1
|
Grigorieva YS, Naumova AA, Nikolaeva SD, Ivlev AP, Chernigovskaya EV, Glazova MV. Abnormal Astrocyte Heterogeneity in the Dentate Gyrus of Rats Prone to Audiogenic Seizures Can Be Corrected by the Nootropic Drug Piracetam. Hippocampus 2025; 35:e23679. [PMID: 39711014 DOI: 10.1002/hipo.23679] [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: 08/06/2024] [Revised: 12/04/2024] [Accepted: 12/11/2024] [Indexed: 12/24/2024]
Abstract
Accumulating evidence indicates that inherited astrocyte dysfunction can be a primary trigger for epilepsy development; however, the available data are rather limited. In addition, astrocytes are considered as a perspective target for the design of novel and improvement of the existing antiepileptic therapy. Piracetam and related nootropic drugs are widely used in the therapy of various epileptic disorders, but detailed mechanisms of racetams action and, in particular, their effects on glial functions are poorly understood. In this study, we explored the functional state of astrocytes in the dentate gyrus (DG) of Krushinsky-Molodkina (KM) rats genetically prone to audiogenic seizures and compared the action of piracetam on the DG astrocytes in KM and normal Wistar rats. Wistar and naïve KM rats which received injections of saline (control) or piracetam (100 mg/kg) for 21 days were recruited in our studies. Comparative analysis of control Wistar and KM rats revealed genetically determined abnormalities in DG astrocytes of KM rats including an increased expression of NFIA but a decreased GFAP, ALDH1L1, EAATs, and glutamine synthetase (GS). Piracetam treatment normalized the expression of all studied markers, except NFIA, in KM rats, while in Wistar rats, it potentiated only GS and NFIA. The results suggested that the nootropic and antiepileptic effects of piracetam may be, at least partially, mediated by the modulation of astroglia functions. In addition, analysis of NFIA and GS colocalization revealed the novel pattern of astrocyte heterogeneity in the DG which was significantly altered in epileptic rats but corrected by piracetam.
Collapse
Affiliation(s)
- Yulia S Grigorieva
- Sechenov Institute of Evolutionary Physiology and Biochemistry, The Russian Academy of Sciences, Saint Petersburg, Russia
| | - Alexandra A Naumova
- Sechenov Institute of Evolutionary Physiology and Biochemistry, The Russian Academy of Sciences, Saint Petersburg, Russia
| | - Svetlana D Nikolaeva
- Sechenov Institute of Evolutionary Physiology and Biochemistry, The Russian Academy of Sciences, Saint Petersburg, Russia
| | - Andrey P Ivlev
- Sechenov Institute of Evolutionary Physiology and Biochemistry, The Russian Academy of Sciences, Saint Petersburg, Russia
| | - Elena V Chernigovskaya
- Sechenov Institute of Evolutionary Physiology and Biochemistry, The Russian Academy of Sciences, Saint Petersburg, Russia
| | - Margarita V Glazova
- Sechenov Institute of Evolutionary Physiology and Biochemistry, The Russian Academy of Sciences, Saint Petersburg, Russia
| |
Collapse
|
2
|
Ohyama K, Shinohara HM, Takayama N, Ogawa R, Omura S, Hayashida M, Takahashi T. Differentiation stage-specific expression of transcriptional regulators for epithelial mesenchymal transition in dentate granule progenitors. Front Neurosci 2024; 18:1425849. [PMID: 39268037 PMCID: PMC11390541 DOI: 10.3389/fnins.2024.1425849] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Accepted: 08/05/2024] [Indexed: 09/15/2024] Open
Abstract
During the development of the mouse dentate gyrus (DG), granule neuronal progenitors (GNPs) arise from glial fibrillary acidic protein (GFAP)-expressing neural stem cells in the dentate notch. However, the transcriptional regulators that control their stepwise differentiation remain poorly defined. Since neurogenesis involves epithelial-to-mesenchymal transition (EMT)-like processes, we investigated the spatio-temporal expression profiles of the EMT transcription factors Zeb1, Scratch2 (Scrt2) and Nkx6-2 in relation to known GNP markers. Our results show that Zeb1 and Scrt2 exhibit sequential, but partially overlapping expression across embryonic and postnatal stages of GNP differentiation. Zeb1 is highly enriched in gfap-GFP+/Sox2+ neural stem/progenitor pools and subsets of Tbr2+/Prox1+/NeuroD+ intermediate GNPs, whereas Scrt2 predominates in Tbr2+/Prox1+/NeuroD+ GNPs. Strikingly, the neuronal EMT regulator Nkx6-2 shows selective expression in postnatal Tbr2+/Prox1+ GNPs, but it is excluded from embryonic counterparts. This temporally coordinated yet distinct expression of Zeb1, Scrt2 and Nkx6-2 reveals discrete transcriptional programs orchestrating GNP differentiation and neurogenic progression at embryonic versus postnatal stages of DG neurogenesis.
Collapse
Affiliation(s)
- Kyoji Ohyama
- Department of Histology and Neuroanatomy, Tokyo Medical University, Tokyo, Japan
| | - Hiroshi M Shinohara
- Department of Histology and Neuroanatomy, Tokyo Medical University, Tokyo, Japan
| | - Natsumi Takayama
- Department of Histology and Neuroanatomy, Tokyo Medical University, Tokyo, Japan
| | - Rina Ogawa
- Department of Histology and Neuroanatomy, Tokyo Medical University, Tokyo, Japan
| | - Shoichiro Omura
- Department of Histology and Neuroanatomy, Tokyo Medical University, Tokyo, Japan
| | - Mio Hayashida
- Department of Histology and Neuroanatomy, Tokyo Medical University, Tokyo, Japan
| | - Tokiharu Takahashi
- Department of Histology and Neuroanatomy, Tokyo Medical University, Tokyo, Japan
| |
Collapse
|
3
|
Jimenez-Cyrus D, Adusumilli VS, Stempel MH, Maday S, Ming GL, Song H, Bond AM. Molecular cascade reveals sequential milestones underlying hippocampal neural stem cell development into an adult state. Cell Rep 2024; 43:114339. [PMID: 38852158 PMCID: PMC11320877 DOI: 10.1016/j.celrep.2024.114339] [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: 05/27/2023] [Revised: 04/16/2024] [Accepted: 05/23/2024] [Indexed: 06/11/2024] Open
Abstract
Quiescent adult neural stem cells (NSCs) in the mammalian brain arise from proliferating NSCs during development. Beyond acquisition of quiescence, an adult NSC hallmark, little is known about the process, milestones, and mechanisms underlying the transition of developmental NSCs to an adult NSC state. Here, we performed targeted single-cell RNA-seq analysis to reveal the molecular cascade underlying NSC development in the early postnatal mouse dentate gyrus. We identified two sequential steps, first a transition to quiescence followed by further maturation, each of which involved distinct changes in metabolic gene expression. Direct metabolic analysis uncovered distinct milestones, including an autophagy burst before NSC quiescence acquisition and cellular reactive oxygen species level elevation along NSC maturation. Functionally, autophagy is important for the NSC transition to quiescence during early postnatal development. Together, our study reveals a multi-step process with defined milestones underlying establishment of the adult NSC pool in the mammalian brain.
Collapse
Affiliation(s)
- Dennisse Jimenez-Cyrus
- Department of Neuroscience and Mahoney Institute for Neurosciences, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Vijay S Adusumilli
- Department of Neuroscience and Mahoney Institute for Neurosciences, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Max H Stempel
- Department of Neuroscience and Mahoney Institute for Neurosciences, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Sandra Maday
- Department of Neuroscience and Mahoney Institute for Neurosciences, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Guo-Li Ming
- Department of Neuroscience and Mahoney Institute for Neurosciences, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Cell and Developmental Biology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA; Institute for Regenerative Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Psychiatry, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Hongjun Song
- Department of Neuroscience and Mahoney Institute for Neurosciences, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Cell and Developmental Biology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA; Institute for Regenerative Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Neurosurgery, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA; The Epigenetics Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA.
| | - Allison M Bond
- Department of Neuroscience and Mahoney Institute for Neurosciences, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA; Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Department of Cell, Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.
| |
Collapse
|
4
|
Butruille L, Batailler M, Vaudin P, Pillon D, Migaud M. GFAP-expressing cells in the adult hypothalamus can generate multiple neural cell lineages in vitro. Neurosci Lett 2024; 824:137674. [PMID: 38355005 DOI: 10.1016/j.neulet.2024.137674] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Revised: 01/05/2024] [Accepted: 02/07/2024] [Indexed: 02/16/2024]
Abstract
Adult neural stem cells (NSCs) located in the two canonical neurogenic niches, the subventricular zone (SVZ) and the subgranular zone (SGZ), express the glial fibrillary acidic protein (GFAP). Recently, proliferative activity has been described in the hypothalamus although the characterization of hypothalamic neural stem/progenitor cells (NSPCs) is still uncertain. We therefore investigated whether hypothalamic GFAP-positive cells, as in the SVZ and SGZ, also have neurogenic potential. We used a transgenic mouse line expressing green fluorescent protein (GFP) under the control of the GFAP promoter. GFAP-GFP expressing cells are localized in the ependymal layer as well as in the parenchyma of the mediobasal hypothalamus (MBH) and express Sox2, a marker for NSCs. Interestingly, no sexual dimorphism was observed in the numbers of GFP + and GFP-Sox2 + cells. After cells sorting, these cells were able to generate neurospheres in vitro and give rise to neurons, astrocytes and oligodendrocytes. Taken together, these results show that hypothalamic GFAP-expressing cells form a population of NSPCs.
Collapse
Affiliation(s)
- Lucile Butruille
- CNRS, IFCE, INRAE, Université de Tours, PRC, F-37380 Nouzilly, France
| | - Martine Batailler
- CNRS, IFCE, INRAE, Université de Tours, PRC, F-37380 Nouzilly, France
| | - Pascal Vaudin
- CNRS, IFCE, INRAE, Université de Tours, PRC, F-37380 Nouzilly, France
| | - Delphine Pillon
- CNRS, IFCE, INRAE, Université de Tours, PRC, F-37380 Nouzilly, France
| | - Martine Migaud
- CNRS, IFCE, INRAE, Université de Tours, PRC, F-37380 Nouzilly, France.
| |
Collapse
|
5
|
Portugal CC. Ascorbate and its transporter SVCT2: The dynamic duo's integrated roles in CNS neurobiology and pathophysiology. Free Radic Biol Med 2024; 212:448-462. [PMID: 38182073 DOI: 10.1016/j.freeradbiomed.2023.12.040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 12/11/2023] [Accepted: 12/27/2023] [Indexed: 01/07/2024]
Abstract
Ascorbate is a small antioxidant molecule essential for the proper development and function of the brain. Ascorbate is transported into the brain and between brain cells via the Sodium vitamin C co-transporter 2 (SVCT2). This review provides an in-depth analysis of ascorbate's physiology, including how ascorbate is absorbed from food into the CNS, emphasizing cellular mechanisms of ascorbate recycling and release in different CNS compartments. Additionally, the review delves into the various functions of ascorbate in the CNS, including its impact on epigenetic modulation, synaptic plasticity, and neurotransmission. It also emphasizes ascorbate's role on neuromodulation and its involvement in neurodevelopmental processes and disorders. Furthermore, it analyzes the relationship between the duo ascorbate/SVCT2 in neuroinflammation, particularly its effects on microglial activation, cytokine release, and oxidative stress responses, highlighting its association with neurodegenerative diseases, such as Alzheimer's disease (AD). Overall, this review emphasizes the crucial role of the dynamic duo ascorbate/SVCT2 in CNS physiology and pathology and the need for further research to fully comprehend its significance in a neurobiological context and its potential therapeutic applications.
Collapse
Affiliation(s)
- Camila C Portugal
- I3s - Instituto de Investigação e Inovação em Saúde da Universidade do Porto and IBMC - Instituto de Biologia Molecular e Celular, Rua Alfredo Allen 208, 4200-135, Porto, Portugal.
| |
Collapse
|
6
|
Zhang H, Ishii K, Shibata T, Ishii S, Hirao M, Lu Z, Takamura R, Kitano S, Miyachi H, Kageyama R, Itakura E, Kobayashi T. Fluctuation of lysosomal protein degradation in neural stem cells of the postnatal mouse brain. Development 2024; 151:dev202231. [PMID: 38265146 PMCID: PMC10911176 DOI: 10.1242/dev.202231] [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: 08/02/2023] [Accepted: 01/16/2024] [Indexed: 01/25/2024]
Abstract
Lysosomes are intracellular organelles responsible for degrading diverse macromolecules delivered from several pathways, including the endo-lysosomal and autophagic pathways. Recent reports have suggested that lysosomes are essential for regulating neural stem cells in developing, adult and aged brains. However, the activity of these lysosomes has yet to be monitored in these brain tissues. Here, we report the development of a new probe to measure lysosomal protein degradation in brain tissue by immunostaining. Our results indicate that lysosomal protein degradation fluctuates in neural stem cells of the hippocampal dentate gyrus, depending on age and brain disorders. Neural stem cells increase their lysosomal activity during hippocampal development in the dentate gyrus, but aging and aging-related disease reduce lysosomal activity. In addition, physical exercise increases lysosomal activity in neural stem cells and astrocytes in the dentate gyrus. We therefore propose that three different stages of lysosomal activity exist: the state of increase during development, the stable state during adulthood and the state of reduction due to damage caused by either age or disease.
Collapse
Affiliation(s)
- He Zhang
- Graduate School of Biostudies, Kyoto University, Kyoto 606-8315, Japan
| | - Karan Ishii
- Graduate School of Biostudies, Kyoto University, Kyoto 606-8315, Japan
| | - Tatsuya Shibata
- Graduate School of Biostudies, Kyoto University, Kyoto 606-8315, Japan
| | - Shunsuke Ishii
- Graduate School of Science, Chiba University, Chiba 263-8522, Japan
| | - Marika Hirao
- Graduate School of Biostudies, Kyoto University, Kyoto 606-8315, Japan
| | - Zhou Lu
- Graduate School of Biostudies, Kyoto University, Kyoto 606-8315, Japan
| | - Risa Takamura
- Graduate School of Biostudies, Kyoto University, Kyoto 606-8315, Japan
| | - Satsuki Kitano
- Institute for Life and Medical Sciences, Kyoto University, Kyoto 606-8507, Japan
| | - Hitoshi Miyachi
- Institute for Life and Medical Sciences, Kyoto University, Kyoto 606-8507, Japan
| | | | - Eisuke Itakura
- Graduate School of Science, Chiba University, Chiba 263-8522, Japan
| | - Taeko Kobayashi
- Graduate School of Biostudies, Kyoto University, Kyoto 606-8315, Japan
- The Institute of Medical Science, The University of Tokyo, Tokyo 108-8639, Japan
| |
Collapse
|
7
|
Pastor-Alonso O, Syeda Zahra A, Kaske B, García-Moreno F, Tetzlaff F, Bockelmann E, Grunwald V, Martín-Suárez S, Riecken K, Witte OW, Encinas JM, Urbach A. Generation of adult hippocampal neural stem cells occurs in the early postnatal dentate gyrus and depends on cyclin D2. EMBO J 2024; 43:317-338. [PMID: 38177500 PMCID: PMC10897295 DOI: 10.1038/s44318-023-00011-2] [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: 01/19/2023] [Revised: 11/03/2023] [Accepted: 11/20/2023] [Indexed: 01/06/2024] Open
Abstract
Lifelong hippocampal neurogenesis is maintained by a pool of multipotent adult neural stem cells (aNSCs) residing in the subgranular zone of the dentate gyrus (DG). The mechanisms guiding transition of NSCs from the developmental to the adult state remain unclear. We show here, by using nestin-based reporter mice deficient for cyclin D2, that the aNSC pool is established through cyclin D2-dependent proliferation during the first two weeks of life. The absence of cyclin D2 does not affect normal development of the dentate gyrus until birth but prevents postnatal formation of radial glia-like aNSCs. Furthermore, retroviral fate mapping reveals that aNSCs are born on-site from precursors located in the dentate gyrus shortly after birth. Taken together, our data identify the critical time window and the spatial location of the precursor divisions that generate the persistent population of aNSCs and demonstrate the central role of cyclin D2 in this process.
Collapse
Affiliation(s)
- Oier Pastor-Alonso
- Laboratory of Neural Stem Cells and Neurogenesis, Achucarro Basque Center for Neuroscience, Scientific Park, 48940, Leioa, Bizkaia, Spain
- Department of Neurology, University of California San Francisco, San Francisco, CA, 94143, USA
| | - Anum Syeda Zahra
- Department of Neurology, Jena University Hospital, 07747, Jena, Germany
| | - Bente Kaske
- Department of Neurology, Jena University Hospital, 07747, Jena, Germany
| | - Fernando García-Moreno
- Laboratory of Neural Stem Cells and Neurogenesis, Achucarro Basque Center for Neuroscience, Scientific Park, 48940, Leioa, Bizkaia, Spain
- IKERBASQUE, The Basque Foundation for Science, Plaza Euskadi 5, 48009, Bilbo, Bizkaia, Spain
- Department of Neurosciences, University of the Basque Country (UPV/EHU), Scientific Park, 48940, Leioa, Bizkaia, Spain
| | - Felix Tetzlaff
- Department of Neurology, Jena University Hospital, 07747, Jena, Germany
| | - Enno Bockelmann
- Department of Neurology, Jena University Hospital, 07747, Jena, Germany
| | - Vanessa Grunwald
- Department of Neurology, Jena University Hospital, 07747, Jena, Germany
| | - Soraya Martín-Suárez
- Laboratory of Neural Stem Cells and Neurogenesis, Achucarro Basque Center for Neuroscience, Scientific Park, 48940, Leioa, Bizkaia, Spain
| | - Kristoffer Riecken
- Research Department Cell and Gene Therapy, Department of Stem Cell Transplantation, University Medical Center Hamburg-Eppendorf, 20246, Hamburg, Germany
| | - Otto Wilhelm Witte
- Department of Neurology, Jena University Hospital, 07747, Jena, Germany
- Jena Centre for Healthy Aging, Jena University Hospital, 07747, Jena, Germany
| | - Juan Manuel Encinas
- Laboratory of Neural Stem Cells and Neurogenesis, Achucarro Basque Center for Neuroscience, Scientific Park, 48940, Leioa, Bizkaia, Spain.
- IKERBASQUE, The Basque Foundation for Science, Plaza Euskadi 5, 48009, Bilbo, Bizkaia, Spain.
- Department of Neurosciences, University of the Basque Country (UPV/EHU), Scientific Park, 48940, Leioa, Bizkaia, Spain.
| | - Anja Urbach
- Department of Neurology, Jena University Hospital, 07747, Jena, Germany.
- Jena Centre for Healthy Aging, Jena University Hospital, 07747, Jena, Germany.
| |
Collapse
|
8
|
Zvozilova A, Reichova A, Mach M, Bakos J, Koprdova R. Effect of a New Substance with Pyridoindole Structure on Adult Neurogenesis, Shape of Neurons, and Behavioral Outcomes in a Chronic Mild Stress Model in Rats. Int J Mol Sci 2024; 25:845. [PMID: 38255918 PMCID: PMC10815319 DOI: 10.3390/ijms25020845] [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: 12/01/2023] [Revised: 01/07/2024] [Accepted: 01/08/2024] [Indexed: 01/24/2024] Open
Abstract
Despite an accumulating number of studies, treatments for depression are currently insufficient. Therefore, the search for new substances with antidepressant potential is very important. In this study, we hypothesized that treatment with a newly synthesized pyridoindole derivative compound SMe1EC2M3 would result in protective and antidepressant-like effects on behavioral outcomes and reverse the impaired adult hippocampal neurogenesis caused by chronic mild stress (CMS). We found that chronic administration of 5 mg/kg and 25 mg/kg SMe1EC2M3 to adult Sprague Dawley rats ameliorated the consequences of CMS on immobility and swimming time in a forced swim test. A slight sedative effect of the highest dose of SMe1EC2M3 in the nonstress group was observed in the open field. SMe1EC2M3 in the highest dose ameliorated CMS-induced decreases in the sucrose preference test. Administration of SMe1EC2M3 significantly increased SOX2-positive cells in the hippocampal dentate gyrus (DG) in CMS compared to control animals. A significant reduction in glial fibrillary acid protein (GFAP)-positive cells in the DG of CMS compared to control animals was observed. Administration of both 5 and 25 mg/kg SMe1EC2M3 significantly increased signal of GFAP-positive cells in the DG of CMS animals. No such effects of SMe1EC2M3 were observed in the cornu ammonis hippocampal area. Additionally, we found that incubation of primary hippocampal neurons in the presence of 1.50 µM SMe1EC2M3 significantly stimulated the length of neurites. Overall, we found that the negative effects of CMS on depression-like behavior are partially reduced by the administration of SMe1EC2M3 and are associated with changes in hippocampal neurogenesis and neuronal differentiation. SMe1EC2M3 represents a potential drug candidate with positive neuroplastic effects and neurogenesis-associated effects in therapeutic approaches to depression.
Collapse
Affiliation(s)
- Alexandra Zvozilova
- Institute of Experimental Pharmacology and Toxicology, Centre of Experimental Medicine, Slovak Academy of Sciences, 841 04 Bratislava, Slovakia; (A.Z.); (R.K.)
| | - Alexandra Reichova
- Institute of Experimental Endocrinology, Biomedical Research Center, Slovak Academy of Sciences, 845 05 Bratislava, Slovakia;
| | - Mojmir Mach
- Institute of Experimental Pharmacology and Toxicology, Centre of Experimental Medicine, Slovak Academy of Sciences, 841 04 Bratislava, Slovakia; (A.Z.); (R.K.)
| | - Jan Bakos
- Institute of Experimental Endocrinology, Biomedical Research Center, Slovak Academy of Sciences, 845 05 Bratislava, Slovakia;
- Institute of Physiology, Faculty of Medicine, Comenius University, 811 08 Bratislava, Slovakia
| | - Romana Koprdova
- Institute of Experimental Pharmacology and Toxicology, Centre of Experimental Medicine, Slovak Academy of Sciences, 841 04 Bratislava, Slovakia; (A.Z.); (R.K.)
| |
Collapse
|
9
|
Ohyama K, Shinohara HM, Omura S, Kawachi T, Sato T, Toda K. PSmad3+/Olig2- expression defines a subpopulation of gfap-GFP+/Sox9+ neural progenitors and radial glia-like cells in mouse dentate gyrus through embryonic and postnatal development. Front Neurosci 2023; 17:1204012. [PMID: 37795190 PMCID: PMC10547214 DOI: 10.3389/fnins.2023.1204012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Accepted: 08/29/2023] [Indexed: 10/06/2023] Open
Abstract
In mouse dentate gyrus, radial glia-like cells (RGLs) persist throughout life and play a critical role in the generation of granule neurons. A large body of evidence has shown that the combinatorial expression of transcription factors (TFs) defines cell types in the developing central nervous system (CNS). As yet, the identification of specific TFs that exclusively define RGLs in the developing mouse dentate gyrus (DG) remains elusive. Here we show that phospho-Smad3 (PSmad3) is expressed in a subpopulation of neural progenitors in the DG. During embryonic stage (E14-15), PSmad3 was predominantly expressed in gfap-GFP-positive (GFP+)/Sox2+ progenitors located at the lower dentate notch (LDN). As the development proceeds (E16-17), the vast majority of PSmad3+ cells were GFP+/Sox2+/Prox1low+/Ki67+ proliferative progenitors that eventually differentiated into granule neurons. During postnatal stage (P1-P6) PSmad3 expression was observed in GFP+ progenitors and astrocytes. Subsequently, at P14-P60, PSmad3 expression was found both in GFP+ RGLs in the subgranular zone (SGZ) and astrocytes in the molecular layer (ML) and hilus. Notably, PSmad3+ SGZ cells did not express proliferation markers such as PCNA and phospho-vimentin, suggesting that they are predominantly quiescent from P14 onwards. Significantly PSmad3+/GFP+ astrocytes, but not SGZ cells, co-expressed Olig2 and S100β. Together, PSmad3+/Olig2- expression serves as an exclusive marker for a specific subpopulation of GFP+ neural progenitors and RGLs in the mouse DG during both embryonic and postnatal period.
Collapse
Affiliation(s)
- Kyoji Ohyama
- Department of Histology and Neuroanatomy, Tokyo Medical University, Tokyo, Japan
| | | | | | | | | | | |
Collapse
|
10
|
Sato RY, Kotake K, Zhang Y, Onishi H, Matsui F, Norimoto H, Zhou Z. Methyl vinyl ketone impairs spatial memory and activates hippocampal glial cells in mice. PLoS One 2023; 18:e0289714. [PMID: 37651419 PMCID: PMC10470879 DOI: 10.1371/journal.pone.0289714] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2023] [Accepted: 07/21/2023] [Indexed: 09/02/2023] Open
Abstract
Memory is a fundamental brain function that can be affected by a variety of external factors including environmental pollutants. One of these pollutants is methyl vinyl ketone (MVK), a hazardous substance found in cigarettes, industrial wastes, and car exhaust. Humans can be exposed to MVK under many circumstances; however, it is unclear whether MVK affects higher-order brain functions such as memory. Here, we examined the memory performances of mice receiving systemic MVK administration. We found that 1 mg/kg of MVK impaired spatial memory. We also showed that 1 mg/kg MVK activated glial cells and altered glial functions in several subregions of the hippocampus, a brain region involved in learning and memory. These results suggest that MVK induces memory deficits and activates glial cells in hippocampal subregions.
Collapse
Affiliation(s)
- Ren Y. Sato
- Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Koki Kotake
- Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Yumin Zhang
- Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Hiraku Onishi
- Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Futaba Matsui
- Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Hiroaki Norimoto
- Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Zhiwen Zhou
- Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| |
Collapse
|
11
|
Olpe C, Jessberger S. Cell population dynamics in the course of adult hippocampal neurogenesis: Remaining unknowns. Hippocampus 2023; 33:402-411. [PMID: 36256493 DOI: 10.1002/hipo.23475] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 09/26/2022] [Accepted: 09/30/2022] [Indexed: 11/08/2022]
Abstract
Neural stem cells (NSCs) generate new neurons throughout life in the mammalian hippocampus. The distinct developmental steps in the course of adult neurogenesis, including NSC activation, expansion, and neuronal integration, are increasingly well characterized down to the molecular level. However, substantial gaps remain in our knowledge about regulators and mechanisms involved in this biological process. This review highlights three long-standing unknowns. First, we discuss potency and identity of NSCs and the quest for a unifying model of short- and long-term self-renewal dynamics. Next, we examine cell death, specifically focusing on the early demise of newborn cells. Then, we outline the current knowledge on cell integration dynamics, discussing which (if any) neurons are replaced by newly added neurons in the hippocampal circuits. For each of these unknowns, we summarize the trajectory of studies leading to the current state of knowledge. Finally, we offer suggestions on how to fill the remaining gaps by taking advantage of novel technology to reveal currently hidden secrets in the course of adult hippocampal neurogenesis.
Collapse
Affiliation(s)
- Cora Olpe
- Brain Research Institute, Faculties of Medicine and Science, University of Zurich, Zurich, Switzerland
| | - Sebastian Jessberger
- Brain Research Institute, Faculties of Medicine and Science, University of Zurich, Zurich, Switzerland
| |
Collapse
|
12
|
Fung CM. Effects of intrauterine growth restriction on embryonic hippocampal dentate gyrus neurogenesis and postnatal critical period of synaptic plasticity that govern learning and memory function. Front Neurosci 2023; 17:1092357. [PMID: 37008232 PMCID: PMC10064986 DOI: 10.3389/fnins.2023.1092357] [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: 11/08/2022] [Accepted: 02/28/2023] [Indexed: 03/18/2023] Open
Abstract
Intrauterine growth restriction (IUGR) complicates up to 10% of human pregnancies and is the second leading cause of perinatal morbidity and mortality after prematurity. The most common etiology of IUGR in developed countries is uteroplacental insufficiency (UPI). For survivors of IUGR pregnancies, long-term studies consistently show a fivefold increased risk for impaired cognition including learning and memory deficits. Among these, only a few human studies have highlighted sex differences with males and females having differing susceptibilities to different impairments. Moreover, it is well established from brain magnetic resonance imaging that IUGR affects both white and gray matter. The hippocampus, composed of the dentate gyrus (DG) and cornu ammonis (CA) subregions, is an important gray matter structure critical to learning and memory, and is particularly vulnerable to the chronic hypoxic-ischemic effects of UPI. Decreased hippocampal volume is a strong predictor for learning and memory deficits. Decreased neuron number and attenuated dendritic and axonal morphologies in both the DG and CA are additionally seen in animal models. What is largely unexplored is the prenatal changes that predispose an IUGR offspring to postnatal learning and memory deficits. This lack of knowledge will continue to hinder the design of future therapy to improve learning and memory. In this review, we will first present the clinical susceptibilities and human epidemiology data regarding the neurological sequelae after IUGR. We will follow with data generated using our laboratory's mouse model of IUGR, that mimics the human IUGR phenotype, to dissect at the cellular and molecular alterations in embryonic hippocampal DG neurogenesis. We will lastly present a newer topic of postnatal neuron development, namely the critical period of synaptic plasticity that is crucial in achieving an excitatory/inhibitory balance in the developing brain. To our knowledge, these findings are the first to describe the prenatal changes that lead to an alteration in postnatal hippocampal excitatory/inhibitory imbalance, a mechanism that is now recognized to be a cause of neurocognitive/neuropsychiatric disorders in at-risk individuals. Studies are ongoing in our laboratory to elucidate additional mechanisms that underlie IUGR-induced learning and memory impairment and to design therapy aimed at ameliorating such impairment.
Collapse
Affiliation(s)
- Camille M. Fung
- Division of Neonatology, Department of Pediatrics, University of Utah School of Medicine, Salt Lake City, UT, United States
| |
Collapse
|
13
|
Sun D, Mei L, Xiong WC. Dorsal Dentate Gyrus, a Key Regulator for Mood and Psychiatric Disorders. Biol Psychiatry 2023:S0006-3223(23)00009-4. [PMID: 36894487 DOI: 10.1016/j.biopsych.2023.01.005] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Revised: 12/06/2022] [Accepted: 01/09/2023] [Indexed: 01/18/2023]
Abstract
The dentate gyrus, a "gate" that controls the flow of information into the hippocampus, is critical for learning, memory, spatial navigation, and mood regulation. Several lines of evidence have demonstrated that deficits in dentate granule cells (DGCs) (e.g., loss of DGCs or genetic mutations in DGCs) contribute to the development of various psychiatric disorders, such as depression and anxiety disorders. Whereas ventral DGCs are believed to be critical for mood regulation, the functions of dorsal DGCs in this regard remain elusive. Here, we review the role of DGCs, in particular the dorsal DGCs, in the regulation of mood, their functional relationships with DGC development, and the contributions of dysfunctional DGCs to mental disorders.
Collapse
Affiliation(s)
- Dong Sun
- Department of Neurosciences, School of Medicine, Case Western Reserve University, Cleveland, Ohio; National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, Jilin, China
| | - Lin Mei
- Department of Neurosciences, School of Medicine, Case Western Reserve University, Cleveland, Ohio
| | - Wen-Cheng Xiong
- Department of Neurosciences, School of Medicine, Case Western Reserve University, Cleveland, Ohio.
| |
Collapse
|
14
|
Tartt AN, Mariani MB, Hen R, Mann JJ, Boldrini M. Dysregulation of adult hippocampal neuroplasticity in major depression: pathogenesis and therapeutic implications. Mol Psychiatry 2022; 27:2689-2699. [PMID: 35354926 PMCID: PMC9167750 DOI: 10.1038/s41380-022-01520-y] [Citation(s) in RCA: 148] [Impact Index Per Article: 49.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Revised: 02/22/2022] [Accepted: 03/09/2022] [Indexed: 02/07/2023]
Abstract
Major depressive disorder (MDD) was previously hypothesized to be a disease of monoamine deficiency in which low levels of monoamines in the synaptic cleft were believed to underlie depressive symptoms. More recently, however, there has been a paradigm shift toward a neuroplasticity hypothesis of depression in which downstream effects of antidepressants, such as increased neurogenesis, contribute to improvements in cognition and mood. This review takes a top-down approach to assess how changes in behavior and hippocampal-dependent circuits may be attributed to abnormalities at the molecular, structural, and synaptic level. We conclude with a discussion of how antidepressant treatments share a common effect in modulating neuroplasticity and consider outstanding questions and future perspectives.
Collapse
Affiliation(s)
| | | | - Rene Hen
- Departments of Psychiatry, Columbia University, New York, NY, USA
- Neuroscience, Columbia University, New York, NY, USA
- Pharmacology, Columbia University, New York, NY, USA
- Integrative Neuroscience, NYS Psychiatric Institute, New York, NY, USA
| | - J John Mann
- Departments of Psychiatry, Columbia University, New York, NY, USA
- Molecular Imaging and Neuropathology, NYS Psychiatric Institute, New York, NY, USA
| | - Maura Boldrini
- Departments of Psychiatry, Columbia University, New York, NY, USA.
- Molecular Imaging and Neuropathology, NYS Psychiatric Institute, New York, NY, USA.
| |
Collapse
|
15
|
Quadros ARAA, Arazola RD, Álvarez AR, Pires J, Meredith RM, Saarloos I, Verhage M, Toonen RF. Neuronal F-Box protein FBXO41 regulates synaptic transmission and hippocampal network maturation. iScience 2022; 25:104069. [PMID: 35372812 PMCID: PMC8971942 DOI: 10.1016/j.isci.2022.104069] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Revised: 02/16/2022] [Accepted: 03/10/2022] [Indexed: 01/04/2023] Open
Abstract
FBXO41 is a neuron-specific E3 ligase subunit implicated in epileptic encephalopathies. Fbxo41 null mutant (KO) mice show behavioral deficits and early lethality. Here, we report that loss of FBXO41 causes defects in synaptic transmission and brain development. Cultured Fbxo41 KO neurons had normal morphology and showed no signs of degeneration. Single-cell electrophysiology showed a lower synaptic vesicle release probability in excitatory neurons. Inhibitory neurons exhibited reduced synaptophysin expression, a smaller readily releasable pool, and decreased charge transfer during repetitive stimulation. In Fbxo41 KO hippocampal slices at postnatal day 6, the dentate gyrus was smaller with fewer radial-glial-like cells and immature neurons. In addition, neuronal migration was delayed. Two-photon calcium imaging showed a delayed increase in network activity and synchronicity. Together, our findings point toward a role for FBXO41 in synaptic transmission and postnatal brain development, before behavioral deficits are detected in Fbxo41 KO mice.
Collapse
Affiliation(s)
- Ana R A A Quadros
- Department of Functional Genomics, Vrije Universiteit (VU) Amsterdam, de Boelelaan 1085, 1081 HV Amsterdam, The Netherlands
| | - Rocío Díez Arazola
- Department of Functional Genomics, Vrije Universiteit (VU) Amsterdam, de Boelelaan 1085, 1081 HV Amsterdam, The Netherlands
| | - Andrea Romaguera Álvarez
- Department of Functional Genomics, Vrije Universiteit (VU) Amsterdam, de Boelelaan 1085, 1081 HV Amsterdam, The Netherlands
| | - Johny Pires
- Department of Integrative Neurophysiology, Vrije Universiteit (VU) Amsterdam, de Boelelaan 1085, 1081 HV Amsterdam, The Netherlands
| | - Rhiannon M Meredith
- Department of Integrative Neurophysiology, Vrije Universiteit (VU) Amsterdam, de Boelelaan 1085, 1081 HV Amsterdam, The Netherlands
| | - Ingrid Saarloos
- Department of Functional Genomics, Vrije Universiteit (VU) Amsterdam, de Boelelaan 1085, 1081 HV Amsterdam, The Netherlands.,Department of Clinical Genetics, Center for Neurogenomics and Cognitive Research (CNCR), Vrije Universiteit (VU) Amsterdam, de Boelelaan 1085, 1081 HV Amsterdam, The Netherlands
| | - Matthijs Verhage
- Department of Functional Genomics, Vrije Universiteit (VU) Amsterdam, de Boelelaan 1085, 1081 HV Amsterdam, The Netherlands.,Department of Clinical Genetics, Center for Neurogenomics and Cognitive Research (CNCR), Vrije Universiteit (VU) Amsterdam, de Boelelaan 1085, 1081 HV Amsterdam, The Netherlands
| | - Ruud F Toonen
- Department of Functional Genomics, Vrije Universiteit (VU) Amsterdam, de Boelelaan 1085, 1081 HV Amsterdam, The Netherlands
| |
Collapse
|
16
|
Early Life Events and Maturation of the Dentate Gyrus: Implications for Neurons and Glial Cells. Int J Mol Sci 2022; 23:ijms23084261. [PMID: 35457079 PMCID: PMC9031216 DOI: 10.3390/ijms23084261] [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: 02/27/2022] [Revised: 04/08/2022] [Accepted: 04/11/2022] [Indexed: 12/15/2022] Open
Abstract
The dentate gyrus (DG), an important part of the hippocampus, plays a significant role in learning, memory, and emotional behavior. Factors potentially influencing normal development of neurons and glial cells in the DG during its maturation can exert long-lasting effects on brain functions. Early life stress may modify maturation of the DG and induce lifelong alterations in its structure and functioning, underlying brain pathologies in adults. In this paper, maturation of neurons and glial cells (microglia and astrocytes) and the effects of early life events on maturation processes in the DG have been comprehensively reviewed. Early postnatal interventions affecting the DG eventually result in an altered number of granule neurons in the DG, ectopic location of neurons and changes in adult neurogenesis. Adverse events in early life provoke proinflammatory changes in hippocampal glia at cellular and molecular levels immediately after stress exposure. Later, the cellular changes may disappear, though alterations in gene expression pattern persist. Additional stressful events later in life contribute to manifestation of glial changes and behavioral deficits. Alterations in the maturation of neuronal and glial cells induced by early life stress are interdependent and influence the development of neural nets, thus predisposing the brain to the development of cognitive and psychiatric disorders.
Collapse
|
17
|
Intrauterine Growth Restriction Causes Abnormal Embryonic Dentate Gyrus Neurogenesis in Mouse Offspring That Leads to Adult Learning and Memory Deficits. eNeuro 2021; 8:ENEURO.0062-21.2021. [PMID: 34544755 PMCID: PMC8503959 DOI: 10.1523/eneuro.0062-21.2021] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2021] [Revised: 09/08/2021] [Accepted: 09/14/2021] [Indexed: 11/25/2022] Open
Abstract
Human infants who suffer from intrauterine growth restriction (IUGR), which is a failure to attain their genetically predetermined weight, are at increased risk for postnatal learning and memory deficits. Hippocampal dentate gyrus (DG) granule neurons play an important role in memory formation; however, it is unknown whether IUGR affects embryonic DG neurogenesis, which could provide a potential mechanism underlying abnormal postnatal learning and memory function. Using a mouse model of the most common cause of IUGR, induced by hypertensive disease of pregnancy, we first assessed adult learning and memory function. We quantified the percentages of embryonic hippocampal DG neural stem cells (NSCs) and progenitor cells and developing glutamatergic granule neurons, as well as hippocampal volumes and neuron cell count and morphology 18 and 40 d after delivery. We characterized the differential embryonic hippocampal transcriptomic pathways between appropriately grown and IUGR mouse offspring. We found that IUGR offspring of both sexes had short-term adult learning and memory deficits. Prenatally, we found that IUGR caused accelerated embryonic DG neurogenesis and Sox2+ neural stem cell depletion. IUGR mice were marked by decreased hippocampal volumes and decreased doublecortin+ neuronal progenitors with increased mean dendritic lengths at postnatal day 18. Consistent with its known molecular role in embryonic DG neurogenesis, we also found evidence for decreased Wnt pathway activity during IUGR. In conclusion, we have discovered that postnatal memory deficits are associated with accelerated NSC differentiation and maturation into glutamatergic granule neurons following IUGR, a phenotype that could be explained by decreased embryonic Wnt signaling.
Collapse
|
18
|
Zhou J, Liu G, Zhang X, Wu C, Ma M, Wu J, Hou L, Yin B, Qiang B, Shu P, Peng X. Comparison of the Spatiotemporal Expression Patterns of Three Cre Lines, Emx1IRES-Cre, D6-Cre and hGFAP-Cre, Commonly Used in Neocortical Development Research. Cereb Cortex 2021; 32:1668-1681. [PMID: 34550336 DOI: 10.1093/cercor/bhab305] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Revised: 08/01/2021] [Accepted: 08/02/2021] [Indexed: 11/14/2022] Open
Abstract
Emx1IRES-Cre, D6-Cre and hGFAP-Cre are commonly used to conditionally manipulate gene expression or lineage tracing because of their specificity in the dorsal telencephalon during early neurogenesis as previously described. However, the spatiotemporal differences in Cre recombinase activity would lead to divergent phenotypes. Here, we compared the patterns of Cre activity in the early embryos among the three lines by mating with reporter mice. The activities of Emx1IRES-Cre, D6-Cre and hGFAP-Cre were observed in the dorsal telencephalon, starting from approximately embryonic day 9.5, 11.5 and 12.5, respectively. Although all the three lines have activity in radial glial cells, Emx1IRES-Cre fully covers the dorsal and medial telencephalon, including the archicortex and cortical hem. D6-Cre is highly restricted to the dorsal telencephalon with anterior-low to posterior-high gradients, partially covers the hippocampus, and absent in the cortical hem. Moreover, both Emx1IRES-Cre and hGFAP-Cre exhibit Cre activity outside the dorsal neocortex. Meanwhile, we used the three Cre lines to mediate Dicer knockout and observed inconsistent phenotypes, including discrepancies in radial glial cell number, survival and neurogenesis in the neocortex and hippocampus. Together we proved differences in Cre activity can perturb the resultant phenotypes, which aid researchers in appropriate experimental design.
Collapse
Affiliation(s)
- Jiafeng Zhou
- The State Key Laboratory of Medical Molecular Biology, Neuroscience Center, Medical Primate Research Center and Department of Molecular Biology and Biochemistry, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, 100005, China
| | - Gaoao Liu
- The State Key Laboratory of Medical Molecular Biology, Neuroscience Center, Medical Primate Research Center and Department of Molecular Biology and Biochemistry, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, 100005, China
| | - Xiaoling Zhang
- The State Key Laboratory of Medical Molecular Biology, Neuroscience Center, Medical Primate Research Center and Department of Molecular Biology and Biochemistry, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, 100005, China
| | - Chao Wu
- The State Key Laboratory of Medical Molecular Biology, Neuroscience Center, Medical Primate Research Center and Department of Molecular Biology and Biochemistry, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, 100005, China
| | - Mengjie Ma
- The State Key Laboratory of Medical Molecular Biology, Neuroscience Center, Medical Primate Research Center and Department of Molecular Biology and Biochemistry, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, 100005, China
| | - Jiarui Wu
- The State Key Laboratory of Medical Molecular Biology, Neuroscience Center, Medical Primate Research Center and Department of Molecular Biology and Biochemistry, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, 100005, China
| | - Lin Hou
- The State Key Laboratory of Medical Molecular Biology, Neuroscience Center, Medical Primate Research Center and Department of Molecular Biology and Biochemistry, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, 100005, China
| | - Bin Yin
- The State Key Laboratory of Medical Molecular Biology, Neuroscience Center, Medical Primate Research Center and Department of Molecular Biology and Biochemistry, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, 100005, China
| | - Boqin Qiang
- The State Key Laboratory of Medical Molecular Biology, Neuroscience Center, Medical Primate Research Center and Department of Molecular Biology and Biochemistry, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, 100005, China
| | - Pengcheng Shu
- The State Key Laboratory of Medical Molecular Biology, Neuroscience Center, Medical Primate Research Center and Department of Molecular Biology and Biochemistry, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, 100005, China.,Chinese Institute for Brain Research, Beijing, 102206, China
| | - Xiaozhong Peng
- The State Key Laboratory of Medical Molecular Biology, Neuroscience Center, Medical Primate Research Center and Department of Molecular Biology and Biochemistry, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, 100005, China.,Institute of Medical Biology of the Chinese Academy of Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming, 650118, China
| |
Collapse
|
19
|
Wang YC, Liu P, Yue LY, Huang F, Xu YX, Zhu CQ. NRSF deficiency leads to abnormal postnatal development of dentate gyrus and impairment of progenitors in subgranular zone of hippocampus. Hippocampus 2021; 31:935-956. [PMID: 33960056 DOI: 10.1002/hipo.23336] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Revised: 04/19/2021] [Accepted: 04/25/2021] [Indexed: 02/05/2023]
Abstract
Neuron-restrictive silencing factor (NRSF) is a zinc-finger transcription factor that regulates expression of a diverse set of genes. However, NRSF function in brain development still remains elusive. In the present study, we generated NRSF-conditional knockout (NRSF-cKO) mice by hGFAP-Cre/loxp system to study the effect of NRSF deficiency on brain development. Results showed that NRSF conditional knockout caused a smaller hippocampus and a thinner granule cell layer (GCL) in mice. Moreover, the reduction and disarrangement of GFAP+ cells in subgranular zone (SGZ) of NRSF-cKO mice was accompanied with the decreased number of premature neurons, neural stem cells (NSCs) and neural progenitor cells (NPCs), as well as compromising the majority of mitotically active cells. The analysis of postnatal development of hippocampus indicated the existence of an abnormality at postnatal day (P) 8, rather than at P1, in NRSF-cKO mice, although the densities of Ki67+ cells with mitotic ability in dentate gyrus were relatively unaffected at P1 and P8. Meanwhile, NRSF deficiency led to abnormal organization of SGZ at P8 during postnatal development. RNA-Seq analysis revealed 79 deregulated genes in hippocampus of NRSF-cKO mice at P8, which were involved in p53 signal transduction, neuron migration and negative regulation of cell proliferation, etc. The deregulation of p53 pathway in NRSF-cKO mice at P1 and P8 was evidenced, of which p21/Cdkn1a was accumulated in a portion of NSCs and NPCs in hippocampus during postnatal development. Together, these results, for the first time, revealed that NRSF could significantly influence the postnatal development of hippocampus, especially the formation of SGZ.
Collapse
Affiliation(s)
- Yan-Cong Wang
- Department of Translational Neuroscience, Jing'an District Center Hospital of Shanghai, State Key Laboratory of Medical Neurobiology and MOE Frontier Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, China
| | - Pu Liu
- Department of Translational Neuroscience, Jing'an District Center Hospital of Shanghai, State Key Laboratory of Medical Neurobiology and MOE Frontier Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, China
| | - Ling-Yun Yue
- Department of Translational Neuroscience, Jing'an District Center Hospital of Shanghai, State Key Laboratory of Medical Neurobiology and MOE Frontier Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, China
| | - Fang Huang
- Department of Translational Neuroscience, Jing'an District Center Hospital of Shanghai, State Key Laboratory of Medical Neurobiology and MOE Frontier Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, China
| | - Yu-Xia Xu
- Department of Translational Neuroscience, Jing'an District Center Hospital of Shanghai, State Key Laboratory of Medical Neurobiology and MOE Frontier Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, China
| | - Cui-Qing Zhu
- Department of Translational Neuroscience, Jing'an District Center Hospital of Shanghai, State Key Laboratory of Medical Neurobiology and MOE Frontier Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, China
| |
Collapse
|
20
|
Gómez-Gálvez Y, Gates MA. Paclitaxel is effective for controlling astrocyte proliferation in vitro: Implications for generating ventral mesencephalic cultures enriched with dopamine neurons. J Neurosci Methods 2020; 351:109065. [PMID: 33387573 DOI: 10.1016/j.jneumeth.2020.109065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Revised: 12/23/2020] [Accepted: 12/27/2020] [Indexed: 11/26/2022]
Abstract
BACKGROUND Primary embryonic ventral mesencephalic (VM) cultures are a high throughput tool for understanding and manipulating dopamine neurons, to study the mechanisms that trigger their degeneration during Parkinson's disease (PD), and to test new drugs aimed at treating the disease. Unfortunately, primary cell cultures are often quickly overwhelmed by dividing astrocytes which both obscure neuronal cells and distort the cellular composition that exists in vivo. NEW METHOD To develop a new in vitro system whereby astrocyte division can be readily controlled while maintaining neuronal integrity, VM cultures were treated with different doses (1.75, 3.5, 7, 14 nM) of the anti-mitotic drug paclitaxel for up to seven days in vitro. The study subsequently sought to determine the importance of astrocytes in dopamine neuron survival when challenged with an exposure to the toxin 6-hydroxydopamine (6-OHDA). RESULTS Optical density (O.D.) measures of GFAP expression and counts of β-III tubulin and tyrosine hydroxylase positive neurons reveals that a low dose of 3.5 nM of paclitaxel significantly reduced the density of GFAP + astrocytes in primary VM cultures, while maintaining the viability of neurons and dopamine neurons. Interestingly, a reduction of GFAP + astrocytes within primary VM cultures did not reveal any statistically significant differences in the number of dopamine neurons surviving treatment with 6-OHDA. CONCLUSIONS These findings detail a quick and simple method for stabilising astrocyte numbers in primary VM cultures, without affecting the viability of dopamine neurons, and suggest that astrocytes may not enhance the survival of dopamine neurons when challenged with the 6-OHDA toxin.
Collapse
Affiliation(s)
- Yolanda Gómez-Gálvez
- School of Pharmacy and Bioengineering, Keele University, Staffordshire, UK; School of Life Sciences, Keele University, Staffordshire, UK; School of Medicine, Keele University, Staffordshire, UK
| | - Monte A Gates
- School of Pharmacy and Bioengineering, Keele University, Staffordshire, UK; School of Medicine, Keele University, Staffordshire, UK.
| |
Collapse
|
21
|
Abstract
Neural stem cells (NSCs) persist into adulthood in the subgranular zone (SGZ) of the dentate gyrus in the hippocampus and in the ventricular-subventricular zone (V-SVZ) of the lateral ventricles, where they generate new neurons and glia cells that contribute to neural plasticity. A better understanding of the developmental process that enables NSCs to persist beyond development will provide insight into factors that determine the size and properties of the adult NSC pool and thus the capacity for life-long neurogenesis in the adult mammalian brain. We review current knowledge regarding the developmental origins of adult NSCs and the developmental process by which embryonic NSCs transition into their adult form. We also discuss potential mechanisms that might regulate proper establishment of the adult NSC pool, and propose future directions of research that will be key to unraveling how NSCs transform to establish the adult NSC pool in the mammalian brain.
Collapse
Affiliation(s)
- Allison M Bond
- Department of Neuroscience and Mahoney Institute for Neurosciences, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Guo-Li Ming
- Department of Neuroscience and Mahoney Institute for Neurosciences, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States; Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States; Institute for Regenerative Medicine, University of Pennsylvania, Philadelphia, PA, United States; Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Hongjun Song
- Department of Neuroscience and Mahoney Institute for Neurosciences, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States; Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States; Institute for Regenerative Medicine, University of Pennsylvania, Philadelphia, PA, United States; The Epigenetics Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States.
| |
Collapse
|
22
|
Mira H, Morante J. Neurogenesis From Embryo to Adult - Lessons From Flies and Mice. Front Cell Dev Biol 2020; 8:533. [PMID: 32695783 PMCID: PMC7339912 DOI: 10.3389/fcell.2020.00533] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2020] [Accepted: 06/08/2020] [Indexed: 12/30/2022] Open
Abstract
The human brain is composed of billions of cells, including neurons and glia, with an undetermined number of subtypes. During the embryonic and early postnatal stages, the vast majority of these cells are generated from neural progenitors and stem cells located in all regions of the neural tube. A smaller number of neurons will continue to be generated throughout our lives, in localized neurogenic zones, mainly confined at least in rodents to the subependymal zone of the lateral ventricles and the subgranular zone of the hippocampal dentate gyrus. During neurogenesis, a combination of extrinsic cues interacting with temporal and regional intrinsic programs are thought to be critical for increasing neuronal diversity, but their underlying mechanisms need further elucidation. In this review, we discuss the recent findings in Drosophila and mammals on the types of cell division and cell interactions used by neural progenitors and stem cells to sustain neurogenesis, and how they are influenced by glia.
Collapse
Affiliation(s)
- Helena Mira
- Instituto de Biomedicina de Valencia, Consejo Superior de Investigaciones Científicas, Valencia, Spain
| | - Javier Morante
- Instituto de Neurociencias, Consejo Superior de Investigaciones Científicas y Universidad Miguel Hernandez, Alicante, Spain
| |
Collapse
|
23
|
Sasaki-Takahashi N, Shinohara H, Shioda S, Seki T. The polarity and properties of radial glia-like neural stem cells are altered by seizures with status epilepticus: Study using an improved mouse pilocarpine model of epilepsy. Hippocampus 2020; 30:250-262. [PMID: 32101365 DOI: 10.1002/hipo.23153] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Revised: 08/04/2019] [Accepted: 08/19/2019] [Indexed: 01/08/2023]
Abstract
In the adult mouse hippocampus, new neurons are produced by radial glia-like (RGL) neural stem cells in the subgranular zone, which extend their apical processes toward the molecular layer, and express the astrocyte marker glial fibrillary acidic protein, but not the astrocyte marker S100β. In rodent models of epilepsy, adult hippocampal neurogenesis was reported to be increased after acute and mild seizures, but to be decreased by chronic and severe epilepsy. In the present study, we investigated how the severity of seizures affects neurogenesis and RGL neural stem cells in acute stages of epilepsy, using an improved mouse pilocarpine model in which pilocarpine-induced hypothermia was prevented by maintaining body temperature, resulting in a high incidence rate of epileptic seizures and low rate of mortality. In mice that experienced seizures without status epilepticus (SE), the number of proliferating progenitors and immature neurons were significantly increased, whereas no changes were observed in RGL cells. In mice that experienced seizures with SE, the number of proliferating progenitors and immature neurons were unchanged, but the number of RGL cells with an apical process was significantly reduced. Furthermore, the processes of the majority of RGL cells extended inversely toward the hilus, and about half of the aberrant RGL cells expressed S100β. These results suggest that seizures with SE lead to changes in the polarity and properties of RGL neural stem cells, which may direct them toward astrocyte differentiation, resulting in the reduction of neural stem cells producing new granule cells. This also suggests the possibility that cell polarity of RGL stem cells is important for maintaining the stemness of adult neural stem cells.
Collapse
Affiliation(s)
| | - Hiroshi Shinohara
- Department of Histology and Neuroanatomy, Tokyo Medical University, Tokyo, Japan
| | - Seiji Shioda
- Institute for Advanced Bioscience Research, Hoshi University, Tokyo, Japan
| | - Tatsunori Seki
- Department of Histology and Neuroanatomy, Tokyo Medical University, Tokyo, Japan
| |
Collapse
|
24
|
Kobayashi T, Piao W, Takamura T, Kori H, Miyachi H, Kitano S, Iwamoto Y, Yamada M, Imayoshi I, Shioda S, Ballabio A, Kageyama R. Enhanced lysosomal degradation maintains the quiescent state of neural stem cells. Nat Commun 2019; 10:5446. [PMID: 31784517 PMCID: PMC6884460 DOI: 10.1038/s41467-019-13203-4] [Citation(s) in RCA: 71] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2018] [Accepted: 10/28/2019] [Indexed: 01/08/2023] Open
Abstract
Quiescence is important for sustaining neural stem cells (NSCs) in the adult brain over the lifespan. Lysosomes are digestive organelles that degrade membrane receptors after they undergo endolysosomal membrane trafficking. Enlarged lysosomes are present in quiescent NSCs (qNSCs) in the subventricular zone of the mouse brain, but it remains largely unknown how lysosomal function is involved in the quiescence. Here we show that qNSCs exhibit higher lysosomal activity and degrade activated EGF receptor by endolysosomal degradation more rapidly than proliferating NSCs. Chemical inhibition of lysosomal degradation in qNSCs prevents degradation of signaling receptors resulting in exit from quiescence. Furthermore, conditional knockout of TFEB, a lysosomal master regulator, delays NSCs quiescence in vitro and increases NSC proliferation in the dentate gyrus of mice. Taken together, our results demonstrate that enhanced lysosomal degradation is an important regulator of qNSC maintenance. It remains unclear why quiescent neural stem cells (qNSCs) in the subventricular zone of the mouse brain have enlarged lysosomes. Here, authors demonstrate that qNSCs exhibit higher lysosomal activity and degrade activated EGF receptor by endolysosomal degradation more rapidly than proliferating NSCs, which prevents the NSC exit from quiescence.
Collapse
Affiliation(s)
- Taeko Kobayashi
- Institute for Frontier Life and Medical Sciences, Kyoto University, Kyoto, 606-8507, Japan. .,Graduate School of Medicine, Kyoto University, Kyoto, 606-8501, Japan. .,Graduate School of Biostudies, Kyoto University, Kyoto, 606-8501, Japan.
| | - Wenhui Piao
- Institute for Frontier Life and Medical Sciences, Kyoto University, Kyoto, 606-8507, Japan.,Graduate School of Biostudies, Kyoto University, Kyoto, 606-8501, Japan
| | - Toshiya Takamura
- Institute for Frontier Life and Medical Sciences, Kyoto University, Kyoto, 606-8507, Japan
| | - Hiroshi Kori
- Department of Complexity Science and Engineering, University of Tokyo, Tokyo, 277-8561, Japan
| | - Hitoshi Miyachi
- Institute for Frontier Life and Medical Sciences, Kyoto University, Kyoto, 606-8507, Japan
| | - Satsuki Kitano
- Institute for Frontier Life and Medical Sciences, Kyoto University, Kyoto, 606-8507, Japan
| | - Yumiko Iwamoto
- Institute for Frontier Life and Medical Sciences, Kyoto University, Kyoto, 606-8507, Japan
| | - Mayumi Yamada
- Institute for Frontier Life and Medical Sciences, Kyoto University, Kyoto, 606-8507, Japan.,Graduate School of Biostudies, Kyoto University, Kyoto, 606-8501, Japan.,Institute for Integrated Cell-Material Sciences (iCeMS), Kyoto University, Kyoto, 606-8501, Japan
| | - Itaru Imayoshi
- Institute for Frontier Life and Medical Sciences, Kyoto University, Kyoto, 606-8507, Japan.,Graduate School of Biostudies, Kyoto University, Kyoto, 606-8501, Japan.,Institute for Integrated Cell-Material Sciences (iCeMS), Kyoto University, Kyoto, 606-8501, Japan
| | - Seiji Shioda
- Peptide Drug Innovation, Global Research Center for Innovative Life Science (GRIL), Hoshi University, 2-4-41 Ebara, Shinagawa-ku, Tokyo, 142-8501, Japan
| | - Andrea Ballabio
- Telethon Institute of Genetics and Medicine, Via Campi Flegrei 34, 80078, Pozzuoli, NA, Italy
| | - Ryoichiro Kageyama
- Institute for Frontier Life and Medical Sciences, Kyoto University, Kyoto, 606-8507, Japan. .,Graduate School of Medicine, Kyoto University, Kyoto, 606-8501, Japan. .,Graduate School of Biostudies, Kyoto University, Kyoto, 606-8501, Japan. .,Institute for Integrated Cell-Material Sciences (iCeMS), Kyoto University, Kyoto, 606-8501, Japan.
| |
Collapse
|
25
|
Namba T, Shinohara H, Seki T. Non-radial tortuous migration with cell polarity alterations of newly generated granule neurons in the neonatal rat dentate gyrus. Brain Struct Funct 2019; 224:3247-3262. [PMID: 31659443 DOI: 10.1007/s00429-019-01971-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Accepted: 10/17/2019] [Indexed: 01/08/2023]
Abstract
To establish functional neuronal circuits, newborn neurons generally migrate from the ventricular germinal zones to their final positions during embryonic periods. However, most excitatory neurons of the hippocampal dentate gyrus are born postnatally in the hilus, far from the lateral ventricle. Newly generated granule neurons must then migrate to the surrounding granule cell layer (GCL), which suggests that newborn granule cells may migrate by unique cellular mechanisms. In the present study, we describe the migratory behaviors of postnatally generated granule neurons using combined retroviral labeling and time-lapse imaging analysis. Our results show that whereas half of the newly generated neurons undergo radial migration, the remainder engages in more complex migratory patterns with veering and turning movements accompanied by process formation and cell polarity alterations. These data reveal a previously unappreciated diversity of mechanisms by which granule neurons distribute throughout the GCL to contribute to hippocampal circuitry.
Collapse
Affiliation(s)
- Takashi Namba
- Department of Anatomy, Juntendo University School of Medicine, Tokyo, 113-8421, Japan.
- Integrative Bioscience and Biomedical Engineering, School of Science and Engineering, Waseda University, Tokyo, 169-8555, Japan.
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany.
| | - Hiroshi Shinohara
- Department of Histology and Neuroanatomy, Tokyo Medical University, Tokyo, 160-8421, Japan
| | - Tatsunori Seki
- Department of Anatomy, Juntendo University School of Medicine, Tokyo, 113-8421, Japan.
- Department of Histology and Neuroanatomy, Tokyo Medical University, Tokyo, 160-8421, Japan.
| |
Collapse
|
26
|
Kozareva DA, Cryan JF, Nolan YM. Born this way: Hippocampal neurogenesis across the lifespan. Aging Cell 2019; 18:e13007. [PMID: 31298475 PMCID: PMC6718573 DOI: 10.1111/acel.13007] [Citation(s) in RCA: 84] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2019] [Revised: 05/31/2019] [Accepted: 06/30/2019] [Indexed: 12/30/2022] Open
Abstract
The capability of the mammalian brain to generate new neurons through the lifespan has gained much attention for the promise of new therapeutic possibilities especially for the aging brain. One of the brain regions that maintains a neurogenesis-permissive environment is the dentate gyrus of the hippocampus. Here, new neurons are generated from a pool of multipotent neural progenitor cells to become fully functional neurons that are integrated into the brain circuitry. A growing body of evidence points to the fact that neurogenesis in the adult hippocampus is necessary for certain memory processes, and in mood regulation, while alterations in hippocampal neurogenesis have been associated with a myriad of neurological and psychiatric disorders. More recently, evidence has come to light that new neurons may differ in their vulnerability to environmental and disease-related influences depending on the time during the life course at which they are exposed. Thus, it has been the topic of intense research in recent years. In this review, we will discuss the complex process and associated functional relevance of hippocampal neurogenesis during the embryonic/postnatal period and in adulthood. We consider the implications of hippocampal neurogenesis during the developmentally critical periods of adolescence and older age. We will further consider the literature surrounding hippocampal neurogenesis and its functional role during these critical periods with a view to providing insight into the potential of harnessing neurogenesis for health and therapeutic benefit.
Collapse
Affiliation(s)
- Danka A. Kozareva
- Department of Anatomy & NeuroscienceUniversity College CorkCorkIreland
| | - John F. Cryan
- Department of Anatomy & NeuroscienceUniversity College CorkCorkIreland
- APC Microbiome IrelandUniversity College CorkCorkIreland
| | - Yvonne M. Nolan
- Department of Anatomy & NeuroscienceUniversity College CorkCorkIreland
| |
Collapse
|
27
|
Cell necrosis, intrinsic apoptosis and senescence contribute to the progression of exencephaly to anencephaly in a mice model of congenital chranioschisis. Cell Death Dis 2019; 10:721. [PMID: 31558708 PMCID: PMC6763477 DOI: 10.1038/s41419-019-1913-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2019] [Revised: 07/08/2019] [Accepted: 08/26/2019] [Indexed: 12/27/2022]
Abstract
Exencephaly/anencephaly is one of the leading causes of neonatal mortality and the most extreme open neural tube defect with no current treatments and limited mechanistic understanding. We hypothesized that exencephaly leads to a local neurodegenerative process in the brain exposed to the amniotic fluid as well as diffuse degeneration in other encephalic areas and the spinal cord. To evaluate the consequences of in utero neural tissue exposure, brain and spinal cord samples from E17 exencephalic murine fetuses (maternal intraperitoneal administration of valproic acid at E8) were analyzed and compared to controls and saline-injected shams (n = 11/group). Expression of apoptosis and senescence genes (p53, p21, p16, Rbl2, Casp3, Casp9) was determined by qRT-PCR and protein expression analyzed by western blot. Apoptosis was measured by TUNEL assay and PI/AV flow cytometry. Valproic acid at E8 induced exencephaly in 22% of fetuses. At E17 the fetuses exhibited the characteristic absence of cranial bones. The brain structures from exencephalic fetuses demonstrated a loss of layers in cortical regions and a complete loss of structural organization in the olfactory bulb, hippocampus, dental gyrus and septal cortex. E17 fetuses had reduced expression of NeuN, GFAP and Oligodendrocytes in the brain with primed microglia. Intrinsic apoptotic activation (p53, Caspase9 and 3) was upregulated and active Caspase3 localized to the layer of brain exposed to the amniotic fluid. Senescence via p21-Rbl2 was increased in the brain and in the spinal cord at the lamina I-II of the somatosensory dorsal horn. The current study characterizes CNS alterations in murine exencephaly and demonstrates that degeneration due to intrinsic apoptosis and senescence occurs in the directly exposed brain but also remotely in the spinal cord.
Collapse
|
28
|
Morales AV, Mira H. Adult Neural Stem Cells: Born to Last. Front Cell Dev Biol 2019; 7:96. [PMID: 31214589 PMCID: PMC6557982 DOI: 10.3389/fcell.2019.00096] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Accepted: 05/20/2019] [Indexed: 01/17/2023] Open
Abstract
The generation of new neurons is a lifelong process in many vertebrate species that provides an extra level of plasticity to several brain circuits. Frequently, neurogenesis in the adult brain is considered a continuation of earlier developmental processes as it relies in the persistence of neural stem cells, similar to radial glia, known as radial glia-like cells (RGLs). However, adult RGLs are not just leftovers of progenitors that remain in hidden niches in the brain after development has finished. Rather, they seem to be specified and set aside at specific times and places during embryonic and postnatal development. The adult RGLs present several cellular and molecular properties that differ from those observed in developmental radial glial cells such as an extended cell cycle length, acquisition of a quiescence state, a more restricted multipotency and distinct transcriptomic programs underlying those cellular processes. In this minireview, we will discuss the recent attempts to determine how, when and where are the adult RGLs specified.
Collapse
Affiliation(s)
- Aixa V Morales
- Instituto Cajal, Consejo Superior de Investigaciones Científicas, Madrid, Spain
| | - Helena Mira
- Instituto de Biomedicina de Valencia, Consejo Superior de Investigaciones Científicas, Valencia, Spain
| |
Collapse
|
29
|
Imura T, Kobayashi Y, Suzutani K, Ichikawa‐Tomikawa N, Chiba H. Differential expression of a stress‐regulated gene Nr4a2 characterizes early‐ and late‐born hippocampal granule cells. Hippocampus 2018; 29:539-549. [DOI: 10.1002/hipo.23045] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2018] [Revised: 10/09/2018] [Accepted: 10/17/2018] [Indexed: 12/22/2022]
Affiliation(s)
- Tetsuya Imura
- Department of Basic PathologyFukushima Medical University School of Medicine Fukushima Japan
- Department of Human PathologyKyoto Prefectural University of Medicine, Graduate School of Medical Sciences Kyoto Japan
- Department of Pathology and Applied NeurobiologyKyoto Prefectural University of Medicine, Graduate School of Medical Sciences Kyoto Japan
| | - Yasuyuki Kobayashi
- Department of Basic PathologyFukushima Medical University School of Medicine Fukushima Japan
| | - Ken Suzutani
- Department of Basic PathologyFukushima Medical University School of Medicine Fukushima Japan
| | - Naoki Ichikawa‐Tomikawa
- Department of Basic PathologyFukushima Medical University School of Medicine Fukushima Japan
| | - Hideki Chiba
- Department of Basic PathologyFukushima Medical University School of Medicine Fukushima Japan
| |
Collapse
|
30
|
Berg DA, Bond AM, Ming GL, Song H. Radial glial cells in the adult dentate gyrus: what are they and where do they come from? F1000Res 2018; 7:277. [PMID: 29568500 PMCID: PMC5840617 DOI: 10.12688/f1000research.12684.1] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/28/2018] [Indexed: 12/26/2022] Open
Abstract
Adult neurogenesis occurs in the dentate gyrus in the mammalian hippocampus. These new neurons arise from neural precursor cells named radial glia-like cells, which are situated in the subgranular zone of the dentate gyrus. Here, we review the emerging topic of precursor heterogeneity in the adult subgranular zone. We also discuss how this heterogeneity may be established during development and focus on the embryonic origin of the dentate gyrus and radial glia-like stem cells. Finally, we discuss recently developed single-cell techniques, which we believe will be critical to comprehensively investigate adult neural stem cell origin and heterogeneity.
Collapse
Affiliation(s)
- Daniel A Berg
- Department of Neuroscience and Mahoney Institute for Neurosciences, Perelman School for Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Allison M Bond
- Department of Neuroscience and Mahoney Institute for Neurosciences, Perelman School for Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Guo-Li Ming
- Department of Neuroscience and Mahoney Institute for Neurosciences, Perelman School for Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA.,Department of Cell and Developmental Biology, Perelman School for Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA.,Institute for Regenerative Medicine, Perelman School for Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Hongjun Song
- Department of Neuroscience and Mahoney Institute for Neurosciences, Perelman School for Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA.,Department of Cell and Developmental Biology, Perelman School for Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA.,Institute for Regenerative Medicine, Perelman School for Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA.,The Epigenetics Institute, Perelman School for Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| |
Collapse
|
31
|
Cataldi S, Arcuri C, Hunot S, Mecca C, Codini M, Laurenti ME, Ferri I, Loreti E, Garcia-Gil M, Traina G, Conte C, Ambesi-Impiombato FS, Beccari T, Curcio F, Albi E. Effect of Vitamin D in HN9.10e Embryonic Hippocampal Cells and in Hippocampus from MPTP-Induced Parkinson's Disease Mouse Model. Front Cell Neurosci 2018; 12:31. [PMID: 29467625 PMCID: PMC5808335 DOI: 10.3389/fncel.2018.00031] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2017] [Accepted: 01/24/2018] [Indexed: 12/14/2022] Open
Abstract
It has long been proven that neurogenesis continues in the adult brains of mammals in the dentatus gyrus of the hippocampus due to the presence of neural stem cells. Although a large number of studies have been carried out to highlight the localization of vitamin D receptor in hippocampus, the expression of vitamin D receptor in neurogenic dentatus gyrus of hippocampus in Parkinson's disease (PD) and the molecular mechanisms triggered by vitamin D underlying the production of differentiated neurons from embryonic cells remain unknown. Thus, we performed a preclinical in vivo study by inducing PD in mice with MPTP and showed a reduction of glial fibrillary acidic protein (GFAP) and vitamin D receptor in the dentatus gyrus of hippocampus. Then, we performed an in vitro study by inducing embryonic hippocampal cell differentiation with vitamin D. Interestingly, vitamin D stimulates the expression of its receptor. Vitamin D receptor is a transcription factor that probably is responsible for the upregulation of microtubule associated protein 2 and neurofilament heavy polypeptide genes. The latter increases heavy neurofilament protein expression, essential for neurofilament growth. Notably N-cadherin, implicated in activity for dendritic outgrowth, is upregulated by vitamin D.
Collapse
Affiliation(s)
- Samuela Cataldi
- Department of Pharmaceutical Sciences, University of Perugia, Perugia, Italy
| | - Cataldo Arcuri
- Department of Experimental Medicine, University of Perugia, Perugia, Italy
| | - Stéphane Hunot
- Institut du Cerveau et de la Moelleépinière, Inserm U 1127, CNRS UMR 7225, UPMC Univ Paris 06, UMR S 1127, Sorbonne Universités, Paris, France
| | - Carmen Mecca
- Department of Experimental Medicine, University of Perugia, Perugia, Italy
| | - Michela Codini
- Department of Pharmaceutical Sciences, University of Perugia, Perugia, Italy
| | - Maria E. Laurenti
- Division of Anatomic Pathology and Histology, Department of Experimental Medicine, School of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Ivana Ferri
- Division of Anatomic Pathology and Histology, Department of Experimental Medicine, School of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Elisabetta Loreti
- Division of Anatomic Pathology and Histology, Department of Experimental Medicine, School of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Mercedes Garcia-Gil
- Department of Biology, University of Pisa, Pisa, Italy
- Interdepartmental Research Center Nutrafood, Nutraceuticals and Food for Health, University of Pisa, Pisa, Italy
| | - Giovanna Traina
- Department of Pharmaceutical Sciences, University of Perugia, Perugia, Italy
| | - Carmela Conte
- Department of Pharmaceutical Sciences, University of Perugia, Perugia, Italy
| | | | - Tommaso Beccari
- Department of Pharmaceutical Sciences, University of Perugia, Perugia, Italy
| | | | - Elisabetta Albi
- Department of Pharmaceutical Sciences, University of Perugia, Perugia, Italy
| |
Collapse
|
32
|
Ruiz-Reig N, Studer M. Rostro-Caudal and Caudo-Rostral Migrations in the Telencephalon: Going Forward or Backward? Front Neurosci 2017; 11:692. [PMID: 29311773 PMCID: PMC5742585 DOI: 10.3389/fnins.2017.00692] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2017] [Accepted: 11/23/2017] [Indexed: 11/13/2022] Open
Abstract
The generation and differentiation of an appropriate number of neurons, as well as its distribution in different parts of the brain, is crucial for the proper establishment, maintenance and plasticity of neural circuitries. Newborn neurons travel along the brain in a process known as neuronal migration, to finalize their correct position in the nervous system. Defects in neuronal migration produce abnormalities in the brain that can generate neurodevelopmental pathologies, such as autism, schizophrenia and intellectual disability. In this review, we present an overview of the developmental origin of the different telencephalic subdivisions and a description of migratory pathways taken by distinct neural populations traveling long distances before reaching their target position in the brain. In addition, we discuss some of the molecules implicated in the guidance of these migratory paths and transcription factors that contribute to the correct migration and integration of these neurons.
Collapse
|
33
|
Radic T, Frieß L, Vijikumar A, Jungenitz T, Deller T, Schwarzacher SW. Differential Postnatal Expression of Neuronal Maturation Markers in the Dentate Gyrus of Mice and Rats. Front Neuroanat 2017; 11:104. [PMID: 29184486 PMCID: PMC5694555 DOI: 10.3389/fnana.2017.00104] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2017] [Accepted: 10/30/2017] [Indexed: 11/13/2022] Open
Abstract
The dentate gyrus (DG) is a unique structure of the hippocampus that is distinguished by ongoing neurogenesis throughout the lifetime of an organism. The development of the DG, which begins during late gestation and continues during the postnatal period, comprises the structural formation of the DG as well as the establishment of the adult neurogenic niche in the subgranular zone (SGZ). We investigated the time course of postnatal maturation of the DG in male C57BL/6J mice and male Sprague-Dawley rats based on the distribution patterns of the immature neuronal marker doublecortin (DCX) and a marker for mature neurons, calbindin (CB). Our findings demonstrate that the postnatal DG is marked by a substantial maturation with a high number of DCX-positive granule cells (GCs) during the first two postnatal weeks followed by a progression toward more mature patterns and increasing numbers of CB-positive GCs within the subsequent 2 weeks. The most substantial shift in maturation of the GC population took place between P7 and P14 in both mice and rats, when young, immature DCX-positive GCs became confined to the innermost part of the GC layer (GCL), indicative of the formation of the SGZ. These results suggest that the first month of postnatal development represents an important transition phase during which DG neurogenesis and the maturation course of the GC population becomes analogous to the process of adult neurogenesis. Therefore, the postnatal DG could serve as an attractive model for studying a growing and functionally maturing neural network. Direct comparisons between mice and rats revealed that the transition from immature DCX-positive to mature CB-positive GCs occurs more rapidly in the rat by approximately 4–6 days. The remarkable species difference in the speed of maturation on the GC population level may have important implications for developmental and neurogenesis research in different rodent species and strains.
Collapse
Affiliation(s)
- Tijana Radic
- Institute of Clinical Neuroanatomy, Neuroscience Center, Goethe University, Frankfurt am Main, Germany
| | - Lara Frieß
- Institute of Clinical Neuroanatomy, Neuroscience Center, Goethe University, Frankfurt am Main, Germany
| | - Aruvi Vijikumar
- Institute of Clinical Neuroanatomy, Neuroscience Center, Goethe University, Frankfurt am Main, Germany
| | - Tassilo Jungenitz
- Institute of Clinical Neuroanatomy, Neuroscience Center, Goethe University, Frankfurt am Main, Germany
| | - Thomas Deller
- Institute of Clinical Neuroanatomy, Neuroscience Center, Goethe University, Frankfurt am Main, Germany
| | - Stephan W Schwarzacher
- Institute of Clinical Neuroanatomy, Neuroscience Center, Goethe University, Frankfurt am Main, Germany
| |
Collapse
|
34
|
Matsue K, Minakawa S, Kashiwagi T, Toda K, Sato T, Shioda S, Seki T. Dentate granule progenitor cell properties are rapidly altered soon after birth. Brain Struct Funct 2017; 223:357-369. [PMID: 28836044 DOI: 10.1007/s00429-017-1499-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Accepted: 08/08/2017] [Indexed: 12/13/2022]
Abstract
Neurogenesis occurs during the embryonic period and ceases soon after birth in the neocortex, but continues to occur in the hippocampus even in the adult. The embryonic neocortex has radial glia or progenitor cells expressing brain lipid-binding protein (BLBP), whereas the adult hippocampus has radial granule progenitor cells expressing BLBP and glial fibrillary acidic protein (GFAP) in the subgranular zone. We previously found that embryonic hippocampal granule progenitor cells express GFAP, but not BLBP, indicating that these cells are different from both embryonic neocortical and adult granule progenitor cells. In the present study, as the first step towards understanding the mechanism of persistent hippocampal neurogenesis, we aimed to determine the stage at which embryonic-type granule progenitors become adult-type progenitors using mouse Gfap-GFP transgenic mice. During the embryonic stages, Gfap-GFP-positive (Gfap-GFP+) cells were distributed in the entire developing dentate gyrus (DG), whereas BLBP-positive (BLBP+) cells were mainly present in the fimbria and subpial region, and to some extent in the DG. Up to postnatal day 0 (P0), double-positive cells were scarcely detected. However, at P1, one-third of the Gfap-GFP+ cells in the DG suddenly began to weakly express BLBP. Thereafter, Gfap-GFP+/BLBP+ cells rapidly increased in number, and extended their radial processes in the inner granular cell layer. At P14 and in the adult, two-thirds of the Gfap-GFP+ cells in the subgranular zone showed BLBP immunoreactivity. These results suggest that the properties of hippocampal granule progenitor cells are rapidly altered from an embryonic to adult type soon after birth.
Collapse
Affiliation(s)
- Kenta Matsue
- Department of Histology and Neuroanatomy, Tokyo Medical University, 6-1-1 Shinjuku, Shinjuku-ku, Tokyo, 160-8402, Japan
| | - Shiori Minakawa
- Department of Histology and Neuroanatomy, Tokyo Medical University, 6-1-1 Shinjuku, Shinjuku-ku, Tokyo, 160-8402, Japan
| | - Taichi Kashiwagi
- Department of Histology and Neuroanatomy, Tokyo Medical University, 6-1-1 Shinjuku, Shinjuku-ku, Tokyo, 160-8402, Japan
| | - Keiko Toda
- Department of Histology and Neuroanatomy, Tokyo Medical University, 6-1-1 Shinjuku, Shinjuku-ku, Tokyo, 160-8402, Japan
| | - Toru Sato
- Department of Histology and Neuroanatomy, Tokyo Medical University, 6-1-1 Shinjuku, Shinjuku-ku, Tokyo, 160-8402, Japan
| | - Seiji Shioda
- Institute for Advanced Bioscience Research, Hoshi University, Tokyo, Japan
| | - Tatsunori Seki
- Department of Histology and Neuroanatomy, Tokyo Medical University, 6-1-1 Shinjuku, Shinjuku-ku, Tokyo, 160-8402, Japan.
| |
Collapse
|
35
|
Dynamics and function of CXCR4 in formation of the granule cell layer during hippocampal development. Sci Rep 2017; 7:5647. [PMID: 28717168 PMCID: PMC5514042 DOI: 10.1038/s41598-017-05738-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2017] [Accepted: 06/02/2017] [Indexed: 01/03/2023] Open
Abstract
In the developing hippocampus, granule cell progenitors (GCPs) arising in the ventricular zone (VZ) migrate to the subpial region, and form the granule cell layer (GCL) of the dentate gyrus (DG). To understand the mechanism of GCL formation, we investigated the dynamics and function of CXCR4 which is expressed by the GCPs and is a receptor of the CXCL12 chemokine secreted by cells surrounding the DG. In the VZ, CXCR4 was expressed on the plasma membrane of the GCPs. During their migration and in the DG, CXCR4 was internalized and accumulated as puncta close to the centrosomes, Golgi apparatus, and lysosomes. Phosphatase analysis suggested that both phosphorylated and dephosphorylated CXCR4 exist on the plasma membrane, whereas CXCR4 in intracellular puncta was mainly dephosphorylated. Intraventricular administration of the CXCR4 antagonist AMD3100 resulted in the disappearance of CXCR4 expression from the intracellular puncta, and its appearance on the plasma membranes. Furthermore, AMD3100 treatment resulted in precocious differentiation, delayed migration, and ectopic GCPs. Taken together, these results suggest that during the development and migration of GCPs, CXCR4 on the plasma membrane is phosphorylated, internalized, sorted to the centrosomes, Golgi apparatus, and lysosomes, and functionally regulates GCP differentiation, migration and positioning.
Collapse
|
36
|
DNA Methyltransferase 1 Is Indispensable for Development of the Hippocampal Dentate Gyrus. J Neurosci 2017; 36:6050-68. [PMID: 27251626 DOI: 10.1523/jneurosci.0512-16.2016] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2016] [Accepted: 04/17/2016] [Indexed: 12/12/2022] Open
Abstract
UNLABELLED Development of the hippocampal dentate gyrus (DG) in the mammalian brain is achieved through multiple processes during late embryonic and postnatal stages, with each developmental step being strictly governed by extracellular cues and intracellular mechanisms. Here, we show that the maintenance DNA methyltransferase 1 (Dnmt1) is critical for development of the DG in the mouse. Deletion of Dnmt1 in neural stem cells (NSCs) at the beginning of DG development led to a smaller size of the granule cell layer in the DG. NSCs lacking Dnmt1 failed to establish proper radial processes or to migrate into the subgranular zone, resulting in aberrant neuronal production in the molecular layer of the DG and a reduction of integrated neurons in the granule cell layer. Interestingly, prenatal deletion of Dnmt1 in NSCs affected not only the developmental progression of the DG but also the properties of NSCs maintained into adulthood: Dnmt1-deficient NSCs displayed impaired neurogenic ability and proliferation. We also found that Dnmt1 deficiency in NSCs decreased the expression of Reelin signaling components in the developing DG and increased that of the cell cycle inhibitors p21 and p57 in the adult DG. Together, these findings led us to propose that Dnmt1 functions as a key regulator to ensure the proper development of the DG, as well as the proper status of NSCs maintained into adulthood, by modulating extracellular signaling and intracellular mechanisms. SIGNIFICANCE STATEMENT Here, we provide evidence that Dnmt1 is required for the proper development of the hippocampal dentate gyrus (DG). Deletion of Dnmt1 in neural stem cells (NSCs) at an early stage of DG development impaired the ability of NSCs to establish secondary radial glial scaffolds and to migrate into the subgranular zone of the DG, leading to aberrant neuronal production in the molecular layer, increased cell death, and decreased granule neuron production. Prenatal deletion of Dnmt1 in NSCs also induced defects in the proliferation and neurogenic ability of adult NSCs. Furthermore, we found that Dnmt1 regulates the expression of key extracellular signaling components during developmental stages while modulating intracellular mechanisms for proliferation and neuronal production of NSCs in the adult.
Collapse
|
37
|
Hao MM, Bergner AJ, Hirst CS, Stamp LA, Casagranda F, Bornstein JC, Boesmans W, Vanden Berghe P, Young HM. Spontaneous calcium waves in the developing enteric nervous system. Dev Biol 2017; 428:74-87. [PMID: 28528728 DOI: 10.1016/j.ydbio.2017.05.018] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2017] [Revised: 05/17/2017] [Accepted: 05/17/2017] [Indexed: 12/20/2022]
Abstract
The enteric nervous system (ENS) is an extensive network of neurons in the gut wall that arises from neural crest-derived cells. Like other populations of neural crest cells, it is known that enteric neural crest-derived cells (ENCCs) influence the behaviour of each other and therefore must communicate. However, little is known about how ENCCs communicate with each other. In this study, we used Ca2+ imaging to examine communication between ENCCs in the embryonic gut, using mice where ENCCs express a genetically-encoded calcium indicator. Spontaneous propagating calcium waves were observed between neighbouring ENCCs, through both neuronal and non-neuronal ENCCs. Pharmacological experiments showed wave propagation was not mediated by gap junctions, but by purinergic signalling via P2 receptors. The expression of several P2X and P2Y receptors was confirmed using RT-PCR. Furthermore, inhibition of P2 receptors altered the morphology of the ENCC network, without affecting neuronal differentiation or ENCC proliferation. It is well established that purines participate in synaptic transmission in the mature ENS. Our results describe, for the first time, purinergic signalling between ENCCs during pre-natal development, which plays roles in the propagation of Ca2+ waves between ENCCs and in ENCC network formation. One previous study has shown that calcium signalling plays a role in sympathetic ganglia formation; our results suggest that calcium waves are likely to be important for enteric ganglia development.
Collapse
Affiliation(s)
- Marlene M Hao
- Department of Anatomy and Neuroscience, University of Melbourne, Australia; Laboratory for Enteric Neuroscience, TARGID, University of Leuven, Belgium.
| | - Annette J Bergner
- Department of Anatomy and Neuroscience, University of Melbourne, Australia
| | - Caroline S Hirst
- Department of Anatomy and Neuroscience, University of Melbourne, Australia
| | - Lincon A Stamp
- Department of Anatomy and Neuroscience, University of Melbourne, Australia
| | - Franca Casagranda
- Department of Anatomy and Neuroscience, University of Melbourne, Australia
| | | | - Werend Boesmans
- Laboratory for Enteric Neuroscience, TARGID, University of Leuven, Belgium
| | | | - Heather M Young
- Department of Anatomy and Neuroscience, University of Melbourne, Australia
| |
Collapse
|
38
|
Time-lapse imaging reveals highly dynamic structural maturation of postnatally born dentate granule cells in organotypic entorhino-hippocampal slice cultures. Sci Rep 2017; 7:43724. [PMID: 28256620 PMCID: PMC5335612 DOI: 10.1038/srep43724] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2016] [Accepted: 01/27/2017] [Indexed: 12/18/2022] Open
Abstract
Neurogenesis of hippocampal granule cells (GCs) persists throughout mammalian life and is important for learning and memory. How newborn GCs differentiate and mature into an existing circuit during this time period is not yet fully understood. We established a method to visualize postnatally generated GCs in organotypic entorhino-hippocampal slice cultures (OTCs) using retroviral (RV) GFP-labeling and performed time-lapse imaging to study their morphological development in vitro. Using anterograde tracing we could, furthermore, demonstrate that the postnatally generated GCs in OTCs, similar to adult born GCs, grow into an existing entorhino-dentate circuitry. RV-labeled GCs were identified and individual cells were followed for up to four weeks post injection. Postnatally born GCs exhibited highly dynamic structural changes, including dendritic growth spurts but also retraction of dendrites and phases of dendritic stabilization. In contrast, older, presumably prenatally born GCs labeled with an adeno-associated virus (AAV), were far less dynamic. We propose that the high degree of structural flexibility seen in our preparations is necessary for the integration of newborn granule cells into an already existing neuronal circuit of the dentate gyrus in which they have to compete for entorhinal input with cells generated and integrated earlier.
Collapse
|
39
|
Miyata M, Maruo T, Kaito A, Wang S, Yamamoto H, Fujiwara T, Mizoguchi A, Mandai K, Takai Y. Roles of afadin in the formation of the cellular architecture of the mouse hippocampus and dentate gyrus. Mol Cell Neurosci 2017; 79:34-44. [DOI: 10.1016/j.mcn.2016.12.007] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2016] [Revised: 11/15/2016] [Accepted: 12/27/2016] [Indexed: 12/19/2022] Open
|
40
|
Namba T, Huttner WB. Neural progenitor cells and their role in the development and evolutionary expansion of the neocortex. WILEY INTERDISCIPLINARY REVIEWS-DEVELOPMENTAL BIOLOGY 2016; 6. [PMID: 27865053 DOI: 10.1002/wdev.256] [Citation(s) in RCA: 85] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/16/2016] [Revised: 09/02/2016] [Accepted: 09/07/2016] [Indexed: 01/16/2023]
Abstract
The evolutionary expansion of the mammalian brain, notably the neocortex, provides a platform for the higher cognitive abilities that characterize humans. Cortical expansion is accompanied by increased folding of the pial surface, which gives rise to a gyrencephalic (folded) rather than lissencephalic (unfolded) neocortex. This expansion reflects the prolonged and increased proliferation of neural stem and progenitor cells (NPCs). Distinct classes of NPCs can be distinguished based on either cell biological criteria (apical progenitors [APs], basal progenitors [BPs]) or lineage (primary progenitors and secondary progenitors). Cortical expansion in development and evolution is linked to an increased abundance and proliferative capacity of BPs, notably basal radial glial cells, a recently characterized type of secondary progenitor derived from apical radial glial cells, the primary progenitors. To gain insight into the molecular basis underlying the prolonged and increased proliferation of NPCs and in particular BPs, comparative genomic and transcriptomic approaches, mostly for human versus mouse, have been employed and applied to specific NPC types and subpopulations. These have revealed two principal sets of molecular changes. One concerns differences in the expression of common genes between species with different degrees of cortical expansion. The other comprises human-specific genes or genomic regulatory sequences. Various systems that allow functional testing of these genomic and gene expression differences between species have emerged, including transient and stable transgenesis, genome editing, cerebral organoids, and organotypic slice cultures. These provide future avenues for uncovering the molecular basis of cortical expansion. WIREs Dev Biol 2017, 6:e256. doi: 10.1002/wdev.256 For further resources related to this article, please visit the WIREs website.
Collapse
Affiliation(s)
- Takashi Namba
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | - Wieland B Huttner
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| |
Collapse
|
41
|
Jourdon A, Gresset A, Spassky N, Charnay P, Topilko P, Santos R. Prss56, a novel marker of adult neurogenesis in the mouse brain. Brain Struct Funct 2015; 221:4411-4427. [DOI: 10.1007/s00429-015-1171-z] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2015] [Accepted: 12/07/2015] [Indexed: 12/25/2022]
|
42
|
Neural stem cells and neuro/gliogenesis in the central nervous system: understanding the structural and functional plasticity of the developing, mature, and diseased brain. J Physiol Sci 2015; 66:197-206. [PMID: 26578509 PMCID: PMC4823343 DOI: 10.1007/s12576-015-0421-4] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2015] [Accepted: 10/18/2015] [Indexed: 11/29/2022]
Abstract
Neurons and glia in the central nervous system (CNS) originate from neural stem cells (NSCs). Knowledge of the mechanisms of neuro/gliogenesis from NSCs is fundamental to our understanding of how complex brain architecture and function develop. NSCs are present not only in the developing brain but also in the mature brain in adults. Adult neurogenesis likely provides remarkable plasticity to the mature brain. In addition, recent progress in basic research in mental disorders suggests an etiological link with impaired neuro/gliogenesis in particular brain regions. Here, we review the recent progress and discuss future directions in stem cell and neuro/gliogenesis biology by introducing several topics presented at a joint meeting of the Japanese Association of Anatomists and the Physiological Society of Japan in 2015. Collectively, these topics indicated that neuro/gliogenesis from NSCs is a common event occurring in many brain regions at various ages in animals. Given that significant structural and functional changes in cells and neural networks are accompanied by neuro/gliogenesis from NSCs and the integration of newly generated cells into the network, stem cell and neuro/gliogenesis biology provides a good platform from which to develop an integrated understanding of the structural and functional plasticity that underlies the development of the CNS, its remodeling in adulthood, and the recovery from diseases that affect it.
Collapse
|
43
|
Hevner RF. Evolution of the mammalian dentate gyrus. J Comp Neurol 2015; 524:578-94. [PMID: 26179319 DOI: 10.1002/cne.23851] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2015] [Revised: 06/02/2015] [Accepted: 07/06/2015] [Indexed: 01/08/2023]
Abstract
The dentate gyrus (DG), a part of the hippocampal formation, has important functions in learning, memory, and adult neurogenesis. Compared with homologous areas in sauropsids (birds and reptiles), the mammalian DG is larger and exhibits qualitatively different phenotypes: 1) folded (C- or V-shaped) granule neuron layer, concave toward the hilus and delimited by a hippocampal fissure; 2) nonperiventricular adult neurogenesis; and 3) prolonged ontogeny, involving extensive abventricular (basal) migration and proliferation of neural stem and progenitor cells (NSPCs). Although gaps remain, available data indicate that these DG traits are present in all orders of mammals, including monotremes and marsupials. The exception is Cetacea (whales, dolphins, and porpoises), in which DG size, convolution, and adult neurogenesis have undergone evolutionary regression. Parsimony suggests that increased growth and convolution of the DG arose in stem mammals concurrently with nonperiventricular adult hippocampal neurogenesis and basal migration of NSPCs during development. These traits could all result from an evolutionary change that enhanced radial migration of NSPCs out of the periventricular zones, possibly by epithelial-mesenchymal transition, to colonize and maintain nonperiventricular proliferative niches. In turn, increased NSPC migration and clonal expansion might be a consequence of growth in the cortical hem (medial patterning center), which produces morphogens such as Wnt3a, generates Cajal-Retzius neurons, and is regulated by Lhx2. Finally, correlations between DG convolution and neocortical gyrification (or capacity for gyrification) suggest that enhanced abventricular migration and proliferation of NSPCs played a transformative role in growth and folding of neocortex as well as archicortex.
Collapse
Affiliation(s)
- Robert F Hevner
- Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, Washington, 98101
- Department of Neurological Surgery, University of Washington, Seattle, Washington, 98104
| |
Collapse
|
44
|
Hayashi K, Kubo KI, Kitazawa A, Nakajima K. Cellular dynamics of neuronal migration in the hippocampus. Front Neurosci 2015; 9:135. [PMID: 25964735 PMCID: PMC4408843 DOI: 10.3389/fnins.2015.00135] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2015] [Accepted: 04/02/2015] [Indexed: 12/30/2022] Open
Abstract
A fine structure of the hippocampus is required for proper functions, and disruption of this formation by neuronal migration defects during development may play a role in some psychiatric illnesses. During hippocampal development in rodents, pyramidal neurons in the Ammon's horn are mostly generated in the ventricular zone (VZ), spent as multipolar cells just above the VZ, and then migrate radially toward the pial surface, ultimately settling into the hippocampal plate. Although this process is similar to that of neocortical projection neurons, these are not identical. In addition to numerous histological studies, the development of novel techniques gives a clear picture of the cellular dynamics of hippocampal neurons, as well as neocortical neurons. In this article, we provide an overview of the cellular mechanisms of rodent hippocampal neuronal migration including those of dentate granule cells, especially focusing on the differences of migration modes between hippocampal neurons and neocortical neurons. The unique migration mode of hippocampal pyramidal neurons might enable clonally related cells in the Ammon's horn to distribute in a horizontal fashion.
Collapse
Affiliation(s)
- Kanehiro Hayashi
- Department of Anatomy, Keio University School of Medicine Tokyo, Japan
| | - Ken-Ichiro Kubo
- Department of Anatomy, Keio University School of Medicine Tokyo, Japan
| | - Ayako Kitazawa
- Department of Anatomy, Keio University School of Medicine Tokyo, Japan
| | - Kazunori Nakajima
- Department of Anatomy, Keio University School of Medicine Tokyo, Japan
| |
Collapse
|
45
|
Urbán N, Guillemot F. Neurogenesis in the embryonic and adult brain: same regulators, different roles. Front Cell Neurosci 2014; 8:396. [PMID: 25505873 PMCID: PMC4245909 DOI: 10.3389/fncel.2014.00396] [Citation(s) in RCA: 340] [Impact Index Per Article: 30.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2014] [Accepted: 11/05/2014] [Indexed: 12/12/2022] Open
Abstract
Neurogenesis persists in adult mammals in specific brain areas, known as neurogenic niches. Adult neurogenesis is highly dynamic and is modulated by multiple physiological stimuli and pathological states. There is a strong interest in understanding how this process is regulated, particularly since active neuronal production has been demonstrated in both the hippocampus and the subventricular zone (SVZ) of adult humans. The molecular mechanisms that control neurogenesis have been extensively studied during embryonic development. Therefore, we have a broad knowledge of the intrinsic factors and extracellular signaling pathways driving proliferation and differentiation of embryonic neural precursors. Many of these factors also play important roles during adult neurogenesis, but essential differences exist in the biological responses of neural precursors in the embryonic and adult contexts. Because adult neural stem cells (NSCs) are normally found in a quiescent state, regulatory pathways can affect adult neurogenesis in ways that have no clear counterpart during embryogenesis. BMP signaling, for instance, regulates NSC behavior both during embryonic and adult neurogenesis. However, this pathway maintains stem cell proliferation in the embryo, while it promotes quiescence to prevent stem cell exhaustion in the adult brain. In this review, we will compare and contrast the functions of transcription factors (TFs) and other regulatory molecules in the embryonic brain and in adult neurogenic regions of the adult brain in the mouse, with a special focus on the hippocampal niche and on the regulation of the balance between quiescence and activation of adult NSCs in this region.
Collapse
Affiliation(s)
- Noelia Urbán
- Department of Molecular Neurobiology, MRC National Institute for Medical Research London, UK
| | - François Guillemot
- Department of Molecular Neurobiology, MRC National Institute for Medical Research London, UK
| |
Collapse
|
46
|
Kirshenbaum GS, Lieberman SR, Briner TJ, Leonardo ED, Dranovsky A. Adolescent but not adult-born neurons are critical for susceptibility to chronic social defeat. Front Behav Neurosci 2014; 8:289. [PMID: 25221485 PMCID: PMC4147831 DOI: 10.3389/fnbeh.2014.00289] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2014] [Accepted: 08/07/2014] [Indexed: 11/26/2022] Open
Abstract
Recent evidence implicates adult hippocampal neurogenesis in regulating behavioral and physiologic responses to stress. Hippocampal neurogenesis occurs across the lifespan, however the rate of cell birth is up to 300% higher in adolescent mice compared to adults. Adolescence is a sensitive period in development where emotional circuitry and stress reactivity undergo plasticity establishing life-long set points. Therefore neurogenesis occurring during adolescence may be particularly important for emotional behavior. However, little is known about the function of hippocampal neurons born during adolescence. In order to assess the contribution of neurons born in adolescence to the adult stress response and depression-related behavior, we transiently reduced cell proliferation either during adolescence, or during adulthood in GFAP-Tk mice. We found that the intervention in adolescence did not change adult baseline behavioral response in the forced swim test, sucrose preference test or social affiliation test, and did not change adult corticosterone responses to an acute stressor. However following chronic social defeat, adult mice with reduced adolescent neurogenesis showed a resilient phenotype. A similar transient reduction in adult neurogenesis did not affect depression-like behaviors or stress induced corticosterone. Our study demonstrates that hippocampal neurons born during adolescence, but not in adulthood are important to confer susceptibility to chronic social defeat.
Collapse
Affiliation(s)
- Greer S Kirshenbaum
- Dranovsky-Leonardo Lab, Department of Psychiatry, Division of Integrative Neuroscience, New York State Psychiatric Institute, Columbia University New York, NY, USA
| | - Sophie R Lieberman
- Dranovsky-Leonardo Lab, Department of Psychiatry, Division of Integrative Neuroscience, New York State Psychiatric Institute, Columbia University New York, NY, USA
| | - Tamara J Briner
- Dranovsky-Leonardo Lab, Department of Psychiatry, Division of Integrative Neuroscience, New York State Psychiatric Institute, Columbia University New York, NY, USA
| | - E David Leonardo
- Dranovsky-Leonardo Lab, Department of Psychiatry, Division of Integrative Neuroscience, New York State Psychiatric Institute, Columbia University New York, NY, USA
| | - Alex Dranovsky
- Dranovsky-Leonardo Lab, Department of Psychiatry, Division of Integrative Neuroscience, New York State Psychiatric Institute, Columbia University New York, NY, USA
| |
Collapse
|
47
|
Sugiyama T, Osumi N, Katsuyama Y. A novel cell migratory zone in the developing hippocampal formation. J Comp Neurol 2014; 522:3520-38. [PMID: 24771490 DOI: 10.1002/cne.23621] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2014] [Revised: 04/25/2014] [Accepted: 04/25/2014] [Indexed: 11/08/2022]
Abstract
The hippocampal formation (HF) is a unique structure in the mammalian brain and is subdivided into the dentate gyrus, Ammon's horn, and subiculum by their functions and connectivity in the neuronal circuit. Because behaviors of the neural stem cells, neuronal progenitors, and the differentiating neurons are complex during hippocampal morphogenesis, the differentiation of these subdivisions has not been well understood. In this study, we investigated embryonic and postnatal expression of the proteins Prox1, Math2, and Ctip2, which clearly indicate principal cells of the dentate gyrus (Prox1 positive) and Ammon's horn (Math2 and Ctip2 positive). Expression patterns of Prox1 and Math2 were consistent with previously suggested localization of migratory pathways of the dentate granule cells and hippocampal pyramidal cells. Interestingly, we found intermingling of Prox1-expressing cells and Math2-expressing cells in a cell migratory stream, suggesting previously unknown behaviors of differentiating cells of the HF.
Collapse
Affiliation(s)
- Taku Sugiyama
- Department of Developmental Neuroscience, Tohoku University Graduate School of Medicine, Sendai, 980-8575, Japan
| | | | | |
Collapse
|
48
|
Takimoto N, Wang L, Itahashi M, Ogawa T, Segawa R, Hara S, Murakami T, Suzuki K, Shibutani M. Maternal single injection of N-methyl-N-nitrosourea to cause microcephaly in offspring induces transient aberration of hippocampal neurogenesis in mice. Toxicol Lett 2014; 226:20-7. [DOI: 10.1016/j.toxlet.2014.01.014] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2013] [Revised: 01/07/2014] [Accepted: 01/08/2014] [Indexed: 10/25/2022]
|
49
|
Kipanyula MJ, Kimaro WH, Yepnjio FN, Aldebasi YH, Farahna M, Nwabo Kamdje AH, Abdel-Magied EM, Seke Etet PF. Signaling pathways bridging fate determination of neural crest cells to glial lineages in the developing peripheral nervous system. Cell Signal 2013; 26:673-82. [PMID: 24378534 DOI: 10.1016/j.cellsig.2013.12.007] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2013] [Revised: 12/13/2013] [Accepted: 12/22/2013] [Indexed: 11/29/2022]
Abstract
Fate determination of neural crest cells is an essential step for the development of different crest cell derivatives. Peripheral glia development is marked by the choice of the neural crest cells to differentiate along glial lineages. The molecular mechanism underlying fate acquisition is poorly understood. However, recent advances have identified different transcription factors and genes required for the complex instructive signaling process that comprise both local environmental and cell intrinsic cues. Among others, at least the roles of Sox10, Notch, and neuregulin 1 have been documented in both in vivo and in vitro models. Cooperative interactions of such factors appear to be necessary for the switch from multipotent neural crest cells to glial lineage precursors in the peripheral nervous system. This review summarizes recent advances in the understanding of fate determination of neural crest cells into different glia subtypes, together with the potential implications in regenerative medicine.
Collapse
Affiliation(s)
- Maulilio John Kipanyula
- Department of Veterinary Anatomy, Faculty of Veterinary Medicine, Sokoine University of Agriculture, P.O. Box 3016, Chuo Kikuu, Morogoro, Tanzania.
| | - Wahabu Hamisi Kimaro
- Department of Veterinary Anatomy, Faculty of Veterinary Medicine, Sokoine University of Agriculture, P.O. Box 3016, Chuo Kikuu, Morogoro, Tanzania
| | - Faustin N Yepnjio
- Neurology Department, Yaoundé Central Hospital, Department of Internal Medicine and Specialties, University of Yaoundé I, P.O. Box 1937, Yaoundé, Cameroon
| | - Yousef H Aldebasi
- Department of Optometry, College of Applied Medical Sciences, Qassim University, 51452 Buraydah, Saudi Arabia
| | - Mohammed Farahna
- Department of Basic Health Sciences, College of Applied Medical Sciences, Qassim University, 51452 Buraydah, Saudi Arabia
| | | | - Eltuhami M Abdel-Magied
- Department of Anatomy and Histology, College of Medicine, Qassim University, 51452 Buraydah, Saudi Arabia
| | - Paul Faustin Seke Etet
- Department of Basic Health Sciences, College of Applied Medical Sciences, Qassim University, 51452 Buraydah, Saudi Arabia.
| |
Collapse
|