1
|
Blair JD, Hartman A, Zenk F, Wahle P, Brancati G, Dalgarno C, Treutlein B, Satija R. Phospho-seq: integrated, multi-modal profiling of intracellular protein dynamics in single cells. Nat Commun 2025; 16:1346. [PMID: 39905064 PMCID: PMC11794950 DOI: 10.1038/s41467-025-56590-7] [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: 11/15/2024] [Accepted: 01/22/2025] [Indexed: 02/06/2025] Open
Abstract
Cell signaling plays a critical role in neurodevelopment, regulating cellular behavior and fate. While multimodal single-cell sequencing technologies are rapidly advancing, scalable and flexible profiling of cell signaling states alongside other molecular modalities remains challenging. Here we present Phospho-seq, an integrated approach that aims to quantify cytoplasmic and nuclear proteins, including those with post-translational modifications, and to connect their activity with cis-regulatory elements and transcriptional targets. We utilize a simplified benchtop antibody conjugation method to create large custom neuro-focused antibody panels for simultaneous protein and scATAC-seq profiling on whole cells, alongside both experimental and computational strategies to incorporate transcriptomic measurements. We apply our workflow to cell lines, induced pluripotent stem cells, and months-old retinal and brain organoids to demonstrate its broad applicability. We show that Phospho-seq can provide insights into cellular states and trajectories, shed light on gene regulatory relationships, and help explore the causes and effects of diverse cell signaling in neurodevelopment.
Collapse
Affiliation(s)
- John D Blair
- New York Genome Center, New York, NY, USA
- Center for Genomics and Systems Biology, New York University, New York, NY, USA
| | | | | | | | | | | | | | - Rahul Satija
- New York Genome Center, New York, NY, USA.
- Center for Genomics and Systems Biology, New York University, New York, NY, USA.
| |
Collapse
|
2
|
Prakash N, Abu Irqeba A, Corbin JG. Development and function of the medial amygdala. Trends Neurosci 2025; 48:22-32. [PMID: 39672784 DOI: 10.1016/j.tins.2024.11.004] [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: 07/12/2024] [Revised: 11/05/2024] [Accepted: 11/19/2024] [Indexed: 12/15/2024]
Abstract
Across studied vertebrates, the medial amygdala (MeA) is a central hub for relaying sensory information with social and/or survival relevance to downstream nuclei such as the bed nucleus of stria terminalis (BNST) and the hypothalamus. MeA-driven behaviors, such as mating, aggression, parenting, and predator avoidance are processed by different molecularly defined inhibitory and excitatory neuronal output populations. Work over the past two decades has deciphered how diverse MeA neurons arise from embryonic development, revealing contributions from multiple telencephalic and diencephalic progenitor domains. Here, we first provide a brief overview of current findings regarding the role of the MeA in social behaviors, followed by a deeper dive into current knowledge of how this complex structure is specified during development. We outline a conceptual model of MeA formation that has emerged based on these findings. We further postulate how embryonic developmental programming of the MeA may inform later emergence of stereotypical circuitry governing hardwired behaviors.
Collapse
Affiliation(s)
- Nandkishore Prakash
- Center for Neuroscience Research, Children's Research Institute, Children's National Hospital, Washington, DC, USA 20010
| | - Ameair Abu Irqeba
- Center for Neuroscience Research, Children's Research Institute, Children's National Hospital, Washington, DC, USA 20010
| | - Joshua G Corbin
- Center for Neuroscience Research, Children's Research Institute, Children's National Hospital, Washington, DC, USA 20010.
| |
Collapse
|
3
|
Morimoto K, Takahashi R, Takahashi G, Miyajima M, Nakajima K. Maternal immunoglobulins are distributed in the offspring's brain to support the maintenance of cortical interneurons in the postnatal period. Inflamm Regen 2024; 44:24. [PMID: 38750507 PMCID: PMC11094934 DOI: 10.1186/s41232-024-00336-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Accepted: 05/01/2024] [Indexed: 05/19/2024] Open
Abstract
It is known that maternal immunoglobulins (Igs) are transferred to the offspring across the placenta. However, receiving maternal Igs, especially before the blood-brain barrier (BBB) is formed in the offspring's brain, carries the risk of transferring some brain-reactive Igs. It is thus hypothesized that there may be some unknown benefit to the offspring's brain that overweighs this risk. In this study, we show that the Ig detected in the embryonic/perinatal mouse brain is IgG not produced by the pups themselves, but is basically transferred from the mother across the placenta using the neonatal Fc receptor (FcRn) during embryonic stages. The amount of IgG in the brain gradually decreases after birth, and almost disappears within 3 weeks postnatally. IgG is detected on axon bundles, microglia, and some meningeal cells, including border-associated macrophages (BAMs), endothelial cells, and fibroblasts. Using Fcer1g knock-out (KO) mice, we show that BAMs and microglia receive maternal IgG in an Fc receptor γ chain (FcRγ)-dependent manner, but IgG on other meningeal cells and axon bundles is received independently of the FcRγ. These results suggest that maternal IgG may be used in multiple ways by different mechanisms. In maternal IgG-deficient mice, the number of interneurons in the cerebral cortex is not altered around birth but is reduced postnatally, suggesting that receipt of maternal IgG is necessary for the maintenance of cortical interneurons in the postnatal period. These data suggest that maternal IgG has an important function in the developing brain, where neither obvious inflammation nor infection is observed.
Collapse
Affiliation(s)
- Keiko Morimoto
- Department of Anatomy, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-Ku, Tokyo, 160-8582, Japan.
| | - Rikuo Takahashi
- Department of Anatomy, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-Ku, Tokyo, 160-8582, Japan
| | - Goro Takahashi
- Department of Anatomy, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-Ku, Tokyo, 160-8582, Japan
| | - Michio Miyajima
- Department of Anatomy, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-Ku, Tokyo, 160-8582, Japan
| | - Kazunori Nakajima
- Department of Anatomy, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-Ku, Tokyo, 160-8582, Japan.
| |
Collapse
|
4
|
Morimoto K, Tabata H, Takahashi R, Nakajima K. Interactions between neural cells and blood vessels in central nervous system development. Bioessays 2024; 46:e2300091. [PMID: 38135890 DOI: 10.1002/bies.202300091] [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: 06/02/2023] [Revised: 08/28/2023] [Accepted: 12/08/2023] [Indexed: 12/24/2023]
Abstract
The sophisticated function of the central nervous system (CNS) is largely supported by proper interactions between neural cells and blood vessels. Accumulating evidence has demonstrated that neurons and glial cells support the formation of blood vessels, which in turn, act as migratory scaffolds for these cell types. Neural progenitors are also involved in the regulation of blood vessel formation. This mutual interaction between neural cells and blood vessels is elegantly controlled by several chemokines, growth factors, extracellular matrix, and adhesion molecules such as integrins. Recent research has revealed that newly migrating cell types along blood vessels repel other preexisting migrating cell types, causing them to detach from the blood vessels. In this review, we discuss vascular formation and cell migration, particularly during development. Moreover, we discuss how the crosstalk between blood vessels and neurons and glial cells could be related to neurodevelopmental disorders.
Collapse
Affiliation(s)
- Keiko Morimoto
- Department of Anatomy, Keio University School of Medicine, Tokyo, Japan
| | - Hidenori Tabata
- Department of Anatomy, Keio University School of Medicine, Tokyo, Japan
- Department of Molecular Neurobiology, Institute for Developmental Research, Aichi Developmental Disability Center, Kasugai, Japan
| | - Rikuo Takahashi
- Department of Anatomy, Keio University School of Medicine, Tokyo, Japan
| | - Kazunori Nakajima
- Department of Anatomy, Keio University School of Medicine, Tokyo, Japan
| |
Collapse
|
5
|
Shin M, Kosodo Y. Brain Sample Preparation After Performing in Utero Electroporation to Proliferating and Differentiated Cells in the Developing Mouse Neocortex. Methods Mol Biol 2024; 2794:187-200. [PMID: 38630230 DOI: 10.1007/978-1-0716-3810-1_16] [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] [Indexed: 04/19/2024]
Abstract
In utero electroporation (IUE) enables labeling and manipulating specific types of cells by introducing DNA plasmids with desired promoters. After the surgery, mouse brains are fixed at any stage and analyzed after staining using specific antibodies. Here, we describe the flow of the IUE experiment from the preparation to microscopic observations.
Collapse
Affiliation(s)
- Minkyung Shin
- Korea Brain Research Institute (KBRI), Daegu, Republic of Korea
| | - Yoichi Kosodo
- Korea Brain Research Institute (KBRI), Daegu, Republic of Korea.
- Department of Brain Sciences, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu, Republic of Korea.
| |
Collapse
|
6
|
Massri AJ, Fitzpatrick M, Cunny H, Li JL, Harry GJ. Differential gene expression profiling implicates altered network development in rat postnatal day 4 cortex following 4-Methylimidazole (4-MeI) induced maternal seizures. Neurotoxicol Teratol 2023; 100:107301. [PMID: 37783441 PMCID: PMC10843020 DOI: 10.1016/j.ntt.2023.107301] [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: 06/23/2023] [Revised: 08/31/2023] [Accepted: 09/19/2023] [Indexed: 10/04/2023]
Abstract
Compromised maternal health leading to maternal seizures can have adverse effects on the healthy development of offspring. This may be the result of inflammation, hypoxia-ischemia, and altered GABA signaling. The current study examined cortical tissue from F2b (2nd litter of the 2nd generation) postnatal day 4 (PND4) offspring of female Harlan SD rats chronically exposed to the seizuregenic compound, 4-Methylimidazole (0, 750, or 2500 ppm 4-MeI). Maternal seizures were evident only at 2500 ppm 4-MeI. GABA related gene expression as examined by qRT-PCR and whole genome microarray showed no indication of disrupted GABA or glutamatergic signaling. Canonical pathway hierarchical clustering and multi-omics combinatory genomic (CNet) plots of differentially expressed genes (DEG) showed alterations in genes associated with regulatory processes of cell development including neuronal differentiation and synaptogenesis. Functional enrichment analysis showed a similarity of cellular processes across the two exposure groups however, the genes comprising each cluster were primarily unique rather than shared and often showed different directionality. A dose-related induction of cytokine signaling was indicated however, pathways associated with individual cytokine signaling were not elevated, suggesting an alternative involvement of cytokine signaling. Pathways related to growth process and cell signaling showed a negative activation supporting an interpretation of disruption or delay in developmental processes at the 2500 ppm 4-MeI exposure level with maternal seizures. Thus, while GABA signaling was not altered as has been observed with maternal seizures, the pattern of DEG suggested a potential for alteration in neuronal network formation.
Collapse
Affiliation(s)
- Abdull J Massri
- Integrative Bioinformatics, Division of Intramural Research, National Institute of Environmental Health Sciences, National Institutes of Health, 111 T. W. Alexander Drive, Research Triangle Park, NC 27709, USA
| | - Mackenzie Fitzpatrick
- Mechanistic Toxicology Branch, National Institute of Environmental Health Sciences, National Institutes of Health, 111 T. W. Alexander Drive, Research Triangle Park, NC 27709, USA
| | - Helen Cunny
- Office of the Scientific Director, Division of Translational Toxicology, National Institute of Environmental Health Sciences, National Institutes of Health, 111 T. W. Alexander Drive, Research Triangle Park, NC 27709, USA
| | - Jian-Liang Li
- Integrative Bioinformatics, Division of Intramural Research, National Institute of Environmental Health Sciences, National Institutes of Health, 111 T. W. Alexander Drive, Research Triangle Park, NC 27709, USA
| | - G Jean Harry
- Mechanistic Toxicology Branch, National Institute of Environmental Health Sciences, National Institutes of Health, 111 T. W. Alexander Drive, Research Triangle Park, NC 27709, USA.
| |
Collapse
|
7
|
Verlinden L, Doms S, Janssens I, Meyer MB, Pike JW, Carmeliet G, Verstuyf A. Neuropilin 2 in osteoblasts regulates trabecular bone mass in male mice. Front Endocrinol (Lausanne) 2023; 14:1223021. [PMID: 37600714 PMCID: PMC10436209 DOI: 10.3389/fendo.2023.1223021] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Accepted: 07/11/2023] [Indexed: 08/22/2023] Open
Abstract
Introduction Neuropilin 2 (NRP2) mediates the effects of class 3 semaphorins and vascular endothelial growth factor and is implicated in axonal guidance and angiogenesis. Moreover, NRP2 expression is suggested to be involved in the regulation of bone homeostasis. Indeed, osteoblasts and osteoclasts express NRP2 and male and female global Nrp2 knockout mice have a reduced bone mass accompanied by reduced osteoblast and increased osteoclast counts. Methods We first examined the in vitro effect of the calciotropic hormone 1,25-dihydroxyvitamin D3 [1,25(OH)2D3] on Nrp2 transcription in osteoblasts. We next generated mice with a conditional deletion of Nrp2 in the osteoblast cell lineage under control of the paired related homeobox 1 promoter and mice with a conditional Nrp2 knockdown in osteoclasts under control of the Lysozyme promoter. Mice were examined under basal conditions or after treatment with either the bone anabolic vitamin D3 analog WY 1048 or with 1,25(OH)2D3. Results and discussion We show that Nrp2 expression is induced by 1,25(OH)2D3 in osteoblasts and is associated with enrichment of the vitamin D receptor in an intronic region of the Nrp2 gene. In male mice, conditional deletion of Nrp2 in osteoblast precursors and mature osteoblasts recapitulated the bone phenotype of global Nrp2 knockout mice, with a reduced cortical cross-sectional tissue area and lower trabecular bone content. However, female mice with reduced osteoblastic Nrp2 expression display a reduced cross-sectional tissue area but have a normal trabecular bone mass. Treatment with the vitamin D3 analog WY 1048 (0.4 μg/kg/d, 14 days, ip) resulted in a similar increase in bone mass in both genotypes and genders. Deleting Nrp2 from the osteoclast lineage did not result in a bone phenotype, even though in vitro osteoclastogenesis of hematopoietic cells derived from mutant mice was significantly increased. Moreover, treatment with a high dose of 1,25(OH)2D3 (0.5 μg/kg/d, 6 days, ip), to induce osteoclast-mediated bone resorption, resulted in a similar reduction in trabecular and cortical bone mass. In conclusion, osteoblastic Nrp2 expression is suggested to regulate bone homeostasis in a sex-specific manner.
Collapse
Affiliation(s)
- Lieve Verlinden
- Department of Chronic Diseases and Metabolism, Clinical and Experimental Endocrinology, KU Leuven, Leuven, Belgium
| | - Stefanie Doms
- Department of Chronic Diseases and Metabolism, Clinical and Experimental Endocrinology, KU Leuven, Leuven, Belgium
| | - Iris Janssens
- Department of Chronic Diseases and Metabolism, Clinical and Experimental Endocrinology, KU Leuven, Leuven, Belgium
| | - Mark B. Meyer
- Department of Nutritional Sciences, University of Wisconsin-Madision, Madison, WI, United States
| | - J. Wesley Pike
- Department of Biochemistry, University of Wisconsin-Madision, Madison, WI, United States
| | - Geert Carmeliet
- Department of Chronic Diseases and Metabolism, Clinical and Experimental Endocrinology, KU Leuven, Leuven, Belgium
| | - Annemieke Verstuyf
- Department of Chronic Diseases and Metabolism, Clinical and Experimental Endocrinology, KU Leuven, Leuven, Belgium
| |
Collapse
|
8
|
You G, Li W, Wang Y, Cao H, Li X, Gao L, Zheng SJ. Reduced NR2F2 Expression in the Host Response to Infectious Bursal Disease Virus Infection Suppressed Viral Replication by Enhancing Type I Interferon Expression by Targeting SOCS5. J Virol 2023; 97:e0066423. [PMID: 37358466 PMCID: PMC10373545 DOI: 10.1128/jvi.00664-23] [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: 05/04/2023] [Accepted: 06/01/2023] [Indexed: 06/27/2023] Open
Abstract
Nuclear receptors are ligand-activated transcription factors that play an important role in regulating innate antiviral immunity and other biological processes. However, the role of nuclear receptors in the host response to infectious bursal disease virus (IBDV) infection remains elusive. In this study, we show that IBDV infection or poly(I·C) treatment of DF-1 or HD11 cells markedly decreased nuclear receptor subfamily 2 group F member 2 (NR2F2) expression. Surprisingly, knockdown, knockout, or inhibition of NR2F2 expression in host cells remarkably inhibited IBDV replication and promoted IBDV/poly(I·C)-induced type I interferon and interferon-stimulated genes expression. Furthermore, our data show that NR2F2 negatively regulates the antiviral innate immune response by promoting the suppressor of cytokine signaling 5 (SOCS5) expression. Thus, reduced NR2F2 expression in the host response to IBDV infection inhibited viral replication by enhancing the expression of type I interferon by targeting SOCS5. These findings reveal that NR2F2 plays a crucial role in antiviral innate immunity, furthering our understanding of the mechanism underlying the host response to viral infection. IMPORTANCE Infectious bursal disease (IBD) is an immunosuppressive disease causing considerable economic losses to the poultry industry worldwide. Nuclear receptors play an important role in regulating innate antiviral immunity. However, the role of nuclear receptors in the host response to IBD virus (IBDV) infection remains elusive. Here, we report that NR2F2 expression decreased in IBDV-infected cells, which consequently reduced SOCS5 expression, promoted type I interferon expression, and suppressed IBDV infection. Thus, NR2F2 serves as a negative factor in the host response to IBDV infection by regulating SOCS5 expression, and intervention in the NR2F2-mediated host response by specific inhibitors might be employed as a strategy for prevention and treatment of IBD.
Collapse
Affiliation(s)
- Guangju You
- National Key Laboratory of Veterinary Public Health Security, College of Veterinary Medicine, China Agricultural University, Beijing, China
- Animal Epidemiology of the Ministry of Agriculture, College of Veterinary Medicine, China Agricultural University, Beijing, China
- College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Wei Li
- National Key Laboratory of Veterinary Public Health Security, College of Veterinary Medicine, China Agricultural University, Beijing, China
- Animal Epidemiology of the Ministry of Agriculture, College of Veterinary Medicine, China Agricultural University, Beijing, China
- College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Yongqiang Wang
- National Key Laboratory of Veterinary Public Health Security, College of Veterinary Medicine, China Agricultural University, Beijing, China
- Animal Epidemiology of the Ministry of Agriculture, College of Veterinary Medicine, China Agricultural University, Beijing, China
- College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Hong Cao
- National Key Laboratory of Veterinary Public Health Security, College of Veterinary Medicine, China Agricultural University, Beijing, China
- Animal Epidemiology of the Ministry of Agriculture, College of Veterinary Medicine, China Agricultural University, Beijing, China
- College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Xiaoqi Li
- College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Li Gao
- National Key Laboratory of Veterinary Public Health Security, College of Veterinary Medicine, China Agricultural University, Beijing, China
- Animal Epidemiology of the Ministry of Agriculture, College of Veterinary Medicine, China Agricultural University, Beijing, China
- College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Shijun J. Zheng
- National Key Laboratory of Veterinary Public Health Security, College of Veterinary Medicine, China Agricultural University, Beijing, China
- Animal Epidemiology of the Ministry of Agriculture, College of Veterinary Medicine, China Agricultural University, Beijing, China
- College of Veterinary Medicine, China Agricultural University, Beijing, China
| |
Collapse
|
9
|
Blair JD, Hartman A, Zenk F, Dalgarno C, Treutlein B, Satija R. Phospho-seq: Integrated, multi-modal profiling of intracellular protein dynamics in single cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.27.534442. [PMID: 37034703 PMCID: PMC10081255 DOI: 10.1101/2023.03.27.534442] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 04/17/2023]
Abstract
Cell signaling plays a critical role in regulating cellular behavior and fate. While multimodal single-cell sequencing technologies are rapidly advancing, scalable and flexible profiling of cell signaling states alongside other molecular modalities remains challenging. Here we present Phospho-seq, an integrated approach that aims to quantify phosphorylated intracellular and intranuclear proteins, and to connect their activity with cis-regulatory elements and transcriptional targets. We utilize a simplified benchtop antibody conjugation method to create large custom antibody panels for simultaneous protein and scATAC-seq profiling on whole cells, and integrate this information with scRNA-seq datasets via bridge integration. We apply our workflow to cell lines, induced pluripotent stem cells, and 3-month-old brain organoids to demonstrate its broad applicability. We demonstrate that Phospho-seq can define cellular states and trajectories, reconstruct gene regulatory relationships, and characterize the causes and consequences of heterogeneous cell signaling in neurodevelopment.
Collapse
Affiliation(s)
- John D. Blair
- New York Genome Center, New York, NY
- New York University, Center for Genomics and Systems Biology, New York, NY
| | | | | | | | | | - Rahul Satija
- New York Genome Center, New York, NY
- New York University, Center for Genomics and Systems Biology, New York, NY
| |
Collapse
|
10
|
A Unique "Reversed" Migration of Neurons in the Developing Claustrum. J Neurosci 2023; 43:693-708. [PMID: 36631266 PMCID: PMC9899091 DOI: 10.1523/jneurosci.0704-22.2022] [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: 04/07/2022] [Revised: 10/24/2022] [Accepted: 12/10/2022] [Indexed: 01/13/2023] Open
Abstract
The claustrum (CLA) is a cluster of neurons located between the insular cortex and striatum. Many studies have shown that the CLA plays an important role in higher brain function. Additionally, growing evidence suggests that CLA dysfunction is associated with neuropsychological symptoms. However, how the CLA is formed during development is not fully understood. In the present study, we analyzed the development of the CLA, especially focusing on the migration profiles of CLA neurons in mice of both sexes. First, we showed that CLA neurons were generated between embryonic day (E) 10.5 and E12.5, but mostly at E11.5. Next, we labeled CLA neurons born at E11.5 using the FlashTag technology and revealed that most neurons reached the brain surface by E13.5 but were distributed deep in the CLA 1 d later at E14.5. Time-lapse imaging of GFP-labeled cells revealed that some CLA neurons first migrated radially outward and then changed their direction inward after reaching the surface. Moreover, we demonstrated that Reelin signal is necessary for the appropriate distribution of CLA neurons. The switch from outward to "reversed" migration of developing CLA neurons is distinct from other migration modes, in which neurons typically migrate in a certain direction, which is simply outward or inward. Future elucidation of the characteristics and precise molecular mechanisms of CLA development may provide insights into the unique cognitive functions of the CLA.SIGNIFICANCE STATEMENT The claustrum (CLA) plays an important role in higher brain function, and its dysfunction is associated with neuropsychological symptoms. Although psychiatric disorders are increasingly being understood as disorders of neurodevelopment, little is known about CLA development, including its neuronal migration profiles and underlying molecular mechanisms. Here, we investigated the migration profiles of CLA neurons during development and found that they migrated radially outward and then inward after reaching the surface. This switch in the migratory direction from outward to inward may be one of the brain's fundamental mechanisms of nuclear formation. Our findings enable us to investigate the relationship between CLA maldevelopment and dysfunction, which may facilitate understanding of the pathogenesis of some psychiatric disorders.
Collapse
|
11
|
Wang M, Yang Y, Xu Y. Brain nuclear receptors and cardiovascular function. Cell Biosci 2023; 13:14. [PMID: 36670468 PMCID: PMC9854230 DOI: 10.1186/s13578-023-00962-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Accepted: 01/12/2023] [Indexed: 01/22/2023] Open
Abstract
Brain-heart interaction has raised up increasing attentions. Nuclear receptors (NRs) are abundantly expressed in the brain, and emerging evidence indicates that a number of these brain NRs regulate multiple aspects of cardiovascular diseases (CVDs), including hypertension, heart failure, atherosclerosis, etc. In this review, we will elaborate recent findings that have established the physiological relevance of brain NRs in the context of cardiovascular function. In addition, we will discuss the currently available evidence regarding the distinct neuronal populations that respond to brain NRs in the cardiovascular control. These findings suggest connections between cardiac control and brain dynamics through NR signaling, which may lead to novel tools for the treatment of pathological changes in the CVDs.
Collapse
Affiliation(s)
- Mengjie Wang
- Department of Pediatrics, USDA/ARS Children’s Nutrition Research Center, Baylor College of Medicine, Houston, TX USA
| | - Yongjie Yang
- Department of Pediatrics, USDA/ARS Children’s Nutrition Research Center, Baylor College of Medicine, Houston, TX USA
| | - Yong Xu
- Department of Pediatrics, USDA/ARS Children’s Nutrition Research Center, Baylor College of Medicine, Houston, TX USA
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX USA
| |
Collapse
|
12
|
Dinh TT, Xiang M, Rajaraman A, Wang Y, Salazar N, Zhu Y, Roper W, Rhee S, Brulois K, O'Hara E, Kiefel H, Dinh TM, Bi Y, Gonzalez D, Bao EP, Red-Horse K, Balogh P, Gábris F, Gaszner B, Berta G, Pan J, Butcher EC. An NKX-COUP-TFII morphogenetic code directs mucosal endothelial addressin expression. Nat Commun 2022; 13:7448. [PMID: 36460642 PMCID: PMC9718832 DOI: 10.1038/s41467-022-34991-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Accepted: 11/14/2022] [Indexed: 12/03/2022] Open
Abstract
Immunoglobulin family and carbohydrate vascular addressins encoded by Madcam1 and St6gal1 control lymphocyte homing into intestinal tissues, regulating immunity and inflammation. The addressins are developmentally programmed to decorate endothelial cells lining gut post-capillary and high endothelial venules (HEV), providing a prototypical example of organ- and segment-specific endothelial specialization. We identify conserved NKX-COUP-TFII composite elements (NCCE) in regulatory regions of Madcam1 and St6gal1 that bind intestinal homeodomain protein NKX2-3 cooperatively with venous nuclear receptor COUP-TFII to activate transcription. The Madcam1 element also integrates repressive signals from arterial/capillary Notch effectors. Pan-endothelial COUP-TFII overexpression induces ectopic addressin expression in NKX2-3+ capillaries, while NKX2-3 deficiency abrogates expression by HEV. Phylogenetically conserved NCCE are enriched in genes involved in neuron migration and morphogenesis of the heart, kidney, pancreas and other organs. Our results define an NKX-COUP-TFII morphogenetic code that targets expression of mucosal vascular addressins.
Collapse
Affiliation(s)
- Thanh Theresa Dinh
- Laboratory of Immunology and Vascular Biology, Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
- Palo Alto Veterans Institute for Research, Palo Alto, CA, USA
| | - Menglan Xiang
- Laboratory of Immunology and Vascular Biology, Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
- Palo Alto Veterans Institute for Research, Palo Alto, CA, USA
| | - Anusha Rajaraman
- Laboratory of Immunology and Vascular Biology, Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
- Palo Alto Veterans Institute for Research, Palo Alto, CA, USA
- Department of Molecular Cell Biology and Immunology, Vrije Universiteit Medical Center, Amsterdam, The Netherlands
| | - Yongzhi Wang
- Laboratory of Immunology and Vascular Biology, Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
- Department of Clinical Science Malmo, Section of Surgery, Lund University, Malmo, Sweden
| | - Nicole Salazar
- Laboratory of Immunology and Vascular Biology, Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Yu Zhu
- Laboratory of Immunology and Vascular Biology, Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Walter Roper
- Columbia University Vagelos College of Physicians and Surgeons, New York City, NY, USA
| | - Siyeon Rhee
- Department of Biology, Stanford University, Stanford, CA, USA
| | - Kevin Brulois
- Laboratory of Immunology and Vascular Biology, Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
- Palo Alto Veterans Institute for Research, Palo Alto, CA, USA
| | - Ed O'Hara
- Palo Alto Veterans Institute for Research, Palo Alto, CA, USA
| | - Helena Kiefel
- Laboratory of Immunology and Vascular Biology, Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Truc M Dinh
- Palo Alto Veterans Institute for Research, Palo Alto, CA, USA
| | - Yuhan Bi
- Laboratory of Immunology and Vascular Biology, Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
- Palo Alto Veterans Institute for Research, Palo Alto, CA, USA
| | | | - Evan P Bao
- Palo Alto Veterans Institute for Research, Palo Alto, CA, USA
| | - Kristy Red-Horse
- Department of Biology, Stanford University, Stanford, CA, USA
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Howard Hughes Medical Institute, Stanford, CA, USA
| | - Peter Balogh
- Department of Immunology and Biotechnology, University of Pécs Medical School, Pécs, Hungary
- Lymphoid Organogenesis Research Team, Szentágothai Research Center, Pécs, Hungary
| | - Fanni Gábris
- Department of Immunology and Biotechnology, University of Pécs Medical School, Pécs, Hungary
- Lymphoid Organogenesis Research Team, Szentágothai Research Center, Pécs, Hungary
| | - Balázs Gaszner
- Department of Anatomy, University of Pécs Medical School, Pécs, Hungary
| | - Gergely Berta
- Department of Medical Biology and Central Electron Microscopy Laboratory, University of Pécs Medical School, Pécs, Hungary
| | - Junliang Pan
- Palo Alto Veterans Institute for Research, Palo Alto, CA, USA.
- The Center for Molecular Biology and Medicine, Veterans Affairs Palo Alto Health Care System, Palo Alto, CA, USA.
| | - Eugene C Butcher
- Laboratory of Immunology and Vascular Biology, Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA.
- Palo Alto Veterans Institute for Research, Palo Alto, CA, USA.
- The Center for Molecular Biology and Medicine, Veterans Affairs Palo Alto Health Care System, Palo Alto, CA, USA.
| |
Collapse
|
13
|
Hazra D, Yoshinaga S, Yoshida K, Takata N, Tanaka KF, Kubo KI, Nakajima K. Rhythmic activation of excitatory neurons in the mouse frontal cortex improves the prefrontal cortex-mediated cognitive function. Cereb Cortex 2022; 32:5243-5258. [PMID: 35136976 DOI: 10.1093/cercor/bhac011] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Revised: 01/07/2022] [Accepted: 01/08/2022] [Indexed: 12/27/2022] Open
Abstract
The prefrontal cortex (PFC) plays essential roles in cognitive processes. Previous studies have suggested the layer and the cell type-specific activation for cognitive enhancement. However, the mechanism by which a temporal pattern of activation affects cognitive function remains to be elucidated. Here, we investigated whether the specific activation of excitatory neurons in the superficial layers mainly in the PFC according to a rhythmic or nonrhythmic pattern could modulate the cognitive functions of normal mice. We used a C128S mutant of channelrhodopsin 2, a step function opsin, and administered two light illumination patterns: (i) alternating pulses of blue and yellow light for rhythmic activation or (ii) pulsed blue light only for nonrhythmic activation. Behavioral analyses were performed to compare the behavioral consequences of these two neural activation patterns. The alternating blue and yellow light pulses, but not the pulsed blue light only, significantly improved spatial working memory and social recognition without affecting motor activity or the anxiety level. These results suggest that the rhythmic, but not the nonrhythmic, activation could enhance cognitive functions. This study indicates that not only the population of neurons that are activated but also the pattern of activation plays a crucial role in the cognitive enhancement.
Collapse
Affiliation(s)
- Debabrata Hazra
- Department of Anatomy, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Satoshi Yoshinaga
- Department of Anatomy, Keio University School of Medicine, Tokyo 160-8582, Japan.,Department of Anatomy, The Jikei University School of Medicine, Tokyo 105-8461, Japan
| | - Keitaro Yoshida
- Department of Neuropsychiatry, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Norio Takata
- Department of Neuropsychiatry, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Kenji F Tanaka
- Department of Neuropsychiatry, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Ken-Ichiro Kubo
- Department of Anatomy, Keio University School of Medicine, Tokyo 160-8582, Japan.,Department of Anatomy, The Jikei University School of Medicine, Tokyo 105-8461, Japan
| | - Kazunori Nakajima
- Department of Anatomy, Keio University School of Medicine, Tokyo 160-8582, Japan
| |
Collapse
|
14
|
KIF5C deficiency causes abnormal cortical neuronal migration, dendritic branching, and spine morphology in mice. Pediatr Res 2022; 92:995-1002. [PMID: 34966180 DOI: 10.1038/s41390-021-01922-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Revised: 11/18/2021] [Accepted: 12/13/2021] [Indexed: 11/08/2022]
Abstract
BACKGROUND Malformation of cortical development (MCD) includes a variety of developmental disorders that are common causes of neurodevelopmental delay and epilepsy. Most recently, clinical studies found that patients carrying KIF5C mutations present early-onset MCD; however, the underlying mechanisms remain elusive. METHODS KIF5C expression level was examined in mouse primary cortical neurons and human ips-derived forebrain organoids. We studied the cortical neuronal migration, dendritic branching, and dendritic spine growth after knocking down the KIF5C gene by electroporation in vitro and in vivo. Then, we studied the transcriptome differences between the knockdown and control groups through RNA sequencing. RESULTS We observed high KIF5C expression in neurons during the early developmental stage in mice and the human brain. Kif5c deficiency results in disturbed cortical neuronal migration, dendritic, and spine growth. Finally, we found that Kif5c knockdown affected several genes associated with cortical neuronal development in vitro. CONCLUSIONS These results suggested a critical role for Kif5c in cortical development, providing insights into underlying pathogenic factors of kinesins in MCD. IMPACT KIF5C mutation-related MCD might be caused by abnormal early cortical neuronal development. Kif5c deficiency led to abnormal cortical neuronal dendritic and spine growth and neuronal migration. Our findings explain how Kif5c deficiency is involved in the aberrant development of cortical neurons and provide a new perspective for the pathology of MCD.
Collapse
|
15
|
Su K, Hao W, Lv Z, Wu M, Li J, Hu Y, Zhang Z, Gao J, Feng X. Electroacupuncture of Baihui and Shenting ameliorates cognitive deficits via Pten/Akt pathway in a rat cerebral ischemia injury model. Front Neurol 2022; 13:855362. [PMID: 36062010 PMCID: PMC9437581 DOI: 10.3389/fneur.2022.855362] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2022] [Accepted: 07/11/2022] [Indexed: 11/23/2022] Open
Abstract
Cerebral ischemic stroke is a huge threat to the health and life of many people. Electroacupuncture (EA) at Baihui (GV20) and Shenting (GV24) acupoints can notably alleviate cerebral ischemia/reperfusion injury (CIRI). However, the molecular basis underlying the effectiveness of EA at the GV20 and GV24 acupoints for CIRI remains largely unknown. Our present study demonstrated that EA treatment at the GV20 and GV24 acupoints markedly alleviated middle cerebral artery occlusion/reperfusion (MCAO/R)-induced cognitive deficits and cerebral infarction in rats. Proteomics analysis revealed that 195 and 218 proteins were dysregulated in rat hippocampal tissues in the MCAO/R vs. sham group and thhhe EA vs. MCAO/R group, respectively. Moreover, 62 proteins with converse alteration trends in MCAO/R vs. sham and EA vs. MCAO/R groups were identified. These proteins might be implicated in the EA-mediated protective effect against MCAO/R-induced cerebral injury. GO enrichment analysis showed that 39 dysregulated proteins in the MCAO/R vs. sham group and 40 dysregulated proteins in the EA vs. MCAO/R group were related to brain and nerve development. Protein–protein interaction analysis of the abovementioned dysregulated proteins associated with brain and nerve development suggested that Pten/Akt pathway-related proteins might play major roles in regulating EA-mediated protective effects against MCAO/R-induced brain and nerve injury. Western blot assays demonstrated that Pak4, Akt3, and Efnb2 were expressed at low levels in the MCAO/R group vs. the sham group but at high levels in the EA group vs. the MCAO/R group. In conclusion, multiple proteins related to the protective effect of EA at the GV20 and GV24 acupoints against CIRI were identified in our study.
Collapse
Affiliation(s)
- Kaiqi Su
- Department of Rehabilitation Medicine, Henan University of Chinese Medicine, Zhengzhou, China
- Rehabilitation Center, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, China
| | - Wenxue Hao
- Department of Rehabilitation, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Zhuan Lv
- Department of Rehabilitation Medicine, Henan University of Chinese Medicine, Zhengzhou, China
- Rehabilitation Center, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, China
| | - Mingli Wu
- Department of Rehabilitation Medicine, Henan University of Chinese Medicine, Zhengzhou, China
- Rehabilitation Center, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, China
| | - Jieying Li
- College of Basic Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| | - Yanchao Hu
- Rehabilitation Center, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, China
| | - Zhenhua Zhang
- Rehabilitation Center, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, China
| | - Jing Gao
- Department of Rehabilitation Medicine, Henan University of Chinese Medicine, Zhengzhou, China
- Rehabilitation Center, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, China
- *Correspondence: Jing Gao
| | - Xiaodong Feng
- Department of Rehabilitation Medicine, Henan University of Chinese Medicine, Zhengzhou, China
- Rehabilitation Center, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, China
- Xiaodong Feng
| |
Collapse
|
16
|
COUP-TFII in Kidneys, from Embryos to Sick Adults. Diagnostics (Basel) 2022; 12:diagnostics12051181. [PMID: 35626336 PMCID: PMC9139597 DOI: 10.3390/diagnostics12051181] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Revised: 05/02/2022] [Accepted: 05/06/2022] [Indexed: 11/16/2022] Open
Abstract
Chicken ovalbumin upstream promoter-transcription factor II (COUP-TFII) is an orphan nuclear hormone receptor of unknown ligands. This molecule has two interesting features: (1) it is a developmental gene, and (2) it is a potential hormone receptor. Here, we describe the possible roles of COUP-TFII in the organogenesis of the kidneys and protection from adult renal diseases, primarily in mouse models. COUP-TFII is highly expressed in embryos, including primordial kidneys, and is essential for the formation of metanephric mesenchyme and the survival of renal precursor cells. Although the expression levels of COUP-TFII are low and its functions are unknown in healthy adults, it serves as a reno-protectant molecule against acute kidney injury. These are good examples of how developmental genes exhibit novel functions in the etiology of adult diseases. We also discuss the ongoing research on the roles of COUP-TFII in podocyte development and diabetic kidney disease. In addition, the identification of potential ligands suggests that COUP-TFII might be a novel therapeutic target for renal diseases in the future.
Collapse
|
17
|
Leung RF, George AM, Roussel EM, Faux MC, Wigle JT, Eisenstat DD. Genetic Regulation of Vertebrate Forebrain Development by Homeobox Genes. Front Neurosci 2022; 16:843794. [PMID: 35546872 PMCID: PMC9081933 DOI: 10.3389/fnins.2022.843794] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Accepted: 03/14/2022] [Indexed: 01/19/2023] Open
Abstract
Forebrain development in vertebrates is regulated by transcription factors encoded by homeobox, bHLH and forkhead gene families throughout the progressive and overlapping stages of neural induction and patterning, regional specification and generation of neurons and glia from central nervous system (CNS) progenitor cells. Moreover, cell fate decisions, differentiation and migration of these committed CNS progenitors are controlled by the gene regulatory networks that are regulated by various homeodomain-containing transcription factors, including but not limited to those of the Pax (paired), Nkx, Otx (orthodenticle), Gsx/Gsh (genetic screened), and Dlx (distal-less) homeobox gene families. This comprehensive review outlines the integral role of key homeobox transcription factors and their target genes on forebrain development, focused primarily on the telencephalon. Furthermore, links of these transcription factors to human diseases, such as neurodevelopmental disorders and brain tumors are provided.
Collapse
Affiliation(s)
- Ryan F. Leung
- Murdoch Children’s Research Institute, The Royal Children’s Hospital Melbourne, Parkville, VIC, Australia
- Department of Paediatrics, University of Melbourne, Parkville, VIC, Australia
| | - Ankita M. George
- Murdoch Children’s Research Institute, The Royal Children’s Hospital Melbourne, Parkville, VIC, Australia
| | - Enola M. Roussel
- Murdoch Children’s Research Institute, The Royal Children’s Hospital Melbourne, Parkville, VIC, Australia
| | - Maree C. Faux
- Murdoch Children’s Research Institute, The Royal Children’s Hospital Melbourne, Parkville, VIC, Australia
- Department of Paediatrics, University of Melbourne, Parkville, VIC, Australia
- Department of Surgery, Royal Melbourne Hospital, The University of Melbourne, Parkville, VIC, Australia
| | - Jeffrey T. Wigle
- Department of Biochemistry and Medical Genetics, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada
- Institute of Cardiovascular Sciences, St. Boniface Hospital Albrechtsen Research Centre, Winnipeg, MB, Canada
| | - David D. Eisenstat
- Murdoch Children’s Research Institute, The Royal Children’s Hospital Melbourne, Parkville, VIC, Australia
- Department of Paediatrics, University of Melbourne, Parkville, VIC, Australia
- Department of Medical Genetics, University of Alberta, Edmonton, AB, Canada
- Department of Pediatrics, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
18
|
Schmidbaur H, Kawaguchi A, Clarence T, Fu X, Hoang OP, Zimmermann B, Ritschard EA, Weissenbacher A, Foster JS, Nyholm SV, Bates PA, Albertin CB, Tanaka E, Simakov O. Emergence of novel cephalopod gene regulation and expression through large-scale genome reorganization. Nat Commun 2022; 13:2172. [PMID: 35449136 PMCID: PMC9023564 DOI: 10.1038/s41467-022-29694-7] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Accepted: 03/28/2022] [Indexed: 12/17/2022] Open
Abstract
Coleoid cephalopods (squid, cuttlefish, octopus) have the largest nervous system among invertebrates that together with many lineage-specific morphological traits enables complex behaviors. The genomic basis underlying these innovations remains unknown. Using comparative and functional genomics in the model squid Euprymna scolopes, we reveal the unique genomic, topological, and regulatory organization of cephalopod genomes. We show that coleoid cephalopod genomes have been extensively restructured compared to other animals, leading to the emergence of hundreds of tightly linked and evolutionary unique gene clusters (microsyntenies). Such novel microsyntenies correspond to topological compartments with a distinct regulatory structure and contribute to complex expression patterns. In particular, we identify a set of microsyntenies associated with cephalopod innovations (MACIs) broadly enriched in cephalopod nervous system expression. We posit that the emergence of MACIs was instrumental to cephalopod nervous system evolution and propose that microsyntenic profiling will be central to understanding cephalopod innovations.
Collapse
Affiliation(s)
- Hannah Schmidbaur
- Department of Neurosciences and Developmental Biology, University of Vienna, Vienna, Austria
| | | | - Tereza Clarence
- Biomolecular Modelling Laboratory, The Francis Crick Institute, London, UK
| | - Xiao Fu
- Biomolecular Modelling Laboratory, The Francis Crick Institute, London, UK
| | - Oi Pui Hoang
- Department of Neurosciences and Developmental Biology, University of Vienna, Vienna, Austria
| | - Bob Zimmermann
- Department of Neurosciences and Developmental Biology, University of Vienna, Vienna, Austria
| | - Elena A Ritschard
- Department of Neurosciences and Developmental Biology, University of Vienna, Vienna, Austria
- Department of Biology and Evolution of Marine Organisms, Stazione Zoologica Anton Dohrn, Naples, Italy
| | | | - Jamie S Foster
- Department of Microbiology and Cell Science, University of Florida, Space Life Science Lab, Merritt Island, FL, USA
| | - Spencer V Nyholm
- Department of Molecular and Cell Biology, University of Connecticut, Storrs, CT, USA
| | - Paul A Bates
- Biomolecular Modelling Laboratory, The Francis Crick Institute, London, UK
| | - Caroline B Albertin
- Bell Center for Regenerative Biology and Tissue Engineering, Marine Biological Laboratory, Woods Hole, MA, USA.
| | - Elly Tanaka
- Institute for Molecular Pathology, Vienna, Austria.
| | - Oleg Simakov
- Department of Neurosciences and Developmental Biology, University of Vienna, Vienna, Austria.
| |
Collapse
|
19
|
Aerts T, Seuntjens E. Novel Perspectives on the Development of the Amygdala in Rodents. Front Neuroanat 2021; 15:786679. [PMID: 34955766 PMCID: PMC8696165 DOI: 10.3389/fnana.2021.786679] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Accepted: 11/10/2021] [Indexed: 12/14/2022] Open
Abstract
The amygdala is a hyperspecialized brain region composed of strongly inter- and intraconnected nuclei involved in emotional learning and behavior. The cellular heterogeneity of the amygdalar nuclei has complicated straightforward conclusions on their developmental origin, and even resulted in contradictory data. Recently, the concentric ring theory of the pallium and the radial histogenetic model of the pallial amygdala have cleared up several uncertainties that plagued previous models of amygdalar development. Here, we provide an extensive overview on the developmental origin of the nuclei of the amygdaloid complex. Starting from older gene expression data, transplantation and lineage tracing studies, we systematically summarize and reinterpret previous findings in light of the novel perspectives on amygdalar development. In addition, migratory routes that these cells take on their way to the amygdala are explored, and known transcription factors and guidance cues that seemingly drive these cells toward the amygdala are emphasized. We propose some future directions for research on amygdalar development and highlight that a better understanding of its development could prove critical for the treatment of several neurodevelopmental and neuropsychiatric disorders.
Collapse
Affiliation(s)
- Tania Aerts
- Laboratory of Developmental Neurobiology, Department of Biology, KU Leuven, Leuven, Belgium
| | - Eve Seuntjens
- Laboratory of Developmental Neurobiology, Department of Biology, KU Leuven, Leuven, Belgium
| |
Collapse
|
20
|
Wang T, Wang Z, de Fabritus L, Tao J, Saied EM, Lee HJ, Ramazanov BR, Jackson B, Burkhardt D, Parker M, Gleinich AS, Wang Z, Seo DE, Zhou T, Xu S, Alecu I, Azadi P, Arenz C, Hornemann T, Krishnaswamy S, van de Pavert SA, Kaech SM, Ivanova NB, Santori FR. 1-deoxysphingolipids bind to COUP-TF to modulate lymphatic and cardiac cell development. Dev Cell 2021; 56:3128-3145.e15. [PMID: 34762852 PMCID: PMC8628544 DOI: 10.1016/j.devcel.2021.10.018] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2020] [Revised: 08/30/2021] [Accepted: 10/19/2021] [Indexed: 12/12/2022]
Abstract
Identification of physiological modulators of nuclear hormone receptor (NHR) activity is paramount for understanding the link between metabolism and transcriptional networks that orchestrate development and cellular physiology. Using libraries of metabolic enzymes alongside their substrates and products, we identify 1-deoxysphingosines as modulators of the activity of NR2F1 and 2 (COUP-TFs), which are orphan NHRs that are critical for development of the nervous system, heart, veins, and lymphatic vessels. We show that these non-canonical alanine-based sphingolipids bind to the NR2F1/2 ligand-binding domains (LBDs) and modulate their transcriptional activity in cell-based assays at physiological concentrations. Furthermore, inhibition of sphingolipid biosynthesis phenocopies NR2F1/2 deficiency in endothelium and cardiomyocytes, and increases in 1-deoxysphingosine levels activate NR2F1/2-dependent differentiation programs. Our findings suggest that 1-deoxysphingosines are physiological regulators of NR2F1/2-mediated transcription.
Collapse
Affiliation(s)
- Ting Wang
- Department of Immunobiology, Yale University, New Haven, CT 06520, USA; Department of Hematology, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Zheng Wang
- Department of Genetics and Cell Biology, Basic Medical College, Qingdao University, Qingdao, Shandong 266071, China; Department of Reproductive Medicine, the Affiliated Hospital of Qingdao University, Qingdao, Shandong 266000, China
| | - Lauriane de Fabritus
- Aix-Marseille Universite, CNRS, INSERM, Centre d'Immunologie de Marseille-Luminy (CIML), 13288 Marseille Cedex 9, France
| | - Jinglian Tao
- Department of Hematology, Tianjin Medical University General Hospital, Tianjin 300052, China; Center for Molecular Medicine, Department of Genetics, University of Georgia, Athens, GA 30602, USA
| | - Essa M Saied
- Institut für Chemie, Humboldt Universität zu Berlin, Berlin 12489, Germany; Chemistry Department, Faculty of Science, Suez Canal University, Ismailia 41522, Egypt
| | - Ho-Joon Lee
- Department of Genetics, Yale University, New Haven, CT 06520, USA; Center for Genome Analysis, Yale University, New Haven, CT 06510, USA
| | - Bulat R Ramazanov
- Department of Cell Biology, Yale University, New Haven, CT 06520, USA
| | - Benjamin Jackson
- Center for Molecular Medicine, Department of Genetics, University of Georgia, Athens, GA 30602, USA
| | - Daniel Burkhardt
- Department of Genetics, Yale University, New Haven, CT 06520, USA
| | - Mikhail Parker
- Center for Molecular Medicine, Department of Genetics, University of Georgia, Athens, GA 30602, USA
| | - Anne S Gleinich
- Complex Carbohydrate Research Center, University of Georgia, Athens, GA 30602, USA
| | - Zhirui Wang
- Complex Carbohydrate Research Center, University of Georgia, Athens, GA 30602, USA
| | - Dong Eun Seo
- Center for Molecular Medicine, Department of Genetics, University of Georgia, Athens, GA 30602, USA
| | - Ting Zhou
- Department of Immunobiology, Yale University, New Haven, CT 06520, USA
| | - Shihao Xu
- NOMIS Center for Immunobiology and Microbial Pathogenesis, the Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Irina Alecu
- Neural Regeneration Laboratory, Department of Biochemistry, Microbiology, and Immunology, University of Ottawa, Ottawa K1H 8M5, Canada
| | - Parastoo Azadi
- Complex Carbohydrate Research Center, University of Georgia, Athens, GA 30602, USA
| | - Christoph Arenz
- Institut für Chemie, Humboldt Universität zu Berlin, Berlin 12489, Germany
| | - Thorsten Hornemann
- Institute of Clinical Chemistry, University and University Hospital of Zurich, Zurich 8091, Switzerland
| | | | - Serge A van de Pavert
- Aix-Marseille Universite, CNRS, INSERM, Centre d'Immunologie de Marseille-Luminy (CIML), 13288 Marseille Cedex 9, France
| | - Susan M Kaech
- NOMIS Center for Immunobiology and Microbial Pathogenesis, the Salk Institute for Biological Studies, La Jolla, CA 92037, USA.
| | - Natalia B Ivanova
- Center for Molecular Medicine, Department of Genetics, University of Georgia, Athens, GA 30602, USA.
| | - Fabio R Santori
- Department of Immunobiology, Yale University, New Haven, CT 06520, USA.
| |
Collapse
|
21
|
Yoshinaga S, Shin M, Kitazawa A, Ishii K, Tanuma M, Kasai A, Hashimoto H, Kubo KI, Nakajima K. Comprehensive characterization of migration profiles of murine cerebral cortical neurons during development using FlashTag labeling. iScience 2021; 24:102277. [PMID: 33851097 PMCID: PMC8022222 DOI: 10.1016/j.isci.2021.102277] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Revised: 12/30/2020] [Accepted: 03/01/2021] [Indexed: 11/26/2022] Open
Abstract
In the mammalian cerebral neocortex, different regions have different cytoarchitecture, neuronal birthdates, and functions. In most regions, neuronal migratory profiles are speculated similar based on observations using thymidine analogs. Few reports have investigated regional migratory differences from mitosis at the ventricular surface. In this study, we applied FlashTag technology, in which dyes are injected intraventricularly, to describe migratory profiles. We revealed a mediolateral regional difference in the migratory profiles of neurons that is dependent on developmental stage; for example, neurons labeled at embryonic day 12.5–15.5 reached their destination earlier dorsomedially than dorsolaterally, even where there were underlying ventricular surfaces, reflecting sojourning below the subplate. This difference was hardly recapitulated by thymidine analogs, which visualize neurogenic gradients, suggesting a biological significance different from the neurogenic gradient. These observations advance our understanding of cortical development and the power of FlashTag in studying migration and are thus resources for future neurodevelopmental studies. FlashTag visualized mediolateral regional differences of cortical migratory profiles Mediolateral differences were observed when neurons were labeled at E12.5–15.5 Late-born neurons transiently sojourned below the dorsolateral subplate (SP) cells The difference was unclear in reeler cortex, where SP cells position superficially
Collapse
Affiliation(s)
- Satoshi Yoshinaga
- Department of Anatomy, Keio University School of Medicine, Shinjuku, Tokyo 160-8582, Japan
| | - Minkyung Shin
- Department of Anatomy, Keio University School of Medicine, Shinjuku, Tokyo 160-8582, Japan
| | - Ayako Kitazawa
- Department of Anatomy, Keio University School of Medicine, Shinjuku, Tokyo 160-8582, Japan
| | - Kazuhiro Ishii
- Department of Anatomy, Keio University School of Medicine, Shinjuku, Tokyo 160-8582, Japan
| | - Masato Tanuma
- Laboratory of Molecular Neuropharmacology, Graduate School of Pharmaceutical Sciences, Osaka University, Suita, Osaka 565-0871, Japan
| | - Atsushi Kasai
- Laboratory of Molecular Neuropharmacology, Graduate School of Pharmaceutical Sciences, Osaka University, Suita, Osaka 565-0871, Japan
| | - Hitoshi Hashimoto
- Laboratory of Molecular Neuropharmacology, Graduate School of Pharmaceutical Sciences, Osaka University, Suita, Osaka 565-0871, Japan.,Molecular Research Center for Children's Mental Development, United Graduate School of Child Development, Osaka University, Kanazawa University, Hamamatsu University School of Medicine, Chiba University, and University of Fukui, Suita, Osaka 565-0871, Japan.,Division of Bioscience, Institute for Datability Science, Osaka University, Suita, Osaka 565-0871, Japan.,Open and Transdisciplinary Research Initiatives, Osaka University, Suita, Osaka 565-0871, Japan.,Department of Molecular Pharmaceutical Sciences, Graduate School of Medicine, Osaka University, Suita, Osaka 565-0871, Japan
| | - Ken-Ichiro Kubo
- Department of Anatomy, Keio University School of Medicine, Shinjuku, Tokyo 160-8582, Japan.,Department of Anatomy, The Jikei University School of Medicine, Minato, Tokyo 105-8461, Japan
| | - Kazunori Nakajima
- Department of Anatomy, Keio University School of Medicine, Shinjuku, Tokyo 160-8582, Japan
| |
Collapse
|
22
|
Limoni G, Niquille M. Semaphorins and Plexins in central nervous system patterning: the key to it all? Curr Opin Neurobiol 2021; 66:224-232. [PMID: 33513538 DOI: 10.1016/j.conb.2020.12.014] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Revised: 12/21/2020] [Accepted: 12/22/2020] [Indexed: 12/17/2022]
Abstract
Semaphorins and Plexins constitute one of the largest family of guidance molecules and receptors involved in setting critical biological steps for central nervous system development. The role of these molecules in axonal development has been extensively characterized but Semaphorins and Plexins are also involved in a variety of other developmental processes, spanning from cell polarization to migration, laminar segregation and neuronal maturation. In this review, we aim to gather discoveries carried in the field of neurodevelopment over the last decade, during which Semaphorin/Plexin complexes have emerged as key regulators of neurogenesis, neural cell migration and adult gliogenesis. As well, we report mechanisms that brought a better understanding of axonal midline crossing.
Collapse
Affiliation(s)
- Greta Limoni
- Department of Basic Neuroscience, University Medical Center, University of Geneva, Rue Michel-Servet 1, 1211 Genève 4, Switzerland.
| | - Mathieu Niquille
- Department of Basic Neuroscience, University Medical Center, University of Geneva, Rue Michel-Servet 1, 1211 Genève 4, Switzerland.
| |
Collapse
|
23
|
Murcia-Ramón R, Company V, Juárez-Leal I, Andreu-Cervera A, Almagro-García F, Martínez S, Echevarría D, Puelles E. Neuronal tangential migration from Nkx2.1-positive hypothalamus. Brain Struct Funct 2020; 225:2857-2869. [PMID: 33145610 PMCID: PMC7674375 DOI: 10.1007/s00429-020-02163-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Accepted: 10/15/2020] [Indexed: 12/28/2022]
Abstract
During the development of the central nervous system, the immature neurons suffer different migration processes. It is well known that Nkx2.1-positive ventricular layer give rise to critical tangential migrations into different regions of the developing forebrain. Our aim was to study this phenomenon in the hypothalamic region. With this purpose, we used a transgenic mouse line that expresses the tdTomato reporter driven by the promotor of Nkx2.1. Analysing the Nkx2.1-positive derivatives at E18.5, we found neural contributions to the prethalamic region, mainly in the zona incerta and in the mes-diencephalic tegmental region. We studied the developing hypothalamus along the embryonic period. From E10.5 we detected that the Nkx2.1 expression domain was narrower than the reporter distribution. Therefore, the Nkx2.1 expression fades in a great number of the early-born neurons from the Nkx2.1-positive territory. At the most caudal positive part, we detected a thin stream of positive neurons migrating caudally into the mes-diencephalic tegmental region using time-lapse experiments on open neural tube explants. Late in development, we found a second migratory stream into the prethalamic territory. All these tangentially migrated neurons developed a gabaergic phenotype. In summary, we have described the contribution of interneurons from the Nkx2.1-positive hypothalamic territory into two different rostrocaudal territories: the mes-diencephalic reticular formation through a caudal tangential migration and the prethalamic zona incerta complex through a dorsocaudal tangential migration.
Collapse
Affiliation(s)
- Raquel Murcia-Ramón
- Instituto de Neurociencias de Alicante, Universidad Miguel Hernández-CSIC, 03550, Sant Joan d'Alacant, Alicante, Spain
| | - Verónica Company
- Instituto de Neurociencias de Alicante, Universidad Miguel Hernández-CSIC, 03550, Sant Joan d'Alacant, Alicante, Spain
| | - Iris Juárez-Leal
- Instituto de Neurociencias de Alicante, Universidad Miguel Hernández-CSIC, 03550, Sant Joan d'Alacant, Alicante, Spain
| | - Abraham Andreu-Cervera
- Instituto de Neurociencias de Alicante, Universidad Miguel Hernández-CSIC, 03550, Sant Joan d'Alacant, Alicante, Spain
| | - Francisca Almagro-García
- Instituto de Neurociencias de Alicante, Universidad Miguel Hernández-CSIC, 03550, Sant Joan d'Alacant, Alicante, Spain
| | - Salvador Martínez
- Instituto de Neurociencias de Alicante, Universidad Miguel Hernández-CSIC, 03550, Sant Joan d'Alacant, Alicante, Spain
| | - Diego Echevarría
- Instituto de Neurociencias de Alicante, Universidad Miguel Hernández-CSIC, 03550, Sant Joan d'Alacant, Alicante, Spain
| | - Eduardo Puelles
- Instituto de Neurociencias de Alicante, Universidad Miguel Hernández-CSIC, 03550, Sant Joan d'Alacant, Alicante, Spain.
| |
Collapse
|
24
|
Reelin-Nrp1 Interaction Regulates Neocortical Dendrite Development in a Context-Specific Manner. J Neurosci 2020; 40:8248-8261. [PMID: 33009002 DOI: 10.1523/jneurosci.1907-20.2020] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Revised: 09/14/2020] [Accepted: 09/23/2020] [Indexed: 11/21/2022] Open
Abstract
Reelin plays versatile roles in neocortical development. The C-terminal region (CTR) of Reelin is required for the correct formation of the superficial structure of the neocortex; however, the mechanisms by which this position-specific effect occurs remain largely unknown. In this study, we demonstrate that Reelin with an intact CTR binds to neuropilin-1 (Nrp1), a transmembrane protein. Both male and female mice were used. Nrp1 is localized with very-low-density lipoprotein receptor (VLDLR), a canonical Reelin receptor, in the superficial layers of the developing neocortex. It forms a complex with VLDLR, and this interaction is modulated by the alternative splicing of VLDLR. Reelin with an intact CTR binds more strongly to the VLDLR/Nrp1 complex than to VLDLR alone. Knockdown of Nrp1 in neurons leads to the accumulation of Dab1 protein. Since the degradation of Dab1 is induced by Reelin signaling, it is suggested that Nrp1 augments Reelin signaling. The interaction between Reelin and Nrp1 is required for normal dendritic development in superficial-layer neurons. All of these characteristics of Reelin are abrogated by proteolytic processing of the six C-terminal amino acid residues of Reelin (0.17% of the whole protein). Therefore, Nrp1 is a coreceptor molecule for Reelin and, together with the proteolytic processing of Reelin, can account for context-specific Reelin function in brain development.SIGNIFICANCE STATEMENT Reelin often exhibits a context-dependent function during brain development; however, its underlying mechanism is not well understood. We found that neuropilin-1 (Nrp1) specifically binds to the CTR of Reelin and acts as a coreceptor for very-low-density lipoprotein receptor (VLDLR). The Nrp1/VLDLR complex is localized in the superficial layers of the neocortex, and its interaction with Reelin is essential for proper dendritic development in superficial-layer neurons. This study provides the first mechanistic evidence of the context-specific function of Reelin (>3400 residues) regulated by the C-terminal residues and Nrp1, a component of the canonical Reelin receptor complex.
Collapse
|
25
|
Kubo KI, Deguchi K. Human neocortical development as a basis to understand mechanisms underlying neurodevelopmental disabilities in extremely preterm infants. J Obstet Gynaecol Res 2020; 46:2242-2250. [PMID: 32924239 DOI: 10.1111/jog.14468] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Accepted: 08/24/2020] [Indexed: 01/21/2023]
Abstract
AIM Recent advances in perinatal and neonatal medicine have resulted in marked improvements in the survival rates of extremely preterm infants (born before 28 gestational weeks) around the world, and Japan is among the countries with the highest reported survival rates of extremely preterm infants. However, it remains a major concern that many survivors develop neurodevelopmental disabilities, including cognitive dysfunctions and neurodevelopmental disorders later in life. In order to understand the pathophysiological mechanisms underlying the neurodevelopmental disabilities observed in the survivors of extremely preterm births, we reviewed recently reported findings about the development of the human neocortex. METHODS First, we have summarized the current knowledge about the development of the neocortex, including recently reported human- and/or primate-specific developmental events. Next, we discussed the possible causal mechanisms underlying the development of neurodevelopmental disabilities in extremely preterm infants. RESULTS Around the birth of extremely preterm infants, neurogenesis and succeeding neuronal migrations are ongoing in the neocortex of human brain. Expansion and maturation of the subplate, which is thought to reflect the axonal wiring in the neocortex, is also prominent at this time. CONCLUSION Brain injuries that occur around the birth of extremely preterm infants are presumed to affect the dynamic developmental events in the neocortex, such as neurogenesis, neuronal migrations and maturation of the subplate, which could underlie the neurodevelopmental disabilities that often develop subsequently in extremely preterm infants. These possibilities should be borne in mind while considering maternal and neonatal care to further improve the long-term outcomes of extremely preterm infants.
Collapse
Affiliation(s)
- Ken-Ichiro Kubo
- Department of Anatomy, Keio University School of Medicine, Tokyo, Japan.,Department of Anatomy, The Jikei University School of Medicine, Tokyo, Japan
| | - Kimiko Deguchi
- Department of Anatomy, Keio University School of Medicine, Tokyo, Japan.,Department of Pediatrics, Deguchi Pediatric Clinic, Omura, Japan
| |
Collapse
|
26
|
Li Z, Jagadapillai R, Gozal E, Barnes G. Deletion of Semaphorin 3F in Interneurons Is Associated with Decreased GABAergic Neurons, Autism-like Behavior, and Increased Oxidative Stress Cascades. Mol Neurobiol 2019; 56:5520-5538. [PMID: 30635860 PMCID: PMC6614133 DOI: 10.1007/s12035-018-1450-9] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2018] [Accepted: 12/07/2018] [Indexed: 12/11/2022]
Abstract
Autism and epilepsy are diseases which have complex genetic inheritance. Genome-wide association and other genetic studies have implicated at least 500+ genes associated with the occurrence of autism spectrum disorders (ASD) including the human semaphorin 3F (Sema 3F) and neuropilin 2 (NRP2) genes. However, the genetic basis of the comorbid occurrence of autism and epilepsy is unknown. The aberrant development of GABAergic circuitry is a possible risk factor in autism and epilepsy. Molecular biological approaches were used to test the hypothesis that cell-specific genetic variation in mouse homologs affects the formation and function of GABAergic circuitry. The empirical analysis with mice homozygous null for one of these genes, Sema 3F, in GABAergic neurons substantiated these predictions. Notably, deletion of Sema 3F in interneurons but not excitatory neurons during early development decreased the number of interneurons/neurites and mRNAs for cell-specific GABAergic markers and increased epileptogenesis and autistic behaviors. Studies of interneuron cell-specific knockout of Sema 3F signaling suggest that deficient Sema 3F signaling may lead to neuroinflammation and oxidative stress. Further studies of mouse KO models of ASD genes such as Sema 3F or NRP2 may be informative to clinical phenotypes contributing to the pathogenesis in autism and epilepsy patients.
Collapse
Affiliation(s)
- Zhu Li
- Department of Neurology, Vanderbilt University School of Medicine, Nashville, TN, USA
- Department of Pediatrics, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Rekha Jagadapillai
- Department of Pediatrics, University of Louisville School of Medicine, Louisville, KY, USA
| | - Evelyne Gozal
- Department of Pediatrics, University of Louisville School of Medicine, Louisville, KY, USA
| | - Gregory Barnes
- Department of Pediatrics, University of Louisville School of Medicine, Louisville, KY, USA.
- Department of Neurology, University of Louisville School of Medicine, Louisville, KY, USA.
- Pediatric Research Institute, University of Louisville Autism Center, 1405 East Burnett Ave, Louisville, KY, 40217, USA.
| |
Collapse
|
27
|
Ruiz-Reig N, Andres B, Lamonerie T, Theil T, Fairén A, Studer M. The caudo-ventral pallium is a novel pallial domain expressing Gdf10 and generating Ebf3-positive neurons of the medial amygdala. Brain Struct Funct 2018; 223:3279-3295. [PMID: 29869132 DOI: 10.1007/s00429-018-1687-0] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Accepted: 05/18/2018] [Indexed: 12/16/2022]
Abstract
In rodents, the medial nucleus of the amygdala receives direct inputs from the accessory olfactory bulbs and is mainly implicated in pheromone-mediated reproductive and defensive behaviors. The principal neurons of the medial amygdala are GABAergic neurons generated principally in the caudo-ventral medial ganglionic eminence and preoptic area. Beside GABAergic neurons, the medial amygdala also contains glutamatergic Otp-expressing neurons cells generated in the lateral hypothalamic neuroepithelium and a non-well characterized Pax6-positive population. In the present work, we describe a novel glutamatergic Ebf3-expressing neuronal subpopulation distributed within the periphery of the postero-ventral medial amygdala. These neurons are generated in a pallial domain characterized by high expression of Gdf10. This territory is topologically the most caudal tier of the ventral pallium and accordingly, we named it Caudo-Ventral Pallium (CVP). In the absence of Pax6, the CVP is disrupted and Ebf3-expressing neurons fail to be generated. Overall, this work proposes a novel model of the neuronal composition of the medial amygdala and unravels for the first time a new novel pallial subpopulation originating from the CVP and expressing the transcription factor Ebf3.
Collapse
Affiliation(s)
- Nuria Ruiz-Reig
- Université Côte d'Azur (UCA), CNRS, Inserm, Institut de Biologie Valrose (iBV), 06108, Nice, France.
- Instituto de Neurociencias (Consejo Superior de Investigaciones Científicas-Universidad Miguel Hernández, CSIC-UMH), 03550, San Juan de Alicante, Spain.
| | - Belen Andres
- Instituto de Neurociencias (Consejo Superior de Investigaciones Científicas-Universidad Miguel Hernández, CSIC-UMH), 03550, San Juan de Alicante, Spain
| | - Thomas Lamonerie
- Université Côte d'Azur (UCA), CNRS, Inserm, Institut de Biologie Valrose (iBV), 06108, Nice, France
| | - Thomas Theil
- Centre for Discovery Brain Sciences, Hugh Robson Building, University of Edinburgh, Edinburgh, EH8 9XD, UK
| | - Alfonso Fairén
- Instituto de Neurociencias (Consejo Superior de Investigaciones Científicas-Universidad Miguel Hernández, CSIC-UMH), 03550, San Juan de Alicante, Spain
- , Palau 11, 03550, San Juan de Alicante, Spain
| | - Michèle Studer
- Université Côte d'Azur (UCA), CNRS, Inserm, Institut de Biologie Valrose (iBV), 06108, Nice, France.
| |
Collapse
|
28
|
Ruiz-Reig N, Studer M. Rostro-Caudal and Caudo-Rostral Migrations in the Telencephalon: Going Forward or Backward? Front Neurosci 2017; 11:692. [PMID: 29311773 PMCID: PMC5742585 DOI: 10.3389/fnins.2017.00692] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2017] [Accepted: 11/23/2017] [Indexed: 11/13/2022] Open
Abstract
The generation and differentiation of an appropriate number of neurons, as well as its distribution in different parts of the brain, is crucial for the proper establishment, maintenance and plasticity of neural circuitries. Newborn neurons travel along the brain in a process known as neuronal migration, to finalize their correct position in the nervous system. Defects in neuronal migration produce abnormalities in the brain that can generate neurodevelopmental pathologies, such as autism, schizophrenia and intellectual disability. In this review, we present an overview of the developmental origin of the different telencephalic subdivisions and a description of migratory pathways taken by distinct neural populations traveling long distances before reaching their target position in the brain. In addition, we discuss some of the molecules implicated in the guidance of these migratory paths and transcription factors that contribute to the correct migration and integration of these neurons.
Collapse
|
29
|
Ephrin-A2 regulates excitatory neuron differentiation and interneuron migration in the developing neocortex. Sci Rep 2017; 7:11813. [PMID: 28924206 PMCID: PMC5603509 DOI: 10.1038/s41598-017-12185-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2017] [Accepted: 09/05/2017] [Indexed: 11/30/2022] Open
Abstract
The development of the neocortex requires co-ordination between proliferation and differentiation, as well as the precise orchestration of neuronal migration. Eph/ephrin signaling is crucial in guiding neurons and their projections during embryonic development. In adult ephrin-A2 knockout mice we consistently observed focal patches of disorganized neocortical laminar architecture, ranging in severity from reduced neuronal density to a complete lack of neurons. Loss of ephrin-A2 in the pre-optic area of the diencephalon reduced the migration of neocortex-bound interneurons from this region. Furthermore, ephrin-A2 participates in the creation of excitatory neurons by inhibiting apical progenitor proliferation in the ventricular zone, with the disruption of ephrin-A2 signaling in these cells recapitulating the abnormal neocortex observed in the knockout. The disturbance to the architecture of the neocortex observed following deletion of ephrin-A2 signaling shares many similarities with defects found in the neocortex of children diagnosed with autism spectrum disorder.
Collapse
|
30
|
Yang X, Feng S, Tang K. COUP-TF Genes, Human Diseases, and the Development of the Central Nervous System in Murine Models. Curr Top Dev Biol 2017; 125:275-301. [PMID: 28527575 DOI: 10.1016/bs.ctdb.2016.12.002] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
COUP-TFI and -TFII are members of the steroid/thyroid nuclear receptor superfamily. Recent clinical studies reveal that COUP-TFI gene mutations are associated with Bosch-Boonstra-Schaaf optic atrophy syndrome displaying symptoms of optic atrophy, intellectual disability, hypotonia, seizure, autism spectrum disorders, oromotor dysfunction, thin corpus callosum, or hearing defects, and COUP-TFII gene mutations lead to congenital heart defects and/or congenital diaphragmatic hernia with developmental delay and mental defects. In this review, we first describe the functions of COUP-TF genes in the morphogenesis of mouse forebrain including cerebral cortex, hippocampus, amygdala complex, hypothalamus, and cortical interneuron. Then, we address their roles in the development of cerebellum, glial cells, neural crest cells, and adult neuronal stem cells. Clearly, the investigations on the functions of COUP-TF genes in the developing mouse central nervous system will benefit not only the understanding of neurodevelopment, but also the etiology of human mental diseases.
Collapse
Affiliation(s)
- Xiong Yang
- Institute of Life Science, Nanchang University, Nanchang, Jiangxi, China
| | - Su Feng
- Institute of Life Science, Nanchang University, Nanchang, Jiangxi, China
| | - Ke Tang
- Institute of Life Science, Nanchang University, Nanchang, Jiangxi, China.
| |
Collapse
|
31
|
Andrews WD, Barber M, Nemitz M, Memi F, Parnavelas JG. Semaphorin3A-neuropilin1 signalling is involved in the generation of cortical interneurons. Brain Struct Funct 2016; 222:2217-2233. [PMID: 27858201 PMCID: PMC5504245 DOI: 10.1007/s00429-016-1337-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2016] [Accepted: 11/11/2016] [Indexed: 01/25/2023]
Abstract
Cortical interneurons are generated predominantly in the medial ganglionic eminence of the ventral telencephalon and migrate to the cortex during embryonic development. These cells express neuropilin (Nrp1 and Nrp2) receptors which mediate their response to the chemorepulsive class 3 semaphorin (Sema) ligands. We show here that semaphorins Sema3A and Sema3F are expressed in layers adjacent to cortical interneuron migratory streams as well as in the striatum, suggesting they may have a role in guiding these cells throughout their journey. Analysis of Sema3A -/- and Sema3F -/- mice during corticogenesis showed that absence of Sema3A, but not Sema3F, leads to aberrant migration of cortical interneurons through the striatum. Reduced number of cortical interneurons was found in the cortex of Sema3A -/-, Nrp1 -/- and Nrp2 -/- mice, as well as altered distribution in Sema3F -/-, Nrp1 -/-, Nrp2 -/- animals and especially in neuropilin double mutants. The observed decrease in interneurons in Sema3A -/- and Nrp1 -/- mice was due to altered proliferative activity of their progenitors highlighted by changes in their mitotic spindle positioning and angle of cleavage plane during cell division. These findings point to a novel role for Sema3A-Nrp1 signalling in progenitor cell dynamics and in the generation of interneurons in the ventral telencephalon.
Collapse
Affiliation(s)
- William D Andrews
- Department of Cell and Developmental Biology, University College London, Gower Street, London, WC1E 6BT, UK.
| | - Melissa Barber
- Department of Cell and Developmental Biology, University College London, Gower Street, London, WC1E 6BT, UK
| | - Marion Nemitz
- Department of Cell and Developmental Biology, University College London, Gower Street, London, WC1E 6BT, UK
| | - Fani Memi
- Department of Cell and Developmental Biology, University College London, Gower Street, London, WC1E 6BT, UK
| | - John G Parnavelas
- Department of Cell and Developmental Biology, University College London, Gower Street, London, WC1E 6BT, UK.
| |
Collapse
|
32
|
Ishii K, Kubo KI, Nakajima K. Reelin and Neuropsychiatric Disorders. Front Cell Neurosci 2016; 10:229. [PMID: 27803648 PMCID: PMC5067484 DOI: 10.3389/fncel.2016.00229] [Citation(s) in RCA: 138] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2016] [Accepted: 09/22/2016] [Indexed: 12/22/2022] Open
Abstract
Proper neuronal migration and laminar formation during corticogenesis is essential for normal brain function. Disruption of these developmental processes is thought to be involved in the pathogenesis of some neuropsychiatric conditions. Especially, Reelin, a glycoprotein mainly secreted by the Cajal-Retzius cells and a subpopulation of GABAergic interneurons, has been shown to play a critical role, both during embryonic and postnatal periods. Indeed, animal studies have clearly revealed that Reelin is an essential molecule for proper migration of cortical neurons and finally regulates the cell positioning in the cortex during embryonic and early postnatal stages; by contrast, Reelin signaling is closely involved in synaptic function in adulthood. In humans, genetic studies have shown that the reelin gene (RELN) is associated with a number of psychiatric diseases, including Schizophrenia (SZ), bipolar disorder (BP) and autistic spectrum disorder. Indeed, Reln haploinsufficiency has been shown to cause cognitive impairment in rodents, suggesting the expression level of the Reelin protein is closely related to the higher brain functions. However, the molecular abnormalities in the Reelin pathway involved in the pathogenesis of psychiatric disorders are not yet fully understood. In this article, we review the current progress in the understanding of the Reelin functions that could be related to the pathogenesis of psychiatric disorders. Furthermore, we discuss the basis for selecting Reelin and molecules in its downstream signaling pathway as potential therapeutic targets for psychiatric illnesses.
Collapse
Affiliation(s)
- Kazuhiro Ishii
- Department of Anatomy, Keio University School of Medicine Tokyo, Japan
| | - Ken-Ichiro Kubo
- Department of Anatomy, Keio University School of Medicine Tokyo, Japan
| | - Kazunori Nakajima
- Department of Anatomy, Keio University School of Medicine Tokyo, Japan
| |
Collapse
|
33
|
Doan RN, Bae BI, Cubelos B, Chang C, Hossain AA, Al-Saad S, Mukaddes NM, Oner O, Al-Saffar M, Balkhy S, Gascon GG, Nieto M, Walsh CA. Mutations in Human Accelerated Regions Disrupt Cognition and Social Behavior. Cell 2016; 167:341-354.e12. [PMID: 27667684 PMCID: PMC5063026 DOI: 10.1016/j.cell.2016.08.071] [Citation(s) in RCA: 228] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2016] [Revised: 05/18/2016] [Accepted: 08/26/2016] [Indexed: 12/15/2022]
Abstract
Comparative analyses have identified genomic regions potentially involved in human evolution but do not directly assess function. Human accelerated regions (HARs) represent conserved genomic loci with elevated divergence in humans. If some HARs regulate human-specific social and behavioral traits, then mutations would likely impact cognitive and social disorders. Strikingly, rare biallelic point mutations-identified by whole-genome and targeted "HAR-ome" sequencing-showed a significant excess in individuals with ASD whose parents share common ancestry compared to familial controls, suggesting a contribution in 5% of consanguineous ASD cases. Using chromatin interaction sequencing, massively parallel reporter assays (MPRA), and transgenic mice, we identified disease-linked, biallelic HAR mutations in active enhancers for CUX1, PTBP2, GPC4, CDKL5, and other genes implicated in neural function, ASD, or both. Our data provide genetic evidence that specific HARs are essential for normal development, consistent with suggestions that their evolutionary changes may have altered social and/or cognitive behavior. PAPERCLIP.
Collapse
Affiliation(s)
- Ryan N Doan
- Division of Genetics and Genomics, Manton Center for Orphan Disease Research, and Howard Hughes Medical Institute, Boston Children's Hospital, Boston, MA 02115, USA
| | - Byoung-Il Bae
- Division of Genetics and Genomics, Manton Center for Orphan Disease Research, and Howard Hughes Medical Institute, Boston Children's Hospital, Boston, MA 02115, USA
| | - Beatriz Cubelos
- Department of Molecular Biology, Centro de Biología Molecular 'Severo Ochoa', Universidad Autonoma de Madrid, UAM-CSIC, Nicolas Cabrera 1, 28049 Madrid, Spain; Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología, CNB-CSIC, Darwin 3, Campus de Cantoblanco, 28049 Madrid, Spain
| | - Cindy Chang
- Division of Genetics and Genomics, Manton Center for Orphan Disease Research, and Howard Hughes Medical Institute, Boston Children's Hospital, Boston, MA 02115, USA
| | - Amer A Hossain
- Division of Genetics and Genomics, Manton Center for Orphan Disease Research, and Howard Hughes Medical Institute, Boston Children's Hospital, Boston, MA 02115, USA
| | | | - Nahit M Mukaddes
- Istanbul Institute of Child and Adolescent Psychiatry, 34365 Istanbul, Turkey
| | - Ozgur Oner
- Department of Child and Adolescent Psychiatry, Bahcesehir University School of Medicine, 34353 Istanbul, Turkey
| | - Muna Al-Saffar
- Division of Genetics and Genomics, Manton Center for Orphan Disease Research, and Howard Hughes Medical Institute, Boston Children's Hospital, Boston, MA 02115, USA; Department of Pediatrics, College of Medicine and Health Sciences, United Arab Emirates University, PO Box 17666, Al-Ain, United Arab Emirates
| | - Soher Balkhy
- Department of Pediatrics, King Faisal Specialist Hospital and Research Center, Jeddah 21499, Kingdom of Saudi Arabia
| | - Generoso G Gascon
- Department of Neurology (Pediatric Neurology), Massachusetts General Hospital, Boston, MA 02114, USA
| | - Marta Nieto
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología, CNB-CSIC, Darwin 3, Campus de Cantoblanco, 28049 Madrid, Spain
| | - Christopher A Walsh
- Division of Genetics and Genomics, Manton Center for Orphan Disease Research, and Howard Hughes Medical Institute, Boston Children's Hospital, Boston, MA 02115, USA; Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Departments of Pediatrics and Neurology, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
34
|
Hegeman DJ, Hong ES, Hernández VM, Chan CS. The external globus pallidus: progress and perspectives. Eur J Neurosci 2016; 43:1239-65. [PMID: 26841063 PMCID: PMC4874844 DOI: 10.1111/ejn.13196] [Citation(s) in RCA: 106] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2015] [Revised: 01/20/2016] [Accepted: 01/27/2016] [Indexed: 12/12/2022]
Abstract
The external globus pallidus (GPe) of the basal ganglia is in a unique and powerful position to influence processing of motor information by virtue of its widespread projections to all basal ganglia nuclei. Despite the clinical importance of the GPe in common motor disorders such as Parkinson's disease, there is only limited information about its cellular composition and organizational principles. In this review, recent advances in the understanding of the diversity in the molecular profile, anatomy, physiology and corresponding behaviour during movement of GPe neurons are described. Importantly, this study attempts to build consensus and highlight commonalities of the cellular classification based on existing but contentious literature. Additionally, an analysis of the literature concerning the intricate reciprocal loops formed between the GPe and major synaptic partners, including both the striatum and the subthalamic nucleus, is provided. In conclusion, the GPe has emerged as a crucial node in the basal ganglia macrocircuit. While subtleties in the cellular makeup and synaptic connection of the GPe create new challenges, modern research tools have shown promise in untangling such complexity, and will provide better understanding of the roles of the GPe in encoding movements and their associated pathologies.
Collapse
Affiliation(s)
- Daniel J Hegeman
- Department of Physiology, Feinberg School of Medicine, Northwestern University, Chicago, IL, 60611, USA
| | - Ellie S Hong
- Department of Physiology, Feinberg School of Medicine, Northwestern University, Chicago, IL, 60611, USA
| | - Vivian M Hernández
- Department of Physiology, Feinberg School of Medicine, Northwestern University, Chicago, IL, 60611, USA
| | - C Savio Chan
- Department of Physiology, Feinberg School of Medicine, Northwestern University, Chicago, IL, 60611, USA
| |
Collapse
|
35
|
Touzot A, Ruiz-Reig N, Vitalis T, Studer M. Molecular control of two novel migratory paths for CGE-derived interneurons in the developing mouse brain. Development 2016; 143:1753-65. [DOI: 10.1242/dev.131102] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2015] [Accepted: 03/17/2016] [Indexed: 01/15/2023]
Abstract
GABAergic interneurons are highly heterogenous and originate in the subpallium mainly from the medial (MGE) and caudal (CGE) ganglionic eminences according to a precise temporal sequence. While MGE-derived cells disperse dorsally and migrate towards all regions of the cortex, little is known on how CGE-derived cells reach their targets during development. Here, we unravel the existence of two novel CGE caudo-rostral migratory streams, one located laterally (LMS) and the other one more medially (MMS) that, together with the well-known caudal migratory stream (CMS), contribute to populate the neocortex, hippocampus and amygdala. These paths appear in a precise temporal sequence and express a distinct combination of transcription factors, such as Sp8, Prox1, COUP-TFI and COUP-TFII. By inactivating COUP-TFI in developing interneurons, the lateral and medial streams are perturbed and expression of Sp8 and COUP-TFII affected. As a consequence, adult mutant neocortices have laminar-specific alterations of distinct cortical interneuron subtypes. Overall, we propose that the existence of spatially and temporally regulated migratory paths in the subpallium contributes to the laminar distribution and specification of distinct interneuron subpopulations in the adult brain.
Collapse
Affiliation(s)
- Audrey Touzot
- Univ. Nice Sophia Antipolis, Inserm, CNRS, iBV, 06100 Nice, France
- iBV, Institut de Biologie Valrose, Univ. Sophia Antipolis, Bâtiment Sciences Naturelles; UFR Sciences; Parc Valrose, 28, avenue Valrose, 06108 Nice Cedex 2, France
| | - Nuria Ruiz-Reig
- Univ. Nice Sophia Antipolis, Inserm, CNRS, iBV, 06100 Nice, France
- iBV, Institut de Biologie Valrose, Univ. Sophia Antipolis, Bâtiment Sciences Naturelles; UFR Sciences; Parc Valrose, 28, avenue Valrose, 06108 Nice Cedex 2, France
- Instituto de Neurociencias de Alicante (Consejo Superior de Investigaciones Cientificas-Universidad Miguel Hernandez, CSIC-UMH), 03550 Alicante, Spain
| | - Tania Vitalis
- Inserm U1141 PROTECT, Hôpital Robert-Debré, 75019 Paris, France
| | - Michèle Studer
- Univ. Nice Sophia Antipolis, Inserm, CNRS, iBV, 06100 Nice, France
- iBV, Institut de Biologie Valrose, Univ. Sophia Antipolis, Bâtiment Sciences Naturelles; UFR Sciences; Parc Valrose, 28, avenue Valrose, 06108 Nice Cedex 2, France
| |
Collapse
|
36
|
Neuronal Heterotopias Affect the Activities of Distant Brain Areas and Lead to Behavioral Deficits. J Neurosci 2015; 35:12432-45. [PMID: 26354912 DOI: 10.1523/jneurosci.3648-14.2015] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Neuronal heterotopia refers to brain malformations resulting from deficits of neuronal migration. Individuals with heterotopias show a high incidence of neurological deficits, such as epilepsy. More recently, it has come to be recognized that focal heterotopias may also show a range of psychiatric problems, including cognitive and behavioral impairments. However, because focal heterotopias are not always located in the brain areas responsible for the symptoms, the causal relationship between the symptoms and heterotopias remains elusive. In this study, we showed that mice with focal heterotopias in the somatosensory cortex generated by in utero electroporation exhibited spatial working memory deficit and low competitive dominance behavior, which have been shown to be closely associated with the activity of the medial prefrontal cortex (mPFC) in rodents. Analysis of the mPFC activity revealed that the immediate-early gene expression was decreased and the local field potentials of the mPFC were altered in the mice with heterotopias compared with the control mice. Moreover, activation of these ectopic and overlying sister neurons using the DREADD (designer receptor exclusively activated by designer drug) system improved the working memory deficits. These findings suggest that cortical regions containing focal heterotopias can affect distant brain regions and give rise to behavioral abnormalities. Significance statement: Recent studies reported that patients with heterotopias have a variety of clinical symptoms, such as cognitive disturbance, psychiatric symptoms, and autistic behavior. However, the causal relationship between the symptoms and heterotopias remains elusive. Here we showed that mice with focal heterotopias in the somatosensory cortex generated by in utero electroporation exhibited behavioral deficits that have been shown to be associated with the mPFC activity in rodents. The existence of heterotopias indeed altered the neural activities of the mPFC, and direct manipulation of the neural activity of the ectopic neurons and their sister neurons in the overlying cortex improved the behavioral deficit. Thus, our results indicate that focal heterotopias could affect the activities of distant brain areas and cause behavioral abnormalities.
Collapse
|