1
|
Herpich ME, de Oliveira Guarnieri L, de Oliveira ACP, Moraes MFD. Bacterial Lipopolysaccharide Post-Conditioning in The kainic acid animal model of Temporal Lobe epilepsy. Epilepsy Behav 2024; 161:110076. [PMID: 39467457 DOI: 10.1016/j.yebeh.2024.110076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Revised: 09/24/2024] [Accepted: 10/03/2024] [Indexed: 10/30/2024]
Abstract
This study used intra-hippocampal injections of Kainic Acid (KA) in Wistar rats to induce spontaneous recurrent seizures (SRS) after a 9-day latent period. A post-conditioning protocol with LPS, injected at the same site 72 h after the initial KA insult, was employed to trigger secondary competing processes. To evaluate the post-conditioning effect of LPS, 25 animals were divided into four groups: SAL-SAL (n = 6), KA-SAL (n = 6), SAL-LPS (n = 7), and KA-LPS (n = 6). SRS occurrence and seizure duration were quantified through video monitoring from days 9 to 17, along with other ictal behaviors, such as tail-chasing and wet-dog-shakes. Behavioral assessments revealed that the KA-LPS group had preserved sucrose preference and intact long-term memory in the object recognition test, indicating reduced depressive-like behavior and cognitive preservation compared to the KA-SAL group. The forced swim test showed increased depressive-like behavior in the SAL-LPS group, with LPS mitigating these effects in the KA group. The marble-burying test showed no significant differences among groups. Animals were euthanized on day 26, and hippocampal slices were analyzed using fluoro-jade staining for cell death and immunofluorescence staining for Iba-1 (microglia) and GFAP (astrocyte) labeling. The results support the hypothesis that epileptogenesis involves a cascade of plastic changes in neural networks and that precise, timely interventions can potentially interfere with this process.
Collapse
Affiliation(s)
- Mateus Eduardo Herpich
- Núcleo de Neurociências, Departamento de Fisiologia e Biofísica, Instituto de Ciências Biológicas - Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Leonardo de Oliveira Guarnieri
- Núcleo de Neurociências, Departamento de Fisiologia e Biofísica, Instituto de Ciências Biológicas - Universidade Federal de Minas Gerais, Belo Horizonte, Brazil; Centro de Tecnologia e Pesquisa em Magneto Ressonância, Programa de Pós-Graduação em Engenharia Elétrica, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | | | - Márcio Flávio Dutra Moraes
- Núcleo de Neurociências, Departamento de Fisiologia e Biofísica, Instituto de Ciências Biológicas - Universidade Federal de Minas Gerais, Belo Horizonte, Brazil; Centro de Tecnologia e Pesquisa em Magneto Ressonância, Programa de Pós-Graduação em Engenharia Elétrica, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil.
| |
Collapse
|
2
|
Pastor-Alonso O, Durá I, Bernardo-Castro S, Varea E, Muro-García T, Martín-Suárez S, Encinas-Pérez JM, Pineda JR. HB-EGF activates EGFR to induce reactive neural stem cells in the mouse hippocampus after seizures. Life Sci Alliance 2024; 7:e202201840. [PMID: 38977310 PMCID: PMC11231495 DOI: 10.26508/lsa.202201840] [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: 11/18/2022] [Revised: 06/18/2024] [Accepted: 06/20/2024] [Indexed: 07/10/2024] Open
Abstract
Hippocampal seizures mimicking mesial temporal lobe epilepsy cause a profound disruption of the adult neurogenic niche in mice. Seizures provoke neural stem cells to switch to a reactive phenotype (reactive neural stem cells, React-NSCs) characterized by multibranched hypertrophic morphology, massive activation to enter mitosis, symmetric division, and final differentiation into reactive astrocytes. As a result, neurogenesis is chronically impaired. Here, using a mouse model of mesial temporal lobe epilepsy, we show that the epidermal growth factor receptor (EGFR) signaling pathway is key for the induction of React-NSCs and that its inhibition exerts a beneficial effect on the neurogenic niche. We show that during the initial days after the induction of seizures by a single intrahippocampal injection of kainic acid, a strong release of zinc and heparin-binding epidermal growth factor, both activators of the EGFR signaling pathway in neural stem cells, is produced. Administration of the EGFR inhibitor gefitinib, a chemotherapeutic in clinical phase IV, prevents the induction of React-NSCs and preserves neurogenesis.
Collapse
Affiliation(s)
- Oier Pastor-Alonso
- Laboratory of Neural Stem Cells and Neurogenesis, Achucarro Basque Center for Neuroscience, Bizkaia, Spain
| | - Irene Durá
- Laboratory of Neural Stem Cells and Neurogenesis, Achucarro Basque Center for Neuroscience, Bizkaia, Spain
| | - Sara Bernardo-Castro
- Laboratory of Neural Stem Cells and Neurogenesis, Achucarro Basque Center for Neuroscience, Bizkaia, Spain
| | - Emilio Varea
- Faculty of Biology, University of Valencia, Valencia, Spain
| | - Teresa Muro-García
- Laboratory of Neural Stem Cells and Neurogenesis, Achucarro Basque Center for Neuroscience, Bizkaia, Spain
| | - Soraya Martín-Suárez
- Laboratory of Neural Stem Cells and Neurogenesis, Achucarro Basque Center for Neuroscience, Bizkaia, Spain
| | - Juan Manuel Encinas-Pérez
- Laboratory of Neural Stem Cells and Neurogenesis, Achucarro Basque Center for Neuroscience, Bizkaia, Spain
- Ikerbasque, The Basque Foundation for Science, Bizkaia, Spain
- Department of Neurosciences, University of the Basque Country (UPV/EHU), Bizkaia, Spain
| | - Jose Ramon Pineda
- Laboratory of Neural Stem Cells and Neurogenesis, Achucarro Basque Center for Neuroscience, Bizkaia, Spain
- Signaling Lab, Department of Cell Biology and Histology, Faculty of Medicine and Nursing, University of the Basque Country (UPV/EHU), Bizkaia, Spain
| |
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
|
Fallahi S, Zangbar HS, Farajdokht F, Rahbarghazi R, Ghiasi F, Mohaddes G. Mesenchymal stem cell-derived exosomes improve neurogenesis and cognitive function of mice with methamphetamine addiction: A novel treatment approach. CNS Neurosci Ther 2024; 30:e14719. [PMID: 38783536 PMCID: PMC11116483 DOI: 10.1111/cns.14719] [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: 10/08/2023] [Revised: 03/01/2024] [Accepted: 03/04/2024] [Indexed: 05/25/2024] Open
Abstract
BACKGROUND Methamphetamine (METH) is a psychostimulant substance with highly addictive and neurotoxic effects, but no ideal treatment option exists to improve METH-induced neurocognitive deficits. Recently, mesenchymal stem cells (MSCs)-derived exosomes have raised many hopes for treating neurodegenerative sequela of brain disorders. This study aimed to determine the therapeutic potential of MSCs-derived exosomes on cognitive function and neurogenesis of METH-addicted rodents. METHODS Male BALB/c mice were subjected to chronic METH addiction, followed by intravenous administration of bone marrow MSCs-derived exosomes. Then, the spatial memory and recognition memory of animals were assessed by the Barnes maze and the novel object recognition test (NORT). The neurogenesis-related factors, including NeuN and DCX, and the expression of Iba-1, a microglial activation marker, were assessed in the hippocampus by immunofluorescence staining. Also, the expression of inflammatory cytokines, including TNF-α and NF-κB, were evaluated by western blotting. RESULTS The results showed that BMSCs-exosomes improved the time spent in the target quadrant and correct-to-wrong relative time in the Barnes maze. Also, NORT's discrimination index (DI) and recognition index (RI) were improved following exosome therapy. Additionally, exosome therapy significantly increased the expression of NeuN and DCX in the hippocampus while decreasing the expression of inflammatory cytokines, including TNF-α and NF-κB. Besides, BMSC-exosomes down-regulated the expression of Iba-1. CONCLUSION Our findings indicate that BMSC-exosomes mitigated METH-caused cognitive dysfunction by improving neurogenesis and inhibiting neuroinflammation in the hippocampus.
Collapse
Affiliation(s)
- Solmaz Fallahi
- Drug Applied Research CenterTabriz University of Medical SciencesTabrizIran
- Department of PhysiologyTabriz University of Medical SciencesTabrizIran
| | - Hamid Soltani Zangbar
- Department of Neuroscience and Cognition, Faculty of Advanced Medical SciencesTabriz University of Medical SciencesTabrizIran
| | - Fereshteh Farajdokht
- Department of PhysiologyTabriz University of Medical SciencesTabrizIran
- Neurosciences Research CenterTabriz University of Medical SciencesTabrizIran
| | - Reza Rahbarghazi
- Department of Applied Cell Sciences, Faculty of Advanced Medical SciencesTabriz University of Medical SciencesTabrizIran
| | - Fariba Ghiasi
- Drug Applied Research CenterTabriz University of Medical SciencesTabrizIran
- Department of PhysiologyTabriz University of Medical SciencesTabrizIran
| | - Gisou Mohaddes
- Drug Applied Research CenterTabriz University of Medical SciencesTabrizIran
- Department of PhysiologyTabriz University of Medical SciencesTabrizIran
- Department of Neuroscience and Cognition, Faculty of Advanced Medical SciencesTabriz University of Medical SciencesTabrizIran
- Department of Biomedical EducationCalifornia Health Sciences University, College of Osteopathic MedicineClovisCaliforniaUSA
| |
Collapse
|
5
|
Ledesma-Corvi S, García-Fuster MJ. Electroconvulsive seizures regulate various stages of hippocampal cell genesis and mBDNF at different times after treatment in adolescent and adult rats of both sexes. Front Mol Neurosci 2023; 16:1275783. [PMID: 37965039 PMCID: PMC10642262 DOI: 10.3389/fnmol.2023.1275783] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Accepted: 10/16/2023] [Indexed: 11/16/2023] Open
Abstract
Electroconvulsive therapy, a fast-acting option for treatment-resistant depression, is modeled at the preclinical level through the induction of electroconvulsive seizures (ECS) in rodents. Recent studies from our group proved sex- and age-differences in the antidepressant-like response elicited by ECS in rats; while an antidepressant-like response was observed in male adolescent and adult rats (although with greater efficacy in adulthood), the same parameters rendered inefficacious in females of any age. To better understand the potential sex differences taking place at the molecular level that might be mediating these behavioral disparities, we evaluated the impact of a repeated treatment with ECS (95 mA for 0.6 s, 100 Hz, 0.6 ms) in adolescent and adult rats of both sexes. Several hippocampal markers of neuroplasticity, commonly regulated by most antidepressants, such as those of neurogenesis (cell proliferation, neurogenic differentiation, long-term cell survival) or mBDNF and associated signaling (e.g., mTOR and ERK1/2) were evaluated at different time-points after treatment (1-, 8-, 15- and up to 30-days post-treatment). The main results demonstrated that ECS improved the survival rate of new cells born in the dentate gryus before treatment. Moreover, ECS increased cell proliferation and neurogenic differentiation at different times post-treatment, paired with persistent increases in mBDNF, observed long after treatment. In general, effects were different for each sex and varied with the age of the animal (adolescent vs. adulthood). The present study is the first-one to demonstrate that such persistent molecular changes induced by ECS in hippocampus, some of them observed up to 30-days post-treatment, also occurred in female rats and adolescence. Although these molecular changes could not justify the lack of ECS efficacy described by these same parameters of ECS in female rats (vs. male rats), they proposed certain beneficial effects common to both sexes, and age periods studied, opening the avenue for further studies. Based on these neurochemical effects, ECS should have displayed similar efficacies for both biological sexes. Therefore, the reason behind these disparities should be further explored to better translate efficacious treatments specific and/or personalized for each sex to the clinic.
Collapse
Affiliation(s)
- Sandra Ledesma-Corvi
- IUNICS, University of the Balearic Islands, Palma, Spain
- Health Research Institute of Balearic Islands (IdISBa), Palma, Spain
| | - M. Julia García-Fuster
- IUNICS, University of the Balearic Islands, Palma, Spain
- Health Research Institute of Balearic Islands (IdISBa), Palma, Spain
- Department of Medicine, University of the Balearic Islands, Palma, Spain
| |
Collapse
|
6
|
Ledesma-Corvi S, García-Fuster MJ. Comparing the antidepressant-like effects of electroconvulsive seizures in adolescent and adult female rats: an intensity dose-response study. Biol Sex Differ 2023; 14:67. [PMID: 37777813 PMCID: PMC10541687 DOI: 10.1186/s13293-023-00552-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Accepted: 09/22/2023] [Indexed: 10/02/2023] Open
Abstract
BACKGROUND The induction of electroconvulsive seizures (ECS) in rodents induces sex- and age-specific disparities in antidepressant-like responses, with females and young age being the most unresponsive ones. Since the electrical charge needed to induce an effective convulsion is also altered by these variables, our aim was to compare different dose-intensities of ECS exclusively in female rats, since there is a lack of preclinical data characterizing this particular sex, while also evaluating efficacy during distinctive age periods of treatment (adolescence vs. adulthood). METHODS Adolescent and adult female Sprague-Dawley rats were exposed to an intensity dose-response study (55, 75 or 95 mA; 0.6 s, 100 Hz, 1 session/day, 5 days). The particular characteristics of the induced convulsions (tonic, clonic, recovery times) were monitored during treatment. Antidepressant-like responses were evaluated under the stress of the forced-swim test 1-, 3-, and 7-days post-treatment (i.e., improved immobility time as an indicative of an antidepressant-like response), and brains were collected 24 h later (8 days post-treatment) to evaluate potential changes in hippocampal neurogenesis (Ki-67 and NeuroD) by immunohistochemistry. RESULTS The lowest intensities tested of ECS (55 and 75 mA) induced an antidepressant-like effect in adult female rats, but rendered insufficient in adolescence. The lack of efficacy observed in adolescent rats paralleled differences in the characteristics of the seizures induced by ECS as compared to adulthood. In line with prior results, different dose-intensities of ECS modulated hippocampal neurogenesis in a comparable fashion with age (i.e., increased survival of neural progenitors 8 days post-treatment). CONCLUSIONS In conjunction, these results reinforce the importance of fine-tuning the parameters of ECS that might render efficacious while considering sex and age as essential variables for treatment response, and suggest that other molecular mechanisms, beside the partial role of hippocampal neurogenesis, might be participating in the antidepressant-like effects induced by ECS.
Collapse
Affiliation(s)
- Sandra Ledesma-Corvi
- IUNICS, University of the Balearic Islands, Cra. de Valldemossa Km 7.5, 07122, Palma, Spain
- Health Research Institute of the Balearic Islands (IdISBa), Palma, Spain
| | - M Julia García-Fuster
- IUNICS, University of the Balearic Islands, Cra. de Valldemossa Km 7.5, 07122, Palma, Spain.
- Health Research Institute of the Balearic Islands (IdISBa), Palma, Spain.
- Department of Medicine, University of the Balearic Islands, Palma, Spain.
| |
Collapse
|
7
|
Li R, Xiong W, Li B, Li Y, Fang B, Wang X, Ren F. Plasmalogen Improves Memory Function by Regulating Neurogenesis in a Mouse Model of Alzheimer's Diseases. Int J Mol Sci 2023; 24:12234. [PMID: 37569610 PMCID: PMC10418626 DOI: 10.3390/ijms241512234] [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: 07/10/2023] [Revised: 07/27/2023] [Accepted: 07/27/2023] [Indexed: 08/13/2023] Open
Abstract
Adult hippocampal neurogenesis (AHN) is associated with hippocampus-dependent cognitive function, and its initiation is attributed to neural stem cells (NSCs). Dysregulated AHN has been identified in Alzheimer's disease (AD) and may underlie impaired cognitive function in AD. Modulating the function of NSCs and stimulating AHN are potential ways to manipulate AD. Plasmalogen (PLA) are a class of cell membrane glycerophospholipids which exhibit neuroprotective properties. However, the effect of PLA on altered AHN in AD has not been investigated. In our study, PLA(10μg/mL) -attenuated Aβ (1-42) (5μM) induced a decrease in NSC viability and neuronal differentiation of NSCs, partially through regulating the Wnt/β-catenin pathway. Additionally, AD mice were supplemented with PLA (67mg/kg/day) for 6 weeks. PLA treatment improved the impaired AHN in AD mice, including increasing the number of neural stem cells (NSCs) and newly generated neurons. The memory function of AD mice was also enhanced after PLA administration. Therefore, it was summarized that PLA could regulate NSC differentiation by activating the Wnt/β-catenin pathway and ameliorate AD-related memory impairment through up-regulating AHN.
Collapse
Affiliation(s)
- Rongzi Li
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing 100083, China; (R.L.); (W.X.); (B.L.); (Y.L.); (B.F.)
| | - Wei Xiong
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing 100083, China; (R.L.); (W.X.); (B.L.); (Y.L.); (B.F.)
- Food Laboratory of Zhongyuan, Luohe 462000, China
| | - Boying Li
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing 100083, China; (R.L.); (W.X.); (B.L.); (Y.L.); (B.F.)
| | - Yixuan Li
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing 100083, China; (R.L.); (W.X.); (B.L.); (Y.L.); (B.F.)
| | - Bing Fang
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing 100083, China; (R.L.); (W.X.); (B.L.); (Y.L.); (B.F.)
| | - Xifan Wang
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing 100083, China; (R.L.); (W.X.); (B.L.); (Y.L.); (B.F.)
| | - Fazheng Ren
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing 100083, China; (R.L.); (W.X.); (B.L.); (Y.L.); (B.F.)
- Food Laboratory of Zhongyuan, Luohe 462000, China
| |
Collapse
|
8
|
Huang Y, Sun W, Gao F, Ma H, Yuan T, Liu Z, Liu H, Hu J, Bai J, Zhang X, Wang R. Brain-Derived Estrogen Regulates Neurogenesis, Learning and Memory with Aging in Female Rats. BIOLOGY 2023; 12:760. [PMID: 37372046 DOI: 10.3390/biology12060760] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Revised: 05/18/2023] [Accepted: 05/18/2023] [Indexed: 06/29/2023]
Abstract
Although 17β-estradiol (E2) can be locally synthesized in the brain, whether and how brain-derived E2 (BDE2) impacts neurogenesis with aging is largely unclear. In this study, we examined the hippocampal neural stem cells, neurogenesis, and gliogenesis of 1, 3, 6, 14, and 18-month (Mon) female rats. Female forebrain neuronal aromatase knockout (FBN-ARO-KO) rats and letrozole-treated rats were also employed. We demonstraed that (1) the number of neural stem cells declined over 14-Mon age, and the differentiation of astrocytes and microglia markedly elevated and exhibited excessive activation. KO rats showed declines in astrocyte A2 subtype and elevation in A1 subtype at 18 Mon; (2) neurogenesis sharply dropped from 1-Mon age; (3) KO suppressed dentate gyrus (DG) neurogenesis at 1, 6 and 18 Mon. Additionally, KO and letrozole treatment led to declined neurogenesis at 1-Mon age, compared to age-matched WT controls; (4) FBN-ARO-KO inhibited CREB-BDNF activation, and decreased protein levels of neurofilament, spinophilin and PSD95. Notably, hippocampal-dependent spatial learning and memory was impaired in juvenile (1 Mon) and adulthood (6 Mon) KO rats. Taken together, we demonstrated that BDE2 plays a pivotal role for hippocampal neurogenesis, as well as learning and memory during female aging, especially in juvenile and middle age.
Collapse
Affiliation(s)
- Yuanyuan Huang
- Neurobiology Institute, School of Public Health, North China University of Science and Technology, Tangshan 063210, China
- School of Basic Medical Science, North China University of Science and Technology, Tangshan 063210, China
| | - Wuxiang Sun
- Neurobiology Institute, School of Public Health, North China University of Science and Technology, Tangshan 063210, China
- School of Basic Medical Science, North China University of Science and Technology, Tangshan 063210, China
| | - Fujia Gao
- Neurobiology Institute, School of Public Health, North China University of Science and Technology, Tangshan 063210, China
| | - Haoran Ma
- Neurobiology Institute, School of Public Health, North China University of Science and Technology, Tangshan 063210, China
| | - Tao Yuan
- Neurobiology Institute, School of Public Health, North China University of Science and Technology, Tangshan 063210, China
| | - Zixuan Liu
- Neurobiology Institute, School of Public Health, North China University of Science and Technology, Tangshan 063210, China
- School of Basic Medical Science, North China University of Science and Technology, Tangshan 063210, China
| | - Huiyu Liu
- Neurobiology Institute, School of Public Health, North China University of Science and Technology, Tangshan 063210, China
| | - Jiewei Hu
- Neurobiology Institute, School of Public Health, North China University of Science and Technology, Tangshan 063210, China
- School of Basic Medical Science, North China University of Science and Technology, Tangshan 063210, China
| | - Jing Bai
- Neurobiology Institute, School of Public Health, North China University of Science and Technology, Tangshan 063210, China
- School of Basic Medical Science, North China University of Science and Technology, Tangshan 063210, China
| | - Xin Zhang
- Neurobiology Institute, School of Public Health, North China University of Science and Technology, Tangshan 063210, China
| | - Ruimin Wang
- Neurobiology Institute, School of Public Health, North China University of Science and Technology, Tangshan 063210, China
- School of Basic Medical Science, North China University of Science and Technology, Tangshan 063210, China
| |
Collapse
|
9
|
Abbate C. The Adult Neurogenesis Theory of Alzheimer's Disease. J Alzheimers Dis 2023:JAD221279. [PMID: 37182879 DOI: 10.3233/jad-221279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/16/2023]
Abstract
Alzheimer's disease starts in neural stem cells (NSCs) in the niches of adult neurogenesis. All primary factors responsible for pathological tau hyperphosphorylation are inherent to adult neurogenesis and migration. However, when amyloid pathology is present, it strongly amplifies tau pathogenesis. Indeed, the progressive accumulation of extracellular amyloid-β deposits in the brain triggers a state of chronic inflammation by microglia. Microglial activation has a significant pro-neurogenic effect that fosters the process of adult neurogenesis and supports neuronal migration. Unfortunately, this "reactive" pro-neurogenic activity ultimately perturbs homeostatic equilibrium in the niches of adult neurogenesis by amplifying tau pathogenesis in AD. This scenario involves NSCs in the subgranular zone of the hippocampal dentate gyrus in late-onset AD (LOAD) and NSCs in the ventricular-subventricular zone along the lateral ventricles in early-onset AD (EOAD), including familial AD (FAD). Neuroblasts carrying the initial seed of tau pathology travel throughout the brain via neuronal migration driven by complex signals and convey the disease from the niches of adult neurogenesis to near (LOAD) or distant (EOAD) brain regions. In these locations, or in close proximity, a focus of degeneration begins to develop. Then, tau pathology spreads from the initial foci to large neuronal networks along neural connections through neuron-to-neuron transmission.
Collapse
Affiliation(s)
- Carlo Abbate
- IRCCS Fondazione Don Carlo Gnocchi ONLUS, Milan, Italy
| |
Collapse
|
10
|
Geribaldi-Doldán N, Carrascal L, Pérez-García P, Oliva-Montero JM, Pardillo-Díaz R, Domínguez-García S, Bernal-Utrera C, Gómez-Oliva R, Martínez-Ortega S, Verástegui C, Nunez-Abades P, Castro C. Migratory Response of Cells in Neurogenic Niches to Neuronal Death: The Onset of Harmonic Repair? Int J Mol Sci 2023; 24:6587. [PMID: 37047560 PMCID: PMC10095545 DOI: 10.3390/ijms24076587] [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: 03/02/2023] [Revised: 03/23/2023] [Accepted: 03/27/2023] [Indexed: 04/05/2023] Open
Abstract
Harmonic mechanisms orchestrate neurogenesis in the healthy brain within specific neurogenic niches, which generate neurons from neural stem cells as a homeostatic mechanism. These newly generated neurons integrate into existing neuronal circuits to participate in different brain tasks. Despite the mechanisms that protect the mammalian brain, this organ is susceptible to many different types of damage that result in the loss of neuronal tissue and therefore in alterations in the functionality of the affected regions. Nevertheless, the mammalian brain has developed mechanisms to respond to these injuries, potentiating its capacity to generate new neurons from neural stem cells and altering the homeostatic processes that occur in neurogenic niches. These alterations may lead to the generation of new neurons within the damaged brain regions. Notwithstanding, the activation of these repair mechanisms, regeneration of neuronal tissue within brain injuries does not naturally occur. In this review, we discuss how the different neurogenic niches respond to different types of brain injuries, focusing on the capacity of the progenitors generated in these niches to migrate to the injured regions and activate repair mechanisms. We conclude that the search for pharmacological drugs that stimulate the migration of newly generated neurons to brain injuries may result in the development of therapies to repair the damaged brain tissue.
Collapse
Affiliation(s)
- Noelia Geribaldi-Doldán
- Departamento de Anatomía y Embriología Humanas, Facultad de Medicina, Universidad de Cádiz, 11003 Cádiz, Spain
- Instituto de Investigación e Innovación Biomédica de Cádiz (INiBICA), 11009 Cádiz, Spain
| | - Livia Carrascal
- Instituto de Investigación e Innovación Biomédica de Cádiz (INiBICA), 11009 Cádiz, Spain
- Departamento de Fisiología, Facultad de Farmacia, Universidad de Sevilla, 41012 Sevilla, Spain
| | - Patricia Pérez-García
- Instituto de Investigación e Innovación Biomédica de Cádiz (INiBICA), 11009 Cádiz, Spain
- Departamento de Biomedicina, Biotecnología y Salud Pública, Área de Fisiología, Facultad de Medicina, Universidad de Cádiz, 11003 Cádiz, Spain
| | - José M. Oliva-Montero
- Instituto de Investigación e Innovación Biomédica de Cádiz (INiBICA), 11009 Cádiz, Spain
- Departamento de Biomedicina, Biotecnología y Salud Pública, Área de Fisiología, Facultad de Medicina, Universidad de Cádiz, 11003 Cádiz, Spain
| | - Ricardo Pardillo-Díaz
- Instituto de Investigación e Innovación Biomédica de Cádiz (INiBICA), 11009 Cádiz, Spain
- Departamento de Biomedicina, Biotecnología y Salud Pública, Área de Fisiología, Facultad de Medicina, Universidad de Cádiz, 11003 Cádiz, Spain
| | - Samuel Domínguez-García
- Instituto de Investigación e Innovación Biomédica de Cádiz (INiBICA), 11009 Cádiz, Spain
- Departamento de Biomedicina, Biotecnología y Salud Pública, Área de Fisiología, Facultad de Medicina, Universidad de Cádiz, 11003 Cádiz, Spain
- Department of Neuroscience, Karolinska Institutet, Biomedicum, 17177 Stockholm, Sweden
| | - Carlos Bernal-Utrera
- Instituto de Investigación e Innovación Biomédica de Cádiz (INiBICA), 11009 Cádiz, Spain
- Departamento de Fisioterapia, Facultad de Enfermería, Fisioterapia y Podología, Universidad de Sevilla, 41009 Sevilla, Spain
| | - Ricardo Gómez-Oliva
- Instituto de Investigación e Innovación Biomédica de Cádiz (INiBICA), 11009 Cádiz, Spain
- Departamento de Biomedicina, Biotecnología y Salud Pública, Área de Fisiología, Facultad de Medicina, Universidad de Cádiz, 11003 Cádiz, Spain
| | - Sergio Martínez-Ortega
- Instituto de Investigación e Innovación Biomédica de Cádiz (INiBICA), 11009 Cádiz, Spain
- Departamento de Biomedicina, Biotecnología y Salud Pública, Área de Fisiología, Facultad de Medicina, Universidad de Cádiz, 11003 Cádiz, Spain
| | - Cristina Verástegui
- Departamento de Anatomía y Embriología Humanas, Facultad de Medicina, Universidad de Cádiz, 11003 Cádiz, Spain
- Instituto de Investigación e Innovación Biomédica de Cádiz (INiBICA), 11009 Cádiz, Spain
| | - Pedro Nunez-Abades
- Instituto de Investigación e Innovación Biomédica de Cádiz (INiBICA), 11009 Cádiz, Spain
- Departamento de Fisiología, Facultad de Farmacia, Universidad de Sevilla, 41012 Sevilla, Spain
| | - Carmen Castro
- Instituto de Investigación e Innovación Biomédica de Cádiz (INiBICA), 11009 Cádiz, Spain
- Departamento de Biomedicina, Biotecnología y Salud Pública, Área de Fisiología, Facultad de Medicina, Universidad de Cádiz, 11003 Cádiz, Spain
| |
Collapse
|
11
|
Post-Stroke Environmental Enrichment Improves Neurogenesis and Cognitive Function and Reduces the Generation of Aberrant Neurons in the Mouse Hippocampus. Cells 2023; 12:cells12040652. [PMID: 36831319 PMCID: PMC9954243 DOI: 10.3390/cells12040652] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Revised: 02/08/2023] [Accepted: 02/09/2023] [Indexed: 02/22/2023] Open
Abstract
Ischemic lesions stimulate adult neurogenesis in the dentate gyrus, however, this is not associated with better cognitive function. Furthermore, increased neurogenesis is associated with the formation of aberrant neurons. In a previous study, we showed that a running task after a stroke not only increases neurogenesis but also the number of aberrant neurons without improving general performance. Here, we determined whether stimulation in an enriched environment after a lesion could increase neurogenesis and cognitive function without enhancing the number of aberrant neurons. After an ischemic stroke induced by MCAO, animals were transferred to an enriched environment containing a running wheel, tunnels and nest materials. A GFP-retroviral vector was delivered on day 3 post-stroke and a modified water maze test was performed 6 weeks after the lesion. We found that the enriched environment significantly increased the number of new neurons compared with the unstimulated stroke group but not the number of aberrant cells after a lesion. Increased neurogenesis after environmental enrichment was associated with improved cognitive function. Our study showed that early placement in an enriched environment after a stroke lesion markedly increased neurogenesis and flexible learning but not the formation of aberrant neurons, indicating that rehabilitative training, as a combination of running wheel training and enriched environment housing, improved functional and structural outcomes after a stroke.
Collapse
|
12
|
Song C, Chen X, Ma J, Buhe H, Liu Y, Saiyin H, Ma L. Construction of a pancreatic cancer nerve invasion system using brain and pancreatic cancer organoids. J Tissue Eng 2023; 14:20417314221147113. [PMID: 36636100 PMCID: PMC9829995 DOI: 10.1177/20417314221147113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Accepted: 12/08/2022] [Indexed: 01/09/2023] Open
Abstract
Pancreatic cancer (PC) is a fatal malignancy in the human abdominal cavity that prefers to invade the surrounding nerve/nerve plexus and even the spine, causing devastating and unbearable pain. The limitation of available in vitro models restricts revealing the molecular mechanism of pain and screening pain-relieving strategies to improve the quality of life of end-stage PC patients. Here, we report a PC nerve invasion model that merged human brain organoids (hBrO) with mouse PC organoids (mPCO). After merging hBrOs with mPCOs, we monitored the structural crosstalk, growth patterns, and mutual interaction dynamics of hBrO with mPCOs for 7 days. After 7 days, we also analyzed the pathophysiological statuses, including proliferation, apoptosis and inflammation. The results showed that mPCOs tend to approximate and intrude into the hBrOs, merge entirely into the hBrOs, and induce the retraction/shrinking of neuronal projections that protrude from the margin of the hBrOs. The approximating of mPCOs to hBrOs accelerated the proliferation of neuronal progenitor cells, intensified the apoptosis of neurons in the hBrOs, and increased the expression of inflammatory molecules in hBrOs, including NLRP3, IL-8, and IL-1β. Our system pathophysiologically replicated the nerve invasions in mouse GEMM (genetically engineered mouse model) primary and human PCs and might have the potential to be applied to reveal the molecular mechanism of nerve invasion and screen therapeutic strategies in PCs.
Collapse
Affiliation(s)
- Chenyun Song
- Department of Anatomy, Histology &
Embryology, School of Basic Medical Science, Fudan University, Shanghai, People’s
Republic of China
| | - Xinyu Chen
- Department of Anatomy, Histology &
Embryology, School of Basic Medical Science, Fudan University, Shanghai, People’s
Republic of China
| | - Jixin Ma
- Department of Anatomy, Histology &
Embryology, School of Basic Medical Science, Fudan University, Shanghai, People’s
Republic of China
| | - Hada Buhe
- The School of Pharmacy, Fujian Medical
University, Fuzhou, People’s Republic of China
| | - Yang Liu
- Department of Anatomy, Histology &
Embryology, School of Basic Medical Science, Fudan University, Shanghai, People’s
Republic of China
| | - Hexige Saiyin
- State Key Laboratory of Genetic
Engineering, School of Life Sciences, Fudan University, Shanghai, People’s Republic
of China,Hexige Saiyin, State Key Laboratory of
Genetic Engineering, School of Life Sciences, Fudan University, Songhu Road,
Shanghai 200438, People’s Republic of China.
| | - Lixiang Ma
- Department of Anatomy, Histology &
Embryology, School of Basic Medical Science, Fudan University, Shanghai, People’s
Republic of China
| |
Collapse
|
13
|
Baazaoui N, Iqbal K. Alzheimer's Disease: Challenges and a Therapeutic Opportunity to Treat It with a Neurotrophic Compound. Biomolecules 2022; 12:biom12101409. [PMID: 36291618 PMCID: PMC9599095 DOI: 10.3390/biom12101409] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 09/27/2022] [Accepted: 09/28/2022] [Indexed: 11/25/2022] Open
Abstract
Alzheimer’s disease (AD) is a progressive neurodegenerative disease with an insidious onset and multifactorial nature. A deficit in neurogenesis and synaptic plasticity are considered the early pathological features associated with neurofibrillary tau and amyloid β pathologies and neuroinflammation. The imbalance of neurotrophic factors with an increase in FGF-2 level and a decrease in brain derived neurotrophic factor (BDNF) and neurotrophin 4 (NT-4) in the hippocampus, frontal cortex and parietal cortex and disruption of the brain micro-environment are other characteristics of AD. Neurotrophic factors are crucial in neuronal differentiation, maturation, and survival. Several attempts to use neurotrophic factors to treat AD were made, but these trials were halted due to their blood-brain barrier (BBB) impermeability, short-half-life, and severe side effects. In the present review we mainly focus on the major etiopathology features of AD and the use of a small neurotrophic and neurogenic peptide mimetic compound; P021 that was discovered in our laboratory and was found to overcome the difficulties faced in the administration of the whole neurotrophic factor proteins. We describe pre-clinical studies on P021 and its potential as a therapeutic drug for AD and related neurodegenerative disorders. Our study is limited because it focuses only on P021 and the relevant literature; a more thorough investigation is required to review studies on various therapeutic approaches and potential drugs that are emerging in the AD field.
Collapse
Affiliation(s)
- Narjes Baazaoui
- Biology Department, College of Sciences and Arts Muhayil Assir, King Khalid University, Abha 61421, Saudi Arabia
| | - Khalid Iqbal
- Department of Neurochemistry, Inge Grundke-Iqbal Research Floor, New York State Institute for Basic Research in Developmental Disabilities, 1050 Forest Hill Road, Staten Island, NY 10314, USA
- Correspondence: ; Tel.: +1-718-494-5259; Fax: +1-718-494-1080
| |
Collapse
|
14
|
Rastegar-moghaddam SH, Ebrahimzadeh-Bideskan A, Shahba S, Malvandi AM, Mohammadipour A. MicroRNA-22: a Novel and Potent Biological Therapeutics in Neurological Disorders. Mol Neurobiol 2022; 59:2694-2701. [DOI: 10.1007/s12035-022-02769-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Accepted: 02/07/2022] [Indexed: 01/03/2023]
|
15
|
Chen P, Chen F, Wu Y, Zhou B. New Insights Into the Role of Aberrant Hippocampal Neurogenesis in Epilepsy. Front Neurol 2022; 12:727065. [PMID: 34975709 PMCID: PMC8714646 DOI: 10.3389/fneur.2021.727065] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Accepted: 11/02/2021] [Indexed: 12/31/2022] Open
Abstract
Data accumulated over the past four decades have confirmed that adult hippocampal neurogenesis (HN) plays a key role in the wide spectrum of hippocampal pathology. Epilepsy is a disorder of the central nervous system characterized by spontaneous recurrent seizures. Although neurogenesis in persistent germinative zones is altered in the adult rodent models of epilepsy, the effects of seizure-induced neurogenesis in the epileptic brain, in terms of either a pathological or reparative role, are only beginning to be explored. In this review, we described the most recent advances in neurogenesis in epilepsy and outlooked future directions for neural stem cells (NSCs) and epilepsy-in-a-dish models. We proposed that it may help in refining the underlying molecular mechanisms of epilepsy and improving the therapies and precision medicine for patients with epilepsy.
Collapse
Affiliation(s)
- Peng Chen
- Department of Pharmacy, Renmin Hospital of Wuhan University, Wuhan, China.,Key Laboratory of Combinatorial Biosynthesis and Drug Discovery, Ministry of Education, Wuhan University School of Pharmaceutical Sciences, Wuhan, China
| | - Fuchao Chen
- Sinopharm Dongfeng General Hospital, Hubei University of Medicine, Shiyan, China
| | - Yue Wu
- Department of Pharmacy, Renmin Hospital of Wuhan University, Wuhan, China
| | - Benhong Zhou
- Department of Pharmacy, Renmin Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
16
|
Chen A, Chen X, Deng J, Zheng X. Research advances in the role of endogenous neurogenesis on neonatal hypoxic-ischemic brain damage. Front Pediatr 2022; 10:986452. [PMID: 36299701 PMCID: PMC9589272 DOI: 10.3389/fped.2022.986452] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Accepted: 09/20/2022] [Indexed: 11/13/2022] Open
Abstract
Hypoxic-ischemic brain damage (HIBD) is the main cause of perinatal mortality and neurologic complications in neonates, but it remains difficult to cure due to scarce treatments and complex molecular mechanisms remaining incompletely explained. Recent, mounting evidence shows that endogenous neurogenesis can improve neonatal neurological dysfunction post-HIBD. However, the capacity for spontaneous endogenous neurogenesis is limited and insufficient for replacing neurons lost to brain damage. Therefore, it is of great clinical value and social significance to seek therapeutic techniques that promote endogenous neurogenesis, to reduce neonatal neurological dysfunction from HIBD. This review summarizes the known neuroprotective effects of, and treatments targeting, endogenous neurogenesis following neonatal HIBD, to provide available targets and directions and a theoretical basis for the treatment of neonatal neurological dysfunction from HIBD.
Collapse
Affiliation(s)
- Andi Chen
- Department of Anesthesiology, Shengli Clinical Medical College of Fujian Medical University, Fujian Provincial Hospital, Fuzhou, China
| | - Xiaohui Chen
- Department of Anesthesiology, Shengli Clinical Medical College of Fujian Medical University, Fujian Provincial Hospital, Fuzhou, China
| | - Jianhui Deng
- Department of Anesthesiology, Shengli Clinical Medical College of Fujian Medical University, Fujian Provincial Hospital, Fuzhou, China
| | - Xiaochun Zheng
- Department of Anesthesiology, Shengli Clinical Medical College of Fujian Medical University, Fujian Provincial Hospital, Fuzhou, China.,Fujian Emergency Medical Center, Fujian Provincial Key Laboratory of Emergency Medicine, Fujian Provincial Key Laboratory of Critical Care Medicine, Fujian Provincial Co-Constructed Laboratory of "Belt and Road", Fuzhou, China
| |
Collapse
|
17
|
Leschik J, Lutz B, Gentile A. Stress-Related Dysfunction of Adult Hippocampal Neurogenesis-An Attempt for Understanding Resilience? Int J Mol Sci 2021; 22:7339. [PMID: 34298958 PMCID: PMC8305135 DOI: 10.3390/ijms22147339] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 07/02/2021] [Accepted: 07/05/2021] [Indexed: 12/16/2022] Open
Abstract
Newborn neurons in the adult hippocampus are regulated by many intrinsic and extrinsic cues. It is well accepted that elevated glucocorticoid levels lead to downregulation of adult neurogenesis, which this review discusses as one reason why psychiatric diseases, such as major depression, develop after long-term stress exposure. In reverse, adult neurogenesis has been suggested to protect against stress-induced major depression, and hence, could serve as a resilience mechanism. In this review, we will summarize current knowledge about the functional relation of adult neurogenesis and stress in health and disease. A special focus will lie on the mechanisms underlying the cascades of events from prolonged high glucocorticoid concentrations to reduced numbers of newborn neurons. In addition to neurotransmitter and neurotrophic factor dysregulation, these mechanisms include immunomodulatory pathways, as well as microbiota changes influencing the gut-brain axis. Finally, we discuss recent findings delineating the role of adult neurogenesis in stress resilience.
Collapse
Affiliation(s)
- Julia Leschik
- Institute of Physiological Chemistry, University Medical Center of the Johannes Gutenberg University Mainz, 55128 Mainz, Germany;
| | - Beat Lutz
- Institute of Physiological Chemistry, University Medical Center of the Johannes Gutenberg University Mainz, 55128 Mainz, Germany;
- Leibniz Institute for Resilience Research (LIR), 55122 Mainz, Germany
| | - Antonietta Gentile
- Synaptic Immunopathology Lab, IRCCS San Raffaele Pisana, 00166 Rome, Italy;
| |
Collapse
|
18
|
Shishkina GT, Kalinina TS, Gulyaeva NV, Lanshakov DA, Dygalo NN. Changes in Gene Expression and Neuroinflammation in the Hippocampus after Focal Brain Ischemia: Involvement in the Long-Term Cognitive and Mental Disorders. BIOCHEMISTRY. BIOKHIMIIA 2021; 86:657-666. [PMID: 34225589 DOI: 10.1134/s0006297921060043] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Ischemic brain injuries are accompanied by the long-term changes in gene expression in the hippocampus, the limbic system structure, involved in the regulation of key aspects of the higher nervous activity, such as cognitive functions and emotions. The altered expression of genes and proteins encoded by them may be related to the development of post-ischemic psycho-emotional and cognitive disturbances. Activation of neuroinflammation following stroke in the hippocampus has been suggested to play an essential role in induction of long-lasting consequences. Identification of changes in the gene expression patterns after ischemia and investigation of the dynamics of these changes in the hippocampus are the necessary first steps toward understanding molecular pathways responsible for the development of post-stroke cognitive impairments and mental pathologies.
Collapse
Affiliation(s)
- Galina T Shishkina
- Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Sciences, Novosibirsk, 630090, Russia.
| | - Tatiana S Kalinina
- Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Sciences, Novosibirsk, 630090, Russia
| | - Natalia V Gulyaeva
- Institute of Higher Nervous Activity and Neurophysiology, Russian Academy of Sciences, Moscow, 117485, Russia
| | - Dmitry A Lanshakov
- Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Sciences, Novosibirsk, 630090, Russia
| | - Nikolay N Dygalo
- Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Sciences, Novosibirsk, 630090, Russia
| |
Collapse
|
19
|
Rudolph M, Schmeer CW, Günther M, Woitke F, Kathner-Schaffert C, Karapetow L, Lindner J, Lehmann T, Jirikowski G, Witte OW, Redecker C, Keiner S. Microglia-mediated phagocytosis of apoptotic nuclei is impaired in the adult murine hippocampus after stroke. Glia 2021; 69:2006-2022. [PMID: 33942391 DOI: 10.1002/glia.24009] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Revised: 04/07/2021] [Accepted: 04/08/2021] [Indexed: 01/02/2023]
Abstract
Following stroke, neuronal death takes place both in the infarct region and in brain areas distal to the lesion site including the hippocampus. The hippocampus is critically involved in learning and memory processes and continuously generates new neurons. Dysregulation of adult neurogenesis may be associated with cognitive decline after a stroke lesion. In particular, proliferation of precursor cells and the formation of new neurons are increased after lesion. Within the first week, many new precursor cells die during development. How dying precursors are removed from the hippocampus and to what extent phagocytosis takes place after stroke is still not clear. Here, we evaluated the effect of a prefrontal stroke lesion on the phagocytic activity of microglia in the dentate gyrus (DG) of the hippocampus. Three-months-old C57BL/6J mice were injected once with the proliferation marker BrdU (250 mg/kg) 6 hr after a middle cerebral artery occlusion or sham surgery. The number of apoptotic cells and the phagocytic capacity of the microglia were evaluated by means of immunohistochemistry, confocal microscopy, and 3D-reconstructions. We found a transient but significant increase in the number of apoptotic cells in the DG early after stroke, associated with impaired removal by microglia. Interestingly, phagocytosis of newly generated precursor cells was not affected. Our study shows that a prefrontal stroke lesion affects phagocytosis of apoptotic cells in the DG, a region distal to the lesion core. Whether disturbed phagocytosis might contribute to inflammatory- and maladaptive processes including cognitive impairment following stroke needs to be further investigated.
Collapse
Affiliation(s)
- Max Rudolph
- Hans-Berger Department of Neurology, Jena University Hospital, Jena, Germany
| | - Christian W Schmeer
- Hans-Berger Department of Neurology, Jena University Hospital, Jena, Germany
| | - Madlen Günther
- Hans-Berger Department of Neurology, Jena University Hospital, Jena, Germany
| | - Florus Woitke
- Hans-Berger Department of Neurology, Jena University Hospital, Jena, Germany
| | | | - Lina Karapetow
- Hans-Berger Department of Neurology, Jena University Hospital, Jena, Germany
| | - Julia Lindner
- Hans-Berger Department of Neurology, Jena University Hospital, Jena, Germany
| | - Thomas Lehmann
- Institute of Medical Statistics and Computer Science, University Hospital Jena, Friedrich Schiller University Jena, Jena, Germany
| | - Gustav Jirikowski
- Health and Medical University Potsdam, University Potsdam, Potsdam, Germany
| | - Otto W Witte
- Hans-Berger Department of Neurology, Jena University Hospital, Jena, Germany
| | - Christoph Redecker
- Hans-Berger Department of Neurology, Jena University Hospital, Jena, Germany.,Department of Neurology, Lippe General Hospital, Lemgo, Germany
| | - Silke Keiner
- Hans-Berger Department of Neurology, Jena University Hospital, Jena, Germany
| |
Collapse
|
20
|
Cuartero MI, García-Culebras A, Torres-López C, Medina V, Fraga E, Vázquez-Reyes S, Jareño-Flores T, García-Segura JM, Lizasoain I, Moro MÁ. Post-stroke Neurogenesis: Friend or Foe? Front Cell Dev Biol 2021; 9:657846. [PMID: 33834025 PMCID: PMC8021779 DOI: 10.3389/fcell.2021.657846] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2021] [Accepted: 02/26/2021] [Indexed: 12/18/2022] Open
Abstract
The substantial clinical burden and disability after stroke injury urges the need to explore therapeutic solutions. Recent compelling evidence supports that neurogenesis persists in the adult mammalian brain and is amenable to regulation in both physiological and pathological situations. Its ability to generate new neurons implies a potential to contribute to recovery after brain injury. However, post-stroke neurogenic response may have different functional consequences. On the one hand, the capacity of newborn neurons to replenish the damaged tissue may be limited. In addition, aberrant forms of neurogenesis have been identified in several insult settings. All these data suggest that adult neurogenesis is at a crossroads between the physiological and the pathological regulation of the neurological function in the injured central nervous system (CNS). Given the complexity of the CNS together with its interaction with the periphery, we ultimately lack in-depth understanding of the key cell types, cell-cell interactions, and molecular pathways involved in the neurogenic response after brain damage and their positive or otherwise deleterious impact. Here we will review the evidence on the stroke-induced neurogenic response and on its potential repercussions on functional outcome. First, we will briefly describe subventricular zone (SVZ) neurogenesis after stroke beside the main evidence supporting its positive role on functional restoration after stroke. Then, we will focus on hippocampal subgranular zone (SGZ) neurogenesis due to the relevance of hippocampus in cognitive functions; we will outline compelling evidence that supports that, after stroke, SGZ neurogenesis may adopt a maladaptive plasticity response further contributing to the development of post-stroke cognitive impairment and dementia. Finally, we will discuss the therapeutic potential of specific steps in the neurogenic cascade that might ameliorate brain malfunctioning and the development of post-stroke cognitive impairment in the chronic phase.
Collapse
Affiliation(s)
- María Isabel Cuartero
- Neurovascular Pathophysiology, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
- Unidad de Investigación Neurovascular, Departamento de Farmacología, Facultad de Medicina, Universidad Complutense de Madrid (UCM), Madrid, Spain
- Instituto Universitario de Investigación en Neuroquímica (IUIN), Universidad Complutense de Madrid (UCM), Madrid, Spain
| | - Alicia García-Culebras
- Neurovascular Pathophysiology, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
- Unidad de Investigación Neurovascular, Departamento de Farmacología, Facultad de Medicina, Universidad Complutense de Madrid (UCM), Madrid, Spain
- Instituto Universitario de Investigación en Neuroquímica (IUIN), Universidad Complutense de Madrid (UCM), Madrid, Spain
| | - Cristina Torres-López
- Neurovascular Pathophysiology, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
- Unidad de Investigación Neurovascular, Departamento de Farmacología, Facultad de Medicina, Universidad Complutense de Madrid (UCM), Madrid, Spain
- Instituto Universitario de Investigación en Neuroquímica (IUIN), Universidad Complutense de Madrid (UCM), Madrid, Spain
| | - Violeta Medina
- Neurovascular Pathophysiology, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
- Unidad de Investigación Neurovascular, Departamento de Farmacología, Facultad de Medicina, Universidad Complutense de Madrid (UCM), Madrid, Spain
- Instituto Universitario de Investigación en Neuroquímica (IUIN), Universidad Complutense de Madrid (UCM), Madrid, Spain
| | - Enrique Fraga
- Neurovascular Pathophysiology, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
- Unidad de Investigación Neurovascular, Departamento de Farmacología, Facultad de Medicina, Universidad Complutense de Madrid (UCM), Madrid, Spain
- Instituto Universitario de Investigación en Neuroquímica (IUIN), Universidad Complutense de Madrid (UCM), Madrid, Spain
| | - Sandra Vázquez-Reyes
- Neurovascular Pathophysiology, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
- Unidad de Investigación Neurovascular, Departamento de Farmacología, Facultad de Medicina, Universidad Complutense de Madrid (UCM), Madrid, Spain
- Instituto Universitario de Investigación en Neuroquímica (IUIN), Universidad Complutense de Madrid (UCM), Madrid, Spain
| | - Tania Jareño-Flores
- Neurovascular Pathophysiology, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
- Unidad de Investigación Neurovascular, Departamento de Farmacología, Facultad de Medicina, Universidad Complutense de Madrid (UCM), Madrid, Spain
- Instituto Universitario de Investigación en Neuroquímica (IUIN), Universidad Complutense de Madrid (UCM), Madrid, Spain
| | - Juan M. García-Segura
- Unidad de Investigación Neurovascular, Departamento de Farmacología, Facultad de Medicina, Universidad Complutense de Madrid (UCM), Madrid, Spain
- Departamento de Bioquímica y Biología Molecular, Facultad de Ciencias Químicas, Universidad Complutense de Madrid (UCM), Madrid, Spain
| | - Ignacio Lizasoain
- Unidad de Investigación Neurovascular, Departamento de Farmacología, Facultad de Medicina, Universidad Complutense de Madrid (UCM), Madrid, Spain
- Instituto Universitario de Investigación en Neuroquímica (IUIN), Universidad Complutense de Madrid (UCM), Madrid, Spain
- Instituto de Investigación Hospital 12 de Octubre (i+12), Madrid, Spain
| | - María Ángeles Moro
- Neurovascular Pathophysiology, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
- Unidad de Investigación Neurovascular, Departamento de Farmacología, Facultad de Medicina, Universidad Complutense de Madrid (UCM), Madrid, Spain
- Instituto Universitario de Investigación en Neuroquímica (IUIN), Universidad Complutense de Madrid (UCM), Madrid, Spain
- Instituto de Investigación Hospital 12 de Octubre (i+12), Madrid, Spain
| |
Collapse
|
21
|
Satta V, Alonso C, Díez P, Martín-Suárez S, Rubio M, Encinas JM, Fernández-Ruiz J, Sagredo O. Neuropathological Characterization of a Dravet Syndrome Knock-In Mouse Model Useful for Investigating Cannabinoid Treatments. Front Mol Neurosci 2021; 13:602801. [PMID: 33584198 PMCID: PMC7879984 DOI: 10.3389/fnmol.2020.602801] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Accepted: 11/30/2020] [Indexed: 12/24/2022] Open
Abstract
Dravet syndrome (DS) is an epileptic syndrome caused by mutations in the Scn1a gene encoding the α1 subunit of the sodium channel Nav1.1, which is associated with febrile seizures that progress to severe tonic-clonic seizures and associated comorbidities. Treatment with cannabidiol has been approved to reduce seizures in DS, but it may also be active against these comorbidities. The aim of this study was to validate a new mouse model of DS having lower mortality than previous models, which may serve to further evaluate therapies for the long-term comorbidities. This new model consists of heterozygous conditional knock-in mice carrying a missense mutation (A1783V) in Scn1a gene expressed exclusively in neurons of the CNS (Syn-Cre/Scn1aWT/A1783V). These mice have been used here to determine the extent and persistence of the behavioral deterioration in different postnatal days (PND), as well as to investigate the alterations that the disease produces in the endocannabinoid system and the contribution of inflammatory events and impaired neurogenesis in the pathology. Syn-Cre/Scn1aWT/A1783V mice showed a strong reduction in hindlimb grasp reflex at PND10, whereas at PND25, they presented spontaneous convulsions and a greater susceptibility to pentylenetetrazole-induced seizures, marked hyperactivity, deficient spatial working memory, lower levels of anxiety, and altered social interaction behavior. These differences disappeared at PND40 and PND60, except the changes in social interaction and anxiety. The analysis of CNS structures associated with these behavioral alterations revealed an elevated glial reactivity in the prefrontal cortex and the dentate gyrus. This was associated in the dentate gyrus with a greater cell proliferation detected with Ki67 immunostaining, whereas double-labeling analyses identified that proliferating cells were GFAP-positive suggesting failed neurogenesis but astrocyte proliferation. The analysis of the endocannabinoid system of Syn-Cre/Scn1aWT/A1783V mice confirmed reductions in CB1 receptors and MAGL and FAAH enzymes, mainly in the cerebellum but also in other areas, whereas CB2 receptors became upregulated in the hippocampus. In conclusion, Syn-Cre/Scn1aWT/A1783V mice showed seizuring susceptibility and several comorbidities (hyperactivity, memory impairment, less anxiety, and altered social behavior), which exhibited a pattern of age expression similar to DS patients. Syn-Cre/Scn1aWT/A1783V mice also exhibited greater glial reactivity and a reactive response in the neurogenic niche, and regional changes in the status of the endocannabinoid signaling, events that could contribute in behavioral impairment.
Collapse
Affiliation(s)
- Valentina Satta
- Instituto Universitario de Investigación en Neuroquímica, Departamento de Bioquímica y Biología Molecular, Facultad de Medicina, Universidad Complutense, Madrid, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain.,Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, Spain
| | - Cristina Alonso
- Instituto Universitario de Investigación en Neuroquímica, Departamento de Bioquímica y Biología Molecular, Facultad de Medicina, Universidad Complutense, Madrid, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain.,Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, Spain
| | - Paula Díez
- Instituto Universitario de Investigación en Neuroquímica, Departamento de Bioquímica y Biología Molecular, Facultad de Medicina, Universidad Complutense, Madrid, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain.,Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, Spain
| | - Soraya Martín-Suárez
- The NSC Cell and Neurogenesis Laboratory, Achucarro Basque Center for Neuroscience, Leioa, Spain
| | - Marta Rubio
- Instituto Universitario de Investigación en Neuroquímica, Departamento de Bioquímica y Biología Molecular, Facultad de Medicina, Universidad Complutense, Madrid, Spain
| | - Juan M Encinas
- The NSC Cell and Neurogenesis Laboratory, Achucarro Basque Center for Neuroscience, Leioa, Spain.,The University of the Basque Country (UPV/EHU), Leioa, Spain.,IKERBASQUE, The Basque Foundation for Science, Bilbao, Spain
| | - Javier Fernández-Ruiz
- Instituto Universitario de Investigación en Neuroquímica, Departamento de Bioquímica y Biología Molecular, Facultad de Medicina, Universidad Complutense, Madrid, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain.,Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, Spain
| | - Onintza Sagredo
- Instituto Universitario de Investigación en Neuroquímica, Departamento de Bioquímica y Biología Molecular, Facultad de Medicina, Universidad Complutense, Madrid, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain.,Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, Spain
| |
Collapse
|
22
|
Clark LR, Yun S, Acquah NK, Kumar PL, Metheny HE, Paixao RCC, Cohen AS, Eisch AJ. Mild Traumatic Brain Injury Induces Transient, Sequential Increases in Proliferation, Neuroblasts/Immature Neurons, and Cell Survival: A Time Course Study in the Male Mouse Dentate Gyrus. Front Neurosci 2021; 14:612749. [PMID: 33488351 PMCID: PMC7817782 DOI: 10.3389/fnins.2020.612749] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Accepted: 12/02/2020] [Indexed: 12/17/2022] Open
Abstract
Mild traumatic brain injuries (mTBIs) are prevalent worldwide. mTBIs can impair hippocampal-based functions such as memory and cause network hyperexcitability of the dentate gyrus (DG), a key entry point to hippocampal circuitry. One candidate for mediating mTBI-induced hippocampal cognitive and physiological dysfunction is injury-induced changes in the process of DG neurogenesis. There are conflicting results on how TBI impacts the process of DG neurogenesis; this is not surprising given that both the neurogenesis process and the post-injury period are dynamic, and that the quantification of neurogenesis varies widely in the literature. Even within the minority of TBI studies focusing specifically on mild injuries, there is disagreement about if and how mTBI changes the process of DG neurogenesis. Here we utilized a clinically relevant rodent model of mTBI (lateral fluid percussion injury, LFPI), gold-standard markers and quantification of the neurogenesis process, and three time points post-injury to generate a comprehensive picture of how mTBI affects adult hippocampal DG neurogenesis. Male C57BL/6J mice (6-8 weeks old) received either sham surgery or mTBI via LFPI. Proliferating cells, neuroblasts/immature neurons, and surviving cells were quantified via stereology in DG subregions (subgranular zone [SGZ], outer granule cell layer [oGCL], molecular layer, and hilus) at short-term (3 days post-injury, dpi), intermediate (7 dpi), and long-term (31 dpi) time points. The data show this model of mTBI induces transient, sequential increases in ipsilateral SGZ/GCL proliferating cells, neuroblasts/immature neurons, and surviving cells which is suggestive of mTBI-induced neurogenesis. In contrast to these ipsilateral hemisphere findings, measures in the contralateral hemisphere were not increased in key neurogenic DG subregions after LFPI. Our work in this mTBI model is in line with most literature on other and more severe models of TBI in showing TBI stimulates the process of DG neurogenesis. However, as our DG data in mTBI provide temporal, subregional, and neurogenesis-stage resolution, these data are important to consider in regard to the functional importance of TBI-induction of the neurogenesis process and future work assessing the potential of replacing and/or repairing DG neurons in the brain after TBI.
Collapse
Affiliation(s)
- Lyles R. Clark
- Department of Anesthesiology and Critical Care Medicine, The Children’s Hospital of Philadelphia (CHOP) Research Institute, Philadelphia, PA, United States
- Mahoney Institute for Neurosciences, Perelman School of Medicine, Philadelphia, PA, United States
| | - Sanghee Yun
- Department of Anesthesiology and Critical Care Medicine, The Children’s Hospital of Philadelphia (CHOP) Research Institute, Philadelphia, PA, United States
- Mahoney Institute for Neurosciences, Perelman School of Medicine, Philadelphia, PA, United States
| | - Nana K. Acquah
- Department of Anesthesiology and Critical Care Medicine, The Children’s Hospital of Philadelphia (CHOP) Research Institute, Philadelphia, PA, United States
- Biological Basis of Behavior Program, University of Pennsylvania, Philadelphia, PA, United States
| | - Priya L. Kumar
- Department of Anesthesiology and Critical Care Medicine, The Children’s Hospital of Philadelphia (CHOP) Research Institute, Philadelphia, PA, United States
- Biomechanical Engineering, University of Pennsylvania, Philadelphia, PA, United States
| | - Hannah E. Metheny
- Department of Anesthesiology and Critical Care Medicine, The Children’s Hospital of Philadelphia (CHOP) Research Institute, Philadelphia, PA, United States
| | - Rikley C. C. Paixao
- Department of Anesthesiology and Critical Care Medicine, The Children’s Hospital of Philadelphia (CHOP) Research Institute, Philadelphia, PA, United States
| | - Akivas S. Cohen
- Department of Anesthesiology and Critical Care Medicine, The Children’s Hospital of Philadelphia (CHOP) Research Institute, Philadelphia, PA, United States
- Mahoney Institute for Neurosciences, Perelman School of Medicine, Philadelphia, PA, United States
| | - Amelia J. Eisch
- Department of Anesthesiology and Critical Care Medicine, The Children’s Hospital of Philadelphia (CHOP) Research Institute, Philadelphia, PA, United States
- Mahoney Institute for Neurosciences, Perelman School of Medicine, Philadelphia, PA, United States
| |
Collapse
|
23
|
Podgorny OV, Gulyaeva NV. Glucocorticoid-mediated mechanisms of hippocampal damage: Contribution of subgranular neurogenesis. J Neurochem 2020; 157:370-392. [PMID: 33301616 DOI: 10.1111/jnc.15265] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Revised: 11/09/2020] [Accepted: 11/30/2020] [Indexed: 12/19/2022]
Abstract
A comprehensive overview of the interplay between glucocorticoids (GCs) and adult hippocampal neurogenesis (AHN) is presented, particularly, in the context of a diseased brain. The effectors of GCs in the dentate gyrus neurogenic niche of the hippocampal are reviewed, and the consequences of the GC signaling on the generation and integration of new neurons are discussed. Recent findings demonstrating how GC signaling mediates impairments of the AHN in various brain pathologies are overviewed. GC-mediated effects on the generation and integration of adult-born neurons in the hippocampal dentate gyrus depend on the nature, severity, and duration of the acting stress factor. GCs realize their effects on the AHN primarily via specific glucocorticoid and mineralocorticoid receptors. Disruption of the reciprocal regulation between the hypothalamic-pituitary-adrenal (HPA) axis and the generation of the adult-born granular neurons is currently considered to be a key mechanism implicating the AHN into the pathogenesis of numerous brain diseases, including those without a direct hippocampal damage. These alterations vary from reduced proliferation of stem and progenitor cells to increased cell death and abnormalities in morphology, connectivity, and localization of young neurons. Although the involvement of the mutual regulation between the HPA axis and the AHN in the pathogenesis of cognitive deficits and mood impairments is evident, several unresolved critical issues are stated. Understanding the details of GC-mediated mechanisms involved in the alterations in AHN could enable the identification of molecular targets for ameliorating pathology-induced imbalance in the HPA axis/AHN mutual regulation to conquer cognitive and psychiatric disturbances.
Collapse
Affiliation(s)
- Oleg V Podgorny
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia.,Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Pirogov Russian National Research Medical University, Moscow, Russia.,Koltzov Institute of Developmental Biology, Russian Academy of Sciences, Moscow, Russia
| | - Natalia V Gulyaeva
- Institute of Higher Nervous Activity and Neurophysiology, Russian Academy of Sciences, Moscow, Russia.,Research and Clinical Center for Neuropsychiatry of Moscow Healthcare Department, Moscow, Russia
| |
Collapse
|
24
|
Nickell CG, Thompson KR, Pauly JR, Nixon K. Recovery of Hippocampal-Dependent Learning Despite Blunting Reactive Adult Neurogenesis After Alcohol Dependence. Brain Plast 2020; 6:83-101. [PMID: 33680848 PMCID: PMC7903006 DOI: 10.3233/bpl-200108] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Background: The excessive alcohol drinking that occurs in alcohol use disorder (AUD) causes neurodegeneration in regions such as the hippocampus, though recovery may occur after a period of abstinence. Mechanisms of recovery are not clear, though reactive neurogenesis has been observed in the hippocampal dentate gyrus following alcohol dependence and correlates to recovery of granule cell number. Objective: We investigated the role of neurons born during reactive neurogenesis in the recovery of hippocampal learning behavior after 4-day binge alcohol exposure, a model of an AUD. We hypothesized that reducing reactive neurogenesis would impair functional recovery. Methods: Adult male rats were subjected to 4-day binge alcohol exposure and two approaches were tested to blunt reactive adult neurogenesis, acute doses of alcohol or the chemotherapy drug, temozolomide (TMZ). Results: Acute 5 g/kg doses of EtOH gavaged T6 and T7 days post binge did not inhibit significantly the number of Bromodeoxyuridine-positive (BrdU+) proliferating cells in EtOH animals receiving 5 g/kg EtOH versus controls. A single cycle of TMZ inhibited reactive proliferation (BrdU+ cells) and neurogenesis (NeuroD+ cells) to that of controls. However, despite this blunting of reactive neurogenesis to basal levels, EtOH-TMZ rats were not impaired in their recovery of acquisition of the Morris water maze (MWM), learning similarly to all other groups 35 days after 4-day binge exposure. Conclusions: These studies show that TMZ is effective in decreasing reactive proliferation/neurogenesis following 4-day binge EtOH exposure, and baseline levels of adult neurogenesis are sufficient to allow recovery of hippocampal function.
Collapse
Affiliation(s)
- Chelsea G Nickell
- University of Kentucky, Department of Pharmaceutical Sciences, Lexington, KY, USA
| | - K Ryan Thompson
- The University of Texas at Austin, College of Pharmacy, Austin, TX, USA
| | - James R Pauly
- University of Kentucky, Department of Pharmaceutical Sciences, Lexington, KY, USA
| | - Kimberly Nixon
- University of Kentucky, Department of Pharmaceutical Sciences, Lexington, KY, USA.,The University of Texas at Austin, College of Pharmacy, Austin, TX, USA
| |
Collapse
|
25
|
García-Cabrerizo R, Ledesma-Corvi S, Bis-Humbert C, García-Fuster MJ. Sex differences in the antidepressant-like potential of repeated electroconvulsive seizures in adolescent and adult rats: Regulation of the early stages of hippocampal neurogenesis. Eur Neuropsychopharmacol 2020; 41:132-145. [PMID: 33160794 DOI: 10.1016/j.euroneuro.2020.10.008] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Revised: 10/06/2020] [Accepted: 10/16/2020] [Indexed: 12/27/2022]
Abstract
Age and sex are critical factors for the diagnosis and treatment of major depression, since there is a well-known age-by-sex difference in the prevalence of major depression (being females the most vulnerable ones) and in antidepressant efficacy (being adolescence a less responsive period than adulthood). Although the induction of electroconvulsive seizures (ECS) is a very old technique in humans, there is not much evidence reporting sex- and age-specific aspects of this treatment. The present study evaluated the antidepressant- and neurogenic-like potential of repeated ECS across time in adolescent and adult rats (naïve or in a model of early life stress capable of mimicking a pro-depressive phenotype), while including a sex perspective. The main results demonstrated age- and sex-specific differences in the antidepressant-like potential of repeated ECS, since it worked when administered during adolescence or adulthood in male rats (although with a shorter length in adolescence), while in females rendered deleterious during adolescence and ineffective in adulthood. Yet, repeated ECS increased cell proliferation and vastly boosted young neuronal survival in a time-dependent manner for both sexes and independently of age. Moreover, pharmacological inhibition of basal cell proliferation prevented the antidepressant-like effect induced by repeated ECS in male rats, but only partially blocked the very robust increase in the initial cell markers of hippocampal neurogenesis. Overall, the present results suggest that the induction of the early phases of neurogenesis by ECS, besides having a role in mediating its antidepressant-like effect, might participate in some other neuroplastic actions, opening the path for future studies.
Collapse
Affiliation(s)
- Rubén García-Cabrerizo
- IUNICS, University of the Balearic Islands, Cra. de Valldemossa km 7.5, E-07122 Palma, Spain; Health Research Institute of the Balearic Islands (IdISBa), Palma, Spain; Present address: APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Sandra Ledesma-Corvi
- IUNICS, University of the Balearic Islands, Cra. de Valldemossa km 7.5, E-07122 Palma, Spain; Health Research Institute of the Balearic Islands (IdISBa), Palma, Spain
| | - Cristian Bis-Humbert
- IUNICS, University of the Balearic Islands, Cra. de Valldemossa km 7.5, E-07122 Palma, Spain; Health Research Institute of the Balearic Islands (IdISBa), Palma, Spain
| | - M Julia García-Fuster
- IUNICS, University of the Balearic Islands, Cra. de Valldemossa km 7.5, E-07122 Palma, Spain; Health Research Institute of the Balearic Islands (IdISBa), Palma, Spain.
| |
Collapse
|
26
|
Lourenço DM, Ribeiro-Rodrigues L, Sebastião AM, Diógenes MJ, Xapelli S. Neural Stem Cells and Cannabinoids in the Spotlight as Potential Therapy for Epilepsy. Int J Mol Sci 2020; 21:E7309. [PMID: 33022963 PMCID: PMC7582633 DOI: 10.3390/ijms21197309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Revised: 09/28/2020] [Accepted: 09/30/2020] [Indexed: 01/18/2023] Open
Abstract
Epilepsy is one of the most common brain diseases worldwide, having a huge burden in society. The main hallmark of epilepsy is the occurrence of spontaneous recurrent seizures, having a tremendous impact on the lives of the patients and of their relatives. Currently, the therapeutic strategies are mostly based on the use of antiepileptic drugs, and because several types of epilepsies are of unknown origin, a high percentage of patients are resistant to the available pharmacotherapy, continuing to experience seizures overtime. Therefore, the search for new drugs and therapeutic targets is highly important. One key aspect to be targeted is the aberrant adult hippocampal neurogenesis (AHN) derived from Neural Stem Cells (NSCs). Indeed, targeting seizure-induced AHN may reduce recurrent seizures and shed some light on the mechanisms of disease. The endocannabinoid system is a known modulator of AHN, and due to the known endogenous antiepileptic properties, it is an interesting candidate for the generation of new antiepileptic drugs. However, further studies and clinical trials are required to investigate the putative mechanisms by which cannabinoids can be used to treat epilepsy. In this manuscript, we will review how cannabinoid-induced modulation of NSCs may promote neural plasticity and whether these drugs can be used as putative antiepileptic treatment.
Collapse
Affiliation(s)
- Diogo M. Lourenço
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisboa, Portugal; (D.M.L.); (L.R.-R.); (A.M.S.); (M.J.D.)
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisboa, Portugal
| | - Leonor Ribeiro-Rodrigues
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisboa, Portugal; (D.M.L.); (L.R.-R.); (A.M.S.); (M.J.D.)
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisboa, Portugal
| | - Ana M. Sebastião
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisboa, Portugal; (D.M.L.); (L.R.-R.); (A.M.S.); (M.J.D.)
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisboa, Portugal
| | - Maria J. Diógenes
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisboa, Portugal; (D.M.L.); (L.R.-R.); (A.M.S.); (M.J.D.)
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisboa, Portugal
| | - Sara Xapelli
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisboa, Portugal; (D.M.L.); (L.R.-R.); (A.M.S.); (M.J.D.)
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisboa, Portugal
| |
Collapse
|
27
|
Zhao X, van Praag H. Steps towards standardized quantification of adult neurogenesis. Nat Commun 2020; 11:4275. [PMID: 32848155 PMCID: PMC7450090 DOI: 10.1038/s41467-020-18046-y] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2020] [Accepted: 08/03/2020] [Indexed: 02/07/2023] Open
Abstract
New neurons are generated in adult mammals. Adult hippocampal neurogenesis is considered to play an important role in cognition and mental health. The number and properties of newly born neurons are regulatable by a broad range of physiological and pathological conditions. To begin to understand the underlying cellular mechanisms and functional relevance of adult neurogenesis, many studies rely on quantification of adult-born neurons. However, lack of standardized methods to quantify new neurons is impeding research reproducibility across laboratories. Here, we review the importance of stereology, and propose why and how it should be applied to the study of adult neurogenesis.
Collapse
Affiliation(s)
- Xinyu Zhao
- Waisman Center and University of Wisconsin-Madison, Madison, WI, 53705, USA.
- Department of Neuroscience, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, 53705, USA.
| | - Henriette van Praag
- Brain Institute and Charles E. Schmidt College of Medicine, Florida Atlantic University, Jupiter, FL, 33458, USA.
| |
Collapse
|
28
|
Taha E, Patil S, Barrera I, Panov J, Khamaisy M, Proud CG, Bramham CR, Rosenblum K. eEF2/eEF2K Pathway in the Mature Dentate Gyrus Determines Neurogenesis Level and Cognition. Curr Biol 2020; 30:3507-3521.e7. [PMID: 32707059 DOI: 10.1016/j.cub.2020.06.061] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Revised: 05/08/2020] [Accepted: 06/18/2020] [Indexed: 12/13/2022]
Abstract
Levels of adult neurogenesis in the dentate gyrus (DG) of the hippocampus are correlated with unique cognitive functions. However, the molecular pathways controlling it are poorly understood. Here, we found that the known physiological ways to enhance neurogenesis converged on the eEF2/eEF2K pathway via AMPK in the DG. Enhancing the elongation phase of mRNA translation in eEF2K-knockout (eEF2K-KO) mice induced the expression of neurogenesis-related proteins in the hippocampus. We thus tested the hypothesis that inducing eEF2K-KO in mature neurons of the DG controls neurogenesis. Indeed, both general eEF2K-KO and targeted KO in DG excitatory mature neurons resulted in enhanced neurogenesis levels and upregulation of neurogenesis-related proteins. Increased neurogenesis was correlated with enhanced performance in DG-dependent learning. Moreover, general and local eEF2K-KO in old mice rejuvenated the DG, paving the way for better mechanistic understanding of how neurogenesis is controlled in the mature DG and possible treatments for incurable aging-associated diseases.
Collapse
Affiliation(s)
- Elham Taha
- Sagol Department of Neurobiology, 199 Aba Khoushy Avenue Mount Carmel, University of Haifa, 3498838 Haifa, Israel
| | - Sudarshan Patil
- Department of Biomedicine and KG Jebsen Centre for Research on Neuropsychiatric Disorders, Jonas Lies vei 91, University of Bergen, 5009 Bergen, Norway
| | - Iliana Barrera
- Sagol Department of Neurobiology, 199 Aba Khoushy Avenue Mount Carmel, University of Haifa, 3498838 Haifa, Israel
| | - Julia Panov
- Sagol Department of Neurobiology, 199 Aba Khoushy Avenue Mount Carmel, University of Haifa, 3498838 Haifa, Israel
| | - Mohammad Khamaisy
- Sagol Department of Neurobiology, 199 Aba Khoushy Avenue Mount Carmel, University of Haifa, 3498838 Haifa, Israel
| | - Christopher G Proud
- School of Biological Sciences, University of Adelaide, Adelaide, SA, Australia; Hopwood Centre for Neurobiology, Lifelong Health, South Australian Health and Medical Research Institute, PO Box 11060, 5001 Adelaide, SA, Australia
| | - Clive R Bramham
- Department of Biomedicine and KG Jebsen Centre for Research on Neuropsychiatric Disorders, Jonas Lies vei 91, University of Bergen, 5009 Bergen, Norway
| | - Kobi Rosenblum
- Sagol Department of Neurobiology, 199 Aba Khoushy Avenue Mount Carmel, University of Haifa, 3498838 Haifa, Israel; Center for Gene Manipulation in the Brain, 199 Aba Khoushy Avenue Mount Carmel, University of Haifa, 3498838 Haifa, Israel.
| |
Collapse
|
29
|
Martín-Suárez S, Abiega O, Ricobaraza A, Hernandez-Alcoceba R, Encinas JM. Alterations of the Hippocampal Neurogenic Niche in a Mouse Model of Dravet Syndrome. Front Cell Dev Biol 2020; 8:654. [PMID: 32793597 PMCID: PMC7385077 DOI: 10.3389/fcell.2020.00654] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Accepted: 07/01/2020] [Indexed: 01/24/2023] Open
Abstract
Hippocampal neurogenesis, the process by which neural stem cells (NSCs) continuously generate new neurons in the dentate gyrus (DG) of most mammals including humans, is chiefly regulated by neuronal activity. Thus, severe alterations have been found in samples from epilepsy patients and in the hippocampal neurogenic niche in mouse models of epilepsy. Reactive-like and gliogenic NSCs plus aberrant newborn neurons with altered migration, morphology, and functional properties are induced by seizures in experimental models of temporal lobe epilepsy. Hippocampal neurogenesis participates in memory and learning and in the control of anxiety and stress. It has been therefore hypothesized that part of the cognitive symptoms associated with epilepsy could be promoted by impaired hippocampal neurogenesis. We here analyze for the first time the alterations of the neurogenic niche in a novel mouse model of Dravet syndrome (DS), a genetic encephalopathy with severe epilepsy in infancy and multiple neurological comorbidities. Scn1aWT/A1783V mice, hereafter referred to as DS, carrying a heterozygous and clinically relevant SCN1A mutation (A1783V) recapitulate the disease at the genetic and phenotypic levels. We demonstrate that in the neurogenic niche of young adult DS mice there are fewer NSCs, they have impaired cell division and bear reactive-like morphology. In addition, there is significant aberrant neurogenesis. Newborn immature neurons migrate abnormally, and several morphological features are drastically changed. Thus, this study shows for the first time important modifications in hippocampal neurogenesis in DS and opens venues for further research on this topic.
Collapse
Affiliation(s)
- Soraya Martín-Suárez
- The Neural Stem Cell and Neurogenesis Laboratory, Achucarro Basque Center for Neuroscience, Leioa, Spain
| | - Oihane Abiega
- The Neural Stem Cell and Neurogenesis Laboratory, Achucarro Basque Center for Neuroscience, Leioa, Spain
| | - Ana Ricobaraza
- Gene Therapy Program CIMA, IdiSNA, Navarra Institute for Health Research, University of Navarra, Pamplona, Spain
| | - Rubén Hernandez-Alcoceba
- Gene Therapy Program CIMA, IdiSNA, Navarra Institute for Health Research, University of Navarra, Pamplona, Spain
| | - Juan Manuel Encinas
- The Neural Stem Cell and Neurogenesis Laboratory, Achucarro Basque Center for Neuroscience, Leioa, Spain.,Department of Neurosciences, Faculty of Medicine and Nursing, University of the Basque Country (UPV/EHU), Leioa, Spain.,IKERBASQUE, The Basque Foundation for Science, Bilbao, Spain
| |
Collapse
|
30
|
Snyder JS, Drew MR. Functional neurogenesis over the years. Behav Brain Res 2020; 382:112470. [PMID: 31917241 PMCID: PMC7769695 DOI: 10.1016/j.bbr.2020.112470] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2019] [Accepted: 01/03/2020] [Indexed: 01/01/2023]
Abstract
There has been interest in the function of adult neurogenesis since its discovery, by Joseph Altman, nearly 60 years ago. While controversy curtailed follow up studies, in the 1990s a second wave of research validated many of Altman's original claims and revealed that factors such as stress and environmental stimulation altered the production of new neurons in the hippocampus. However, only with the advent of tools for manipulating neurogenesis did it become possible to perform causal tests of the function of newborn neurons. Here, we identify approximately 100 studies in which adult neurogenesis was manipulated to study its function. A majority of these studies demonstrate functions for adult neurogenesis in classic hippocampal behaviors such as context learning and spatial memory, as well as emotional behaviors related to stress, anxiety and depression. However, a closer look reveals a number of other, arguably understudied, functions in decision making, temporal association memory, and addiction. In this special issue, we present 16 new studies and review articles that continue to address and clarify the function of adult neurogenesis in behaviors as diverse as memory formation, consolidation and forgetting, pattern separation and discrimination behaviors, addiction, and attention. Reviews of stem cell dynamics and regenerative properties provide insights into the mechanisms by which neurogenesis may be controlled to offset age- and disease-related brain injury. Finally, translation-oriented reviews identify next steps for minimizing the gap between discoveries made in animals and applications for human health. The articles in this issue synthesize and extend what we have learned in the last half century of functional neurogenesis research and identify themes that will define its future.
Collapse
Affiliation(s)
- Jason S Snyder
- Department of Psychology, Djavad Mowafaghian Centre for Brain Health, University of British Columbia, 2211 Wesbrook Mall, Vancouver, British Columbia, V6T 2B5, Canada.
| | - Michael R Drew
- Center for Learning and Memory, Department of Neuroscience, University of Texas at Austin, Austin, Texas, 78712, USA
| |
Collapse
|
31
|
Neuroinflammation and Neurogenesis in Alzheimer's Disease and Potential Therapeutic Approaches. Int J Mol Sci 2020; 21:ijms21030701. [PMID: 31973106 PMCID: PMC7037892 DOI: 10.3390/ijms21030701] [Citation(s) in RCA: 108] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2019] [Revised: 01/17/2020] [Accepted: 01/19/2020] [Indexed: 12/17/2022] Open
Abstract
In adult brain, new neurons are generated throughout adulthood in the subventricular zone and the dentate gyrus; this process is commonly known as adult neurogenesis. The regulation or modulation of adult neurogenesis includes various intrinsic pathways (signal transduction pathway and epigenetic or genetic modulation pathways) or extrinsic pathways (metabolic growth factor modulation, vascular, and immune system pathways). Altered neurogenesis has been identified in Alzheimer's disease (AD), in both human AD brains and AD rodent models. The exact mechanism of the dysregulation of adult neurogenesis in AD has not been completely elucidated. However, neuroinflammation has been demonstrated to alter adult neurogenesis. The presence of various inflammatory components, such as immune cells, cytokines, or chemokines, plays a role in regulating the survival, proliferation, and maturation of neural stem cells. Neuroinflammation has also been considered as a hallmark neuropathological feature of AD. In this review, we summarize current, state-of-the art perspectives on adult neurogenesis, neuroinflammation, and the relationship between these two phenomena in AD. Furthermore, we discuss the potential therapeutic approaches, focusing on the anti-inflammatory and proneurogenic interventions that have been reported in this field.
Collapse
|
32
|
Kathner-Schaffert C, Karapetow L, Günther M, Rudolph M, Dahab M, Baum E, Lehmann T, Witte OW, Redecker C, Schmeer CW, Keiner S. Early Stroke Induces Long-Term Impairment of Adult Neurogenesis Accompanied by Hippocampal-Mediated Cognitive Decline. Cells 2019; 8:cells8121654. [PMID: 31861141 PMCID: PMC6953020 DOI: 10.3390/cells8121654] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Revised: 11/29/2019] [Accepted: 12/12/2019] [Indexed: 12/20/2022] Open
Abstract
Stroke increases neurogenesis in the adult dentate gyrus in the short term, however, long-term effects at the cellular and functional level are poorly understood. Here we evaluated the impact of an early stroke lesion on neurogenesis and cognitive function of the aging brain. We hypothesized that a stroke disturbs dentate neurogenesis during aging correlate with impaired flexible learning. To address this issue a stroke was induced in 3-month-old C57Bl/6 mice by a middle cerebral artery occlusion (MCAO). To verify long-term changes of adult neurogenesis the thymidine analogue BrdU (5-Bromo-2′-deoxyuridine) was administrated at different time points during aging. One and half months after BrdU injections learning and memory performance were assessed with a modified version of the Morris water maze (MWM) that includes the re-learning paradigm, as well as hippocampus-dependent and -independent search strategies. After MWM performance mice were transcardially perfused. To further evaluate in detail the stroke-mediated changes on stem- and progenitor cells as well as endogenous proliferation nestin-green-fluorescent protein (GFP) mice were used. Adult nestin-GFP mice received a retroviral vector injection in the hippocampus to evaluate changes in the neuronal morphology. At an age of 20 month the nestin-GFP mice were transcardially perfused after MWM performance and BrdU application 1.5 months later. The early stroke lesion significantly decreased neurogenesis in 7.5- and 9-month-old animals and also endogenous proliferation in the latter group. Furthermore, immature doublecortin (DCX)-positive neurons were reduced in 20-month-old nestin-GFP mice after lesion. All MCAO groups showed an impaired performance in the MWM and mostly relied on hippocampal-independent search strategies. These findings indicate that an early ischemic insult leads to a dramatical decline of neurogenesis during aging that correlates with a premature development of hippocampal-dependent deficits. Our study supports the notion that an early stroke might lead to long-term cognitive deficits as observed in human patients after lesion.
Collapse
Affiliation(s)
- Carolin Kathner-Schaffert
- Hans-Berger Department of Neurology, Jena University Hospital, Am Klinikum 1, 07747 Jena, Germany; (C.K.-S.); (L.K.); (M.G.); (M.R.); (M.D.); (E.B.); (O.W.W.); (C.R.); (C.W.S.)
| | - Lina Karapetow
- Hans-Berger Department of Neurology, Jena University Hospital, Am Klinikum 1, 07747 Jena, Germany; (C.K.-S.); (L.K.); (M.G.); (M.R.); (M.D.); (E.B.); (O.W.W.); (C.R.); (C.W.S.)
| | - Madlen Günther
- Hans-Berger Department of Neurology, Jena University Hospital, Am Klinikum 1, 07747 Jena, Germany; (C.K.-S.); (L.K.); (M.G.); (M.R.); (M.D.); (E.B.); (O.W.W.); (C.R.); (C.W.S.)
| | - Max Rudolph
- Hans-Berger Department of Neurology, Jena University Hospital, Am Klinikum 1, 07747 Jena, Germany; (C.K.-S.); (L.K.); (M.G.); (M.R.); (M.D.); (E.B.); (O.W.W.); (C.R.); (C.W.S.)
| | - Mahmoud Dahab
- Hans-Berger Department of Neurology, Jena University Hospital, Am Klinikum 1, 07747 Jena, Germany; (C.K.-S.); (L.K.); (M.G.); (M.R.); (M.D.); (E.B.); (O.W.W.); (C.R.); (C.W.S.)
| | - Eileen Baum
- Hans-Berger Department of Neurology, Jena University Hospital, Am Klinikum 1, 07747 Jena, Germany; (C.K.-S.); (L.K.); (M.G.); (M.R.); (M.D.); (E.B.); (O.W.W.); (C.R.); (C.W.S.)
| | - Thomas Lehmann
- Institute of Medical Statistics and Computer Science, University Hospital Jena, Friedrich Schiller University Jena, 07743 Jena, Germany;
| | - Otto W. Witte
- Hans-Berger Department of Neurology, Jena University Hospital, Am Klinikum 1, 07747 Jena, Germany; (C.K.-S.); (L.K.); (M.G.); (M.R.); (M.D.); (E.B.); (O.W.W.); (C.R.); (C.W.S.)
| | - Christoph Redecker
- Hans-Berger Department of Neurology, Jena University Hospital, Am Klinikum 1, 07747 Jena, Germany; (C.K.-S.); (L.K.); (M.G.); (M.R.); (M.D.); (E.B.); (O.W.W.); (C.R.); (C.W.S.)
| | - Christian W. Schmeer
- Hans-Berger Department of Neurology, Jena University Hospital, Am Klinikum 1, 07747 Jena, Germany; (C.K.-S.); (L.K.); (M.G.); (M.R.); (M.D.); (E.B.); (O.W.W.); (C.R.); (C.W.S.)
| | - Silke Keiner
- Hans-Berger Department of Neurology, Jena University Hospital, Am Klinikum 1, 07747 Jena, Germany; (C.K.-S.); (L.K.); (M.G.); (M.R.); (M.D.); (E.B.); (O.W.W.); (C.R.); (C.W.S.)
- Correspondence: ; Tel.: +49-364-1932-5914
| |
Collapse
|