1
|
Dittmann NL, Chen L, Voronova A. Regulation of neural stem cells by innervating neurons. J Neurochem 2025; 169:e16287. [PMID: 39775528 PMCID: PMC11707326 DOI: 10.1111/jnc.16287] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Revised: 10/25/2024] [Accepted: 10/28/2024] [Indexed: 01/11/2025]
Abstract
The adult central nervous system (CNS) hosts several niches, in which the neural stem and precursor cells (NPCs) reside. The subventricular zone (SVZ) lines the lateral brain ventricles and the subgranular zone (SGZ) is located in the dentate gyrus of the hippocampus. SVZ and SGZ NPCs replace neurons and glia in the homeostatic as well as diseased or injured states. Recently, NPCs have been found to express neurotransmitter receptors, respond to electrical stimulation and interact with neurons, suggesting that neuron-NPC communication is an emerging critical regulator of NPC biology. In this review, we discuss reports that demonstrate neuronal innervation and control of the neurogenic niches. We discuss the role of innervating neurons in regulating NPC fates, such as activation, proliferation, and differentiation. Our review focuses primarily on the innervation of the SVZ niche by the following neuronal types: glutamatergic, GABAergic projection and interneurons, cholinergic, dopaminergic, serotonergic, neuropeptidergic, nitrergic, and noradrenergic. We also discuss the origins of SVZ niche innervating neurons, such as striatum, cortex, basal ganglia, raphe nuclei, substantia nigra and ventral tegmental area, hypothalamus, and locus coeruleus. Our review highlights the various roles of innervating neurons in SVZ NPC fates in a spatiotemporal manner and emphasizes a need for future investigation into the impact of neuronal innervation on NPC gliogenesis.
Collapse
Affiliation(s)
| | - Lauren Chen
- Department of Medical Genetics, Faculty of Medicine & DentistryUniversity of AlbertaEdmontonAlbertaCanada
- Department of Cell Biology, Faculty of Medicine & DentistryUniversity of AlbertaEdmontonAlbertaCanada
| | - Anastassia Voronova
- Neurosciences and Mental Health InstituteUniversity of AlbertaEdmontonAlbertaCanada
- Department of Medical Genetics, Faculty of Medicine & DentistryUniversity of AlbertaEdmontonAlbertaCanada
- Department of Cell Biology, Faculty of Medicine & DentistryUniversity of AlbertaEdmontonAlbertaCanada
- Faculty of Medicine & Dentistry, MS CentreUniversity of AlbertaEdmontonAlbertaCanada
- Women and Children's Health Research InstituteUniversity of AlbertaEdmontonAlbertaCanada
| |
Collapse
|
2
|
Louhivuori LM, Turunen PM, Louhivuori V, Al Rayyes I, Nordström T, Uhlén P, Åkerman KE. Neurotransmitters and Endothelins Acting on Radial Glial G-Protein-Coupled Receptors Are, Through Proteolytic NRG/ErbB4 Activation, Able to Modify the Migratory Behavior of Neocortical Cells and Mediate Bipolar-to-Multipolar Transition. Stem Cells Dev 2020; 29:1160-1177. [PMID: 31941419 DOI: 10.1089/scd.2019.0133] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Cell-cell communication plays a central role in the guidance of migrating neurons during the development of the cerebral cortex. Neuregulins (NRGs) are essential mediators for migration and maintenance of the radial glial scaffold. We show, in this study that soluble NRG reduces neuronal motility, causes transition of bipolar cells to multipolar ones, and induces neuronal mitosis. Blocking the NRG receptor, ErbB4, results in reduction of neuron-neuron and neuron-radial glial contacts and causes an increase in neuronal motility. Blocking the radial glial metabotropic glutamate receptor 5 (mGluR5), the nonselective cation channel transient receptor potential 3 (TRPC3), or matrix metalloproteinases (MMPs) results in similar effects as ErbB4 blockade. Soluble NRG counteract the changes in motility pattern. Stimulation of other radial glial G-protein-coupled receptors (GPCRs), such as muscarinic acetylcholine receptors or endothelin receptors counteract all the effect of mGluR5 blockade, but not that of ErbB4, TRPC3, and MMP blockade. The results indicate that neurotransmitters and endothelins acting on radial glial GPCRs are, through proteolytic NRG/ErbB4 activation, able to modify the migratory behavior of neurons.
Collapse
Affiliation(s)
- Lauri M Louhivuori
- Department of Physiology, Faculty of Medicine, Biomedicum Helsinki, University of Helsinki, Helsinki, Finland.,Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Pauli M Turunen
- Department of Physiology, Faculty of Medicine, Biomedicum Helsinki, University of Helsinki, Helsinki, Finland
| | - Verna Louhivuori
- Department of Physiology, Faculty of Medicine, Biomedicum Helsinki, University of Helsinki, Helsinki, Finland
| | - Ibrahim Al Rayyes
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Tommy Nordström
- Department of Physiology, Faculty of Medicine, Biomedicum Helsinki, University of Helsinki, Helsinki, Finland
| | - Per Uhlén
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Karl E Åkerman
- Department of Physiology, Faculty of Medicine, Biomedicum Helsinki, University of Helsinki, Helsinki, Finland
| |
Collapse
|
3
|
Tannic acid inhibits electrogenic Na+/HCO3- co-transporter activity in embryonic neural stem cell-derived radial glial-like cells. Neuroreport 2020; 31:57-63. [PMID: 31714480 PMCID: PMC6903378 DOI: 10.1097/wnr.0000000000001372] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Self-renewing neural stem cells and progenitor cells are cell populations that generate radial glial cells and neurons through asymmetric division. Regulation of intracellular pH in stem cells with high metabolic activity is critical for both cell signaling and proliferation. We have recently found that a S0859-inhibitable electrogenic Na+/HCO3− co-transporter (NBCe1, Slc4a4), is the primary pHi regulatory mechanism in stem cell-derived radial glial-like cells. Here we show, by using the voltage-sensitive fluorescent dye DiBAC4(3) and BCECF, a pH-sensitive dye, that an antioxidant, tannic acid (100 µM), can inhibit potassium- and calcium-dependent rapid changes in membrane potential and NBCe1 mediated pHi regulation in brain-derived glial-like cells in vitro. Furthermore, neural stem cell differentiation and neurosphere formation (proliferation) were completely inhibited by tannic acid. The present study provides evidence that tannic acid is a natural inhibitor of NBCe1. It is tempting to speculate that tannic acid or related compounds that inhibits NBCe1-mediated pHi regulation in glial-like cells may also have bearing on the treatment of glial neoplasms.
Collapse
|
4
|
Thompson EG, Sontheimer H. Acetylcholine Receptor Activation as a Modulator of Glioblastoma Invasion. Cells 2019; 8:cells8101203. [PMID: 31590360 PMCID: PMC6829263 DOI: 10.3390/cells8101203] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2019] [Revised: 09/30/2019] [Accepted: 10/01/2019] [Indexed: 12/20/2022] Open
Abstract
Grade IV astrocytomas, or glioblastomas (GBMs), are the most common malignant primary brain tumor in adults. The median GBM patient survival of 12–15 months has remained stagnant, in spite of treatment strategies, making GBMs a tremendous challenge clinically. This is at least in part due to the complex interaction of GBM cells with the brain microenvironment and their tendency to aggressively infiltrate normal brain tissue. GBMs frequently invade supratentorial brain regions that are richly innervated by neurotransmitter projections, most notably acetylcholine (ACh). Here, we asked whether ACh signaling influences the biology of GBMs. We examined the expression and function of known ACh receptors (AChRs) in large GBM datasets, as well as, human GBM cell lines and patient-derived xenograft lines. Using RNA-Seq data from the “The Cancer Genome Atlas” (TCGA), we confirmed the expression of AChRs and demonstrated the functionality of these receptors in GBM cells with time-lapse calcium imaging. AChR activation did not alter cell proliferation or migration, however, it significantly increased cell invasion through complex extracellular matrices. This was due to the enhanced activity of matrix metalloproteinase-9 (MMP-9) from GBM cells, which we found to be dependent on an intracellular calcium-dependent mechanism. Consistent with these findings, AChRs were significantly upregulated in regions of GBM infiltration in situ (Ivy Glioblastoma Atlas Project) and elevated expression of muscarinic AChR M3 correlated with reduced patient survival (TCGA). Data from the Repository for Molecular Brain Neoplasia Data (REMBRANDT) dataset also showed the co-expression of choline transporters, choline acetyltransferase, and vesicular acetylcholine transporters, suggesting that GBMs express all the proteins required for ACh synthesis and release. These findings identify ACh as a modulator of GBM behavior and posit that GBMs may utilize ACh as an autocrine signaling molecule.
Collapse
Affiliation(s)
- Emily G Thompson
- Glial Biology in Health, Disease and Cancer Center, Fralin Biomedical Institute at Virginia Tech-Carilion, Roanoke, VA 24016, USA.
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA.
| | - Harald Sontheimer
- Glial Biology in Health, Disease and Cancer Center, Fralin Biomedical Institute at Virginia Tech-Carilion, Roanoke, VA 24016, USA.
- School of Neuroscience, Virginia Polytechnic Institute and State University, Blacksburg, VA 24061, USA.
| |
Collapse
|
5
|
Nordström T, Andersson LC, Åkerman KE. Regulation of intracellular pH by electrogenic Na+/HCO3– co-transporters in embryonic neural stem cell-derived radial glia-like cells. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2019; 1861:1037-1048. [DOI: 10.1016/j.bbamem.2019.03.007] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/01/2018] [Revised: 03/11/2019] [Accepted: 03/14/2019] [Indexed: 01/19/2023]
|
6
|
Achuta VS, Möykkynen T, Peteri UK, Turconi G, Rivera C, Keinänen K, Castrén ML. Functional changes of AMPA responses in human induced pluripotent stem cell-derived neural progenitors in fragile X syndrome. Sci Signal 2018; 11:11/513/eaan8784. [PMID: 29339535 DOI: 10.1126/scisignal.aan8784] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Altered neuronal network formation and function involving dysregulated excitatory and inhibitory circuits are associated with fragile X syndrome (FXS). We examined functional maturation of the excitatory transmission system in FXS by investigating the response of FXS patient-derived neural progenitor cells to the glutamate analog (AMPA). Neural progenitors derived from induced pluripotent stem cell (iPSC) lines generated from boys with FXS had augmented intracellular Ca2+ responses to AMPA and kainate that were mediated by Ca2+-permeable AMPA receptors (CP-AMPARs) lacking the GluA2 subunit. Together with the enhanced differentiation of glutamate-responsive cells, the proportion of CP-AMPAR and N-methyl-d-aspartate (NMDA) receptor-coexpressing cells was increased in human FXS progenitors. Differentiation of cells lacking GluA2 was also increased and paralleled the increased inward rectification in neural progenitors derived from Fmr1-knockout mice (the FXS mouse model). Human FXS progenitors had increased the expression of the precursor and mature forms of miR-181a, a microRNA that represses translation of the transcript encoding GluA2. Blocking GluA2-lacking, CP-AMPARs reduced the neurite length of human iPSC-derived control progenitors and further reduced the shortened length of neurites in human FXS progenitors, supporting the contribution of CP-AMPARs to the regulation of progenitor differentiation. Furthermore, we observed reduced expression of Gria2 (the GluA2-encoding gene) in the frontal lobe of FXS mice, consistent with functional changes of AMPARs in FXS. Increased Ca2+ influx through CP-AMPARs may increase the vulnerability and affect the differentiation and migration of distinct cell populations, which may interfere with normal circuit formation in FXS.
Collapse
Affiliation(s)
- Venkat Swaroop Achuta
- Department of Physiology, Faculty of Medicine, University of Helsinki, P.O. Box 63, FIN-00014 Helsinki, Finland
| | - Tommi Möykkynen
- Division of Biochemistry and Biotechnology, Department of Biosciences, University of Helsinki, P.O. Box 56, FIN-00014, Helsinki, Finland
| | - Ulla-Kaisa Peteri
- Department of Physiology, Faculty of Medicine, University of Helsinki, P.O. Box 63, FIN-00014 Helsinki, Finland
| | - Giorgio Turconi
- Department of Physiology, Faculty of Medicine, University of Helsinki, P.O. Box 63, FIN-00014 Helsinki, Finland
| | - Claudio Rivera
- Neuroscience Center, University of Helsinki, P.O. Box 56, FIN-00014 Helsinki, Finland.,Institut de Neurobiologie de la Méditerranée, INSERM, Unité 901, 13009 Marseille, France.,Aix-Marseille Université, Unité Mixte de Recherche 901, 13273 Marseille, France
| | - Kari Keinänen
- Division of Biochemistry and Biotechnology, Department of Biosciences, University of Helsinki, P.O. Box 56, FIN-00014, Helsinki, Finland
| | - Maija L Castrén
- Department of Physiology, Faculty of Medicine, University of Helsinki, P.O. Box 63, FIN-00014 Helsinki, Finland. .,Rinnekoti Foundation, Rinnekodintie 10, FIN-02980 Espoo, Finland.,Autism Foundation, Kuortaneenkatu 7B, FIN-00520 Helsinki, Finland
| |
Collapse
|
7
|
Theisen U, Hennig C, Ring T, Schnabel R, Köster RW. Neurotransmitter-mediated activity spatially controls neuronal migration in the zebrafish cerebellum. PLoS Biol 2018; 16:e2002226. [PMID: 29300740 PMCID: PMC5754045 DOI: 10.1371/journal.pbio.2002226] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2017] [Accepted: 11/22/2017] [Indexed: 11/27/2022] Open
Abstract
Neuronal migration during embryonic development contributes to functional brain circuitry. Many neurons migrate in morphologically distinct stages that coincide with differentiation, requiring tight spatial regulation. It had been proposed that neurotransmitter-mediated activity could exert this control. Here, we demonstrate that intracellular calcium transients occur in cerebellar neurons of zebrafish embryos during migration. We show that depolarization increases and hyperpolarization reduces the speed of tegmental hindbrain neurons using optogenetic tools and advanced track analysis optimized for in vivo migration. Finally, we introduce a compound screening assay to identify acetylcholine (ACh), glutamate, and glycine as regulators of migration, which act regionally along the neurons’ route. We summarize our findings in a model describing how different neurotransmitters spatially interact to control neuronal migration. The high evolutionary conservation of the cerebellum and hindbrain makes it likely that polarization state-driven motility constitutes an important principle in building a functional brain. Postmitotic neurons migrate from their site of origin to their final destination in the developing brain to form functional structures. These neurons typically follow defined routes through the tissue. Previous studies investigating progress along such route have identified neurotransmitters—chemicals that transmit the signals between neurons—as important regulators in neuronal migration using mostly rodent brain slice cultures and cultivated neurons. In this study, we use live zebrafish embryos to test the influence of neurotransmitters on migrating hindbrain neurons. First, we demonstrate that calcium transients can be measured in these neurons using genetically encoded reporters. Next, we use optogenetic channels to specifically de- or hyperpolarize the plasma membrane of the neurons to show that the polarization state is linked to migratory speed. Finally, we use a screening method to identify the neurotransmitter systems involved in migration progress control. We summarize these findings in a model that suggests that there are regions of influence for different neurotransmitters that act successively on the neurons to ensure their timely arrival at their destination.
Collapse
Affiliation(s)
- Ulrike Theisen
- Technische Universität Braunschweig, Zoological Institute, Cellular and Molecular Neurobiology, Braunschweig, Germany
| | - Christian Hennig
- Technische Universität Braunschweig, Institute for Genetics, Braunschweig, Germany
| | - Tobias Ring
- Technische Universität Braunschweig, Institute for Engineering Design, Vibroacoustics, Braunschweig, Germany
| | - Ralf Schnabel
- Technische Universität Braunschweig, Institute for Genetics, Braunschweig, Germany
| | - Reinhard W. Köster
- Technische Universität Braunschweig, Zoological Institute, Cellular and Molecular Neurobiology, Braunschweig, Germany
- * E-mail:
| |
Collapse
|
8
|
Louhivuori LM, Turunen PM, Louhivuori V, Yellapragada V, Nordström T, Uhlén P, Åkerman KE. Regulation of radial glial process growth by glutamate via mGluR5/TRPC3 and neuregulin/ErbB4. Glia 2017; 66:94-107. [PMID: 28887860 DOI: 10.1002/glia.23230] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2016] [Revised: 08/18/2017] [Accepted: 08/24/2017] [Indexed: 11/09/2022]
Abstract
Radial glial cells play an essential role through their function as guides for neuronal migration during development. Disruption of metabotropic glutamate receptor 5 (mGluR5) function retards the growth of radial glial processes in vitro. Neuregulins (NRG) are activated by proteolytic cleavage and regulate (radial) glial maintenance via ErbB3/ErbB4 receptors. We show here that blocking ErbB4 disrupts radial process extension. Soluble NRG acting on ErbB4 receptors is able to promote radial process extension in particular where process elongation has been impeded by blockade of mGluR5, the nonselective cation channel canonical transient receptor potential 3 (TRPC3), or matrix metalloproteases (MMP). NRG does not restore retarded process growth caused by ErbB4 blockade. Stimulation of muscarinic receptors restores process elongation due to mGluR5 blockade but not that caused by TRPC3, MMP or ErbB4 blockade suggesting that muscarinic receptors can replace mGluR5 with respect to radial process extension. Additionally, NRG/ErbB4 causes Ca2+ mobilization in a population of cells through cooperation with ErbB1 receptors. Our results indicate that mGluR5 promotes radial process growth via NRG activation by a mechanism involving TRPC3 channels and MMPs. Thus neurotransmitters acting on G-protein coupled receptors could play a central role in the maintenance of the radial glial scaffold through activation of NRG/ErbB4 signaling.
Collapse
Affiliation(s)
- Lauri M Louhivuori
- University of Helsinki, Biomedicum, Medicum/Physiology, Helsinki, FIN-00014, Finland.,Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, SE-171 77, Sweden
| | - Pauli M Turunen
- University of Helsinki, Biomedicum, Medicum/Physiology, Helsinki, FIN-00014, Finland
| | - Verna Louhivuori
- University of Helsinki, Biomedicum, Medicum/Physiology, Helsinki, FIN-00014, Finland
| | | | - Tommy Nordström
- University of Helsinki, Biomedicum, Medicum/Physiology, Helsinki, FIN-00014, Finland
| | - Per Uhlén
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, SE-171 77, Sweden
| | - Karl E Åkerman
- University of Helsinki, Biomedicum, Medicum/Physiology, Helsinki, FIN-00014, Finland
| |
Collapse
|
9
|
Kärkkäinen V, Pomeshchik Y, Savchenko E, Dhungana H, Kurronen A, Lehtonen S, Naumenko N, Tavi P, Levonen AL, Yamamoto M, Malm T, Magga J, Kanninen KM, Koistinaho J. Nrf2 regulates neurogenesis and protects neural progenitor cells against Aβ toxicity. Stem Cells 2015; 32:1904-16. [PMID: 24753106 DOI: 10.1002/stem.1666] [Citation(s) in RCA: 113] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2013] [Accepted: 01/15/2014] [Indexed: 12/25/2022]
Abstract
Neural stem/progenitor cells (NPCs) proliferate and produce new neurons in neurogenic areas throughout the lifetime. While these cells represent potential therapeutic treatment of neurodegenerative diseases, regulation of neurogenesis is not completely understood. We show that deficiency of nuclear factor erythroid 2-related factor (Nrf2), a transcription factor induced in response to oxidative stress, prevents the ischemia-induced increase in newborn neurons in the subgranular zone of the dentate gyrus. Consistent with this finding, the growth of NPC neurospheres was increased by lentivirus-mediated overexpression of Nrf2 gene or by treatment with pyrrolidine dithiocarbamate (PDTC), an Nrf2 activating compound. Also, neuronal differentiation of NPCs was increased by Nrf2 overexpression or PDTC treatment but reduced by Nrf2 deficiency. To investigate the impact of Nrf2 on NPCs in Alzheimer's disease (AD), we treated NPCs with amyloid beta (Aβ), a toxic peptide associated with neurodegeneration and cognitive abnormalities in AD. We found that Aβ1-42-induced toxicity and reduction in neurosphere proliferation were prevented by Nrf2 overexpression, while Nrf2 deficiency enhanced the Aβ1-42-induced reduction of neuronal differentiation. On the other hand, Aβ1-40 had no effect on neurosphere proliferation in wt NPCs but increased the proliferation of Nrf2 overexpressing neurospheres and reduced it in Nrf2-deficient neurospheres. These results suggest that Nrf2 is essential for neuronal differentiation of NPCs, regulates injury-induced neurogenesis and provides protection against Aβ-induced NPC toxicity.
Collapse
Affiliation(s)
- Virve Kärkkäinen
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
10
|
Louhivuori LM, Jansson L, Turunen PM, Jäntti MH, Nordström T, Louhivuori V, Åkerman KE. Transient receptor potential channels and their role in modulating radial glial-neuronal interaction: a signaling pathway involving mGluR5. Stem Cells Dev 2014; 24:701-13. [PMID: 25347706 DOI: 10.1089/scd.2014.0209] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
The guidance of developing neurons to the right position in the central nervous system is of central importance in brain development. Canonical transient receptor potential (TRPC) channels are thought to mediate turning responses of growth cones to guidance cues through fine control of calcium transients. Proliferating and 1- to 5-day-differentiated neural progenitor cells (NPCs) showed expression of Trpc1 and Trpc3 mRNA, while Trpc4-7 was not clearly detected. Time-lapse imaging showed that the motility pattern of neuronal cells was phasic with bursts of rapid movement (>60 μm/h), changes in direction, and intermittent slow phases or stallings (<40 μm/h), which frequently occurred in close contact with radial glial processes. Genetic interference with the TRPC3 and TRPC1 channel enhanced the motility of NPCs (burst frequency/stalling frequency). TRPC3-deficient cells or cells treated with the TRPC3 blocker pyr3 infrequently changed direction and seldom contacted radial glial processes. TRPC channels are also activated by group I metabotropic glutamate receptors (mGluR1 and mGluR5). As shown here, pyr3 blocked the calcium response mediated through mGluR5 in radial glial processes. Furthermore, 2-methyl-6-(phenylethynyl)pyridine, a blocker of mGluR5, affected the motility pattern in a similar way as TRPC3/6 double knockout or pyr3. The results suggest that radial glial cells exert attractant signals to migrating neuronal cells, which alter their motility pattern. Our results suggest that mGluR5 acting through TRPC3 is of central importance in radial glial-mediated neuronal guidance.
Collapse
Affiliation(s)
- Lauri M Louhivuori
- Biomedicum Helsinki, Institute of Biomedicine/Physiology, University of Helsinki , Helsinki, Finland
| | | | | | | | | | | | | |
Collapse
|
11
|
Erichsen JL, Blaabjerg M, Bogetofte H, Serrano AM, Meyer M. Group I Metabotropic Glutamate Receptors: A Potential Target for Regulation of Proliferation and Differentiation of an Immortalized Human Neural Stem Cell Line. Basic Clin Pharmacol Toxicol 2014; 116:329-36. [DOI: 10.1111/bcpt.12324] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2014] [Accepted: 09/03/2014] [Indexed: 11/28/2022]
Affiliation(s)
- Julie Ladeby Erichsen
- Department of Neurobiology Research; Institute of Molecular Medicine; University of Southern Denmark; Odense Denmark
| | - Morten Blaabjerg
- Department of Neurology; Odense University Hospital; Odense Denmark
| | - Helle Bogetofte
- Department of Neurobiology Research; Institute of Molecular Medicine; University of Southern Denmark; Odense Denmark
| | - Alberto Martinez Serrano
- Department of Molecular Biology and Center of Molecular Biology Severo Ochoa; University Autonoma Madrid-C.S.I.C. Campus Cantoblanco; Madrid Spain
| | - Morten Meyer
- Department of Neurobiology Research; Institute of Molecular Medicine; University of Southern Denmark; Odense Denmark
| |
Collapse
|
12
|
Tissue plasminogen activator contributes to alterations of neuronal migration and activity-dependent responses in fragile X mice. J Neurosci 2014; 34:1916-23. [PMID: 24478370 DOI: 10.1523/jneurosci.3753-13.2014] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022] Open
Abstract
Fragile X syndrome (FXS) is the most common inherited neurodevelopmental disorder with intellectual disability. Here, we show that the expression of tissue plasminogen activator (tPA) is increased in glial cells differentiated from neural progenitors of Fmr1 knock-out mice, a mouse model for FXS, and that tPA is involved in the altered migration and differentiation of these progenitors lacking FMR1 protein (FMRP). When tPA function is blocked with an antibody, enhanced migration of doublecortin-immunoreactive neurons in 1 d differentiated FMRP-deficient neurospheres is normalized. In time-lapse imaging, blocking the tPA function promotes early glial differentiation and reduces the velocity of nuclear movement of FMRP-deficient radial glia. In addition, we show that enhanced intracellular Ca(2+) responses to depolarization with potassium are prevented by the treatment with the tPA-neutralizing antibody in FMRP-deficient cells during early neural progenitor differentiation. Alterations of the tPA expression in the embryonic, postnatal, and adult brain of Fmr1 knock-out mice suggest an important role for tPA in the abnormal neuronal differentiation and plasticity in FXS. Altogether, the results indicate that tPA may prove to be an interesting potential target for pharmacological intervention in FXS.
Collapse
|
13
|
Jansson LC, Åkerman KE. The role of glutamate and its receptors in the proliferation, migration, differentiation and survival of neural progenitor cells. J Neural Transm (Vienna) 2014; 121:819-36. [DOI: 10.1007/s00702-014-1174-6] [Citation(s) in RCA: 88] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2013] [Accepted: 02/04/2014] [Indexed: 12/19/2022]
|
14
|
Schwartz L, Spitsin SV, Meshki J, Tuluc F, Douglas SD, Wolfe JH. Substance P enhances HIV-1 infection in human fetal brain cell cultures expressing full-length neurokinin-1 receptor. J Neurovirol 2013; 19:219-27. [PMID: 23765222 PMCID: PMC3719168 DOI: 10.1007/s13365-013-0166-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2012] [Revised: 04/05/2013] [Accepted: 04/16/2013] [Indexed: 12/12/2022]
Abstract
The associations between the neurokinin-1 receptor (NK-1R), substance P (SP), and HIV-1 were investigated in neurosphere-derived cultures of microglial-depleted human fetal brain cells (HFBC). Full-length NK-1R was identified in HFBC cultures. SP treatment of the HFBC increased intracellular calcium mobilization and decreased electrical impedance, both of which were blocked by the NK-1R antagonist aprepitant. SP treatment of HIV-1-infected HFBC upregulated HIV-1 expression. These data show that human neural cells grown from neurospheres express functional full length NK-1R that is responsive to SP, and that SP enhanced HIV-1 infection in HBFC.
Collapse
Affiliation(s)
- Lynnae Schwartz
- Research Institute, Children's Hospital of Philadelphia, Suite 1208, 3615 Civic Center Boulevard, Philadelphia, PA 19104, USA.
| | | | | | | | | | | |
Collapse
|
15
|
Castrén ML, Castrén E. BDNF in fragile X syndrome. Neuropharmacology 2013; 76 Pt C:729-36. [PMID: 23727436 DOI: 10.1016/j.neuropharm.2013.05.018] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2013] [Revised: 05/07/2013] [Accepted: 05/08/2013] [Indexed: 12/12/2022]
Abstract
Fragile X syndrome (FXS) is a monogenic disorder that is caused by the absence of FMR1 protein (FMRP). FXS serves as an excellent model disorder for studies investigating disturbed molecular mechanisms and synapse function underlying cognitive impairment, autism, and behavioral disturbance. Abnormalities in dendritic spines and synaptic transmission in the brain of FXS individuals and mouse models for FXS indicate perturbations in the development, maintenance, and plasticity of neuronal network connectivity. However, numerous alterations are found during the early development in FXS, including abnormal differentiation of neural progenitors and impaired migration of newly born neurons. Several aspects of FMRP function are modulated by brain-derived neurotrophic factor (BDNF) signaling. Here, we review the evidence of the role for BDNF in the developing and adult FXS brain. This article is part of the Special Issue entitled 'BDNF Regulation of Synaptic Structure, Function, and Plasticity'.
Collapse
Affiliation(s)
- Maija L Castrén
- Institute of Biomedicine/Physiology, University of Helsinki, P.O. Box 63, FIN-00014 Helsinki, Finland; Rinnekoti Foundation, Rinnekodintie 10, FIN-02980 Espoo, Finland.
| | | |
Collapse
|
16
|
Louhivuori LM, Louhivuori V, Wigren HK, Hakala E, Jansson LC, Nordström T, Castrén ML, Akerman KE. Role of low voltage activated calcium channels in neuritogenesis and active migration of embryonic neural progenitor cells. Stem Cells Dev 2013; 22:1206-19. [PMID: 23234460 DOI: 10.1089/scd.2012.0234] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
The central role of calcium influx and electrical activity in embryonic development raises important questions about the role and regulation of voltage-dependent calcium influx. Using cultured neural progenitor cell (NPC) preparations, we recorded barium currents through voltage-activated channels using the whole-cell configuration of the patch-clamp technique and monitored intracellular free calcium concentrations with Fura-2 digital imaging. We found that NPCs as well as expressing high-voltage-activated (HVA) calcium channels express functional low-threshold voltage-dependent calcium channels in the very early stages of differentiation (5 h to 1 day). The size of the currents recorded at -50 versus -20 mV after 1 day in differentiation was dependent on the nature of the charge carrier. Peak currents measured at -20 mV in the presence 10 mM Ca2+ instead of 10 mM Ba2+ had a tendency to be smaller, whereas the nature of the divalent species did not influence the amplitude measured at -50 mV. The T-type channel blockers mibefradil and NNC 55-0396 significantly reduced the calcium responses elicited by depolarizing with extracellular potassium, while the overall effect of the HVA calcium channel blockers was small at differentiation day 1. At differentiation day 20, the calcium responses were effectively blocked by nifedipine. Time-lapse imaging of differentiating neurospheres cultured in the presence of low-voltage-activated (LVA) blockers showed a significant decrease in the number of active migrating neuron-like cells and neurite extensions. Together, these data provide evidence that LVA calcium channels are involved in the physiology of differentiating and migrating NPCs.
Collapse
Affiliation(s)
- Lauri M Louhivuori
- Biomedicum Helsinki, Institute of Biomedicine/Physiology, University of Helsinki, Helsinki, Finland.
| | | | | | | | | | | | | | | |
Collapse
|
17
|
Jansson LC, Louhivuori L, Wigren HK, Nordström T, Louhivuori V, Castrén ML, Åkerman KE. Effect of glutamate receptor antagonists on migrating neural progenitor cells. Eur J Neurosci 2013; 37:1369-82. [PMID: 23383979 DOI: 10.1111/ejn.12152] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2012] [Revised: 01/10/2013] [Accepted: 01/10/2013] [Indexed: 02/03/2023]
Abstract
Neurotransmitters such as glutamate are potential regulators of neurogenesis. Interference with defined glutamate receptor subtypes affects proliferation, migration and differentiation of neural progenitor cells. The cellular targets for the actions of different glutamate receptor ligands are less well known. In this study we have combined calcium imaging, measurement of membrane potential, time-lapse imaging and immunocytochemistry to obtain a spatial overview of migrating mouse embryonic neural progenitor cell-derived cells responding to glutamate receptor agonists and antagonists. Responses via metabotropic glutamate receptor 5 correlated with radial glial cells and dominated in the inner migration zones close to the neurosphere. Block of metabotropic glutamate receptor 5 resulted in shorter radial glial processes, a transient increase in neuron-like cells emerging from the neurosphere and increased motility of neuron-like cells. α-Amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA)/kainate receptors are present on the majority of migrating neuronal cells, which with time accumulate at the outer edge of the migration zone. Blocking these receptors leads to an enhanced extension of radial glial processes and a reduced motility of neuron-like cells. Our results indicate that functional glutamate receptors have profound effects on the motility of neural progenitor cells. The main target for metabotropic glutamate receptor 5 appears to be radial glial cells while AMPA/kainate receptors are mainly expressed in newborn neuronal cells and regulate the migratory progress of these cells. The results suggest that both metabotropic glutamate receptor 5 and AMPA/kainate receptors are of importance for the guidance of migrating embryonic progenitor cells.
Collapse
Affiliation(s)
- Linda C Jansson
- Biomedicum Helsinki, Institute of Biomedicine/Physiology, University of Helsinki, Helsinki, Finland
| | | | | | | | | | | | | |
Collapse
|
18
|
Li F, Ohtani A, Senzaki K, Shiga T. Receptor-dependent regulation of dendrite formation of noradrenaline and dopamine in non-GABAergic cerebral cortical neurons. Dev Neurobiol 2012; 73:370-83. [PMID: 23135899 DOI: 10.1002/dneu.22065] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2012] [Revised: 09/18/2012] [Accepted: 10/31/2012] [Indexed: 12/12/2022]
Abstract
The present study characterized the receptor-dependent regulation of dendrite formation of noradrenaline (NA) and dopamine (DA) in cultured neurons obtained from embryonic day 16 rat cerebral cortex. Morphological diversity of cortical dendrites was analyzed on various features: dendrite initiation, dendrite outgrowth, and dendrite branching. Using a combination of immunocytochemical markers of dendrites and GABAergic neurons, we focused on the dendrite morphology of non-GABAergic neurons. Our results showed that (1) NA inhibited the dendrite branching, (2) β adrenergic receptor (β-AR) agonist inhibited the dendrite initiation, while promoted the dendrite outgrowth, (3) β1-AR and β2-AR were present in all the cultured neurons, and both agonists inhibited the dendrite initiation, while only β1-AR agonist induced the dendrite branching; (4) DA inhibited the dendrite outgrowth, (5) D1 receptor agonist inhibited the dendrite initiation, while promoted the dendrite branching. In conclusion, this study compared the effects of NA, DA and their receptors and showed that NA and DA regulate different features on the dendrite formation of non-GABAergic cortical neurons, depending on the receptors.
Collapse
Affiliation(s)
- Fei Li
- Graduate School of Comprehensive Human Sciences, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, 305-8577, Ibaraki, Japan
| | | | | | | |
Collapse
|
19
|
Jansson L, Louhivuori L, Wigren HK, Nordström T, Louhivuori V, Castrén M, Åkerman K. Brain-derived neurotrophic factor increases the motility of a particular N-methyl-d-aspartate /GABA-responsive subset of neural progenitor cells. Neuroscience 2012; 224:223-34. [DOI: 10.1016/j.neuroscience.2012.08.038] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2012] [Revised: 08/15/2012] [Accepted: 08/16/2012] [Indexed: 10/28/2022]
|
20
|
Abstract
OBJECTIVE High doses or prolonged exposure to ketamine increase neuronal apoptosis in the developing brain, although effects on neural stem progenitor cells remain unexplored. This study investigated dose- and time-dependent responses to ketamine on cell death and neurogenesis in cultured rat fetal cortical neural stem progenitor cells. DESIGN Laboratory-based study. SETTING University research laboratory. SUBJECT Sprague-Dawley rats. INTERVENTIONS Neural stem progenitor cells were isolated from the cortex of Sprague-Dawley rat fetuses on embryonic day 17. In dose-response experiments, cultured neural stem progenitor cells were exposed to different concentrations of ketamine (0-100 µM) for 24 hrs. In time-course experiments, neural stem progenitor cells cultures were exposed to 10 µM ketamine for different durations (0-48 hrs). MEASUREMENTS AND MAIN RESULTS Apoptosis and necrosis in neural stem progenitor cells were assessed using activated caspase-3 immunostaining and lactate dehydrogenase assays, respectively. Proliferative changes in neural stem progenitor cells were detected using bromo-deoxyuridine incorporation and Ki67 immunostaining. Neuronal differentiation was assessed using Tuj-1 immunostaining. Cultured neural stem progenitor cells were resistant to apoptosis and necrosis following all concentrations and durations of ketamine exposure tested. Ketamine inhibited proliferation with decreased numbers of bromo-deoxyuridine-positive cells following ketamine exposure to 100 µM for 24 hrs (p<.005) or 10 µM for 48 hrs (p< .01), and reduced numbers of Ki67-positive cells following exposure to ketamine concentration>10 µM for 24 hrs (p<.001) or at 10 µM for 48 hrs (p<.01). Ketamine enhanced neuronal differentiation, with all ketamine concentrations increasing Tuj-1-positive neurons (p<.001) after 24-hrs of exposure. This also occurred with all exposures to 10 µM ketamine for >8 hrs (p<.001). CONCLUSIONS Clinically relevant concentrations of ketamine do not induce cell death in neural stem progenitor cells via apoptosis or necrosis. Ketamine alters the proliferation and increases the neuronal differentiation of neural stem progenitor cells isolated from the rat neocortex. These studies imply that ketamine exposure during fetal or neonatal life may alter neurogenesis and subsequent brain development.
Collapse
|
21
|
Angioneural crosstalk in scaffolds with oriented microchannels for regenerative spinal cord injury repair. J Mol Neurosci 2012; 49:334-46. [PMID: 22878912 DOI: 10.1007/s12031-012-9863-9] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2012] [Accepted: 07/23/2012] [Indexed: 01/15/2023]
Abstract
The aim of our work is to utilize the crosstalk between the vascular and the neuronal system to enhance directed neuritogenesis in uniaxial guidance scaffolds for the repair of spinal cord injury. In this study, we describe a method for angioneural regenerative engineering, i.e., for generating biodegradable scaffolds, produced by a combination of controlled freezing (freeze-casting) and lyophilization, which contain longitudinally oriented channels, and provide uniaxial directionality to support and guide neuritogenesis from neuronal cells in the presence of endothelial cells. The optimized scaffolds, composed of 2.5 % gelatin and 1 % genipin crosslinked, were characterized by an elastic modulus of ~51 kPa and longitudinal channels of ~50 μm diameter. The scaffolds support the growth of endothelial cells, undifferentiated or NGF-differentiated PC12 cells, and primary cultures of fetal chick forebrain neurons. The angioneural crosstalk, as generated by first forming endothelial cell monolayers in the scaffolds followed by injection of neuronal cells, leads to the outgrowth of long aligned neurites in the PC12/endothelial cell co-cultures also in the absence of exogenously added nerve growth factor. Neuritogenesis was not observed in the scaffolds in the absence of the endothelial cells. This methodology is a promising approach for neural tissue engineering and may be applicable for regenerative spinal cord injury repair.
Collapse
|
22
|
Abstract
The importance of adult neurogenesis has only recently been accepted, resulting in a completely new field of investigation within stem cell biology. The regulation and functional significance of adult neurogenesis is currently an area of highly active research. G-protein-coupled receptors (GPCRs) have emerged as potential modulators of adult neurogenesis. GPCRs represent a class of proteins with significant clinical importance, because approximately 30% of all modern therapeutic treatments target these receptors. GPCRs bind to a large class of neurotransmitters and neuromodulators such as norepinephrine, dopamine, and serotonin. Besides their typical role in cellular communication, GPCRs are expressed on adult neural stem cells and their progenitors that relay specific signals to regulate the neurogenic process. This review summarizes the field of adult neurogenesis and its methods and specifies the roles of various GPCRs and their signal transduction pathways that are involved in the regulation of adult neural stem cells and their progenitors. Current evidence supporting adult neurogenesis as a model for self-repair in neuropathologic conditions, adult neural stem cell therapeutic strategies, and potential avenues for GPCR-based therapeutics are also discussed.
Collapse
Affiliation(s)
- Van A Doze
- Department of Molecular Cardiology, NB50, Lerner Research Institute, The Cleveland Clinic Foundation, 9500 Euclid Ave., Cleveland, OH 44195, USA
| | | |
Collapse
|
23
|
Effects of acute hypoxia/acidosis on intracellular pH in differentiating neural progenitor cells. Brain Res 2012; 1461:10-23. [PMID: 22608071 DOI: 10.1016/j.brainres.2012.04.043] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2011] [Revised: 03/26/2012] [Accepted: 04/20/2012] [Indexed: 01/19/2023]
Abstract
The response of differentiating mouse neural progenitor cells, migrating out from neurospheres, to conditions simulating ischemia (hypoxia and extracellular or intracellular acidosis) was studied. We show here, by using BCECF and single cell imaging to monitor intracellular pH (pH(i)), that two main populations can be distinguished by exposing migrating neural progenitor cells to low extracellular pH or by performing an acidifying ammonium prepulse. The cells dominating at the periphery of the neurosphere culture, which were positive for neuron specific markers MAP-2, calbindin and NeuN had lower initial resting pH(i) and could also easily be further acidified by lowering the extracellular pH. Moreover, in this population, a more profound acidification was seen when the cells were acidified using the ammonium prepulse technique. However, when the cell population was exposed to depolarizing potassium concentrations no alterations in pH(i) took place in this population. In contrast, depolarization caused an increase in pH(i) (by 0.5 pH units) in the cell population closer to the neurosphere body, which region was positive for the radial cell marker (GLAST). This cell population, having higher resting pH(i) (pH 6.9-7.1) also responded to acute hypoxia. During hypoxic treatment the resting pH(i) decreased by 0.1 pH units and recovered rapidly after reoxygenation. Our results show that migrating neural progenitor cells are highly sensitive to extracellular acidosis and that irreversible damage becomes evident at pH 6.2. Moreover, our results show that a response to acidosis clearly distinguishes two individual cell populations probably representing neuronal and radial cells.
Collapse
|
24
|
Abstract
Neural stem/progenitor cell (NPC) cultures are a tool to study the differentiation of neuronal cells and can be used to model disease conditions in studies investigating the pathological mechanisms affecting the development and cellular plasticity of the central nervous system. There is evidence that abnormalities of NPCs and their differentiation contribute to the pathophysiology of fragile X syndrome. The results obtained with NPC cultures derived from human and mouse brain tissue with the fragile X mutation are in line with the abnormalities of Fmr1-knockout mouse brain in vivo indicating that NPC cultures can be useful as a model for fragile X syndrome.
Collapse
Affiliation(s)
- Maija Castrén
- Institute of Biomedicine/Physiology, University of Helsinki, Helsinki, Finland.
| |
Collapse
|
25
|
Louhivuori V, Vicario A, Uutela M, Rantamäki T, Louhivuori LM, Castrén E, Tongiorgi E, Akerman KE, Castrén ML. BDNF and TrkB in neuronal differentiation of Fmr1-knockout mouse. Neurobiol Dis 2010; 41:469-80. [PMID: 21047554 DOI: 10.1016/j.nbd.2010.10.018] [Citation(s) in RCA: 70] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2010] [Revised: 10/14/2010] [Accepted: 10/27/2010] [Indexed: 01/19/2023] Open
Abstract
Fragile X syndrome (FXS) is a common cause of inherited mental retardation and the best characterized form of autistic spectrum disorders. FXS is caused by the loss of functional fragile X mental retardation protein (FMRP), which leads to abnormalities in the differentiation of neural progenitor cells (NPCs) and in the development of dendritic spines and neuronal circuits. Brain-derived neurotrophic factor (BDNF) and its TrkB receptors play a central role in neuronal maturation and plasticity. We studied BDNF/TrkB actions in the absence of FMRP and show that an increase in catalytic TrkB expression in undifferentiated NPCs of Fmr1-knockout (KO) mice, a mouse model for FXS, is associated with changes in the differentiation and migration of neurons expressing TrkB in neurosphere cultures and in the developing cortex. Aberrant intracellular calcium responses to BDNF and ATP in subpopulations of differentiating NPCs combined with changes in the expression of BDNF and TrkB suggest cell subtype-specific alterations during early neuronal maturation in the absence of FMRP. Furthermore, we show that dendritic targeting of Bdnf mRNA was increased under basal conditions and further enhanced in cortical layer V and hippocampal CA1 neurons of Fmr1-KO mice by pilocarpine-induced neuronal activity represented by convulsive seizures, suggesting that BDNF/TrkB-mediated feedback mechanisms for strengthening the synapses were compromised in the absence of FMRP. Pilocarpine-induced seizures caused an accumulation of Bdnf mRNA transcripts in the most proximal segments of dendrites in cortical but not in hippocampal neurons of Fmr1-KO mice. In addition, BDNF protein levels were increased in the hippocampus but reduced in the cortex of Fmr1-KO mice in line with regional differences of synaptic plasticity in the brain of Fmr1-KO mice. Altogether, the present data suggest that alterations in the BDNF/TrkB signaling modulate brain development and impair synaptic plasticity in FXS.
Collapse
Affiliation(s)
- Verna Louhivuori
- Department of Biomedicine/Physiology, University of Helsinki, PO Box 63, FIN-00014 Helsinki, Finland
| | | | | | | | | | | | | | | | | |
Collapse
|