1
|
Hosaka S, Hosokawa M, Hibi M, Shimizu T. The Zebrafish Cerebellar Neural Circuits Are Involved in Orienting Behavior. eNeuro 2024; 11:ENEURO.0141-24.2024. [PMID: 39406478 PMCID: PMC11521796 DOI: 10.1523/eneuro.0141-24.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Revised: 07/22/2024] [Accepted: 08/13/2024] [Indexed: 11/01/2024] Open
Abstract
Deficits in social behavior are found in neurodevelopmental disorders, including autism spectrum disorders (ASDs). Since abnormalities in cerebellar morphology and function are observed in ASD patients, the cerebellum is thought to play a role in social behavior. However, it remains unknown whether the cerebellum is involved in social behavior in other animals and how cerebellar circuits control social behavior. To address this issue, we employed zebrafish stereotyped orienting behavior as a model of social behaviors, in which a pair of adult zebrafish in two separate tanks approach each other, with one swimming at synchronized angles (orienting angles) with the other. We harnessed transgenic zebrafish that express botulinum toxin, which inhibits the release of neurotransmitters, in either granule cells or Purkinje cells (PCs), and zebrafish mutants of reelin, which is involved in the positioning of cerebellar neurons, including PCs. These zebrafish, deficient in the function or formation of cerebellar neural circuits, showed a significantly shorter period of orienting behavior compared with their control siblings. We found an increase in c-fos and egr1 expression in the cerebellum after the orienting behavior. These results suggest that zebrafish cerebellar circuits play an important role in social orienting behavior.
Collapse
Affiliation(s)
- Shiori Hosaka
- Department of Biological Science, Graduate School of Science, Nagoya University, Nagoya, Aichi 464-8602, Japan
| | - Miu Hosokawa
- Department of Biological Science, Graduate School of Science, Nagoya University, Nagoya, Aichi 464-8602, Japan
| | - Masahiko Hibi
- Department of Biological Science, Graduate School of Science, Nagoya University, Nagoya, Aichi 464-8602, Japan
| | - Takashi Shimizu
- Department of Biological Science, Graduate School of Science, Nagoya University, Nagoya, Aichi 464-8602, Japan
| |
Collapse
|
2
|
Skelin L, Racetin A, Kelam N, Ogorevc M, Znaor L, Saraga-Babić M, Filipović N, Katsuyama Y, Pogorelić Z, Vukojević K. Connexin Expression Is Altered in the Eye Development of Yotari Mice: A Preliminary Study. Biomolecules 2024; 14:1174. [PMID: 39334940 PMCID: PMC11430515 DOI: 10.3390/biom14091174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Revised: 09/12/2024] [Accepted: 09/13/2024] [Indexed: 09/30/2024] Open
Abstract
This study aimed to explore how Dab1 functional silencing influences the expression patterns of different connexins in the developing yotari (yot) mice eyes as potential determinants of retinogenesis. Using immunofluorescence staining, the protein expression of Dab1, Reelin, and connexin 37, 40, 43, and 45 (Cx37, Cx40, Cx43, and Cx45) in the wild-type (wt) and yot eyes at embryonic days 13.5 and 15.5 (E13.5 and E15.5) were analyzed. Different expression patterns of Cx37 were seen between the wt and yot groups. The highest fluorescence intensity of Cx37 was observed in the yot animals at E15.5. Cx40 had higher expression at the E13.5 when differentiation of retinal layers was still beginning, whereas it decreased at the E15.5 when differentiation was at the advanced stage. Higher expression of Cx43 was found in the yot group at both time points. Cx45 was predominantly expressed at E13.5 in both groups. Our results reveal the altered expression of connexins during retinogenesis in yot mice and their potential involvement in retinal pathology, where they might serve as prospective therapeutic targets.
Collapse
Affiliation(s)
- Ljubica Skelin
- Clinical Department of Ophthalmology, University Hospital of Split, 21000 Split, Croatia
| | - Anita Racetin
- Department of Anatomy, Histology and Embryology, University of Split School of Medicine, Šoltanska 2A, 21000 Split, Croatia
- Center for Translational Research in Biomedicine, University of Split School of Medicine, Šoltanska 2A, 21000 Split, Croatia
| | - Nela Kelam
- Department of Anatomy, Histology and Embryology, University of Split School of Medicine, Šoltanska 2A, 21000 Split, Croatia
- Center for Translational Research in Biomedicine, University of Split School of Medicine, Šoltanska 2A, 21000 Split, Croatia
| | - Marin Ogorevc
- Department of Anatomy, Histology and Embryology, University of Split School of Medicine, Šoltanska 2A, 21000 Split, Croatia
- Center for Translational Research in Biomedicine, University of Split School of Medicine, Šoltanska 2A, 21000 Split, Croatia
| | - Ljubo Znaor
- Clinical Department of Ophthalmology, University Hospital of Split, 21000 Split, Croatia
- Department of Ophthalmology, University of Split School of Medicine, Šoltanska 2A, 21000 Split, Croatia
| | - Mirna Saraga-Babić
- Department of Anatomy, Histology and Embryology, University of Split School of Medicine, Šoltanska 2A, 21000 Split, Croatia
| | - Natalija Filipović
- Department of Anatomy, Histology and Embryology, University of Split School of Medicine, Šoltanska 2A, 21000 Split, Croatia
| | - Yu Katsuyama
- Department of Anatomy, Shiga University of Medical Science, Otsu 520-2192, Japan
| | - Zenon Pogorelić
- Department of Pediatric Surgery, Split University Hospital, 21000 Split, Croatia
| | - Katarina Vukojević
- Department of Anatomy, Histology and Embryology, University of Split School of Medicine, Šoltanska 2A, 21000 Split, Croatia
- Center for Translational Research in Biomedicine, University of Split School of Medicine, Šoltanska 2A, 21000 Split, Croatia
| |
Collapse
|
3
|
Cui Y, Rolova T, Fagerholm SC. The role of integrins in brain health and neurodegenerative diseases. Eur J Cell Biol 2024; 103:151441. [PMID: 39002282 DOI: 10.1016/j.ejcb.2024.151441] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 06/28/2024] [Accepted: 07/02/2024] [Indexed: 07/15/2024] Open
Abstract
Integrins are heterodimeric membrane proteins expressed on the surface of most cells. They mediate adhesion and signaling processes relevant for a wealth of physiological processes, including nervous system development and function. Interestingly, integrins are also recognized therapeutic targets for inflammatory diseases, such as multiple sclerosis. Here, we discuss the role of integrins in brain development and function, as well as in neurodegenerative diseases affecting the brain (Alzheimer's disease, multiple sclerosis, stroke). Furthermore, we discuss therapeutic targeting of these adhesion receptors in inflammatory diseases of the brain.
Collapse
Affiliation(s)
- Yunhao Cui
- Molecular and Integrative Biosciences Research Programme, Faculty of Biological and Environmental Sciences, University of Helsinki, Helsinki 00790, Finland
| | - Taisia Rolova
- Neuroscience Center, HiLIFE, University of Helsinki, Helsinki 00290, Finland
| | - Susanna C Fagerholm
- Molecular and Integrative Biosciences Research Programme, Faculty of Biological and Environmental Sciences, University of Helsinki, Helsinki 00790, Finland.
| |
Collapse
|
4
|
Sultana OF, Bandaru M, Islam MA, Reddy PH. Unraveling the complexity of human brain: Structure, function in healthy and disease states. Ageing Res Rev 2024; 100:102414. [PMID: 39002647 PMCID: PMC11384519 DOI: 10.1016/j.arr.2024.102414] [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: 05/23/2024] [Revised: 06/29/2024] [Accepted: 07/05/2024] [Indexed: 07/15/2024]
Abstract
The human brain stands as an intricate organ, embodying a nexus of structure, function, development, and diversity. This review delves into the multifaceted landscape of the brain, spanning its anatomical intricacies, diverse functional capacities, dynamic developmental trajectories, and inherent variability across individuals. The dynamic process of brain development, from early embryonic stages to adulthood, highlights the nuanced changes that occur throughout the lifespan. The brain, a remarkably complex organ, is composed of various anatomical regions, each contributing uniquely to its overall functionality. Through an exploration of neuroanatomy, neurophysiology, and electrophysiology, this review elucidates how different brain structures interact to support a wide array of cognitive processes, sensory perception, motor control, and emotional regulation. Moreover, it addresses the impact of age, sex, and ethnic background on brain structure and function, and gender differences profoundly influence the onset, progression, and manifestation of brain disorders shaped by genetic, hormonal, environmental, and social factors. Delving into the complexities of the human brain, it investigates how variations in anatomical configuration correspond to diverse functional capacities across individuals. Furthermore, it examines the impact of neurodegenerative diseases on the structural and functional integrity of the brain. Specifically, our article explores the pathological processes underlying neurodegenerative diseases, such as Alzheimer's, Parkinson's, and Huntington's diseases, shedding light on the structural alterations and functional impairments that accompany these conditions. We will also explore the current research trends in neurodegenerative diseases and identify the existing gaps in the literature. Overall, this article deepens our understanding of the fundamental principles governing brain structure and function and paves the way for a deeper understanding of individual differences and tailored approaches in neuroscience and clinical practice-additionally, a comprehensive understanding of structural and functional changes that manifest in neurodegenerative diseases.
Collapse
Affiliation(s)
- Omme Fatema Sultana
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
| | - Madhuri Bandaru
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
| | - Md Ariful Islam
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
| | - P Hemachandra Reddy
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; Nutritional Sciences Department, College of Human Sciences, Texas Tech University, Lubbock, TX 79409, USA; Department of Pharmacology and Neuroscience, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; Department of Neurology, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA 5. Department of Public Health, Graduate School of Biomedical Sciences, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; Department of Speech, Language, and Hearing Sciences, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA.
| |
Collapse
|
5
|
Primak A, Bozov K, Rubina K, Dzhauari S, Neyfeld E, Illarionova M, Semina E, Sheleg D, Tkachuk V, Karagyaur M. Morphogenetic theory of mental and cognitive disorders: the role of neurotrophic and guidance molecules. Front Mol Neurosci 2024; 17:1361764. [PMID: 38646100 PMCID: PMC11027769 DOI: 10.3389/fnmol.2024.1361764] [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: 12/26/2023] [Accepted: 03/04/2024] [Indexed: 04/23/2024] Open
Abstract
Mental illness and cognitive disorders represent a serious problem for the modern society. Many studies indicate that mental disorders are polygenic and that impaired brain development may lay the ground for their manifestation. Neural tissue development is a complex and multistage process that involves a large number of distant and contact molecules. In this review, we have considered the key steps of brain morphogenesis, and the major molecule families involved in these process. The review provides many indications of the important contribution of the brain development process and correct functioning of certain genes to human mental health. To our knowledge, this comprehensive review is one of the first in this field. We suppose that this review may be useful to novice researchers and clinicians wishing to navigate the field.
Collapse
Affiliation(s)
- Alexandra Primak
- Faculty of Medicine, Lomonosov Moscow State University, Moscow, Russia
| | - Kirill Bozov
- Faculty of Medicine, Lomonosov Moscow State University, Moscow, Russia
| | - Kseniya Rubina
- Faculty of Medicine, Lomonosov Moscow State University, Moscow, Russia
| | - Stalik Dzhauari
- Faculty of Medicine, Lomonosov Moscow State University, Moscow, Russia
| | - Elena Neyfeld
- Faculty of Medicine, Lomonosov Moscow State University, Moscow, Russia
- Federal State Budgetary Educational Institution of the Higher Education “A.I. Yevdokimov Moscow State University of Medicine and Dentistry” of the Ministry of Healthcare of the Russian Federation, Moscow, Russia
| | - Maria Illarionova
- Faculty of Medicine, Lomonosov Moscow State University, Moscow, Russia
| | - Ekaterina Semina
- Faculty of Medicine, Lomonosov Moscow State University, Moscow, Russia
| | - Dmitriy Sheleg
- Faculty of Medicine, Lomonosov Moscow State University, Moscow, Russia
- Federal State Budgetary Educational Institution of the Higher Education “A.I. Yevdokimov Moscow State University of Medicine and Dentistry” of the Ministry of Healthcare of the Russian Federation, Moscow, Russia
| | - Vsevolod Tkachuk
- Faculty of Medicine, Lomonosov Moscow State University, Moscow, Russia
- Institute for Regenerative Medicine, Medical Research and Education Center, Lomonosov Moscow State University, Moscow, Russia
| | - Maxim Karagyaur
- Faculty of Medicine, Lomonosov Moscow State University, Moscow, Russia
- Institute for Regenerative Medicine, Medical Research and Education Center, Lomonosov Moscow State University, Moscow, Russia
| |
Collapse
|
6
|
Pavlou M, Probst M, Blasdel N, Prieve AR, Reh TA. The impact of timing and injury mode on induced neurogenesis in the adult mammalian retina. Stem Cell Reports 2024; 19:239-253. [PMID: 38278154 PMCID: PMC10874861 DOI: 10.1016/j.stemcr.2023.12.010] [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: 11/29/2023] [Revised: 12/20/2023] [Accepted: 12/21/2023] [Indexed: 01/28/2024] Open
Abstract
Regeneration of neurons has important implications for human health, and the retina provides an accessible system to study the potential of replacing neurons following injury. In previous work, we generated transgenic mice in which neurogenic transcription factors were expressed in Müller glia (MG) and showed that they stimulated neurogenesis following inner retinal damage. It was unknown, however, whether the timing or mode of injury mattered in this process. Here, we explored these parameters on induced neurogenesis from MG and show that MG expressing Ascl1 will generate new bipolar neurons with similar efficiency irrespective of injury mode or timing. However, MG that express Ascl1-Atoh1 produce a new type of retinal ganglion-like cell after outer retinal damage, which is absent with inner retinal damage. Our data suggest that although cell fate is primarily dictated by neurogenic transcription factors, the inflammatory state of MG relative to injury can influence the outcome of induced neurogenesis.
Collapse
Affiliation(s)
- Marina Pavlou
- Department of Biological Structure, University of Washington, Seattle, WA, USA; Institute for Stem Cells and Regenerative Medicine, University of Washington, Seattle, WA, USA
| | - Marlene Probst
- Department of Biological Structure, University of Washington, Seattle, WA, USA
| | - Nicolai Blasdel
- Department of Biological Structure, University of Washington, Seattle, WA, USA
| | - Aric R Prieve
- Department of Biological Structure, University of Washington, Seattle, WA, USA
| | - Thomas A Reh
- Department of Biological Structure, University of Washington, Seattle, WA, USA; Institute for Stem Cells and Regenerative Medicine, University of Washington, Seattle, WA, USA.
| |
Collapse
|
7
|
Pei L, Ouyang Z, Zhang H, Huang S, Jiang R, Liu B, Tang Y, Feng M, Yuan M, Wang H, Yao S, Shi S, Yu Z, Xu D, Gong G, Wei K. Thrombospondin 1 and Reelin act through Vldlr to regulate cardiac growth and repair. Basic Res Cardiol 2024; 119:169-192. [PMID: 38147128 DOI: 10.1007/s00395-023-01021-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Revised: 11/01/2023] [Accepted: 11/01/2023] [Indexed: 12/27/2023]
Abstract
Adult mammalian cardiomyocytes have minimal cell cycle capacity, which leads to poor regeneration after cardiac injury such as myocardial infarction. Many positive regulators of cardiomyocyte cell cycle and cardioprotective signals have been identified, but extracellular signals that suppress cardiomyocyte proliferation are poorly understood. We profiled receptors enriched in postnatal cardiomyocytes, and found that very-low-density-lipoprotein receptor (Vldlr) inhibits neonatal cardiomyocyte cell cycle. Paradoxically, Reelin, the well-known Vldlr ligand, expressed in cardiac Schwann cells and lymphatic endothelial cells, promotes neonatal cardiomyocyte proliferation. Thrombospondin1 (TSP-1), another ligand of Vldlr highly expressed in adult heart, was then found to inhibit cardiomyocyte proliferation through Vldlr, and may contribute to Vldlr's overall repression on proliferation. Mechanistically, Rac1 and subsequent Yap phosphorylation and nucleus translocation mediate the regulation of the cardiomyocyte cell cycle by TSP-1/Reelin-Vldlr signaling. Importantly, Reln mutant neonatal mice displayed impaired cardiomyocyte proliferation and cardiac regeneration after apical resection, while cardiac-specific Thbs1 deletion and cardiomyocyte-specific Vldlr deletion promote cardiomyocyte proliferation and are cardioprotective after myocardial infarction. Our results identified a novel role of Vldlr in consolidating extracellular signals to regulate cardiomyocyte cell cycle activity and survival, and the overall suppressive TSP-1-Vldlr signal may contribute to the poor cardiac repair capacity of adult mammals.
Collapse
Affiliation(s)
- Lijuan Pei
- School of Life Sciences and Technology, Institute for Regenerative Medicine, Shanghai Key Laboratory of Signaling and Disease Research, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Shanghai East Hospital, Shanghai Institute of Stem Cell Research and Clinical Translation, Tongji University, 1239 Siping Road, Shanghai, 200092, China
| | - Zhaohui Ouyang
- School of Life Sciences and Technology, Institute for Regenerative Medicine, Shanghai Key Laboratory of Signaling and Disease Research, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Shanghai East Hospital, Shanghai Institute of Stem Cell Research and Clinical Translation, Tongji University, 1239 Siping Road, Shanghai, 200092, China
| | - Hongjie Zhang
- School of Life Sciences and Technology, Institute for Regenerative Medicine, Shanghai Key Laboratory of Signaling and Disease Research, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Shanghai East Hospital, Shanghai Institute of Stem Cell Research and Clinical Translation, Tongji University, 1239 Siping Road, Shanghai, 200092, China
| | - Shiqi Huang
- School of Life Sciences and Technology, Institute for Regenerative Medicine, Shanghai Key Laboratory of Signaling and Disease Research, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Shanghai East Hospital, Shanghai Institute of Stem Cell Research and Clinical Translation, Tongji University, 1239 Siping Road, Shanghai, 200092, China
| | - Rui Jiang
- School of Life Sciences and Technology, Institute for Regenerative Medicine, Shanghai Key Laboratory of Signaling and Disease Research, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Shanghai East Hospital, Shanghai Institute of Stem Cell Research and Clinical Translation, Tongji University, 1239 Siping Road, Shanghai, 200092, China
| | - Bilin Liu
- Institute for Regenerative Medicine, School of Life Sciences and Technology, Shanghai East Hospital, Tongji University, Shanghai, 200092, China
| | - Yansong Tang
- Department of Cardiology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, 200072, China
| | - Mengying Feng
- School of Life Sciences and Technology, Institute for Regenerative Medicine, Shanghai Key Laboratory of Signaling and Disease Research, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Shanghai East Hospital, Shanghai Institute of Stem Cell Research and Clinical Translation, Tongji University, 1239 Siping Road, Shanghai, 200092, China
| | - Min Yuan
- School of Life Sciences and Technology, Institute for Regenerative Medicine, Shanghai Key Laboratory of Signaling and Disease Research, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Shanghai East Hospital, Shanghai Institute of Stem Cell Research and Clinical Translation, Tongji University, 1239 Siping Road, Shanghai, 200092, China
| | - Haocun Wang
- School of Life Sciences and Technology, Institute for Regenerative Medicine, Shanghai Key Laboratory of Signaling and Disease Research, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Shanghai East Hospital, Shanghai Institute of Stem Cell Research and Clinical Translation, Tongji University, 1239 Siping Road, Shanghai, 200092, China
| | - Su Yao
- School of Life Sciences and Technology, Institute for Regenerative Medicine, Shanghai Key Laboratory of Signaling and Disease Research, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Shanghai East Hospital, Shanghai Institute of Stem Cell Research and Clinical Translation, Tongji University, 1239 Siping Road, Shanghai, 200092, China
| | - Shuyue Shi
- School of Life Sciences and Technology, Institute for Regenerative Medicine, Shanghai Key Laboratory of Signaling and Disease Research, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Shanghai East Hospital, Shanghai Institute of Stem Cell Research and Clinical Translation, Tongji University, 1239 Siping Road, Shanghai, 200092, China
| | - Zhao Yu
- School of Life Sciences and Technology, Institute for Regenerative Medicine, Shanghai Key Laboratory of Signaling and Disease Research, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Shanghai East Hospital, Shanghai Institute of Stem Cell Research and Clinical Translation, Tongji University, 1239 Siping Road, Shanghai, 200092, China
| | - Dachun Xu
- Department of Cardiology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, 200072, China
| | - Guohua Gong
- Institute for Regenerative Medicine, School of Life Sciences and Technology, Shanghai East Hospital, Tongji University, Shanghai, 200092, China
| | - Ke Wei
- School of Life Sciences and Technology, Institute for Regenerative Medicine, Shanghai Key Laboratory of Signaling and Disease Research, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Shanghai East Hospital, Shanghai Institute of Stem Cell Research and Clinical Translation, Tongji University, 1239 Siping Road, Shanghai, 200092, China.
| |
Collapse
|
8
|
Mingo-Moreno N, Truschow P, Staiger JF, Wagener RJ. Caudally pronounced deficiencies in preplate splitting and migration underly a rostro-caudal progression of cortical lamination defects in the reeler brain. Cereb Cortex 2024; 34:bhae023. [PMID: 38383722 DOI: 10.1093/cercor/bhae023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Revised: 12/30/2023] [Accepted: 12/31/2023] [Indexed: 02/23/2024] Open
Abstract
In mammalian neocortex development, every cohort of newborn neurons is guided toward the marginal zone, leading to an "inside-out" organization of the 6 neocortical layers. This migratory pattern is regulated by the extracellular glycoprotein Reelin. The reeler mouse shows a homozygous mutation of the reelin gene. Using RNA in situ hybridization we could demonstrate that the Reelin-deficient mouse cortex (male and female) displays an increasing lamination defect along the rostro-caudal axis that is characterized by strong cellular intermingling, but roughly reproduces the "inside-out" pattern in rostral cortex, while caudal cortex shows a relative inversion of neuronal positioning ("outside-in"). We found that in development of the reeler cortex, preplate-splitting is also defective with an increasing severity along the rostro-caudal axis. This leads to a misplacement of subplate neurons that are crucial for a switch in migration mode within the cortical plate. Using Flash Tag labeling and nucleoside analog pulse-chasing, we found an according migration defect within the cortical plate, again with a progressive severity along the rostro-caudal axis. Thus, loss of one key player in neocortical development leads to highly area-specific (caudally pronounced) developmental deficiencies that result in multiple roughly opposite rostral versus caudal adult neocortical phenotypes.
Collapse
Affiliation(s)
- Nieves Mingo-Moreno
- Institute for Neuroanatomy, University Medical Center Göttingen, Göttingen 37075, Germany
- Center for Nanoscale Microscopy and Molecular Physiology of the Brain, Göttingen 37073, Germany
| | - Pavel Truschow
- Institute for Neuroanatomy, University Medical Center Göttingen, Göttingen 37075, Germany
| | - Jochen F Staiger
- Institute for Neuroanatomy, University Medical Center Göttingen, Göttingen 37075, Germany
- Center for Nanoscale Microscopy and Molecular Physiology of the Brain, Göttingen 37073, Germany
| | - Robin J Wagener
- Institute for Neuroanatomy, University Medical Center Göttingen, Göttingen 37075, Germany
- Department of Neurology, University Hospital Heidelberg, Heidelberg 69120, Germany
- Clinical Cooperation Unit Neurooncology, German Cancer Research Center (DKFZ), Heidelberg 69120, Germany
| |
Collapse
|
9
|
Zhang ML, Zhou KM, Wang XW. Identification and characterization of a Reeler domain containing protein in Procambarus clarkii provides new insights into antibacterial immunity in crustacean. FISH AND SHELLFISH IMMUNOLOGY REPORTS 2023; 4:100094. [PMID: 37131543 PMCID: PMC10149183 DOI: 10.1016/j.fsirep.2023.100094] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 04/16/2023] [Accepted: 04/17/2023] [Indexed: 05/04/2023] Open
Abstract
Crayfish, as an invertebrate, relies only on the innate immune system to resist external pathogens. In this study, a molecule containing a single Reeler domain was identified from red swamp crayfish Procambarus clarkii (named as PcReeler). Tissue distribution analysis showed that PcReeler was highly expressed in gills and its expression was induced by bacterial stimulation. Inhibiting the expression of PcReeler by RNA interference led to a significant increase in the bacterial abundance in the gills of crayfish, and a significant increase in the crayfish mortality. Silencing of PcReeler influenced the stability of the microbiota in the gills revealed by 16S rDNA high-throughput sequencing. Recombinant PcReeler showed the ability to bind microbial polysaccharide and bacteria and to inhibit the formation of bacterial biofilms. These results provided direct evidence for the involvement of PcReeler in the antibacterial immune mechanism of P. clarkii.
Collapse
Affiliation(s)
- Ming-Lu Zhang
- Shandong Provincial Key Laboratory of Animal Cells and Developmental Biology, School of Life Sciences, Shandong University, Qingdao, China
| | - Kai-Min Zhou
- Shandong Provincial Key Laboratory of Animal Cells and Developmental Biology, School of Life Sciences, Shandong University, Qingdao, China
| | - Xian-Wei Wang
- Shandong Provincial Key Laboratory of Animal Cells and Developmental Biology, School of Life Sciences, Shandong University, Qingdao, China
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao, China
- Corresponding author at: School of Life Sciences, Shandong University, Qingdao 266237, China.
| |
Collapse
|
10
|
Matrone C, Ferretti G. Semaphorin 3A influences neuronal processes that are altered in patients with autism spectrum disorder: Potential diagnostic and therapeutic implications. Neurosci Biobehav Rev 2023; 153:105338. [PMID: 37524141 DOI: 10.1016/j.neubiorev.2023.105338] [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: 02/16/2023] [Revised: 07/27/2023] [Accepted: 07/28/2023] [Indexed: 08/02/2023]
Abstract
Autism spectrum disorder (ASD) is a pervasive disorder that most frequently manifests in early childhood and lasts for their entire lifespan. Several behavioural traits characterise the phenotype of patients with ASD, including difficulties in reciprocal social communication as well as compulsive/repetitive stereotyped verbal and non-verbal behaviours. Although multiple hypotheses have been proposed to explain the aetiology of ASD and many resources have been used to improve our understanding of ASD, several aspects remain largely unexplored. Class 3 semaphorins (SEMA3) are secreted proteins involved in the organisation of structural and functional connectivity in the brain that regulate synaptic and dendritic development. Alterations in brain connectivity and aberrant neuronal development have been described in some patients with ASD. Mutations and polymorphisms in SEMA3A and alterations in its receptors and signalling have been associated with some neurological disorders such as schizophrenia and epilepsy, which are comorbidities in ASD, but also with ASD itself. In addition, SEMA3A is a key regulator of the immune response and neuroinflammatory processes, which have been found to be dysregulated in mothers of children who develop ASD and in affected patients. In this review, we highlight neurodevelopmental-related processes in which SEMA3A is involved, which are altered in ASD, and provide a viewpoint emphasising the development of strategies targeting changes in the SEMA3A signal to identify patterns of anomalies distinctive of ASD or to predict the prognosis of affected patients.
Collapse
Affiliation(s)
- Carmela Matrone
- Division of Pharmacology, Department of Neuroscience, School of Medicine, University of Naples "Federico II", Via Pansini 5, 80131 Naples, Italy.
| | - Gabriella Ferretti
- Division of Pharmacology, Department of Neuroscience, School of Medicine, University of Naples "Federico II", Via Pansini 5, 80131 Naples, Italy
| |
Collapse
|
11
|
Nakagawa N, Iwasato T. Golgi polarity shift instructs dendritic refinement in the neonatal cortex by mediating NMDA receptor signaling. Cell Rep 2023; 42:112843. [PMID: 37516101 DOI: 10.1016/j.celrep.2023.112843] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 05/15/2023] [Accepted: 07/05/2023] [Indexed: 07/31/2023] Open
Abstract
Dendritic refinement is a critical component of activity-dependent neuronal circuit maturation, through which individual neurons establish specific connectivity with their target axons. Here, we demonstrate that the developmental shift of Golgi polarity is a key process in dendritic refinement. During neonatal development, the Golgi apparatus in layer 4 spiny stellate (SS) neurons in the mouse barrel cortex lose their original apical positioning and acquire laterally polarized distributions. This lateral Golgi polarity, which is oriented toward the barrel center, peaks on postnatal days 5-7 (P5-P7) and disappears by P15, which aligns with the developmental time course of SS neuron dendritic refinement. Genetic ablation of N-methyl-D-aspartate (NMDA) receptors, key players in dendritic refinement, disturbs the lateral Golgi polarity. Golgi polarity manipulation disrupts the asymmetric dendritic projection pattern and the primary-whisker-specific response of SS neurons. Our results elucidate activity-dependent Golgi dynamics and their critical role in developmental neuronal circuit refinement.
Collapse
Affiliation(s)
- Naoki Nakagawa
- Laboratory of Mammalian Neural Circuits, National Institute of Genetics (NIG), Mishima, Shizuoka 411-8540, Japan; Graduate Institute for Advanced Studies, SOKENDAI, Mishima, Shizuoka 411-8540, Japan.
| | - Takuji Iwasato
- Laboratory of Mammalian Neural Circuits, National Institute of Genetics (NIG), Mishima, Shizuoka 411-8540, Japan; Graduate Institute for Advanced Studies, SOKENDAI, Mishima, Shizuoka 411-8540, Japan.
| |
Collapse
|
12
|
Zhao J, Lang M. New insight into protein glycosylation in the development of Alzheimer's disease. Cell Death Discov 2023; 9:314. [PMID: 37626031 PMCID: PMC10457297 DOI: 10.1038/s41420-023-01617-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Revised: 08/09/2023] [Accepted: 08/16/2023] [Indexed: 08/27/2023] Open
Abstract
Alzheimer's disease (AD) is a chronic neurodegenerative disease that seriously endangers the physical and mental health of patients, however, there are still no effective drugs or methods to cure this disease up to now. Protein glycosylation is the most common modifications of the translated proteins in eukaryotic cells. Recently many researches disclosed that aberrant glycosylation happens in some important AD-related proteins, such as APP, Tau, Reelin and CRMP-2, etc, suggesting a close link between abnormal protein glycosylation and AD. Because of its complexity and diversity, glycosylation is thus considered a completely new entry point for understanding the precise cause of AD. This review comprehensively summarized the currently discovered changes in protein glycosylation patterns in AD, and especially introduced the latest progress on the mechanism of protein glycosylation affecting the progression of AD and the potential application of protein glycosylation in AD detection and treatment, thereby providing a wide range of opportunities for uncovering the pathogenesis of AD and promoting the translation of glycosylation research into future clinical applications.
Collapse
Affiliation(s)
- Jingwei Zhao
- CAS Center for Excellence in Biotic Interactions, College of Life Science, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Minglin Lang
- CAS Center for Excellence in Biotic Interactions, College of Life Science, University of Chinese Academy of Sciences, Beijing, 100049, China.
- College of Life Science, Agricultural University of Hebei, Baoding, 071000, China.
| |
Collapse
|
13
|
Kim M, Jun S, Park H, Tanaka-Yamamoto K, Yamamoto Y. Regulation of cerebellar network development by granule cells and their molecules. Front Mol Neurosci 2023; 16:1236015. [PMID: 37520428 PMCID: PMC10375027 DOI: 10.3389/fnmol.2023.1236015] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Accepted: 06/27/2023] [Indexed: 08/01/2023] Open
Abstract
The well-organized cerebellar structures and neuronal networks are likely crucial for their functions in motor coordination, motor learning, cognition, and emotion. Such cerebellar structures and neuronal networks are formed during developmental periods through orchestrated mechanisms, which include not only cell-autonomous programs but also interactions between the same or different types of neurons. Cerebellar granule cells (GCs) are the most numerous neurons in the brain and are generated through intensive cell division of GC precursors (GCPs) during postnatal developmental periods. While GCs go through their own developmental processes of proliferation, differentiation, migration, and maturation, they also play a crucial role in cerebellar development. One of the best-characterized contributions is the enlargement and foliation of the cerebellum through massive proliferation of GCPs. In addition to this contribution, studies have shown that immature GCs and GCPs regulate multiple factors in the developing cerebellum, such as the development of other types of cerebellar neurons or the establishment of afferent innervations. These studies have often found impairments of cerebellar development in animals lacking expression of certain molecules in GCs, suggesting that the regulations are mediated by molecules that are secreted from or present in GCs. Given the growing recognition of GCs as regulators of cerebellar development, this review will summarize our current understanding of cerebellar development regulated by GCs and molecules in GCs, based on accumulated studies and recent findings, and will discuss their potential further contributions.
Collapse
Affiliation(s)
- Muwoong Kim
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, Republic of Korea
- Division of Bio-Medical Science and Technology, KIST School, University of Science and Technology (UST), Seoul, Republic of Korea
| | - Soyoung Jun
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, Republic of Korea
- Division of Bio-Medical Science and Technology, KIST School, University of Science and Technology (UST), Seoul, Republic of Korea
| | - Heeyoun Park
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, Republic of Korea
| | - Keiko Tanaka-Yamamoto
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, Republic of Korea
- Division of Bio-Medical Science and Technology, KIST School, University of Science and Technology (UST), Seoul, Republic of Korea
| | - Yukio Yamamoto
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, Republic of Korea
| |
Collapse
|
14
|
Demir R, Deveci R. In silico analysis of the possible crosstalk between O-linked β-GlcNAcylation and phosphorylation sites of Disabled 1 adaptor protein in vertebrates. Amino Acids 2023:10.1007/s00726-023-03266-5. [PMID: 37067567 DOI: 10.1007/s00726-023-03266-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Accepted: 04/10/2023] [Indexed: 04/18/2023]
Abstract
Disabled 1 (Dab1) is an adaptor protein with essential functions regulated by reelin signaling and affects many biological processes in the nervous system, including cell motility, adhesion, cortical development, maturation, and synaptic plasticity. Posttranslational modifications directly guide the fates of cytoplasmic proteins to complete their functions correctly. Reciprocal crosstalk between O-GlcNAcylation and phosphorylation is a dynamic modification in cytoplasmic proteins. It modulates the functions of the proteins by regulating their interactions with other molecules in response to the continuously changeable cell microenvironment. Although Dab1 contains conserved recognition sites for phosphorylation in their N-terminal protein interaction domain, the O-β-GlcNAcylation and phosphorylation sites of human Dab1 sequence, their reciprocal crosstalk, and potential kinases catalyzing the phosphorylation remain unknown. In this study, we determined potential thirty-seven O-β-GlcNAcylation and sixty-seven phosphorylation sites. Conserved twenty-one residues of these glycosylated sites were also phosphorylated with various kinases, including ATM, CKI, DNAPK, GSK3, PKC, PKG, RSK, cdc2, cdk5, and p38MAPK. In addition, we analyzed these conserved sites at our constructed two- and three-dimensional structures of human Dab1 protein. Dab1 protein models were frequently composed of coil structures as well as α-helix and β-strands. Many of these conserved crosstalk sites between O-β-GlcNAcylation and phosphorylation were localized at the coil region of the protein model. These findings may guide biochemical, genetic, and glyco-biology based on further experiments about the Dab1 signaling process. Understanding these modifications might change the point of view of the Dab1 signaling process and treatment for pathological conditions in neurodegenerative diseases such as Alzheimer's disease.
Collapse
Affiliation(s)
- Ramiz Demir
- Molecular Biology Section, Department of Biology, Faculty of Science, Ege University, Bornova, 35040,, Izmir, Turkey
- Graduate School of Health Science, Koç University Research Center for Translational Medicine (KUTTAM), Koç University, 34010, Istanbul, Turkey
| | - Remziye Deveci
- Molecular Biology Section, Department of Biology, Faculty of Science, Ege University, Bornova, 35040,, Izmir, Turkey.
| |
Collapse
|
15
|
Immunoexpression Pattern of Autophagy Markers in Developing and Postnatal Kidneys of Dab1−/−(yotari) Mice. Biomolecules 2023; 13:biom13030402. [PMID: 36979337 PMCID: PMC10046325 DOI: 10.3390/biom13030402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Revised: 02/17/2023] [Accepted: 02/19/2023] [Indexed: 02/24/2023] Open
Abstract
The purpose of this study was to compare the immunofluorescence patterns of autophagic markers: Light chain 3 beta (LC3B), Glucose regulating protein 78 (GRP78), Heat shock cognate 71 (HSC70) and Lysosomal-associated membrane protein 2A (LAMP2A) in the developing and postnatal kidneys of Dab1−/− (yotari) mice to those of wild-type samples. Embryos were obtained on gestation days 13.5 and 15.5 (E13.5 and E15.5), and adult animals were sacrificed at postnatal days 4, 11 and 14 (P4, P11, and P14). After fixation and dehydration, paraffin-embedded kidney tissues were sectioned and incubated with specific antibodies. Using an immunofluorescence microscope, sections were analyzed. For statistical analysis, a two-way ANOVA test and a Tukey’s multiple comparison test were performed with a probability level of p < 0.05. A significant increase in GRP78 and LAMP2A expression was observed in the renal vesicles and convoluted tubules of yotari in embryonic stages. In postnatal kidneys, all observed proteins showed higher signal intensities in proximal and distal convoluted tubules of yotari, while a higher percentage of LC3B-positive cells was also observed in glomeruli. Our findings suggest that all of the examined autophagic markers play an important role in normal kidney development, as well as the potential importance of these proteins in renal pathology, where they primarily serve a protective function and thus may be used as diagnostic and therapeutic targets.
Collapse
|
16
|
XiaoHong Z, Shuo Y, GeHong D, AnChao Y, Ce W, YunYun D, Can W, SiJie H, Feng C, WenBin L. Inflammatory brain lesions preceding primary central nervous system lymphoma: a case report and genetic analysis. Neurol Sci 2023; 44:1555-1561. [PMID: 36599976 PMCID: PMC10102048 DOI: 10.1007/s10072-022-06587-7] [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: 09/18/2022] [Accepted: 12/24/2022] [Indexed: 01/06/2023]
Abstract
BACKGROUND Primary central nervous system lymphoma (PCNSL) is an aggressive extranodal lymphoma exclusively occurring within the central nervous system. Inflammatory brain lesions as "sentinel lesions" of PCNSL are very rare. We present a rare case of PCNSL with preceding inflammatory lesions in an immunocompetent patient who underwent two biopsies, one craniotomy and two genetic testing. CASE REPORT A 66-year-old male patient presented with left limb weakness and ataxia. Brain magnetic resonance imaging showed a contrast-enhancing lesion with perifocal brain edema in the near midline of right frontal lobe. Histological examination of a brain biopsy specimen revealed inflammatory lesion characteristics with infiltration of T-cell dominant lymphocytes and few B-cell. Given that the patient developed cerebral hematoma after biopsy, lesion resection by craniotomy was performed. An excised sample demonstrated mixed T-cell and B-cell infiltrating inflammatory lesions. Four months after total resection of the right frontal lobe lesion, another lesion appeared in the left frontal parietal lobe, which was diagnosed as diffuse large B-cell lymphoma by biopsy. In addition, genetic testing of the lesions at two different locations was performed, and the results showed that the inflammatory lesions had the same three gene (RELN, PCLO, and CREBBP) mutations as PCNSL. Interestingly, the three mutated genes are associated with tumor. CONCLUSION Our present case is the first to demonstrate inflammatory brain lesions heralding PCNSL from genetic and pathological perspectives. This may help clinicians to select new auxiliary diagnostic methods for timely diagnosis of patients with suspected PCNSL.
Collapse
Affiliation(s)
- Zheng XiaoHong
- Department of Neuro-Oncology, Cancer Center, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Yin Shuo
- Department of Neuro-Oncology, Cancer Center, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Dong GeHong
- Department of Pathology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Yang AnChao
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Wang Ce
- Department of Neuro-Oncology, Cancer Center, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Duan YunYun
- Department of Radiology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Wang Can
- Department of Neuro-Oncology, Cancer Center, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Huang SiJie
- Department of Neuro-Oncology, Cancer Center, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Chen Feng
- Department of Neuro-Oncology, Cancer Center, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.
| | - Li WenBin
- Department of Neuro-Oncology, Cancer Center, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.
| |
Collapse
|
17
|
Pardo M, Gregorio S, Montalban E, Pujadas L, Elias-Tersa A, Masachs N, Vílchez-Acosta A, Parent A, Auladell C, Girault JA, Vila M, Nairn AC, Manso Y, Soriano E. Adult-specific Reelin expression alters striatal neuronal organization: implications for neuropsychiatric disorders. Front Cell Neurosci 2023; 17:1143319. [PMID: 37153634 PMCID: PMC10157100 DOI: 10.3389/fncel.2023.1143319] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Accepted: 03/27/2023] [Indexed: 05/10/2023] Open
Abstract
In addition to neuronal migration, brain development, and adult plasticity, the extracellular matrix protein Reelin has been extensively implicated in human psychiatric disorders such as schizophrenia, bipolar disorder, and autism spectrum disorder. Moreover, heterozygous reeler mice exhibit features reminiscent of these disorders, while overexpression of Reelin protects against its manifestation. However, how Reelin influences the structure and circuits of the striatal complex, a key region for the above-mentioned disorders, is far from being understood, especially when altered Reelin expression levels are found at adult stages. In the present study, we took advantage of complementary conditional gain- and loss-of-function mouse models to investigate how Reelin levels may modify adult brain striatal structure and neuronal composition. Using immunohistochemical techniques, we determined that Reelin does not seem to influence the striatal patch and matrix organization (studied by μ-opioid receptor immunohistochemistry) nor the density of medium spiny neurons (MSNs, studied with DARPP-32). We show that overexpression of Reelin leads to increased numbers of striatal parvalbumin- and cholinergic-interneurons, and to a slight increase in tyrosine hydroxylase-positive projections. We conclude that increased Reelin levels might modulate the numbers of striatal interneurons and the density of the nigrostriatal dopaminergic projections, suggesting that these changes may be involved in the protection of Reelin against neuropsychiatric disorders.
Collapse
Affiliation(s)
- Mònica Pardo
- Developmental Neurobiology and Regeneration Laboratory, Department of Cell Biology, Physiology and Immunology, Institute of Neurosciences, Universitat de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
| | - Sara Gregorio
- Developmental Neurobiology and Regeneration Laboratory, Department of Cell Biology, Physiology and Immunology, Institute of Neurosciences, Universitat de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
| | - Enrica Montalban
- Institut du Fer à Moulin UMR-S 1270, INSERM, Sorbonne University, Paris, France
| | - Lluís Pujadas
- Developmental Neurobiology and Regeneration Laboratory, Department of Cell Biology, Physiology and Immunology, Institute of Neurosciences, Universitat de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
- Department of Experimental Sciences and Methodology, Faculty of Health Science and Welfare, University of Vic – Central University of Catalonia (UVic-UCC), Vic, Spain
- Tissue Repair and Regeneration Laboratory (TR2Lab), Institut de Recerca i Innovació en Ciències de la Vida i de la Salut a la Catalunya Central (IRIS-CC), Barcelona, Spain
| | - Alba Elias-Tersa
- Developmental Neurobiology and Regeneration Laboratory, Department of Cell Biology, Physiology and Immunology, Institute of Neurosciences, Universitat de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
| | - Núria Masachs
- Developmental Neurobiology and Regeneration Laboratory, Department of Cell Biology, Physiology and Immunology, Institute of Neurosciences, Universitat de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
| | - Alba Vílchez-Acosta
- Developmental Neurobiology and Regeneration Laboratory, Department of Cell Biology, Physiology and Immunology, Institute of Neurosciences, Universitat de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
| | - Annabelle Parent
- Neurodegenerative Diseases Research Group, Vall d’Hebron Research Institute, Barcelona, Spain
| | - Carme Auladell
- Developmental Neurobiology and Regeneration Laboratory, Department of Cell Biology, Physiology and Immunology, Institute of Neurosciences, Universitat de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
| | | | - Miquel Vila
- Centro de Investigación Biomédica en Red Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
- Neurodegenerative Diseases Research Group, Vall d’Hebron Research Institute, Barcelona, Spain
- Department of Biochemistry and Molecular Biology, Autonomous University of Barcelona (UAB), Barcelona, Spain
- Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD, United States
| | - Angus C. Nairn
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, United States
| | - Yasmina Manso
- Developmental Neurobiology and Regeneration Laboratory, Department of Cell Biology, Physiology and Immunology, Institute of Neurosciences, Universitat de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
- Yasmina Manso,
| | - Eduardo Soriano
- Developmental Neurobiology and Regeneration Laboratory, Department of Cell Biology, Physiology and Immunology, Institute of Neurosciences, Universitat de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
- *Correspondence: Eduardo Soriano,
| |
Collapse
|
18
|
Demir R, Deveci R. In silico analysis of glycosylation pattern in 5 th-6 th repeat sequence of reelin glycoprotein. J Biomol Struct Dyn 2022; 40:10065-10073. [PMID: 34121615 DOI: 10.1080/07391102.2021.1938682] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Reelin is an extracellular matrix glycoprotein that plays a key role in cortical development, maturation, synaptic plasticity, and memory formation in the adult mammalian brain. Glycosylation is a significant post- and co-translational modification of proteins. Although glycosylation contributes to the characteristic of proteins from their production to molecular interactions, the knowledge about the glycosylation pattern of reelin is very limited. In this study, we aimed to predict the potential glycosylation pattern of the 5th-6th repeat of central reelin fragment that responsible for their signaling, by using in silico methods. We found that the predicted glycosylation pattern of the 5th-6th repeat of human reelin was highly conserved between vertebrate species. However, this conservation was not observed in analyzed invertebrates. For the first time, we described the sites of glycosylation at a three-dimensional protein structure in human reelin. Because the sites were very closed to EGF-like repeats and receptor binding sites, they could contribute the interaction with a partner of reelin in addition to the effect of thermostability to protein. Many of the residues related glycosylation were also conserved in analyzed species. These findings may guide biochemical, genetic, and glycobiology base on further experiments about reelin glycosylation. The understanding of reelin glycosylation might change the point of view of treatment for many pathological conditions in neurodegenerative diseases such as Alzheimer's disease. Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Ramiz Demir
- Faculty of Science, Department of Biology, Molecular Biology Section, Ege University, Izmir, Turkey.,The Graduate School of Health Sciences, Koç University Research Center for Translational Medicine (KUTTAM), Koç University, Istanbul, Turkey
| | - Remziye Deveci
- Faculty of Science, Department of Biology, Molecular Biology Section, Ege University, Izmir, Turkey
| |
Collapse
|
19
|
Cho E, Kim K, Kim H, Cho SR. Reelin protects against pathological α-synuclein accumulation and dopaminergic neurodegeneration after environmental enrichment in Parkinson's disease. Neurobiol Dis 2022; 175:105898. [DOI: 10.1016/j.nbd.2022.105898] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Revised: 09/25/2022] [Accepted: 10/12/2022] [Indexed: 11/05/2022] Open
|
20
|
Fisher E, Feng J. RNA splicing regulators play critical roles in neurogenesis. WILEY INTERDISCIPLINARY REVIEWS. RNA 2022; 13:e1728. [PMID: 35388651 DOI: 10.1002/wrna.1728] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Revised: 03/07/2022] [Accepted: 03/09/2022] [Indexed: 06/14/2023]
Abstract
Alternative RNA splicing increases transcript diversity in different cell types and under varying conditions. It is executed with the help of RNA splicing regulators (RSRs), which are operationally defined as RNA-binding proteins (RBPs) that regulate alternative splicing, but not directly catalyzing the chemical reactions of splicing. By systematically searching for RBPs and manually identifying those that regulate splicing, we curated 305 RSRs in the human genome. Surprisingly, most of the RSRs are involved in neurogenesis. Among these RSRs, we focus on nine families (PTBP, NOVA, RBFOX, ELAVL, CELF, DBHS, MSI, PCBP, and MBNL) that play essential roles in the neurogenic pathway. A better understanding of their functions will provide novel insights into the role of splicing in brain development, health, and disease. This comprehensive review serves as a stepping-stone to explore the diverse and complex set of RSRs as fundamental regulators of neural development. This article is categorized under: RNA-Based Catalysis > RNA Catalysis in Splicing and Translation RNA Interactions with Proteins and Other Molecules > Protein-RNA Interactions: Functional Implications RNA Processing > Splicing Regulation/Alternative Splicing.
Collapse
Affiliation(s)
- Emily Fisher
- Department of Physiology and Biophysics, State University of New York at Buffalo, Buffalo, New York, USA
- Veterans Affairs Western New York Healthcare System, Buffalo, New York, USA
| | - Jian Feng
- Department of Physiology and Biophysics, State University of New York at Buffalo, Buffalo, New York, USA
- Veterans Affairs Western New York Healthcare System, Buffalo, New York, USA
| |
Collapse
|
21
|
Nguyen H, Sokpor G, Parichha A, Pham L, Saikhedkar N, Xie Y, Ulmke PA, Rosenbusch J, Pirouz M, Behr R, Stoykova A, Brand-Saberi B, Nguyen HP, Staiger JF, Tole S, Tuoc T. BAF (mSWI/SNF) complex regulates mediolateral cortical patterning in the developing forebrain. Front Cell Dev Biol 2022; 10:1011109. [PMID: 36263009 PMCID: PMC9573979 DOI: 10.3389/fcell.2022.1011109] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Accepted: 09/16/2022] [Indexed: 11/24/2022] Open
Abstract
Early forebrain patterning entails the correct regional designation of the neuroepithelium, and appropriate specification, generation, and distribution of neural cells during brain development. Specific signaling and transcription factors are known to tightly regulate patterning of the dorsal telencephalon to afford proper structural/functional cortical arealization and morphogenesis. Nevertheless, whether and how changes of the chromatin structure link to the transcriptional program(s) that control cortical patterning remains elusive. Here, we report that the BAF chromatin remodeling complex regulates the spatiotemporal patterning of the mouse dorsal telencephalon. To determine whether and how the BAF complex regulates cortical patterning, we conditionally deleted the BAF complex scaffolding subunits BAF155 and BAF170 in the mouse dorsal telencephalic neuroepithelium. Morphological and cellular changes in the BAF mutant forebrain were examined using immunohistochemistry and in situ hybridization. RNA sequencing, Co-immunoprecipitation, and mass spectrometry were used to investigate the molecular basis of BAF complex involvement in forebrain patterning. We found that conditional ablation of BAF complex in the dorsal telencephalon neuroepithelium caused expansion of the cortical hem and medial cortex beyond their developmental boundaries. Consequently, the hippocampal primordium is not specified, the mediolateral cortical patterning is compromised, and the cortical identity is disturbed in the absence of BAF complex. The BAF complex was found to interact with the cortical hem suppressor LHX2. The BAF complex suppresses cortical hem fate to permit proper forebrain patterning. We provide evidence that BAF complex modulates mediolateral cortical patterning possibly by interacting with the transcription factor LHX2 to drive the LHX2-dependent transcriptional program essential for dorsal telencephalon patterning. Our data suggest a putative mechanistic synergy between BAF chromatin remodeling complex and LHX2 in regulating forebrain patterning and ontogeny.
Collapse
Affiliation(s)
- Huong Nguyen
- Institute for Neuroanatomy, University Medical Center, Georg-August-University Goettingen, Goettingen, Germany
- Faculty of Biotechnology, Thai Nguyen University of Sciences, Thai Nguyen, Vietnam
| | - Godwin Sokpor
- Institute for Neuroanatomy, University Medical Center, Georg-August-University Goettingen, Goettingen, Germany
- Department of Human Genetics, Ruhr University Bochum, Bochum, Germany
- Department of Anatomy and Molecular Embryology, Ruhr University Bochum, Bochum, Germany
| | | | - Linh Pham
- Institute for Neuroanatomy, University Medical Center, Georg-August-University Goettingen, Goettingen, Germany
- Department of Human Genetics, Ruhr University Bochum, Bochum, Germany
| | | | - Yuanbin Xie
- Institute for Neuroanatomy, University Medical Center, Georg-August-University Goettingen, Goettingen, Germany
| | - Pauline Antonie Ulmke
- Institute for Neuroanatomy, University Medical Center, Georg-August-University Goettingen, Goettingen, Germany
- Department of Human Genetics, Ruhr University Bochum, Bochum, Germany
| | - Joachim Rosenbusch
- Institute for Neuroanatomy, University Medical Center, Georg-August-University Goettingen, Goettingen, Germany
| | - Mehdi Pirouz
- Max Planck Institute for Multidisciplinary Sciences, Goettingen, Germany
- Harvard Stem Cell Institute, Harvard Medical School, Boston, MA, United States
| | - Rüdiger Behr
- German Primate Center-Leibniz Institute for Primate Research, Goettingen, Germany
| | | | - Beate Brand-Saberi
- Department of Anatomy and Molecular Embryology, Ruhr University Bochum, Bochum, Germany
| | - Huu Phuc Nguyen
- Department of Human Genetics, Ruhr University Bochum, Bochum, Germany
| | - Jochen F. Staiger
- Institute for Neuroanatomy, University Medical Center, Georg-August-University Goettingen, Goettingen, Germany
| | - Shubha Tole
- Tata Institute of Fundamental Research, Mumbai, India
- *Correspondence: Shubha Tole, ; Tran Tuoc,
| | - Tran Tuoc
- Institute for Neuroanatomy, University Medical Center, Georg-August-University Goettingen, Goettingen, Germany
- Department of Human Genetics, Ruhr University Bochum, Bochum, Germany
- *Correspondence: Shubha Tole, ; Tran Tuoc,
| |
Collapse
|
22
|
Mezheritskiy MI, Dyakonova VE. Direct and Inherited Epigenetic Changes in the Nervous System Caused by Intensive Locomotion: Possible Adaptive Significance. Russ J Dev Biol 2022. [DOI: 10.1134/s1062360422050058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Abstract
This review is devoted to the analysis of works that investigated the long-term effects of species-specific forms of intensive locomotion on the cognitive functions of animals and humans, which can be transmitted to the next generation. To date, the anxiolytic and cognitive-enhancing long-term effects of intensive locomotion have been demonstrated in humans, rodents, fish, insects, mollusks, and nematodes. In rodents, changes in the central nervous system caused by intense locomotion can be transmitted through the maternal and paternal line to the descendants of the first generation. These include reduced anxiety, improved spatial learning and memory, increased levels of brain neurotrophic factor and vascular endothelial growth factor in the hippocampus and frontal cortex. The shift of the balance of histone acetylation in the hippocampus of rodents towards hyperacetylation, and the balance of DNA methylation towards demethylation manifests itself both as a direct and as a first-generation inherited effect of motor activity. The question about the mechanisms that link locomotion with an increase in the plasticity of a genome in the brain of descendants remains poorly understood, and invertebrate model organisms can be an ideal object for its study. Currently, there is a lack of a theoretical model explaining why motor activity leads to long-term improvement of some cognitive functions that can be transmitted to the next generation and why such an influence could have appeared in evolution. The answer to these questions is not only of fundamental interest, but it is necessary for predicting therapeutic and possible side effects of motor activity in humans. In this regard, the article pays special attention to the review of ideas on the evolutionary aspects of the problem. We propose our own hypothesis, according to which the activating effect of intensive locomotion on the function of the nervous system could have been formed in evolution as a preadaptation to a possible entry into a new environment.
Collapse
|
23
|
Specific contribution of Reelin expressed by Cajal-Retzius cells or GABAergic interneurons to cortical lamination. Proc Natl Acad Sci U S A 2022; 119:e2120079119. [PMID: 36067316 PMCID: PMC9477240 DOI: 10.1073/pnas.2120079119] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The extracellular protein Reelin, expressed by Cajal-Retzius (CR) cells at early stages of cortical development and at late stages by GABAergic interneurons, regulates radial migration and the "inside-out" pattern of positioning. Current models of Reelin functions in corticogenesis focus on early CR cell-derived Reelin in layer I. However, developmental disorders linked to Reelin deficits, such as schizophrenia and autism, are related to GABAergic interneuron-derived Reelin, although its role in migration has not been established. Here we selectively inactivated the Reln gene in CR cells or GABAergic interneurons. We show that CR cells have a major role in the inside-out order of migration, while CR and GABAergic cells sequentially cooperate to prevent invasion of cortical neurons into layer I. Furthermore, GABAergic cell-derived Reelin compensates some features of the reeler phenotype and is needed for the fine tuning of the layer-specific distribution of cortical neurons. In the hippocampus, the inactivation of Reelin in CR cells causes dramatic alterations in the dentate gyrus and mild defects in the hippocampus proper. These findings lead to a model in which both CR and GABAergic cell-derived Reelin cooperate to build the inside-out order of corticogenesis, which might provide a better understanding of the mechanisms involved in the pathogenesis of neuropsychiatric disorders linked to abnormal migration and Reelin deficits.
Collapse
|
24
|
de Guglielmo G, Iemolo A, Nur A, Turner A, Montilla-Perez P, Martinez A, Crook C, Roberts A, Telese F. Reelin deficiency exacerbates cocaine-induced hyperlocomotion by enhancing neuronal activity in the dorsomedial striatum. GENES, BRAIN, AND BEHAVIOR 2022; 21:e12828. [PMID: 35906757 PMCID: PMC9744517 DOI: 10.1111/gbb.12828] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Revised: 07/15/2022] [Accepted: 07/20/2022] [Indexed: 11/30/2022]
Abstract
The Reln gene encodes for the extracellular glycoprotein Reelin, which regulates several brain functions from development to adulthood, including neuronal migration, dendritic growth and branching and synapse formation and plasticity. Human studies have implicated Reelin signaling in several neurodevelopmental and psychiatric disorders. Mouse studies using the heterozygous Reeler (HR) mice have shown that reduced levels of Reln expression are associated with deficits in learning and memory and increased disinhibition. Although these traits are relevant to substance use disorders, the role of Reelin in cellular and behavioral responses to addictive drugs remains largely unknown. Here, we compared HR mice to wild-type (WT) littermate controls to investigate whether Reelin signaling contributes to the hyperlocomotor and rewarding effects of cocaine. After a single or repeated cocaine injections, HR mice showed enhanced cocaine-induced locomotor activity compared with WT controls. This effect persisted after withdrawal. In contrast, Reelin deficiency did not induce cocaine sensitization, and did not affect the rewarding effects of cocaine measured in the conditioned place preference assay. The elevated cocaine-induced hyperlocomotion in HR mice was associated with increased protein Fos expression in the dorsal medial striatum (DMS) compared with WT. Lastly, we performed an RNA fluorescent in situ hybridization experiment and found that Reln was highly co-expressed with the Drd1 gene, which encodes for the dopamine receptor D1, in the DMS. These findings show that Reelin signaling contributes to the locomotor effects of cocaine and improve our understanding of the neurobiological mechanisms underlying the cellular and behavioral effects of cocaine.
Collapse
Affiliation(s)
- Giordano de Guglielmo
- Department of Psychiatry, University of California, San Diego, La Jolla, California, USA
| | - Attilio Iemolo
- Department of Medicine, University of California, San Diego, La Jolla, California, USA
| | - Aisha Nur
- Department of Medicine, University of California, San Diego, La Jolla, California, USA
| | - Andrew Turner
- Department of Medicine, University of California, San Diego, La Jolla, California, USA
| | | | - Angelica Martinez
- Department of Psychiatry, University of California, San Diego, La Jolla, California, USA
| | - Caitlin Crook
- Department of Psychiatry, University of California, San Diego, La Jolla, California, USA
| | - Amanda Roberts
- Animal Models Core Facility, The Scripps Research Institute, La Jolla, California, USA
| | - Francesca Telese
- Department of Medicine, University of California, San Diego, La Jolla, California, USA
| |
Collapse
|
25
|
López-Mengual A, Segura-Feliu M, Sunyer R, Sanz-Fraile H, Otero J, Mesquida-Veny F, Gil V, Hervera A, Ferrer I, Soriano J, Trepat X, Farré R, Navajas D, Del Río JA. Involvement of Mechanical Cues in the Migration of Cajal-Retzius Cells in the Marginal Zone During Neocortical Development. Front Cell Dev Biol 2022; 10:886110. [PMID: 35652101 PMCID: PMC9150848 DOI: 10.3389/fcell.2022.886110] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Accepted: 04/25/2022] [Indexed: 12/24/2022] Open
Abstract
Emerging evidence points to coordinated action of chemical and mechanical cues during brain development. At early stages of neocortical development, angiogenic factors and chemokines such as CXCL12, ephrins, and semaphorins assume crucial roles in orchestrating neuronal migration and axon elongation of postmitotic neurons. Here we explore the intrinsic mechanical properties of the developing marginal zone of the pallium in the migratory pathways and brain distribution of the pioneer Cajal-Retzius cells. These neurons are generated in several proliferative regions in the developing brain (e.g., the cortical hem and the pallial subpallial boundary) and migrate tangentially in the preplate/marginal zone covering the upper portion of the developing cortex. These cells play crucial roles in correct neocortical layer formation by secreting several molecules such as Reelin. Our results indicate that the motogenic properties of Cajal-Retzius cells and their perinatal distribution in the marginal zone are modulated by both chemical and mechanical factors, by the specific mechanical properties of Cajal-Retzius cells, and by the differential stiffness of the migratory routes. Indeed, cells originating in the cortical hem display higher migratory capacities than those generated in the pallial subpallial boundary which may be involved in the differential distribution of these cells in the dorsal-lateral axis in the developing marginal zone.
Collapse
Affiliation(s)
- Ana López-Mengual
- Molecular and Cellular Neurobiotechnology, Institute for Bioengineering of Catalonia (IBEC), Barcelona, Spain.,Department of Cell Biology, Physiology and Immunology, Universitat de Barcelona, Barcelona, Spain.,Network Centre of Biomedical Research of Neurodegenerative Diseases (CIBERNED), Institute of Health Carlos III, Madrid, Spain.,Institute of Neuroscience, University of Barcelona, Barcelona, Spain
| | - Miriam Segura-Feliu
- Molecular and Cellular Neurobiotechnology, Institute for Bioengineering of Catalonia (IBEC), Barcelona, Spain.,Department of Cell Biology, Physiology and Immunology, Universitat de Barcelona, Barcelona, Spain.,Network Centre of Biomedical Research of Neurodegenerative Diseases (CIBERNED), Institute of Health Carlos III, Madrid, Spain.,Institute of Neuroscience, University of Barcelona, Barcelona, Spain
| | - Raimon Sunyer
- Unitat de Biofísica I Bioenginyeria, Universitat de Barcelona, Barcelona, Spain
| | - Héctor Sanz-Fraile
- Unitat de Biofísica I Bioenginyeria, Universitat de Barcelona, Barcelona, Spain
| | - Jorge Otero
- Unitat de Biofísica I Bioenginyeria, Universitat de Barcelona, Barcelona, Spain.,Centro de Investigación Biomédica en Red en Enfermedades Respiratorias, Madrid, Spain
| | - Francina Mesquida-Veny
- Molecular and Cellular Neurobiotechnology, Institute for Bioengineering of Catalonia (IBEC), Barcelona, Spain.,Department of Cell Biology, Physiology and Immunology, Universitat de Barcelona, Barcelona, Spain.,Network Centre of Biomedical Research of Neurodegenerative Diseases (CIBERNED), Institute of Health Carlos III, Madrid, Spain.,Institute of Neuroscience, University of Barcelona, Barcelona, Spain
| | - Vanessa Gil
- Molecular and Cellular Neurobiotechnology, Institute for Bioengineering of Catalonia (IBEC), Barcelona, Spain.,Department of Cell Biology, Physiology and Immunology, Universitat de Barcelona, Barcelona, Spain.,Network Centre of Biomedical Research of Neurodegenerative Diseases (CIBERNED), Institute of Health Carlos III, Madrid, Spain.,Institute of Neuroscience, University of Barcelona, Barcelona, Spain
| | - Arnau Hervera
- Molecular and Cellular Neurobiotechnology, Institute for Bioengineering of Catalonia (IBEC), Barcelona, Spain.,Department of Cell Biology, Physiology and Immunology, Universitat de Barcelona, Barcelona, Spain.,Network Centre of Biomedical Research of Neurodegenerative Diseases (CIBERNED), Institute of Health Carlos III, Madrid, Spain.,Institute of Neuroscience, University of Barcelona, Barcelona, Spain
| | - Isidre Ferrer
- Institute of Neuroscience, University of Barcelona, Barcelona, Spain.,Senior Consultant, Bellvitge University Hospital, Hospitalet de Llobregat, Barcelona, Spain.,Department of Pathology and Experimental Therapeutics, University of Barcelona, Barcelona, Spain
| | - Jordi Soriano
- Departament de Física de La Matèria Condensada, Universitat de Barcelona, Barcelona, Spain.,University of Barcelona Institute of Complex Systems (UBICS), Barcelona, Spain
| | - Xavier Trepat
- Unitat de Biofísica I Bioenginyeria, Universitat de Barcelona, Barcelona, Spain.,Integrative Cell and Tissue Dynamics, Institute for Bioengineering of Catalonia (IBEC), Parc Científic de Barcelona, Barcelona, Spain.,Center for Networked Biomedical Research on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Madrid, Spain.,Institució Catalana de Recerca I Estudis Avançats, University of Barcelona, Barcelona, Spain
| | - Ramon Farré
- Unitat de Biofísica I Bioenginyeria, Universitat de Barcelona, Barcelona, Spain.,Centro de Investigación Biomédica en Red en Enfermedades Respiratorias, Madrid, Spain.,Institut D'Investigacions Biomèdiques August Pi Sunyer, Barcelona, Spain
| | - Daniel Navajas
- Unitat de Biofísica I Bioenginyeria, Universitat de Barcelona, Barcelona, Spain.,Centro de Investigación Biomédica en Red en Enfermedades Respiratorias, Madrid, Spain.,Cellular and Respiratory Biomechanics, Institute for Bioengineering of Catalonia (IBEC), Barcelona, Spain
| | - José Antonio Del Río
- Molecular and Cellular Neurobiotechnology, Institute for Bioengineering of Catalonia (IBEC), Barcelona, Spain.,Department of Cell Biology, Physiology and Immunology, Universitat de Barcelona, Barcelona, Spain.,Network Centre of Biomedical Research of Neurodegenerative Diseases (CIBERNED), Institute of Health Carlos III, Madrid, Spain.,Institute of Neuroscience, University of Barcelona, Barcelona, Spain
| |
Collapse
|
26
|
Iyshwarya B, Vajagathali M, Ramakrishnan V. Investigation of Genetic Polymorphism in Autism Spectrum Disorder: a Pathogenesis of the Neurodevelopmental Disorder. ADVANCES IN NEURODEVELOPMENTAL DISORDERS 2022; 6:136-146. [DOI: 10.1007/s41252-022-00251-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 03/12/2022] [Indexed: 12/07/2023]
|
27
|
Anstötz M, Lee SK, Maccaferri G. Glutamate released by Cajal-Retzius cells impacts specific hippocampal circuits and behaviors. Cell Rep 2022; 39:110822. [PMID: 35584670 PMCID: PMC9190441 DOI: 10.1016/j.celrep.2022.110822] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 03/23/2022] [Accepted: 04/23/2022] [Indexed: 12/31/2022] Open
Abstract
The impact of Cajal-Retzius cells on the regulation of hippocampal circuits and related behaviors is unresolved. Here, we directly address this issue by impairing the glutamatergic output of Cajal-Retzius cells with the conditional ablation of vGluT2, which is their main vesicular glutamate transporter. Although two distinct conditional knockout lines do not reveal major alterations in hippocampal-layer organization and dendritic length of principal neurons or GABAergic cells, we find parallel deficits in specific hippocampal-dependent behaviors and in their putative underlying microcircuits. First, conditional knockout animals show increased innate anxiety and decreased feedforward GABAergic inhibition on dentate gyrus granule cells. Second, we observe impaired spatial memory processing, which is associated with decreased spine density and reduced AMPA/NMDA ratio of postsynaptic responses at the perforant- and entorhino-hippocampal pathways. We conclude that glutamate synaptically released by Cajal-Retzius cells is critical for the regulation of hippocampal microcircuits and specific types of behaviors. Anstötz et al. report that postnatal hippocampal Cajal-Retzius cells use vGluT2 as their main glutamate vesicular transporter. Conditional inactivation of vGluT2 in mice reveals both behavioral and network alterations. The observed results indicate the involvement of Cajal-Retzius cells in the regulation of innate anxiety/spatial memory and in potentially related neuronal circuits.
Collapse
Affiliation(s)
- Max Anstötz
- Department of Neuroscience, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA; Institute of Anatomy II, Medical Faculty, Heinrich-Heine-University, Düsseldorf 40225, Germany.
| | - Sun Kyong Lee
- Department of Neuroscience, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Gianmaria Maccaferri
- Department of Neuroscience, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA.
| |
Collapse
|
28
|
Romero-Morales AI, Gama V. Revealing the Impact of Mitochondrial Fitness During Early Neural Development Using Human Brain Organoids. Front Mol Neurosci 2022; 15:840265. [PMID: 35571368 PMCID: PMC9102998 DOI: 10.3389/fnmol.2022.840265] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Accepted: 04/04/2022] [Indexed: 11/13/2022] Open
Abstract
Mitochondrial homeostasis -including function, morphology, and inter-organelle communication- provides guidance to the intrinsic developmental programs of corticogenesis, while also being responsive to environmental and intercellular signals. Two- and three-dimensional platforms have become useful tools to interrogate the capacity of cells to generate neuronal and glia progeny in a background of metabolic dysregulation, but the mechanistic underpinnings underlying the role of mitochondria during human neurogenesis remain unexplored. Here we provide a concise overview of cortical development and the use of pluripotent stem cell models that have contributed to our understanding of mitochondrial and metabolic regulation of early human brain development. We finally discuss the effects of mitochondrial fitness dysregulation seen under stress conditions such as metabolic dysregulation, absence of developmental apoptosis, and hypoxia; and the avenues of research that can be explored with the use of brain organoids.
Collapse
Affiliation(s)
| | - Vivian Gama
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN, United States
- Vanderbilt Center for Stem Cell Biology, Vanderbilt University, Nashville, TN, United States
- Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN, United States
| |
Collapse
|
29
|
Mapelli L, Soda T, D’Angelo E, Prestori F. The Cerebellar Involvement in Autism Spectrum Disorders: From the Social Brain to Mouse Models. Int J Mol Sci 2022; 23:ijms23073894. [PMID: 35409253 PMCID: PMC8998980 DOI: 10.3390/ijms23073894] [Citation(s) in RCA: 50] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Revised: 03/28/2022] [Accepted: 03/29/2022] [Indexed: 02/04/2023] Open
Abstract
Autism spectrum disorders (ASD) are pervasive neurodevelopmental disorders that include a variety of forms and clinical phenotypes. This heterogeneity complicates the clinical and experimental approaches to ASD etiology and pathophysiology. To date, a unifying theory of these diseases is still missing. Nevertheless, the intense work of researchers and clinicians in the last decades has identified some ASD hallmarks and the primary brain areas involved. Not surprisingly, the areas that are part of the so-called “social brain”, and those strictly connected to them, were found to be crucial, such as the prefrontal cortex, amygdala, hippocampus, limbic system, and dopaminergic pathways. With the recent acknowledgment of the cerebellar contribution to cognitive functions and the social brain, its involvement in ASD has become unmistakable, though its extent is still to be elucidated. In most cases, significant advances were made possible by recent technological developments in structural/functional assessment of the human brain and by using mouse models of ASD. Mouse models are an invaluable tool to get insights into the molecular and cellular counterparts of the disease, acting on the specific genetic background generating ASD-like phenotype. Given the multifaceted nature of ASD and related studies, it is often difficult to navigate the literature and limit the huge content to specific questions. This review fulfills the need for an organized, clear, and state-of-the-art perspective on cerebellar involvement in ASD, from its connections to the social brain areas (which are the primary sites of ASD impairments) to the use of monogenic mouse models.
Collapse
Affiliation(s)
- Lisa Mapelli
- Department of Brain and Behavioral Sciences, University of Pavia, 27100 Pavia, Italy; (T.S.); (E.D.)
- Correspondence: (L.M.); (F.P.)
| | - Teresa Soda
- Department of Brain and Behavioral Sciences, University of Pavia, 27100 Pavia, Italy; (T.S.); (E.D.)
| | - Egidio D’Angelo
- Department of Brain and Behavioral Sciences, University of Pavia, 27100 Pavia, Italy; (T.S.); (E.D.)
- Brain Connectivity Center, IRCCS Mondino Foundation, 27100 Pavia, Italy
| | - Francesca Prestori
- Department of Brain and Behavioral Sciences, University of Pavia, 27100 Pavia, Italy; (T.S.); (E.D.)
- Correspondence: (L.M.); (F.P.)
| |
Collapse
|
30
|
Guo J, Riley KW, Durham T, Margolis AE, Wang S, Perera F, Herbstman JB. Association Studies of Environmental Exposures, DNA Methylation and Children’s Cognitive, Behavioral, and Mental Health Problems. Front Genet 2022; 13:871820. [PMID: 35528545 PMCID: PMC9074894 DOI: 10.3389/fgene.2022.871820] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Accepted: 03/14/2022] [Indexed: 11/21/2022] Open
Abstract
Introduction: Prenatal environmental exposures have been associated with children’s cognitive, behavioral, and mental health problems, and alterations in DNA methylation have been hypothesized as an underlying biological mechanism. However, when testing this hypothesis, it is often difficult to overcome the problem of multiple comparisons in statistical testing when evaluating a large number of developmental outcomes and DNA methylation sites as potential mediators. The objective of this study is to implement a ‘meet-in-the-middle’ approach with a sequential roadmap to address this concern. Methods: In the Columbia Center for Children’s Environmental Health birth cohort study, we implemented a 5-step sequential process for identifying CpG sites that mediate associations between prenatal environmental exposures and cognitive, behavioral, and mental health problems as measured by the Wechsler Intelligence Scale for Children-Fourth Edition (WISC-IV) and the Child Behavior Checklist (CBCL). These steps include 1) the identification of biological pathways that are relevant to each outcome of interest; 2) selection of a set of genes and CpGs on genes that are significantly associated with the outcomes; 3) identification of exposures that are significantly associated with selected CpGs; 4) examination of exposure-outcome relationships among those where significant CpGs were identified; and 5) mediation analysis of the selected exposures and corresponding outcomes. In this study, we considered a spectrum of environmental exposure classes including environmental phenols, pesticides, phthalates, flame retardants and air pollutants. Results: Among all considered exposures and outcomes, we found one CpG site (cg27510182) on gene (DAB1) that potentially mediates the effect of exposure to PAH on CBCL social problems at children aged 7. Conclusion: This ‘meet-in-the-middle’ approach attenuates concerns regarding multiple comparisons by focusing on genes and pathways that are biologically relevant for the hypothesis.
Collapse
Affiliation(s)
- Jia Guo
- Columbia Center for Children’s Environmental Health, Mailman School of Public Health, Columbia University, New York, NY, United States
- Department of Biostatistics, Mailman School of Public Health, Columbia University, New York, NY, United States
| | - Kylie W. Riley
- Columbia Center for Children’s Environmental Health, Mailman School of Public Health, Columbia University, New York, NY, United States
- Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University, New York, NY, United States
| | - Teresa Durham
- Columbia Center for Children’s Environmental Health, Mailman School of Public Health, Columbia University, New York, NY, United States
- Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University, New York, NY, United States
| | - Amy E. Margolis
- Columbia Center for Children’s Environmental Health, Mailman School of Public Health, Columbia University, New York, NY, United States
- Division of Child and Adolescent Psychiatry, Columbia University Irving Medical Center, New York, NY, United States
| | - Shuang Wang
- Columbia Center for Children’s Environmental Health, Mailman School of Public Health, Columbia University, New York, NY, United States
- Department of Biostatistics, Mailman School of Public Health, Columbia University, New York, NY, United States
| | - Frederica Perera
- Columbia Center for Children’s Environmental Health, Mailman School of Public Health, Columbia University, New York, NY, United States
- Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University, New York, NY, United States
| | - Julie B. Herbstman
- Columbia Center for Children’s Environmental Health, Mailman School of Public Health, Columbia University, New York, NY, United States
- Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University, New York, NY, United States
- *Correspondence: Julie B. Herbstman,
| |
Collapse
|
31
|
Long KR, Huttner WB. The Role of the Extracellular Matrix in Neural Progenitor Cell Proliferation and Cortical Folding During Human Neocortex Development. Front Cell Neurosci 2022; 15:804649. [PMID: 35140590 PMCID: PMC8818730 DOI: 10.3389/fncel.2021.804649] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Accepted: 12/20/2021] [Indexed: 12/12/2022] Open
Abstract
Extracellular matrix (ECM) has long been known to regulate many aspects of neural development in many different species. However, the role of the ECM in the development of the human neocortex is not yet fully understood. In this review we discuss the role of the ECM in human neocortex development and the different model systems that can be used to investigate this. In particular, we will focus on how the ECM regulates human neural stem and progenitor cell proliferation and differentiation, how the ECM regulates the architecture of the developing human neocortex and the effect of mutations in ECM and ECM-associated genes in neurodevelopmental disorders.
Collapse
Affiliation(s)
- Katherine R. Long
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, United Kingdom
- MRC Centre for Neurodevelopmental Disorders, King’s College London, London, United Kingdom
| | - Wieland B. Huttner
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| |
Collapse
|
32
|
Transcription factor 4 controls positioning of cortical projection neurons through regulation of cell adhesion. Mol Psychiatry 2021; 26:6562-6577. [PMID: 33963287 DOI: 10.1038/s41380-021-01119-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Revised: 04/02/2021] [Accepted: 04/13/2021] [Indexed: 02/07/2023]
Abstract
The establishment of neural circuits depends on precise neuronal positioning in the cortex, which occurs via a tightly coordinated process of neuronal differentiation, migration, and terminal localization. Deficits in this process have been implicated in several psychiatric disorders. Here, we show that the transcription factor Tcf4 controls neuronal positioning during brain development. Tcf4-deficient neurons become mispositioned in clusters when their migration to the cortical plate is complete. We reveal that Tcf4 regulates the expression of cell adhesion molecules to control neuronal positioning. Furthermore, through in vivo extracellular electrophysiology, we show that neuronal functions are disrupted after the loss of Tcf4. TCF4 mutations are strongly associated with schizophrenia and cause Pitt-Hopkins syndrome, which is characterized by severe intellectual disability. Thus, our results not only reveal the importance of neuronal positioning in brain development but also provide new insights into the potential mechanisms underlying neurological defects linked to TCF4 mutations.
Collapse
|
33
|
Abstract
The human brain is characterized by the large size and intricate folding of its cerebral cortex, which are fundamental for our higher cognitive function and frequently altered in pathological dysfunction. Cortex folding is not unique to humans, nor even to primates, but is common across mammals. Cortical growth and folding are the result of complex developmental processes that involve neural stem and progenitor cells and their cellular lineages, the migration and differentiation of neurons, and the genetic programs that regulate and fine-tune these processes. All these factors combined generate mechanical stress and strain on the developing neural tissue, which ultimately drives orderly cortical deformation and folding. In this review we examine and summarize the current knowledge on the molecular, cellular, histogenic and mechanical mechanisms that are involved in and influence folding of the cerebral cortex, and how they emerged and changed during mammalian evolution. We discuss the main types of pathological malformations of human cortex folding, their specific developmental origin, and how investigating their genetic causes has illuminated our understanding of key events involved. We close our review by presenting the state-of-the-art animal and in vitro models of cortex folding that are currently used to study these devastating developmental brain disorders in children, and what are the main challenges that remain ahead of us to fully understand brain folding.
Collapse
Affiliation(s)
- Lucia Del Valle Anton
- Instituto de Neurociencias, Agencia Estatal Consejo Superior de Investigaciones Científicas, San Juan de Alicante, Alicante, Spain
| | - Victor Borrell
- Instituto de Neurociencias, Agencia Estatal Consejo Superior de Investigaciones Científicas, San Juan de Alicante, Alicante, Spain
| |
Collapse
|
34
|
Orcinha C, Kilias A, Paschen E, Follo M, Haas CA. Reelin Is Required for Maintenance of Granule Cell Lamination in the Healthy and Epileptic Hippocampus. Front Mol Neurosci 2021; 14:730811. [PMID: 34483838 PMCID: PMC8414139 DOI: 10.3389/fnmol.2021.730811] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Accepted: 07/26/2021] [Indexed: 11/21/2022] Open
Abstract
One characteristic feature of mesial temporal lobe epilepsy is granule cell dispersion (GCD), a pathological widening of the granule cell layer in the dentate gyrus. The loss of the extracellular matrix protein Reelin, an important positional cue for neurons, correlates with GCD formation in MTLE patients and in rodent epilepsy models. Here, we used organotypic hippocampal slice cultures (OHSC) from transgenic mice expressing enhanced green fluorescent protein (eGFP) in differentiated granule cells (GCs) to monitor GCD formation dynamically by live cell video microscopy and to investigate the role of Reelin in this process. We present evidence that following treatment with the glutamate receptor agonist kainate (KA), eGFP-positive GCs migrated mainly toward the hilar region. In the hilus, Reelin-producing neurons were rapidly lost following KA treatment as shown in a detailed time series. Addition of recombinant Reelin fragments to the medium effectively prevented the KA-triggered movement of eGFP-positive GCs. Placement of Reelin-coated beads into the hilus of KA-treated cultures stopped the migration of GCs in a distance-dependent manner. In addition, quantitative Western blot analysis revealed that KA treatment affects the Reelin signal transduction pathway by increasing intracellular adaptor protein Disabled-1 synthesis and reducing the phosphorylation of cofilin, a downstream target of the Reelin pathway. Both events were normalized by addition of recombinant Reelin fragments. Finally, following neutralization of Reelin in healthy OHSC by incubation with the function-blocking CR-50 Reelin antibody, GCs started to migrate without any direction preference. Together, our findings demonstrate that normotopic position of Reelin is essential for the maintenance of GC lamination in the dentate gyrus and that GCD is the result of a local Reelin deficiency.
Collapse
Affiliation(s)
- Catarina Orcinha
- Experimental Epilepsy Research, Department of Neurosurgery, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg im Breisgau, Germany
| | - Antje Kilias
- Biomicrotechnology, Department of Microsystems Engineering (IMTEK), University of Freiburg, Freiburg im Breisgau, Germany
| | - Enya Paschen
- Experimental Epilepsy Research, Department of Neurosurgery, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg im Breisgau, Germany.,Faculty of Biology, University of Freiburg, Freiburg im Breisgau, Germany
| | - Marie Follo
- Lighthouse Core Facility, Department of Internal Medicine I, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg im Breisgau, Germany
| | - Carola A Haas
- Experimental Epilepsy Research, Department of Neurosurgery, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg im Breisgau, Germany.,Center for Basics in NeuroModulation, Faculty of Medicine, University of Freiburg, Freiburg im Breisgau, Germany
| |
Collapse
|
35
|
Ahmed S, Rakib A, Uddin MMN, Islam MS, Ullah SA, Emran TB. Association of reelin gene (RELN) polymorphism with autism spectrum disorder in the Bangladeshi population. Meta Gene 2021. [DOI: 10.1016/j.mgene.2021.100901] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
|
36
|
Reelin Alleviates Mesenchymal Stem Cell Senescence and Reduces Pathological α-Synuclein Expression in an In Vitro Model of Parkinson's Disease. Genes (Basel) 2021; 12:genes12071066. [PMID: 34356083 PMCID: PMC8308051 DOI: 10.3390/genes12071066] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 07/06/2021] [Accepted: 07/09/2021] [Indexed: 02/08/2023] Open
Abstract
Parkinson's disease (PD) is one of the most common neurodegenerative diseases. The mechanisms underlying PD remain to be fully elucidated, and research into treatments for this condition is ongoing. Recent advances in genetic research have shed light on the mechanisms underlying PD. In this study, we used PD and control mesenchymal stem cells (MSCs) obtained from adipose tissues to confirm the differences between groups at the cellular and molecular levels. The results revealed that in PD MSCs, cell viability was clearly lower, and the rate of cell senescence was higher compared to the controls. Next, to compare the gene expression in PD and control cells, transcriptome analysis was performed. Genes in pathways, including extracellular matrix (ECM) receptor interaction, P53 signaling, and focal adhesion, were down-regulated in PD. Among genes related to ECM receptor interaction, RELN gene expression was markedly decreased in PD cells; however, after being treated with recombinant Reelin protein, a significant increase in cell viability and a decrease in α-Synuclein aggregation and cell senescence were observed. In conclusion, Reelin affects PD by positively influencing the cell characteristics. Our findings will facilitate research into new treatments for PD.
Collapse
|
37
|
Liu Y, Wang Y, Yuan W, Dong F, Zhen F, Liu J, Yang L, Qu X, Yao R. Reelin promotes oligodendrocyte precursors migration via the Wnt/β-catenin signaling pathway. Neurol Res 2021; 43:543-552. [PMID: 33616025 DOI: 10.1080/01616412.2021.1888604] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Accepted: 02/07/2021] [Indexed: 10/22/2022]
Abstract
Objectives: The extracellular matrix glycoprotein Reelin plays an important role in the development of the central nervous system and is involved in neurogenesis, neuronal polarization and migration. Although it has been reported that Reelin and its receptor are expressed in oligodendrocyte precursors (OPCs), the main functions and possible mechanism of Reelin in OPCs remain unclear.Methods: In this study, immunofluorescence staining was used to detect the expressions of A2B5, PDGFRα, Reelin, VLDLR and Dab1 in OPCs. The expression of p-Dab1 in OPCs which was treated with Reelin at different concentrations was assayed by western blot. Effects of Reelin on the proliferation of OPCs was measured by EdU and CCK-8. Annexin V-FITC/PI assayed the effect of Reelin on the apoptosis of OPCs. Effects of Reelin on the migration ability of OPCs were detected by the scratch test and transwell experiments. Immunoblotting was used to measure the activation of Wnt/β-catenin signaling with Reelin, while transwell experiments were performed to verify the migration of OPCs under the activation of Wnt/β-catenin signaling.Results: Results showed that the receptor of Reelin, very-low-density lipoprotein receptor (VLDLR), and its adaptor protein, Dab1, are highly expressed in A2B5/PDGFRα double-positive OPCs. Recombinant Reelin protein promoted OPCs migration in vitro but had no obvious effects on proliferation or apoptosis. Reelin also promoted the phosphorylation of Dab1 and increased the expression of β-catenin in OPCs. WIKI4, an inhibitor of Wnt/β-catenin signaling, suppressed the migration of OPCs induced by Reelin.Conclusion: The present study indicated that Reelin promotes OPCs migration via the Wnt/β-catenin pathway.
Collapse
Affiliation(s)
- Yaping Liu
- Laboratory of National Experimental Teaching and Demonstration Center of Basic Medicine, Xuzhou Medical University, Xuzhou, Jiangsu, PRC
| | - Yuanyuan Wang
- Pediatrics, Nanjing Tongren Hospital, Nanjing, Jiangsu, PRC
| | - Wen Yuan
- Department of Cell Biology and Neurobiology, Xuzhou Key Laboratory of Neurobiology, Xuzhou Medical University, Xuzhou, Jiangsu, PRC
| | - Fuxing Dong
- Department of Cell Biology and Neurobiology, Xuzhou Key Laboratory of Neurobiology, Xuzhou Medical University, Xuzhou, Jiangsu, PRC
| | - Fei Zhen
- Department of Pathology, Hongze District People's Hospital, Huai 'an, Jiangsu, PRC
| | - Jing Liu
- Department of Cell Biology and Neurobiology, Xuzhou Key Laboratory of Neurobiology, Xuzhou Medical University, Xuzhou, Jiangsu, PRC
| | - Lihua Yang
- Department of Cell Biology and Neurobiology, Xuzhou Key Laboratory of Neurobiology, Xuzhou Medical University, Xuzhou, Jiangsu, PRC
| | - Xuebin Qu
- Department of Cell Biology and Neurobiology, Xuzhou Key Laboratory of Neurobiology, Xuzhou Medical University, Xuzhou, Jiangsu, PRC
| | - Ruiqin Yao
- Department of Cell Biology and Neurobiology, Xuzhou Key Laboratory of Neurobiology, Xuzhou Medical University, Xuzhou, Jiangsu, PRC
| |
Collapse
|
38
|
Pingale TD, Gupta GL. Novel therapeutic approaches for Parkinson's disease by targeting brain cholesterol homeostasis. J Pharm Pharmacol 2021; 73:862-873. [PMID: 33822122 DOI: 10.1093/jpp/rgaa063] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Accepted: 12/17/2020] [Indexed: 01/25/2023]
Abstract
OBJECTIVES Human brain is composed of 25% of the cholesterol & any dysfunction in brain cholesterol homeostasis contributes to neurodegenerative disorders such as Parkinson, Alzheimer's, Huntington's disease, etc. A growing literature indicates that alteration in neurotransmission & brain cholesterol metabolism takes place in the early stage of the disease. The current paper summarizes the role of cholesterol & its homeostasis in the pathophysiology of Parkinson's disease. KEY FINDINGS Literature findings suggest the possible role of lipids such as oxysterols, lipoproteins, etc. in Parkinson's disease pathophysiology. Cholesterol performs a diverse role in the brain but any deviation in its levels leads to neurodegeneration. Dysregulation of lipid caused by oxidative stress & inflammation leads to α-synuclein trafficking which contributes to Parkinson's disease progression. Also, α-synuclein by binding to membrane lipid forms lipid-protein complex & results in its aggregation. Different targets such as Phospholipase A2, Stearoyl-CoA desaturase enzyme, proprotein convertase subtilisin/kexin type 9, etc. have been identified as a potential novel approach for Parkinson's disease treatment. SUMMARY In the current review, we have discussed the possible molecular role of cholesterol homeostasis in Parkinson's disease progression. We also identified potential therapeutic targets that need to be evaluated clinically for the development of Parkinson's treatment.
Collapse
Affiliation(s)
- Tanvi Dayanand Pingale
- Department of Pharmacology, School of Pharmacy and Technology Management, SVKM'S NMIMS, Shirpur, Maharashtra, India
| | - Girdhari Lal Gupta
- Department of Pharmacology, School of Pharmacy and Technology Management, SVKM'S NMIMS, Shirpur, Maharashtra, India
| |
Collapse
|
39
|
Camblor-Perujo S, Kononenko NL. Brain-specific functions of the endocytic machinery. FEBS J 2021; 289:2219-2246. [PMID: 33896112 DOI: 10.1111/febs.15897] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Revised: 03/29/2021] [Indexed: 12/12/2022]
Abstract
Endocytosis is an essential cellular process required for multiple physiological functions, including communication with the extracellular environment, nutrient uptake, and signaling by the cell surface receptors. In a broad sense, endocytosis is accomplished through either constitutive or ligand-induced invagination of the plasma membrane, which results in the formation of the plasma membrane-retrieved endocytic vesicles, which can either be sent for degradation to the lysosomes or recycled back to the PM. This additional function of endocytosis in membrane retrieval has been adopted by excitable cells, such as neurons, for membrane equilibrium maintenance at synapses. The last two decades were especially productive with respect to the identification of brain-specific functions of the endocytic machinery, which additionally include but not limited to regulation of neuronal differentiation and migration, maintenance of neuron morphology and synaptic plasticity, and prevention of neurotoxic aggregates spreading. In this review, we highlight the current knowledge of brain-specific functions of endocytic machinery with a specific focus on three brain cell types, neuronal progenitor cells, neurons, and glial cells.
Collapse
Affiliation(s)
| | - Natalia L Kononenko
- CECAD Cluster of Excellence, University of Cologne, Germany.,Center for Physiology & Pathophysiology, Medical Faculty, University of Cologne, Germany
| |
Collapse
|
40
|
Dong X, Yang L, Liu K, Ji X, Tang C, Li W, Ma L, Mei Y, Peng T, Feng B, Wu Z, Tang Q, Gao Y, Yan K, Zhou W, Xiong M. Transcriptional networks identify synaptotagmin-like 3 as a regulator of cortical neuronal migration during early neurodevelopment. Cell Rep 2021; 34:108802. [PMID: 33657377 DOI: 10.1016/j.celrep.2021.108802] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2020] [Revised: 12/25/2020] [Accepted: 02/08/2021] [Indexed: 02/07/2023] Open
Abstract
Human brain development is a complex process involving neural proliferation, differentiation, and migration that are directed by many essential cellular factors and drivers. Here, using the NetBID2 algorithm and developing human brain RNA sequencing dataset, we identify synaptotagmin-like 3 (SYTL3) as one of the top drivers of early human brain development. Interestingly, SYTL3 exhibits high activity but low expression in both early developmental human cortex and human embryonic stem cell (hESC)-derived neurons. Knockout of SYTL3 (SYTL3-KO) in human neurons or knockdown of Sytl3 in embryonic mouse cortex markedly promotes neuronal migration. SYTL3-KO causes an abnormal distribution of deep-layer neurons in brain organoids and reduces presynaptic neurotransmitter release in hESC-derived neurons. We further demonstrate that SYTL3-KO-accelerated neuronal migration is modulated by high expression of matrix metalloproteinases. Together, based on bioinformatics and biological experiments, we identify SYTL3 as a regulator of cortical neuronal migration in human and mouse developing brains.
Collapse
Affiliation(s)
- Xinran Dong
- Molecular Medical Center, Children's Hospital of Fudan University, 399 Wanyuan Road, Shanghai 201102, China
| | - Lin Yang
- Molecular Medical Center, Children's Hospital of Fudan University, 399 Wanyuan Road, Shanghai 201102, China
| | - Kaiyi Liu
- Molecular Medical Center, Children's Hospital of Fudan University, 399 Wanyuan Road, Shanghai 201102, China
| | - Xiaoli Ji
- Molecular Medical Center, Children's Hospital of Fudan University, 399 Wanyuan Road, Shanghai 201102, China; Stem Cell Center, Children's Hospital of Fudan University, Shanghai 201102, China
| | - Chuanqing Tang
- Stem Cell Center, Children's Hospital of Fudan University, Shanghai 201102, China
| | - Wanxing Li
- Molecular Medical Center, Children's Hospital of Fudan University, 399 Wanyuan Road, Shanghai 201102, China
| | - Ling Ma
- Stem Cell Center, Children's Hospital of Fudan University, Shanghai 201102, China
| | - Yuting Mei
- Stem Cell Center, Children's Hospital of Fudan University, Shanghai 201102, China
| | - Ting Peng
- Key Laboratory of Neonatal Diseases, Ministry of Health, Children's Hospital of Fudan University, Shanghai 201102, China
| | - Ban Feng
- Institute of Neuroscience, Chinese Academy of Sciences, Shanghai 200031, Shanghai, China
| | - Ziyan Wu
- Institute of Neuroscience, Chinese Academy of Sciences, Shanghai 200031, Shanghai, China
| | - Qingyuan Tang
- Stem Cell Center, Children's Hospital of Fudan University, Shanghai 201102, China
| | - Yanyan Gao
- Ultrasonography Department, Children's Hospital of Fudan University, Shanghai 201102, China
| | - Kai Yan
- Key Laboratory of Neonatal Diseases, Ministry of Health, Children's Hospital of Fudan University, Shanghai 201102, China
| | - Wenhao Zhou
- Molecular Medical Center, Children's Hospital of Fudan University, 399 Wanyuan Road, Shanghai 201102, China; Key Laboratory of Neonatal Diseases, Ministry of Health, Children's Hospital of Fudan University, Shanghai 201102, China.
| | - Man Xiong
- Stem Cell Center, Children's Hospital of Fudan University, Shanghai 201102, China.
| |
Collapse
|
41
|
Weninger J, Meseke M, Rana S, Förster E. Heat-Shock Induces Granule Cell Dispersion and Microgliosis in Hippocampal Slice Cultures. Front Cell Dev Biol 2021; 9:626704. [PMID: 33693000 PMCID: PMC7937632 DOI: 10.3389/fcell.2021.626704] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Accepted: 02/03/2021] [Indexed: 11/13/2022] Open
Abstract
Granule cell dispersion (GCD) has been found in the dentate gyrus (dg) of patients with temporal lobe epilepsy (TLE) and a history of febrile seizures but was also recently observed in pediatric patients that did not suffer from epilepsy. This indicates that GCD might not always be disease related, but instead could reflect normal morphological variation. Thus, distribution of newborn granule cells within the hilar region is part of normal dg development at early stages but could be misinterpreted as pathological GCD. In turn, pathological GCD may be caused, for example, by genetic mutations, such as the reeler mutation. GCD in the reeler mutant goes along with an increased susceptibility to epileptiform activity. Pathological GCD in combination with epilepsy is caused by experimental administration of the glutamate receptor agonist kainic acid in rodents. In consequence, the interpretation of GCD and the role of febrile seizures remain controversial. Here, we asked whether febrile temperatures alone might be sufficient to trigger GCD and used hippocampal slice cultures as in vitro model to analyze the effect of a transient temperature increase on the dg morphology. We found that a heat-shock of 41°C for 6 h was sufficient to induce GCD and degeneration of a fraction of granule cells. Both of these factors, broadening of the granule cell layer (gcl) and increased neuronal cell death within the gcl, contributed to the development of a significantly reduced packaging density of granule cells. In contrast, Reelin expressing Cajal–Retzius (CR) cells in the molecular layer were heat-shock resistant. Thus, their number was not reduced, and we did not detect degenerating CR cells after heat-shock, implying that GCD was not caused by the loss of CR cells. Importantly, the heat-shock-induced deterioration of dg morphology was accompanied by a massive microgliosis, reflecting a robust heat-shock-induced immune response. In contrast, in the study that reported on GCD as a non-specific finding in pediatric patients, no microglia reaction was observed. Thus, our findings underpin the importance of microglia as a marker to distinguish pathological GCD from normal morphological variation.
Collapse
Affiliation(s)
- Jasmin Weninger
- Institute of Anatomy, Department of Neuroanatomy and Molecular Brain Research, Ruhr-Universität Bochum, Bochum, Germany
| | - Maurice Meseke
- Institute of Anatomy, Department of Neuroanatomy and Molecular Brain Research, Ruhr-Universität Bochum, Bochum, Germany
| | - Shaleen Rana
- Institute of Anatomy, Department of Neuroanatomy and Molecular Brain Research, Ruhr-Universität Bochum, Bochum, Germany
| | - Eckart Förster
- Institute of Anatomy, Department of Neuroanatomy and Molecular Brain Research, Ruhr-Universität Bochum, Bochum, Germany
| |
Collapse
|
42
|
de Agustín-Durán D, Mateos-White I, Fabra-Beser J, Gil-Sanz C. Stick around: Cell-Cell Adhesion Molecules during Neocortical Development. Cells 2021; 10:118. [PMID: 33435191 PMCID: PMC7826847 DOI: 10.3390/cells10010118] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Revised: 12/29/2020] [Accepted: 01/07/2021] [Indexed: 12/21/2022] Open
Abstract
The neocortex is an exquisitely organized structure achieved through complex cellular processes from the generation of neural cells to their integration into cortical circuits after complex migration processes. During this long journey, neural cells need to establish and release adhesive interactions through cell surface receptors known as cell adhesion molecules (CAMs). Several types of CAMs have been described regulating different aspects of neurodevelopment. Whereas some of them mediate interactions with the extracellular matrix, others allow contact with additional cells. In this review, we will focus on the role of two important families of cell-cell adhesion molecules (C-CAMs), classical cadherins and nectins, as well as in their effectors, in the control of fundamental processes related with corticogenesis, with special attention in the cooperative actions among the two families of C-CAMs.
Collapse
Affiliation(s)
| | | | | | - Cristina Gil-Sanz
- Neural Development Laboratory, Instituto Universitario de Biomedicina y Biotecnología (BIOTECMED) and Departamento de Biología Celular, Facultat de Biología, Universidad de Valencia, 46100 Burjassot, Spain; (D.d.A.-D.); (I.M.-W.); (J.F.-B.)
| |
Collapse
|
43
|
Amin S, Borrell V. The Extracellular Matrix in the Evolution of Cortical Development and Folding. Front Cell Dev Biol 2020; 8:604448. [PMID: 33344456 PMCID: PMC7744631 DOI: 10.3389/fcell.2020.604448] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Accepted: 11/12/2020] [Indexed: 02/02/2023] Open
Abstract
The evolution of the mammalian cerebral cortex leading to humans involved a remarkable sophistication of developmental mechanisms. Specific adaptations of progenitor cell proliferation and neuronal migration mechanisms have been proposed to play major roles in this evolution of neocortical development. One of the central elements influencing neocortex development is the extracellular matrix (ECM). The ECM provides both a structural framework during tissue formation and to present signaling molecules to cells, which directly influences cell behavior and movement. Here we review recent advances in the understanding of the role of ECM molecules on progenitor cell proliferation and neuronal migration, and how these contribute to cerebral cortex expansion and folding. We discuss how transcriptomic studies in human, ferret and mouse identify components of ECM as being candidate key players in cortex expansion during development and evolution. Then we focus on recent functional studies showing that ECM components regulate cortical progenitor cell proliferation, neuron migration and the mechanical properties of the developing cortex. Finally, we discuss how these features differ between lissencephalic and gyrencephalic species, and how the molecular evolution of ECM components and their expression profiles may have been fundamental in the emergence and evolution of cortex folding across mammalian phylogeny.
Collapse
Affiliation(s)
| | - Víctor Borrell
- Instituto de Neurociencias, Consejo Superior de Investigaciones Científicas and Universidad Miguel Hernández, Sant Joan d’Alacant, Spain
| |
Collapse
|
44
|
Philippot G, Hellsten SV, Viberg H, Fredriksson R. Evaluation of the dentate gyrus in adult mice exposed to acetaminophen (paracetamol) on postnatal day 10. Int J Dev Neurosci 2020; 81:91-97. [PMID: 33222217 DOI: 10.1002/jdn.10079] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Revised: 11/16/2020] [Accepted: 11/16/2020] [Indexed: 11/09/2022] Open
Abstract
Acetaminophen (AAP; or paracetamol) is a widely used nonprescription drug with antipyretic and analgesic properties. Alarmingly, there is an increasing body of evidence showing that developmental exposure to AAP is associated with adverse behavioural outcomes later in life. We have previously shown that relevant doses of AAP in 10-day-old mice affected memory, learning and locomotor activity in the adult animals. Interestingly, the neurons of the dentate gyrus (DG) have a relatively late time of origin as they are generated during the first two weeks of postnatal life in rodents. Since the generation of these cells, which are important for memory processing, coincides with our AAP exposure, we aim to investigate if the cytoarchitecture of the DG is affected by postnatal day 10 AAP treatment. In addition, we investigate if markers for differentiation and migration in the hippocampus were affected by the same treatment. We did not observe any visual effects in adult DG cytoarchitecture, nor any changes of markers for differentiation/migration in the hippocampus in 24 hr after exposure. Even though a large effect size was estimated on adult DG thickness following AAP exposure, the estimated 95% CIs around the differences of the means reveal no significant effect. Hence, larger sample sizes are warranted to clarify if neonatal AAP exposure affects adult DG thickness in mice.
Collapse
Affiliation(s)
- Gaëtan Philippot
- Department of Pharmaceutical Biosciences, Molecular Neuropharmacology, Biomedical Center, Uppsala University, Uppsala, Sweden
| | - Sofie V Hellsten
- Department of Pharmaceutical Biosciences, Molecular Neuropharmacology, Biomedical Center, Uppsala University, Uppsala, Sweden
| | - Henrik Viberg
- Department of Organismal Biology, Environmental Toxicology, Evolutionary Biology Centre, Uppsala University, Uppsala, Sweden
| | - Robert Fredriksson
- Department of Pharmaceutical Biosciences, Molecular Neuropharmacology, Biomedical Center, Uppsala University, Uppsala, Sweden
| |
Collapse
|
45
|
Faini G, Del Bene F, Albadri S. Reelin functions beyond neuronal migration: from synaptogenesis to network activity modulation. Curr Opin Neurobiol 2020; 66:135-143. [PMID: 33197872 DOI: 10.1016/j.conb.2020.10.009] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Revised: 10/08/2020] [Accepted: 10/11/2020] [Indexed: 01/05/2023]
Abstract
Reelin, a glycoprotein of the extracellular matrix, has been the focus of several studies over the years, mostly for its role in cell migration. Here we report the role of this molecule and of its downstream pathways in post-mitotic neurons and how they contribute to neural circuit assembly, refinement and function. Accumulating evidence has pointed at a major role for Reelin in axonal guidance, synaptogenesis and dendritic spine formation. In particular, new evidence points at a direct role in axonal targeting and refinement at the target site. In addition, recent advances highlight new functions of Reelin in the modulation of synaptic activity, plasticity and behavior and in the direct regulation of GABA receptors expression and stability. We discuss these findings in the context of neurodevelopmental disorders.
Collapse
Affiliation(s)
- Giulia Faini
- Institut de la Vision, Sorbonne Université, INSERM, CNRS, Paris, France
| | - Filippo Del Bene
- Institut de la Vision, Sorbonne Université, INSERM, CNRS, Paris, France.
| | - Shahad Albadri
- Institut de la Vision, Sorbonne Université, INSERM, CNRS, Paris, France
| |
Collapse
|
46
|
Nawa Y, Kimura H, Mori D, Kato H, Toyama M, Furuta S, Yu Y, Ishizuka K, Kushima I, Aleksic B, Arioka Y, Morikawa M, Okada T, Inada T, Kaibuchi K, Ikeda M, Iwata N, Suzuki M, Okahisa Y, Egawa J, Someya T, Nishimura F, Sasaki T, Ozaki N. Rare single-nucleotide DAB1 variants and their contribution to Schizophrenia and autism spectrum disorder susceptibility. Hum Genome Var 2020; 7:37. [PMID: 33298905 PMCID: PMC7655853 DOI: 10.1038/s41439-020-00125-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 09/24/2020] [Accepted: 09/27/2020] [Indexed: 01/09/2023] Open
Abstract
Disabled 1 (DAB1) is an intracellular adaptor protein in the Reelin signaling pathway and plays an essential role in correct neuronal migration and layer formation in the developing brain. DAB1 has been repeatedly reported to be associated with neurodevelopmental disorders including schizophrenia (SCZ) and autism spectrum disorders (ASD) in genetic, animal, and postmortem studies. Recently, increasing attention has been given to rare single-nucleotide variants (SNVs) found by deep sequencing of candidate genes. In this study, we performed exon-targeted resequencing of DAB1 in 370 SCZ and 192 ASD patients using next-generation sequencing technology to identify rare SNVs with a minor allele frequency <1%. We detected two rare missense mutations (G382C, V129I) and then performed a genetic association study in a sample comprising 1763 SCZ, 380 ASD, and 2190 healthy control subjects. Although no statistically significant association with the detected mutations was observed for either SCZ or ASD, G382C was found only in the case group, and in silico analyses and in vitro functional assays suggested that G382C alters the function of the DAB1 protein. The rare variants of DAB1 found in the present study should be studied further to elucidate their potential functional relevance to the pathophysiology of SCZ and ASD.
Collapse
Affiliation(s)
- Yoshihiro Nawa
- Department of Psychiatry, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan
| | - Hiroki Kimura
- Department of Psychiatry, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan.
| | - Daisuke Mori
- Department of Psychiatry, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan
- Brain and Mind Research Center, Nagoya University, Nagoya, Aichi, Japan
| | - Hidekazu Kato
- Department of Psychiatry, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan
| | - Miho Toyama
- Department of Psychiatry, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan
| | - Sho Furuta
- Department of Psychiatry, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan
| | - Yanjie Yu
- Department of Psychiatry, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan
| | - Kanako Ishizuka
- Department of Psychiatry, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan
| | - Itaru Kushima
- Department of Psychiatry, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan
- Institute for Advanced Research, Nagoya University, Nagoya, Aichi, Japan
| | - Branko Aleksic
- Department of Psychiatry, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan
| | - Yuko Arioka
- Department of Psychiatry, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan
- Institute for Advanced Research, Nagoya University, Nagoya, Aichi, Japan
- Center for Advanced Medicine and Clinical Research, Nagoya University Hospital, Nagoya, Aichi, Japan
| | - Mako Morikawa
- Department of Psychiatry, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan
| | - Takashi Okada
- Department of Psychiatry, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan
| | - Toshiya Inada
- Department of Psychiatry, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan
| | - Kozo Kaibuchi
- Department of Cell Pharmacology, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan
| | - Masashi Ikeda
- Department of Psychiatry, Fujita Health University School of Medicine, Toyoake, Aichi, Japan
| | - Nakao Iwata
- Department of Psychiatry, Fujita Health University School of Medicine, Toyoake, Aichi, Japan
| | - Michio Suzuki
- Department of Neuropsychiatry, University of Toyama Graduate School of Medicine and Pharmaceutical Sciences, Toyama, Japan
| | - Yuko Okahisa
- Department of Neuropsychiatry, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Jun Egawa
- Department of Psychiatry, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Toshiyuki Someya
- Department of Psychiatry, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Fumichika Nishimura
- Office for Mental Health Support, Center for Research on Counseling and Support Services, The University of Tokyo, Tokyo, Japan
| | - Tsukasa Sasaki
- Department of Physical and Health Education, Graduate School of Education, The University of Tokyo, Tokyo, Japan
| | - Norio Ozaki
- Department of Psychiatry, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan
| |
Collapse
|
47
|
Demir R, Şahar U, Deveci R. Determination of terminal glycan and total monosaccharide profiles of reelin glycoprotein in SH-SY5Y neuroblastoma cell line by lectin blotting and capillary liquid chromatography electrospray ionization-ion trap tandem mass spectrometry system. BIOCHIMICA ET BIOPHYSICA ACTA-PROTEINS AND PROTEOMICS 2020; 1869:140559. [PMID: 33130090 DOI: 10.1016/j.bbapap.2020.140559] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Revised: 10/23/2020] [Accepted: 10/27/2020] [Indexed: 12/30/2022]
Abstract
Reelin (400 kDa) is an extracellular matrix glycoprotein that is a key regulator of the many significant biological processes including the brain formation, cell aggregation, and dendrite formation. The glycosylation contributes to the nature of the protein through folding, localization and trafficking, solubility, antigenicity, biological activity, and half-life. Although reelin is to be known as a glycoprotein, the knowledge of its glycosylation is very limited. In this study, we aimed to characterize the terminal glycan profile of reelin by lectin blotting and monosaccharide analysis of glycan chains by capillary liquid chromatography electrospray ionization ion trap tandem mass spectrometry (CapLC-ESI-MS/MS) in SH-SY5Y neuroblastoma cell line. According to our results, reelin was detected in different protein fragments (310, 250, and 85 kDa) in addition to full-length form (400 kDa) in the cell line. The reelin glycoprotein was found to carry the β-N-Acetylglucosamine, α-Mannose, β-Galactose, and α-2,3 and α2,6 linked sialic acids by lectin blotting. Nevertheless, these terminal monosaccharides were found in different intensity according to reelin fragments. Besides, we purified a reelin fragment (250 kDa), and we analyzed it for their monosaccharide by CapLC-ESI-MS/MS. We found that reelin contained five types of monosaccharides, which were consisted of N-Acetylgalactosamine, N-Acetylglucosamine, Galactose, Glucose, Mannose and Sialic acid, from high to low abundance respectively. The present results provide a valuable guide for biochemical, genetic, and glycobiology based further experiments about reelin glycosylation in cancer perspective.
Collapse
Affiliation(s)
- Ramiz Demir
- Ege University, Faculty of Science, Department of Biology, Molecular Biology Section, Izmir, Turkey
| | - Umut Şahar
- Ege University, Faculty of Science, Department of Biology, Molecular Biology Section, Izmir, Turkey.
| | - Remziye Deveci
- Ege University, Faculty of Science, Department of Biology, Molecular Biology Section, Izmir, Turkey.
| |
Collapse
|
48
|
Zhang L, Jing H, Li H, Chen W, Luo B, Zhang H, Dong Z, Li L, Su H, Xiong WC, Mei L. Neddylation is critical to cortical development by regulating Wnt/β-catenin signaling. Proc Natl Acad Sci U S A 2020; 117:26448-26459. [PMID: 33020269 PMCID: PMC7584916 DOI: 10.1073/pnas.2005395117] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Wnt signaling plays a critical role in production and differentiation of neurons and undergoes a progressive reduction during cortical development. However, how Wnt signaling is regulated is not well understood. Here we provide evidence for an indispensable role of neddylation, a ubiquitylation-like protein modification, in inhibiting Wnt/β-catenin signaling. We show that β-catenin is neddylated; and inhibiting β-catenin neddylation increases its nuclear accumulation and Wnt/β-catenin signaling. To test this hypothesis in vivo, we mutated Nae1, an obligative subunit of the E1 for neddylation in cortical progenitors. The mutation leads to eventual reduction in radial glia progenitors (RGPs). Consequently, the production of intermediate progenitors (IPs) and neurons is reduced, and neuron migration is impaired, resulting in disorganization of the cerebral cortex. These phenotypes are similar to those of β-catenin gain-of-function mice. Finally, suppressing β-catenin expression is able to rescue deficits of Nae1 mutant mice. Together, these observations identified a mechanism to regulate Wnt/β-catenin signaling in cortical development.
Collapse
Affiliation(s)
- Lei Zhang
- Department of Neurosciences, School of Medicine, Case Western Reserve University, Cleveland, OH 44106
| | - Hongyang Jing
- Department of Neurosciences, School of Medicine, Case Western Reserve University, Cleveland, OH 44106
| | - Haiwen Li
- Department of Neurosciences, School of Medicine, Case Western Reserve University, Cleveland, OH 44106
| | - Wenbing Chen
- Department of Neurosciences, School of Medicine, Case Western Reserve University, Cleveland, OH 44106
| | - Bin Luo
- Department of Neurosciences, School of Medicine, Case Western Reserve University, Cleveland, OH 44106
| | - Hongsheng Zhang
- Department of Neurosciences, School of Medicine, Case Western Reserve University, Cleveland, OH 44106
| | - Zhaoqi Dong
- Department of Neurosciences, School of Medicine, Case Western Reserve University, Cleveland, OH 44106
| | - Lei Li
- Department of Neurosciences, School of Medicine, Case Western Reserve University, Cleveland, OH 44106
| | - Huabo Su
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, GA 30912
| | - Wen-Cheng Xiong
- Department of Neurosciences, School of Medicine, Case Western Reserve University, Cleveland, OH 44106
- Louis Stokes Cleveland Veterans Affairs Medical Center, Cleveland, OH 44106
| | - Lin Mei
- Department of Neurosciences, School of Medicine, Case Western Reserve University, Cleveland, OH 44106;
- Louis Stokes Cleveland Veterans Affairs Medical Center, Cleveland, OH 44106
| |
Collapse
|
49
|
Son AI, Mohammad S, Sasaki T, Ishii S, Yamashita S, Hashimoto-Torii K, Torii M. Dual Role of Rbpj in the Maintenance of Neural Progenitor Cells and Neuronal Migration in Cortical Development. Cereb Cortex 2020; 30:6444-6457. [PMID: 32780108 DOI: 10.1093/cercor/bhaa206] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2016] [Revised: 06/09/2020] [Accepted: 07/06/2020] [Indexed: 12/30/2022] Open
Abstract
The development of the cerebral cortex is directed by a series of methodically precise events, including progenitor cell proliferation, neural differentiation, and cell positioning. Over the past decade, many studies have demonstrated the critical contributions of Notch signaling in neurogenesis, including that in the developing telencephalon. However, in vivo evidence for the role of Notch signaling in cortical development still remains limited partly due to the redundant functions of four mammalian Notch paralogues and embryonic lethality of the knockout mice. Here, we utilized the conditional deletion and in vivo gene manipulation of Rbpj, a transcription factor that mediates signaling by all four Notch receptors, to overcome these challenges and examined the specific roles of Rbpj in cortical development. We report severe structural abnormalities in the embryonic and postnatal cerebral cortex in Rbpj conditional knockout mice, which provide strong in vivo corroboration of previously reported functions of Notch signaling in neural development. Our results also provide evidence for a novel dual role of Rbpj in cell type-specific regulation of two key developmental events in the cerebral cortex: the maintenance of the undifferentiated state of neural progenitor cells, and the radial and tangential allocation of neurons, possibly through stage-dependent differential regulation of Ngn1.
Collapse
Affiliation(s)
- Alexander I Son
- Center for Neuroscience Research, Children's Research Institute, Children's National Hospital, Washington, DC 20010, USA
| | - Shahid Mohammad
- Center for Neuroscience Research, Children's Research Institute, Children's National Hospital, Washington, DC 20010, USA
| | - Toru Sasaki
- Center for Neuroscience Research, Children's Research Institute, Children's National Hospital, Washington, DC 20010, USA
| | - Seiji Ishii
- Center for Neuroscience Research, Children's Research Institute, Children's National Hospital, Washington, DC 20010, USA
| | - Satoshi Yamashita
- Center for Neuroscience Research, Children's Research Institute, Children's National Hospital, Washington, DC 20010, USA
| | - Kazue Hashimoto-Torii
- Center for Neuroscience Research, Children's Research Institute, Children's National Hospital, Washington, DC 20010, USA.,Department of Pediatrics, Pharmacology and Physiology, School of Medicine and Health Sciences, The George Washington University, Washington, DC 20052, USA
| | - Masaaki Torii
- Center for Neuroscience Research, Children's Research Institute, Children's National Hospital, Washington, DC 20010, USA.,Department of Pediatrics, Pharmacology and Physiology, School of Medicine and Health Sciences, The George Washington University, Washington, DC 20052, USA
| |
Collapse
|
50
|
Rahimi-Balaei M, Jiao X, Dalvand A, Shabanipour S, Chung SH, Amiri S, Kong J, Marzban H. Mutations in the Reelin pathway are associated with abnormal expression of microglial IgG FC receptors in the cerebellar cortex. Mol Biol Rep 2020; 47:5323-5331. [PMID: 32594343 DOI: 10.1007/s11033-020-05614-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2020] [Accepted: 06/20/2020] [Indexed: 11/29/2022]
Abstract
Microglia are the immune cells of the central nervous system involved in a variety of developmental processes, such as regulation of cell death and survival, spatial patterning, and contribute to the development of Purkinje cells (PCs) during migration. Microglia express immunoglobulin G Fc receptors (FcgRs). In this report, we describe microglial FcgR expression and its relation to abnormal PC migration in the cerebellum during development. To detect microglial FcgR, the direct anti-IgG (secondary antisera) and high concentrations of Triton X-100 were applied as a method for labeling microglial cells without the use of any specific primary antiserum. By using Acp2-/- mice, which show an excessive PC migration into the molecular layer (ml), and 3 different types of mice with a null to alter the Reelin pathway (Reeler-, Dab1 (SCM)-, and Apoer mutant mice), we studied the location of PCs and the expression of FcgRs. Wild type littermates were used as controls in all studies. We show that the expression of microglial FcgRs was absent and PCs were ectopically located in the white matter in the cerebella of all mutant mice, except for the Acp2-/- mice (PCs were located in the ml). These results suggest a role for FcgRs in the Reelin signaling pathway, not in regulating PC migration, but rather in the adaptation to an environment with a relatively large number of ectopically located PCs. However, the exact correlation between the ectopic location of PCs and lack of FcgRs in Reeler, SCM, and Apoer-/- mice and the presence of FcgRs and directed PC location in the ml in Acp2-/- mice are yet to be determined.
Collapse
Affiliation(s)
- Maryam Rahimi-Balaei
- Department of Human Anatomy and Cell Science, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada.,The Children's Hospital Research Institute of Manitoba (CHRIM), Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada
| | - Xiaodan Jiao
- Department of Human Anatomy and Cell Science, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada
| | - Azadeh Dalvand
- Department of Human Anatomy and Cell Science, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada
| | - Shahin Shabanipour
- Department of Human Anatomy and Cell Science, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada
| | - Seung H Chung
- Department of Oral Biology, University of Illinois at Chicago, Chicago, IL, USA
| | - Shayan Amiri
- Department of Human Anatomy and Cell Science, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada
| | - Jimig Kong
- Department of Human Anatomy and Cell Science, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada
| | - Hassan Marzban
- Department of Human Anatomy and Cell Science, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada. .,The Children's Hospital Research Institute of Manitoba (CHRIM), Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada.
| |
Collapse
|