cho
- Developmental Biology Program, Sloan Kettering Institute for Cancer Research, New York, NY 10065, USA
- Center for Stem Cell Biology, Sloan Kettering Institute for Cancer Research, New York, NY 10065, USA
| | - Thomas Vierbuchen
- Developmental Biology Program, Sloan Kettering Institute for Cancer Research, New York, NY 10065, USA
- Center for Stem Cell Biology, Sloan Kettering Institute for Cancer Research, New York, NY 10065, USA
| |
Collapse
|
5
|
Cunningham JG, Scripter JD, Nti SA, Tucker ES. Early construction of the thalamocortical axon pathway requires c-Jun N-terminal kinase signaling within the ventral forebrain. Dev Dyn 2022; 251:459-480. [PMID: 34494344 PMCID: PMC8891049 DOI: 10.1002/dvdy.416] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2021] [Revised: 09/03/2021] [Accepted: 09/03/2021] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND Thalamocortical connectivity is essential for normal brain function. This important pathway is established during development, when thalamic axons extend a long distance through the forebrain before reaching the cerebral cortex. In this study, we identify a novel role for the c-Jun N-terminal kinase (JNK) signaling pathway in guiding thalamocortical axons through intermediate target territories. RESULTS Complete genetic removal of JNK signaling from the Distal-less 5/6 (Dlx5/6) domain in mice prevents thalamocortical axons from crossing the diencephalon-telencephalon boundary (DTB) and the internal capsule fails to form. Ventral telencephalic cells critical for thalamocortical axon extensions including corridor and guidepost neurons are also disrupted. In addition, corticothalamic, striatonigral, and nigrostriatal axons fail to cross the DTB. Analyses of different JNK mutants demonstrate that thalamocortical axon pathfinding has a non-autonomous requirement for JNK signaling. CONCLUSIONS We conclude that JNK signaling within the Dlx5/6 territory enables the construction of major axonal pathways in the developing forebrain. Further exploration of this intermediate axon guidance territory is needed to uncover mechanisms of axonal pathfinding during normal brain development and to elucidate how this vital process may be compromised in neurodevelopmental disorders.
Collapse
Affiliation(s)
- Jessica G. Cunningham
- Department of Neuroscience, West Virginia University School of Medicine, Morgantown, WV 26506,Neuroscience Graduate Program, West Virginia University School of Medicine, Morgantown, WV 26506,Rockefeller Neuroscience Institute, West Virginia University School of Medicine, Morgantown, WV 26506
| | - James D. Scripter
- Department of Neuroscience, West Virginia University School of Medicine, Morgantown, WV 26506,Neuroscience Graduate Program, West Virginia University School of Medicine, Morgantown, WV 26506,Rockefeller Neuroscience Institute, West Virginia University School of Medicine, Morgantown, WV 26506
| | - Stephany A. Nti
- Department of Neuroscience, West Virginia University School of Medicine, Morgantown, WV 26506,Rockefeller Neuroscience Institute, West Virginia University School of Medicine, Morgantown, WV 26506
| | - Eric S. Tucker
- Department of Neuroscience, West Virginia University School of Medicine, Morgantown, WV 26506,Rockefeller Neuroscience Institute, West Virginia University School of Medicine, Morgantown, WV 26506
| |
Collapse
|
6
|
Qin J, Wang M, Zhao T, Xiao X, Li X, Yang J, Yi L, Goffinet AM, Qu Y, Zhou L. Early Forebrain Neurons and Scaffold Fibers in Human Embryos. Cereb Cortex 2021; 30:913-928. [PMID: 31298263 DOI: 10.1093/cercor/bhz136] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Revised: 05/21/2019] [Accepted: 05/31/2019] [Indexed: 12/24/2022] Open
Abstract
Neural progenitor proliferation, neuronal migration, areal organization, and pioneer axon wiring are critical events during early forebrain development, yet remain incompletely understood, especially in human. Here, we studied forebrain development in human embryos aged 5 to 8 postconceptional weeks (WPC5-8), stages that correspond to the neuroepithelium/early marginal zone (WPC5), telencephalic preplate (WPC6 & 7), and incipient cortical plate (WPC8). We show that early telencephalic neurons are formed at the neuroepithelial stage; the most precocious ones originate from local telencephalic neuroepithelium and possibly from the olfactory placode. At the preplate stage, forebrain organization is quite similar in human and mouse in terms of areal organization and of differentiation of Cajal-Retzius cells, pioneer neurons, and axons. Like in mice, axons from pioneer neurons in prethalamus, ventral telencephalon, and cortical preplate cross the diencephalon-telencephalon junction and the pallial-subpallial boundary, forming scaffolds that could guide thalamic and cortical axons at later stages. In accord with this model, at the early cortical plate stage, corticofugal axons run in ventral telencephalon in close contact with scaffold neurons, which express CELSR3 and FZD3, two molecules that regulates formation of similar scaffolds in mice.
Collapse
Affiliation(s)
- Jingwen Qin
- Guangdong-Hongkong-Macau Institute of CNS Regeneration, Ministry of Education CNS Regeneration Collaborative Joint Laboratory Jinan University Guangzhou, P R China
| | - Meizhi Wang
- Guangdong-Hongkong-Macau Institute of CNS Regeneration, Ministry of Education CNS Regeneration Collaborative Joint Laboratory Jinan University Guangzhou, P R China
| | - Tianyun Zhao
- Department of Anesthesiology, Guangzhou Women and Children's Medical Center Guangzhou Medical University Guangzhou, P R China
| | - Xue Xiao
- Guangdong-Hongkong-Macau Institute of CNS Regeneration, Ministry of Education CNS Regeneration Collaborative Joint Laboratory Jinan University Guangzhou, P R China
| | - Xuejun Li
- Guangdong-Hongkong-Macau Institute of CNS Regeneration, Ministry of Education CNS Regeneration Collaborative Joint Laboratory Jinan University Guangzhou, P R China
| | - Jieping Yang
- Department of Obstetrics and Gynecology, Guangzhou Women and Children's Medical Center Guangzhou Medical University Guangzhou, P R China
| | - Lisha Yi
- Department of Obstetrics and Gynecology, Guangzhou Women and Children's Medical Center Guangzhou Medical University Guangzhou, P R China
| | - Andre M Goffinet
- Guangdong-Hongkong-Macau Institute of CNS Regeneration, Ministry of Education CNS Regeneration Collaborative Joint Laboratory Jinan University Guangzhou, P R China
| | - Yibo Qu
- Guangdong-Hongkong-Macau Institute of CNS Regeneration, Ministry of Education CNS Regeneration Collaborative Joint Laboratory Jinan University Guangzhou, P R China.,Guangzhou Regenerative Medicine and Health Guangdong Laboratory Guangzhou, P R China
| | - Libing Zhou
- Guangdong-Hongkong-Macau Institute of CNS Regeneration, Ministry of Education CNS Regeneration Collaborative Joint Laboratory Jinan University Guangzhou, P R China.,Guangzhou Regenerative Medicine and Health Guangdong Laboratory Guangzhou, P R China.,Key Laboratory of Neuroscience, School of Basic Medical Sciences; Institute of Neuroscience, The Second Affiliated Hospital Guangzhou Medical University Guangzhou, P R China
| |
Collapse
|
7
|
Wojtowicz WM, Vielmetter J, Fernandes RA, Siepe DH, Eastman CL, Chisholm GB, Cox S, Klock H, Anderson PW, Rue SM, Miller JJ, Glaser SM, Bragstad ML, Vance J, Lam AW, Lesley SA, Zinn K, Garcia KC. A Human IgSF Cell-Surface Interactome Reveals a Complex Network of Protein-Protein Interactions. Cell 2021; 182:1027-1043.e17. [PMID: 32822567 PMCID: PMC7440162 DOI: 10.1016/j.cell.2020.07.025] [Citation(s) in RCA: 60] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Revised: 05/19/2020] [Accepted: 07/17/2020] [Indexed: 12/17/2022]
Abstract
Cell-surface protein-protein interactions (PPIs) mediate cell-cell communication, recognition, and responses. We executed an interactome screen of 564 human cell-surface and secreted proteins, most of which are immunoglobulin superfamily (IgSF) proteins, using a high-throughput, automated ELISA-based screening platform employing a pooled-protein strategy to test all 318,096 PPI combinations. Screen results, augmented by phylogenetic homology analysis, revealed ∼380 previously unreported PPIs. We validated a subset using surface plasmon resonance and cell binding assays. Observed PPIs reveal a large and complex network of interactions both within and across biological systems. We identified new PPIs for receptors with well-characterized ligands and binding partners for “orphan” receptors. New PPIs include proteins expressed on multiple cell types and involved in diverse processes including immune and nervous system development and function, differentiation/proliferation, metabolism, vascularization, and reproduction. These PPIs provide a resource for further biological investigation into their functional relevance and may offer new therapeutic drug targets. Human IgSF interactome reveals complex network of cell-surface protein interactions Phylogenetic homology analysis predicts protein-protein interactions ∼380 previously unknown protein-protein interactions identified Deorphanization of receptors and new binding partners for well-studied receptors
Collapse
Affiliation(s)
- Woj M Wojtowicz
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA 94305, USA.
| | - Jost Vielmetter
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - Ricardo A Fernandes
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Dirk H Siepe
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA 94305, USA; Howard Hughes Medical Institute, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Catharine L Eastman
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Gregory B Chisholm
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - Sarah Cox
- The Genomics Institute of the Novartis Research Foundation, San Diego, CA 92121, USA
| | - Heath Klock
- The Genomics Institute of the Novartis Research Foundation, San Diego, CA 92121, USA
| | - Paul W Anderson
- The Genomics Institute of the Novartis Research Foundation, San Diego, CA 92121, USA
| | - Sarah M Rue
- The Genomics Institute of the Novartis Research Foundation, San Diego, CA 92121, USA
| | - Jessica J Miller
- The Genomics Institute of the Novartis Research Foundation, San Diego, CA 92121, USA
| | - Scott M Glaser
- The Genomics Institute of the Novartis Research Foundation, San Diego, CA 92121, USA
| | - Melisa L Bragstad
- The Genomics Institute of the Novartis Research Foundation, San Diego, CA 92121, USA
| | - Julie Vance
- The Genomics Institute of the Novartis Research Foundation, San Diego, CA 92121, USA
| | - Annie W Lam
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - Scott A Lesley
- The Genomics Institute of the Novartis Research Foundation, San Diego, CA 92121, USA
| | - Kai Zinn
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - K Christopher Garcia
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA 94305, USA; Howard Hughes Medical Institute, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Structural Biology, Stanford University School of Medicine, Stanford, CA 94305, USA.
| |
Collapse
|
8
|
Marfull-Oromí P, Fleitas C, Zammou B, Rocandio D, Ballester-Lurbe B, Terrado J, Perez-Roger I, Espinet C, Egea J. Genetic ablation of the Rho GTPase Rnd3 triggers developmental defects in internal capsule and the globus pallidus formation. J Neurochem 2021; 158:197-216. [PMID: 33576044 DOI: 10.1111/jnc.15322] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Revised: 01/20/2021] [Accepted: 02/08/2021] [Indexed: 12/24/2022]
Abstract
The forebrain includes the cerebral cortex, the thalamus, and the striatum and globus pallidus (GP) in the subpallium. The formation of these structures and their interconnections by specific axonal tracts take place in a precise and orchestrated time and spatial-dependent manner during development. However, the knowledge of the molecular and cellular mechanisms that are involved is rather limited. Moreover, while many extracellular cues and specific receptors have been shown to play a role in different aspects of nervous system development, including neuron migration and axon guidance, examples of intracellular signaling effectors involved in these processes are sparse. In the present work, we have shown that the atypical RhoGTPase, Rnd3, is expressed very early during brain development and keeps a dynamic expression in several brain regions including the cortex, the thalamus, and the subpallium. By using a gene-trap allele (Rnd3gt ) and immunological techniques, we have shown that Rnd3gt/gt embryos display severe defects in striatal and thalamocortical axonal projections (SAs and TCAs, respectively) and defects in GP formation already at early stages. Surprisingly, the corridor, an important intermediate target for TCAs is still present in these mutants. Mechanistically, a conditional genetic deletion approach revealed that Rnd3 is primarily required for the normal development of Medial Ganglionic Eminence-derived structures, such as the GP, and therefore acts non-cell autonomously in SAs and TCAs. In conclusion, we have demonstrated the important role of Rnd3 as an early regulator of subpallium development in vivo and revealed new insights about SAs and TCAs development.
Collapse
Affiliation(s)
| | | | | | | | - Begoña Ballester-Lurbe
- Departamento de Medicina y Cirugía Animal, Facultad de Veterinaria, Universidad CEU Cardenal Herrera, Valencia, Spain
| | - Jose Terrado
- Departamento de Medicina y Cirugía Animal, Facultad de Veterinaria, Universidad CEU Cardenal Herrera, Valencia, Spain
| | - Ignacio Perez-Roger
- Departamento de Medicina y Cirugía Animal, Facultad de Veterinaria, Universidad CEU Cardenal Herrera, Valencia, Spain
| | | | - Joaquim Egea
- IRBLLEIDA/Universitat de Lleida, Serra Húnter associate professor, Lleida, Spain
| |
Collapse
|
9
|
A gene expression atlas for different kinds of stress in the mouse brain. Sci Data 2020; 7:437. [PMID: 33328476 PMCID: PMC7744580 DOI: 10.1038/s41597-020-00772-z] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Accepted: 11/25/2020] [Indexed: 12/17/2022] Open
Abstract
Stressful experiences are part of everyday life and animals have evolved physiological and behavioral responses aimed at coping with stress and maintaining homeostasis. However, repeated or intense stress can induce maladaptive reactions leading to behavioral disorders. Adaptations in the brain, mediated by changes in gene expression, have a crucial role in the stress response. Recent years have seen a tremendous increase in studies on the transcriptional effects of stress. The input raw data are freely available from public repositories and represent a wealth of information for further global and integrative retrospective analyses. We downloaded from the Sequence Read Archive 751 samples (SRA-experiments), from 18 independent BioProjects studying the effects of different stressors on the brain transcriptome in mice. We performed a massive bioinformatics re-analysis applying a single, standardized pipeline for computing differential gene expression. This data mining allowed the identification of novel candidate stress-related genes and specific signatures associated with different stress conditions. The large amount of computational results produced was systematized in the interactive “Stress Mice Portal”.
Collapse
|
10
|
Corley MJ, Vargas-Maya N, Pang APS, Lum-Jones A, Li D, Khadka V, Sultana R, Blanchard DC, Maunakea AK. Epigenetic Delay in the Neurodevelopmental Trajectory of DNA Methylation States in Autism Spectrum Disorders. Front Genet 2019; 10:907. [PMID: 31681403 PMCID: PMC6797928 DOI: 10.3389/fgene.2019.00907] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2019] [Accepted: 08/28/2019] [Indexed: 12/16/2022] Open
Abstract
Autism spectrum disorders (ASD) are hypothesized to originate in utero from perturbations in neural stem cell niche regions of the developing brain. Dynamic epigenetic processes including DNA methylation are integral to coordinating typical brain development. However, the extent and consequences of alterations to DNA methylation states in neural stem cell compartments in ASD are unknown. Here, we report significant DNA methylation defects in the subventricular zone of the lateral ventricles from postmortem brain of 17 autism diagnosed compared to 17 age- and gender-matched typically developing individuals. Both array- and sequencing-based genome-wide methylome analyses independently revealed that these alterations were preferentially targeted to intragenic and bivalently modified chromatin domains of genes predominately involved in neurodevelopment, which associated with aberrant precursor messenger RNA splicing events of ASD-relevant genes. Integrative analysis of our ASD and typically developing postmortem brain methylome datasets with that from fetal brain at different neurodevelopmental stages revealed that the methylation states of differentially methylated loci associated with ASD remarkably resemble the methylation states at earlier time points in fetal brain development. This observation was confirmed using additional methylome datasets from three other brain regions. Altogether, these findings implicate an epigenetic delay in the trajectory of normal DNA methylation states during the course of brain development that may consequently lead to deleterious transcriptomic events in ASD and support the hypothesis of an early developmental origin of ASD.
Collapse
Affiliation(s)
- Michael J Corley
- Department of Native Hawaiian Health, John A. Burns School of Medicine, University of Hawaii, Honolulu, HI, United States
| | - Nauru Vargas-Maya
- Department of Native Hawaiian Health, John A. Burns School of Medicine, University of Hawaii, Honolulu, HI, United States
| | - Alina P S Pang
- Department of Native Hawaiian Health, John A. Burns School of Medicine, University of Hawaii, Honolulu, HI, United States
| | - Annette Lum-Jones
- Department of Native Hawaiian Health, John A. Burns School of Medicine, University of Hawaii, Honolulu, HI, United States
| | - Dongmei Li
- Department of Clinical and Translational Research, University of Rochester Medical Center, Rochester, NY, United States
| | - Vedbar Khadka
- Office of Biostatistics & Quantitative Health Sciences, John A. Burns School of Medicine, University of Hawaii, Honolulu, HI, United States
| | - Razvan Sultana
- Department of Native Hawaiian Health, John A. Burns School of Medicine, University of Hawaii, Honolulu, HI, United States
| | - D Caroline Blanchard
- Bekesy Neurobiology Laboratory, Pacific Biosciences Research Center, University of Hawaii at Manoa, Honolulu, HI, United States
| | - Alika K Maunakea
- Department of Native Hawaiian Health, John A. Burns School of Medicine, University of Hawaii, Honolulu, HI, United States
| |
Collapse
|
11
|
Lees JA, Ferwerda B, Kremer PHC, Wheeler NE, Serón MV, Croucher NJ, Gladstone RA, Bootsma HJ, Rots NY, Wijmega-Monsuur AJ, Sanders EAM, Trzciński K, Wyllie AL, Zwinderman AH, van den Berg LH, van Rheenen W, Veldink JH, Harboe ZB, Lundbo LF, de Groot LCPGM, van Schoor NM, van der Velde N, Ängquist LH, Sørensen TIA, Nohr EA, Mentzer AJ, Mills TC, Knight JC, du Plessis M, Nzenze S, Weiser JN, Parkhill J, Madhi S, Benfield T, von Gottberg A, van der Ende A, Brouwer MC, Barrett JC, Bentley SD, van de Beek D. Joint sequencing of human and pathogen genomes reveals the genetics of pneumococcal meningitis. Nat Commun 2019; 10:2176. [PMID: 31092817 PMCID: PMC6520353 DOI: 10.1038/s41467-019-09976-3] [Citation(s) in RCA: 69] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Accepted: 04/11/2019] [Indexed: 12/21/2022] Open
Abstract
Streptococcus pneumoniae is a common nasopharyngeal colonizer, but can also cause life-threatening invasive diseases such as empyema, bacteremia and meningitis. Genetic variation of host and pathogen is known to play a role in invasive pneumococcal disease, though to what extent is unknown. In a genome-wide association study of human and pathogen we show that human variation explains almost half of variation in susceptibility to pneumococcal meningitis and one-third of variation in severity, identifying variants in CCDC33 associated with susceptibility. Pneumococcal genetic variation explains a large amount of invasive potential (70%), but has no effect on severity. Serotype alone is insufficient to explain invasiveness, suggesting other pneumococcal factors are involved in progression to invasive disease. We identify pneumococcal genes involved in invasiveness including pspC and zmpD, and perform a human-bacteria interaction analysis. These genes are potential candidates for the development of more broadly-acting pneumococcal vaccines.
Collapse
Affiliation(s)
- John A Lees
- Department of Microbiology, New York University School of Medicine, New York, NY, 10016, USA
- Parasites and Microbes, Wellcome Sanger Institute, Cambridge, CB10 1SA, UK
| | - Bart Ferwerda
- Amsterdam UMC, University of Amsterdam, Department of Neurology, Amsterdam Neuroscience, Meibergdreef 9, Amsterdam, 1105 AZ, The Netherlands
| | - Philip H C Kremer
- Amsterdam UMC, University of Amsterdam, Department of Neurology, Amsterdam Neuroscience, Meibergdreef 9, Amsterdam, 1105 AZ, The Netherlands
| | - Nicole E Wheeler
- Parasites and Microbes, Wellcome Sanger Institute, Cambridge, CB10 1SA, UK
- The Centre for Genomic Pathogen Surveillance, Wellcome Sanger Institute, Cambridge, CB10 1SA, UK
| | - Mercedes Valls Serón
- Amsterdam UMC, University of Amsterdam, Department of Neurology, Amsterdam Neuroscience, Meibergdreef 9, Amsterdam, 1105 AZ, The Netherlands
| | - Nicholas J Croucher
- MRC Centre for Global Infectious Disease Analysis, Department of Infectious Disease Epidemiology, Imperial College London, London, W2 1PG, UK
| | | | - Hester J Bootsma
- Centre for Infectious Disease Control, National Institute for Public Health and the Environment, Bilthoven, 3721 MA, The Netherlands
| | - Nynke Y Rots
- Centre for Infectious Disease Control, National Institute for Public Health and the Environment, Bilthoven, 3721 MA, The Netherlands
| | - Alienke J Wijmega-Monsuur
- Centre for Infectious Disease Control, National Institute for Public Health and the Environment, Bilthoven, 3721 MA, The Netherlands
| | - Elisabeth A M Sanders
- Centre for Infectious Disease Control, National Institute for Public Health and the Environment, Bilthoven, 3721 MA, The Netherlands
- Department of Pediatric Immunology and Infectious Diseases, Wilhelmina Children's Hospital, University Medical Centre Utrecht, Utrecht, 3508 AB, The Netherlands
| | - Krzysztof Trzciński
- Department of Pediatric Immunology and Infectious Diseases, Wilhelmina Children's Hospital, University Medical Centre Utrecht, Utrecht, 3508 AB, The Netherlands
| | - Anne L Wyllie
- Department of Pediatric Immunology and Infectious Diseases, Wilhelmina Children's Hospital, University Medical Centre Utrecht, Utrecht, 3508 AB, The Netherlands
- Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, CT, 06520, USA
| | - Aeilko H Zwinderman
- Amsterdam UMC, University of Amsterdam, Department of Clinical Epidemiology, Biostatistics and Bioinformatics, Amsterdam Public Health, Meibergdreef 9, Amsterdam, 1105 AZ, The Netherlands
| | - Leonard H van den Berg
- Department of Neurology, Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht, 3584 CG, The Netherlands
| | - Wouter van Rheenen
- Department of Neurology, Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht, 3584 CG, The Netherlands
| | - Jan H Veldink
- Department of Neurology, Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht, 3584 CG, The Netherlands
| | - Zitta B Harboe
- Department of Microbiological Surveillance and Research, Statens Serum Institut, Copenhagen, DK-2300, Denmark
| | - Lene F Lundbo
- Department of Infectious Diseases, Hvidovre Hospital, University of Copenhagen, Hvidovre, 2650, Denmark
| | - Lisette C P G M de Groot
- Department of Human Nutrition, Wageningen University, P.O. Box 17, 6700 AA, Wageningen, The Netherlands
| | - Natasja M van Schoor
- Amsterdam UMC, VU University, Department of Epidemiology and Biostatistics, Amsterdam Public Health, Van der Boechorststraat 7, Amsterdam, 1007 MB, The Netherlands
| | - Nathalie van der Velde
- Amsterdam UMC, University of Amsterdam, Department of Internal Medicine, Geriatrics, Amsterdam Public Health, Meibergdreef 9, Amsterdam, 1105 AZ, The Netherlands
- Department of Internal Medicine, Erasmus MC, University Medical Centre Rotterdam, P.O. Box 2040, 3000 CA, Rotterdam, The Netherlands
| | - Lars H Ängquist
- Center for Clinical Research and Disease Prevention, Bispebjerg and Frederiksberg Hospitals, The Capital Region, Copenhagen, DK-2000, Denmark
| | - Thorkild I A Sørensen
- The Novo Nordisk Foundation Center for Basic Metabolic Research, Section of Metabolic Genetics, Copenhagen, DK-2200, Denmark
- The Department of Public Health, Section of Epidemiology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, DK-1014, Denmark
| | - Ellen A Nohr
- Institute of Clinical Research, University of Southern Denmark, Odense, DK-5000, Denmark
| | - Alexander J Mentzer
- Wellcome Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, OX3 7BN, UK
| | - Tara C Mills
- Wellcome Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, OX3 7BN, UK
| | - Julian C Knight
- Wellcome Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, OX3 7BN, UK
| | - Mignon du Plessis
- School of Pathology, Faculty of Health Sciences, University of Witwatersrand, Johannesburg, 2000, South Africa
| | - Susan Nzenze
- School of Pathology, Faculty of Health Sciences, University of Witwatersrand, Johannesburg, 2000, South Africa
| | - Jeffrey N Weiser
- Department of Microbiology, New York University School of Medicine, New York, NY, 10016, USA
| | - Julian Parkhill
- Parasites and Microbes, Wellcome Sanger Institute, Cambridge, CB10 1SA, UK
| | - Shabir Madhi
- National Institute for Communicable Diseases, Johannesburg, 2192, South Africa
| | - Thomas Benfield
- Department of Infectious Diseases, Hvidovre Hospital, University of Copenhagen, Hvidovre, 2650, Denmark
| | - Anne von Gottberg
- School of Pathology, Faculty of Health Sciences, University of Witwatersrand, Johannesburg, 2000, South Africa
- National Institute for Communicable Diseases, Johannesburg, 2192, South Africa
| | - Arie van der Ende
- Amsterdam UMC, University of Amsterdam, Department of Medical Microbiology, Amsterdam Infection and Immunity, Meibergdreef 9, Amsterdam, 1105 AZ, The Netherlands
- Netherlands Reference Laboratory for Bacterial Meningitis, Amsterdam UMC/RIVM, University of Amsterdam, Meibergdreef 9, Amsterdam, 1105 AZ, The Netherlands
| | - Matthijs C Brouwer
- Amsterdam UMC, University of Amsterdam, Department of Neurology, Amsterdam Neuroscience, Meibergdreef 9, Amsterdam, 1105 AZ, The Netherlands
| | - Jeffrey C Barrett
- Parasites and Microbes, Wellcome Sanger Institute, Cambridge, CB10 1SA, UK
- Genomics Plc, East Road, Cambridge, CB1 1BH, UK
| | - Stephen D Bentley
- Parasites and Microbes, Wellcome Sanger Institute, Cambridge, CB10 1SA, UK.
| | - Diederik van de Beek
- Amsterdam UMC, University of Amsterdam, Department of Neurology, Amsterdam Neuroscience, Meibergdreef 9, Amsterdam, 1105 AZ, The Netherlands.
| |
Collapse
|
12
|
Schafer ST, Paquola ACM, Stern S, Gosselin D, Ku M, Pena M, Kuret TJM, Liyanage M, Mansour AA, Jaeger BN, Marchetto MC, Glass CK, Mertens J, Gage FH. Pathological priming causes developmental gene network heterochronicity in autistic subject-derived neurons. Nat Neurosci 2019; 22:243-255. [PMID: 30617258 PMCID: PMC6402576 DOI: 10.1038/s41593-018-0295-x] [Citation(s) in RCA: 177] [Impact Index Per Article: 29.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2018] [Accepted: 11/13/2018] [Indexed: 12/16/2022]
Abstract
Autism spectrum disorder (ASD) is thought to emerge during early cortical development. However, the exact developmental stages and associated molecular networks that prime disease propensity are elusive. To profile early neurodevelopmental alterations in ASD with macrocephaly, we monitored subject-derived induced pluripotent stem cells (iPSCs) throughout the recapitulation of cortical development. Our analysis revealed ASD-associated changes in the maturational sequence of early neuron development, involving temporal dysregulation of specific gene networks and morphological growth acceleration. The observed changes tracked back to a pathologically primed stage in neural stem cells (NSCs), reflected by altered chromatin accessibility. Concerted over-representation of network factors in control NSCs was sufficient to trigger ASD-like features, and circumventing the NSC stage by direct conversion of ASD iPSCs into induced neurons abolished ASD-associated phenotypes. Our findings identify heterochronic dynamics of a gene network that, while established earlier in development, contributes to subsequent neurodevelopmental aberrations in ASD.
Collapse
Affiliation(s)
- Simon T Schafer
- Laboratory of Genetics, The Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Apua C M Paquola
- Laboratory of Genetics, The Salk Institute for Biological Studies, La Jolla, CA, USA
- Lieber Institute for Brain Development, Baltimore, MD, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Shani Stern
- Laboratory of Genetics, The Salk Institute for Biological Studies, La Jolla, CA, USA
| | - David Gosselin
- Centre de Recherche du Centre Hospitalier Universitaire de Québec-Université Laval, Département de Médecine Moléculaire, Faculté de Médecine, Université Laval, Québec, Canada
| | - Manching Ku
- Next Generation Sequencing Core, The Salk Institute for Biological Studies, La Jolla, CA, USA
- Division of Pediatric Hematology and Oncology, Department of Pediatric and Adolescent Medicine, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Monique Pena
- Laboratory of Genetics, The Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Thomas J M Kuret
- Laboratory of Genetics, The Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Marvin Liyanage
- Laboratory of Genetics, The Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Abed AlFatah Mansour
- Laboratory of Genetics, The Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Baptiste N Jaeger
- Laboratory of Genetics, The Salk Institute for Biological Studies, La Jolla, CA, USA
- Laboratory of Neural Plasticity, Faculties of Medicine and Science, Brain Research Institute, University of Zurich, Zurich, Switzerland
| | - Maria C Marchetto
- Laboratory of Genetics, The Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Christopher K Glass
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Jerome Mertens
- Laboratory of Genetics, The Salk Institute for Biological Studies, La Jolla, CA, USA
- Department of Genomics, Stem Cell Biology and Regenerative Medicine, Institute of Molecular Biology & CMBI, University of Innsbruck, Innsbruck, Austria
| | - Fred H Gage
- Laboratory of Genetics, The Salk Institute for Biological Studies, La Jolla, CA, USA.
| |
Collapse
|
13
|
A novel ISLR2-linked autosomal recessive syndrome of congenital hydrocephalus, arthrogryposis and abdominal distension. Hum Genet 2018; 138:105-107. [DOI: 10.1007/s00439-018-1963-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2018] [Accepted: 11/21/2018] [Indexed: 01/22/2023]
|
14
|
Essential Role of Linx/Islr2 in the Development of the Forebrain Anterior Commissure. Sci Rep 2018; 8:7292. [PMID: 29739947 PMCID: PMC5940738 DOI: 10.1038/s41598-018-24064-0] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2017] [Accepted: 03/27/2018] [Indexed: 12/14/2022] Open
Abstract
Linx is a member of the leucine-rich repeat and immunoglobulin family of membrane proteins which has critical roles in the development of the peripheral nervous system and forebrain connectivity. A previous study showed that Linx is expressed in projection neurons in the cortex and in cells that comprise the passage to the prethalamus that form the internal capsule, indicating the involvement of Linx in axon guidance and cell-cell communication. In this study, we found that Linx-deficient mice develop severe hydrocephalus and die perinatally by unknown mechanisms. Importantly, mice heterozygous for the linx gene exhibited defects in the development of the anterior commissure in addition to hydrocephalus, indicating haploinsufficiency of the linx gene in forebrain development. In N1E-115 neuroblastoma cells and primary cultured hippocampal neurons, Linx depletion led to impaired neurite extension and an increase in cell body size. Consistent with this, but of unknown significance, we found that Linx interacts with and upregulates the activity of Rho-kinase, a modulator of many cellular processes including cytoskeletal organization. These data suggest a role for Linx in the regulation of complex forebrain connectivity, and future identification of its extracellular ligand(s) will help clarify this function.
Collapse
|
15
|
Seven pass Cadherins CELSR1-3. Semin Cell Dev Biol 2017; 69:102-110. [PMID: 28716607 DOI: 10.1016/j.semcdb.2017.07.014] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2017] [Revised: 07/12/2017] [Accepted: 07/13/2017] [Indexed: 11/20/2022]
Abstract
Cadherin EGF LAG seven-pass G-type receptors 1, 2 and 3 (CELSR1-3) form a family of three atypical cadherins with multiple functions in epithelia and in the nervous system. During the past decade, evidence has accumulated for a key role of CELSR1 in epithelial planar cell polarity (PCP), and for CELSR2 and CELSR3 in ciliogenesis and neural development, especially neuron migration and axon guidance in the central, peripheral and enteric nervous systems. Phenotypes in mutant mice indicate that CELSR proteins work in concert with FZD3 and FZD6, but several questions remain. Apart from PCP signaling pathways implicating CELSR1 that begin to be unraveled, little is known about other signals generated by CELSR2 and CELSR3. A crucial question concerns the putative ligands that trigger signaling, in particular what is the role of WNT factors. Another critical issue is the identification of novel intracellular pathways and effectors that relay and transmit signals in receptive cells? Answers to those questions should further our understanding of the role of those important molecules not only in development but also in regeneration and disease.
Collapse
|
16
|
Martini FJ, Moreno-Juan V, Filipchuk A, Valdeolmillos M, López-Bendito G. Impact of thalamocortical input on barrel cortex development. Neuroscience 2017; 368:246-255. [PMID: 28412498 DOI: 10.1016/j.neuroscience.2017.04.005] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2017] [Revised: 03/31/2017] [Accepted: 04/03/2017] [Indexed: 01/22/2023]
Abstract
The development of cortical maps requires the balanced interaction between genetically determined programs and input/activity-dependent signals generated spontaneously or triggered from the environment. The somatosensory pathway of mice provides an excellent scenario to study cortical map development because of its highly organized cytoarchitecture, known as the barrel field. This precise organization makes evident even small alterations in the cortical map layout. In this review, we will specially focus on the thalamic factors that control barrel field development. We will summarize the role of thalamic input integration and identity, neurotransmission and spontaneous activity in cortical map formation and early cross-modal plasticity.
Collapse
Affiliation(s)
- Francisco J Martini
- Instituto de Neurociencias de Alicante, Universidad Miguel Hernández-Consejo Superior de Investigaciones Científicas (UMH-CSIC), Sant Joan d'Alacant, Spain.
| | - Verónica Moreno-Juan
- Instituto de Neurociencias de Alicante, Universidad Miguel Hernández-Consejo Superior de Investigaciones Científicas (UMH-CSIC), Sant Joan d'Alacant, Spain
| | - Anton Filipchuk
- Instituto de Neurociencias de Alicante, Universidad Miguel Hernández-Consejo Superior de Investigaciones Científicas (UMH-CSIC), Sant Joan d'Alacant, Spain
| | - Miguel Valdeolmillos
- Instituto de Neurociencias de Alicante, Universidad Miguel Hernández-Consejo Superior de Investigaciones Científicas (UMH-CSIC), Sant Joan d'Alacant, Spain
| | - Guillermina López-Bendito
- Instituto de Neurociencias de Alicante, Universidad Miguel Hernández-Consejo Superior de Investigaciones Científicas (UMH-CSIC), Sant Joan d'Alacant, Spain.
| |
Collapse
|
17
|
Gezelius H, López-Bendito G. Thalamic neuronal specification and early circuit formation. Dev Neurobiol 2016; 77:830-843. [PMID: 27739248 DOI: 10.1002/dneu.22460] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2016] [Revised: 09/16/2016] [Accepted: 10/10/2016] [Indexed: 12/12/2022]
Abstract
The thalamus is a central structure of the brain, primarily recognized for the relay of incoming sensory and motor information to the cerebral cortex but also key in high order intracortical communication. It consists of glutamatergic projection neurons organized in several distinct nuclei, each having a stereotype connectivity pattern and functional roles. In the adult, these nuclei can be appreciated by architectural boundaries, although their developmental origin and specification is only recently beginning to be revealed. Here, we summarize the current knowledge on the specification of the distinct thalamic neurons and nuclei, starting from early embryonic patterning until the postnatal days when active sensory experience is initiated and the overall system connectivity is already established. We also include an overview of the guidance processes important for establishing thalamocortical connections, with emphasis on the early topographical specification. The extensively studied thalamocortical axon branching in the cortex is briefly mentioned; however, the maturation and plasticity of this connection are beyond the scope of this review. In separate chapters, additional mechanisms and/or features that influence the specification and development of thalamic neurons and their circuits are also discussed. Finally, an outlook of future directions is given. © 2016 Wiley Periodicals, Inc. Develop Neurobiol 77: 830-843, 2017.
Collapse
Affiliation(s)
- Henrik Gezelius
- Instituto de Neurociencias de Alicante, Universidad Miguel Hernández-Consejo Superior de Investigaciones Científicas (UMH-CSIC), Avenida Ramón y Cajal, s/n, Sant Joan d'Alacant, Spain
| | - Guillermina López-Bendito
- Instituto de Neurociencias de Alicante, Universidad Miguel Hernández-Consejo Superior de Investigaciones Científicas (UMH-CSIC), Avenida Ramón y Cajal, s/n, Sant Joan d'Alacant, Spain
| |
Collapse
|
18
|
Ledda F, Paratcha G. Assembly of Neuronal Connectivity by Neurotrophic Factors and Leucine-Rich Repeat Proteins. Front Cell Neurosci 2016; 10:199. [PMID: 27555809 PMCID: PMC4977320 DOI: 10.3389/fncel.2016.00199] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2016] [Accepted: 07/29/2016] [Indexed: 11/13/2022] Open
Abstract
Proper function of the nervous system critically relies on sophisticated neuronal networks interconnected in a highly specific pattern. The architecture of these connections arises from sequential developmental steps such as axonal growth and guidance, dendrite development, target determination, synapse formation and plasticity. Leucine-rich repeat (LRR) transmembrane proteins have been involved in cell-type specific signaling pathways that underlie these developmental processes. The members of this superfamily of proteins execute their functions acting as trans-synaptic cell adhesion molecules involved in target specificity and synapse formation or working in cis as cell-intrinsic modulators of neurotrophic factor receptor trafficking and signaling. In this review, we will focus on novel physiological mechanisms through which LRR proteins regulate neurotrophic factor receptor signaling, highlighting the importance of these modulatory events for proper axonal extension and guidance, tissue innervation and dendrite morphogenesis. Additionally, we discuss few examples linking this set of LRR proteins to neurodevelopmental and psychiatric disorders.
Collapse
Affiliation(s)
- Fernanda Ledda
- Division of Molecular and Cellular Neuroscience, Institute of Cell Biology and Neuroscience (IBCN)-CONICET, School of Medicine-University of Buenos Aires (UBA) Buenos Aires, Argentina
| | - Gustavo Paratcha
- Division of Molecular and Cellular Neuroscience, Institute of Cell Biology and Neuroscience (IBCN)-CONICET, School of Medicine-University of Buenos Aires (UBA) Buenos Aires, Argentina
| |
Collapse
|
19
|
Feng J, Xian Q, Guan T, Hu J, Wang M, Huang Y, So KF, Evans SM, Chai G, Goffinet AM, Qu Y, Zhou L. Celsr3 and Fzd3 Organize a Pioneer Neuron Scaffold to Steer Growing Thalamocortical Axons. Cereb Cortex 2016; 26:3323-34. [PMID: 27170656 PMCID: PMC4898681 DOI: 10.1093/cercor/bhw132] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Celsr3 and Fzd3 regulate the development of reciprocal thalamocortical projections independently of their expression in cortical or thalamic neurons. To understand this cell non autonomous mechanism further, we tested whether Celsr3 and Fzd3 could act via Isl1-positive guidepost cells. Isl1-positive cells appear in the forebrain at embryonic day (E) 9.5-E10.5 and, from E12.5, they form 2 contingents in ventral telencephalon and prethalamus. In control mice, corticothalamic axons run in the ventral telencephalic corridor in close contact with Isl1-positive cells. When Celsr3 or Fzd3 is inactivated in Isl1-expressing cells, corticofugal fibers stall and loop in the ventral telencephalic corridor of high Isl1 expression, and thalamic axons fail to cross the diencephalon–telencephalon junction (DTJ). At E12.5, before thalamic and cortical axons emerge, pioneer projections from Isl1-positive cells cross the DTJ from both sides in control but not mutant embryos. These early projections appear to act like a bridge to guide later growing thalamic axons through the DTJ. Our data suggest that Celsr3 and Fzd3 orchestrate the formation of a scaffold of pioneer neurons and their axons. This scaffold extends from prethalamus to ventral telencephalon and subcortex, and steers reciprocal corticothalamic fibers.
Collapse
Affiliation(s)
- Jia Feng
- Guangdong-Hongkong-Macau Institute of CNS Regeneration, Ministry of Education CNS Regeneration Collaborative Joint Laboratory Guangdong key Laboratory of Brain Function and Diseases, Jinan University, Guangzhou 510632, PR China
| | - Quanxiang Xian
- Guangdong-Hongkong-Macau Institute of CNS Regeneration, Ministry of Education CNS Regeneration Collaborative Joint Laboratory Guangdong key Laboratory of Brain Function and Diseases, Jinan University, Guangzhou 510632, PR China
| | - Tingting Guan
- Guangdong-Hongkong-Macau Institute of CNS Regeneration, Ministry of Education CNS Regeneration Collaborative Joint Laboratory Guangdong key Laboratory of Brain Function and Diseases, Jinan University, Guangzhou 510632, PR China
| | - Jing Hu
- Guangdong-Hongkong-Macau Institute of CNS Regeneration, Ministry of Education CNS Regeneration Collaborative Joint Laboratory Guangdong key Laboratory of Brain Function and Diseases, Jinan University, Guangzhou 510632, PR China
| | - Meizhi Wang
- Guangdong-Hongkong-Macau Institute of CNS Regeneration, Ministry of Education CNS Regeneration Collaborative Joint Laboratory Guangdong key Laboratory of Brain Function and Diseases, Jinan University, Guangzhou 510632, PR China
| | - Yuhua Huang
- Guangdong-Hongkong-Macau Institute of CNS Regeneration, Ministry of Education CNS Regeneration Collaborative Joint Laboratory Guangdong key Laboratory of Brain Function and Diseases, Jinan University, Guangzhou 510632, PR China
| | - Kwok-Fai So
- Guangdong-Hongkong-Macau Institute of CNS Regeneration, Ministry of Education CNS Regeneration Collaborative Joint Laboratory Guangdong key Laboratory of Brain Function and Diseases, Jinan University, Guangzhou 510632, PR China Department of Anatomy, The University of Hong Kong Pokfulam, Hong Kong SAR, PR China
| | - Sylvia M Evans
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA 92093, USA
| | - Guoliang Chai
- Institute of Neuroscience, Université catholique de Louvain, Brussels B1200, Belgium
| | - Andre M Goffinet
- Institute of Neuroscience, Université catholique de Louvain, Brussels B1200, Belgium
| | - Yibo Qu
- Guangdong-Hongkong-Macau Institute of CNS Regeneration, Ministry of Education CNS Regeneration Collaborative Joint Laboratory Guangdong key Laboratory of Brain Function and Diseases, Jinan University, Guangzhou 510632, PR China Co-innovation Center of Neuroregeneration, Jiangsu, China
| | - Libing Zhou
- Guangdong-Hongkong-Macau Institute of CNS Regeneration, Ministry of Education CNS Regeneration Collaborative Joint Laboratory Guangdong key Laboratory of Brain Function and Diseases, Jinan University, Guangzhou 510632, PR China Co-innovation Center of Neuroregeneration, Jiangsu, China
| |
Collapse
|
20
|
Martinez-Martin N, Ramani SR, Hackney JA, Tom I, Wranik BJ, Chan M, Wu J, Paluch MT, Takeda K, Hass PE, Clark H, Gonzalez LC. The extracellular interactome of the human adenovirus family reveals diverse strategies for immunomodulation. Nat Commun 2016; 7:11473. [PMID: 27145901 PMCID: PMC4858740 DOI: 10.1038/ncomms11473] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2015] [Accepted: 03/30/2016] [Indexed: 01/06/2023] Open
Abstract
Viruses encode secreted and cell-surface expressed proteins essential to modulate host immune defenses and establish productive infections. However, to date there has been no systematic study of the extracellular interactome of any human virus. Here we utilize the E3 proteins, diverse and rapidly evolving transmembrane-containing proteins encoded by human adenoviruses, as a model system to survey the extracellular immunomodulatory landscape. From a large-scale protein interaction screen against a microarray of more than 1,500 human proteins, we find and validate 51 previously unidentified virus–host interactions. Our results uncover conserved strategies as well as substantial diversity and multifunctionality in host targeting within and between viral species. Prominent modulation of the leukocyte immunoglobulin-like and signalling lymphocyte activation molecule families and a number of inhibitory receptors were identified as hubs for viral perturbation, suggesting unrecognized immunoregulatory strategies. We describe a virus–host extracellular interaction map of unprecedented scale that provides new insights into viral immunomodulation. Viruses interact with their hosts via secreted and membrane-bound proteins to affect host immune responses and virulence. Here the authors contribute to our understanding of this relationship with an extracellular interaction map of human and adenoviral E3 immunomodulatory proteins.
Collapse
Affiliation(s)
- Nadia Martinez-Martin
- Department of Protein Chemistry, Genentech, 470 East Grand Avenue, South San Francisco, California 94080, USA
| | - Sree R Ramani
- Department of Protein Chemistry, Genentech, 470 East Grand Avenue, South San Francisco, California 94080, USA
| | - Jason A Hackney
- Department of Bioinformatics and Computational Biology, Genentech, 455 East Grand Avenue, South San Francisco, California 94080, USA
| | - Irene Tom
- Department of Protein Chemistry, Genentech, 470 East Grand Avenue, South San Francisco, California 94080, USA
| | - Bernd J Wranik
- Department of Protein Chemistry, Genentech, 470 East Grand Avenue, South San Francisco, California 94080, USA
| | - Michelle Chan
- Department of Protein Chemistry, Genentech, 470 East Grand Avenue, South San Francisco, California 94080, USA
| | - Johnny Wu
- Department of Bioinformatics and Computational Biology, Genentech, 455 East Grand Avenue, South San Francisco, California 94080, USA
| | - Maciej T Paluch
- Department of Protein Chemistry, Genentech, 470 East Grand Avenue, South San Francisco, California 94080, USA
| | - Kentaro Takeda
- Department of Protein Chemistry, Genentech, 470 East Grand Avenue, South San Francisco, California 94080, USA
| | - Philip E Hass
- Department of Protein Chemistry, Genentech, 470 East Grand Avenue, South San Francisco, California 94080, USA
| | - Hilary Clark
- Department of Bioinformatics and Computational Biology, Genentech, 455 East Grand Avenue, South San Francisco, California 94080, USA
| | - Lino C Gonzalez
- Department of Protein Chemistry, Genentech, 470 East Grand Avenue, South San Francisco, California 94080, USA
| |
Collapse
|
21
|
Wang W, Jossin Y, Chai G, Lien WH, Tissir F, Goffinet AM. Feedback regulation of apical progenitor fate by immature neurons through Wnt7-Celsr3-Fzd3 signalling. Nat Commun 2016; 7:10936. [PMID: 26939553 PMCID: PMC4786577 DOI: 10.1038/ncomms10936] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2015] [Accepted: 02/03/2016] [Indexed: 11/09/2022] Open
Abstract
Sequential generation of neurons and glial cells during development is critical for the wiring and function of the cerebral cortex. This process requires accurate coordination of neural progenitor cell (NPC) fate decisions, by NPC-autonomous mechanisms as well as by negative feedback from neurons. Here, we show that neurogenesis is protracted and gliogenesis decreased in mice with mutations of genes Celsr3 and Fzd3. This phenotype is not due to gene inactivation in progenitors, but rather in immature cortical neurons. Mutant neurons are unable to upregulate expression of Jag1 in response to cortical Wnt7, resulting in blunted activation of Notch signalling in NPC. Thus, Celsr3 and Fzd3 enable immature neurons to respond to Wnt7, upregulate Jag1 and thereby facilitate feedback signals that tune the timing of NPC fate decisions via Notch activation.
Collapse
Affiliation(s)
- Wei Wang
- Université catholique de Louvain, Institute of Neuroscience, 73 Avenue Mounier, Box B1.7316, 1200 Brussels, Belgium
| | - Yves Jossin
- Université catholique de Louvain, Institute of Neuroscience, 73 Avenue Mounier, Box B1.7316, 1200 Brussels, Belgium
| | - Guoliang Chai
- Université catholique de Louvain, Institute of Neuroscience, 73 Avenue Mounier, Box B1.7316, 1200 Brussels, Belgium
| | - Wen-Hui Lien
- Université catholique de Louvain, de Duve Institute, Avenue Hippocrate 74, Box B1.74.09, 1200 Brussels, Belgium
| | - Fadel Tissir
- Université catholique de Louvain, Institute of Neuroscience, 73 Avenue Mounier, Box B1.7316, 1200 Brussels, Belgium
| | - Andre M Goffinet
- Université catholique de Louvain, Institute of Neuroscience, 73 Avenue Mounier, Box B1.7316, 1200 Brussels, Belgium.,WELBIO, 6 Avenue Pasteur, 1300 Wavre, Belgium
| |
Collapse
|
22
|
Maeda K, Enomoto A, Hara A, Asai N, Kobayashi T, Horinouchi A, Maruyama S, Ishikawa Y, Nishiyama T, Kiyoi H, Kato T, Ando K, Weng L, Mii S, Asai M, Mizutani Y, Watanabe O, Hirooka Y, Goto H, Takahashi M. Identification of Meflin as a Potential Marker for Mesenchymal Stromal Cells. Sci Rep 2016; 6:22288. [PMID: 26924503 PMCID: PMC4770287 DOI: 10.1038/srep22288] [Citation(s) in RCA: 77] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2015] [Accepted: 02/11/2016] [Indexed: 01/14/2023] Open
Abstract
Bone marrow-derived mesenchymal stromal cells (BM-MSCs) in culture are derived from BM stromal cells or skeletal stem cells. Whereas MSCs have been exploited in clinical medicine, the identification of MSC-specific markers has been limited. Here, we report that a cell surface and secreted protein, Meflin, is expressed in cultured MSCs, fibroblasts and pericytes, but not other types of cells including epithelial, endothelial and smooth muscle cells. In vivo, Meflin is expressed by immature osteoblasts and chondroblasts. In addition, Meflin is found on stromal cells distributed throughout the BM, and on pericytes and perivascular cells in multiple organs. Meflin maintains the undifferentiated state of cultured MSCs and is downregulated upon their differentiation, consistent with the observation that Meflin-deficient mice exhibit increased number of osteoblasts and accelerated bone development. In the bone and BM, Meflin is more highly expressed in primitive stromal cells that express platelet-derived growth factor receptor α and Sca-1 than the Sca-1-negative adipo-osteogenic progenitors, which create a niche for hematopoiesis. Those results are consistent with a decrease in the number of clonogenic colony-forming unit-fibroblasts within the BM of Meflin-deficient mice. These preliminary data suggest that Meflin is a potential marker for cultured MSCs and their source cells in vivo.
Collapse
Affiliation(s)
- Keiko Maeda
- Department of Pathology, 65 Tsurumai-cho, Showa-ku, Nagoya 466-8550, Japan.,Department of Gastroenterology, 65 Tsurumai-cho, Showa-ku, Nagoya 466-8550, Japan
| | - Atsushi Enomoto
- Department of Pathology, 65 Tsurumai-cho, Showa-ku, Nagoya 466-8550, Japan
| | - Akitoshi Hara
- Department of Pathology, 65 Tsurumai-cho, Showa-ku, Nagoya 466-8550, Japan
| | - Naoya Asai
- Department of Pathology, 65 Tsurumai-cho, Showa-ku, Nagoya 466-8550, Japan
| | - Takeshi Kobayashi
- Department of Physiology, 65 Tsurumai-cho, Showa-ku, Nagoya 466-8550, Japan
| | - Asuka Horinouchi
- Department of Nephrology, 65 Tsurumai-cho, Showa-ku, Nagoya 466-8550, Japan
| | - Shoichi Maruyama
- Department of Nephrology, 65 Tsurumai-cho, Showa-ku, Nagoya 466-8550, Japan
| | - Yuichi Ishikawa
- Department of Hematology and Oncology, Nagoya University Graduate School of Medicine, , 65 Tsurumai-cho, Showa-ku, Nagoya 466-8550, Japan
| | - Takahiro Nishiyama
- Department of Hematology and Oncology, Nagoya University Graduate School of Medicine, , 65 Tsurumai-cho, Showa-ku, Nagoya 466-8550, Japan
| | - Hitoshi Kiyoi
- Department of Hematology and Oncology, Nagoya University Graduate School of Medicine, , 65 Tsurumai-cho, Showa-ku, Nagoya 466-8550, Japan
| | - Takuya Kato
- Tumour Cell Biology Laboratory, The Francis-Crick Institute, 44 Lincoln's Inn Fields, London, WC2A 3LY, United Kingdom
| | - Kenju Ando
- Department of Pathology, 65 Tsurumai-cho, Showa-ku, Nagoya 466-8550, Japan
| | - Liang Weng
- Department of Pathology, 65 Tsurumai-cho, Showa-ku, Nagoya 466-8550, Japan
| | - Shinji Mii
- Department of Pathology, 65 Tsurumai-cho, Showa-ku, Nagoya 466-8550, Japan
| | - Masato Asai
- Department of Pathology, 65 Tsurumai-cho, Showa-ku, Nagoya 466-8550, Japan
| | - Yasuyuki Mizutani
- Department of Pathology, 65 Tsurumai-cho, Showa-ku, Nagoya 466-8550, Japan.,Department of Gastroenterology, 65 Tsurumai-cho, Showa-ku, Nagoya 466-8550, Japan
| | - Osamu Watanabe
- Department of Gastroenterology, 65 Tsurumai-cho, Showa-ku, Nagoya 466-8550, Japan
| | - Yoshiki Hirooka
- Department of Gastroenterology, 65 Tsurumai-cho, Showa-ku, Nagoya 466-8550, Japan
| | - Hidemi Goto
- Department of Gastroenterology, 65 Tsurumai-cho, Showa-ku, Nagoya 466-8550, Japan
| | - Masahide Takahashi
- Department of Pathology, 65 Tsurumai-cho, Showa-ku, Nagoya 466-8550, Japan
| |
Collapse
|
23
|
Draxin from neocortical neurons controls the guidance of thalamocortical projections into the neocortex. Nat Commun 2015; 6:10232. [PMID: 26659141 PMCID: PMC4682175 DOI: 10.1038/ncomms10232] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2015] [Accepted: 11/19/2015] [Indexed: 11/21/2022] Open
Abstract
The thalamocortical tract carries sensory information to the neocortex. It has long been recognized that the neocortical pioneer axons of subplate neurons are essential for thalamocortical development. Herein we report that an axon guidance cue, draxin, is expressed in early-born neocortical neurons, including subplate neurons, and is necessary for thalamocortical development. In draxin−/− mice, thalamocortical axons do not enter the neocortex. This phenotype is sufficiently rescued by the transgenic expression of draxin in neocortical neurons. Genetic interaction data suggest that draxin acts through Deleted in colorectal cancer (DCC) and Neogenin (Neo1), to regulate thalamocortical projections in vivo. Draxin promotes the outgrowth of thalamic axons in vitro and this effect is abolished in thalamic neurons from Dcc and Neo1 double mutants. These results suggest that draxin from neocortical neurons controls thalamocortical projections into the neocortex, and that this effect is mediated through the DCC and Neo1 receptors. During neural development thalamocortical axons follow corticofugal projections into the neocortex. Here, using a combination of knock down and rescue experiments, the authors show that Draxin expression in neocortical cells promotes thalamic axon projections from the internal capsule.
Collapse
|
24
|
Panza P, Sitko AA, Maischein HM, Koch I, Flötenmeyer M, Wright GJ, Mandai K, Mason CA, Söllner C. The LRR receptor Islr2 is required for retinal axon routing at the vertebrate optic chiasm. Neural Dev 2015; 10:23. [PMID: 26492970 PMCID: PMC4618557 DOI: 10.1186/s13064-015-0050-x] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2015] [Accepted: 10/01/2015] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND In the visual system of most binocular vertebrates, the axons of retinal ganglion cells (RGCs) diverge at the diencephalic midline and extend to targets on both ipsi- and contralateral sides of the brain. While a molecular mechanism explaining ipsilateral guidance decisions has been characterized, less is known of how RGC axons cross the midline. RESULTS Here, we took advantage of the zebrafish, in which all RGC axons project contralaterally at the optic chiasm, to characterize Islr2 as an RGC receptor required for complete retinal axon midline crossing. We used a systematic extracellular protein-protein interaction screening assay to identify two Vasorin paralogs, Vasna and Vasnb, as specific Islr2 ligands. Antibodies against Vasna and Vasnb reveal cellular populations surrounding the retinal axon pathway, suggesting the involvement of these proteins in guidance decisions made by axons of the optic nerve. Specifically, Vasnb marks the membranes of a cellular barricade located anteriorly to the optic chiasm, a structure termed the "glial knot" in higher vertebrates. Loss of function mutations in either vasorin paralog, individually or combined, however, do not exhibit an overt retinal axon projection phenotype, suggesting that additional midline factors, acting either independently or redundantly, compensate for their loss. Analysis of Islr2 knockout mice supports a scenario in which Islr2 controls the coherence of RGC axons through the ventral midline and optic tract. CONCLUSIONS Although stereotypic guidance of RGC axons at the vertebrate optic chiasm is controlled by multiple, redundant mechanisms, and despite the differences in ventral diencephalic tissue architecture, we identify a novel role for the LRR receptor Islr2 in ensuring proper axon navigation at the optic chiasm of both zebrafish and mouse.
Collapse
Affiliation(s)
- Paolo Panza
- Max-Planck-Institut für Entwicklungsbiologie, Abteilung Genetik, Spemannstraße 35, 72076, Tübingen, Germany.
| | - Austen A Sitko
- Department of Neuroscience, Columbia University, College of Physicians and Surgeons, 630 West 168th Street, New York, NY, 10032, USA
| | - Hans-Martin Maischein
- Max-Planck-Institut für Entwicklungsbiologie, Abteilung Genetik, Spemannstraße 35, 72076, Tübingen, Germany.,Present address: Max-Planck-Institut für Herz- und Lungenforschung, Abteilung Genetik der Entwicklung, Ludwigstraße 43, 61231, Bad Nauheim, Germany
| | - Iris Koch
- Max-Planck-Institut für Entwicklungsbiologie, Elektronenmikroskopie, Spemannstraße 35, 72076, Tübingen, Germany
| | - Matthias Flötenmeyer
- Max-Planck-Institut für Entwicklungsbiologie, Elektronenmikroskopie, Spemannstraße 35, 72076, Tübingen, Germany
| | - Gavin J Wright
- Wellcome Trust Sanger Institute, Cell Surface Signalling Laboratory, Hinxton, Cambridge, CB10 1HH, UK
| | - Kenji Mandai
- Department of Biochemistry and Molecular Biology, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-cho, Chuo-ku, Kobe, Hyogo, 650-0017, Japan
| | - Carol A Mason
- Department of Pathology & Cell Biology, Columbia University, College of Physicians and Surgeons, 630 West 168th Street, New York, NY, 10032, USA.,Department of Neuroscience, Columbia University, College of Physicians and Surgeons, 630 West 168th Street, New York, NY, 10032, USA
| | - Christian Söllner
- Max-Planck-Institut für Entwicklungsbiologie, Abteilung Genetik, Spemannstraße 35, 72076, Tübingen, Germany
| |
Collapse
|
25
|
Squarzoni P, Thion MS, Garel S. Neuronal and microglial regulators of cortical wiring: usual and novel guideposts. Front Neurosci 2015; 9:248. [PMID: 26236185 PMCID: PMC4505395 DOI: 10.3389/fnins.2015.00248] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2015] [Accepted: 06/30/2015] [Indexed: 12/17/2022] Open
Abstract
Neocortex functioning relies on the formation of complex networks that begin to be assembled during embryogenesis by highly stereotyped processes of cell migration and axonal navigation. The guidance of cells and axons is driven by extracellular cues, released along by final targets or intermediate targets located along specific pathways. In particular, guidepost cells, originally described in the grasshopper, are considered discrete, specialized cell populations located at crucial decision points along axonal trajectories that regulate tract formation. These cells are usually early-born, transient and act at short-range or via cell-cell contact. The vast majority of guidepost cells initially identified were glial cells, which play a role in the formation of important axonal tracts in the forebrain, such as the corpus callosum, anterior, and post-optic commissures as well as optic chiasm. In the last decades, tangential migrating neurons have also been found to participate in the guidance of principal axonal tracts in the forebrain. This is the case for several examples such as guideposts for the lateral olfactory tract (LOT), corridor cells, which open an internal path for thalamo-cortical axons and Cajal-Retzius cells that have been involved in the formation of the entorhino-hippocampal connections. More recently, microglia, the resident macrophages of the brain, were specifically observed at the crossroads of important neuronal migratory routes and axonal tract pathways during forebrain development. We furthermore found that microglia participate to the shaping of prenatal forebrain circuits, thereby opening novel perspectives on forebrain development and wiring. Here we will review the last findings on already known guidepost cell populations and will discuss the role of microglia as a potentially new class of atypical guidepost cells.
Collapse
Affiliation(s)
- Paola Squarzoni
- Centre National de la Recherche Scientifique UMR8197, Ecole Normale Supérieure, Institut de Biologie, Institut National de la Santé et de la Recherche Médicale U1024 Paris, France
| | - Morgane S Thion
- Centre National de la Recherche Scientifique UMR8197, Ecole Normale Supérieure, Institut de Biologie, Institut National de la Santé et de la Recherche Médicale U1024 Paris, France
| | - Sonia Garel
- Centre National de la Recherche Scientifique UMR8197, Ecole Normale Supérieure, Institut de Biologie, Institut National de la Santé et de la Recherche Médicale U1024 Paris, France
| |
Collapse
|
26
|
Chai G, Goffinet AM, Tissir F. Celsr3 and Fzd3 in axon guidance. Int J Biochem Cell Biol 2015; 64:11-4. [PMID: 25813877 DOI: 10.1016/j.biocel.2015.03.013] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2015] [Revised: 03/10/2015] [Accepted: 03/16/2015] [Indexed: 12/21/2022]
Abstract
The assembly of functional neuronal circuits depends on the correct wiring of axons and dendrites. To reach their targets, axons are guided by a variety of extracellular guidance cues, including Netrins, Ephrins, Semaphorins and Slits. Corresponding receptors in the growth cone, the dynamic structure at the tip of the growing axon, sense and integrate these positional signals, and activate downstream effectors to regulate cytoskeletal organization. In addition to the four canonical families of axon guidance cues mentioned above, some proteins that regulate planar cell polarity were recently found to be critical for axon guidance. The seven-transmembrane domain receptors Celsr3 and Fzd3, in particular, control the development of most longitudinal tracts in the central nervous system, and axon navigation in the peripheral, sympathetic and enteric nervous systems. Despite their unequivocally important role, however, underlying molecular mechanisms remain elusive. We do not know which extracellular ligands they recognize, whether they have co-receptors in the growth cone, and what their downstream effectors are. Here, we review some recent advances and discuss future trends in this emerging field.
Collapse
Affiliation(s)
- Guoliang Chai
- Institute of Neuroscience, Université catholique de Louvain, 73 Avenue Mounier, B1.73.16, Brussels 1200, Belgium
| | - Andre M Goffinet
- Institute of Neuroscience, Université catholique de Louvain, 73 Avenue Mounier, B1.73.16, Brussels 1200, Belgium.
| | - Fadel Tissir
- Institute of Neuroscience, Université catholique de Louvain, 73 Avenue Mounier, B1.73.16, Brussels 1200, Belgium.
| |
Collapse
|
27
|
Lokmane L, Garel S. Map transfer from the thalamus to the neocortex: inputs from the barrel field. Semin Cell Dev Biol 2014; 35:147-55. [PMID: 25020201 DOI: 10.1016/j.semcdb.2014.07.005] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2014] [Revised: 07/03/2014] [Accepted: 07/04/2014] [Indexed: 01/05/2023]
Abstract
Sensory perception relies on the formation of stereotyped maps inside the brain. This feature is particularly well illustrated in the mammalian neocortex, which is subdivided into distinct cortical sensory areas that comprise topological maps, such as the somatosensory homunculus in humans or the barrel field of the large whiskers in rodents. How somatosensory maps are formed and relayed into the neocortex remain essential questions in developmental neuroscience. Here, we will present our current knowledge on whisker map transfer in the mouse model, with the goal of linking embryonic and postnatal studies into a comprehensive framework.
Collapse
Affiliation(s)
- Ludmilla Lokmane
- Ecole Normale Supérieure, Institut de Biologie de l'ENS, IBENS, 46 rue d'Ulm, Paris F-75005, France; Inserm, U1024, Paris F-75005, France; CNRS, UMR 8197, Paris F-75005, France.
| | - Sonia Garel
- Ecole Normale Supérieure, Institut de Biologie de l'ENS, IBENS, 46 rue d'Ulm, Paris F-75005, France; Inserm, U1024, Paris F-75005, France; CNRS, UMR 8197, Paris F-75005, France.
| |
Collapse
|