1
|
Graciani AL, Gutierre MU, Coppi AA, Arida RM, Gutierre RC. MYELIN, AGING, AND PHYSICAL EXERCISE. Neurobiol Aging 2023; 127:70-81. [PMID: 37116408 DOI: 10.1016/j.neurobiolaging.2023.03.009] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 03/22/2023] [Accepted: 03/24/2023] [Indexed: 04/03/2023]
Abstract
Myelin sheath is a structure in neurons fabricated by oligodendrocytes and Schwann cells responsible for increasing the efficiency of neural synapsis, impulse transmission, and providing metabolic support to the axon. They present morpho-functional changes during health aging as deformities of the sheath and its fragmentation, causing an increased load on microglial phagocytosis, with Alzheimer's disease aggravating. Physical exercise has been studied as a possible protective agent for the nervous system, offering benefits to neuroplasticity. In this regard, studies in animal models for Alzheimer's and depression reported the efficiency of physical exercise in protecting against myelin degeneration. A reduction of myelin damage during aging has also been observed in healthy humans. Physical activity promotes oligodendrocyte proliferation and myelin preservation during old age, although some controversies remain. In this review, we will address how effective physical exercise can be as a protective agent of the myelin sheath against the effects of aging in physiological and pathological conditions.
Collapse
|
2
|
Coviello S, Gramuntell Y, Klimczak P, Varea E, Blasco-Ibañez JM, Crespo C, Gutierrez A, Nacher J. Phenotype and Distribution of Immature Neurons in the Human Cerebral Cortex Layer II. Front Neuroanat 2022; 16:851432. [PMID: 35464133 PMCID: PMC9027810 DOI: 10.3389/fnana.2022.851432] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2022] [Accepted: 03/22/2022] [Indexed: 11/13/2022] Open
Abstract
This work provides evidence of the presence of immature neurons in the human brain, specifically in the layer II of the cerebral cortex. Using surgical samples from epileptic patients and post-mortem tissue, we have found cells with different levels of dendritic complexity (type I and type II cells) expressing DCX and PSA-NCAM and lacking expression of the mature neuronal marker NeuN. These immature cells belonged to the excitatory lineage, as demonstrated both by the expression of CUX1, CTIP2, and TBR1 transcription factors and by the lack of the inhibitory marker GAD67. The type II cells had some puncta expressing inhibitory and excitatory synaptic markers apposed to their perisomatic and peridendritic regions and ultrastructural analysis suggest the presence of synaptic contacts. These cells did not present glial cell markers, although astroglial and microglial processes were found in close apposition to their somata and dendrites, particularly on type I cells. Our findings confirm the presence of immature neurons in several regions of the cerebral cortex of humans of different ages and define their lineage. The presence of some mature features in some of these cells suggests the possibility of a progressively integration as excitatory neurons, as described in the olfactory cortex of rodents.
Collapse
Affiliation(s)
- Simona Coviello
- Neurobiology Unit, Program in Neurosciences and Institute of Biotechnology and Biomedicine (BIOTECMED), Universitat de València, Burjassot, Spain
| | - Yaiza Gramuntell
- Neurobiology Unit, Program in Neurosciences and Institute of Biotechnology and Biomedicine (BIOTECMED), Universitat de València, Burjassot, Spain
| | - Patrycja Klimczak
- Neurobiology Unit, Program in Neurosciences and Institute of Biotechnology and Biomedicine (BIOTECMED), Universitat de València, Burjassot, Spain
- Spanish National Network for Research in Mental Health, Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Madrid, Spain
| | - Emilio Varea
- Neurobiology Unit, Program in Neurosciences and Institute of Biotechnology and Biomedicine (BIOTECMED), Universitat de València, Burjassot, Spain
| | - José Miguel Blasco-Ibañez
- Neurobiology Unit, Program in Neurosciences and Institute of Biotechnology and Biomedicine (BIOTECMED), Universitat de València, Burjassot, Spain
| | - Carlos Crespo
- Neurobiology Unit, Program in Neurosciences and Institute of Biotechnology and Biomedicine (BIOTECMED), Universitat de València, Burjassot, Spain
| | - Antonio Gutierrez
- Unidad de Cirugía de la Epilepsia, Hospital Universitario La Fe, Valencia, Spain
| | - Juan Nacher
- Neurobiology Unit, Program in Neurosciences and Institute of Biotechnology and Biomedicine (BIOTECMED), Universitat de València, Burjassot, Spain
- Spanish National Network for Research in Mental Health, Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Madrid, Spain
- Fundación Investigación Hospital Clínico de Valencia (INCLIVA), Valencia, Spain
- *Correspondence: Juan Nacher,
| |
Collapse
|
3
|
Bunk EC, König HG, Prehn JHM, Kirby BP. p53 upregulated mediator of apoptosis (Puma) deficiency increases survival of adult neural stem cells generated physiologically in the hippocampus, but does not protect stem cells generated in surplus after an excitotoxic lesion. J Basic Clin Physiol Pharmacol 2020; 32:57-66. [PMID: 33155994 DOI: 10.1515/jbcpp-2020-0109] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Accepted: 10/09/2020] [Indexed: 11/15/2022]
Abstract
OBJECTIVES Neurogenesis occurs in the mammalian brain throughout adulthood and increases in response to metabolic, toxic or traumatic insults. To remove potentially superfluous or unwanted neural stem cells/neuronal progenitors, their rate of proliferation and differentiation is fine-tuned against their rate of apoptosis. Apoptosis requires the transcriptional and posttranslational activation of Bcl-2-homolgy domain 3 (BH3)-only proteins. Previously, we demonstrated that the BH3-only protein p53-upregulated mediator of apoptosis (Puma) controls the physiological rate of apoptosis of neural precursor cells in the adult mouse hippocampus. Puma's role in controlling a lesion-induced increase in neural stem cells is currently not known. METHODS We employed a model of local, N-methyl-D-asparte (NMDA)-induced excitotoxic injury to the CA1 hippocampal subfield and immunofluorescence labelling to produce increased neural stem cell proliferation/ neurogenesis in the dentate gyrus at two survival times following the excitotoxic lesion. RESULTS Deletion of puma failed to rescue any NMDA-induced increase in adult born cells as assessed by BrdU or Doublecortin labelling in the long-term. No difference in the proportion of BrdU/NeuN-positive cells comparing the different genotypes and treatments suggested that the phenotypic fate of the cells was preserved regardless of the genotype and the treatment. CONCLUSIONS While neurogenesis is up-regulated in puma-deficient animals following NMDA-induced excitotoxicity to the hippocampal CA1 subfield, puma deficiency could not protect this surplus of newly generated cells from apoptotic cell death.
Collapse
Affiliation(s)
- Eva C Bunk
- Department of Physiology and Medical Physics, RCSI University of Medicine and Health Sciences, Dublin 2, Ireland.,Klinik für Neurochirurgie, Universitätsklinikum Münster, Albert-Schweitzer-Campus 1, Gebäude A1, 48149, Münster, Germany
| | - Hans-Georg König
- Department of Physiology and Medical Physics, RCSI University of Medicine and Health Sciences, Dublin 2, Ireland
| | - Jochen H M Prehn
- Department of Physiology and Medical Physics, RCSI University of Medicine and Health Sciences, Dublin 2, Ireland.,Future-Neuro SFI Research Centre, RCSI University of Medicine and Health Sciences, Dublin 2, Ireland
| | - Brian P Kirby
- School of Pharmacy and Biomolecular Sciences, RCSI University of Medicine and Health Sciences, Dublin 2, Ireland
| |
Collapse
|
4
|
Ojeda J, Ávila A. Early Actions of Neurotransmitters During Cortex Development and Maturation of Reprogrammed Neurons. Front Synaptic Neurosci 2019; 11:33. [PMID: 31824293 PMCID: PMC6881277 DOI: 10.3389/fnsyn.2019.00033] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Accepted: 11/07/2019] [Indexed: 12/12/2022] Open
Abstract
The development of the brain is shaped by a myriad of factors among which neurotransmitters play remarkable roles before and during the formation and maturation of synaptic circuits. Cellular processes such as neurogenesis, morphological development, synaptogenesis and maturation of synapses are temporary and spatially regulated by the local or distal influence of neurotransmitters in the developing cortex. Thus, research on this area has contributed to the understanding of fundamental mechanisms of brain development and to shed light on the etiology of various human neurodevelopmental disorders such as autism and Rett syndrome (RTT), among others. Recently, the field of neuroscience has been shaken by an explosive advance of experimental approaches linked to the use of induced pluripotent stem cells and reprogrammed neurons. This new technology has allowed researchers for the first time to model in the lab the unique events that take place during early human brain development and to explore the mechanisms that cause synaptopathies. In this context, the role of neurotransmitters during early stages of cortex development is beginning to be re-evaluated and a revision of the state of the art has become necessary in a time when new protocols are being worked out to differentiate stem cells into functional neurons. New perspectives on reconsidering the function of neurotransmitters include opportunities for methodological advances, a better understanding of the origin of mental disorders and the potential for development of new treatments.
Collapse
Affiliation(s)
- Jorge Ojeda
- Developmental Neurobiology Unit, Biomedical Sciences Research Laboratory, Basic Sciences Department, Faculty of Medicine, Universidad Católica de la Santísima Concepción, Concepción, Chile
| | - Ariel Ávila
- Developmental Neurobiology Unit, Biomedical Sciences Research Laboratory, Basic Sciences Department, Faculty of Medicine, Universidad Católica de la Santísima Concepción, Concepción, Chile
| |
Collapse
|
5
|
Amidfar M, Woelfer M, Réus GZ, Quevedo J, Walter M, Kim YK. The role of NMDA receptor in neurobiology and treatment of major depressive disorder: Evidence from translational research. Prog Neuropsychopharmacol Biol Psychiatry 2019; 94:109668. [PMID: 31207274 DOI: 10.1016/j.pnpbp.2019.109668] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2019] [Revised: 05/24/2019] [Accepted: 06/11/2019] [Indexed: 12/16/2022]
Abstract
There is accumulating evidence demonstrating that dysfunction of glutamatergic neurotransmission, particularly via N-methyl-d-aspartate (NMDA) receptors, is involved in the pathophysiology of major depressive disorder (MDD). Several studies have revealed an altered expression of NMDA receptor subtypes and impaired NMDA receptor-mediated intracellular signaling pathways in brain circuits of patients with MDD. Clinical studies have demonstrated that NMDA receptor antagonists, particularly ketamine, have rapid antidepressant effects in treatment-resistant depression, however, neurobiological mechanisms are not completely understood. Growing body of evidence suggest that signal transduction pathways involved in synaptic plasticity play critical role in molecular mechanisms underlying rapidly acting antidepressant properties of ketamine and other NMDAR antagonists in MDD. Discovering the molecular mechanisms underlying the unique antidepressant actions of ketamine will facilitate the development of novel fast acting antidepressants which lack undesirable effects of ketamine. This review provides a critical examination of the NMDA receptor involvement in the neurobiology of MDD including analyses of alterations in NMDA receptor subtypes and their interactive signaling cascades revealed by postmortem studies. Furthermore, to elucidate mechanisms underlying rapid-acting antidepressant properties of NMDA receptor antagonists we discussed their effects on the neuroplasticity, mostly based on signaling systems involved in synaptic plasticity of mood-related neurocircuitries.
Collapse
Affiliation(s)
| | - Marie Woelfer
- Clinical Affective Neuroimaging Laboratory, University Magdeburg, Germany; New Jersey Institute of Technology, Newark, NJ, USA
| | - Gislaine Z Réus
- Translational Psychiatry Laboratory, Graduate Program in Health Sciences, Health Sciences Unit, University of Southern Santa Catarina, Criciúma, SC, Brazil
| | - João Quevedo
- Translational Psychiatry Laboratory, Graduate Program in Health Sciences, Health Sciences Unit, University of Southern Santa Catarina, Criciúma, SC, Brazil; Translational Psychiatry Program, Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA; Center of Excellence on Mood Disorders, Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA; Neuroscience Graduate Program, The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX, USA
| | - Martin Walter
- Clinical Affective Neuroimaging Laboratory, University Magdeburg, Germany; Department of Psychiatry, University Tuebingen, Germany
| | - Yong-Ku Kim
- Department of Psychiatry, College of Medicine, Korea University, Seoul, South Korea
| |
Collapse
|
6
|
The Neurogenesis Actuator and NR2B/NMDA Receptor Antagonist Ro25-6981 Consistently Improves Spatial Memory Retraining Via Brain Region-Specific Gene Expression. J Mol Neurosci 2018; 65:167-178. [PMID: 29790100 PMCID: PMC6061165 DOI: 10.1007/s12031-018-1083-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2018] [Accepted: 05/09/2018] [Indexed: 12/30/2022]
Abstract
NR2B-containing NMDA (NR2B/NMDA) receptors are important in controlling neurogenesis and are involved in generating spatial memory. Ro25-6981 is a selective antagonist at these receptors and actuates neurogenesis and spatial memory. Inter-structural neuroanatomical profiles of gene expression regulating adult neurogenesis and neuroapoptosis require examination in the context of memory retrieval and reversal learning. The aim was to investigate spatial memory retrieval and reversal learning in relation to gene expression-linked neurogenetic processes following blockade of NR2B/NMDA receptors by Ro25-6981. Rats were trained in Morris water maze (MWM) platform location for 5 days. Ro25-6981 was administered (protocol days 6–7) followed by retraining (days 15–18 or 29–32). Platform location was tested (on days 19 or 33) then post-mortem brain tissue sampling (on days 20 or 34). The expression of three genes known to regulate cell proliferation (S100a6), differentiation (Ascl1), and apoptosis (Casp-3) were concomitantly evaluated in the hippocampus, prefrontal cortex, and cerebellum in relation to the MWM performance protocol. Following initial training, Ro25-6981 enhanced visuospatial memory retrieval performance during further retraining (protocol days 29–32) but did not influence visuospatial reversal learning (day 33). Hippocampal Ascl1 and Casp-3 expressions were correspondingly increased and decreased while cerebellar S100a6 and Casp-3 activities were decreased and increased respectively 27 days after Ro25-6981 treatment. Chronological analysis indicated a possible involvement of new mature neurons in the reconfiguration of memory processes. This was attended by behavioral/gene correlations which revealed direct links between spatial memory retrieval enhancement and modified gene activity induced by NR2B/NMDA receptor blockade and upregulation.
Collapse
|
7
|
Laszczyk AM, Fox-Quick S, Vo HT, Nettles D, Pugh PC, Overstreet-Wadiche L, King GD. Klotho regulates postnatal neurogenesis and protects against age-related spatial memory loss. Neurobiol Aging 2017; 59:41-54. [PMID: 28837861 PMCID: PMC5612914 DOI: 10.1016/j.neurobiolaging.2017.07.008] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2017] [Revised: 06/22/2017] [Accepted: 07/21/2017] [Indexed: 12/29/2022]
Abstract
Although the absence of the age-regulating klotho protein causes klotho-deficient mice to rapidly develop cognitive impairment and increasing klotho enhances hippocampal-dependent memory, the cellular effects of klotho that mediate hippocampal-dependent memory function are unknown. Here, we show premature aging of the klotho-deficient hippocampal neurogenic niche as evidenced by reduced numbers of neural stem cells, decreased proliferation, and impaired maturation of immature neurons. Klotho-deficient neurospheres show reduced proliferation and size that is rescued by supplementation with shed klotho protein. Conversely, 6-month-old klotho-overexpressing mice exhibit increased numbers of neural stem cells, increased proliferation, and more immature neurons with enhanced dendritic arborization. Protection from normal age-related loss of object location memory with klotho overexpression and loss of spatial memory when klotho is reduced by even half suggests direct, local effects of the protein. Together, these data show that klotho is a novel regulator of postnatal neurogenesis affecting neural stem cell proliferation and maturation sufficient to impact hippocampal-dependent spatial memory function.
Collapse
Affiliation(s)
- Ann M Laszczyk
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Stephanie Fox-Quick
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Hai T Vo
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Dailey Nettles
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Phyllis C Pugh
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, AL, USA
| | | | - Gwendalyn D King
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, AL, USA.
| |
Collapse
|
8
|
|
9
|
Perez-Rando M, Castillo-Gómez E, Guirado R, Blasco-Ibañez JM, Crespo C, Varea E, Nacher J. NMDA Receptors Regulate the Structural Plasticity of Spines and Axonal Boutons in Hippocampal Interneurons. Front Cell Neurosci 2017; 11:166. [PMID: 28659763 PMCID: PMC5466979 DOI: 10.3389/fncel.2017.00166] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2017] [Accepted: 05/29/2017] [Indexed: 11/13/2022] Open
Abstract
N-methyl-D-aspartate receptors (NMDARs) are present in both pyramidal neurons and interneurons of the hippocampus. These receptors play an important role in the adult structural plasticity of excitatory neurons, but their impact on the remodeling of interneurons is unknown. Among hippocampal interneurons, somatostatin-expressing cells located in the stratum oriens are of special interest because of their functional importance and structural characteristics: they display dendritic spines, which change density in response to different stimuli. In order to understand the role of NMDARs on the structural plasticity of these interneurons, we have injected acutely MK-801, an NMDAR antagonist, to adult mice which constitutively express enhanced green fluorescent protein (EGFP) in these cells. We have behaviorally tested the animals, confirming effects of the drug on locomotion and anxiety-related behaviors. NMDARs were expressed in the somata and dendritic spines of somatostatin-expressing interneurons. Twenty-four hours after the injection, the density of spines did not vary, but we found a significant increase in the density of their en passant boutons (EPB). We have also used entorhino-hippocampal organotypic cultures to study these interneurons in real-time. There was a rapid decrease in the apparition rate of spines after MK-801 administration, which persisted for 24 h and returned to basal levels afterwards. A similar reversible decrease was detected in spine density. Our results show that both spines and axons of interneurons can undergo remodeling and highlight NMDARs as regulators of this plasticity. These results are specially relevant given the importance of all these players on hippocampal physiology and the etiopathology of certain psychiatric disorders.
Collapse
Affiliation(s)
- Marta Perez-Rando
- Neurobiology Unit, Department of Cell Biology, Interdisciplinary Research Structure for Biotechnology and Biomedicine (BIOTECMED), Universitat de ValènciaValència, Spain
| | - Esther Castillo-Gómez
- Neurobiology Unit, Department of Cell Biology, Interdisciplinary Research Structure for Biotechnology and Biomedicine (BIOTECMED), Universitat de ValènciaValència, Spain
| | - Ramon Guirado
- Neurobiology Unit, Department of Cell Biology, Interdisciplinary Research Structure for Biotechnology and Biomedicine (BIOTECMED), Universitat de ValènciaValència, Spain
| | - José Miguel Blasco-Ibañez
- Neurobiology Unit, Department of Cell Biology, Interdisciplinary Research Structure for Biotechnology and Biomedicine (BIOTECMED), Universitat de ValènciaValència, Spain
| | - Carlos Crespo
- Neurobiology Unit, Department of Cell Biology, Interdisciplinary Research Structure for Biotechnology and Biomedicine (BIOTECMED), Universitat de ValènciaValència, Spain
| | - Emilio Varea
- Neurobiology Unit, Department of Cell Biology, Interdisciplinary Research Structure for Biotechnology and Biomedicine (BIOTECMED), Universitat de ValènciaValència, Spain
| | - Juan Nacher
- Neurobiology Unit, Department of Cell Biology, Interdisciplinary Research Structure for Biotechnology and Biomedicine (BIOTECMED), Universitat de ValènciaValència, Spain.,CIBERSAM: Spanish National Network for Research in Mental HealthMadrid, Spain.,Fundación Investigación Hospital Clínico de Valencia, Instituto de Investigación Sanitaria (INCLIVA)València, Spain
| |
Collapse
|
10
|
Teng L, Lei HM, Sun F, An SM, Tang YB, Meng S, Wang CH, Shen Y, Chen HZ, Zhu L. Autocrine glutamatergic transmission for the regulation of embryonal carcinoma stem cells. Oncotarget 2016; 7:49552-49564. [PMID: 27322683 PMCID: PMC5226528 DOI: 10.18632/oncotarget.9973] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2015] [Accepted: 05/30/2016] [Indexed: 12/17/2022] Open
Abstract
Glutamate behaves as the principal excitatory neurotransmitter in the vertebrate central nervous system and recently demonstrates intercellular signaling activities in periphery cancer cells. How the glutamatergic transmission is organized and operated in cancer stem cells remains undefined. We have identified a glutamatergic transmission circuit in embryonal carcinoma stem cells. The circuit is organized and operated in an autocrine mechanism and suppresses the cell proliferation and motility. Biological analyses determined a repertoire of glutamatergic transmission components, glutaminase, vesicular glutamate transporter, glutamate NMDA receptor, and cell membrane excitatory amino-acid transporter, for glutamate biosynthesis, package for secretion, reaction, and reuptake in mouse and human embryonal carcinoma stem cells. The glutamatergic components were also identified in mouse transplanted teratocarcinoma and in human primary teratocarcinoma tissues. Released glutamate acting as the signal was directly quantified by liquid chromatography coupled with tandem mass spectrometry (LC-MS/MS). Genetic and pharmacological abolishment of the endogenously released glutamate-induced tonic activation of the NMDA receptors increased the cell proliferation and motility. The finding suggests that embryonal carcinoma stem cells can be actively regulated by establishing a glutamatergic autocrine/paracrine niche via releasing and responding to the transmitter.
Collapse
Affiliation(s)
- Lin Teng
- Department of Pharmacology and Chemical Biology, Basic Medicine Faculty of Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China.,Present address: Department of Cardiology, The First College of Clinical Medical Sciences, China Three Gorges University, Hubei 443003, China
| | - Hui-Min Lei
- Department of Pharmacology and Chemical Biology, Basic Medicine Faculty of Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Fan Sun
- Department of Pharmacology and Chemical Biology, Basic Medicine Faculty of Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China.,Department of Pharmacy, Renji Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China
| | - Shi-Min An
- Department of Pharmacology and Chemical Biology, Basic Medicine Faculty of Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China.,Shanghai Universities Collaborative Innovation Center for Translational Medicine, Shanghai 200025, China
| | - Ya-Bin Tang
- Department of Pharmacology and Chemical Biology, Basic Medicine Faculty of Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China.,Shanghai Universities Collaborative Innovation Center for Translational Medicine, Shanghai 200025, China
| | - Shuang Meng
- Department of Pharmacology and Chemical Biology, Basic Medicine Faculty of Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Cong-Hui Wang
- Department of Pharmacology and Chemical Biology, Basic Medicine Faculty of Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China.,Shanghai Universities Collaborative Innovation Center for Translational Medicine, Shanghai 200025, China
| | - Ying Shen
- Department of Pharmacology and Chemical Biology, Basic Medicine Faculty of Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China.,Shanghai Universities Collaborative Innovation Center for Translational Medicine, Shanghai 200025, China
| | - Hong-Zhuan Chen
- Department of Pharmacology and Chemical Biology, Basic Medicine Faculty of Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China.,Shanghai Universities Collaborative Innovation Center for Translational Medicine, Shanghai 200025, China
| | - Liang Zhu
- Department of Pharmacology and Chemical Biology, Basic Medicine Faculty of Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China.,Shanghai Universities Collaborative Innovation Center for Translational Medicine, Shanghai 200025, China
| |
Collapse
|
11
|
López-Hidalgo R, Ballestín R, Vega J, Blasco-Ibáñez JM, Crespo C, Gilabert-Juan J, Nácher J, Varea E. Hypocellularity in the Murine Model for Down Syndrome Ts65Dn Is Not Affected by Adult Neurogenesis. Front Neurosci 2016; 10:75. [PMID: 26973453 PMCID: PMC4773601 DOI: 10.3389/fnins.2016.00075] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2015] [Accepted: 02/17/2016] [Indexed: 01/08/2023] Open
Abstract
Down syndrome (DS) is caused by the presence of an extra copy of the chromosome 21 and it is the most common aneuploidy producing intellectual disability. Neural mechanisms underlying this alteration may include defects in the formation of neuronal networks, information processing and brain plasticity. The murine model for DS, Ts65Dn, presents reduced adult neurogenesis. This reduction has been suggested to underlie the hypocellularity of the hippocampus as well as the deficit in olfactory learning in the Ts65Dn mice. Similar alterations have also been observed in individuals with DS. To determine whether the impairment in adult neurogenesis is, in fact, responsible for the hypocellularity in the hippocampus and physiology of the olfactory bulb, we have analyzed cell proliferation and neuronal maturation in the two major adult neurogenic niches in the Ts656Dn mice: the subgranular zone (SGZ) of the hippocampus and the subventricular zone (SVZ). Additionally, we carried out a study to determine the survival rate and phenotypic fate of newly generated cells in both regions, injecting 5'BrdU and sacrificing the mice 21 days later, and analyzing the number and phenotype of the remaining 5'BrdU-positive cells. We observed a reduction in the number of proliferating (Ki67 positive) cells and immature (doublecortin positive) neurons in the subgranular and SVZ of Ts65Dn mice, but we did not observe changes in the number of surviving cells or in their phenotype. These data correlated with a lower number of apoptotic cells (cleaved caspase 3 positive) in Ts65Dn. We conclude that although adult Ts65Dn mice have a lower number of proliferating cells, it is compensated by a lower level of cell death. This higher survival rate in Ts65Dn produces a final number of mature cells similar to controls. Therefore, the reduction of adult neurogenesis cannot be held responsible for the neuronal hypocellularity in the hippocampus or for the olfactory learning deficit of Ts65Dn mice.
Collapse
Affiliation(s)
- Rosa López-Hidalgo
- Neurobiology Unit and Program in Basic and Applied Neurosciences, Cell Biology Department, Universitat de ValènciaValència, Spain
- Estructura de Recerca Interdisciplinar en Biotecnologia i Biomedicina (BIOTECMED), Universitat de ValènciaValència, Spain
| | - Raul Ballestín
- Neurobiology Unit and Program in Basic and Applied Neurosciences, Cell Biology Department, Universitat de ValènciaValència, Spain
- Estructura de Recerca Interdisciplinar en Biotecnologia i Biomedicina (BIOTECMED), Universitat de ValènciaValència, Spain
| | - Jessica Vega
- Neurobiology Unit and Program in Basic and Applied Neurosciences, Cell Biology Department, Universitat de ValènciaValència, Spain
- Estructura de Recerca Interdisciplinar en Biotecnologia i Biomedicina (BIOTECMED), Universitat de ValènciaValència, Spain
| | - José M. Blasco-Ibáñez
- Neurobiology Unit and Program in Basic and Applied Neurosciences, Cell Biology Department, Universitat de ValènciaValència, Spain
- Estructura de Recerca Interdisciplinar en Biotecnologia i Biomedicina (BIOTECMED), Universitat de ValènciaValència, Spain
| | - Carlos Crespo
- Neurobiology Unit and Program in Basic and Applied Neurosciences, Cell Biology Department, Universitat de ValènciaValència, Spain
- Estructura de Recerca Interdisciplinar en Biotecnologia i Biomedicina (BIOTECMED), Universitat de ValènciaValència, Spain
| | - Javier Gilabert-Juan
- Neurobiology Unit and Program in Basic and Applied Neurosciences, Cell Biology Department, Universitat de ValènciaValència, Spain
- Estructura de Recerca Interdisciplinar en Biotecnologia i Biomedicina (BIOTECMED), Universitat de ValènciaValència, Spain
- Fundación Investigación Hospital Clínico de Valencia, INCLIVAValència, Spain
- CIBERSAM, Spanish National Network for Research in Mental HealthValència, Spain
- Genetics Department, CIBERSAM, Universitat de ValènciaValència, Spain
| | - Juan Nácher
- Neurobiology Unit and Program in Basic and Applied Neurosciences, Cell Biology Department, Universitat de ValènciaValència, Spain
- Estructura de Recerca Interdisciplinar en Biotecnologia i Biomedicina (BIOTECMED), Universitat de ValènciaValència, Spain
- Fundación Investigación Hospital Clínico de Valencia, INCLIVAValència, Spain
- CIBERSAM, Spanish National Network for Research in Mental HealthValència, Spain
- Genetics Department, CIBERSAM, Universitat de ValènciaValència, Spain
| | - Emilio Varea
- Neurobiology Unit and Program in Basic and Applied Neurosciences, Cell Biology Department, Universitat de ValènciaValència, Spain
- Estructura de Recerca Interdisciplinar en Biotecnologia i Biomedicina (BIOTECMED), Universitat de ValènciaValència, Spain
| |
Collapse
|
12
|
|
13
|
Staples MC, Mandyam CD. Thinking after Drinking: Impaired Hippocampal-Dependent Cognition in Human Alcoholics and Animal Models of Alcohol Dependence. Front Psychiatry 2016; 7:162. [PMID: 27746746 PMCID: PMC5043052 DOI: 10.3389/fpsyt.2016.00162] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2016] [Accepted: 09/13/2016] [Indexed: 12/05/2022] Open
Abstract
Alcohol use disorder currently affects approximately 18 million Americans, with at least half of these individuals having significant cognitive impairments subsequent to their chronic alcohol use. This is most widely apparent as frontal cortex-dependent cognitive dysfunction, where executive function and decision-making are severely compromised, as well as hippocampus-dependent cognitive dysfunction, where contextual and temporal reasoning are negatively impacted. This review discusses the relevant clinical literature to support the theory that cognitive recovery in tasks dependent on the prefrontal cortex and hippocampus is temporally different across extended periods of abstinence from alcohol. Additional studies from preclinical models are discussed to support clinical findings. Finally, the unique cellular composition of the hippocampus and cognitive impairment dependent on the hippocampus is highlighted in the context of alcohol dependence.
Collapse
Affiliation(s)
- Miranda C Staples
- Committee on the Neurobiology of Addictive Disorders, The Scripps Research Institute , La Jolla, CA , USA
| | - Chitra D Mandyam
- Committee on the Neurobiology of Addictive Disorders, The Scripps Research Institute , La Jolla, CA , USA
| |
Collapse
|
14
|
Korte SM, Prins J, Krajnc AM, Hendriksen H, Oosting RS, Westphal KG, Korte-Bouws GA, Olivier B. The many different faces of major depression: It is time for personalized medicine. Eur J Pharmacol 2015; 753:88-104. [DOI: 10.1016/j.ejphar.2014.11.045] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2014] [Revised: 10/16/2014] [Accepted: 11/26/2014] [Indexed: 01/11/2023]
|
15
|
Geil CR, Hayes DM, McClain JA, Liput DJ, Marshall SA, Chen KY, Nixon K. Alcohol and adult hippocampal neurogenesis: promiscuous drug, wanton effects. Prog Neuropsychopharmacol Biol Psychiatry 2014; 54:103-13. [PMID: 24842804 PMCID: PMC4134968 DOI: 10.1016/j.pnpbp.2014.05.003] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2014] [Revised: 05/01/2014] [Accepted: 05/08/2014] [Indexed: 01/29/2023]
Abstract
Adult neurogenesis is now widely accepted as an important contributor to hippocampal integrity and function but also dysfunction when adult neurogenesis is affected in neuropsychiatric diseases such as alcohol use disorders. Excessive alcohol consumption, the defining characteristic of alcohol use disorders, results in a variety of cognitive and behavioral impairments related wholly or in part to hippocampal structure and function. Recent preclinical work has shown that adult neurogenesis may be one route by which alcohol produces hippocampal neuropathology. Alcohol is a pharmacologically promiscuous drug capable of interfering with adult neurogenesis through multiple mechanisms. This review will discuss the primary mechanisms underlying alcohol-induced changes in adult hippocampal neurogenesis including alcohol's effects on neurotransmitters, CREB and its downstream effectors, and the neurogenic niche.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Kimberly Nixon
- Department of Pharmaceutical Sciences, University of Kentucky, Lexington, KY 40536, United States.
| |
Collapse
|
16
|
Kannangara TS, Bostrom CA, Ratzlaff A, Thompson L, Cater RM, Gil-Mohapel J, Christie BR. Deletion of the NMDA receptor GluN2A subunit significantly decreases dendritic growth in maturing dentate granule neurons. PLoS One 2014; 9:e103155. [PMID: 25083703 PMCID: PMC4118862 DOI: 10.1371/journal.pone.0103155] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2014] [Accepted: 06/26/2014] [Indexed: 11/18/2022] Open
Abstract
It is known that NMDA receptors can modulate adult hippocampal neurogenesis, but the contribution of specific regulatory GluN2 subunits has been difficult to determine. Here we demonstrate that mice lacking GluN2A (formerly NR2A) do not show altered cell proliferation or neuronal differentiation, but present significant changes in neuronal morphology in dentate granule cells. Specifically, GluN2A deletion significantly decreased total dendritic length and dendritic complexity in DG neurons located in the inner granular zone. Furthermore, the absence of GluN2A also resulted in a localized increase in spine density in the middle molecular layer, a region innervated by the medial perforant path. Interestingly, alterations in dendritic morphology and spine density were never seen in dentate granule cells located in the outer granular zone, a region that has been hypothesized to contain older, more mature, neurons. These results indicate that although the GluN2A subunit is not critical for the cell proliferation and differentiation stages of the neurogenic process, it does appear to play a role in establishing synaptic and dendritic morphology in maturing dentate granule cells localized in the inner granular zone.
Collapse
Affiliation(s)
- Timal S. Kannangara
- Division of Medical Sciences, University of Victoria, Victoria, Canada
- Department of Cellular and Physiological Sciences, University of British Columbia, Victoria, Canada
- Graduate Program of Neuroscience and The Brain Research Centre, University of British Columbia, Victoria, Canada
| | - Crystal A. Bostrom
- Division of Medical Sciences, University of Victoria, Victoria, Canada
- Department of Biology, University of Victoria, Victoria, Canada
| | - Andrea Ratzlaff
- Division of Medical Sciences, University of Victoria, Victoria, Canada
- Department of Biology, University of Victoria, Victoria, Canada
| | - Lee Thompson
- Division of Medical Sciences, University of Victoria, Victoria, Canada
- Department of Biology, University of Victoria, Victoria, Canada
| | - Robyn M. Cater
- Division of Medical Sciences, University of Victoria, Victoria, Canada
- Department of Biology, University of Victoria, Victoria, Canada
| | - Joana Gil-Mohapel
- Division of Medical Sciences, University of Victoria, Victoria, Canada
| | - Brian R. Christie
- Division of Medical Sciences, University of Victoria, Victoria, Canada
- Department of Biology, University of Victoria, Victoria, Canada
- Neuroscience Graduate Program, University of Victoria, Victoria, Canada
- Department of Cellular and Physiological Sciences, University of British Columbia, Victoria, Canada
- Graduate Program of Neuroscience and The Brain Research Centre, University of British Columbia, Victoria, Canada
- * E-mail:
| |
Collapse
|
17
|
Leptin reverses corticosterone-induced inhibition of neural stem cell proliferation through activating the NR2B subunits of NMDA receptors. Biochem Biophys Res Commun 2014; 447:95-100. [DOI: 10.1016/j.bbrc.2014.03.112] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2014] [Accepted: 03/22/2014] [Indexed: 12/31/2022]
|
18
|
The role of the N-methyl-d-aspartate receptor in the proliferation of adult hippocampal neural stem and precursor cells. SCIENCE CHINA-LIFE SCIENCES 2014; 57:403-11. [DOI: 10.1007/s11427-014-4637-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/15/2014] [Accepted: 02/26/2014] [Indexed: 12/29/2022]
|
19
|
Kannangara TS, Eadie BD, Bostrom CA, Morch K, Brocardo PS, Christie BR. GluN2A-/- Mice Lack Bidirectional Synaptic Plasticity in the Dentate Gyrus and Perform Poorly on Spatial Pattern Separation Tasks. Cereb Cortex 2014; 25:2102-13. [PMID: 24554729 DOI: 10.1093/cercor/bhu017] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
The different secondary subunits of the N-methyl-d-aspartate (NMDA) receptor each convey unique biophysical properties to the receptor complex, and may be key in determining the functional role played by NMDA receptors. In the hippocampus, the GluN2A and GluN2B subunits are particularly abundant; however, their exact roles in synaptic plasticity and behavior remain controversial. Here, we show that mice carrying a deletion for the GluN2A subunit (GluN2A(-/-)) demonstrate a severely compromised NMDA to AMPA receptor current ratio in granule cells from the dentate gyrus (DG), while granule cell morphology is unaltered. This deficit is accompanied by significant impairments in both LTP and LTD in the DG, whereas only minor impairments are observed in the CA1. In accordance with these hippocampal region-specific deficits, GluN2A(-/-) mice show impaired performance on the DG-associated task of spatial pattern separation. In contrast, GluN2A(-/-) mice show no deficit in temporal pattern separation, a process associated with CA1 functioning. Thus, our results establish the GluN2A subunit as a significant contributor to both bidirectional synaptic plasticity and spatial pattern separation in the DG.
Collapse
Affiliation(s)
- Timal S Kannangara
- Division of Medical Sciences Department of Cellular and Physiological Sciences Graduate Programs of Neuroscience and the Brain Research Centre, University of British Columbia, Vancouver, BC, Canada, V6T 1Z3
| | - Brennan D Eadie
- Division of Medical Sciences Department of Cellular and Physiological Sciences Graduate Programs of Neuroscience and the Brain Research Centre, University of British Columbia, Vancouver, BC, Canada, V6T 1Z3
| | - Crystal A Bostrom
- Division of Medical Sciences Department of Biology, University of Victoria, BC, Canada, V8P 5C2 and
| | | | | | - Brian R Christie
- Division of Medical Sciences Department of Biology, University of Victoria, BC, Canada, V8P 5C2 and Department of Cellular and Physiological Sciences Graduate Programs of Neuroscience and the Brain Research Centre, University of British Columbia, Vancouver, BC, Canada, V6T 1Z3
| |
Collapse
|
20
|
Impact of lipid nutrition on neural stem/progenitor cells. Stem Cells Int 2013; 2013:973508. [PMID: 24260036 PMCID: PMC3821937 DOI: 10.1155/2013/973508] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2012] [Accepted: 09/09/2013] [Indexed: 11/17/2022] Open
Abstract
The neural system originates from neural stem/progenitor cells (NSPCs). Embryonic NSPCs first proliferate to increase their numbers and then produce neurons and glial cells that compose the complex neural circuits in the brain. New neurons are continually produced even after birth from adult NSPCs in the inner wall of the lateral ventricle and in the hippocampal dentate gyrus. These adult-born neurons are involved in various brain functions, including olfaction-related functions, learning and memory, pattern separation, and mood control. NSPCs are regulated by various intrinsic and extrinsic factors. Diet is one of such important extrinsic factors. Of dietary nutrients, lipids are important because they constitute the cell membrane, are a source of energy, and function as signaling molecules. Metabolites of some lipids can be strong lipid mediators that also regulate various biological activities. Recent findings have revealed that lipids are important regulators of both embryonic and adult NSPCs. We and other groups have shown that lipid signals including fat, fatty acids, their metabolites and intracellular carriers, cholesterol, and vitamins affect proliferation and differentiation of embryonic and adult NSPCs. A better understanding of the NSPCs regulation by lipids may provide important insight into the neural development and brain function.
Collapse
|
21
|
Platel JC, Kelsch W. Role of NMDA receptors in adult neurogenesis: an ontogenetic (re)view on activity-dependent development. Cell Mol Life Sci 2013; 70:3591-601. [PMID: 23397131 PMCID: PMC11113726 DOI: 10.1007/s00018-013-1262-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2012] [Revised: 12/18/2012] [Accepted: 01/03/2013] [Indexed: 12/27/2022]
Abstract
It is now widely accepted that neurogenesis continues throughout life. Accumulating evidence suggests that neurotransmitters are essential signaling molecules that control the different steps of neurogenesis. Nevertheless, we are only beginning to understand the precise role of neurotransmitter receptors and in particular excitatory glutamatergic transmission in the differentiation of adult-born neurons. Recent technical advances allow single-cell gene deletion to study cell-autonomous effects during the maturation of adult-born neurons. Single-cell gene deletion overcomes some of the difficulties in interpreting global gene deletion effects on entire brain areas or systemic pharmacological approaches that might result in compensatory circuit effects. The aim of this review is to summarize recent advances in the understanding of the role of NMDA receptors (NMDARs) during the differentiation of adult-born neurons and put them in perspective with previous findings on cortical development.
Collapse
Affiliation(s)
- Jean-Claude Platel
- Grenoble Institut des Neurosciences U836, Université Joseph Fourier, Site Santé La Tronche BP 170, 38042 Grenoble Cedex 9, France
| | - Wolfgang Kelsch
- Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, J5, 68159 Mannheim, Germany
| |
Collapse
|
22
|
Sultan S, Gebara EG, Moullec K, Toni N. D-serine increases adult hippocampal neurogenesis. Front Neurosci 2013; 7:155. [PMID: 24009551 PMCID: PMC3756301 DOI: 10.3389/fnins.2013.00155] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2013] [Accepted: 08/12/2013] [Indexed: 12/16/2022] Open
Abstract
Adult hippocampal neurogenesis results in the continuous formation of new neurons and is a process of brain plasticity involved in learning and memory. The neurogenic niche regulates the stem cell proliferation and the differentiation and survival of new neurons and a major contributor to the neurogenic niche are astrocytes. Among the molecules secreted by astrocytes, D-serine is an important gliotransmitter and is a co-agonist of the glutamate, N-methyl-D-aspartate (NMDA) receptor. D-serine has been shown to enhance the proliferation of neural stem cells in vitro, but its effect on adult neurogenesis in vivo is unknown. Here, we tested the effect of exogenous administration of D-serine on adult neurogenesis in the mouse dentate gyrus. We found that 1 week of treatment with D-serine increased cell proliferation in vivo and in vitro and increased the density of neural stem cells and transit amplifying progenitors. Furthermore, D-serine increased the survival of newborn neurons. Together, these results indicate that D-serine treatment resulted in the improvement of several steps of adult neurogenesis in vivo.
Collapse
Affiliation(s)
- Sebastien Sultan
- Department of Fundamental Neurosciences, University of Lausanne Lausanne, Switzerland
| | | | | | | |
Collapse
|
23
|
Impact of N-tau on adult hippocampal neurogenesis, anxiety, and memory. Neurobiol Aging 2013; 34:2551-63. [PMID: 23769395 DOI: 10.1016/j.neurobiolaging.2013.05.010] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2012] [Revised: 04/11/2013] [Accepted: 05/08/2013] [Indexed: 01/12/2023]
Abstract
Different pathological tau species are involved in memory loss in Alzheimer's disease, the most common cause of dementia among older people. However, little is known about how tau pathology directly affects adult hippocampal neurogenesis, a unique form of structural plasticity implicated in hippocampus-dependent spatial learning and mood-related behavior. To this aim, we generated a transgenic mouse model conditionally expressing a pathological tau fragment (26-230 aa of the longest human tau isoform, or N-tau) in nestin-positive stem/progenitor cells. We found that N-tau reduced the proliferation of progenitor cells in the adult dentate gyrus, reduced cell survival and increased cell death by a caspase-3-independent mechanism, and recruited microglia. Although the number of terminally differentiated neurons was reduced, these showed an increased dendritic arborization and spine density. This resulted in an increase of anxiety-related behavior and an impairment of episodic-like memory, whereas less complex forms of spatial learning remained unaltered. Understanding how pathological tau species directly affect neurogenesis is important for developing potential therapeutic strategies to direct neurogenic instructive cues for hippocampal function repair.
Collapse
|
24
|
Hofacer RD, Deng M, Ward CG, Joseph B, Hughes EA, Jiang C, Danzer SC, Loepke AW. Cell age-specific vulnerability of neurons to anesthetic toxicity. Ann Neurol 2013; 73:695-704. [PMID: 23526697 DOI: 10.1002/ana.23892] [Citation(s) in RCA: 87] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2012] [Revised: 01/29/2013] [Accepted: 03/01/2013] [Indexed: 11/08/2022]
Abstract
OBJECTIVE Anesthetics have been linked to widespread neuronal cell death in neonatal animals. Epidemiological human studies have associated early childhood anesthesia with long-term neurobehavioral abnormalities, raising substantial concerns that anesthetics may cause similar cell death in young children. However, key aspects of the phenomenon remain unclear, such as why certain neurons die, whereas immediately adjacent neurons are seemingly unaffected, and why the immature brain is exquisitely vulnerable, whereas the mature brain seems resistant. Elucidating these questions is critical for assessing the phenomenon's applicability to humans, defining the susceptible age, predicting vulnerable neuronal populations, and devising mitigating strategies. METHODS This study examines the effects of anesthetic exposure on late- and adult-generated neurons in newborn, juvenile, and adult mice, and characterizes vulnerable cells using birth-dating and immunohistochemical techniques. RESULTS We identify a critical period of cellular developmental during which neurons are susceptible to anesthesia-induced apoptosis. Importantly, we demonstrate that anesthetic neurotoxicity can extend into adulthood in brain regions with ongoing neurogenesis, such as dentate gyrus and olfactory bulb. INTERPRETATION Our findings suggest that anesthetic vulnerability reflects the age of the neuron, not the age of the organism, and therefore may potentially not only be relevant to children but also to adults undergoing anesthesia. This observation further predicts differential heightened regional vulnerability to anesthetic neuroapoptosis to closely follow the distinct regional peaks in neurogenesis. This knowledge may help guide neurocognitive testing of specific neurological domains in humans following exposure to anesthesia, dependent on the individual's age during exposure.
Collapse
Affiliation(s)
- Rylon D Hofacer
- Department of Anesthesiology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | | | | | | | | | | | | | | |
Collapse
|
25
|
Abstract
Plasticity in the adult brain enables lifelong learning. The fundamental mechanism of adult neural plasticity is activity-dependent reorganization of pre-existing structure, in contrast to the widespread cellular proliferation and migration that occurs during development. Whereas adult hippocampal dentate gyrus continuously generates cohorts of neurons, and newborn neurons integrate into the existing neural circuit under the regulation of existing global and local neural activity, demonstrating a unique cellular and synaptic flexibility in adult brain. Exhibiting an enhanced structural and synaptic plasticity during the maturation, adult-born hippocampal neurons may represent a unique population for hippocampal function. Current evidence indicates that lifelong addition of new hippocampal neurons may extend early developmental plasticity to adulthood, which continuously rejuvenates adult brain. We reviewed most recent advancements in exploring the circuit and behavioral role of adult-born hippocampal neurons.
Collapse
Affiliation(s)
- Yan Gu
- Department of Neurobiology and Behavior, Stony Brook University, 100 Nicolls Road, Stony Brook, NY, 11794, USA,
| | | | | |
Collapse
|
26
|
Shin JY, Ahn YH, Paik MJ, Park HJ, Sohn YH, Lee PH. Elevated homocysteine by levodopa is detrimental to neurogenesis in parkinsonian model. PLoS One 2012; 7:e50496. [PMID: 23209759 PMCID: PMC3509089 DOI: 10.1371/journal.pone.0050496] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2012] [Accepted: 10/25/2012] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Modulation of neurogenesis that acts as an endogenous repair mechanism would have a significant impact on future therapeutic strategies for Parkinson's disease (PD). Several studies demonstrated dopaminergic modulation of neurogenesis in the subventricular zone (SVZ) of the adult brain. Levodopa, the gold standard therapy for PD, causes an increase in homocysteine levels that induces neuronal death via N-methyl-D-aspartate (NMDA) receptor. The present study investigated whether elevated homocysteine by levodopa treatment in a parkinsonian model would modulate neurogenesis via NMDA receptor signal cascade and compared the effect of levodopa and pramipexol (PPX) on neurogenic activity. METHODOLOGY/PRINCIPAL FINDINGS Neurogenesis was assessed in vitro using neural progenitor cells (NPCs) isolated from the SVZ and in vivo with the BrdU-injected animal model of PD using 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine. Modulation of homocysteine levels was evaluated using co-cultures of NPCs and astrocytes and PD animals. Immunochemical and Western blot analyses were used to measure neurogenesis and determine the cell death signaling. Levodopa treatment increased release of homocysteine on astrocytes culture media as well as in plasma and brain of PD animals. Increased homocysteine by levodopa led to increased apoptosis of NPCs through the NMDA receptor-dependent the extracellular signal-regulated kinase (ERK) signaling pathways. The administration of a NMDA antagonist significantly attenuated apoptotic cell death in levodopa-treated NPCs and markedly increased the number of BrdU-positive cells in the SVZ of levodopa-treated PD animals. Comparative analysis revealed that PPX treatment significantly increased the number of NPCs and BrdU-positive cells in the SVZ of PD animals compared to levodopa treatment. Our present study demonstrated that increased homocysteine by levodopa has a detrimental effect on neurogenesis through NMDA receptor-mediated ERK signaling pathway. CONCLUSIONS/SIGNIFICANCE Modulation of levodopa-induced elevated homocysteine by NMDA antagonist or dopamine agonist has a clinical relevance for PD treatment in terms of adult neurogenesis.
Collapse
Affiliation(s)
- Jin Young Shin
- Department of Neurology and Brain Research Institute, Yonsei University College of Medicine, Seoul, Korea
| | - Young-Hwan Ahn
- Department of Neurosurgery, Ajou University School of Medicine, Suwon, South Korea
| | - Man-Jeong Paik
- Department of Molecular Science and Technology, Ajou University, Suwon, South Korea
| | - Hyun Jung Park
- Department of Neurology and Brain Research Institute, Yonsei University College of Medicine, Seoul, Korea
- Severance Biomedical Science Institute, Yonsei University, Seoul, Korea
| | - Young H. Sohn
- Department of Neurology and Brain Research Institute, Yonsei University College of Medicine, Seoul, Korea
| | - Phil Hyu Lee
- Department of Neurology and Brain Research Institute, Yonsei University College of Medicine, Seoul, Korea
- Severance Biomedical Science Institute, Yonsei University, Seoul, Korea
- * E-mail:
| |
Collapse
|
27
|
Neurogenic differentiation of human adipose-derived stem cells: relevance of different signaling molecules, transcription factors, and key marker genes. Gene 2012; 511:427-36. [PMID: 23000064 DOI: 10.1016/j.gene.2012.09.038] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2012] [Revised: 07/16/2012] [Accepted: 09/12/2012] [Indexed: 01/13/2023]
Abstract
Since numerous diseases affect the central nervous system and it has limited self-repair capability, a great interest in using stem cells as an alternative cell source is generated. Previous reports have shown the differentiation of adipose-derived stem cells in neuron-like cells and it has also been proved that the expression pattern of patterning, proneural, and neural factors, such as Pax6, Mash1, Ngn2, NeuroD1, Tbr2 and Tbr1, regulates and defines adult neurogenesis. Regarding this, we hypothesize that a functional parallelism between adult neurogenesis and neuronal differentiation of human adipose-derived stem cells exists. In this study we differentiate human adipose-derived stem cells into neuron-like cells and analyze the expression pattern of different patterning, proneural, neural and neurotransmitter genes, before and after neuronal differentiation. The neuron-like cells expressed neuronal markers, patterning and proneural factors characteristics of intermediate stages of neuronal differentiation. Thus we demonstrated that it is possible to differentiate adipose-derived stem cells in vitro into immature neuron-like cells and that this process is regulated in a similar way to adult neurogenesis. This may contribute to elucidate molecular mechanisms involved in neuronal differentiation of adult human non-neural cells, in aid of the development of potential therapeutic tools for diseases of the nervous system.
Collapse
|
28
|
Faigle R, Song H. Signaling mechanisms regulating adult neural stem cells and neurogenesis. Biochim Biophys Acta Gen Subj 2012; 1830:2435-48. [PMID: 22982587 DOI: 10.1016/j.bbagen.2012.09.002] [Citation(s) in RCA: 253] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2012] [Revised: 08/11/2012] [Accepted: 09/05/2012] [Indexed: 12/13/2022]
Abstract
BACKGROUND Adult neurogenesis occurs throughout life in discrete regions of the mammalian brain and is tightly regulated via both extrinsic environmental influences and intrinsic genetic factors. In recent years, several crucial signaling pathways have been identified in regulating self-renewal, proliferation, and differentiation of neural stem cells, as well as migration and functional integration of developing neurons in the adult brain. SCOPE OF REVIEW Here we review our current understanding of signaling mechanisms, including Wnt, notch, sonic hedgehog, growth and neurotrophic factors, bone morphogenetic proteins, neurotransmitters, transcription factors, and epigenetic modulators, and crosstalk between these signaling pathways in the regulation of adult neurogenesis. We also highlight emerging principles in the vastly growing field of adult neural stem cell biology and neural plasticity. MAJOR CONCLUSIONS Recent methodological advances have enabled the field to identify signaling mechanisms that fine-tune and coordinate neurogenesis in the adult brain, leading to a better characterization of both cell-intrinsic and environmental cues defining the neurogenic niche. Significant questions related to niche cell identity and underlying regulatory mechanisms remain to be fully addressed and will be the focus of future studies. GENERAL SIGNIFICANCE A full understanding of the role and function of individual signaling pathways in regulating neural stem cells and generation and integration of newborn neurons in the adult brain may lead to targeted new therapies for neurological diseases in humans. This article is part of a Special Issue entitled Biochemistry of Stem Cells.
Collapse
Affiliation(s)
- Roland Faigle
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA.
| | | |
Collapse
|
29
|
Hodes GE, Brookshire BR, Hill-Smith TE, Teegarden SL, Berton O, Lucki I. Strain differences in the effects of chronic corticosterone exposure in the hippocampus. Neuroscience 2012; 222:269-80. [PMID: 22735575 DOI: 10.1016/j.neuroscience.2012.06.017] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2012] [Revised: 05/18/2012] [Accepted: 06/07/2012] [Indexed: 10/28/2022]
Abstract
Stress hormones are thought to be involved in the etiology of depression, in part, because animal models show they cause morphological damage to the brain, an effect that can be reversed by chronic antidepressant treatment. The current study examined two mouse strains selected for naturalistic variation of tissue regeneration after injury for resistance to the effects of chronic corticosterone (CORT) exposure on cell proliferation and neurotrophin mobilization. The wound healer MRL/MpJ and control C57BL/6J mice were implanted subcutaneously with pellets that released CORT for 7 days. MRL/MpJ mice were resistant to reductions of hippocampal cell proliferation by chronic exposure to CORT when compared to vulnerable C57BL/6J mice. Chronic CORT exposure also reduced protein levels of brain-derived neurotrophic factor (BDNF) in the hippocampus of C57BL/6J but not MRL/MpJ mice. CORT pellet exposure increased circulating levels of CORT in the plasma of both strains in a dose-dependent manner although MRL/MpJ mice may have larger changes from baseline. The strains did not differ in circulating levels of corticosterone binding globulin (CBG). There were also no strain differences in CORT levels in the hippocampus, nor did CORT exposure alter glucocorticoid receptor or mineralocorticoid receptor expression in a strain-dependent manner. Strain differences were found in the N-methyl-D-aspartate (NMDA) receptor, and BDNF I and IV promoters. Strain and CORT exposure interacted to alter tropomyosine-receptor-kinase B (TrkB) expression and this may be a potential mechanism protecting MRL/MpJ mice. In addition, differences in the inflammatory response of matrix metalloproteinases (MMPs) may also contribute to these strain differences in resistance to the deleterious effects of CORT to the brain.
Collapse
Affiliation(s)
- G E Hodes
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA 19104, United States
| | | | | | | | | | | |
Collapse
|
30
|
Fan H, Gao J, Wang W, Li X, Xu T, Yin X. Expression of NMDA receptor and its effect on cell proliferation in the subventricular zone of neonatal rat brain. Cell Biochem Biophys 2012; 62:305-16. [PMID: 21964542 DOI: 10.1007/s12013-011-9302-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Abstract
We investigated the involvement of N-methyl D-aspartate receptor (NMDAR) in neurogenesis of rat's subventricular zone (SVZ). For this purpose, we determined expression of the NMDAR subunits NR1, NR2A, and NR2B in SVZ of the neonatal Sprague-Dawley rats using immunohistochemical techniques. All three NMDAR subunits were expressed during postnatal day (PND)-1 to PND-28 whereas each subunit showed a distinct expression pattern. We also examined the functional effect of this receptor on cell proliferation in this region and, in this regard, the animals received either intraperitoneal injection of NMDAR agonist NMDA (2 mg/kg/day) or selective non-competitive NMDAR antagonist MK-801 (10 mg/kg) or NR2B antagonist Ro25-6981 (40 mg/kg), respectively, at PND-3. A significant developmental increase of the total cell density was observed at PND-7 (P < 0.05) while proliferating cell nuclear antigen-positive cell density was significantly increased at PND-14 (P < 0.05) and at PND-28 (P < 0.05) in the SVZ after NMDA (2 mg/kg/day) injection. Our data show that the NMDAR activation promoted the cell proliferation in SVZ during the neonatal period. We, therefore, inferred that NMDAR is expressed in SVZ of the neonatal rat brain and can promote neurogenesis, as through cell proliferation process in that region, and can thus be used as a potential therapeutic target in neurodegenerative diseases.
Collapse
Affiliation(s)
- Hongbin Fan
- Jiangsu Key Laboratory of Neurodegeneration, Department of Pharmacology, Nanjing Medical University, Nanjing 210029, China
| | | | | | | | | | | |
Collapse
|
31
|
Jhaveri DJ, Taylor CJ, Bartlett PF. Activation of different neural precursor populations in the adult hippocampus: Does this lead to new neurons with discrete functions? Dev Neurobiol 2012; 72:1044-58. [DOI: 10.1002/dneu.22027] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
|
32
|
Bonfanti L, Nacher J. New scenarios for neuronal structural plasticity in non-neurogenic brain parenchyma: the case of cortical layer II immature neurons. Prog Neurobiol 2012; 98:1-15. [PMID: 22609484 DOI: 10.1016/j.pneurobio.2012.05.002] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2011] [Revised: 04/25/2012] [Accepted: 05/08/2012] [Indexed: 11/20/2022]
Abstract
The mammalian central nervous system, due to its interaction with the environment, must be endowed with plasticity. Conversely, the nervous tissue must be substantially static to ensure connectional invariability. Structural plasticity can be viewed as a compromise between these requirements. In adult mammals, brain structural plasticity is strongly reduced with respect to other animal groups in the phylogenetic tree. It persists under different forms, which mainly consist of remodeling of neuronal shape and connectivity, and, to a lesser extent, the production of new neurons. Adult neurogenesis is mainly restricted within two neurogenic niches, yet some gliogenic and neurogenic processes also occur in the so-called non-neurogenic tissue, starting from parenchymal progenitors. In this review we focus on a population of immature, non-newly generated neurons in layer II of the cerebral cortex, which were previously thought to be newly generated since they heavily express the polysialylated form of the neural cell adhesion molecule and doublecortin. These unusual neurons exhibit characteristics defining an additional type of structural plasticity, different from either synaptic plasticity or adult neurogenesis. Evidences concerning their morphology, antigenic features, ultrastructure, phenotype, origin, fate, and reaction to different kind of stimulations are gathered and analyzed. Their possible role is discussed in the context of an enriched complexity and heterogeneity of mammalian brain structural plasticity.
Collapse
Affiliation(s)
- Luca Bonfanti
- Neuroscience Institute Cavalieri Ottolenghi (NICO), Regione Gonzole 10, Orbassano (TO), and Department of Veterinary Morphophysiology, University of Turin, Turin, Italy.
| | | |
Collapse
|
33
|
Yao J, Mu Y, Gage FH. Neural stem cells: mechanisms and modeling. Protein Cell 2012; 3:251-61. [PMID: 22549585 PMCID: PMC4875476 DOI: 10.1007/s13238-012-2033-6] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2012] [Accepted: 02/21/2012] [Indexed: 12/17/2022] Open
Abstract
In the adult brain, neural stem cells have been found in two major niches: the dentate gyrus and the subventricular zone [corrected]. Neurons derived from these stem cells contribute to learning, memory, and the autonomous repair of the brain under pathological conditions. Hence, the physiology of adult neural stem cells has become a significant component of research on synaptic plasticity and neuronal disorders. In addition, the recently developed induced pluripotent stem cell technique provides a powerful tool for researchers engaged in the pathological and pharmacological study of neuronal disorders. In this review, we briefly summarize the research progress in neural stem cells in the adult brain and in the neuropathological application of the induced pluripotent stem cell technique.
Collapse
Affiliation(s)
- Jun Yao
- Laboratory of Genetics, The Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | | | | |
Collapse
|
34
|
Genius J, Benninghoff J, Reuter N, Braun I, Giegling I, Hartmann A, Möller HJ, Rujescu D. Dysequilibrium of neuronal proliferation and apoptosis in a pharmacological animal model of psychosis. Methods 2012; 56:519-27. [DOI: 10.1016/j.ymeth.2012.04.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2012] [Revised: 04/03/2012] [Accepted: 04/04/2012] [Indexed: 11/26/2022] Open
|
35
|
Gilley JA, Kernie SG. Excitatory amino acid transporter 2 and excitatory amino acid transporter 1 negatively regulate calcium-dependent proliferation of hippocampal neural progenitor cells and are persistently upregulated after injury. Eur J Neurosci 2011; 34:1712-23. [PMID: 22092549 DOI: 10.1111/j.1460-9568.2011.07888.x] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Using a transgenic mouse (Mus musculus) in which nestin-expressing progenitors are labeled with enhanced green fluorescent protein, we previously characterized the expression of excitatory amino acid transporter 2 (GltI) and excitatory amino acid transporter 1 (Glast) on early neural progenitors in vivo. To address their functional role in this cell population, we manipulated their expression in P7 neurospheres isolated from the dentate gyrus. We observed that knockdown of GltI or Glast was associated with decreased bromodeoxyuridine incorporation and neurosphere formation. Moreover, we determined that both glutamate transporters regulated progenitor proliferation in a calcium-dependent and metabotropic glutamate receptor-dependent manner. To address the relevance of this in vivo, we utilized models of acquired brain injury, which are known to induce hippocampal neurogenesis. We observed that GltI and Glast were specifically upregulated in progenitors following brain injury, and that this increased expression was maintained for many weeks. Additionally, we found that recurrently injured animals with increased expression of glutamate transporters within the progenitor population were resistant to subsequent injury-induced proliferation. These findings demonstrate that GltI and Glast negatively regulate calcium-dependent proliferation in vitro and that their upregulation after injury is associated with decreased proliferation after brain trauma.
Collapse
Affiliation(s)
- Jennifer A Gilley
- Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | | |
Collapse
|
36
|
Li M, Zhang DQ, Wang XZ, Xu TJ. NR2B-containing NMDA receptors promote neural progenitor cell proliferation through CaMKIV/CREB pathway. Biochem Biophys Res Commun 2011; 411:667-72. [DOI: 10.1016/j.bbrc.2011.06.170] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2011] [Accepted: 06/27/2011] [Indexed: 01/09/2023]
|
37
|
Cheyne JE, Grant L, Butler-Munro C, Foote JW, Connor B, Montgomery JM. Synaptic integration of newly generated neurons in rat dissociated hippocampal cultures. Mol Cell Neurosci 2011; 47:203-14. [DOI: 10.1016/j.mcn.2011.04.006] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2010] [Revised: 04/20/2011] [Accepted: 04/26/2011] [Indexed: 10/18/2022] Open
|
38
|
|
39
|
Choi JH, Yoo KY, Lee CH, Yi SS, Yoo DY, Seong JK, Yoon YS, Hwang IK, Won MH. Effects of treadmill exercise combined with MK 801 treatment on neuroblast differentiation in the dentate gyrus in rats. Cell Mol Neurobiol 2011; 31:285-92. [PMID: 21052826 PMCID: PMC11498510 DOI: 10.1007/s10571-010-9619-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2010] [Accepted: 10/12/2010] [Indexed: 02/02/2023]
Abstract
N-methyl-D-aspartate receptor (NR) is involved in activity-dependent synaptic plasticity, such as associative long-term potentiation, and in related central functions, such as learning and memory. In this study, we observed effects of treadmill exercise on NR1 and doublecortin (DCX, a marker for neuroblast differentiation) in the subgranular zone of the dentate gyrus (DG). At 6 weeks of age, rats were put on a treadmill with or without running for 1 h/day for 5 consecutive days at 22 m/min for 5 weeks. Exercise increased NR1 immunoreactivity and protein level in the hippocampus. To identify the correlations between NR and neuroblasts, we intraperitoneally administered a NR antagonist, MK-801, to the exercised rats. MK-801 treatment reduced NR1 protein level in the hippocampus of the exercised rats. In addition, in the MK-801-treated group, the number of DCX cells was significantly decreased in the subgranular zone of the DG. These results suggest that NR may be one of the important factors that modulate neuroblast differentiation during exercise in rats.
Collapse
Affiliation(s)
- Jung Hoon Choi
- Department of Anatomy, College of Veterinary Medicine, Kangwon National University, Chuncheon, 200-701 South Korea
| | - Ki-Yeon Yoo
- Institute of Natural Medicine, Hallym University, Chuncheon, 200-702 South Korea
| | - Choong Hyun Lee
- Laboratory of Veterinary Pharmacology, College of Veterinary Medicine, Seoul National University, Seoul, 151-742 South Korea
| | - Sun Shin Yi
- Department of Anatomy and Cell Biology, College of Veterinary Medicine, and Research Institute for Veterinary Science, Seoul National University, Seoul, 151-742 South Korea
| | - Dae Young Yoo
- Department of Anatomy and Cell Biology, College of Veterinary Medicine, and Research Institute for Veterinary Science, Seoul National University, Seoul, 151-742 South Korea
| | - Je Kyung Seong
- Department of Anatomy and Cell Biology, College of Veterinary Medicine, and Research Institute for Veterinary Science, Seoul National University, Seoul, 151-742 South Korea
| | - Yeo Sung Yoon
- Department of Anatomy and Cell Biology, College of Veterinary Medicine, and Research Institute for Veterinary Science, Seoul National University, Seoul, 151-742 South Korea
| | - In Koo Hwang
- Department of Anatomy and Cell Biology, College of Veterinary Medicine, and Research Institute for Veterinary Science, Seoul National University, Seoul, 151-742 South Korea
| | - Moo-Ho Won
- Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon, 200-701 South Korea
| |
Collapse
|
40
|
Kempermann G, Fabel K, Ehninger D, Babu H, Leal-Galicia P, Garthe A, Wolf SA. Why and how physical activity promotes experience-induced brain plasticity. Front Neurosci 2010; 4:189. [PMID: 21151782 PMCID: PMC3000002 DOI: 10.3389/fnins.2010.00189] [Citation(s) in RCA: 206] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2010] [Accepted: 10/23/2010] [Indexed: 12/17/2022] Open
Abstract
Adult hippocampal neurogenesis is an unusual case of brain plasticity, since new neurons (and not just neurites and synapses) are added to the network in an activity-dependent way. At the behavioral level the plasticity-inducing stimuli include both physical and cognitive activity. In reductionistic animal studies these types of activity can be studied separately in paradigms like voluntary wheel running and environmental enrichment. In both of these, adult neurogenesis is increased but the net effect is primarily due to different mechanisms at the cellular level. Locomotion appears to stimulate the precursor cells, from which adult neurogenesis originates, to increased proliferation and maintenance over time, whereas environmental enrichment, as well as learning, predominantly promotes survival of immature neurons, that is the progeny of the proliferating precursor cells. Surprisingly, these effects are additive: boosting the potential for adult neurogenesis by physical activity increases the recruitment of cells following cognitive stimulation in an enriched environment. Why is that? We argue that locomotion actually serves as an intrinsic feedback mechanism, signaling to the brain, including its neural precursor cells, increasing the likelihood of cognitive challenges. In the wild (other than in front of a TV), no separation of physical and cognitive activity occurs. Physical activity might thus be much more than a generally healthy garnish to leading "an active life" but an evolutionarily fundamental aspect of "activity," which is needed to provide the brain and its systems of plastic adaptation with the appropriate regulatory input and feedback.
Collapse
Affiliation(s)
- Gerd Kempermann
- Center for Regenerative Therapies Dresden, German Research FoundationDresden, Germany
- German Center for Neurodegenerative DiseasesDresden, Germany
| | - Klaus Fabel
- Center for Regenerative Therapies Dresden, German Research FoundationDresden, Germany
- German Center for Neurodegenerative DiseasesDresden, Germany
| | - Dan Ehninger
- German Center for Neurodegenerative DiseasesBonn, Germany
| | - Harish Babu
- Department of Neurosurgery, Stanford UniversityStanford, CA, USA
| | - Perla Leal-Galicia
- Center for Regenerative Therapies Dresden, German Research FoundationDresden, Germany
| | - Alexander Garthe
- Center for Regenerative Therapies Dresden, German Research FoundationDresden, Germany
| | - Susanne A. Wolf
- Department of Cell and Neurobiology, Institute of Anatomy, University ZürichZürich, Switzerland
| |
Collapse
|
41
|
Bunk EC, König HG, Bonner HP, Kirby BP, Prehn JH. NMDA-induced injury of mouse organotypic hippocampal slice cultures triggers delayed neuroblast proliferation in the dentate gyrus: An in vitro model for the study of neural precursor cell proliferation. Brain Res 2010; 1359:22-32. [DOI: 10.1016/j.brainres.2010.05.024] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2009] [Revised: 05/03/2010] [Accepted: 05/06/2010] [Indexed: 01/18/2023]
|
42
|
Spatial learning sculpts the dendritic arbor of adult-born hippocampal neurons. Proc Natl Acad Sci U S A 2010; 107:7963-8. [PMID: 20375283 DOI: 10.1073/pnas.0914613107] [Citation(s) in RCA: 161] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Neurogenesis in the hippocampus is characterized by the birth of thousand of cells that generate neurons throughout life. The fate of these adult newborn neurons depends on life experiences. In particular, spatial learning promotes the survival and death of new neurons. Whether learning influences the development of the dendritic tree of the surviving neurons (a key parameter for synaptic integration and signal processing) is unknown. Here we show that learning accelerates the maturation of their dendritic trees and their integration into the hippocampal network. We demonstrate that these learning effects on dendritic arbors are homeostatically regulated, persist for several months, and are specific to neurons born during adulthood. Finally, we show that this dendritic shaping depends on the cognitive demand and relies on the activation of NMDA receptors. In the search for the structural changes underlying long-term memory, these findings lead to the conclusion that shaping neo-networks is important in forming spatial memories.
Collapse
|
43
|
Platel JC, Dave KA, Gordon V, Lacar B, Rubio ME, Bordey A. NMDA receptors activated by subventricular zone astrocytic glutamate are critical for neuroblast survival prior to entering a synaptic network. Neuron 2010; 65:859-72. [PMID: 20346761 PMCID: PMC2861893 DOI: 10.1016/j.neuron.2010.03.009] [Citation(s) in RCA: 175] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/26/2010] [Indexed: 11/23/2022]
Abstract
Even before integrating into existing circuitry, adult-born neurons express receptors for neurotransmitters, but the intercellular mechanisms and their impact on neurogenesis remain largely unexplored. Here, we show that neuroblasts born in the postnatal subventricular zone (SVZ) acquire NMDA receptors (NMDARs) during their migration to the olfactory bulb. Along their route, neuroblasts are ensheathed by astrocyte-like cells expressing vesicular glutamate release machinery. Increasing calcium in these specialized astrocytes induced NMDAR activity in neuroblasts, and blocking astrocytic vesicular release eliminated spontaneous NMDAR activity. Single-cell knockout of NMDARs using neonatal electroporation resulted in neuroblast apoptosis at the time of NMDAR acquisition. This cumulated in a 40% loss of neuroblasts along their migratory route, demonstrating that NMDAR acquisition is critical for neuroblast survival prior to entering a synaptic network. In addition, our findings suggest an unexpected mechanism wherein SVZ astrocytes use glutamate signaling through NMDARs to control the number of adult-born neurons reaching their final destination.
Collapse
Affiliation(s)
- Jean-Claude Platel
- Departments of Neurosurgery, and Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, CT 06520-8082
| | - Kathleen A. Dave
- Departments of Neurosurgery, and Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, CT 06520-8082
| | - Valerie Gordon
- Departments of Neurosurgery, and Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, CT 06520-8082
| | - Benjamin Lacar
- Departments of Neurosurgery, and Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, CT 06520-8082
| | - Maria E. Rubio
- Department of Physiology and Neurobiology, University of Connecticut, Storrs, Connecticut 06269
| | - Angélique Bordey
- Departments of Neurosurgery, and Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, CT 06520-8082
| |
Collapse
|
44
|
Jhaveri DJ, Mackay EW, Hamlin AS, Marathe SV, Nandam LS, Vaidya VA, Bartlett PF. Norepinephrine directly activates adult hippocampal precursors via beta3-adrenergic receptors. J Neurosci 2010; 30:2795-806. [PMID: 20164362 PMCID: PMC2837927 DOI: 10.1523/jneurosci.3780-09.2010] [Citation(s) in RCA: 129] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2009] [Revised: 12/01/2009] [Accepted: 01/11/2010] [Indexed: 11/21/2022] Open
Abstract
Adult hippocampal neurogenesis is a critical form of cellular plasticity that is greatly influenced by neural activity. Among the neurotransmitters that are widely implicated in regulating this process are serotonin and norepinephrine, levels of which are modulated by stress, depression and clinical antidepressants. However, studies to date have failed to address a direct role for either neurotransmitter in regulating hippocampal precursor activity. Here we show that norepinephrine but not serotonin directly activates self-renewing and multipotent neural precursors, including stem cells, from the hippocampus of adult mice. Mechanistically, we provide evidence that beta(3)-adrenergic receptors, which are preferentially expressed on a Hes5-expressing precursor population in the subgranular zone (SGZ), mediate this norepinephrine-dependent activation. Moreover, intrahippocampal injection of a selective beta(3)-adrenergic receptor agonist in vivo increases the number of proliferating cells in the SGZ. Similarly, systemic injection of the beta-adrenergic receptor agonist isoproterenol not only results in enhancement of proliferation in the SGZ but also leads to an increase in the percentage of nestin/glial fibrillary acidic protein double-positive neural precursors in vivo. Finally, using a novel ex vivo "slice-sphere" assay that maintains an intact neurogenic niche, we demonstrate that antidepressants that selectively block the reuptake of norepinephrine, but not serotonin, robustly increase hippocampal precursor activity via beta-adrenergic receptors. These findings suggest that the activation of neurogenic precursors and stem cells via beta(3)-adrenergic receptors could be a potent mechanism to increase neuronal production, providing a putative target for the development of novel antidepressants.
Collapse
MESH Headings
- Adrenergic Agonists/pharmacology
- Adrenergic Antagonists/pharmacology
- Adrenergic alpha-Agonists/pharmacology
- Adrenergic beta-3 Receptor Agonists
- Adrenergic beta-3 Receptor Antagonists
- Adult Stem Cells/drug effects
- Animals
- Animals, Newborn
- Cells, Cultured
- Flow Cytometry/methods
- Glial Fibrillary Acidic Protein/metabolism
- Green Fluorescent Proteins/genetics
- Hippocampus/cytology
- In Vitro Techniques
- Male
- Mice
- Mice, Inbred C57BL
- Mice, Transgenic
- Myelin Basic Protein/metabolism
- Norepinephrine/pharmacology
- Rats
- Rats, Wistar
- Receptors, Adrenergic, beta-3/genetics
- Receptors, Adrenergic, beta-3/physiology
- Statistics, Nonparametric
- Tubulin/metabolism
Collapse
Affiliation(s)
- Dhanisha J. Jhaveri
- The Queensland Brain Institute, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Eirinn W. Mackay
- The Queensland Brain Institute, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Adam S. Hamlin
- The Queensland Brain Institute, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Swananda V. Marathe
- Department of Biological Sciences, Tata Institute of Fundamental Research, Mumbai 400005, India
| | - L. Sanjay Nandam
- The Queensland Brain Institute, The University of Queensland, Brisbane, Queensland 4072, Australia
- The Prince Charles Hospital, Brisbane, Queensland 4032, Australia, and
| | - Vidita A. Vaidya
- Department of Biological Sciences, Tata Institute of Fundamental Research, Mumbai 400005, India
| | - Perry F. Bartlett
- The Queensland Brain Institute, The University of Queensland, Brisbane, Queensland 4072, Australia
| |
Collapse
|
45
|
Pathania M, Yan LD, Bordey A. A symphony of signals conducts early and late stages of adult neurogenesis. Neuropharmacology 2010; 58:865-76. [PMID: 20097213 DOI: 10.1016/j.neuropharm.2010.01.010] [Citation(s) in RCA: 84] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2009] [Revised: 01/15/2010] [Accepted: 01/15/2010] [Indexed: 02/06/2023]
Abstract
Neurogenesis is continually occurring in two regions within the mammalian central nervous system (CNS) and increasing evidence suggests that it is important for selective learning and memory. How this plasticity is maintained in isolated niches within mature networks has been extensively studied in recent years, and a large body of evidence has accumulated describing many different regulatory factors and points of regulation. In this review, we attempt to organize the current research by summarizing findings affecting early neurogenesis: during proliferation, fate commitment and migration, versus late neurogenesis: including dendritic development, synaptic integration, and survival. We discuss the roles of three different classes of factors regulating early and late phases of neurogenesis: intrinsic factors, extrinsic factors, and neurotransmitters. Finally, we suggest that neurotransmitters may act upstream from extracellular other factors and cell-intrinsic mechanisms by coupling network activity to the niche microenvironment and intracellular machinery to ultimately regulate neurogenesis.
Collapse
Affiliation(s)
- Manavendra Pathania
- Departments of Neurosurgery, and Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, Connecticut 06520-8082, USA
| | | | | |
Collapse
|
46
|
Abstract
The identification of neural stem cells (NSCs) and their contribution to continuous neurogenesis has shown that the hippocampus and olfactory bulb are plastic. Brain plasticity, achieved at the level of cell genesis, has an essential role in the maintenance of brain homeostasis. Via combinatorial functions of extrinsic signals and intrinsic programs, adult neurogenesis is tightly regulated in a specialized microenvironment, a niche. Misregulated neurogenesis is detrimental to normal brain functions and, in extreme cases, pathogenic. Hence, understanding signaling in adult neurogenesis is not only important to understand the physiological roles of neurogenesis, but also to provide knowledge that is essential for developing therapeutic applications using NSCs to intervene in the progression of brain diseases.
Collapse
Affiliation(s)
- Hoonkyo Suh
- Laboratory of Genetics, The Salk Institute for Biological Studies, La Jolla, California 92037, USA
| | | | | |
Collapse
|
47
|
Wegner F, Kraft R, Busse K, Schaarschmidt G, Härtig W, Schwarz SC, Schwarz J. Glutamate receptor properties of human mesencephalic neural progenitor cells: NMDA enhances dopaminergic neurogenesisin vitro. J Neurochem 2009; 111:204-16. [DOI: 10.1111/j.1471-4159.2009.06315.x] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
48
|
Namba T, Maekawa M, Yuasa S, Kohsaka S, Uchino S. The Alzheimer's disease drug memantine increases the number of radial glia-like progenitor cells in adult hippocampus. Glia 2009; 57:1082-90. [PMID: 19115386 DOI: 10.1002/glia.20831] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
New neurons are continuously generated in the hippocampus of the adult mammalian brain, and N-methyl-D-aspartate receptor (NMDA-R) antagonists have been found to increase the number of newly generated neurons in the dentate gyrus (DG) of the adult hippocampus. In this study, we examined the effect of memantine, an NMDA-R antagonist that is clinically used for the treatment of Alzheimer's disease, on primary progenitor cells exhibiting a radial glia-like (RGL) morphology in the DG. We injected 3-month-old mice with memantine (50 mg/kg body weight, intraperitoneally [i.p.]); 3 days later, we injected the mice with 5-bromo-2-deoxyuridine (BrdU; 75 mg/kg body weight, i.p.). We then counted the number of BrdU-labeled RGL progenitor cells in the DG 1 or 7 days after the BrdU-injection. The number of BrdU-labeled RGL progenitor cells had increased significantly by 5.1-fold on day 1 and by 13.7-fold on day 7 after BrdU-injection. Immunohistochemical staining revealed that the BrdU-labeled RGL progenitor cells expressed two primary progenitor cell marker proteins, nestin and Sox2. These results clearly demonstrated that memantine promotes the proliferation of RGL progenitor cells. We also found that memantine increased the ratio of horizontally aligned RGL progenitor cells, which are probably produced by symmetric division. These findings suggest that memantine increases the proliferation of primary progenitor cells and expands the primary progenitor cell pool in the adult hippocampus by stimulating symmetric division.
Collapse
Affiliation(s)
- Takashi Namba
- Department of Neurochemistry, National Institute of Neuroscience, Kodaira, Tokyo, Japan
| | | | | | | | | |
Collapse
|
49
|
Fernández-Lamo I, Montero-Pedrazuela A, Delgado-García JM, Guadaño-Ferraz A, Gruart A. Effects of thyroid hormone replacement on associative learning and hippocampal synaptic plasticity in adult hypothyroid rats. Eur J Neurosci 2009; 30:679-92. [PMID: 19686470 DOI: 10.1111/j.1460-9568.2009.06862.x] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Activity-dependent changes taking place at the hippocampal perforant pathway-dentate gyrus synapse during classical eyeblink conditioning were recorded in adult thyroidectomized (hypothyroid) and control (euthyroid) rats, and in animals treated with thyroid hormones 20 days after thyroidectomy (recovery rats). The aim was to determine the contribution of thyroid hormones and the consequences of adult-onset hypothyroidism to both associative learning and the physiological potentiation of hippocampal synapses during the actual learning process in alert behaving animals. Control and recovery rats presented similar learning curves, whereas hypothyroid animals presented lower values. A single pulse presented to the perforant pathway during the conditioned-unconditioned inter-stimulus interval evoked a monosynaptic field excitatory postsynaptic potential in dentate granule cells (whose slope was linearly related to the rate of acquisition in the control group), but not in hypothyroid and recovery animals. Input-output relationships and long-term potentiation evoked by train stimulation of the perforant pathway were significantly depressed in hypothyroid animals. Thyroid hormone treatment failed to normalize these two neurophysiological abnormalities observed in hypothyroid animals. In contrast, paired-pulse facilitation was not affected by thyroidectomy. The results indicate that thyroid hormone treatment after a short period of adult hypothyroidism helps to restore some hippocampally dependent functions, such as classical conditioning, but not other hippocampal properties, such as the synaptic plasticity evoked during associative learning and during experimentally induced long-term potentiation. The present results have important clinical implications for the handling of patients with adult-onset thyroid diseases.
Collapse
Affiliation(s)
- Iván Fernández-Lamo
- Department of Nervous System and Endocrine Pathophysiology, Instituto de Investigaciones Biomédicas Alberto Sols, Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid, Madrid, Spain
| | | | | | | | | |
Collapse
|
50
|
Balu DT, Lucki I. Adult hippocampal neurogenesis: regulation, functional implications, and contribution to disease pathology. Neurosci Biobehav Rev 2009; 33:232-52. [PMID: 18786562 PMCID: PMC2671071 DOI: 10.1016/j.neubiorev.2008.08.007] [Citation(s) in RCA: 278] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2008] [Revised: 08/13/2008] [Accepted: 08/14/2008] [Indexed: 12/28/2022]
Abstract
It is now well established that the mammalian brain has the capacity to produce new neurons into adulthood. One such region that provides the proper milieu to sustain progenitor cells and is permissive to neuronal fate determination is located in the dentate gyrus of the hippocampus. This review will discuss in detail the complex process of adult hippocampal neurogenesis, including proliferation, differentiation, survival, and incorporation into neuronal networks. The regulation of this phenomenon by a number of factors is described, including neurotransmitter systems, growth factors, paracrine signaling molecules, neuropeptides, transcription factors, endogenous psychotropic systems, sex hormones, stress, and others. This review also addresses the functional significance of adult born hippocampal granule cells with regard to hippocampal circuitry dynamics and behavior. Furthermore, the relevance of perturbations in adult hippocampal neurogenesis to the pathophysiology of various disease states, including depression, schizophrenia, epilepsy, and diabetes are examined. Finally, this review discusses the potential of using hippocampal neurogenesis as a therapeutic target for these disorders.
Collapse
Affiliation(s)
- Darrick T. Balu
- Department of Pharmacology, University of Pennsylvania, Philadelphia, PA, USA
| | - Irwin Lucki
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|