1
|
Mardones MD, Rostam KD, Nickerson MC, Gupta K. Canonical Wnt activator Chir99021 prevents epileptogenesis in the intrahippocampal kainate mouse model of temporal lobe epilepsy. Exp Neurol 2024; 376:114767. [PMID: 38522659 PMCID: PMC11058011 DOI: 10.1016/j.expneurol.2024.114767] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 02/29/2024] [Accepted: 03/21/2024] [Indexed: 03/26/2024]
Abstract
The Wnt signaling pathway mediates the development of dentate granule cell neurons in the hippocampus. These neurons are central to the development of temporal lobe epilepsy and undergo structural and physiological remodeling during epileptogenesis, which results in the formation of epileptic circuits. The pathways responsible for granule cell remodeling during epileptogenesis have yet to be well defined, and represent therapeutic targets for the prevention of epilepsy. The current study explores Wnt signaling during epileptogenesis and for the first time describes the effect of Wnt activation using Wnt activator Chir99021 as a novel anti-epileptogenic therapeutic approach. Focal mesial temporal lobe epilepsy was induced by intrahippocampal kainate (IHK) injection in wild-type and POMC-eGFP transgenic mice. Wnt activator Chir99021 was administered daily, beginning 3 h after seizure induction, and continued up to 21-days. Immature granule cell morphology was quantified in the ipsilateral epileptogenic zone and the contralateral peri-ictal zone 14 days after IHK, targeting the end of the latent period. Bilateral hippocampal electrocorticographic recordings were performed for 28-days, 7-days beyond treatment cessation. Hippocampal behavioral tests were performed after completion of Chir99021 treatment. Consistent with previous studies, IHK resulted in the development of epilepsy after a 14 day latent period in this well-described mouse model. Activation of the canonical Wnt pathway with Chir99021 significantly reduced bilateral hippocampal seizure number and duration. Critically, this effect was retained after treatment cessation, suggesting a durable antiepileptogenic change in epileptic circuitry. Morphological analyses demonstrated that Wnt activation prevented pathological remodeling of the primary dendrite in both the epileptogenic zone and peri-ictal zone, changes in which may serve as a biomarker of epileptogenesis and anti-epileptogenic treatment response in pre-clinical studies. These findings were associated with improved object location memory with Chir99021 treatment after IHK. This study provides novel evidence that canonical Wnt activation prevents epileptogenesis in the IHK mouse model of mesial temporal lobe epilepsy, preventing pathological remodeling of dentate granule cells. Wnt signaling may therefore play a key role in mesial temporal lobe epileptogenesis, and Wnt modulation may represent a novel therapeutic strategy in the prevention of epilepsy.
Collapse
Affiliation(s)
- Muriel D Mardones
- Indiana University, Stark Neurosciences Research Institute, W 15th St, Indianapolis, IN 46202, United States of America; Indiana University, Department of Neurosurgery, W 16th St, Indianapolis, IN 46202, United States of America.
| | - Kevin D Rostam
- Indiana University, Stark Neurosciences Research Institute, W 15th St, Indianapolis, IN 46202, United States of America.
| | - Margaret C Nickerson
- Indiana University, Stark Neurosciences Research Institute, W 15th St, Indianapolis, IN 46202, United States of America.
| | - Kunal Gupta
- Medical College of Wisconsin, Department of Neurosurgery, 8701 Watertown Plank Rd, Milwaukee, WI 53226, United States of America; Medical College of Wisconsin, Neuroscience Research Center, 8701 Watertown Plank Rd, Milwaukee, WI 53226, United States of America; Indiana University, Stark Neurosciences Research Institute, W 15th St, Indianapolis, IN 46202, United States of America; Indiana University, Department of Neurosurgery, W 16th St, Indianapolis, IN 46202, United States of America.
| |
Collapse
|
2
|
Chang WL, Hen R. Adult Neurogenesis, Context Encoding, and Pattern Separation: A Pathway for Treating Overgeneralization. ADVANCES IN NEUROBIOLOGY 2024; 38:163-193. [PMID: 39008016 DOI: 10.1007/978-3-031-62983-9_10] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/16/2024]
Abstract
In mammals, the subgranular zone of the dentate gyrus is one of two brain regions (with the subventricular zone of the olfactory bulb) that continues to generate new neurons throughout adulthood, a phenomenon known as adult hippocampal neurogenesis (AHN) (Eriksson et al., Nat Med 4:1313-1317, 1998; García-Verdugo et al., J Neurobiol 36:234-248, 1998). The integration of these new neurons into the dentate gyrus (DG) has implications for memory encoding, with unique firing and wiring properties of immature neurons that affect how the hippocampal network encodes and stores attributes of memory. In this chapter, we will describe the process of AHN and properties of adult-born cells as they integrate into the hippocampal circuit and mature. Then, we will discuss some methodological considerations before we review evidence for the role of AHN in two major processes supporting memory that are performed by the DG. First, we will discuss encoding of contextual information for episodic memories and how this is facilitated by AHN. Second, will discuss pattern separation, a major role of the DG that reduces interference for the formation of new memories. Finally, we will review clinical and translational considerations, suggesting that stimulation of AHN may help decrease overgeneralization-a common endophenotype of mood, anxiety, trauma-related, and age-related disorders.
Collapse
Affiliation(s)
- Wei-Li Chang
- Departments of Psychiatry and Neuroscience, Columbia University, New York, NY, USA
- Division of Systems Neuroscience, New York State Psychiatric Institute, New York, NY, USA
| | - Rene Hen
- Departments of Psychiatry and Neuroscience, Columbia University, New York, NY, USA.
- Division of Systems Neuroscience, New York State Psychiatric Institute, New York, NY, USA.
| |
Collapse
|
3
|
Jiang YK, Dong FY, Dong YB, Zhu XY, Pan LH, Hu LB, Xu L, Xu XF, Xu LM, Zhang XQ. Lateral septal nucleus, dorsal part, and dentate gyrus are necessary for spatial and object recognition memory, respectively, in mice. Front Behav Neurosci 2023; 17:1139737. [PMID: 37064302 PMCID: PMC10102498 DOI: 10.3389/fnbeh.2023.1139737] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2023] [Accepted: 03/14/2023] [Indexed: 04/03/2023] Open
Abstract
IntroductionCognitive impairment includes the abnormality of learning, memory and judgment, resulting in severe learning and memory impairment and social activity impairment, which greatly affects the life quality of individuals. However, the specific mechanisms underlying cognitive impairment in different behavioral paradigms remain to be elucidated.MethodsThe study utilized two behavioral paradigms, novel location recognition (NLR) and novel object recognition (NOR), to investigate the brain regions involved in cognitive function. These tests comprised two phases: mice were presented with two identical objects for familiarization during the training phase, and a novel (experiment) or familiar (control) object/location was presented during testing. Immunostaining quantification of c-Fos, an immediate early gene used as a neuronal activity marker, was performed in eight different brain regions after the NLR or NOR test.ResultsThe number of c-Fos-positive cells was significantly higher in the dorsal part of the lateral septal nucleus (LSD) in the NLR and dentate gyrus (DG) in the NOR experiment group than in the control group. We further bilaterally lesioned these regions using excitotoxic ibotenic acid and replenished the damaged areas using an antisense oligonucleotide (ASO) strategy.DiscussionThese data reinforced the importance of LSD and DG in regulating spatial and object recognition memory, respectively. Thus, the study provides insight into the roles of these brain regions and suggests potential intervention targets for impaired spatial and object recognition memory.
Collapse
Affiliation(s)
- Ying-Ke Jiang
- Department of Pharmacology, Health Science Center, Ningbo University, Ningbo, Zhejiang, China
| | - Fei-Yuan Dong
- Department of Pharmacology, Health Science Center, Ningbo University, Ningbo, Zhejiang, China
| | - Yi-Bei Dong
- Department of Pharmacology, Health Science Center, Ningbo University, Ningbo, Zhejiang, China
| | - Xin-Yi Zhu
- Department of Pharmacology, Health Science Center, Ningbo University, Ningbo, Zhejiang, China
| | - Lu-Hui Pan
- Department of Pharmacology, Health Science Center, Ningbo University, Ningbo, Zhejiang, China
| | - Lin-Bo Hu
- Department of Pharmacology, Health Science Center, Ningbo University, Ningbo, Zhejiang, China
| | - Le Xu
- Department of Pharmacology, Health Science Center, Ningbo University, Ningbo, Zhejiang, China
| | - Xiao-Fan Xu
- Department of Pharmacology, Health Science Center, Ningbo University, Ningbo, Zhejiang, China
| | - Li-Min Xu
- Ningbo Women and Children’s Hospital, Ningbo, Zhejiang, China
- Li-Min Xu,
| | - Xiao-Qin Zhang
- Department of Pharmacology, Health Science Center, Ningbo University, Ningbo, Zhejiang, China
- *Correspondence: Xiao-Qin Zhang,
| |
Collapse
|
4
|
Gonzalez JC, Lee H, Vincent AM, Hill AL, Goode LK, King GD, Gamble KL, Wadiche JI, Overstreet-Wadiche L. Circadian regulation of dentate gyrus excitability mediated by G-protein signaling. Cell Rep 2023; 42:112039. [PMID: 36749664 PMCID: PMC10404305 DOI: 10.1016/j.celrep.2023.112039] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Revised: 10/27/2022] [Accepted: 01/12/2023] [Indexed: 02/08/2023] Open
Abstract
The central circadian regulator within the suprachiasmatic nucleus transmits time of day information by a diurnal spiking rhythm driven by molecular clock genes controlling membrane excitability. Most brain regions, including the hippocampus, harbor similar intrinsic circadian transcriptional machinery, but whether these molecular programs generate oscillations of membrane properties is unclear. Here, we show that intrinsic excitability of mouse dentate granule neurons exhibits a 24-h oscillation that controls spiking probability. Diurnal changes in excitability are mediated by antiphase G-protein regulation of potassium and sodium currents that reduce excitability during the Light phase. Disruption of the circadian transcriptional machinery by conditional deletion of Bmal1 enhances excitability selectively during the Light phase by removing G-protein regulation. These results reveal that circadian transcriptional machinery regulates intrinsic excitability by coordinated regulation of ion channels by G-protein signaling, highlighting a potential novel mechanism of cell-autonomous oscillations.
Collapse
Affiliation(s)
- Jose Carlos Gonzalez
- Department of Neurobiology and McKnight Brain Institute, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Haeun Lee
- Department of Neurobiology and McKnight Brain Institute, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Angela M Vincent
- Department of Neurobiology and McKnight Brain Institute, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Angela L Hill
- Department of Neurobiology and McKnight Brain Institute, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Lacy K Goode
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Gwendalyn D King
- Department of Biology, Creighton University, Omaha, NE 68178, USA
| | - Karen L Gamble
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Jacques I Wadiche
- Department of Neurobiology and McKnight Brain Institute, University of Alabama at Birmingham, Birmingham, AL 35294, USA.
| | - Linda Overstreet-Wadiche
- Department of Neurobiology and McKnight Brain Institute, University of Alabama at Birmingham, Birmingham, AL 35294, USA.
| |
Collapse
|
5
|
Pinson A, Sevrin E, Chatzi C, Le Gac B, Thiry M, Westbrook GL, Parent AS. Induction of Oxidative Stress and Alteration of Synaptic Gene Expression in Newborn Hippocampal Granule Cells after Developmental Exposure to Aroclor 1254. Neuroendocrinology 2022; 113:1248-1261. [PMID: 36257292 PMCID: PMC10110769 DOI: 10.1159/000527576] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Accepted: 10/10/2022] [Indexed: 11/19/2022]
Abstract
INTRODUCTION Hippocampal newborn neurons integrate into functional circuits where they play an important role in learning and memory. We previously showed that perinatal exposure to Aroclor 1254, a commercial mixture of polychlorinated biphenyls (PCBs) associated with alterations of cognitive function in children, disrupted the normal maturation of excitatory synapses in the dentate gyrus. We hypothesized that hippocampal immature neurons underlie some of the cognitive effects of PCBs. METHODS We used newly generated neurons to examine the effects of PCBs in mice following maternal exposure. Newborn dentate granule cells were tagged with enhanced green fluorescent protein using a transgenic mouse line. The transcriptome of the newly generated granule cells was assessed using RNA sequencing. RESULTS Gestational and lactational exposure to 6 mg/kg/day of Aroclor 1254 disrupted the mRNA expression of 1,308 genes in newborn granule cells. Genes involved in mitochondrial functions were highly enriched with 154 genes significantly increased in exposed compared to control mice. The upregulation of genes involved in oxidative phosphorylation was accompanied by signs of endoplasmic reticulum stress and an increase in lipid peroxidation, a marker of oxidative stress, in the subgranular zone of the dentate gyrus but not in mature granule cells in the granular zone. Aroclor 1254 exposure also disrupted the expression of synaptic genes. Using laser-captured subgranular and granular zones, this effect was restricted to the subgranular zone, where newborn neurons are located. CONCLUSION Our data suggest that gene expression in newborn granule cells is disrupted by Aroclor 1254 and provide clues to the effects of endocrine-disrupting chemicals on the brain.
Collapse
Affiliation(s)
- Anneline Pinson
- Neuroendocrinology Unit, GIGA-Neurosciences, University of Liège, Liège, Belgium
| | - Elena Sevrin
- Neuroendocrinology Unit, GIGA-Neurosciences, University of Liège, Liège, Belgium
| | - Christina Chatzi
- Vollum Institute, Oregon Health and Science University, Portland, OR, USA
| | - Benjamin Le Gac
- Neuroendocrinology Unit, GIGA-Neurosciences, University of Liège, Liège, Belgium
| | - Marc Thiry
- Cellular and tissular biology, GIGA-Neurosciences, University of Liège, Liège, Belgium
| | - Gary L Westbrook
- Vollum Institute, Oregon Health and Science University, Portland, OR, USA
| | - Anne-Simone Parent
- Neuroendocrinology Unit, GIGA-Neurosciences, University of Liège, Liège, Belgium
| |
Collapse
|
6
|
Osorno T, Rudolph S, Nguyen T, Kozareva V, Nadaf NM, Norton A, Macosko EZ, Lee WCA, Regehr WG. Candelabrum cells are ubiquitous cerebellar cortex interneurons with specialized circuit properties. Nat Neurosci 2022; 25:702-713. [PMID: 35578131 PMCID: PMC9548381 DOI: 10.1038/s41593-022-01057-x] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Accepted: 03/21/2022] [Indexed: 01/22/2023]
Abstract
To understand how the cerebellar cortex transforms mossy fiber (MF) inputs into Purkinje cell (PC) outputs, it is vital to delineate the elements of this circuit. Candelabrum cells (CCs) are enigmatic interneurons of the cerebellar cortex that have been identified based on their morphology, but their electrophysiological properties, synaptic connections and function remain unknown. Here, we clarify these properties using electrophysiology, single-nucleus RNA sequencing, in situ hybridization and serial electron microscopy in mice. We find that CCs are the most abundant PC layer interneuron. They are GABAergic, molecularly distinct and present in all cerebellar lobules. Their high resistance renders CC firing highly sensitive to synaptic inputs. CCs are excited by MFs and granule cells and are strongly inhibited by PCs. CCs in turn primarily inhibit molecular layer interneurons, which leads to PC disinhibition. Thus, inputs, outputs and local signals converge onto CCs to allow them to assume a unique role in controlling cerebellar output.
Collapse
Affiliation(s)
- Tomas Osorno
- Department of Neurobiology, Harvard Medical School, Boston, MA, USA
| | - Stephanie Rudolph
- Department of Neurobiology, Harvard Medical School, Boston, MA, USA
- Albert Einstein College of Medicine, New York, NY, USA
| | - Tri Nguyen
- Department of Neurobiology, Harvard Medical School, Boston, MA, USA
| | - Velina Kozareva
- Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Naeem M Nadaf
- Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Aliya Norton
- Department of Neurobiology, Harvard Medical School, Boston, MA, USA
| | - Evan Z Macosko
- Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Wei-Chung Allen Lee
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Wade G Regehr
- Department of Neurobiology, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
7
|
Edem EE, Ihaza BE, Fafure AA, Ishola AO, Nebo KE, Enye LA, Akinluyi ET. Virgin coconut oil abrogates depression-associated cognitive deficits by modulating hippocampal antioxidant balance, GABAergic and glutamatergic receptors in mice. Drug Metab Pers Ther 2021; 37:177-190. [PMID: 34881837 DOI: 10.1515/dmpt-2021-0126] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Accepted: 08/31/2021] [Indexed: 12/14/2022]
Abstract
OBJECTIVES GABA and glutamate neurotransmission play critical roles in both the neurobiology of depression and cognition; and Virgin coconut oil (VCO) is reported to support brain health. The present study investigated the effect of VCO on depression-associated cognitive deficits in mice. METHODS Thirty male mice divided into five groups were either exposed to chronic unpredicted mild stress (CUMS) protocol for 28 days or pre-treated with 3 mL/kg b. wt. of VCO for 21 days or post-treated with 3 mL/kg b. wt. of VCO for 21 days following 28 days of CUMS exposure. Mice were subjected to behavioural assessments for depressive-like behaviours and short-term memory, and thereafter euthanised. Hippocampal tissue was dissected from the harvested whole brain for biochemical and immunohistochemical evaluations. RESULTS Our results showed that CUMS exposure produced depressive-like behaviours, cognitive deficits and altered hippocampal redox balance. However, treatment with VCO abrogated depression-associated cognitive impairment, and enhanced hippocampal antioxidant concentration. Furthermore, immunohistochemical evaluation revealed significant improvement in GABAA and mGluR1a immunoreactivity following treatment with VCO in the depressed mice. CONCLUSIONS Therefore, findings from this study support the dietary application of VCO to enhance neural resilience in patients with depression and related disorders.
Collapse
Affiliation(s)
- Edem Ekpenyong Edem
- Department of Anatomy, College of Medicine and Health Sciences, Afe Babalola University, Ado-Ekiti, Ekiti State, Nigeria
| | - Blessing Eghosa Ihaza
- Department of Anatomy, College of Medicine and Health Sciences, Afe Babalola University, Ado-Ekiti, Ekiti State, Nigeria
| | - Adedamola Adediran Fafure
- Department of Anatomy, College of Medicine and Health Sciences, Afe Babalola University, Ado-Ekiti, Ekiti State, Nigeria
| | - Azeez Olakunle Ishola
- Department of Anatomy, College of Medicine and Health Sciences, Afe Babalola University, Ado-Ekiti, Ekiti State, Nigeria
| | - Kate Eberechukwu Nebo
- Department of Anatomy, College of Medicine and Health Sciences, Afe Babalola University, Ado-Ekiti, Ekiti State, Nigeria
| | - Linus Anderson Enye
- Department of Anatomy, College of Medicine and Health Sciences, Afe Babalola University, Ado-Ekiti, Ekiti State, Nigeria
| | - Elizabeth Toyin Akinluyi
- Department of Pharmacology and Therapeutics, College of Medicine and Health Sciences, Afe Babalola University, Ado-Ekiti, Ekiti State, Nigeria
| |
Collapse
|
8
|
Forte N, Boccella S, Tunisi L, Fernández-Rilo AC, Imperatore R, Iannotti FA, De Risi M, Iannotta M, Piscitelli F, Capasso R, De Girolamo P, De Leonibus E, Maione S, Di Marzo V, Cristino L. Orexin-A and endocannabinoids are involved in obesity-associated alteration of hippocampal neurogenesis, plasticity, and episodic memory in mice. Nat Commun 2021; 12:6137. [PMID: 34675233 PMCID: PMC8531398 DOI: 10.1038/s41467-021-26388-4] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2020] [Accepted: 09/30/2021] [Indexed: 11/20/2022] Open
Abstract
The mammalian brain stores and distinguishes among episodic memories, i.e. memories formed during the personal experience, through a mechanism of pattern separation computed in the hippocampal dentate gyrus. Decision-making for food-related behaviors, such as the choice and intake of food, might be affected in obese subjects by alterations in the retrieval of episodic memories. Adult neurogenesis in the dentate gyrus regulates the pattern separation. Several molecular factors affect adult neurogenesis and exert a critical role in the development and plasticity of newborn neurons. Orexin-A/hypocretin-1 and downstream endocannabinoid 2-arachidonoylglycerol signaling are altered in obese mice. Here, we show that excessive orexin-A/2-arachidonoylglycerol/cannabinoid receptor type-1 signaling leads to the dysfunction of adult hippocampal neurogenesis and the subsequent inhibition of plasticity and impairment of pattern separation. By inhibiting orexin-A action at orexin-1 receptors we rescued both plasticity and pattern separation impairment in obese mice, thus providing a molecular and functional mechanism to explain alterations in episodic memory in obesity. The authors show that adult hippocampal neurogenesis is altered in the dentate gyrus of obese mice with subsequent inhibition of long-term potentiation and impairment of pattern separation. Inhibition of orexin-A action at orexin-1 receptors rescued both impairments in obese mice.
Collapse
Affiliation(s)
- Nicola Forte
- Institute of Biomolecular Chemistry, Consiglio Nazionale delle Ricerche (CNR), Pozzuoli, NA, Italy
| | - Serena Boccella
- Department of Experimental Medicine, Division of Pharmacology, University of Campania Luigi Vanvitelli, Napoli, Italy
| | - Lea Tunisi
- Institute of Biomolecular Chemistry, Consiglio Nazionale delle Ricerche (CNR), Pozzuoli, NA, Italy
| | | | - Roberta Imperatore
- Department of Science and Technology, University of Sannio, Benevento, Italy
| | - Fabio Arturo Iannotti
- Institute of Biomolecular Chemistry, Consiglio Nazionale delle Ricerche (CNR), Pozzuoli, NA, Italy
| | - Maria De Risi
- Telethon Institute of Genetics and Medicine, Pozzuoli, Naples, Italy.,Institute of Biochemistry and Cell Biology, Consiglio Nazionale delle Ricerche (CNR), Monterotondo Scalo, Rome, Italy
| | - Monica Iannotta
- Department of Experimental Medicine, Division of Pharmacology, University of Campania Luigi Vanvitelli, Napoli, Italy
| | - Fabiana Piscitelli
- Institute of Biomolecular Chemistry, Consiglio Nazionale delle Ricerche (CNR), Pozzuoli, NA, Italy
| | - Raffaele Capasso
- Department of Agricultural Sciences, University of Naples Federico II, Portici, NA, Italy
| | - Paolo De Girolamo
- Department of Veterinary Medicine and Animal Productions, University Federico II, Napoli, Italy
| | - Elvira De Leonibus
- Telethon Institute of Genetics and Medicine, Pozzuoli, Naples, Italy.,Institute of Biochemistry and Cell Biology, Consiglio Nazionale delle Ricerche (CNR), Monterotondo Scalo, Rome, Italy
| | - Sabatino Maione
- Department of Experimental Medicine, Division of Pharmacology, University of Campania Luigi Vanvitelli, Napoli, Italy.,I.R.C.S.S., Neuromed, 86077, Pozzilli, Italy
| | - Vincenzo Di Marzo
- Institute of Biomolecular Chemistry, Consiglio Nazionale delle Ricerche (CNR), Pozzuoli, NA, Italy. .,Heart and Lung Research Institute of Université Laval, Québec City, QC, Canada. .,Institute for Nutrition and Functional Foods, Centre NUTRISS, Université Laval, Québec City, QC, Canada. .,Canada Excellence Research Chair on the Microbiome-Endocannabinoidome Axis in Metabolic Health, Université Laval, Québec City, QC, 61V0AG, Canada.
| | - Luigia Cristino
- Institute of Biomolecular Chemistry, Consiglio Nazionale delle Ricerche (CNR), Pozzuoli, NA, Italy.
| |
Collapse
|
9
|
Olson B, Zhu X, Norgard MA, Diba P, Levasseur PR, Buenafe AC, Huisman C, Burfeind KG, Michaelis KA, Kong G, Braun T, Marks DL. Chronic cerebral lipocalin 2 exposure elicits hippocampal neuronal dysfunction and cognitive impairment. Brain Behav Immun 2021; 97:102-118. [PMID: 34245812 PMCID: PMC8453133 DOI: 10.1016/j.bbi.2021.07.002] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2021] [Revised: 06/21/2021] [Accepted: 07/03/2021] [Indexed: 12/12/2022] Open
Abstract
Lipocalin 2 (LCN2) is a pleiotropic molecule that is induced in the central nervous system (CNS) in several acute and chronic pathologies. The acute induction of LCN2 evolved as a beneficial process, aimed at combating bacterial infection through the sequestration of iron from pathogens, while the role of LCN2 during chronic, non-infectious disease remains unclear, and recent studies suggest that LCN2 is neurotoxic. However, whether LCN2 is sufficient to induce behavioral and cognitive alterations remains unclear. In this paper, we sought to address the role of cerebral LCN2 on cognition in both acute and chronic settings. We demonstrate that LCN2 is robustly induced in the CNS during both acute and chronic inflammatory conditions, including LPS-based sepsis and cancer cachexia. In vivo, LPS challenge results in a global induction of LCN2 in the central nervous system, while cancer cachexia results in a distribution specific to the vasculature. Similar to these in vivo observations, in vitro modeling demonstrated that both glia and cerebral endothelium produce and secrete LCN2 when challenged with LPS, while only cerebral endothelium secrete LCN2 when challenged with cancer-conditioned medium. Chronic, but not short-term, cerebral LCN2 exposure resulted in reduced hippocampal neuron staining intensity, an increase in newborn neurons, microglial activation, and increased CNS immune cell infiltration, while gene set analyses suggested these effects were mediated through melanocortin-4 receptor independent mechanisms. RNA sequencing analyses of primary hippocampal neurons revealed a distinct transcriptome associated with prolonged LCN2 exposure, and ontology analysis was suggestive of altered neurite growth and abnormal spatial learning. Indeed, LCN2-treated hippocampal neurons display blunted neurite processes, and mice exposed to prolonged cerebral LCN2 levels experienced a reduction in spatial reference memory as indicated by Y-maze assessment. These findings implicate LCN2 as a pathologic mediator of cognitive decline in the setting of chronic disease.
Collapse
Affiliation(s)
- Brennan Olson
- Papé Family Pediatric Research Institute, Oregon Health & Science University, Portland, OR USA, Medical Scientist Training Program, Oregon Health & Science University, Portland, OR USA
| | - Xinxia Zhu
- Papé Family Pediatric Research Institute, Oregon Health & Science University, Portland, OR USA
| | - Mason A Norgard
- Papé Family Pediatric Research Institute, Oregon Health & Science University, Portland, OR USA
| | - Parham Diba
- Papé Family Pediatric Research Institute, Oregon Health & Science University, Portland, OR USA, Medical Scientist Training Program, Oregon Health & Science University, Portland, OR USA
| | - Peter R Levasseur
- Papé Family Pediatric Research Institute, Oregon Health & Science University, Portland, OR USA
| | - Abby C Buenafe
- Papé Family Pediatric Research Institute, Oregon Health & Science University, Portland, OR USA
| | - Christian Huisman
- Papé Family Pediatric Research Institute, Oregon Health & Science University, Portland, OR USA
| | - Kevin G Burfeind
- Papé Family Pediatric Research Institute, Oregon Health & Science University, Portland, OR USA, Medical Scientist Training Program, Oregon Health & Science University, Portland, OR USA
| | - Katherine A Michaelis
- Papé Family Pediatric Research Institute, Oregon Health & Science University, Portland, OR USA, Medical Scientist Training Program, Oregon Health & Science University, Portland, OR USA
| | - Garth Kong
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR USA
| | - Theodore Braun
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR USA
| | - Daniel L Marks
- Papé Family Pediatric Research Institute, Oregon Health & Science University, Portland, OR, USA; Brenden-Colson Center for Pancreatic Care, Oregon Health and & Science University Portland, OR, USA; Knight Cancer Institute, Oregon Health & Science University, Portland, OR, USA.
| |
Collapse
|
10
|
Domínguez-Rivas E, Ávila-Muñoz E, Schwarzacher SW, Zepeda A. Adult hippocampal neurogenesis in the context of lipopolysaccharide-induced neuroinflammation: A molecular, cellular and behavioral review. Brain Behav Immun 2021; 97:286-302. [PMID: 34174334 DOI: 10.1016/j.bbi.2021.06.014] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2020] [Revised: 06/17/2021] [Accepted: 06/19/2021] [Indexed: 12/17/2022] Open
Abstract
The continuous generation of new neurons occurs in at least two well-defined niches in the adult rodent brain. One of these areas is the subgranular zone of the dentate gyrus (DG) in the hippocampus. While the DG is associated with contextual and spatial learning and memory, hippocampal neurogenesis is necessary for pattern separation. Hippocampal neurogenesis begins with the activation of neural stem cells and culminates with the maturation and functional integration of a portion of the newly generated glutamatergic neurons into the hippocampal circuits. The neurogenic process is continuously modulated by intrinsic factors, one of which is neuroinflammation. The administration of lipopolysaccharide (LPS) has been widely used as a model of neuroinflammation and has yielded a body of evidence for unveiling the detrimental impact of inflammation upon the neurogenic process. This work aims to provide a comprehensive overview of the current knowledge on the effects of the systemic and central administration of LPS upon the different stages of neurogenesis and discuss their effects at the molecular, cellular, and behavioral levels.
Collapse
Affiliation(s)
- Eduardo Domínguez-Rivas
- Departamento de Medicina Genómica y Toxicología Ambiental, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Evangelina Ávila-Muñoz
- Departamento de Medicina Genómica y Toxicología Ambiental, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Stephan W Schwarzacher
- Institute of Clinical Neuroanatomy, Neuroscience Center, Goethe University Frankfurt, Frankfurt am Main, Germany
| | - Angélica Zepeda
- Departamento de Medicina Genómica y Toxicología Ambiental, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City, Mexico; Institute of Clinical Neuroanatomy, Neuroscience Center, Goethe University Frankfurt, Frankfurt am Main, Germany.
| |
Collapse
|
11
|
Martins-Macedo J, Salgado AJ, Gomes ED, Pinto L. Adult brain cytogenesis in the context of mood disorders: From neurogenesis to the emergent role of gliogenesis. Neurosci Biobehav Rev 2021; 131:411-428. [PMID: 34555383 DOI: 10.1016/j.neubiorev.2021.09.030] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Revised: 09/06/2021] [Accepted: 09/16/2021] [Indexed: 12/18/2022]
Abstract
Psychiatric disorders severely impact patients' lives. Motivational, cognitive and emotional deficits are the most common symptoms observed in these patients and no effective treatment is still available, either due to the adverse side effects or the low rate of efficacy of currently available drugs. Neurogenesis recovery has been one important focus in the treatment of psychiatric disorders, which undeniably contributes to the therapeutic action of antidepressants. However, glial plasticity is emerging as a new strategy to explore the deficits observed in mood disorders and the efficacy of therapeutic interventions. Thus, it is crucial to understand the mechanisms behind glio- and neurogenesis to better define treatments and preventive therapies, once adult cytogenesis is of pivotal importance to cognitive and emotional components of behavior, both in healthy and pathological contexts, including in psychiatric disorders. Here, we review the concepts and history of neuro- and gliogenesis, providing as well a reflection on the functional importance of cytogenesis in the context of disease.
Collapse
Affiliation(s)
- Joana Martins-Macedo
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus de Gualtar, 4710-057 Braga, Portugal; ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal.
| | - António J Salgado
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus de Gualtar, 4710-057 Braga, Portugal; ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal.
| | - Eduardo D Gomes
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus de Gualtar, 4710-057 Braga, Portugal; ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal.
| | - Luísa Pinto
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus de Gualtar, 4710-057 Braga, Portugal; ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal.
| |
Collapse
|
12
|
Role of NMDA Receptors in Adult Neurogenesis and Normal Development of the Dentate Gyrus. eNeuro 2021; 8:ENEURO.0566-20.2021. [PMID: 34266965 PMCID: PMC8354713 DOI: 10.1523/eneuro.0566-20.2021] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 05/28/2021] [Accepted: 06/19/2021] [Indexed: 01/21/2023] Open
Abstract
The NMDA receptors are a type of glutamate receptors, which is involved in neuronal function, plasticity and development in the mammalian brain. However, how the NMDA receptors contribute to adult neurogenesis and development of the dentate gyrus is unclear. In this study, we investigate this question by examining a region-specific knock-out mouse line that lacks the NR1 gene, which encodes the essential subunit of the NMDA receptors, in granule cells of the dentate gyrus (DG-NR1KO mice). We found that the survival of newly-generated granule cells, cell proliferation and the size of the granule cell layer are significantly reduced in the dorsal dentate gyrus of adult DG-NR1KO mice. Our results also show a significant reduction in the number of immature neurons and in the volume of the granule cell layer, starting from three weeks of postnatal age. DG-NR1KO mice also showed impairment in the expression of an immediate early gene, Arc, and behavior during the novelty-suppressed feeding and open field test. These results suggest that the NMDA receptors in granule cells have a role in adult neurogenesis in the adult brain and contributes to the normal development of the dentate gyrus.
Collapse
|
13
|
Pahle J, Muhia M, Wagener RJ, Tippmann A, Bock HH, Graw J, Herz J, Staiger JF, Drakew A, Kneussel M, Rune GM, Frotscher M, Brunne B. Selective Inactivation of Reelin in Inhibitory Interneurons Leads to Subtle Changes in the Dentate Gyrus But Leaves Cortical Layering and Behavior Unaffected. Cereb Cortex 2021; 30:1688-1707. [PMID: 31667489 PMCID: PMC7132935 DOI: 10.1093/cercor/bhz196] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
Reelin is an extracellular matrix protein, known for its dual role in neuronal migration during brain development and in synaptic plasticity at adult stages. During the perinatal phase, Reelin expression switches from Cajal-Retzius (CR) cells, its main source before birth, to inhibitory interneurons (IN), the main source of Reelin in the adult forebrain. IN-derived Reelin has been associated with schizophrenia and temporal lobe epilepsy; however, the functional role of Reelin from INs is presently unclear. In this study, we used conditional knockout mice, which lack Reelin expression specifically in inhibitory INs, leading to a substantial reduction in total Reelin expression in the neocortex and dentate gyrus. Our results show that IN-specific Reelin knockout mice exhibit normal neuronal layering and normal behavior, including spatial reference memory. Although INs are the major source of Reelin within the adult stem cell niche, Reelin from INs does not contribute substantially to normal adult neurogenesis. While a closer look at the dentate gyrus revealed some unexpected alterations at the cellular level, including an increase in the number of Reelin expressing CR cells, overall our data suggest that Reelin derived from INs is less critical for cortex development and function than Reelin expressed by CR cells.
Collapse
Affiliation(s)
- Jasmine Pahle
- Institute for Structural Neurobiology, Center for Molecular Neurobiology, ZMNH, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Mary Muhia
- Institute of Molecular Neurogenetics, Center for Molecular Neurobiology, ZMNH, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Robin J Wagener
- Neurology Clinic, University Hospital Heidelberg, Im Neuenheimer Feld 400, 69120 Heidelberg, Germany
| | - Anja Tippmann
- Institute for Structural Neurobiology, Center for Molecular Neurobiology, ZMNH, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany.,Department of Systems Neuroscience, Johann-Friedrich-Blumenbach Institute for Zoology and Anthropology, University of Göttingen, 37075 Göttingen, Germany
| | - Hans H Bock
- Clinic of Gastroenterology and Hepatology, Heinrich-Heine-University Düsseldorf, 40225 Düsseldorf, Germany
| | - Janice Graw
- Institute of Neuroanatomy, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Joachim Herz
- Department of Molecular Genetics, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Jochen F Staiger
- Institute for Neuroanatomy, University Medical Center Göttingen, Georg-August-University Göttingen, 37075 Göttingen, Germany
| | - Alexander Drakew
- Institute for Structural Neurobiology, Center for Molecular Neurobiology, ZMNH, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany.,Institute of Clinical Neuroanatomy, Faculty of Medicine, 60590 Frankfurt, Germany
| | - Matthias Kneussel
- Institute of Molecular Neurogenetics, Center for Molecular Neurobiology, ZMNH, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Gabriele M Rune
- Institute of Neuroanatomy, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Michael Frotscher
- Institute for Structural Neurobiology, Center for Molecular Neurobiology, ZMNH, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Bianka Brunne
- Institute for Structural Neurobiology, Center for Molecular Neurobiology, ZMNH, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany.,Institute of Neuroanatomy, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| |
Collapse
|
14
|
Chou SM, Li KX, Huang MY, Chen C, Lin King YH, Li GG, Zhou W, Teo CF, Jan YN, Jan LY, Yang SB. Kv1.1 channels regulate early postnatal neurogenesis in mouse hippocampus via the TrkB signaling pathway. eLife 2021; 10:e58779. [PMID: 34018923 PMCID: PMC8208815 DOI: 10.7554/elife.58779] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Accepted: 05/20/2021] [Indexed: 12/20/2022] Open
Abstract
In the postnatal brain, neurogenesis occurs only within a few regions, such as the hippocampal sub-granular zone (SGZ). Postnatal neurogenesis is tightly regulated by factors that balance stem cell renewal with differentiation, and it gives rise to neurons that participate in learning and memory formation. The Kv1.1 channel, a voltage-gated potassium channel, was previously shown to suppress postnatal neurogenesis in the SGZ in a cell-autonomous manner. In this study, we have clarified the physiological and molecular mechanisms underlying Kv1.1-dependent postnatal neurogenesis. First, we discovered that the membrane potential of neural progenitor cells is highly dynamic during development. We further established a multinomial logistic regression model for cell-type classification based on the biophysical characteristics and corresponding cell markers. We found that the loss of Kv1.1 channel activity causes significant depolarization of type 2b neural progenitor cells. This depolarization is associated with increased tropomyosin receptor kinase B (TrkB) signaling and proliferation of neural progenitor cells; suppressing TrkB signaling reduces the extent of postnatal neurogenesis. Thus, our study defines the role of the Kv1.1 potassium channel in regulating the proliferation of postnatal neural progenitor cells in mouse hippocampus.
Collapse
Affiliation(s)
- Shu-Min Chou
- Institute of Biomedical Sciences, Academia SinicaTaipeiTaiwan
| | - Ke-Xin Li
- Howard Hughes Medical Institute, Departments of Physiology, Biochemistry and Biophysics, University of California, San FranciscoSan FranciscoUnited States
| | | | - Chao Chen
- Howard Hughes Medical Institute, Departments of Physiology, Biochemistry and Biophysics, University of California, San FranciscoSan FranciscoUnited States
| | - Yuan-Hung Lin King
- Howard Hughes Medical Institute, Departments of Physiology, Biochemistry and Biophysics, University of California, San FranciscoSan FranciscoUnited States
- Neuroscience Graduate Program, University of California, San FranciscoSan FranciscoUnited States
| | | | - Wei Zhou
- Department of Anesthesia and Perioperative Care, University of California, San FranciscoSan FranciscoUnited States
| | - Chin Fen Teo
- Howard Hughes Medical Institute, Departments of Physiology, Biochemistry and Biophysics, University of California, San FranciscoSan FranciscoUnited States
| | - Yuh Nung Jan
- Howard Hughes Medical Institute, Departments of Physiology, Biochemistry and Biophysics, University of California, San FranciscoSan FranciscoUnited States
| | - Lily Yeh Jan
- Howard Hughes Medical Institute, Departments of Physiology, Biochemistry and Biophysics, University of California, San FranciscoSan FranciscoUnited States
| | - Shi-Bing Yang
- Institute of Biomedical Sciences, Academia SinicaTaipeiTaiwan
- Neuroscience Program of Academia Sinica, Academia SinicaTaipeiTaiwan
| |
Collapse
|
15
|
Pten is a key intrinsic factor regulating raphe 5-HT neuronal plasticity and depressive behaviors in mice. Transl Psychiatry 2021; 11:186. [PMID: 33771970 PMCID: PMC7998026 DOI: 10.1038/s41398-021-01303-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2020] [Revised: 02/20/2021] [Accepted: 03/03/2021] [Indexed: 12/14/2022] Open
Abstract
Serotonin (5-HT)-based antidepressants, selective serotonin reuptake inhibitors (SSRIs) aim to enhance serotonergic activity by blocking its reuptake. We propose PTEN as a target for an alternative approach for regulating 5-HT neuron activity in the brain and depressive behaviors. We show that PTEN is elevated in central 5-HT neurons in the raphe nucleus by chronic stress in mice, and selective deletion of Pten in the 5-HT neurons induces its structural plasticity shown by increases of dendritic branching and density of PSD95-positive puncta in the dendrites. 5-HT levels are elevated and electrical stimulation of raphe neurons evokes more 5-HT release in the brain of condition knockout (cKO) mice with Pten-deficient 5-HT neurons. In addition, the 5-HT neurons remain normal electrophysiological properties but have increased excitatory synaptic inputs. Single-cell RNA sequencing revealed gene transcript alterations that may underlay morphological and functional changes in Pten-deficient 5-HT neurons. Finally, Pten cKO mice and wild-type mice treated with systemic application of PTEN inhibitor display reduced depression-like behaviors. Thus, PTEN is an intrinsic regulator of 5-HT neuron activity, representing a novel therapeutic strategy for producing antidepressant action.
Collapse
|
16
|
Sanders M, Petrasch-Parwez E, Habbes HW, Düring MV, Förster E. Postnatal Developmental Expression Profile Classifies the Indusium Griseum as a Distinct Subfield of the Hippocampal Formation. Front Cell Dev Biol 2021; 8:615571. [PMID: 33511122 PMCID: PMC7835525 DOI: 10.3389/fcell.2020.615571] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Accepted: 12/07/2020] [Indexed: 11/13/2022] Open
Abstract
The indusium griseum (IG) is a cortical structure overlying the corpus callosum along its anterior–posterior extent. It has been classified either as a vestige of the hippocampus or as an extension of the dentate gyrus via the fasciola cinerea, but its attribution to a specific hippocampal subregion is still under debate. To specify the identity of IG neurons more precisely, we investigated the spatiotemporal expression of calbindin, secretagogin, Necab2, PCP4, and Prox1 in the postnatal mouse IG, fasciola cinerea, and hippocampus. We identified the calcium-binding protein Necab2 as a first reliable marker for the IG and fasciola cinerea throughout postnatal development into adulthood. In contrast, calbindin, secretagogin, and PCP4 were expressed each with a different individual time course during maturation, and at no time point, IG or fasciola cinerea principal neurons expressed Prox1, a transcription factor known to define dentate granule cell fate. Concordantly, in a transgenic mouse line expressing enhanced green fluorescent protein (eGFP) in dentate granule cells, neurons of IG and fasciola cinerea were eGFP-negative. Our findings preclude that IG neurons represent dentate granule cells, as earlier hypothesized, and strongly support the view that the IG is an own hippocampal subfield composed of a distinct neuronal population.
Collapse
Affiliation(s)
- Marie Sanders
- Department of Neuroanatomy and Molecular Brain Research, Ruhr-University Bochum, Bochum, Germany
| | | | - Hans-Werner Habbes
- Department of Neuroanatomy and Molecular Brain Research, Ruhr-University Bochum, Bochum, Germany
| | - Monika V Düring
- Department of Neuroanatomy and Molecular Brain Research, Ruhr-University Bochum, Bochum, Germany
| | - Eckart Förster
- Department of Neuroanatomy and Molecular Brain Research, Ruhr-University Bochum, Bochum, Germany
| |
Collapse
|
17
|
Zhang X, Mei Y, He Y, Wang D, Wang J, Wei X, Yang E, Zhou D, Shen H, Peng G, Shu Q, Li X, Luo B, Zhou Y, Sun B. Ablating Adult Neural Stem Cells Improves Synaptic and Cognitive Functions in Alzheimer Models. Stem Cell Reports 2020; 16:89-105. [PMID: 33382977 PMCID: PMC7897582 DOI: 10.1016/j.stemcr.2020.12.003] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Revised: 11/30/2020] [Accepted: 12/01/2020] [Indexed: 01/26/2023] Open
Abstract
Adult neurogenesis is impaired in the hippocampus of patients with Alzheimer disease (AD) as well as AD models. However, it is far from clear how modulating adult neurogenesis affects AD neuropathology. We confirm that adult hippocampal neurogenesis is impaired in two AD models. Surprisingly, however, cognitive functions are improved in AD models after ablating adult neural stem cells (aNSCs). Ablation of aNSCs does not affect the levels of amyloid β but restores the normal synaptic transmission in the dentate gyrus (DG) granule cells of AD models. Furthermore, calbindin depletion in the DG of AD mice is ameliorated after aNSC ablation, and knocking down calbindin abolishes the effects of aNSC ablation on synaptic and cognitive functions of AD mice. Together, our data suggest that cognitive functions of AD mice are improved after aNSC ablation, which is associated with the restoration of synaptic transmission in the DG granule cells with calbindin as an important mediator. Adult hippocampal neurogenesis was impaired in two AD models Cognitive functions were improved in AD models after ablation of aNSCs Ablating aNSCs restored the normal synaptic transmission in the DG granule cells of AD models Altered expression of calbindin mediated the effects of ablating aNSCs on synaptic and cognitive functions in AD mice
Collapse
Affiliation(s)
- Xiaoqin Zhang
- Department of Neurobiology and Department of Neurology of the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, 310058, China; Department of Physiology and Pharmacology, Medical School of Ningbo University, Ningbo, Zhejiang Province, 315211, China
| | - Yufei Mei
- Department of Neurobiology and Department of Neurology of the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, 310058, China; NHC and CAMS Key Laboratory of Medical Neurobiology, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, Zhejiang Province, 310058, China
| | - Yang He
- Department of Neurobiology and Department of Neurology of the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, 310058, China; NHC and CAMS Key Laboratory of Medical Neurobiology, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, Zhejiang Province, 310058, China
| | - Dongpi Wang
- Children's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, 310052, China
| | - Jing Wang
- Department of Neurobiology and Department of Neurology of the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, 310058, China; NHC and CAMS Key Laboratory of Medical Neurobiology, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, Zhejiang Province, 310058, China
| | - Xiaojie Wei
- Department of Neurobiology and Department of Neurology of the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, 310058, China; NHC and CAMS Key Laboratory of Medical Neurobiology, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, Zhejiang Province, 310058, China
| | - Enlu Yang
- Department of Neurobiology and Department of Neurology of the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, 310058, China; NHC and CAMS Key Laboratory of Medical Neurobiology, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, Zhejiang Province, 310058, China
| | - Dongming Zhou
- Children's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, 310052, China
| | - Haowei Shen
- Department of Physiology and Pharmacology, Medical School of Ningbo University, Ningbo, Zhejiang Province, 315211, China
| | - Guoping Peng
- Department of Neurobiology and Department of Neurology of the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, 310058, China
| | - Qiang Shu
- Children's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, 310052, China
| | - Xuekun Li
- Children's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, 310052, China; Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, 310029, China
| | - Benyan Luo
- Department of Neurobiology and Department of Neurology of the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, 310058, China
| | - Yudong Zhou
- Department of Neurobiology and Department of Neurology of the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, 310058, China; NHC and CAMS Key Laboratory of Medical Neurobiology, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, Zhejiang Province, 310058, China.
| | - Binggui Sun
- Department of Neurobiology and Department of Neurology of the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, 310058, China; NHC and CAMS Key Laboratory of Medical Neurobiology, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, Zhejiang Province, 310058, China.
| |
Collapse
|
18
|
Recruitment of parvalbumin and somatostatin interneuron inputs to adult born dentate granule neurons. Sci Rep 2020; 10:17522. [PMID: 33067500 PMCID: PMC7568561 DOI: 10.1038/s41598-020-74385-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Accepted: 09/25/2020] [Indexed: 12/28/2022] Open
Abstract
GABA is a key regulator of adult-born dentate granule cell (abDGC) maturation so mapping the functional connectivity between abDGCs and local interneurons is required to understand their development and integration into the hippocampal circuit. We recorded from birthdated abDGCs in mice and photoactivated parvalbumin (PV) and somatostatin (SST) interneurons to map the timing and strength of inputs to abDGCs during the first 4 weeks after differentiation. abDGCs received input from PV interneurons in the first week, but SST inputs were not detected until the second week. Analysis of desynchronized quantal events established that the number of GABAergic synapses onto abDGCs increased with maturation, whereas individual synaptic strength was constant. Voluntary wheel running in mice scaled the GABAergic input to abDGCs by increasing the number of synaptic contacts from both interneuron types. This demonstrates that GABAergic innervation to abDGCs develops during a prolonged post-mitotic period and running scales both SST and PV synaptic afferents.
Collapse
|
19
|
Nam MH, Han KS, Lee J, Won W, Koh W, Bae JY, Woo J, Kim J, Kwong E, Choi TY, Chun H, Lee SE, Kim SB, Park KD, Choi SY, Bae YC, Lee CJ. Activation of Astrocytic μ-Opioid Receptor Causes Conditioned Place Preference. Cell Rep 2020; 28:1154-1166.e5. [PMID: 31365861 DOI: 10.1016/j.celrep.2019.06.071] [Citation(s) in RCA: 67] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2019] [Revised: 05/22/2019] [Accepted: 06/20/2019] [Indexed: 11/19/2022] Open
Abstract
The underlying mechanisms of how positive emotional valence (e.g., pleasure) causes preference of an associated context is poorly understood. Here, we show that activation of astrocytic μ-opioid receptor (MOR) drives conditioned place preference (CPP) by means of specific modulation of astrocytic MOR, an exemplar endogenous Gi protein-coupled receptor (Gi-GPCR), in the CA1 hippocampus. Long-term potentiation (LTP) induced by a subthreshold stimulation with the activation of astrocytic MOR at the Schaffer collateral pathway accounts for the memory acquisition to induce CPP. This astrocytic MOR-mediated LTP induction is dependent on astrocytic glutamate released upon activation of the astrocytic MOR and the consequent activation of the presynaptic mGluR1. The astrocytic MOR-dependent LTP and CPP were recapitulated by a chemogenetic activation of astrocyte-specifically expressed Gi-DREADD hM4Di. Our study reveals that the transduction of inhibitory Gi-signaling into augmented excitatory synaptic transmission through astrocytic glutamate is critical for the acquisition of contextual memory for CPP.
Collapse
MESH Headings
- Animals
- Astrocytes/metabolism
- CA1 Region, Hippocampal/metabolism
- GTP-Binding Protein alpha Subunits, Gi-Go/genetics
- GTP-Binding Protein alpha Subunits, Gi-Go/metabolism
- Memory
- Mice
- Mice, Knockout
- Receptors, Metabotropic Glutamate/genetics
- Receptors, Metabotropic Glutamate/metabolism
- Receptors, Opioid, mu/genetics
- Receptors, Opioid, mu/metabolism
Collapse
Affiliation(s)
- Min-Ho Nam
- Center for Neuroscience, Korea Institute of Science and Technology (KIST), Seoul 02792, Korea; Department of Science in Korean Medicine, Graduate School, Kyung Hee University, Seoul 02447, Korea
| | - Kyung-Seok Han
- Center for Neuroscience, Korea Institute of Science and Technology (KIST), Seoul 02792, Korea; Department of Neuroscience, Division of Bio-Medical Science & Technology, KIST School, Korea University of Science and Technology, Seoul 02792, Korea
| | - Jaekwang Lee
- Center for Neuroscience, Korea Institute of Science and Technology (KIST), Seoul 02792, Korea
| | - Woojin Won
- Center for Neuroscience, Korea Institute of Science and Technology (KIST), Seoul 02792, Korea; KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul 02841, Korea; Center for Cognition and Sociality, Institute for Basic Science, Daejeon 34126, Korea
| | - Wuhyun Koh
- Center for Neuroscience, Korea Institute of Science and Technology (KIST), Seoul 02792, Korea; Department of Neuroscience, Division of Bio-Medical Science & Technology, KIST School, Korea University of Science and Technology, Seoul 02792, Korea; Center for Cognition and Sociality, Institute for Basic Science, Daejeon 34126, Korea
| | - Jin Young Bae
- Department of Anatomy and Neurobiology, School of Dentistry, Kyungpook National University, Daegu 41940, Korea
| | - Junsung Woo
- Center for Neuroscience, Korea Institute of Science and Technology (KIST), Seoul 02792, Korea; Department of Neuroscience, Division of Bio-Medical Science & Technology, KIST School, Korea University of Science and Technology, Seoul 02792, Korea
| | - Jayoung Kim
- Center for Neuroscience, Korea Institute of Science and Technology (KIST), Seoul 02792, Korea
| | - Elliot Kwong
- Center for Neuroscience, Korea Institute of Science and Technology (KIST), Seoul 02792, Korea
| | - Tae-Yong Choi
- Department of Physiology and Dental Research Institute, Seoul National University School of Dentistry, Seoul 03080, Korea
| | - Heejung Chun
- Center for Neuroscience, Korea Institute of Science and Technology (KIST), Seoul 02792, Korea; Center for Cognition and Sociality, Institute for Basic Science, Daejeon 34126, Korea
| | - Seung Eun Lee
- Virus Facility, Research Animal Resource Center, KIST, Seoul 02792, Korea
| | - Sang-Bum Kim
- New Drug Development Center, Daegu-Gyeongbuk Medical Innovation Foundation, Daegu 41061, Korea
| | - Ki Duk Park
- Department of Neuroscience, Division of Bio-Medical Science & Technology, KIST School, Korea University of Science and Technology, Seoul 02792, Korea; KHU-KIST Department of Converging Science and Technology, Kyung Hee University, Seoul 02447, Korea; Convergence Research Center for Diagnosis, Treatment and Care System of Dementia, KIST, Seoul 02792, Korea
| | - Se-Young Choi
- Department of Physiology and Dental Research Institute, Seoul National University School of Dentistry, Seoul 03080, Korea
| | - Yong Chul Bae
- Department of Anatomy and Neurobiology, School of Dentistry, Kyungpook National University, Daegu 41940, Korea.
| | - C Justin Lee
- Center for Neuroscience, Korea Institute of Science and Technology (KIST), Seoul 02792, Korea; Department of Neuroscience, Division of Bio-Medical Science & Technology, KIST School, Korea University of Science and Technology, Seoul 02792, Korea; Center for Cognition and Sociality, Institute for Basic Science, Daejeon 34126, Korea.
| |
Collapse
|
20
|
Chai X, Zhang W, Li L, Wu Y, Zhu X, Zhao S. Profile of MIF in Developing Hippocampus: Association With Cell Proliferation and Neurite Outgrowth. Front Mol Neurosci 2020; 13:147. [PMID: 32903462 PMCID: PMC7434973 DOI: 10.3389/fnmol.2020.00147] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Accepted: 07/15/2020] [Indexed: 11/13/2022] Open
Abstract
Proinflammatory cytokine macrophage migration inhibitory factor (MIF) is a multifunctional cytokine and has been found involved in many neurological diseases such as Alzheimer disease (AD), epilepsy, and multiple sclerosis. Previous studies have shown that MIF is expressed in neocortex, hippocampus, hypothalamus, cerebellum, and spinal cord in adult mice. It is expressed by astrocytes and activates microglias in neuroinflammation. Further studies have shown that MIF is detected in moss fibers of dentate granule cells and in apical dendrites of pyramidal neurons in adult hippocampus. Only NeuroD-positive immature granule neurons but not NeuN-positive mature neurons express MIF. These findings led us eager to know the exact role of MIF in the development of hippocampus. Therefore, we systematically checked the spatial and temporal expression pattern of MIF and characterized MIF-positive cells in hippocampus from mice aged from postnatal day 0 (P0) to 3 months. Our results showed that the lowest level of MIF protein occurred at P7 and mif mRNA increased from P0, reached a peak at P7, and stably expressed until P30 before declining dramatically at 3 months. MIF was localized in fibers of GFAP- and BLBP-positive radial glial precursor cells in dentate gyrus (DG). DCX-expressing newly generated neurons were MIF-negative. Inhibition of MIF by MIF antagonist S, R-3-(4-hydroxyphenyl)-4, 5-dihydro-5-isoxazole acetic acid methyl ester (ISO-1) reduced BrdU-positive cells. Interestingly, MIF was expressed by NeuN-positive GABAergic interneurons including parvalbumin-and Reelin-expressing cells in the DG. Neither NeuN-positive granule cells nor NeuN-positive pyramidal neurons expressed MIF. In transgenic mice, POMC-EGFP–positive immature dentate granule cells and Thy1-EGFP–positive mature granule cells were MIF-negative. Treatment of neuronal cultures with ISO-1 inhibited neurite outgrowth. Therefore, we conclude that MIF might be important for feature maintenance of neural stem cells and neurite outgrowth during hippocampal development.
Collapse
Affiliation(s)
- Xuejun Chai
- College of Basic Medicine, Xi'an Medical University, Xi'an, China
| | - Wei Zhang
- College of Veterinary Medicine, Northwest A&F University, Yangling, China
| | - Lingling Li
- College of Veterinary Medicine, Northwest A&F University, Yangling, China
| | - Yongji Wu
- College of Veterinary Medicine, Northwest A&F University, Yangling, China
| | - Xiaoyan Zhu
- College of Veterinary Medicine, Northwest A&F University, Yangling, China
| | - Shanting Zhao
- College of Veterinary Medicine, Northwest A&F University, Yangling, China
| |
Collapse
|
21
|
Ethanol has concentration-dependent effects on hypothalamic POMC neuronal excitability. Alcohol 2020; 86:103-112. [PMID: 32304714 DOI: 10.1016/j.alcohol.2020.04.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Revised: 03/31/2020] [Accepted: 04/01/2020] [Indexed: 11/21/2022]
Abstract
Alcohol abuse is a worldwide public health concern, yet the precise molecular targets of alcohol in the brain are still not fully understood. Alcohol may promote its euphoric and motivational effects, in part, by activating the endogenous opioid system. One particular component of this system consists of pro-opiomelanocortin (POMC) -producing neurons in the arcuate nucleus (ArcN) of the hypothalamus, which project to reward-related brain areas. To identify the physiological effects of ethanol on ArcN POMC neurons, we utilized whole cell patch-clamp recordings and bath application of ethanol (5-40 mM) to identify alterations in spontaneous baseline activity, rheobase, spiking characteristics, or intrinsic neuronal properties. We found that 10 mM ethanol increased the number of depolarization-induced spikes in the majority of recorded cells, whereas higher concentrations of ethanol (20-40 mM) decreased the number of spikes. Interestingly, we found that basal firing rates of ArcN POMC neurons may predict physiological responding to ethanol. Rheobase and spontaneous activity, measured by spontaneous excitatory post-synaptic potentials (EPSPs) at rest, were unchanged after exposure to ethanol, regardless of concentration. These results suggest that ethanol has concentration-dependent modulatory effects on ArcN POMC neuronal activity, which may be relevant to treatments for alcohol use disorders that target endogenous opioid systems.
Collapse
|
22
|
Vaden RJ, Gonzalez JC, Tsai MC, Niver AJ, Fusilier AR, Griffith CM, Kramer RH, Wadiche JI, Overstreet-Wadiche L. Parvalbumin interneurons provide spillover to newborn and mature dentate granule cells. eLife 2020; 9:54125. [PMID: 32602839 PMCID: PMC7326496 DOI: 10.7554/elife.54125] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Accepted: 06/19/2020] [Indexed: 01/09/2023] Open
Abstract
Parvalbumin-expressing interneurons (PVs) in the dentate gyrus provide activity-dependent regulation of adult neurogenesis as well as maintain inhibitory control of mature neurons. In mature neurons, PVs evoke GABAA postsynaptic currents (GPSCs) with fast rise and decay phases that allow precise control of spike timing, yet synaptic currents with fast kinetics do not appear in adult-born neurons until several weeks after cell birth. Here we used mouse hippocampal slices to address how PVs signal to newborn neurons prior to the appearance of fast GPSCs. Whereas PV-evoked currents in mature neurons exhibit hallmark fast rise and decay phases, newborn neurons display slow GPSCs with characteristics of spillover signaling. We also unmasked slow spillover currents in mature neurons in the absence of fast GPSCs. Our results suggest that PVs mediate slow spillover signaling in addition to conventional fast synaptic signaling, and that spillover transmission mediates activity-dependent regulation of early events in adult neurogenesis.
Collapse
Affiliation(s)
- Ryan J Vaden
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, United States
| | - Jose Carlos Gonzalez
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, United States
| | - Ming-Chi Tsai
- Department of Molecular and Cellular Biology, University of California Berkeley, Berkeley, United States
| | - Anastasia J Niver
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, United States
| | - Allison R Fusilier
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, United States
| | - Chelsea M Griffith
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, United States
| | - Richard H Kramer
- Department of Molecular and Cellular Biology, University of California Berkeley, Berkeley, United States
| | - Jacques I Wadiche
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, United States
| | | |
Collapse
|
23
|
Neuronal network remodeling and Wnt pathway dysregulation in the intra-hippocampal kainate mouse model of temporal lobe epilepsy. PLoS One 2019; 14:e0215789. [PMID: 31596871 PMCID: PMC6785072 DOI: 10.1371/journal.pone.0215789] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2019] [Accepted: 09/20/2019] [Indexed: 01/19/2023] Open
Abstract
Mouse models of mesial temporal lobe epilepsy recapitulate aspects of human epilepsy, which is characterized by neuronal network remodeling in the hippocampal dentate gyrus. Observational studies suggest that this remodeling is associated with altered Wnt pathway signaling, although this has not been experimentally examined. We used the well-characterized mouse intrahippocampal kainate model of temporal lobe epilepsy to examine associations between hippocampal neurogenesis and altered Wnt signaling after seizure induction. Tissue was analyzed using immunohistochemistry and confocal microscopy, and gene expression analysis was performed by RT-qPCR on RNA extracted from anatomically micro-dissected dentate gyri. Seizures increased neurogenesis and dendritic arborization of newborn hippocampal dentate granule cells in peri-ictal regions, and decreased neurogenesis in the ictal zone, 2-weeks after kainate injection. Interestingly, administration of the novel canonical Wnt pathway inhibitor XAV939 daily for 2-weeks after kainate injection further increased dendritic arborization in peri-ictal regions after seizure, without an effect on baseline neurogenesis in control animals. Transcriptome analysis of dentate gyri demonstrated significant canonical Wnt gene dysregulation in kainate-injected mice across all regions for Wnt3, 5a and 9a. Intriguingly, certain Wnt genes demonstrated differential patterns of dysregulation between the ictal and peri-ictal zones, most notably Wnt5B, 7B and DKK-1. Together, these results demonstrate regional variation in Wnt pathway dysregulation early after seizure induction, and surprisingly, suggest that some Wnt-mediated effects might actually temper aberrant neurogenesis after seizures. The Wnt pathway may therefore provide suitable targets for novel therapies that prevent network remodeling and the development of epileptic foci in high-risk patients.
Collapse
|
24
|
Tunc-Ozcan E, Peng CY, Zhu Y, Dunlop SR, Contractor A, Kessler JA. Activating newborn neurons suppresses depression and anxiety-like behaviors. Nat Commun 2019; 10:3768. [PMID: 31434877 PMCID: PMC6704083 DOI: 10.1038/s41467-019-11641-8] [Citation(s) in RCA: 112] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Accepted: 07/26/2019] [Indexed: 12/18/2022] Open
Abstract
The etiology of major depressive disorder (MDD), the leading cause of worldwide disability, is unknown. The neurogenic hypothesis proposes that MDD is linked to impairments of adult neurogenesis in the hippocampal dentate gyrus (DG), while the effects of antidepressants are mediated by increased neurogenesis. However, alterations in neurogenesis and endophenotypes are not always causally linked, and the relationship between increased neurogenesis and altered behavior is controversial. To address causality, we used chemogenetics in transgenic mice to selectively manipulate activity of newborn DG neurons. Suppressing excitability of newborn neurons without altering neurogenesis abolish the antidepressant effects of fluoxetine. Remarkably, activating these neurons is sufficient to alleviate depression-like behavior and reverse the adverse effects of unpredictable chronic mild stress. Our results demonstrate a direct causal relationship between newborn neuronal activity and affective behavior. Thus, strategies that target not only neurogenesis but also activity of newborn neurons may lead to more effective antidepressants. It is unclear if there is a causal link between increased neurogenesis and altered affective behaviors in major depressive disorders. Here, the authors show that selectively suppressing the excitability of newborn neurons, without altering neurogenesis, abolishes the antidepressant effects of fluoxetine.
Collapse
Affiliation(s)
- Elif Tunc-Ozcan
- Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA.
| | - Chian-Yu Peng
- Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Yiwen Zhu
- Department of Physiology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Sara R Dunlop
- Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Anis Contractor
- Department of Physiology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - John A Kessler
- Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| |
Collapse
|
25
|
Defining the Vulnerability Window of Anesthesia-Induced Neuroapoptosis in Developing Dentate Gyrus Granule Cells - A Transgenic Approach Utilizing POMC-EGFP Mice. Neuroscience 2019; 415:59-69. [PMID: 31301366 DOI: 10.1016/j.neuroscience.2019.07.009] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2019] [Revised: 05/20/2019] [Accepted: 07/02/2019] [Indexed: 11/22/2022]
Abstract
Exposure to commonly used anesthetics is associated with widespread neuroapoptosis in neonatal animals. Vulnerability of developing hippocampal dentate gyrus granule cells to anesthetic neurotoxicity peaks approximately 2 weeks after cell birth, as measured by bromodeoxyuridine birth dating, regardless of the age of the animal. The present study examined whether the vulnerable window can be further characterized by utilizing a transgenic approach. Proopiomelanocortin enhanced green fluorescent protein (POMC-EGFP) mice (postnatal day 21) were exposed to 3% sevoflurane for 6 h. Following exposure, cleaved caspase 3, expression of EGFP and differential maturational markers were quantified and compared with unanesthetized littermates. Electrophysiological properties of EGFP+ and EGFP- cells in the subgranular zone and the inner half of the granule cell layer were recorded by whole-cell patch-clamp. We found that sevoflurane significantly increased apoptosis of POMC-EGFP+ granule cells that accounted for approximate 1/3 of all apoptotic cells in dentate gyrus. Apoptotic EGFP- granule cells more frequently expressed the immature neuronal marker calretinin (75.4% vs 45.0%, P < 0.001) and less frequently the late progenitor marker NeuroD1 (21.9% vs 87.9%, P < 0.001) than EGFP+ granule cells. Although EGFP- granule cells were more mature in immunostaining than EGFP+ granule cells, their electrophysiological properties partially overlapped in terms of input resistance, resting membrane potential and action potential amplitude. Our results revealed the POMC stage, when GABA acts as an excitatory neurotransmitter, only partly captures susceptibility to anesthetic neurotoxicity, suggesting the vulnerable window of anesthesia-induced neuroapoptosis extends from the end of POMC+ stage to the post-POMC+ stage when depolarizing glutamatergic inputs emerge.
Collapse
|
26
|
Trivino-Paredes JS, Nahirney PC, Pinar C, Grandes P, Christie BR. Acute slice preparation for electrophysiology increases spine numbers equivalently in the male and female juvenile hippocampus: a DiI labeling study. J Neurophysiol 2019; 122:958-969. [PMID: 31268808 DOI: 10.1152/jn.00332.2019] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Hippocampal slices are widely used for in vitro electrophysiological experiments to study underlying mechanisms for synaptic transmission and plasticity, and there is a growing appreciation for sex differences in synaptic plasticity. To date, several studies have shown that the process of making slices from male animals can induce synaptogenesis in cornu ammonis area 1 (CA1) pyramidal cells, but there is a paucity of data for females and other brain regions. In the current study we use microcrystals of the lipophilic carbocyanine dye DiI (1,1'-dioctadecyl-3,3,3',3'-tetramethylindocarbocyanine perchlorate) to stain individual neurons in the CA1 and dentate gyrus (DG) hippocampal subfields of postnatal day 21 male and female rats. We show that the preparation of sections for electrophysiology produces significant increases in spines in sections obtained from females, similar to that observed in males. We also show that the procedures used for in vitro electrophysiology also result in significant spine increases in the DG and CA1 subfields. These results demonstrate the utility of this refined DiI procedure for staining neuronal dendrites and spines. They also show, for the first time, that in vitro electrophysiology slice preparations enhance spine numbers on hippocampal cells equivalently in both juvenile females and males.NEW & NOTEWORTHY This study introduces a new DiI technique that elucidates differences in spine numbers in juvenile female and male hippocampus, and shows that slice preparations for hippocampal electrophysiology in vitro may mask these differences.
Collapse
Affiliation(s)
- J S Trivino-Paredes
- Division of Medical Sciences, University of Victoria, Victoria, British Columbia, Canada
| | - P C Nahirney
- Division of Medical Sciences, University of Victoria, Victoria, British Columbia, Canada.,Island Medical Program, University of British Columbia, Victoria, British Columbia, Canada.,Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, British Columbia, Canada
| | - C Pinar
- Division of Medical Sciences, University of Victoria, Victoria, British Columbia, Canada
| | - P Grandes
- Division of Medical Sciences, University of Victoria, Victoria, British Columbia, Canada.,Department of Neurosciences, Faculty of Medicine and Nursing, University of the Basque Country UPV/EHU, Leioa, Vizcaya, Spain.,Achucarro Basque Center for Neuroscience, Science Park of the UPV/EHU, Leioa, Vizcaya, Spain
| | - B R Christie
- Division of Medical Sciences, University of Victoria, Victoria, British Columbia, Canada.,Island Medical Program, University of British Columbia, Victoria, British Columbia, Canada.,Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
27
|
Jung D, Kim S, Sariev A, Sharif F, Kim D, Royer S. Dentate granule and mossy cells exhibit distinct spatiotemporal responses to local change in a one-dimensional landscape of visual-tactile cues. Sci Rep 2019; 9:9545. [PMID: 31267019 PMCID: PMC6606600 DOI: 10.1038/s41598-019-45983-6] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Accepted: 06/19/2019] [Indexed: 11/09/2022] Open
Abstract
The dentate gyrus (DG) is critical for detecting changes in environments; however, how granule cells (GCs) and mossy cells (MCs), the two excitatory cell types of the DG, respond to small changes in the object layout is unclear. Here, we recorded GCs and MCs, identified by spike feature and optogenetic tagging, as mice ran on a treadmill belt enriched with visual-tactile cues. We observed that fixing a new cue on the belt induced a reconfiguration of GC and MC spatial representations via the emergence, extinction and rate alteration of firing fields. For both GCs and MCs, the response was maximal near the cue and spread over the entire belt. However, compared to the GC response, the MC response was stronger and more immediate, peaked at a slightly earlier belt position, and exhibited a transient component reminiscent of neuromodulatory activity. A competitive neural network model reproduced the GC response contingent on both the introduction of new object-vector inputs and the reconfiguration of MC activity, the former being critical for spreading the GC response in locations distant from the cue. These findings suggest that GCs operate as a competitive network and that MCs precede GCs in detecting changes and help expand the range of GC pattern separation.
Collapse
Affiliation(s)
- Dajung Jung
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon, 34141, Republic of Korea
- Center for Functional Connectomics, Korea Institute of Science and Technology, Seoul, 02792, Republic of Korea
| | - Soyoun Kim
- Center for Functional Connectomics, Korea Institute of Science and Technology, Seoul, 02792, Republic of Korea
| | - Anvar Sariev
- Center for Functional Connectomics, Korea Institute of Science and Technology, Seoul, 02792, Republic of Korea
- Division of Bio-Medical Science and Technology, KIST School, Korea University of Science and Technology, Seoul, 02792, Republic of Korea
| | - Farnaz Sharif
- Center for Functional Connectomics, Korea Institute of Science and Technology, Seoul, 02792, Republic of Korea
| | - Daesoo Kim
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon, 34141, Republic of Korea
| | - Sebastien Royer
- Center for Functional Connectomics, Korea Institute of Science and Technology, Seoul, 02792, Republic of Korea.
- Division of Bio-Medical Science and Technology, KIST School, Korea University of Science and Technology, Seoul, 02792, Republic of Korea.
| |
Collapse
|
28
|
Chatzi C, Zhang Y, Hendricks WD, Chen Y, Schnell E, Goodman RH, Westbrook GL. Exercise-induced enhancement of synaptic function triggered by the inverse BAR protein, Mtss1L. eLife 2019; 8:45920. [PMID: 31232686 PMCID: PMC6609409 DOI: 10.7554/elife.45920] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2019] [Accepted: 06/22/2019] [Indexed: 01/11/2023] Open
Abstract
Exercise is a potent enhancer of learning and memory, yet we know little of the underlying mechanisms that likely include alterations in synaptic efficacy in the hippocampus. To address this issue, we exposed mice to a single episode of voluntary exercise, and permanently marked activated mature hippocampal dentate granule cells using conditional Fos-TRAP mice. Exercise-activated neurons (Fos-TRAPed) showed an input-selective increase in dendritic spines and excitatory postsynaptic currents at 3 days post-exercise, indicative of exercise-induced structural plasticity. Laser-capture microdissection and RNASeq of activated neurons revealed that the most highly induced transcript was Mtss1L, a little-studied I-BAR domain-containing gene, which we hypothesized could be involved in membrane curvature and dendritic spine formation. shRNA-mediated Mtss1L knockdown in vivo prevented the exercise-induced increases in spines and excitatory postsynaptic currents. Our results link short-term effects of exercise to activity-dependent expression of Mtss1L, which we propose as a novel effector of activity-dependent rearrangement of synapses.
Collapse
Affiliation(s)
- Christina Chatzi
- Vollum Institute, Oregon Health & Science University, Portland, United States
| | - Yingyu Zhang
- Vollum Institute, Oregon Health & Science University, Portland, United States
| | - Wiiliam D Hendricks
- Vollum Institute, Oregon Health & Science University, Portland, United States.,Neuroscience Graduate Program, Vollum Institute, Oregon Health & Science University, Portland, United States
| | - Yang Chen
- Vollum Institute, Oregon Health & Science University, Portland, United States.,Neuroscience Graduate Program, Vollum Institute, Oregon Health & Science University, Portland, United States
| | - Eric Schnell
- Department of Anesthesiology and Perioperative Medicine, Oregon Health & Science University, Portland, United States.,Portland VA Health Care System, Portland, United States
| | - Richard H Goodman
- Vollum Institute, Oregon Health & Science University, Portland, United States
| | - Gary L Westbrook
- Vollum Institute, Oregon Health & Science University, Portland, United States
| |
Collapse
|
29
|
Synaptic properties of newly generated granule cells support sparse coding in the adult hippocampus. Behav Brain Res 2019; 372:112036. [PMID: 31201871 DOI: 10.1016/j.bbr.2019.112036] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Revised: 06/06/2019] [Accepted: 06/11/2019] [Indexed: 12/14/2022]
Abstract
In the adult hippocampus new neurons are continuously generated throughout life and integrate into the existing network via the formation of thousands of new synapses. Adult-born granule cells are known to improve learning and memory at about 3-6 weeks post mitosis by enhancing the brains ability to discriminate similar memory items. However, the underlying mechanisms are still controversial. Here we review the distinct functional properties of the newborn young neurons, including enhanced excitability, reduced GABAergic inhibition, NMDA-receptor dependent electrogenesis and enhanced synaptic plasticity. Although these cellular properties provide a competitive advantage for synapse formation, they do not generate 'hyperactivity' of young neurons. By contrast, in vivo evidence from immediate early gene expression and calcium imaging indicates that young neurons show sparse activity during learning. Similarly, in vitro data show a low number of high-impact synapses, leading to activation young cells by distinct subsets of afferent fibers with minimal overlap. Overall, the enhanced excitability of young cells does not generate hyperactivity but rather counterbalance the low number of excitatory input synapses. Finally, sparse coding in young neurons has been shown to be crucial for neurogenesis-dependent improvement of learning behavior. Taken together, converging evidence from cell physiology and behavioral studies suggests a mechanism that can explain the beneficial effects of adult neurogenesis on brain function.
Collapse
|
30
|
Villasana LE, Peters A, McCallum R, Liu C, Schnell E. Diazepam Inhibits Post-Traumatic Neurogenesis and Blocks Aberrant Dendritic Development. J Neurotrauma 2019; 36:2454-2467. [PMID: 30794026 DOI: 10.1089/neu.2018.6162] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Traumatic brain injury (TBI) triggers a robust increase in neurogenesis within the dentate gyrus of the hippocampus, but these new neurons undergo aberrant maturation and dendritic outgrowth. Because gamma-aminobutyric acid (GABA)A receptors (GABAARs) modulate dendritic outgrowth during constitutive neurogenesis and GABAAR-modulating sedatives are often administered to human patients after TBI, we investigated whether the benzodiazepine, diazepam (DZP), alters post-injury hippocampal neurogenesis. We used a controlled cortical impact (CCI) model of TBI in adult mice, and administered DZP or vehicle continuously for 1 week after injury via osmotic pump. Although DZP did not affect the neurogenesis rate in control mice, it almost completely prevented the TBI-induced increase in hippocampal neurogenesis as well as the aberrant dendritic growth of neurons born after TBI. DZP did not reduce cortical injury, reactive gliosis, or cell proliferation early after injury, but decreased c-Fos activation in the dentate gyrus at both early and late time-points after TBI, suggesting an association between neuronal activity and post-injury neurogenesis. Because DZP blocks post-injury neurogenesis, further studies are warranted to assess whether benzodiazepines alter cognitive recovery or the development of complications after TBI.
Collapse
Affiliation(s)
- Laura E Villasana
- 1Department of Anesthesiology and Perioperative Medicine, Oregon Health & Science University, Portland, Oregon
| | - Austin Peters
- 1Department of Anesthesiology and Perioperative Medicine, Oregon Health & Science University, Portland, Oregon
| | - Raluca McCallum
- 2Operative Care Division, VA Portland Health Care System, Portland, Oregon
| | - Chang Liu
- 1Department of Anesthesiology and Perioperative Medicine, Oregon Health & Science University, Portland, Oregon
| | - Eric Schnell
- 1Department of Anesthesiology and Perioperative Medicine, Oregon Health & Science University, Portland, Oregon.,2Operative Care Division, VA Portland Health Care System, Portland, Oregon
| |
Collapse
|
31
|
Yau SY, Bettio L, Chiu J, Chiu C, Christie BR. Fragile-X Syndrome Is Associated With NMDA Receptor Hypofunction and Reduced Dendritic Complexity in Mature Dentate Granule Cells. Front Mol Neurosci 2019; 11:495. [PMID: 30705620 PMCID: PMC6344420 DOI: 10.3389/fnmol.2018.00495] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2018] [Accepted: 12/27/2018] [Indexed: 12/11/2022] Open
Abstract
Fragile X syndrome (FXS) is the most common form of inherited intellectual disability. It is caused by the overexpansion of cytosine-guanine-guanine (CGG) trinucleotide in Fmr1 gene, resulting in complete loss of the fragile X mental retardation protein (FMRP). Previous studies using Fmr1 knockout (Fmr1 KO) mice have suggested that a N-methyl-D-aspartate receptors (NMDAR) hypofunction in the hippocampal dentate gyrus may partly contribute to cognitive impairments in FXS. Since activation of NMDAR plays an important role in dendritic arborization during neuronal development, we examined whether deficits in NMDAR function are associated with alterations in dendritic complexity in the hippocampal dentate region. The dentate granule cell layer (GCL) presents active postnatal neurogenesis, and consists of a heterogenous neuronal population with gradient ages from the superficial to its deep layer. Here, we show that neurons with multiple primary dendrites that reside in the outer GCL of Fmr1 KO mice display significantly smaller NMDAR excitatory post-synaptic currents (EPSCs) and a higher α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) to NMDA ratio in comparison to their wild-type counterparts. These deficits were associated with a significant decrease in dendritic complexity, with both dendritic length and number of intersections being significantly reduced. In contrast, although neurons with a single primary dendrite resided in the inner GCL of Fmr1 KO mice had a trend toward a reduction in NMDAR EPSCs and a higher AMPA/NMDA ratio, no alterations were found in dendritic complexity at this developmental stage. Our data indicate that the loss of FMRP causes NMDAR deficits and reduced dendritic complexity in granule neurons with multiple primary dendrites which are thought to be more mature in the GCL.
Collapse
Affiliation(s)
- Suk-Yu Yau
- Division of Medical Sciences, Island Medical Program, University of Victoria, Victoria, BC, Canada
| | - Luis Bettio
- Division of Medical Sciences, Island Medical Program, University of Victoria, Victoria, BC, Canada
| | - Jason Chiu
- Division of Medical Sciences, Island Medical Program, University of Victoria, Victoria, BC, Canada
| | - Christine Chiu
- Division of Medical Sciences, Island Medical Program, University of Victoria, Victoria, BC, Canada
| | - Brian R Christie
- Division of Medical Sciences, Island Medical Program, University of Victoria, Victoria, BC, Canada
| |
Collapse
|
32
|
Development of GABAergic Inputs Is Not Altered in Early Maturation of Adult Born Dentate Granule Neurons in Fragile X Mice. eNeuro 2018; 5:eN-NRS-0137-18. [PMID: 30627633 PMCID: PMC6325535 DOI: 10.1523/eneuro.0137-18.2018] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2018] [Revised: 09/27/2018] [Accepted: 10/17/2018] [Indexed: 02/08/2023] Open
Abstract
Fragile X syndrome (FXS) is the most common form of inherited mental retardation and the most common known cause of autism. Loss of fragile X mental retardation protein (FMRP) in mice (Fmr1 KO) leads to altered synaptic and circuit maturation in the hippocampus that is correlated with alterations in hippocampal-dependent behaviors. Previous studies have demonstrated that loss of FMRP increased the rate of proliferation of progenitor cells and altered their fate specification in adult Fmr1 KO mice. While these studies clearly demonstrate a role for FMRP in adult neurogenesis in the hippocampus, it is not known whether the functional synaptic maturation and integration of adult-born dentate granule cells (abDGCs) into hippocampal circuits is affected in Fmr1 KO mice. Here, we used retroviral labeling to birthdate abDGCs in Fmr1 KO mice which allowed us to perform targeted patch clamp recording to measure the development of synaptic inputs to these neurons at precise time points after differentiation. The frequency and amplitude of spontaneous GABAergic events increased over the first three weeks after differentiation; however, this normal development of GABAergic synapses was not altered in Fmr1 KO mice. Furthermore, the relatively depolarized GABA reversal potential (EGABA) in immature abDGCs was unaffected by loss of FMRP as was the development of dendritic arbor of the adult generated neurons. These studies systematically characterized the functional development of abDGCs during the first four weeks after differentiation and demonstrate that the maturation of GABAergic synaptic inputs to these neurons is not grossly affected by the loss of FMRP.
Collapse
|
33
|
Benito N, Gaborieau E, Sanz Diez A, Kosar S, Foucault L, Raineteau O, De Saint Jan D. A Pool of Postnatally Generated Interneurons Persists in an Immature Stage in the Olfactory Bulb. J Neurosci 2018; 38:9870-9882. [PMID: 30282727 PMCID: PMC6596244 DOI: 10.1523/jneurosci.1216-18.2018] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2018] [Revised: 07/31/2018] [Accepted: 08/09/2018] [Indexed: 02/01/2023] Open
Abstract
Calretinin (CR)-expressing periglomerular (PG) cells are the most abundant interneurons in the glomerular layer of the olfactory bulb. They are predominately generated postnatally from the septal and dorsal subventricular zones that continue producing them well into adulthood. Yet, little is known about their properties and functions. Using transgenic approaches and patch-clamp recording in mice of both sexes we show that CR(+) PG cells of both septal and dorsal origin have homogeneous morphological and electrophysiological properties. However, unlike other PG cells, these axonless neurons express a surprisingly small repertoire of voltage-activated channels and do not fire or fire at most a single and often small action potential. Moreover, they are not innervated by olfactory sensory neurons and receive little synaptic inputs from mitral or tufted cells at excitatory synapses where NMDA receptors predominate. These membrane and synaptic properties, that resemble those of newborn immature neurons not yet integrated in the network, persist over time and limit the recruitment of CR(+) PG cells by afferent inputs that strongly drive local network activity. Together, our results show that postnatally generated CR(+) PG cells continuously supply a large pool of neurons with unconventional properties. These data also question the contribution of CR(+) PG cells in olfactory bulb computation.SIGNIFICANCE STATEMENT Calretinin-expressing PG cells are by far the most abundant interneurons in the glomerular layer of the olfactory bulb. They are continuously produced during postnatal life, including adulthood, from neural stem cells located in the subventricular zones. Surprisingly, unlike other postnatally generated newborn neurons that quickly integrate into preexisting olfactory bulb networks, calretinin-expressing PG cells retain immature properties that limit their recruitment in local network activity for weeks, if not months, as if they would never fully mature. The function of this so far unsuspected pool of latent neurons is still unknown.
Collapse
Affiliation(s)
- Nuria Benito
- Institut des Neurosciences Cellulaires et Intégratives, Centre National de la Recherche Scientifique, Université de Strasbourg, 67084 Strasbourg, France, and
| | - Elodie Gaborieau
- Univ Lyon, Université Claude Bernard Lyon 1, Inserm, Stem Cell and Brain Research Institute U1208, 69500 Bron, France
| | - Alvaro Sanz Diez
- Institut des Neurosciences Cellulaires et Intégratives, Centre National de la Recherche Scientifique, Université de Strasbourg, 67084 Strasbourg, France, and
| | - Seher Kosar
- Institut des Neurosciences Cellulaires et Intégratives, Centre National de la Recherche Scientifique, Université de Strasbourg, 67084 Strasbourg, France, and
| | - Louis Foucault
- Univ Lyon, Université Claude Bernard Lyon 1, Inserm, Stem Cell and Brain Research Institute U1208, 69500 Bron, France
| | - Olivier Raineteau
- Univ Lyon, Université Claude Bernard Lyon 1, Inserm, Stem Cell and Brain Research Institute U1208, 69500 Bron, France
| | - Didier De Saint Jan
- Institut des Neurosciences Cellulaires et Intégratives, Centre National de la Recherche Scientifique, Université de Strasbourg, 67084 Strasbourg, France, and
| |
Collapse
|
34
|
Zhang H, He X, Mei Y, Ling Q. Ablation of ErbB4 in parvalbumin-positive interneurons inhibits adult hippocampal neurogenesis through down-regulating BDNF/TrkB expression. J Comp Neurol 2018; 526:2482-2492. [PMID: 30329159 DOI: 10.1002/cne.24506] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2018] [Revised: 07/06/2018] [Accepted: 07/24/2018] [Indexed: 12/20/2022]
Abstract
Parvalbumin (PV) positive interneurons in the subgranular zone (SGZ) can regulate adult hippocampal neurogenesis. ErbB4 is mainly expressed in PV neurons in the hippocampus and is crucial for keeping normal function of PV neurons. However, whether ErbB4 in PV interneurons affects the adult hippocampal neurogenesis remains unknown. In the present study, we deleted ErbB4 specifically in PV neurons by crossing PV-Cre mice with ErbB4f/f mice. Results of BrdU labeling and NeuN staining revealed that the proliferation of neural progenitors was increased but the survival and maturation of newborn neurons were decreased in the hippocampus of mice after deleting ErbB4 in PV neurons, suggesting that ErbB4 in PV neurons is closely associated with the process of adult hippocampal neurogenesis. Interestingly, the expression of brain-derived neurotrophic factor (BDNF) and its receptor, tropomyosin-related kinase B (TrkB), was significantly decreased in the hippocampus of ErbB4-deleted mice. Together, our data suggested that ErbB4 in PV neurons might modulate adult hippocampal neurogenesis by affecting BDNF-TrkB signaling pathway.
Collapse
Affiliation(s)
- Heng Zhang
- Department of Basic Medicine, School of Medicine, Shaoxing University, Shaoxing, Zhejiang, China.,Department of Neurobiology, Key Laboratory of Medical Neurobiology (Ministry of Health of China), Key Laboratory of Neurobiology of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Xiao He
- Department of Neurobiology, Key Laboratory of Medical Neurobiology (Ministry of Health of China), Key Laboratory of Neurobiology of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.,Department of Nuclear Medicine, The Second Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.,Zhejiang University Medical PET Center, Hangzhou, Zhejiang, China
| | - Yufei Mei
- Department of Neurobiology, Key Laboratory of Medical Neurobiology (Ministry of Health of China), Key Laboratory of Neurobiology of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Qingzhou Ling
- Human resources office, Shaoxing University, Shaoxing, Zhejiang, China
| |
Collapse
|
35
|
Uneven balance of power between hypothalamic peptidergic neurons in the control of feeding. Proc Natl Acad Sci U S A 2018; 115:E9489-E9498. [PMID: 30224492 PMCID: PMC6176613 DOI: 10.1073/pnas.1802237115] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
The interplay between the anorexigenic and orexigenic neurons in the arcuate nucleus that contributes to the control of feeding remains elusive. Using optogenetic stimulation, we show that activation of POMC neurons rapidly inhibits feeding behavior in fasted animals. However, simultaneous stimulation of both POMC neurons and a subset of the orexigenic neurons that express AgRP is sufficient to reverse that inhibition and trigger intense feeding behavior. We used 3D imaging and functional studies to illuminate the anatomical underpinning of both the inhibitory and excitatory events. Our work suggests that translational applications that aim to control appetite need to target the activation rather than the inhibition mechanisms. Two classes of peptide-producing neurons in the arcuate nucleus (Arc) of the hypothalamus are known to exert opposing actions on feeding: the anorexigenic neurons that express proopiomelanocortin (POMC) and the orexigenic neurons that express agouti-related protein (AgRP) and neuropeptide Y (NPY). These neurons are thought to arise from a common embryonic progenitor, but our anatomical and functional understanding of the interplay of these two peptidergic systems that contribute to the control of feeding remains incomplete. The present study uses a combination of optogenetic stimulation with viral and transgenic approaches, coupled with neural activity mapping and brain transparency visualization to demonstrate the following: (i) selective activation of Arc POMC neurons inhibits food consumption rapidly in unsated animals; (ii) activation of Arc neurons arising from POMC-expressing progenitors, including POMC and a subset of AgRP neurons, triggers robust feeding behavior, even in the face of satiety signals from POMC neurons; (iii) the opposing effects on food intake are associated with distinct neuronal projection and activation patterns of adult hypothalamic POMC neurons versus Arc neurons derived from POMC-expressing lineages; and (iv) the increased food intake following the activation of orexigenic neurons derived from POMC-expressing progenitors engages an extensive neural network that involves the endogenous opioid system. Together, these findings shed further light on the dynamic balance between two peptidergic systems in the moment-to-moment regulation of feeding behavior.
Collapse
|
36
|
Constitutive and Synaptic Activation of GIRK Channels Differentiates Mature and Newborn Dentate Granule Cells. J Neurosci 2018; 38:6513-6526. [PMID: 29915136 DOI: 10.1523/jneurosci.0674-18.2018] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Revised: 05/30/2018] [Accepted: 05/30/2018] [Indexed: 12/21/2022] Open
Abstract
Sparse neural activity in the dentate gyrus is enforced by powerful networks of inhibitory GABAergic interneurons in combination with low intrinsic excitability of the principal neurons, the dentate granule cells (GCs). Although the cellular and circuit properties that dictate synaptic inhibition are well studied, less is known about mechanisms that confer low GC intrinsic excitability. Here we demonstrate that intact G protein-mediated signaling contributes to the characteristic low resting membrane potential that differentiates mature dentate GCs from CA1 pyramidal cells and developing adult-born GCs. In mature GCs from male and female mice, intact G protein signaling robustly reduces intrinsic excitability, whereas deletion of G protein-activated inwardly rectifying potassium channel 2 (GIRK2) increases excitability and blocks the effects of G protein signaling on intrinsic properties. Similarly, pharmacological manipulation of GABAB receptors (GABABRs) or GIRK channels alters intrinsic excitability and GC spiking behavior. However, adult-born new GCs lack functional GIRK activity, with phasic and constitutive GABABR-mediated GIRK signaling appearing after several weeks of maturation. Phasic activation is interneuron specific, arising primarily from nNOS-expressing interneurons rather than parvalbumin- or somatostatin-expressing interneurons. Together, these results demonstrate that G protein signaling contributes to the intrinsic excitability that differentiates mature and developing dentate GCs and further suggest that late maturation of GIRK channel activity is poised to convert early developmental functions of GABAB receptor signaling into GABABR-mediated inhibition.SIGNIFICANCE STATEMENT The dentate gyrus exhibits sparse neural activity that is essential for the computational function of pattern separation. Sparse activity is ascribed to strong local inhibitory circuits in combination with low intrinsic excitability of the principal neurons, the granule cells. Here we show that constitutive activity of G protein-coupled inwardly rectifying potassium channels (GIRKs) underlies to the hallmark low resting membrane potential and input resistance of mature dentate neurons. Adult-born neurons initially lack functional GIRK channels, with constitutive and phasic GABAB receptor-mediated GIRK inhibition developing in tandem after several weeks of maturation. Our results reveal that GABAB/GIRK activity is an important determinant of low excitability of mature dentate granule cells that may contribute to sparse DG activity in vivo.
Collapse
|
37
|
A novel pyramidal cell type promotes sharp-wave synchronization in the hippocampus. Nat Neurosci 2018; 21:985-995. [PMID: 29915194 DOI: 10.1038/s41593-018-0172-7] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2017] [Accepted: 04/26/2018] [Indexed: 01/05/2023]
Abstract
To support cognitive function, the CA3 region of the hippocampus performs computations involving attractor dynamics. Understanding how cellular and ensemble activities of CA3 neurons enable computation is critical for elucidating the neural correlates of cognition. Here we show that CA3 comprises not only classically described pyramid cells with thorny excrescences, but also includes previously unidentified 'athorny' pyramid cells that lack mossy-fiber input. Moreover, the two neuron types have distinct morphological and physiological phenotypes and are differentially modulated by acetylcholine. To understand the contribution of these athorny pyramid neurons to circuit function, we measured cell-type-specific firing patterns during sharp-wave synchronization events in vivo and recapitulated these dynamics with an attractor network model comprising two principal cell types. Our data and simulations reveal a key role for athorny cell bursting in the initiation of sharp waves: transient network attractor states that signify the execution of pattern completion computations vital to cognitive function.
Collapse
|
38
|
Preferential Targeting of Lateral Entorhinal Inputs onto Newly Integrated Granule Cells. J Neurosci 2018; 38:5843-5853. [PMID: 29793975 DOI: 10.1523/jneurosci.1737-17.2018] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2017] [Revised: 04/26/2018] [Accepted: 05/17/2018] [Indexed: 12/20/2022] Open
Abstract
Mature dentate granule cells in the hippocampus receive input from the entorhinal cortex via the perforant path in precisely arranged lamina, with medial entorhinal axons innervating the middle molecular layer and lateral entorhinal cortex axons innervating the outer molecular layer. Although vastly outnumbered by mature granule cells, adult-generated newborn granule cells play a unique role in hippocampal function, which has largely been attributed to their enhanced excitability and plasticity (Schmidt-Hieber et al., 2004; Ge et al., 2007). Inputs from the medial and lateral entorhinal cortex carry different informational content. Thus, the distribution of inputs onto newly integrated granule cells will affect their function in the circuit. Using retroviral labeling in combination with selective optogenetic activation of medial or lateral entorhinal inputs, we examined the functional innervation and synaptic maturation of newly generated dentate granule cells in the mouse hippocampus. Our results indicate that lateral entorhinal inputs provide the majority of functional innervation of newly integrated granule cells at 21 d postmitosis. Despite preferential functional targeting, the dendritic spine density of immature granule cells was similar in the outer and middle molecular layers, which we speculate could reflect an unequal distribution of shaft synapses. However, chronic blockade of neurotransmitter release of medial entorhinal axons with tetanus toxin disrupted normal synapse development of both medial and lateral entorhinal inputs. Our results support a role for preferential lateral perforant path input onto newly generated neurons in mediating pattern separation, but also indicate that medial perforant path input is necessary for normal synaptic development.SIGNIFICANCE STATEMENT The formation of episodic memories involves the integration of contextual and spatial information. Newly integrated neurons in the dentate gyrus of the hippocampus play a critical role in this process, despite constituting only a minor fraction of the total number of granule cells. Here we demonstrate that these neurons preferentially receive information thought to convey the context of an experience. Each newly integrated granule cell plays this unique role for ∼1 month before reaching maturity.
Collapse
|
39
|
Yau SY, Li A, Tong JB, Bostrom C, Christie BR, Lee TMC, So KF. Chronic corticosterone administration reduces dendritic complexity in mature, but not young granule cells in the rat dentate gyrus. Restor Neurol Neurosci 2018; 34:849-57. [PMID: 27567758 PMCID: PMC5345640 DOI: 10.3233/rnn-160662] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Background: Our previous work has shown that exposure to the stress hormone corticosterone (40 mg/kg CORT) for two weeks induces dendritic atrophy of pyramidal neurons in the hippocampal CA3 region and behavioral deficits. However, it is unclear whether this treatment also affects the dentate gyrus (DG), a subregion of the hippocampus comprising a heterogeneous population of young and mature neurons. Objective: We examined the effect of CORT treatment on the dendritic complexity of mature and young granule cells in the DG. Methods: We utilized a Golgi staining method to investigate the dendritic morphology and spine density of young neurons in the inner granular cell layer (GCL) and mature neurons in the outer GCL in response to CORT application. The expressions of glucocorticoid receptors during neuronal maturation were examined using Western blot analysis in a primary hippocampal neuronal culture. Results: Sholl analysis revealed that CORT treatment decreased the number of intersections and shortened the dendritic length in mature, but not young, granule cells. However, the spine density of mature and young neurons was not affected. Western blot analysis showed a progressive increase in the protein levels of glucocorticoid receptors (GRs) in the cultured primary hippocampal neurons during neuronal maturation. Conclusion: These data suggest that mature neurons are likely more vulnerable to chronic exposure to CORT; this may be due to their higher expression of GRs when compared to younger DG neurons.
Collapse
Affiliation(s)
- Suk-Yu Yau
- Department of Rehabilitation Sciences, The Hong Kong Polytechnic University, Hong Kong SAR
| | - Ang Li
- Guangdong-Hong Kong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou, China
| | - Jian-Bin Tong
- Department of Anesthesiology, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Crystal Bostrom
- Division of Medical Sciences, The University of Victoria, British Columbia, Canada
| | - Brian R Christie
- Division of Medical Sciences, The University of Victoria, British Columbia, Canada
| | - Tatia M C Lee
- State Key Laboratory of Brain and Cognitive Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR.,Laboratory of Neuropsychology, The University of Hong Kong, Hong Kong SAR.,Laboratory of Cognitive Affective Neuroscience, The University of Hong Kong, Hong Kong SAR
| | - Kwok-Fai So
- Guangdong-Hong Kong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou, China.,State Key Laboratory of Brain and Cognitive Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR.,Department of Ophthalmology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR.,Co-innovation Center of Neuroregeneration, Jiangsu, China
| |
Collapse
|
40
|
Trajectory Analysis Unveils Reelin's Role in the Directed Migration of Granule Cells in the Dentate Gyrus. J Neurosci 2017; 38:137-148. [PMID: 29138282 DOI: 10.1523/jneurosci.0988-17.2017] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2017] [Revised: 11/01/2017] [Accepted: 11/03/2017] [Indexed: 11/21/2022] Open
Abstract
Reelin controls neuronal migration and layer formation. Previous studies in reeler mice deficient in Reelin focused on the result of the developmental process in fixed tissue sections. It has remained unclear whether Reelin affects the migratory process, migration directionality, or migrating neurons guided by the radial glial scaffold. Moreover, Reelin has been regarded as an attractive signal because newly generated neurons migrate toward the Reelin-containing marginal zone. Conversely, Reelin might be a stop signal because migrating neurons in reeler, but not in wild-type mice, invade the marginal zone. Here, we monitored the migration of newly generated proopiomelanocortin-EGFP-expressing dentate granule cells in slice cultures from reeler, reeler-like mutants and wild-type mice of either sex using real-time microscopy. We discovered that not the actual migratory process and migratory speed, but migration directionality of the granule cells is controlled by Reelin. While wild-type granule cells migrated toward the marginal zone of the dentate gyrus, neurons in cultures from reeler and reeler-like mutants migrated randomly in all directions as revealed by vector analyses of migratory trajectories. Moreover, live imaging of granule cells in reeler slices cocultured to wild-type dentate gyrus showed that the reeler neurons changed their directions and migrated toward the Reelin-containing marginal zone of the wild-type culture, thus forming a compact granule cell layer. In contrast, directed migration was not observed when Reelin was ubiquitously present in the medium of reeler slices. These results indicate that topographically administered Reelin controls the formation of a granule cell layer.SIGNIFICANCE STATEMENT Neuronal migration and the various factors controlling its onset, speed, directionality, and arrest are poorly understood. Slice cultures offer a unique model to study the migration of individual neurons in an almost natural environment. In the present study, we took advantage of the expression of proopiomelanocortin-EGFP by newly generated, migrating granule cells to analyze their migratory trajectories in hippocampal slice cultures from wild-type mice and mutants deficient in Reelin signaling. We show that the compartmentalized presence of Reelin is essential for the directionality, but not the actual migratory process or speed, of migrating granule cells leading to their characteristic lamination in the dentate gyrus.
Collapse
|
41
|
Zhang Y, Kibaly C, Xu C, Loh HH, Law PY. Temporal effect of manipulating NeuroD1 expression with the synthetic small molecule KHS101 on morphine contextual memory. Neuropharmacology 2017; 126:58-69. [DOI: 10.1016/j.neuropharm.2017.08.030] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2017] [Revised: 08/10/2017] [Accepted: 08/21/2017] [Indexed: 12/20/2022]
|
42
|
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
|
43
|
Encinas JM, Fitzsimons CP. Gene regulation in adult neural stem cells. Current challenges and possible applications. Adv Drug Deliv Rev 2017; 120:118-132. [PMID: 28751200 DOI: 10.1016/j.addr.2017.07.016] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2017] [Revised: 07/17/2017] [Accepted: 07/19/2017] [Indexed: 12/13/2022]
Abstract
Adult neural stem and progenitor cells (NSPCs) offer a unique opportunity for neural regeneration and niche modification in physiopathological conditions, harnessing the capability to modify from neuronal circuits to glial scar. Findings exposing the vast plasticity and potential of NSPCs have accumulated over the past years and we currently know that adult NSPCs can naturally give rise not only to neurons but also to astrocytes and reactive astrocytes, and eventually to oligodendrocytes through genetic manipulation. We can consider NSPCs as endogenous flexible tools to fight against neurodegenerative and neurological disorders and aging. In addition, NSPCs can be considered as active agents contributing to chronic brain alterations and as relevant cell populations to be preserved, so that their main function, neurogenesis, is not lost in damage or disease. Altogether we believe that learning to manipulate NSPC is essential to prevent, ameliorate or restore some of the cognitive deficits associated with brain disease and injury, and therefore should be considered as target for future therapeutic strategies. The first step to accomplish this goal is to target them specifically, by unveiling and understanding their unique markers and signaling pathways.
Collapse
Affiliation(s)
- Juan Manuel Encinas
- Achucarro Basque Center for Neuroscience, Bizkaia Science and Technology Park, 205, 48170 Zamudio, Spain; Ikerbasque, The Basque Science Foundation, María Díaz de Haro 3, 6(th) Floor, 48013 Bilbao, Spain; University of the Basque Country (UPV/EHU), Barrio Sarriena s/n, 48940 Leioa, Spain.
| | - Carlos P Fitzsimons
- Neuroscience Program, Swammerdam Institute for Life Sciences, Faculty of Sciences, University of Amsterdam, SciencePark 904, 1098XH Amsterdam, The Netherlands.
| |
Collapse
|
44
|
Eliwa H, Belzung C, Surget A. Adult hippocampal neurogenesis: Is it the alpha and omega of antidepressant action? Biochem Pharmacol 2017; 141:86-99. [DOI: 10.1016/j.bcp.2017.08.005] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2017] [Accepted: 08/04/2017] [Indexed: 01/08/2023]
|
45
|
Neural mechanisms underlying GABAergic regulation of adult hippocampal neurogenesis. Cell Tissue Res 2017; 371:33-46. [PMID: 28948349 DOI: 10.1007/s00441-017-2668-y] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2017] [Accepted: 07/01/2017] [Indexed: 12/25/2022]
Abstract
Within the dentate gyrus of the adult hippocampus is the subgranular zone, which contains a neurogenic niche for radial-glia like cells, the most primitive neural stem cells in the adult brain. The quiescence of neural stem cells is maintained by tonic gamma-aminobutyric acid (GABA) released from local interneurons. Once these cells differentiate into neural progenitor cells, GABA continues to regulate their development into mature granule cells, the principal cell type of the dentate gyrus. Here, we review the role of GABA circuits, signaling, and receptors in regulating development of adult-born cells, as well as the molecular players that modulate GABA signaling. Furthermore, we review recent findings linking dysregulation of adult hippocampal neurogenesis to the altered GABAergic circuitry and signaling under various pathological conditions.
Collapse
|
46
|
Sah N, Peterson BD, Lubejko ST, Vivar C, van Praag H. Running reorganizes the circuitry of one-week-old adult-born hippocampal neurons. Sci Rep 2017; 7:10903. [PMID: 28883658 PMCID: PMC5589841 DOI: 10.1038/s41598-017-11268-z] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2016] [Accepted: 07/05/2017] [Indexed: 12/23/2022] Open
Abstract
Adult hippocampal neurogenesis is an important form of structural and functional plasticity in the mature mammalian brain. The existing consensus is that GABA regulates the initial integration of adult-born neurons, similar to neuronal development during embryogenesis. Surprisingly, virus-based anatomical tracing revealed that very young, one-week-old, new granule cells in male C57Bl/6 mice receive input not only from GABAergic interneurons, but also from multiple glutamatergic cell types, including mature dentate granule cells, area CA1-3 pyramidal cells and mossy cells. Consistently, patch-clamp recordings from retrovirally labeled new granule cells at 7-8 days post retroviral injection (dpi) show that these cells respond to NMDA application with tonic currents, and that both electrical and optogenetic stimulation can evoke NMDA-mediated synaptic responses. Furthermore, new dentate granule cell number, morphology and excitatory synaptic inputs at 7 dpi are modified by voluntary wheel running. Overall, glutamatergic and GABAergic innervation of newly born neurons in the adult hippocampus develops concurrently, and excitatory input is reorganized by exercise.
Collapse
Affiliation(s)
- Nirnath Sah
- Neuroplasticity and Behavior Unit, Laboratory of Neurosciences, Intramural Research Program, National Institute on Aging, National Institutes of Health, Baltimore, MD, 21224, USA
| | - Benjamin D Peterson
- Neuroplasticity and Behavior Unit, Laboratory of Neurosciences, Intramural Research Program, National Institute on Aging, National Institutes of Health, Baltimore, MD, 21224, USA
| | - Susan T Lubejko
- Neuroplasticity and Behavior Unit, Laboratory of Neurosciences, Intramural Research Program, National Institute on Aging, National Institutes of Health, Baltimore, MD, 21224, USA
| | - Carmen Vivar
- Laboratory of Neurogenesis and Neuroplasticity, Department of Physiology, Biophysics, and Neuroscience, Center for Research and Advanced Studies of the National Polytechnic Institute, Mexico City, 07360, Mexico.
| | - Henriette van Praag
- Neuroplasticity and Behavior Unit, Laboratory of Neurosciences, Intramural Research Program, National Institute on Aging, National Institutes of Health, Baltimore, MD, 21224, USA.
| |
Collapse
|
47
|
Vidal V, García-Cerro S, Martínez P, Corrales A, Lantigua S, Vidal R, Rueda N, Ozmen L, Hernández MC, Martínez-Cué C. Decreasing the Expression of GABA A α5 Subunit-Containing Receptors Partially Improves Cognitive, Electrophysiological, and Morphological Hippocampal Defects in the Ts65Dn Model of Down Syndrome. Mol Neurobiol 2017; 55:4745-4762. [PMID: 28717969 DOI: 10.1007/s12035-017-0675-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2017] [Accepted: 06/27/2017] [Indexed: 12/31/2022]
Abstract
Trisomy 21 or Down syndrome (DS) is the most common cause of intellectual disability of a genetic origin. The Ts65Dn (TS) mouse, which is the most commonly used and best-characterized mouse model of DS, displays many of the cognitive, neuromorphological, and biochemical anomalies that are found in the human condition. One of the mechanisms that have been proposed to be responsible for the cognitive deficits in this mouse model is impaired GABA-mediated inhibition. Because of the well-known modulatory role of GABAA α5 subunit-containing receptors in cognitive processes, these receptors are considered to be potential targets for improving the intellectual disability in DS. The chronic administration of GABAA α5-negative allosteric modulators has been shown to be procognitive without anxiogenic or proconvulsant side effects. In the present study, we use a genetic approach to evaluate the contribution of GABAA α5 subunit-containing receptors to the cognitive, electrophysiological, and neuromorphological deficits in TS mice. We show that reducing the expression of GABAA α5 receptors by deleting one or two copies of the Gabra5 gene in TS mice partially ameliorated the cognitive impairments, improved long-term potentiation, enhanced neural differentiation and maturation, and normalized the density of the GABAergic synapse markers. Reducing the gene dosage of Gabra5 in TS mice did not induce motor alterations and anxiety or affect the viability of the mice. Our results provide further evidence of the role of GABAA α5 receptor-mediated inhibition in cognitive impairment in the TS mouse model of DS.
Collapse
Affiliation(s)
- Verónica Vidal
- Departamento de Fisiología y Farmacología, Facultad de Medicina, Universidad deCantabria, Santander, Spain
| | - Susana García-Cerro
- Departamento de Fundamentos Clínicos, Unidad de Farmacología, Universitat de Barcelona, Barcelona, Spain
| | - Paula Martínez
- Departamento de Fisiología y Farmacología, Facultad de Medicina, Universidad deCantabria, Santander, Spain
| | - Andrea Corrales
- Departamento de Fisiología y Farmacología, Facultad de Medicina, Universidad deCantabria, Santander, Spain
| | - Sara Lantigua
- Departamento de Fisiología y Farmacología, Facultad de Medicina, Universidad deCantabria, Santander, Spain
| | - Rebeca Vidal
- Departamento de Fisiología y Farmacología, Facultad de Medicina, Universidad deCantabria, Santander, Spain.,Instituto de Biomedicina y Biotecnología de Cantabria, IBBTEC (Universidad de Cantabria, CSIC, SODERCAN), Santander, Spain.,Centro de Investigacion Biomédica en Red de Salud Mental (CIBERSAM), Instituto de Salud Carlos III, Madrid, Spain
| | - Noemí Rueda
- Departamento de Fisiología y Farmacología, Facultad de Medicina, Universidad deCantabria, Santander, Spain
| | - Laurence Ozmen
- Pharma Research and Early Development, Hoffman-La Roche Ltd., Basel, Switzerland
| | | | - Carmen Martínez-Cué
- Departamento de Fisiología y Farmacología, Facultad de Medicina, Universidad deCantabria, Santander, Spain.
| |
Collapse
|
48
|
Adlaf EW, Vaden RJ, Niver AJ, Manuel AF, Onyilo VC, Araujo MT, Dieni CV, Vo HT, King GD, Wadiche JI, Overstreet-Wadiche L. Adult-born neurons modify excitatory synaptic transmission to existing neurons. eLife 2017; 6:19886. [PMID: 28135190 PMCID: PMC5279947 DOI: 10.7554/elife.19886] [Citation(s) in RCA: 67] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2016] [Accepted: 01/05/2017] [Indexed: 12/31/2022] Open
Abstract
Adult-born neurons are continually produced in the dentate gyrus but it is unclear whether synaptic integration of new neurons affects the pre-existing circuit. Here we investigated how manipulating neurogenesis in adult mice alters excitatory synaptic transmission to mature dentate neurons. Enhancing neurogenesis by conditional deletion of the pro-apoptotic gene Bax in stem cells reduced excitatory postsynaptic currents (EPSCs) and spine density in mature neurons, whereas genetic ablation of neurogenesis increased EPSCs in mature neurons. Unexpectedly, we found that Bax deletion in developing and mature dentate neurons increased EPSCs and prevented neurogenesis-induced synaptic suppression. Together these results show that neurogenesis modifies synaptic transmission to mature neurons in a manner consistent with a redistribution of pre-existing synapses to newly integrating neurons and that a non-apoptotic function of the Bax signaling pathway contributes to ongoing synaptic refinement within the dentate circuit. DOI:http://dx.doi.org/10.7554/eLife.19886.001 Neurogenesis, the creation of new brain cells called neurons, occurs primarily before birth. However, a region of the brain called the dentate gyrus, which is involved in memory, continues to produce new neurons throughout life. Recent studies suggest that adding neurons to the dentate gyrus helps the brain to distinguish between similar sights, sounds and smells. This in turn makes it easier to encode similar experiences as distinct memories. The brain’s outer layer, called the cortex, processes information from our senses and sends it, along with information about our location in space, to the dentate gyrus. By combining this sensory and spatial information, the dentate gyrus is able to generate a unique memory of an experience. But how does neurogenesis affect this process? As the dentate gyrus accumulates more neurons, the number of neurons in the cortex remains unchanged. Do some cortical neurons transfer their connections – called synapses – to the new neurons? Or does the brain generate additional synapses to accommodate the newborn cells? Adlaf et al. set out to answer this question by genetically modifying mice to alter the number of new neurons that could form in the dentate gyrus. Increasing the number of newborn neurons reduced the number of synapses between the cortex and the mature neurons in the dentate gyrus. Conversely, killing off newborn neurons had the opposite effect, increasing the strength of the synaptic connections to older cells. This suggests that new synapses are not formed to accommodate new neurons, but rather that there is a redistribution of synapses between old and new neurons in the dentate gyrus. Further work is required to determine how this redistribution of synapses contributes to how the dentate gyrus works. Does redistributing synapses disrupt existing memories? And how do these findings relate to the effects of exercise – does this natural way of increasing neurogenesis increase the overall number of synapses in the system, potentially creating enough connections for both new and old neurons? DOI:http://dx.doi.org/10.7554/eLife.19886.002
Collapse
Affiliation(s)
- Elena W Adlaf
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, United States
| | - Ryan J Vaden
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, United States
| | - Anastasia J Niver
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, United States
| | - Allison F Manuel
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, United States
| | - Vincent C Onyilo
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, United States
| | - Matheus T Araujo
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, United States
| | - Cristina V Dieni
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, United States
| | - Hai T Vo
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, United States
| | - Gwendalyn D King
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, United States
| | - Jacques I Wadiche
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, United States
| | | |
Collapse
|
49
|
Parent AS, Pinson A, Woods N, Chatzi C, Vaaga CE, Bensen A, Gérard A, Thome JP, Bourguignon JP, Westbrook GL. Early exposure to Aroclor 1254 in vivo disrupts the functional synaptic development of newborn hippocampal granule cells. Eur J Neurosci 2016; 44:3001-3010. [PMID: 27740705 DOI: 10.1111/ejn.13437] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2015] [Revised: 10/10/2016] [Accepted: 10/11/2016] [Indexed: 02/05/2023]
Abstract
Neurogenesis in the dentate gyrus is sensitive to endogenous and exogenous factors that influence hippocampal function. Ongoing neurogenesis and the integration of these new neurons throughout life thus may provide a sensitive indicator of environmental stress. We examined the effects of Aroclor 1254 (A1254), a mixture of polychlorinated biphenyls (PCBs), on the development and function of newly generated dentate granule cells. Early exposure to A1254 has been associated with learning impairment in children, suggesting potential impact on the development of hippocampus and/or cortical circuits. Oral A1254 (from the 6th day of gestation to postnatal day 21) produced the expected increase in PCB levels in brain at postnatal day 21, which persisted at lower levels into adulthood. A1254 did not affect the proliferation or survival of newborn neurons in immature animals nor did it cause overt changes in neuronal morphology. However, A1254 occluded the normal developmental increase in sEPSC frequency in the third post-mitotic week without altering the average sEPSC amplitude. Our results suggest that early exposure to PCBs can disrupt excitatory synaptic function during a period of active synaptogenesis, and thus could contribute to the cognitive effects noted in children exposed to PCBs.
Collapse
Affiliation(s)
- A S Parent
- Neuroendocrinology Unit, GIGA-N, University of Liège, CHU Sart Tilman, 4000, Liège, Belgium
| | - A Pinson
- Neuroendocrinology Unit, GIGA-N, University of Liège, CHU Sart Tilman, 4000, Liège, Belgium
| | - N Woods
- Vollum Institute, Portland, OR, USA
| | - C Chatzi
- Vollum Institute, Portland, OR, USA
| | | | - A Bensen
- Vollum Institute, Portland, OR, USA
| | - A Gérard
- Neuroendocrinology Unit, GIGA-N, University of Liège, CHU Sart Tilman, 4000, Liège, Belgium
| | - J P Thome
- Laboratory of Animal Ecology and Ecotoxicology, University of Liège, Liège, Belgium
| | - J P Bourguignon
- Neuroendocrinology Unit, GIGA-N, University of Liège, CHU Sart Tilman, 4000, Liège, Belgium
| | | |
Collapse
|
50
|
Pan H, Wang D, Zhang X, Zhou D, Zhang H, Qian Q, He X, Liu Z, Liu Y, Zheng T, Zhang L, Wang M, Sun B. Amyloid β Is Not the Major Factor Accounting for Impaired Adult Hippocampal Neurogenesis in Mice Overexpressing Amyloid Precursor Protein. Stem Cell Reports 2016; 7:707-718. [PMID: 27693425 PMCID: PMC5063569 DOI: 10.1016/j.stemcr.2016.08.019] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2016] [Revised: 08/30/2016] [Accepted: 08/31/2016] [Indexed: 11/22/2022] Open
Abstract
Adult hippocampal neurogenesis was impaired in several Alzheimer's disease models overexpressing mutant human amyloid precursor protein (hAPP). However, the effects of wild-type hAPP on adult neurogenesis and whether the impaired adult hippocampal neurogenesis was caused by amyloid β (Aβ) or APP remained unclear. Here, we found that neurogenesis was impaired in the dentate gyrus (DG) of adult mice overexpressing wild-type hAPP (hAPP-I5) compared with controls. However, the adult hippocampal neurogenesis was more severely impaired in hAPP-I5 than that in hAPP-J20 mice, which express similar levels of hAPP mRNA but much higher levels of Aβ. Furthermore, reducing Aβ levels did not affect the number of doublecortin-positive cells in the DG of hAPP-J20 mice. Our results suggested that hAPP was more likely an important factor inhibiting adult neurogenesis, and Aβ was not the major factor affecting neurogenesis in the adult hippocampus of hAPP mice. Overexpression of wild-type hAPP impairs adult hippocampal neurogenesis Adult hippocampal neurogenesis was more severely impaired in hAPP-I5 than in hAPP-J20 Reducing Aβ levels did not affect adult neurogenesis in hAPP-J20 mice Aβ is not the major factor accounting for impaired adult neurogenesis in hAPP mice
Collapse
Affiliation(s)
- Hongyu Pan
- Department of Neurobiology, School of Basic Medical Sciences, Key Laboratory of Medical Neurobiology (Ministry of Health of China), Key Laboratory of Neurobiology of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province 310058, China
| | - Dongpi Wang
- Department of Neurobiology, School of Basic Medical Sciences, Key Laboratory of Medical Neurobiology (Ministry of Health of China), Key Laboratory of Neurobiology of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province 310058, China
| | - Xiaoqin Zhang
- Department of Neurobiology, School of Basic Medical Sciences, Key Laboratory of Medical Neurobiology (Ministry of Health of China), Key Laboratory of Neurobiology of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province 310058, China
| | - Dongming Zhou
- Department of Neurobiology, School of Basic Medical Sciences, Key Laboratory of Medical Neurobiology (Ministry of Health of China), Key Laboratory of Neurobiology of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province 310058, China
| | - Heng Zhang
- Department of Neurobiology, School of Basic Medical Sciences, Key Laboratory of Medical Neurobiology (Ministry of Health of China), Key Laboratory of Neurobiology of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province 310058, China
| | - Qi Qian
- Department of Neurology, Brain Medical Center, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province 310058, China
| | - Xiao He
- Department of Neurobiology, School of Basic Medical Sciences, Key Laboratory of Medical Neurobiology (Ministry of Health of China), Key Laboratory of Neurobiology of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province 310058, China
| | - Zhaoling Liu
- Department of Neurobiology, School of Basic Medical Sciences, Key Laboratory of Medical Neurobiology (Ministry of Health of China), Key Laboratory of Neurobiology of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province 310058, China
| | - Yunjin Liu
- Department of Neurobiology, School of Basic Medical Sciences, Key Laboratory of Medical Neurobiology (Ministry of Health of China), Key Laboratory of Neurobiology of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province 310058, China
| | - Tingting Zheng
- Department of Neurology, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province 310058, China
| | - Ling Zhang
- Department of Neurobiology, School of Basic Medical Sciences, Key Laboratory of Medical Neurobiology (Ministry of Health of China), Key Laboratory of Neurobiology of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province 310058, China
| | - Mingkai Wang
- Department of Neurobiology, School of Basic Medical Sciences, Key Laboratory of Medical Neurobiology (Ministry of Health of China), Key Laboratory of Neurobiology of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province 310058, China
| | - Binggui Sun
- Department of Neurobiology, School of Basic Medical Sciences, Key Laboratory of Medical Neurobiology (Ministry of Health of China), Key Laboratory of Neurobiology of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province 310058, China.
| |
Collapse
|