1
|
Liu Y, Chen L, Lin L, Xu C, Xiong Y, Qiu H, Li X, Li S, Cao H. Unveiling the hidden pathways: Exploring astrocytes as a key target for depression therapy. J Psychiatr Res 2024; 174:101-113. [PMID: 38626560 DOI: 10.1016/j.jpsychires.2024.04.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Revised: 03/07/2024] [Accepted: 04/02/2024] [Indexed: 04/18/2024]
Abstract
Depressive disorders are widely debilitating psychiatric disease. Despite the considerable progress in the field of depression therapy, extensive research spanning many decades has failed to uncover pathogenic pathways that might aid in the creation of long-acting and rapid-acting antidepressants. Consequently, it is imperative to reconsider existing approaches and explore other targets to improve this area of study. In contemporary times, several scholarly investigations have unveiled that persons who have received a diagnosis of depression, as well as animal models employed to study depression, demonstrate a decrease in both the quantity as well as density of astrocytes, accompanied by alterations in gene expression and morphological attributes. Astrocytes rely on a diverse array of channels and receptors to facilitate their neurotransmitter transmission inside tripartite synapses. This study aimed to investigate the potential processes behind the development of depression, specifically focusing on astrocyte-associated neuroinflammation and the involvement of several molecular components such as connexin 43, potassium channel Kir4.1, aquaporin 4, glutamatergic aspartic acid transporter protein, SLC1A2 or GLT-1, glucocorticoid receptors, 5-hydroxytryptamine receptor 2B, and autophagy, that localized on the surface of astrocytes. The study also explores novel approaches in the treatment of depression, with a focus on astrocytes, offering innovative perspectives on potential antidepressant medications.
Collapse
Affiliation(s)
- Ying Liu
- Department of Psychiatry, The School of Clinical Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, 410208, China; Department of Psychiatry, Brain Hospital of Hunan Province (The Second People's Hospital of Hunan Province), Changsha, Hunan, 410007, China.
| | - Lu Chen
- Department of Gastroenterology, The School of Clinical Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, 410208, China; Department of Gastroenterology, Brain Hospital of Hunan Province (The Second People's Hospital of Hunan Province), Changsha, Hunan, 410007, China.
| | - Lin Lin
- Scientific Research Management Department, Brain Hospital of Hunan Province (The Second People's Hospital of Hunan Province), Changsha, Hunan, 410007, China.
| | - Caijuan Xu
- Department of Psychiatry, The School of Clinical Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, 410208, China; Department of Psychiatry, Brain Hospital of Hunan Province (The Second People's Hospital of Hunan Province), Changsha, Hunan, 410007, China.
| | - Yifan Xiong
- Department of Psychiatry, The School of Clinical Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, 410208, China; Department of Psychiatry, Brain Hospital of Hunan Province (The Second People's Hospital of Hunan Province), Changsha, Hunan, 410007, China.
| | - Huiwen Qiu
- Department of Psychiatry, The School of Clinical Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, 410208, China; Department of Psychiatry, Brain Hospital of Hunan Province (The Second People's Hospital of Hunan Province), Changsha, Hunan, 410007, China.
| | - Xinyu Li
- Department of Psychiatry, The School of Clinical Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, 410208, China; Department of Psychiatry, Brain Hospital of Hunan Province (The Second People's Hospital of Hunan Province), Changsha, Hunan, 410007, China.
| | - Sixin Li
- Department of Psychiatry, The School of Clinical Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, 410208, China; Department of Psychiatry, Brain Hospital of Hunan Province (The Second People's Hospital of Hunan Province), Changsha, Hunan, 410007, China.
| | - Hui Cao
- Department of Psychiatry, The School of Clinical Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, 410208, China; Department of Psychiatry, Brain Hospital of Hunan Province (The Second People's Hospital of Hunan Province), Changsha, Hunan, 410007, China.
| |
Collapse
|
2
|
Gonda S, Riedel C, Reiner A, Köhler I, Wahle P. Axons of cortical basket cells originating from dendrites develop higher local complexity than axons emerging from basket cell somata. Development 2023; 150:dev202305. [PMID: 37902086 PMCID: PMC10690106 DOI: 10.1242/dev.202305] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Accepted: 10/24/2023] [Indexed: 10/31/2023]
Abstract
Neuronal differentiation is regulated by neuronal activity. Here, we analyzed dendritic and axonal growth of Basket cells (BCs) and non-Basket cells (non-BCs) using sparse transfection of channelrhodopsin-YFP and repetitive optogenetic stimulation in slice cultures of rat visual cortex. Neocortical interneurons often display axon-carrying dendrites (AcDs). We found that the AcDs of BCs and non-BCs were, on average, the most complex dendrites. Further, the AcD configuration had an influence on BC axonal development. Axons originating from an AcD formed denser arborizations with more terminal endings within the dendritic field of the parent cell. Intriguingly, this occurred already in unstimulated BCs, and complexity was not increased further by optogenetic stimulation. However, optogenetic stimulation exerted a growth-promoting effect on axons emerging from BC somata. The axons of non-BCs neither responded to the AcD configuration nor to the optogenetic stimulation. The results suggest that the formation of locally dense BC plexuses is regulated by spontaneous activity. Moreover, in the AcD configuration, the AcD and the axon it carries mutually support each other's growth.
Collapse
Affiliation(s)
- Steffen Gonda
- Developmental Neurobiology, Faculty of Biology and Biotechnology, Ruhr University Bochum, 44801 Bochum, Germany
| | - Christian Riedel
- Developmental Neurobiology, Faculty of Biology and Biotechnology, Ruhr University Bochum, 44801 Bochum, Germany
| | - Andreas Reiner
- Cellular Neurobiology, Faculty of Biology and Biotechnology, Ruhr University Bochum, 44801 Bochum, Germany
| | - Ina Köhler
- Developmental Neurobiology, Faculty of Biology and Biotechnology, Ruhr University Bochum, 44801 Bochum, Germany
| | - Petra Wahle
- Developmental Neurobiology, Faculty of Biology and Biotechnology, Ruhr University Bochum, 44801 Bochum, Germany
| |
Collapse
|
3
|
Hearing Vocalizations during First Social Experience with Pups Increase Bdnf Transcription in Mouse Auditory Cortex. Neural Plast 2023; 2023:5225952. [PMID: 36845359 PMCID: PMC9946766 DOI: 10.1155/2023/5225952] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 12/30/2022] [Accepted: 02/01/2023] [Indexed: 02/17/2023] Open
Abstract
While infant cues are often assumed to innately motivate maternal response, recent research highlights how the neural coding of infant cues is altered through maternal care. Infant vocalizations are important social signals for caregivers, and evidence from mice suggests that experience caring for mouse pups induces inhibitory plasticity in the auditory cortex (AC), though the molecular mediators for such AC plasticity during the initial pup experience are not well delineated. Here, we used the maternal mouse communication model to explore whether transcription in AC of a specific, inhibition-linked, memory-associated gene, brain-derived neurotrophic factor (Bdnf) changes due to the very first pup caring experience hearing vocalizations, while controlling for the systemic influence of the hormone estrogen. Ovariectomized and estradiol or blank-implanted virgin female mice hearing pup calls with pups present had significantly higher AC exon IV Bdnf mRNA compared to females without pups present, suggesting that the social context of vocalizations induces immediate molecular changes at the site of auditory cortical processing. E2 influenced the rate of maternal behavior but did not significantly affect Bdnf mRNA transcription in the AC. To our knowledge, this is the first time Bdnf has been associated with processing social vocalizations in the AC, and our results suggest that it is a potential molecular component responsible for enhancing future recognition of infant cues by contributing to AC plasticity.
Collapse
|
4
|
Rodriguez LA, Kim SH, Page SC, Nguyen CV, Pattie EA, Hallock HL, Valerino J, Maynard KR, Jaffe AE, Martinowich K. The basolateral amygdala to lateral septum circuit is critical for regulating social novelty in mice. Neuropsychopharmacology 2023; 48:529-539. [PMID: 36369482 PMCID: PMC9852457 DOI: 10.1038/s41386-022-01487-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Revised: 10/07/2022] [Accepted: 10/24/2022] [Indexed: 11/13/2022]
Abstract
The lateral septum (LS) is a basal forebrain GABAergic region that is implicated in social novelty. However, the neural circuits and cell signaling pathways that converge on the LS to mediate social behaviors aren't well understood. Multiple lines of evidence suggest that signaling of brain-derived neurotrophic factor (BDNF) through its receptor TrkB plays important roles in social behavior. BDNF is not locally produced in LS, but we demonstrate that nearly all LS GABAergic neurons express TrkB. Local TrkB knock-down in LS neurons decreased social novelty recognition and reduced recruitment of neural activity in LS neurons in response to social novelty. Since BDNF is not synthesized in LS, we investigated which inputs to LS could serve as potential BDNF sources for controlling social novelty recognition. We demonstrate that selectively ablating inputs to LS from the basolateral amygdala (BLA), but not from ventral CA1 (vCA1), impairs social novelty recognition. Moreover, depleting BDNF selectively in BLA-LS projection neurons phenocopied the decrease in social novelty recognition caused by either local LS TrkB knockdown or ablation of BLA-LS inputs. These data support the hypothesis that BLA-LS projection neurons serve as a critical source of BDNF for activating TrkB signaling in LS neurons to control social novelty recognition.
Collapse
Affiliation(s)
- Lionel A Rodriguez
- Department of Neuroscience, Johns Hopkins School of Medicine, Baltimore, MD, 21205, USA
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, 21205, USA
| | - Sun-Hong Kim
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, 21205, USA
| | - Stephanie C Page
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, 21205, USA
| | - Claudia V Nguyen
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, 21205, USA
| | - Elizabeth A Pattie
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, 21205, USA
| | - Henry L Hallock
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, 21205, USA
| | - Jessica Valerino
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, 21205, USA
| | - Kristen R Maynard
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, 21205, USA
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine, Baltimore, MD, 21205, USA
| | - Andrew E Jaffe
- Department of Neuroscience, Johns Hopkins School of Medicine, Baltimore, MD, 21205, USA
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, 21205, USA
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine, Baltimore, MD, 21205, USA
- Department of Genetic Medicine, McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
- Center for Computational Biology, Johns Hopkins University, Baltimore, MD, 21205, USA
- Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, 21205, USA
- Department of Mental Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, 21205, USA
| | - Keri Martinowich
- Department of Neuroscience, Johns Hopkins School of Medicine, Baltimore, MD, 21205, USA.
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, 21205, USA.
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine, Baltimore, MD, 21205, USA.
- The Kavli Neuroscience Discovery Institute, Johns Hopkins University, Baltimore, USA.
| |
Collapse
|
5
|
Sharma V, Singh TG, Kaur A, Mannan A, Dhiman S. Brain-Derived Neurotrophic Factor: A Novel Dynamically Regulated Therapeutic Modulator in Neurological Disorders. Neurochem Res 2023; 48:317-339. [PMID: 36308619 DOI: 10.1007/s11064-022-03755-1] [Citation(s) in RCA: 21] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Revised: 09/02/2022] [Accepted: 09/03/2022] [Indexed: 02/04/2023]
Abstract
The growth factor brain-derived neurotrophic factor (BDNF), and its receptor tropomyosin-related kinase receptor type B (TrkB) play an active role in numerous areas of the adult brain, where they regulate the neuronal activity, function, and survival. Upregulation and downregulation of BDNF expression are critical for the physiology of neuronal circuits and functioning in the brain. Loss of BDNF function has been reported in the brains of patients with neurodegenerative or psychiatric disorders. This article reviews the BDNF gene structure, transport, secretion, expression and functions in the brain. This article also implicates BDNF in several brain-related disorders, including Alzheimer's disease, Parkinson's disease, Huntington's disease, amyotrophic lateral sclerosis, major depressive disorder, schizophrenia, epilepsy and bipolar disorder.
Collapse
Affiliation(s)
- Veerta Sharma
- Chitkara College of Pharmacy, Chitkara University, 140401, Rajpura, Punjab, India
| | - Thakur Gurjeet Singh
- Chitkara College of Pharmacy, Chitkara University, 140401, Rajpura, Punjab, India.
| | - Amarjot Kaur
- Chitkara College of Pharmacy, Chitkara University, 140401, Rajpura, Punjab, India
| | - Ashi Mannan
- Chitkara College of Pharmacy, Chitkara University, 140401, Rajpura, Punjab, India
| | - Sonia Dhiman
- Chitkara College of Pharmacy, Chitkara University, 140401, Rajpura, Punjab, India
| |
Collapse
|
6
|
Koizumi S. Glial Purinergic Signals and Psychiatric Disorders. Front Cell Neurosci 2022; 15:822614. [PMID: 35069121 PMCID: PMC8766327 DOI: 10.3389/fncel.2021.822614] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Accepted: 12/08/2021] [Indexed: 12/30/2022] Open
Abstract
Emotion-related neural networks are regulated in part by the activity of glial cells, and glial dysfunction can be directly related to emotional diseases such as depression. Here, we discuss three different therapeutic strategies involving astrocytes that are effective for treating depression. First, the antidepressant, fluoxetine, acts on astrocytes and increases exocytosis of ATP. This has therapeutic effects via brain-derived neurotrophic factor-dependent mechanisms. Second, electroconvulsive therapy is a well-known treatment for drug-resistant depression. Electroconvulsive therapy releases ATP from astrocytes to induce leukemia inhibitory factors and fibroblast growth factor 2, which leads to antidepressive actions. Finally, sleep deprivation therapy is well-known to cause antidepressive effects. Sleep deprivation also increases release of ATP, whose metabolite, adenosine, has antidepressive effects. These independent treatments share the same mechanism, i.e., ATP release from astrocytes, indicating an essential role of glial purinergic signals in the pathogenesis of depression.
Collapse
Affiliation(s)
- Schuichi Koizumi
- Department of Neuropharmacology, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Yamanashi, Japan
- GLIA Center, University of Yamanashi, Yamanashi, Japan
- *Correspondence: Schuichi Koizumi
| |
Collapse
|
7
|
Tomoda T, Sumitomo A, Shukla R, Hirota-Tsuyada Y, Miyachi H, Oh H, French L, Sibille E. BDNF controls GABA AR trafficking and related cognitive processes via autophagic regulation of p62. Neuropsychopharmacology 2022; 47:553-563. [PMID: 34341497 PMCID: PMC8674239 DOI: 10.1038/s41386-021-01116-0] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 07/14/2021] [Accepted: 07/18/2021] [Indexed: 02/07/2023]
Abstract
Reduced brain-derived neurotrophic factor (BDNF) and gamma-aminobutyric acid (GABA) neurotransmission co-occur in brain conditions (depression, schizophrenia and age-related disorders) and are associated with symptomatology. Rodent studies show they are causally linked, suggesting the presence of biological pathways mediating this link. Here we first show that reduced BDNF and GABA also co-occur with attenuated autophagy in human depression. Using mice, we then show that reducing Bdnf levels (Bdnf+/-) leads to upregulated sequestosome-1/p62, a key autophagy-associated adaptor protein, whose levels are inversely correlated with autophagic activity. Reduced Bdnf levels also caused reduced surface presentation of α5 subunit-containing GABAA receptor (α5-GABAAR) in prefrontal cortex (PFC) pyramidal neurons. Reducing p62 gene dosage restored α5-GABAAR surface expression and rescued PFC-relevant behavioral deficits of Bdnf+/- mice, including cognitive inflexibility and reduced sensorimotor gating. Increasing p62 levels was sufficient to recreate the molecular and behavioral profiles of Bdnf+/- mice. Collectively, the data reveal a novel mechanism by which deficient BDNF leads to targeted reduced GABAergic signaling through autophagic dysregulation of p62, potentially underlying cognitive impairment across brain conditions.
Collapse
Affiliation(s)
- Toshifumi Tomoda
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health (CAMH), Toronto, ON, Canada. .,Department of Research and Drug Discovery, Medical Innovation Center, Kyoto University Graduate School of Medicine, Kyoto, Japan.
| | - Akiko Sumitomo
- grid.155956.b0000 0000 8793 5925Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health (CAMH), Toronto, ON Canada ,grid.258799.80000 0004 0372 2033Department of Research and Drug Discovery, Medical Innovation Center, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Rammohan Shukla
- grid.155956.b0000 0000 8793 5925Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health (CAMH), Toronto, ON Canada ,grid.267337.40000 0001 2184 944XDepartment of Neurosciences, University of Toledo, Toledo, OH USA
| | - Yuki Hirota-Tsuyada
- grid.258799.80000 0004 0372 2033Department of Research and Drug Discovery, Medical Innovation Center, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Hitoshi Miyachi
- grid.258799.80000 0004 0372 2033Institute for Virus Research, Kyoto University, Kyoto, Japan
| | - Hyunjung Oh
- grid.155956.b0000 0000 8793 5925Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health (CAMH), Toronto, ON Canada
| | - Leon French
- grid.155956.b0000 0000 8793 5925Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health (CAMH), Toronto, ON Canada ,grid.17063.330000 0001 2157 2938Department of Psychiatry, University of Toronto, Toronto, ON Canada
| | - Etienne Sibille
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health (CAMH), Toronto, ON, Canada. .,Department of Psychiatry, University of Toronto, Toronto, ON, Canada. .,Department of Pharmacology and Toxicology, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
8
|
Molecular Basis of Late-Life Depression. Int J Mol Sci 2021; 22:ijms22147421. [PMID: 34299040 PMCID: PMC8303929 DOI: 10.3390/ijms22147421] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Revised: 07/05/2021] [Accepted: 07/08/2021] [Indexed: 12/14/2022] Open
Abstract
Late-life depression (LLD), compared to depression at a young age, is more likely to have poor prognosis and high risk of progression to dementia. A recent systemic review and meta-analysis of the present antidepressants for LLD showed that the treatment response rate was 48% and the remission rate was only 33.7%, thus implying the need to improve the treatment with other approaches in the future. Recently, agents modulating the glutamatergic system have been tested for mental disorders such as schizophrenia, dementia, and depressive disorder. Ketamine, a noncompetitive NMDA receptor (NMDAR) antagonist, requires more evidence from randomized clinical trials (RCTs) to prove its efficacy and safety in treating LLD. The metabotropic receptors (mGluRs) of the glutamatergic system are family G-protein-coupled receptors, and inhibition of the Group II mGluRs subtypes (mGlu2 and mGlu3) was found to be as effective as ketamine in exerting rapid antidepressant activity in some animal studies. Inflammation has been thought to contribute to depression for a long time. The cytokine levels not only increase with age but also decrease serotonin. Regarding LLD, interleukin 6 (IL-6) and tumor necrosis factor α (TNF-α) released in vivo are likely to contribute to the reduced serotonin level. Brain-derived neurotrophic factor (BDNF), a growth factor and a modulator in the tropomyosin receptor kinase (Trk) family of tyrosine kinase receptors, probably declines quantitatively with age. Recent studies suggest that BDNF/TrkB decrement may contribute to learning deficits and memory impairment. In the process of aging, physiological changes in combination with geriatric diseases such as vascular diseases result in poorer prognosis of LLD in comparison with that of young-age depression. Treatments with present antidepressants have been generally unsatisfactory. Novel treatments such as anti-inflammatory agents or NMDAR agonists/antagonists require more studies in LLD. Last but not least, LLD and dementia, which share common pathways and interrelate reciprocally, are a great concern. If it is possible to enhance the treatment of LDD, dementia can be prevented or delated.
Collapse
|
9
|
Guyon N, Zacharias LR, van Lunteren JA, Immenschuh J, Fuzik J, Märtin A, Xuan Y, Zilberter M, Kim H, Meletis K, Lopes-Aguiar C, Carlén M. Adult trkB Signaling in Parvalbumin Interneurons is Essential to Prefrontal Network Dynamics. J Neurosci 2021; 41:3120-3141. [PMID: 33593856 PMCID: PMC8026352 DOI: 10.1523/jneurosci.1848-20.2021] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Revised: 01/16/2021] [Accepted: 01/25/2021] [Indexed: 01/29/2023] Open
Abstract
Inhibitory interneurons expressing parvalbumin (PV) are central to cortical network dynamics, generation of γ oscillations, and cognition. Dysfunction of PV interneurons disrupts cortical information processing and cognitive behavior. Brain-derived neurotrophic factor (BDNF)/tyrosine receptor kinase B (trkB) signaling regulates the maturation of cortical PV interneurons but is also implicated in their adult multidimensional functions. Using a novel viral strategy for cell-type-specific and spatially restricted expression of a dominant-negative trkB (trkB.DN), we show that BDNF/trkB signaling is essential to the integrity and maintenance of prefrontal PV interneurons in adult male and female mice. Reduced BDNF/trkB signaling in PV interneurons in the medial prefrontal cortex (mPFC) resulted in deficient PV inhibition and increased baseline local field potential (LFP) activity in a broad frequency band. The altered network activity was particularly pronounced during increased activation of the prefrontal network and was associated with changed dynamics of local excitatory neurons, as well as decreased modulation of the LFP, abnormalities that appeared to generalize across stimuli and brain states. In addition, our findings link reduced BDNF/trkB signaling in prefrontal PV interneurons to increased aggression. Together our investigations demonstrate that BDNF/trkB signaling in PV interneurons in the adult mPFC is essential to local network dynamics and cognitive behavior. Our data provide direct support for the suggested association between decreased trkB signaling, deficient PV inhibition, and altered prefrontal circuitry.SIGNIFICANCE STATEMENT Brain-derived neurotrophic factor (BDNF)/tyrosine receptor kinase B (trkB) signaling promotes the maturation of inhibitory parvalbumin (PV) interneurons, neurons central to local cortical dynamics, γ rhythms, and cognition. Here, we used a novel viral approach for reduced BDNF/trkB signaling in PV interneurons in the medial prefrontal cortex (mPFC) to establish the role of BDNF/trkB signaling in adult prefrontal network activities. Reduced BDNF/trkB signaling caused pronounced morphologic alterations, reduced PV inhibition, and deficient prefrontal network dynamics. The altered network activity appeared to manifest across stimuli and brain states and was associated with aberrant local field potential (LFP) activities and increased aggression. The results demonstrate that adult BDNF/trkB signaling is essential to PV inhibition and prefrontal circuit function and directly links BDNF/trkB signaling to network integrity in the adult brain.
Collapse
Affiliation(s)
- Nicolas Guyon
- Department of Neuroscience, Karolinska Institutet, Stockholm 17177, Sweden
| | - Leonardo Rakauskas Zacharias
- Department of Neuroscience and Behavioral Sciences, Ribeirão Preto Medical School, Universidade de São Paulo, Ribeirão Preto 14049-900, Brazil
| | | | - Jana Immenschuh
- Department of Neuroscience, Karolinska Institutet, Stockholm 17177, Sweden
| | - Janos Fuzik
- Department of Neuroscience, Karolinska Institutet, Stockholm 17177, Sweden
| | - Antje Märtin
- Department of Neuroscience, Karolinska Institutet, Stockholm 17177, Sweden
| | - Yang Xuan
- Department of Neuroscience, Karolinska Institutet, Stockholm 17177, Sweden
| | - Misha Zilberter
- Department of Neuroscience, Karolinska Institutet, Stockholm 17177, Sweden
| | - Hoseok Kim
- Department of Neuroscience, Karolinska Institutet, Stockholm 17177, Sweden
| | | | - Cleiton Lopes-Aguiar
- Department of Physiology and Biophysics, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, Brazil
| | - Marie Carlén
- Department of Neuroscience, Karolinska Institutet, Stockholm 17177, Sweden
- Department of Biosciences and Nutrition, Karolinska Institutet, Huddinge 14183, Sweden
| |
Collapse
|
10
|
Griego E, Herrera-López G, Gómez-Lira G, Barrionuevo G, Gutiérrez R, Galván EJ. Functional expression of TrkB receptors on interneurones and pyramidal cells of area CA3 of the rat hippocampus. Neuropharmacology 2020; 182:108379. [PMID: 33130041 DOI: 10.1016/j.neuropharm.2020.108379] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Revised: 10/09/2020] [Accepted: 10/29/2020] [Indexed: 12/13/2022]
Abstract
The dentate gyrus and hippocampal area CA3 region of the mammalian brain contains the highest levels of brain-derived neurotrophic factor (BDNF) and its canonical membrane receptor, tropomyosin-related kinase B (TrkB). Therefore, the present study examines the expression and physiological responses triggered by activation of TrkB on hippocampal area CA3 interneurones and pyramidal cells of the rat hippocampus. Triple immunolabelling for TrkB, glutamate decarboxylase 67, and the calcium-binding proteins parvalbumin, calbindin or calretinin confirms the somatic expression of TrkB in all CA3 sublayers. TrkB-positive interneurones with fast-spiking discharge are restricted to strata oriens and lucidum, whereas regular-spiking interneurones are found in the strata lucidum, radiatum and lacunosum-moleculare. Activation of TrkB receptors with 7,8-dihydroxyflavone (DHF) modulates amplitude and frequency of spontaneous synaptic currents recorded from CA3 interneurones. Furthermore, the isolated excitatory postsynaptic currents (EPSC) of CA3 interneurones evoked by the mossy fibres (MF) or commissural/associational (C/A) axons, show input-specific synaptic potentiation in response to TrkB stimulation. On CA3 pyramidal cells, stimulation with DHF potentiates the MF synaptic transmission and increases the MF-EPSP - spike coupling. The latter exhibits a dramatic increase when picrotoxin is bath perfused after DHF, indicating that local interneurones restrain the excitability mediated by activation of TrkB. Therefore, we propose that release of BDNF on area CA3 reshapes the output of this hippocampal region by simultaneous activation of TrkB on GABAergic interneurones and pyramidal cells.
Collapse
Affiliation(s)
- Ernesto Griego
- Departamento de Farmacobiología, Cinvestav Sur, México City, México
| | | | | | - Germán Barrionuevo
- Department of Neuroscience, University of Pittsburgh, Pittsburgh, PA, 15260, United States
| | - Rafael Gutiérrez
- Departamento de Farmacobiología, Cinvestav Sur, México City, México
| | - Emilio J Galván
- Departamento de Farmacobiología, Cinvestav Sur, México City, México.
| |
Collapse
|
11
|
TrkB Signaling Influences Gene Expression in Cortistatin-Expressing Interneurons. eNeuro 2020; 7:ENEURO.0310-19.2019. [PMID: 31941661 PMCID: PMC7031852 DOI: 10.1523/eneuro.0310-19.2019] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Revised: 11/14/2019] [Accepted: 12/04/2019] [Indexed: 01/02/2023] Open
Abstract
Brain-derived neurotrophic factor (BDNF) signals through its cognate receptor tropomyosin receptor kinase B (TrkB) to promote the function of several classes of inhibitory interneurons. We previously reported that loss of BDNF-TrkB signaling in cortistatin (Cort)-expressing interneurons leads to behavioral hyperactivity and spontaneous seizures in mice. We performed bulk RNA sequencing (RNA-seq) from the cortex of mice with disruption of BDNF-TrkB signaling in cortistatin interneurons, and identified differential expression of genes important for excitatory neuron function. Using translating ribosome affinity purification and RNA-seq, we define a molecular profile for Cort-expressing inhibitory neurons and subsequently compare the translatome of normal and TrkB-depleted Cort neurons, revealing alterations in calcium signaling and axon development. Several of the genes enriched in Cort neurons and differentially expressed in TrkB-depleted neurons are also implicated in autism and epilepsy. Our findings highlight TrkB-dependent molecular pathways as critical for the maturation of inhibitory interneurons and support the hypothesis that loss of BDNF signaling in Cort interneurons leads to altered excitatory/inhibitory balance.
Collapse
|
12
|
Sex-specific spatial memory deficits in mice with a conditional TrkB deletion on parvalbumin interneurons. Behav Brain Res 2019; 372:111984. [DOI: 10.1016/j.bbr.2019.111984] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2019] [Revised: 05/23/2019] [Accepted: 05/27/2019] [Indexed: 12/12/2022]
|
13
|
Prenatal Androgenization Induces Anxiety-Like Behavior in Female Rats, Associated with Reduction of Inhibitory Interneurons and Increased BDNF in Hippocampus and Cortex. BIOMED RESEARCH INTERNATIONAL 2019; 2019:3426092. [PMID: 31281833 PMCID: PMC6590533 DOI: 10.1155/2019/3426092] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/05/2019] [Accepted: 04/16/2019] [Indexed: 12/22/2022]
Abstract
Anxiety is one of the most frequent psychiatric disorders. Despite the fact that most studies describe an anxiolytic effect of testosterone, hyperandrogenemia in mothers is assumed to be related to an increased risk of mood disorders in their offspring. An increasing body of scientific evidence suggests that an altered expression of interneuronal markers of the hippocampus may be the cause of anxiety. The aim of this study was to examine the influence of maternal hyperandrogenemia on behavioral parameters of anxiety-like behavior, neuropeptide Y (NPY) and parvalbumin (PV) expression in the hippocampus, and the level of the brain-derived neurotrophic factor (BDNF) in the hippocampus and cerebral cortex. Pregnant female Wistar albino rats were treated with testosterone undecanoate on the 20th day of gestation. Anxiety-like behavior in adult female offspring was evaluated by the elevated plus maze test and the open field. The number of PV and NPY immunoreactive cells in the hippocampus was determined immunohistochemically. The level of BDNF expression in the hippocampus and cerebral cortex was analyzed with the Western blot test. Prenatal hyperandrogenization increased anxiety-like behavior in female offspring and decreased expression of NPY+ and PV+ in the CA1 region of the hippocampus as compared to the control group. BDNF expression in the hippocampus and cerebral cortex of prenatally androgenized female offspring was significantly increased in comparison with the controls. Prenatal hyperandrogenization may be the cause of anxiety-like behavior in female offspring. Decrease in NPY and PV expression in the hippocampus may explain the possible mechanism of hyperandrogenization induced anxiety.
Collapse
|
14
|
The Role of Dendritic Brain-Derived Neurotrophic Factor Transcripts on Altered Inhibitory Circuitry in Depression. Biol Psychiatry 2019; 85:517-526. [PMID: 30449530 PMCID: PMC6380918 DOI: 10.1016/j.biopsych.2018.09.026] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2018] [Revised: 08/24/2018] [Accepted: 09/08/2018] [Indexed: 01/10/2023]
Abstract
BACKGROUND A parallel downregulation of brain-derived neurotrophic factor (BDNF) and somatostatin (SST), a marker of inhibitory gamma-aminobutyric acid interneurons that target pyramidal cell dendrites, has been reported in several brain areas of subjects with major depressive disorder (MDD). Rodent genetic studies suggest that they are linked and that both contribute to the illness. However, the mechanism by which they contribute to the pathophysiology of the illness has remained elusive. METHODS With quantitative polymerase chain reaction, we determined the expression level of BDNF transcript variants and synaptic markers in the prefrontal cortex of patients with MDD and matched control subjects (n = 19/group) and of C57BL/6J mice exposed to chronic stress or control conditions (n = 12/group). We next suppressed Bdnf transcripts with long 3' untranslated region (L-3'-UTR) using short hairpin RNA and investigated changes in cell morphology, gene expression, and behavior. RESULTS L-3'-UTRs containing BDNF messenger RNAs, which migrate to distal dendrites of pyramidal neurons, are selectively reduced, and their expression was highly correlated with SST expression in the prefrontal cortex of subjects with MDD. A similar downregulation occurs in mice submitted to chronic stress. We next show that Bdnf L-3'-UTR knockdown is sufficient to induce 1) dendritic shrinkage in cortical neurons, 2) cell-specific MDD-like gene changes (including Sst downregulation), and 3) depressive- and anxiety-like behaviors. The translational validity of the Bdnf L-3'-UTR short hairpin RNA-treated mice was confirmed by significant cross-species correlation of changes in MDD-associated gene expression. CONCLUSIONS These findings provide evidence for a novel MDD-related pathological mechanism linking local neurotrophic support, pyramidal cell structure, dendritic inhibition, and mood regulation.
Collapse
|
15
|
The primate-specific peptide Y-P30 regulates morphological maturation of neocortical dendritic spines. PLoS One 2019; 14:e0211151. [PMID: 30759095 PMCID: PMC6373909 DOI: 10.1371/journal.pone.0211151] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Accepted: 01/08/2019] [Indexed: 12/20/2022] Open
Abstract
The 30-amino acid peptide Y-P30 corresponds to the N-terminus of the primate-specific, sweat gland-derived dermcidin prepropeptide. Previous work has revealed that Y-P30 enhances the interaction of pleiotrophin and syndecans-2/3, and thus represents a natural ligand to study this signaling pathway. In immature neurons, Y-P30 activates the c-Src and p42/44 ERK kinase pathway, increases the amount of F-actin in axonal growth cones, and promotes neuronal survival, cell migration and axonal elongation. The action of Y-P30 on axonal growth requires syndecan-3 and heparan sulfate side chains. Whether Y-P30 has the potential to influence dendrites and dendritic protrusions has not been explored. The latter is suggested by the observations that syndecan-2 expression increases during postnatal development, that syndecan-2 becomes enriched in dendritic spines, and that overexpression of syndecan-2 in immature neurons results in a premature morphological maturation of dendritic spines. Here, analysing rat cortical pyramidal and non-pyramidal neurons in organotypic cultures, we show that Y-P30 does not alter the development of the dendritic arborization patterns. However, Y-P30 treatment decreases the density of apical, but not basal dendritic protrusions at the expense of the filopodia. Analysis of spine morphology revealed an unchanged mushroom/stubby-to-thin spine ratio and a shortening of the longest decile of dendritic protrusions. Whole-cell recordings from cortical principal neurons in dissociated cultures grown in the presence of Y-P30 demonstrated a decrease in the frequency of glutamatergic mEPSCs. Despite these differences in protrusion morphology and synaptic transmission, the latter likely attributable to presynaptic effects, calcium event rate and amplitude recorded in pyramidal neurons in organotypic cultures were not altered by Y-P30 treatment. Together, our data suggest that Y-P30 has the capacity to decelerate spinogenesis and to promote morphological, but not synaptic, maturation of dendritic protrusions.
Collapse
|
16
|
Hill JL, Jimenez DV, Mai Y, Ren M, Hallock HL, Maynard KR, Chen HY, Hardy NF, Schloesser RJ, Maher BJ, Yang F, Martinowich K. Cortistatin-expressing interneurons require TrkB signaling to suppress neural hyper-excitability. Brain Struct Funct 2018; 224:471-483. [PMID: 30377803 DOI: 10.1007/s00429-018-1783-1] [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] [Received: 02/09/2018] [Accepted: 10/21/2018] [Indexed: 01/12/2023]
Abstract
Signaling of brain-derived neurotrophic factor (BDNF) via tropomyosin receptor kinase B (TrkB) plays a critical role in the maturation of cortical inhibition and controls expression of inhibitory interneuron markers, including the neuropeptide cortistatin (CST). CST is expressed exclusively in a subset of cortical and hippocampal GABAergic interneurons, where it has anticonvulsant effects and controls sleep slow-wave activity (SWA). We hypothesized that CST-expressing interneurons play a critical role in regulating excitatory/inhibitory balance, and that BDNF, signaling through TrkB receptors on CST-expressing interneurons, is required for this function. Ablation of CST-expressing cells caused generalized seizures and premature death during early postnatal development, demonstrating a critical role for these cells in providing inhibition. Mice in which TrkB was selectively deleted from CST-expressing interneurons were hyperactive, slept less and developed spontaneous seizures. Frequencies of spontaneous excitatory post-synaptic currents (sEPSCs) on CST-expressing interneurons were attenuated in these mice. These data suggest that BDNF, signaling through TrkB receptors on CST-expressing cells, promotes excitatory drive onto these cells. Loss of excitatory drive onto CST-expressing cells that lack TrkB receptors may contribute to observed hyperexcitability and epileptogenesis.
Collapse
Affiliation(s)
- Julia L Hill
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, 855 North Wolfe Street, Suite 300, Baltimore, MD, 21205, USA
| | - Dennisse V Jimenez
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, 855 North Wolfe Street, Suite 300, Baltimore, MD, 21205, USA
| | - Yishan Mai
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, 855 North Wolfe Street, Suite 300, Baltimore, MD, 21205, USA
| | - Ming Ren
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, 855 North Wolfe Street, Suite 300, Baltimore, MD, 21205, USA
| | - Henry L Hallock
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, 855 North Wolfe Street, Suite 300, Baltimore, MD, 21205, USA
| | - Kristen R Maynard
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, 855 North Wolfe Street, Suite 300, Baltimore, MD, 21205, USA
| | - Huei-Ying Chen
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, 855 North Wolfe Street, Suite 300, Baltimore, MD, 21205, USA
| | - Nicholas F Hardy
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, 855 North Wolfe Street, Suite 300, Baltimore, MD, 21205, USA
| | | | - Brady J Maher
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, 855 North Wolfe Street, Suite 300, Baltimore, MD, 21205, USA.,Departments of Psychiatry and Behavioral Sciences, and Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Feng Yang
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, 855 North Wolfe Street, Suite 300, Baltimore, MD, 21205, USA
| | - Keri Martinowich
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, 855 North Wolfe Street, Suite 300, Baltimore, MD, 21205, USA. .,Departments of Psychiatry and Behavioral Sciences, and Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA.
| |
Collapse
|
17
|
Ohno Y, Kinboshi M, Shimizu S. Inwardly Rectifying Potassium Channel Kir4.1 as a Novel Modulator of BDNF Expression in Astrocytes. Int J Mol Sci 2018; 19:ijms19113313. [PMID: 30356026 PMCID: PMC6274740 DOI: 10.3390/ijms19113313] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2018] [Revised: 10/19/2018] [Accepted: 10/22/2018] [Indexed: 12/02/2022] Open
Abstract
Brain-derived neurotrophic factor (BDNF) is a key molecule essential for neural plasticity and development, and is implicated in the pathophysiology of various central nervous system (CNS) disorders. It is now documented that BDNF is synthesized not only in neurons, but also in astrocytes which actively regulate neuronal activities by forming tripartite synapses. Inwardly rectifying potassium (Kir) channel subunit Kir4.1, which is specifically expressed in astrocytes, constructs Kir4.1 and Kir4.1/5.1 channels, and mediates the spatial potassium (K+) buffering action of astrocytes. Recent evidence illustrates that Kir4.1 channels play important roles in bringing about the actions of antidepressant drugs and modulating BDNF expression in astrocytes. Although the precise mechanisms remain to be clarified, it seems likely that inhibition (down-regulation or blockade) of astrocytic Kir4.1 channels attenuates K+ buffering, increases neuronal excitability by elevating extracellular K+ and glutamate, and facilitates BDNF expression. Conversely, activation (up-regulation or opening) of Kir4.1 channels reduces neuronal excitability by lowering extracellular K+ and glutamate, and attenuates BDNF expression. Particularly, the former pathophysiological alterations seem to be important in epileptogenesis and pain sensitization, and the latter in the pathogenesis of depressive disorders. In this article, we review the functions of Kir4.1 channels, with a focus on their regulation of spatial K+ buffering and BDNF expression in astrocytes, and discuss the role of the astrocytic Kir4.1-BDNF system in modulating CNS disorders.
Collapse
Affiliation(s)
- Yukihiro Ohno
- Department of Pharmacology, Osaka University of Pharmaceutical Sciences, 4-20-1 Nasahara, Takatsuki, Osaka 569-1094, Japan.
| | - Masato Kinboshi
- Department of Pharmacology, Osaka University of Pharmaceutical Sciences, 4-20-1 Nasahara, Takatsuki, Osaka 569-1094, Japan.
| | - Saki Shimizu
- Department of Pharmacology, Osaka University of Pharmaceutical Sciences, 4-20-1 Nasahara, Takatsuki, Osaka 569-1094, Japan.
| |
Collapse
|
18
|
Postnatal TrkB ablation in corticolimbic interneurons induces social dominance in male mice. Proc Natl Acad Sci U S A 2018; 115:E9909-E9915. [PMID: 30282736 DOI: 10.1073/pnas.1812083115] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
The tight balance between synaptic excitation and inhibition (E/I) within neocortical circuits in the mammalian brain is important for complex behavior. Many loss-of-function studies have demonstrated that brain-derived neurotrophic factor (BDNF) and its cognate receptor tropomyosin receptor kinase B (TrkB) are essential for the development of inhibitory GABAergic neurons. However, behavioral consequences of impaired BDNF/TrkB signaling in GABAergic neurons remain unclear, largely due to confounding motor function deficits observed in previous animal models. In this study, we generated conditional knockout mice (TrkB cKO) in which TrkB was ablated from a majority of corticolimbic GABAergic interneurons postnatally. These mice showed intact motor coordination and movement, but exhibited enhanced dominance over other mice in a group-housed setting. In addition, immature fast-spiking GABAergic neurons of TrkB cKO mice resulted in an E/I imbalance in layer 5 microcircuits within the medial prefrontal cortex (mPFC), a key region regulating social dominance. Restoring the E/I imbalance via optogenetic modulation in the mPFC of TrkB cKO mice normalized their social dominance behavior. Taken together, our results provide strong evidence for a role of BDNF/TrkB signaling in inhibitory synaptic modulation and social dominance behavior in mice.
Collapse
|
19
|
Marathe SV, D'almeida PL, Virmani G, Bathini P, Alberi L. Effects of Monoamines and Antidepressants on Astrocyte Physiology: Implications for Monoamine Hypothesis of Depression. J Exp Neurosci 2018; 12:1179069518789149. [PMID: 30046253 PMCID: PMC6056786 DOI: 10.1177/1179069518789149] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2018] [Accepted: 06/19/2018] [Indexed: 01/17/2023] Open
Abstract
Major depressive disorder (MDD) is one of the most common neuropsychiatric
disorders affecting over one-fifth of the population worldwide. Owing to our
limited understanding of the pathophysiology of MDD, the quest for finding novel
antidepressant drug targets is severely impeded. Monoamine hypothesis of MDD
provides a robust theoretical framework, forming the core of a large jigsaw
puzzle, around which we must look for the vital missing pieces. Growing evidence
suggests that the glial loss observed in key regions of the limbic system in
depressed patients, at least partly, accounts for the structural and cognitive
manifestations of MDD. Studies in animal models have subsequently hinted at the
possibility that the glial atrophy may play a causative role in the
precipitation of depressive symptoms. Antidepressants as well as monoamine
neurotransmitters exert profound effects on the gene expression and metabolism
in astrocytes. This raises an intriguing possibility that the astrocytes may
play a central role alongside neurons in the behavioral effects of
antidepressant drugs. In this article, we discuss the gene expression and
metabolic changes brought about by antidepressants in astrocytes, which could be
of relevance to synaptic plasticity and behavioral effects of antidepressant
treatments.
Collapse
Affiliation(s)
| | | | - Garima Virmani
- Centre for Neuroscience, Indian Institute of Science, Bangalore, India
| | - Praveen Bathini
- Department of Medicine University of Fribourg, Fribourg, Switzerland.,Swiss Integrative Center for Human Health SA (SICHH), Fribourg, Switzerland
| | - Lavinia Alberi
- Department of Medicine University of Fribourg, Fribourg, Switzerland.,Swiss Integrative Center for Human Health SA (SICHH), Fribourg, Switzerland
| |
Collapse
|
20
|
Hing B, Sathyaputri L, Potash JB. A comprehensive review of genetic and epigenetic mechanisms that regulate BDNF expression and function with relevance to major depressive disorder. Am J Med Genet B Neuropsychiatr Genet 2018; 177:143-167. [PMID: 29243873 DOI: 10.1002/ajmg.b.32616] [Citation(s) in RCA: 83] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2017] [Accepted: 11/21/2017] [Indexed: 12/11/2022]
Abstract
Major depressive disorder (MDD) is a mood disorder that affects behavior and impairs cognition. A gene potentially important to this disorder is the brain derived neurotrophic factor (BDNF) as it is involved in processes controlling neuroplasticity. Various mechanisms exist to regulate BDNF's expression level, subcellular localization, and sorting to appropriate secretory pathways. Alterations to these processes by genetic factors and negative stressors can dysregulate its expression, with possible implications for MDD. Here, we review the mechanisms governing the regulation of BDNF expression, and discuss how disease-associated single nucleotide polymorphisms (SNPs) can alter these mechanisms, and influence MDD. As negative stressors increase the likelihood of MDD, we will also discuss the impact of these stressors on BDNF expression, the cellular effect of such a change, and its impact on behavior in animal models of stress. We will also describe epigenetic processes that mediate this change in BDNF expression. Similarities in BDNF expression between animal models of stress and those in MDD will be highlighted. We will also contrast epigenetic patterns at the BDNF locus between animal models of stress, and MDD patients, and address limitations to current clinical studies. Future work should focus on validating current genetic and epigenetic findings in tightly controlled clinical studies. Regions outside of BDNF promoters should also be explored, as should other epigenetic marks, to improve identification of biomarkers for MDD.
Collapse
Affiliation(s)
- Benjamin Hing
- Department of Psychiatry, Carver College of Medicine, University of Iowa, Iowa City, Iowa
| | - Leela Sathyaputri
- Department of Psychiatry, Carver College of Medicine, University of Iowa, Iowa City, Iowa
| | - James B Potash
- Department of Psychiatry, Carver College of Medicine, University of Iowa, Iowa City, Iowa
| |
Collapse
|
21
|
Höfflin F, Jack A, Riedel C, Mack-Bucher J, Roos J, Corcelli C, Schultz C, Wahle P, Engelhardt M. Heterogeneity of the Axon Initial Segment in Interneurons and Pyramidal Cells of Rodent Visual Cortex. Front Cell Neurosci 2017; 11:332. [PMID: 29170630 PMCID: PMC5684645 DOI: 10.3389/fncel.2017.00332] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2017] [Accepted: 10/09/2017] [Indexed: 11/13/2022] Open
Abstract
The microdomain that orchestrates action potential initiation in neurons is the axon initial segment (AIS). It has long been considered to be a rather homogeneous domain at the very proximal axon hillock with relatively stable length, particularly in cortical pyramidal cells. However, studies in other brain regions paint a different picture. In hippocampal CA1, up to 50% of axons emerge from basal dendrites. Further, in about 30% of thick-tufted layer V pyramidal neurons in rat somatosensory cortex, axons have a dendritic origin. Consequently, the AIS is separated from the soma. Recent in vitro and in vivo studies have shown that cellular excitability is a function of AIS length/position and somatodendritic morphology, undermining a potentially significant impact of AIS heterogeneity for neuronal function. We therefore investigated neocortical axon morphology and AIS composition, hypothesizing that the initial observation of seemingly homogeneous AIS is inadequate and needs to take into account neuronal cell types. Here, we biolistically transfected cortical neurons in organotypic cultures to visualize the entire neuron and classify cell types in combination with immunolabeling against AIS markers. Using confocal microscopy and morphometric analysis, we investigated axon origin, AIS position, length, diameter as well as distance to the soma. We find a substantial AIS heterogeneity in visual cortical neurons, classified into three groups: (I) axons with somatic origin with proximal AIS at the axon hillock; (II) axons with somatic origin with distal AIS, with a discernible gap between the AIS and the soma; and (III) axons with dendritic origin (axon-carrying dendrite cell, AcD cell) and an AIS either starting directly at the axon origin or more distal to that point. Pyramidal cells have significantly longer AIS than interneurons. Interneurons with vertical columnar axonal projections have significantly more distal AIS locations than all other cells with their prevailing phenotype as an AcD cell. In contrast, neurons with perisomatic terminations display most often an axon originating from the soma. Our data contribute to the emerging understanding that AIS morphology is highly variable, and potentially a function of the cell type.
Collapse
Affiliation(s)
- Felix Höfflin
- Institute of Neuroanatomy, Medical Faculty Mannheim, Center for Biomedicine and Medical Technology Mannheim (CBTM), Heidelberg University, Heidelberg, Germany
| | - Alexander Jack
- Developmental Neurobiology, Department of Zoology and Neurobiology, Ruhr-University Bochum, Bochum, Germany
| | - Christian Riedel
- Developmental Neurobiology, Department of Zoology and Neurobiology, Ruhr-University Bochum, Bochum, Germany
| | - Julia Mack-Bucher
- Live Cell Imaging Core Mannheim (LIMA), Medical Faculty Mannheim, Center for Biomedicine and Medical Technology Mannheim (CBTM), Heidelberg University, Heidelberg, Germany
| | - Johannes Roos
- Institute of Neuroanatomy, Medical Faculty Mannheim, Center for Biomedicine and Medical Technology Mannheim (CBTM), Heidelberg University, Heidelberg, Germany
| | - Corinna Corcelli
- Institute of Neuroanatomy, Medical Faculty Mannheim, Center for Biomedicine and Medical Technology Mannheim (CBTM), Heidelberg University, Heidelberg, Germany
| | - Christian Schultz
- Institute of Neuroanatomy, Medical Faculty Mannheim, Center for Biomedicine and Medical Technology Mannheim (CBTM), Heidelberg University, Heidelberg, Germany
| | - Petra Wahle
- Developmental Neurobiology, Department of Zoology and Neurobiology, Ruhr-University Bochum, Bochum, Germany
| | - Maren Engelhardt
- Institute of Neuroanatomy, Medical Faculty Mannheim, Center for Biomedicine and Medical Technology Mannheim (CBTM), Heidelberg University, Heidelberg, Germany
| |
Collapse
|
22
|
Leukemia inhibitory factor impairs structural and neurochemical development of rat visual cortex in vivo. Mol Cell Neurosci 2017; 79:81-92. [PMID: 28088609 DOI: 10.1016/j.mcn.2016.12.008] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2016] [Revised: 11/25/2016] [Accepted: 12/29/2016] [Indexed: 11/23/2022] Open
Abstract
Minipump infusions into visual cortex in vivo at the onset of the critical period have revealed that the proinflammatory cytokine leukemia inhibitory factor (LIF) delays the maturation of thalamocortical projection neurons of the lateral geniculate nucleus, and tecto-thalamic projection neurons of the superior colliculus, and cortical layer IV spiny stellates and layer VI pyramidal neurons. Here, we report that P12-20 LIF infusion inhibits somatic maturation of pyramidal neurons and of all interneuron types in vivo. Likewise, DIV 12-20 LIF treatment in organotypic cultures prevents somatic growth GABA-ergic neurons. Further, while NPY expression is increased in the LIF-infused hemispheres, the expression of parvalbumin mRNA and protein, Kv3.1 mRNA, calbindin D-28k protein, and GAD-65 mRNA, but not of GAD-67 mRNA or calretinin protein is substantially reduced. Also, LIF treatment decreases parvalbumin, Kv3.1, Kv3.2 and GAD-65, but not GAD-67 mRNA expression in OTC. Developing cortical neurons are known to depend on neurotrophins. Indeed, LIF alters neurotrophin mRNA expression, and prevents the growth promoting action of neurotophin-4 in GABA-ergic neurons. The results imply that LIF, by altering neurotrophin expression and/or signaling, could counteract neurotrophin-dependent growth and neurochemical differentiation of cortical neurons.
Collapse
|
23
|
Oh H, Lewis DA, Sibille E. The Role of BDNF in Age-Dependent Changes of Excitatory and Inhibitory Synaptic Markers in the Human Prefrontal Cortex. Neuropsychopharmacology 2016; 41:3080-3091. [PMID: 27417517 PMCID: PMC5101556 DOI: 10.1038/npp.2016.126] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2015] [Revised: 06/10/2016] [Accepted: 07/04/2016] [Indexed: 01/07/2023]
Abstract
Reduced brain-derived neurotrophic factor (BDNF) may underlie age-related synaptic loss, in turn contributing to cerebral atrophy, cognitive decline, and increased risk for psychiatric disorders. However, the specific contribution of BDNF to the age-related expression changes in synaptic markers and their temporal trajectories remain uncharacterized. Using microarray data from orbitofrontal cortex of control subjects (n=209; 16-96 years), we identified genes whose expression positively correlates with BDNF (r>0.575; n=200 genes) and analyzed them for enriched biological pathways. qPCR was performed to measure the expression level of transcript variants of BDNF, NTRK2, and selected BDNF-coexpressed genes in younger and older subjects. We confirmed age-related downregulation of BDNF and show 78 of the top 200 BDNF-coexpressed genes are associated with synaptic function. Both excitatory and inhibitory synaptic genes show decreased expression with age and are positively correlated with BDNF and NTRK2 expression and negatively correlated with dominant-negative truncated NTRK2 level. Results were validated at the RNA level in an independent cohort and at the protein level for selected findings. We next tested the causal link between the correlative human findings using mice with conditional blockade of BDNF/NTRK2 signaling. Blockade of NTRK2 activity in adult mice recapitulate the age-like pattern in the expression of markers for inhibitory presynaptic but notably not for excitatory synaptic genes. Together, these findings suggest that age-dependent decrease in BDNF signaling may cause synaptic alterations through an initial and preferential effect on GABA presynaptic genes. These results have implications for neuropsychiatric disorders characterized by accelerated aging molecular profiles, such as major depression.
Collapse
Affiliation(s)
- Hyunjung Oh
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health (CAMH), Toronto, Ontario, Canada
| | - David A Lewis
- Department of Psychiatry, Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA, USA
| | - Etienne Sibille
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health (CAMH), Toronto, Ontario, Canada,Department of Psychiatry, Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA, USA,Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada,Department of Pharmacology and Toxicology, University of Toronto, Toronto, Ontario, Canada,Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health (CAMH), 250 College Street, Room 134, Toronto, Ontario M5T 1R8, Canada, Tel: +1 416 535 8501, E-mail:
| |
Collapse
|
24
|
Benítez-Temiño B, Davis-López de Carrizosa MA, Morcuende S, Matarredona ER, de la Cruz RR, Pastor AM. Functional Diversity of Neurotrophin Actions on the Oculomotor System. Int J Mol Sci 2016; 17:E2016. [PMID: 27916956 PMCID: PMC5187816 DOI: 10.3390/ijms17122016] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2016] [Revised: 11/24/2016] [Accepted: 11/25/2016] [Indexed: 11/16/2022] Open
Abstract
Neurotrophins play a principal role in neuronal survival and differentiation during development, but also in the maintenance of appropriate adult neuronal circuits and phenotypes. In the oculomotor system, we have demonstrated that neurotrophins are key regulators of developing and adult neuronal properties, but with peculiarities depending on each neurotrophin. For instance, the administration of NGF (nerve growth factor), BDNF (brain-derived neurotrophic factor) or NT-3 (neurotrophin-3) protects neonatal extraocular motoneurons from cell death after axotomy, but only NGF and BDNF prevent the downregulation in ChAT (choline acetyltransferase). In the adult, in vivo recordings of axotomized extraocular motoneurons have demonstrated that the delivery of NGF, BDNF or NT-3 recovers different components of the firing discharge activity of these cells, with some particularities in the case of NGF. All neurotrophins have also synaptotrophic activity, although to different degrees. Accordingly, neurotrophins can restore the axotomy-induced alterations acting selectively on different properties of the motoneuron. In this review, we summarize these evidences and discuss them in the context of other motor systems.
Collapse
Affiliation(s)
- Beatriz Benítez-Temiño
- Departamento de Fisiología, Facultad de Biología, Universidad de Sevilla, 41012 Sevilla, Spain.
| | | | - Sara Morcuende
- Departamento de Fisiología, Facultad de Biología, Universidad de Sevilla, 41012 Sevilla, Spain.
| | - Esperanza R Matarredona
- Departamento de Fisiología, Facultad de Biología, Universidad de Sevilla, 41012 Sevilla, Spain.
| | - Rosa R de la Cruz
- Departamento de Fisiología, Facultad de Biología, Universidad de Sevilla, 41012 Sevilla, Spain.
| | - Angel M Pastor
- Departamento de Fisiología, Facultad de Biología, Universidad de Sevilla, 41012 Sevilla, Spain.
| |
Collapse
|
25
|
Castillo-Gómez E, Pérez-Rando M, Vidueira S, Nacher J. Polysialic Acid Acute Depletion Induces Structural Plasticity in Interneurons and Impairs the Excitation/Inhibition Balance in Medial Prefrontal Cortex Organotypic Cultures. Front Cell Neurosci 2016; 10:170. [PMID: 27445697 PMCID: PMC4925659 DOI: 10.3389/fncel.2016.00170] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2016] [Accepted: 06/10/2016] [Indexed: 01/08/2023] Open
Abstract
The structure and function of the medial prefrontal cortex (mPFC) is affected in several neuropsychiatric disorders, including schizophrenia and major depression. Recent studies suggest that imbalances between excitatory and inhibitory activity (E/I) may be responsible for this cortical dysfunction and therefore, may underlie the core symptoms of these diseases. This E/I imbalance seems to be correlated with alterations in the plasticity of interneurons but there is still scarce information on the mechanisms that may link these phenomena. The polysialylated form of the neural cell adhesion molecule (PSA-NCAM) is a good candidate, because it modulates the neuronal plasticity of interneurons and its expression is altered in schizophrenia and major depression. To address this question, we have developed an in vitro model using mPFC organotypic cultures of transgenic mice displaying fluorescent spiny interneurons. After enzymatic depletion of PSA, the spine density of interneurons, the number of synaptic puncta surrounding pyramidal neuron somata and the E/I ratio were strongly affected. These results point to the polysialylation of NCAM as an important factor in the maintenance of E/I balance and the structural plasticity of interneurons. This may be particularly relevant for better understanding the etiology of schizophrenia and major depression.
Collapse
Affiliation(s)
- Esther Castillo-Gómez
- Neurobiology Unit/BIOTECMED, Cell Biology Department, Universitat de ValènciaValencia, Spain; Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM): Spanish National Network for Research in Mental HealthMadrid, Spain
| | - Marta Pérez-Rando
- Neurobiology Unit/BIOTECMED, Cell Biology Department, Universitat de València Valencia, Spain
| | - Sandra Vidueira
- Neurobiology Unit/BIOTECMED, Cell Biology Department, Universitat de València Valencia, Spain
| | - Juan Nacher
- Neurobiology Unit/BIOTECMED, Cell Biology Department, Universitat de ValènciaValencia, Spain; Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM): Spanish National Network for Research in Mental HealthMadrid, Spain; Fundación Investigación Hospital Clínico de Valencia, INCLIVAValencia, Spain
| |
Collapse
|
26
|
BDNF-induced presynaptic facilitation of GABAergic transmission in the hippocampus of young adults is dependent of TrkB and adenosine A2A receptors. Purinergic Signal 2016; 12:283-94. [PMID: 26897393 DOI: 10.1007/s11302-016-9502-y] [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: 11/25/2015] [Accepted: 02/10/2016] [Indexed: 01/03/2023] Open
Abstract
Brain-derived neurotrophic factor (BDNF) and adenosine are widely recognized as neuromodulators of glutamatergic transmission in the adult brain. Most BDNF actions upon excitatory plasticity phenomena are under control of adenosine A2A receptors (A2ARs). Concerning gamma-aminobutyric acid (GABA)-mediated transmission, the available information refers to the control of GABA transporters. We now focused on the influence of BDNF and the interplay with adenosine on phasic GABAergic transmission. To assess this, we evaluated evoked and spontaneous synaptic currents recorded from CA1 pyramidal cells in acute hippocampal slices from adult rat brains (6 to 10 weeks old). BDNF (10-100 ng/mL) increased miniature inhibitory postsynaptic current (mIPSC) frequency, but not amplitude, as well as increased the amplitude of inhibitory postsynaptic currents (IPSCs) evoked by afferent stimulation. The facilitatory action of BDNF upon GABAergic transmission was lost in the presence of a Trk inhibitor (K252a, 200 nM), but not upon p75(NTR) blockade (anti-p75(NTR) IgG, 50 μg/mL). Moreover, the facilitatory action of BDNF onto GABAergic transmission was also prevented upon A2AR antagonism (SCH 58261, 50 nM). We conclude that BDNF facilitates GABAergic signaling at the adult hippocampus via a presynaptic mechanism that depends on TrkB and adenosine A2AR activation.
Collapse
|
27
|
Hill JL, Martinowich K. Activity-dependent signaling: influence on plasticity in circuits controlling fear-related behavior. Curr Opin Neurobiol 2016; 36:59-65. [PMID: 26485574 PMCID: PMC4738053 DOI: 10.1016/j.conb.2015.10.001] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2015] [Revised: 09/30/2015] [Accepted: 10/01/2015] [Indexed: 10/22/2022]
Abstract
Fear regulation is impaired in anxiety and trauma-related disorders. Patients experience heightened fear expression and reduced ability to extinguish fear memories. Because fear regulation is abnormal in these disorders and extinction recapitulates current treatment strategies, understanding the underlying mechanisms is vital for developing new treatments. This is critical because although extinction-based exposure therapy is a mainstay of treatment, relapse is common. We examine recent findings describing changes in network activity and functional connectivity within limbic circuits during fear regulation, and explore how activity-dependent signaling contributes to the neural activity patterns that control fear and anxiety. We review the role of the prototypical activity-dependent molecule, brain-derived neurotrophic factor (BDNF), whose signaling has been critically linked to regulation of fear behavior.
Collapse
Affiliation(s)
- Julia L Hill
- Lieber Institute for Brain Development, Baltimore, MD 21205, United States
| | - Keri Martinowich
- Lieber Institute for Brain Development, Baltimore, MD 21205, United States; Department of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine, Baltimore, MD 21205, United States.
| |
Collapse
|
28
|
Purves-Tyson TD, Allen K, Fung S, Rothmond D, Noble PL, Handelsman DJ, Shannon Weickert C. Adolescent testosterone influences BDNF and TrkB mRNA and neurotrophin-interneuron marker relationships in mammalian frontal cortex. Schizophr Res 2015; 168:661-70. [PMID: 26088421 DOI: 10.1016/j.schres.2015.05.040] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2015] [Revised: 05/11/2015] [Accepted: 05/28/2015] [Indexed: 11/24/2022]
Abstract
Late adolescence in males is a period of increased susceptibility for the onset of schizophrenia, coinciding with increased circulating testosterone. The cognitive deficits prevalent in schizophrenia may be related to unhealthy cortical interneurons, which are trophically dependent on brain derived neurotrophic factor. We investigated, under conditions of depleted (monkey and rat) and replaced (rat) testosterone over adolescence, changes in gene expression of cortical BDNF and TrkB transcripts and interneuron markers and the relationships between these mRNAs and circulating testosterone. Testosterone removal by gonadectomy reduced gene expression of some BDNF transcripts in monkey and rat frontal cortices and the BDNF mRNA reduction was prevented by testosterone replacement. In rat, testosterone replacement increased the potential for classical TrkB signalling by increasing the full length to truncated TrkB mRNA ratio, whereas in the monkey cortex, circulating testosterone was negatively correlated with the TrkB full length/truncated mRNA ratio. We did not identify changes in interneuron gene expression in monkey frontal cortex in response to gonadectomy, and in rat, we showed that only somatostatin mRNA was decreased by gonadectomy but not restored by testosterone replacement. We identified complex and possibly species-specific, relationships between BDNF/TrkB gene expression and interneuron marker gene expression that appear to be dependent on the presence of testosterone at adolescence in rat and monkey frontal cortices. Taken together, our findings suggest there are dynamic relationships between BDNF/TrkB and interneuron markers that are dependent on the presence of testosterone but that this may not be a straightforward increase in testosterone leading to changes in BDNF/TrkB that contributes to interneuron health.
Collapse
Affiliation(s)
- Tertia D Purves-Tyson
- Schizophrenia Research Institute, Sydney 2021, Australia; Neuroscience Research Australia, Sydney 2031, Australia; School of Medical Sciences, University of New South Wales, Sydney 2031, Australia
| | - Katherine Allen
- Schizophrenia Research Institute, Sydney 2021, Australia; Neuroscience Research Australia, Sydney 2031, Australia; School of Psychiatry, University of New South Wales, Sydney 2031, Australia
| | - Samantha Fung
- Schizophrenia Research Institute, Sydney 2021, Australia; Neuroscience Research Australia, Sydney 2031, Australia; School of Psychiatry, University of New South Wales, Sydney 2031, Australia
| | - Debora Rothmond
- Schizophrenia Research Institute, Sydney 2021, Australia; Neuroscience Research Australia, Sydney 2031, Australia
| | | | | | - Cynthia Shannon Weickert
- Schizophrenia Research Institute, Sydney 2021, Australia; Neuroscience Research Australia, Sydney 2031, Australia; School of Psychiatry, University of New South Wales, Sydney 2031, Australia
| |
Collapse
|
29
|
Hanno-Iijima Y, Tanaka M, Iijima T. Activity-Dependent Bidirectional Regulation of GAD Expression in a Homeostatic Fashion Is Mediated by BDNF-Dependent and Independent Pathways. PLoS One 2015; 10:e0134296. [PMID: 26241953 PMCID: PMC4524701 DOI: 10.1371/journal.pone.0134296] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2015] [Accepted: 07/07/2015] [Indexed: 11/23/2022] Open
Abstract
Homeostatic synaptic plasticity, or synaptic scaling, is a mechanism that tunes neuronal transmission to compensate for prolonged, excessive changes in neuronal activity. Both excitatory and inhibitory neurons undergo homeostatic changes based on synaptic transmission strength, which could effectively contribute to a fine-tuning of circuit activity. However, gene regulation that underlies homeostatic synaptic plasticity in GABAergic (GABA, gamma aminobutyric) neurons is still poorly understood. The present study demonstrated activity-dependent dynamic scaling in which NMDA-R (N-methyl-D-aspartic acid receptor) activity regulated the expression of GABA synthetic enzymes: glutamic acid decarboxylase 65 and 67 (GAD65 and GAD67). Results revealed that activity-regulated BDNF (brain-derived neurotrophic factor) release is necessary, but not sufficient, for activity-dependent up-scaling of these GAD isoforms. Bidirectional forms of activity-dependent GAD expression require both BDNF-dependent and BDNF-independent pathways, both triggered by NMDA-R activity. Additional results indicated that these two GAD genes differ in their responsiveness to chronic changes in neuronal activity, which could be partially caused by differential dependence on BDNF. In parallel to activity-dependent bidirectional scaling in GAD expression, the present study further observed that a chronic change in neuronal activity leads to an alteration in neurotransmitter release from GABAergic neurons in a homeostatic, bidirectional fashion. Therefore, the differential expression of GAD65 and 67 during prolonged changes in neuronal activity may be implicated in some aspects of bidirectional homeostatic plasticity within mature GABAergic presynapses.
Collapse
Affiliation(s)
- Yoko Hanno-Iijima
- Tokai University Institute of Innovative Science and Technology, Medical Division, Kanagawa, Japan
- School of Medicine, Tokai University, Kanagawa, Japan
| | - Masami Tanaka
- Tokai University Institute of Innovative Science and Technology, Medical Division, Kanagawa, Japan
- School of Medicine, Tokai University, Kanagawa, Japan
| | - Takatoshi Iijima
- Tokai University Institute of Innovative Science and Technology, Medical Division, Kanagawa, Japan
- School of Medicine, Tokai University, Kanagawa, Japan
- * E-mail:
| |
Collapse
|
30
|
Pickering C, Alsiö J, Morud J, Ericson M, Robbins TW, Söderpalm B. Ethanol impairment of spontaneous alternation behaviour and associated changes in medial prefrontal glutamatergic gene expression precede putative markers of dependence. Pharmacol Biochem Behav 2015; 132:63-70. [DOI: 10.1016/j.pbb.2015.02.021] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2014] [Revised: 02/17/2015] [Accepted: 02/21/2015] [Indexed: 12/21/2022]
|
31
|
Abstract
The endogenous cannabinoid (endocannabinoid) system is an important regulator of synaptic function. Endocannabinoids acutely modulate inhibitory and excitatory transmission, and also mediate long-term depression at GABAergic and glutamatergic synapses. Typically, endocannabinoid synthesis and release is stimulated by depolarization-induced calcium influx and/or activation of phospholipase-C (PLC) signaling triggered by mGluR activation. Recently it has been shown that brain-derived neurotrophic factor (BDNF) can also induce endocannabinoid release. Although there is growing evidence for cross-talk between BDNF and endocannabinoid signaling, little is known about the functional relevance of these interactions. In the present studies, we examined BDNF - endocannabinoid interactions in regulating activity-dependent long-term depression at inhibitory synapses (iLTD). We found that theta burst stimulation (TBS) in layer 2/3 of mouse somatosensory cortical slices can induce a form of endocannabinoid-mediated iLTD that is independent of metabotropic glutamate receptor (mGluR) activation. This endocannabinoid-dependent iLTD, however, requires endogenous BDNF-trkB signaling, as it is blocked by a trk tyrosine kinase inhibitor and by a trkB receptor antagonist, and also requires activation of diacylglycerol lipase (DAG-lipase, DGL). In addition, endocannabinoid-mediated iLTD can be induced by combining a subthreshold concentration of exogenous BDNF with weak TBS stimulation that by itself is insufficient to induce iLTD. Taken together, our results suggest that TBS can induce the release of endogenous BDNF, which triggers DGL-dependent endocannabinoid mobilization and cannabinoid receptor-dependent iLTD at layer 2/3 cortical synapses.
Collapse
|
32
|
Vakalopoulos C. The effect of deficient muscarinic signaling on commonly reported biochemical effects in schizophrenia and convergence with genetic susceptibility loci in explaining symptom dimensions of psychosis. Front Pharmacol 2014; 5:277. [PMID: 25566074 PMCID: PMC4266038 DOI: 10.3389/fphar.2014.00277] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2014] [Accepted: 11/27/2014] [Indexed: 11/13/2022] Open
Abstract
With the advent of DSM 5 criticism has generally centered on a lack of biological validity of the diagnostic criteria. Part of the problem in describing a nosology of psychosis is the tacit assumption of multiple genetic causes each with an incremental loading on the clinical picture that fails to differentiate a clear underlying pathophysiology of high impact. The aim of this paper is to consolidate a primary theory of deficient muscarinic signaling underlying key clinical features of schizophrenia and its regulation by several important genetic associations including neuregulin, DISC and dysbindin. Secondary reductions in markers for GABAergic function and changes in the levels of interneuron calcium binding proteins parvalbumin and calbindin can be attributed to dysfunctional muscarinic transduction. A parallel association exists for cytokine production. The convergent pathway hypothesis is likewise used to model dopaminergic and glutamatergic theories of schizophrenia. The negative symptom dimension is correlated with dysfunction of Akt and ERK transduction, a major point of convergence. The present paradigm predicts the importance of a recent finding of a deletion in a copy number variant of PLCB1 and its potential use if replicated, as one of the first testable biological markers differentiating schizophrenia from bipolar disorder and further subtyping of schizophrenia into deficit and non-deficit. Potential limitations of PLCB1 as a prospective marker are also discussed.
Collapse
|
33
|
Lucas EK, Jegarl A, Clem RL. Mice lacking TrkB in parvalbumin-positive cells exhibit sexually dimorphic behavioral phenotypes. Behav Brain Res 2014; 274:219-25. [PMID: 25127683 DOI: 10.1016/j.bbr.2014.08.011] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2014] [Revised: 07/09/2014] [Accepted: 08/04/2014] [Indexed: 12/20/2022]
Abstract
Activity-dependent brain-derived neurotrophic factor (BDNF) signaling through receptor tyrosine kinase B (TrkB) is required for cued fear memory consolidation and extinction. Although BDNF is primarily secreted from glutamatergic neurons, TrkB is expressed by other genetically defined cells whose contributions to the behavioral effects of BDNF remain poorly understood. Parvalbumin (PV)-positive interneurons, which are highly enriched in TrkB, are emerging as key regulators of fear memory expression. We therefore hypothesized that activity-dependent BDNF signaling in PV-interneurons may modulate emotional learning. To test this hypothesis, we utilized the LoxP/Cre system for conditional deletion of TrkB in PV-positive cells to examine the impact of cell-autonomous BDNF signaling on Pavlovian fear conditioning and extinction. However, behavioral abnormalities indicative of vestibular dysfunction precluded the use of homozygous conditional knockouts in tests of higher cognitive functioning. While vestibular dysfunction was apparent in both sexes, female conditional knockouts exhibited an exacerbated phenotype, including extreme motor hyperactivity and circling behavior, compared to their male littermates. Heterozygous conditional knockouts were spared of vestibular dysfunction. While fear memory consolidation was unaffected in heterozygotes of both sexes, males exhibited impaired extinction consolidation compared to their littermate controls. Our findings complement evidence from human and rodent studies suggesting that BDNF signaling promotes consolidation of extinction and point to PV-positive neurons as a discrete population that mediates these effects in a sex-specific manner.
Collapse
Affiliation(s)
- Elizabeth K Lucas
- Fishberg Department of Neuroscience and the Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, One Gustave Levy Place, Box 1065, New York, NY 10029, USA
| | - Anita Jegarl
- Fishberg Department of Neuroscience and the Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, One Gustave Levy Place, Box 1065, New York, NY 10029, USA
| | - Roger L Clem
- Fishberg Department of Neuroscience and the Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, One Gustave Levy Place, Box 1065, New York, NY 10029, USA.
| |
Collapse
|
34
|
Aoki C, Wable G, Chowdhury TG, Sabaliauskas NA, Laurino K, Barbarich-Marsteller NC. α4βδ-GABAARs in the hippocampal CA1 as a biomarker for resilience to activity-based anorexia. Neuroscience 2014; 265:108-23. [PMID: 24444828 PMCID: PMC3996507 DOI: 10.1016/j.neuroscience.2014.01.011] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2013] [Revised: 12/09/2013] [Accepted: 01/07/2014] [Indexed: 12/01/2022]
Abstract
Anorexia nervosa (AN) is a psychiatric illness characterized by restricted eating and an intense fear of gaining weight. Most individuals with AN are females, diagnosed first during adolescence, 40-80% of whom exhibit excessive exercise, and an equally high number with a history of anxiety disorder. We sought to determine the cellular basis for individual differences in AN vulnerability by using an animal model, activity-based anorexia (ABA), that is induced by combining food restriction (FR) with access to a running wheel that allows voluntary exercise. Previously, we showed that by the fourth day of FR, the ABA group of adolescent female rats exhibit >500% greater levels of non-synaptic α4βδ-GABAARs at the plasma membrane of hippocampal CA1 pyramidal cell spines, relative to the levels found in age-matched controls that are not FR and without wheel access. Here, we show that the ABA group exhibits individual differences in body weight loss, with some losing nearly 30%, while others lose only 15%. The individual differences in weight loss are ascribable to individual differences in wheel activity that both precedes and concurs with days of FR. Moreover, the increase in activity during FR correlates strongly and negatively with α4βδ-GABAAR levels (R=-0.9, p<0.01). This negative correlation is evident within 2days of FR, before body weight loss approaches life-threatening levels for any individual. These findings suggest that increased shunting inhibition by α4βδ-GABAARs in spines of CA1 pyramidal neurons may participate in the protection against the ABA-inducing environmental factors of severe weight loss by suppressing excitability of the CA1 pyramidal neurons which, in turn, is related indirectly to suppression of excessive exercise. The data also indicate that, although exercise has many health benefits, it can be maladaptive to individuals with low levels of α4βδ-GABAARs in the CA1, particularly when combined with FR.
Collapse
Affiliation(s)
- C Aoki
- Center for Neural Science, New York University, New York, NY 10003, United States.
| | - G Wable
- Center for Neural Science, New York University, New York, NY 10003, United States
| | - T G Chowdhury
- Center for Neural Science, New York University, New York, NY 10003, United States
| | - N A Sabaliauskas
- Center for Neural Science, New York University, New York, NY 10003, United States
| | - K Laurino
- Department of Psychiatry, College of Physicians and Surgeons of Columbia University, New York, NY 10032, United States; Department of Psychiatry, New York State Psychiatric Institute, New York, NY 10032, United States
| | - N C Barbarich-Marsteller
- Department of Psychiatry, College of Physicians and Surgeons of Columbia University, New York, NY 10032, United States; Department of Psychiatry, New York State Psychiatric Institute, New York, NY 10032, United States
| |
Collapse
|
35
|
Liu H, Xu H, Yu T, Yao J, Zhao C, Yin ZQ. Expression of perineuronal nets, parvalbumin and protein tyrosine phosphatase σ in the rat visual cortex during development and after BFD. Curr Eye Res 2013; 38:1083-94. [PMID: 23718120 DOI: 10.3109/02713683.2013.803287] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
UNLABELLED Abstract Purpose of the Study: Protein tyrosine phosphatase σ (PTPσ) acts as a neuronal receptor for chondroitin sulfate proteoglycans (CSPGs). CSPGs have inhibitory effects on experience-dependent plasticity and usually form lattice-like cell coatings that surround the parvalbumin (PV) interneurons in the visual cortex (VC). We investigated developmental changes and the effect of binocular form deprivation (BFD) on PTPσ, perineuronal nets (PNNs) and their tempo-spatial relationships with PV neurons in the VC. MATERIALS AND METHODS Double-immunostaining was used to observe the coexpression pattern of PNNs staining by biotinylated wisteria floribunda lectin (WFA) with PV neurons. The expression of PTPσ in the VC of Long Evans rats was detected by real-time quantitative PCR, immunohistochemistry and western blots. The changes in the number of PV/WFA/PTPσ labeled cells in layer IV of the VC and its proportion of PV neurons were examined during development and after BFD. RESULTS The expression of PV neurons wrapped by PNNs was increased, particularly in the first half of the critical period, and the ratio for PV neurons reached the highest level (over 75%) at adulthood, indicating that PNNs may play an important role in the maturation of PV neurons during the critical period. BFD decreased the density of PNNs and the percentage of PV neurons with PNNs. This result suggests that the number of PNNs surrounding PV neurons may be experience-dependent. Meanwhile, the CSPG receptor PTPσ was maintained at its lowest level during the critical period and could be modulated by BFD after the critical period. The percentage of PV/WFA/PTPσ-positive cells in PV population increased during development and reached its highest ratio at adulthood, which could also be reversed by BFD. CONCLUSIONS The changes in the coexpression of PNNs, PV and PTPσ provide valuable insights into the connection between CSPGs and PV neurons.
Collapse
Affiliation(s)
- Hui Liu
- Southwest Eye Hospital, Southwest Hospital, Third Military Medical University, Chongqing, China
| | | | | | | | | | | |
Collapse
|
36
|
Kelsom C, Lu W. Development and specification of GABAergic cortical interneurons. Cell Biosci 2013; 3:19. [PMID: 23618463 PMCID: PMC3668182 DOI: 10.1186/2045-3701-3-19] [Citation(s) in RCA: 115] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2013] [Accepted: 03/28/2013] [Indexed: 12/21/2022] Open
Abstract
GABAergic interneurons are inhibitory neurons of the nervous system that play a vital role in neural circuitry and activity. They are so named due to their release of the neurotransmitter gamma-aminobutyric acid (GABA), and occupy different areas of the brain. This review will focus primarily on GABAergic interneurons of the mammalian cerebral cortex from a developmental standpoint. There is a diverse amount of cortical interneuronal subtypes that may be categorized by a number of characteristics; this review will classify them largely by the protein markers they express. The developmental origins of GABAergic interneurons will be discussed, as well as factors that influence the complex migration routes that these interneurons must take in order to ultimately localize in the cerebral cortex where they will integrate with the neural circuitry set in place. This review will also place an emphasis on the transcriptional network of genes that play a role in the specification and maintenance of GABAergic interneuron fate. Gaining an understanding of the different aspects of cortical interneuron development and specification, especially in humans, has many useful clinical applications that may serve to treat various neurological disorders linked to alterations in interneuron populations.
Collapse
Affiliation(s)
- Corey Kelsom
- Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, Department of Biochemistry and Molecular Biology, University of Southern California, 1425 San Pablo Street, Los Angeles, CA 90033, USA.
| | | |
Collapse
|
37
|
Intrinsically determined cell death of developing cortical interneurons. Nature 2012; 491:109-13. [PMID: 23041929 PMCID: PMC3726009 DOI: 10.1038/nature11523] [Citation(s) in RCA: 246] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2009] [Accepted: 08/17/2012] [Indexed: 12/29/2022]
Abstract
Cortical inhibitory circuits are formed by GABAergic interneurons, a cell population that originates far from the cerebral cortex in the embryonic ventral forebrain. Given their distant developmental origins, it is intriguing how the number of cortical interneurons is ultimately determined. One possibility, suggested by the neurotrophic hypothesis1-5, is that cortical interneurons are overproduced, and then following their migration into cortex, excess interneurons are eliminated through a competition for extrinsically derived trophic signals. Here we have characterized the developmental cell death of mouse cortical interneurons in vivo, in vitro, and following transplantation. We found that 40% of developing cortical interneurons were eliminated through Bax- (Bcl-2 associated X-) dependent apoptosis during postnatal life. When cultured in vitro or transplanted into the cortex, interneuron precursors died at a cellular age similar to that at which endogenous interneurons died during normal development. Remarkably, over transplant sizes that varied 200-fold, a constant fraction of the transplanted population underwent cell death. The death of transplanted neurons was not affected by the cell-autonomous disruption of TrkB (tropomyosin kinase receptor B), the main neurotrophin receptor expressed by central nervous system (CNS) neurons6-8. Transplantation expanded the cortical interneuron population by up to 35%, but the frequency of inhibitory synaptic events did not scale with the number of transplanted interneurons. Together, our findings indicate that interneuron cell death is intrinsically determined, either cell-autonomously, or through a population-autonomous competition for survival signals derived from other interneurons.
Collapse
|
38
|
Dynamic plasticity: the role of glucocorticoids, brain-derived neurotrophic factor and other trophic factors. Neuroscience 2012; 239:214-27. [PMID: 22922121 DOI: 10.1016/j.neuroscience.2012.08.034] [Citation(s) in RCA: 170] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2012] [Revised: 08/15/2012] [Accepted: 08/16/2012] [Indexed: 12/12/2022]
Abstract
Brain-derived neurotrophic factor (BDNF) is a secreted protein that has been linked to numerous aspects of plasticity in the central nervous system (CNS). Stress-induced remodeling of the hippocampus, prefrontal cortex and amygdala is coincident with changes in the levels of BDNF, which has been shown to act as a trophic factor facilitating the survival of existing and newly born neurons. Initially, hippocampal atrophy after chronic stress was associated with reduced BDNF, leading to the hypothesis that stress-related learning deficits resulted from suppressed hippocampal neurogenesis. However, recent evidence suggests that BDNF also plays a rapid and essential role in regulating synaptic plasticity, providing another mechanism through which BDNF can modulate learning and memory after a stressful event. Numerous reports have shown BDNF levels are highly dynamic in response to stress, and not only vary across brain regions but also fluctuate rapidly, both immediately after a stressor and over the course of a chronic stress paradigm. Yet, BDNF alone is not sufficient to effect many of the changes observed after stress. Glucocorticoids and other molecules have been shown to act in conjunction with BDNF to facilitate both the morphological and molecular changes that occur, particularly changes in spine density and gene expression. This review briefly summarizes the evidence supporting BDNF's role as a trophic factor modulating neuronal survival, and will primarily focus on the interactions between BDNF and other systems within the brain to facilitate synaptic plasticity. This growing body of evidence suggests a more nuanced role for BDNF in stress-related learning and memory, where it acts primarily as a facilitator of plasticity and is dependent upon the coactivation of glucocorticoids and other factors as the determinants of the final cellular response.
Collapse
|
39
|
Schizophrenia. Transl Neurosci 2012. [DOI: 10.1017/cbo9780511980053.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
|
40
|
Faux C, Rakic S, Andrews W, Britto JM. Neurons on the move: migration and lamination of cortical interneurons. Neurosignals 2012; 20:168-89. [PMID: 22572780 DOI: 10.1159/000334489] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The modulation of cortical activity by GABAergic interneurons is required for normal brain function and is achieved through the immense level of heterogeneity within this neuronal population. Cortical interneurons share a common origin in the ventral telencephalon, yet during the maturation process diverse subtypes are generated that form the characteristic laminar arrangement observed in the adult brain. The long distance tangential and short-range radial migration into the cortical plate is regulated by a combination of intrinsic and extrinsic signalling mechanisms, and a great deal of progress has been made to understand these developmental events. In this review, we will summarize current findings regarding the molecular control of subtype specification and provide a detailed account of the migratory cues influencing interneuron migration and lamination. Furthermore, a dysfunctional GABAergic system is associated with a number of neurological and psychiatric conditions, and some of these may have a developmental aetiology with alterations in interneuron generation and migration. We will discuss the notion of additional sources of interneuron progenitors found in human and non-human primates and illustrate how the disruption of early developmental events can instigate a loss in GABAergic function.
Collapse
Affiliation(s)
- Clare Faux
- Centre for Neuroscience, University of Melbourne, Parkville, Vic, Australia
| | | | | | | |
Collapse
|
41
|
Langlois A, Diabira D, Ferrand N, Porcher C, Gaiarsa JL. NMDA-dependent switch of proBDNF actions on developing GABAergic synapses. Cereb Cortex 2012; 23:1085-96. [PMID: 22510533 DOI: 10.1093/cercor/bhs071] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
The brain-derived neurotrophic factor (BDNF) has emerged as an important messenger for activity-dependent development of neuronal network. Recent findings have suggested that a significant proportion of BDNF can be secreted as a precursor (proBDNF) and cleaved by extracellular proteases to yield the mature form. While the actions of proBDNF on maturation and plasticity of excitatory synapses have been studied, the effect of the precursor on developing GABAergic synapses remains largely unknown. Here, we show that regulated secretion of proBDNF exerts a bidirectional control of GABAergic synaptic activity with NMDA receptors driving the polarity of the plasticity. When NMDA receptors are activated during ongoing synaptic activity, regulated Ca(2+)-dependent secretion of proBDNF signals via p75(NTR) to depress GABAergic synaptic activity, while in the absence of NMDA receptors activation, secreted proBDNF induces a p75(NTR)-dependent potentiation of GABAergic synaptic activity. These results revealed a new function for proBDNF-p75(NTR) signaling in synaptic plasticity and a novel mechanism by which synaptic activity can modulate the development of GABAergic synaptic connections.
Collapse
Affiliation(s)
- Anais Langlois
- Institut National de la Santé et de la Recherche Médicale Unité 901, Marseille 13009, France
| | | | | | | | | |
Collapse
|
42
|
A key mechanism underlying sensory experience-dependent maturation of neocortical GABAergic circuits in vivo. Proc Natl Acad Sci U S A 2011; 108:12131-6. [PMID: 21730187 DOI: 10.1073/pnas.1105296108] [Citation(s) in RCA: 67] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Mechanisms underlying experience-dependent refinement of cortical connections, especially GABAergic inhibitory circuits, are unknown. By using a line of mutant mice that lack activity-dependent BDNF expression (bdnf-KIV), we show that experience regulation of cortical GABAergic network is mediated by activity-driven BDNF expression. Levels of endogenous BDNF protein in the barrel cortex are strongly regulated by sensory inputs from whiskers. There is a severe alteration of excitation and inhibition balance in the barrel cortex of bdnf-KIV mice as a result of reduced inhibitory but not excitatory conductance. Within the inhibitory circuits, the mutant barrel cortex exhibits significantly reduced levels of GABA release only from the parvalbumin-expressing fast-spiking (FS) interneurons, but not other interneuron subtypes. Postnatal deprivation of sensory inputs markedly decreased perisomatic inhibition selectively from FS cells in wild-type but not bdnf-KIV mice. These results suggest that postnatal experience, through activity-driven BDNF expression, controls cortical development by regulating FS cell-mediated perisomatic inhibition in vivo.
Collapse
|
43
|
Martinowich K, Schloesser RJ, Jimenez DV, Weinberger DR, Lu B. Activity-dependent brain-derived neurotrophic factor expression regulates cortistatin-interneurons and sleep behavior. Mol Brain 2011; 4:11. [PMID: 21388527 PMCID: PMC3061911 DOI: 10.1186/1756-6606-4-11] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2011] [Accepted: 03/09/2011] [Indexed: 01/19/2023] Open
Abstract
Background Sleep homeostasis is characterized by a positive correlation between sleep length and intensity with the duration of the prior waking period. A causal role for brain-derived neurotrophic factor (BDNF) in sleep homeostasis has been suggested, but the underlying mechanisms remain unclear. Cortistatin, a neuropeptide expressed primarily in a subset of cortical GABAergic interneurons, is another molecule implicated in sleep homeostasis. Results We confirmed that sleep deprivation leads to an increase in cortical cortistatin mRNA expression. Disruption of activity-dependent BDNF expression in a genetically modified mouse line impairs both baseline levels of cortistatin mRNA as well as its levels following sleep deprivation. Disruption of activity-dependent BDNF also leads to a decrease in sleep time during the active (dark) phase. Conclusion Our studies suggest that regulation of cortistatin-expressing interneurons by activity-dependent BDNF expression may contribute to regulation of sleep behavior.
Collapse
Affiliation(s)
- Keri Martinowich
- Genes, Cognition and Psychosis Program (GCAP), National Institute of Mental Health (NIMH), Bethesda, MD 20892, USA
| | | | | | | | | |
Collapse
|
44
|
Balu DT, Coyle JT. Neuroplasticity signaling pathways linked to the pathophysiology of schizophrenia. Neurosci Biobehav Rev 2011; 35:848-70. [PMID: 20951727 PMCID: PMC3005823 DOI: 10.1016/j.neubiorev.2010.10.005] [Citation(s) in RCA: 126] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2010] [Revised: 10/06/2010] [Accepted: 10/10/2010] [Indexed: 12/15/2022]
Abstract
Schizophrenia is a severe mental illness that afflicts nearly 1% of the world's population. One of the cardinal pathological features of schizophrenia is perturbation in synaptic connectivity. Although the etiology of schizophrenia is unknown, it appears to be a developmental disorder involving the interaction of a potentially large number of risk genes, with no one gene producing a strong effect except rare, highly penetrant copy number variants. The purpose of this review is to detail how putative schizophrenia risk genes (DISC-1, neuregulin/ErbB4, dysbindin, Akt1, BDNF, and the NMDA receptor) are involved in regulating neuroplasticity and how alterations in their expression may contribute to the disconnectivity observed in schizophrenia. Moreover, this review highlights how many of these risk genes converge to regulate common neurotransmitter systems and signaling pathways. Future studies aimed at elucidating the functions of these risk genes will provide new insights into the pathophysiology of schizophrenia and will likely lead to the nomination of novel therapeutic targets for restoring proper synaptic connectivity in the brain in schizophrenia and related disorders.
Collapse
Affiliation(s)
- Darrick T Balu
- Department of Psychiatry, Harvard Medical School, Belmont, MA, USA.
| | | |
Collapse
|
45
|
Eagleson KL, Campbell DB, Thompson BL, Bergman MY, Levitt P. The autism risk genes MET and PLAUR differentially impact cortical development. Autism Res 2010; 4:68-83. [PMID: 21328570 DOI: 10.1002/aur.172] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2010] [Accepted: 11/01/2010] [Indexed: 02/06/2023]
Abstract
Candidate risk genes for autism spectrum disorder (ASD) have been identified, but the challenge of determining their contribution to pathogenesis remains. We previously identified two ASD risk genes encoding the receptor tyrosine kinase MET and the urokinase plasminogen activator receptor (PLAUR), which is thought to modulate availability of the MET ligand. We also reported a role for Met signaling in cortical interneuron development in vitro and a reduction of these neurons in uPAR (mouse ortholog of PLAUR) null mice, suggesting that disruption of either gene impacts cortical development similarly. Here, we modify this conclusion, reporting that interneuron numbers are unchanged in the neocortex of Met(fx/fx) / Dlx5/6(cre) mice, in which Met is ablated from cells arising from the ventral telencephalon (VTel). Consistent with this, Met transcript is not detected in the VTel during interneuron genesis and migration; furthermore, during the postnatal period of interneuron maturation, Met is co-expressed in glutamatergic projection neurons, but not interneurons. Low levels of Met protein are expressed in the VTel at E12.5 and E14.5, likely reflecting the arrival of Met containing corticofugal axons. Met expression, however, is induced in E12.5 VTel cells after 2 days in vitro, perhaps underlying discrepancies between observations in vitro and in Met(fx/fx) / Dlx5/6(cre) mice. We suggest that, in vivo, Met impacts the development of cortical projection neurons, whereas uPAR influences interneuron maturation. An altered balance between excitation and inhibition has been postulated as a biological mechanism for ASD; this imbalance could arise from different risk genes differentially affecting either or both elements.
Collapse
Affiliation(s)
- Kathie L Eagleson
- Zilkha Neurogenetic Institute, Keck School of Medicine at USC, Los Angeles, California 90033, USA.
| | | | | | | | | |
Collapse
|
46
|
Woo TUW, Spencer K, McCarley RM. Gamma oscillation deficits and the onset and early progression of schizophrenia. Harv Rev Psychiatry 2010; 18:173-89. [PMID: 20415633 PMCID: PMC2860612 DOI: 10.3109/10673221003747609] [Citation(s) in RCA: 80] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
A fascinating convergence of evidence in recent years has implicated the disturbances of neural synchrony in the gamma frequency band (30-100 Hz) as a major pathophysiologic feature of schizophrenia. Evidence suggests that reduced glutamatergic neurotransmission via the N-methyl-D-aspartate (NMDA) receptors that are localized to inhibitory interneurons, perhaps especially the fast-spiking cells that contain the calcium-binding protein parvalbumin (PV), may contribute to gamma band synchrony deficits. These deficits may underlie the brain's failure to integrate information and hence the manifestations of many symptoms and deficits of schizophrenia. Furthermore, because gamma oscillations are thought to provide the temporal structure that is necessary for synaptic plasticity, gamma oscillation deficits may disturb the developmental synaptic reorganization process that is occurring during the period of late adolescence and early adulthood. This disturbance may contribute to the onset of schizophrenia and the functional deterioration that is characteristic of the early stage of the illness. Finally, reduced NMDA neurotransmission on inhibitory interneurons, including the PV-containing cells, may inflict excitotoxic or oxidative injury to downstream pyramidal neurons, leading to further loss of synapses and dendritic branchings. Hence, a key element in the conceptualization of rational early-intervention and prevention strategies for schizophrenia may involve correcting the abnormal NMDA neurotransmission on inhibitory interneurons-possibly that on the PV-containing neurons, in particular-thereby normalizing gamma oscillation deficits and attenuating downstream neuronal pathology.
Collapse
Affiliation(s)
- Tsung-Ung W. Woo
- Laboratory of Translational Psychiatry, Mailman Research Center McLean Hospital Belmont, MA 02478,Department of Psychiatry, Beth Israel Deaconess Medical Center, Boston, MA 02215,Department of Psychiatry, Harvard Medical School, Boston, MA 02115
| | - Kevin Spencer
- Department of Psychiatry, VA Boston Healthcare System, Brockton, MA 02301,Department of Psychiatry, Harvard Medical School, Boston, MA 02115
| | - Robert M. McCarley
- Laboratory of Translational Psychiatry, Mailman Research Center McLean Hospital Belmont, MA 02478,Department of Psychiatry, VA Boston Healthcare System, Brockton, MA 02301,Department of Psychiatry, Harvard Medical School, Boston, MA 02115
| |
Collapse
|
47
|
Molecules and mechanisms involved in the generation and migration of cortical interneurons. ASN Neuro 2010; 2:e00031. [PMID: 20360946 PMCID: PMC2847827 DOI: 10.1042/an20090053] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2009] [Revised: 03/04/2010] [Accepted: 03/05/2010] [Indexed: 11/30/2022] Open
Abstract
The GABA (γ-aminobutyric acid)-containing interneurons of the neocortex are largely derived from the ganglionic eminences in the subpallium. Numerous studies have previously defined the migratory paths travelled by these neurons from their origins to their destinations in the cortex. We review here results of studies that have identified many of the genes expressed in the subpallium that are involved in the specification of the subtypes of cortical interneurons, and the numerous transcription factors, motogenic factors and guidance molecules that are involved in their migration.
Collapse
Key Words
- 5-HT, 5-hydroxytryptamine
- AEP, anterior entopeduncular
- BDNF, brain-derived neurotrophic factor
- CGE, caudal ganglionic eminence
- CP, cortical plate
- CR, calretinin
- CXCR, CXC chemokine receptor
- E, embryonic day
- GABA, γ-aminobutyric acid
- GABAR, GABA receptor
- HGF/SF, hepatocyte growth factor/scatter factor
- IZ, intermediate zone
- LGE, lateral ganglionic eminence
- MGE, medial ganglionic eminence
- MZ, marginal zone
- NGR, neuregulin
- NPY, neuropeptide Y
- Nrp, neuropilin
- POA, preoptic area
- PV, paravalbumin
- Robo, Roundabout
- SDF-1, stromal-derived factor 1
- SHH, sonic hedgehog
- SST, somatostatin
- SVZ, subventricular zone
- VZ, ventricular zone
- gene expression
- interneuron
- migration
- neocortex
- neuronal specification
- subpallium
Collapse
|
48
|
Cell death and proliferation in acute slices and organotypic cultures of mammalian CNS. Prog Neurobiol 2009; 88:221-45. [DOI: 10.1016/j.pneurobio.2009.01.002] [Citation(s) in RCA: 112] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2008] [Revised: 12/09/2008] [Accepted: 01/07/2009] [Indexed: 11/24/2022]
|
49
|
Cell and receptor type-specific alterations in markers of GABA neurotransmission in the prefrontal cortex of subjects with schizophrenia. Neurotox Res 2009; 14:237-48. [PMID: 19073429 DOI: 10.1007/bf03033813] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
Impairments in cognitive control, such as those involved in working memory, are associated with dysfunction of the dorsolateral prefrontal cortex (DLPFC) in individuals with schizophrenia. This dysfunction appears to result, at least in part, from abnormalities in GABA-mediated neurotransmission. In this paper, we review recent findings indicating that the altered DLPFC circuitry in subjects with schizophrenia reflects changes in the expression of genes that encode selective presynaptic and postsynaptic components of GABA neurotransmission. Specifically, using a combination of methods, we found that subjects with schizophrenia exhibited expression deficits in GABA-related transcripts encoding presynaptic regulators of GABA neurotransmission, neuropeptide markers of specific subpopulations of GABA neurons, and certain subunits of the GABA(A) receptor. In particular, alterations in the expression of the neuropeptide somatostatin suggested that GABA neurotransmission is impaired in the Martinotti subset of GABA neurons that target the dendrites of pyramidal cells. In contrast, none of the GABA-related transcripts assessed to date were altered in the DLPFC of monkeys chronically exposed to antipsychotic medications, suggesting that the effects observed in the human studies reflect the disease process and not its treatment. In concert with previous findings, these data suggest that working memory dysfunction in schizophrenia may be attributable to altered GABA neurotransmission in specific DLPFC microcircuits.
Collapse
|
50
|
Critical role of promoter IV-driven BDNF transcription in GABAergic transmission and synaptic plasticity in the prefrontal cortex. Proc Natl Acad Sci U S A 2009; 106:5942-7. [PMID: 19293383 DOI: 10.1073/pnas.0811431106] [Citation(s) in RCA: 160] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Transcription of Bdnf is controlled by multiple promoters, which drive expression of multiple transcripts encoding for the same protein. Promoter IV contributes significantly to activity-dependent brain-derived neurotrophic factor (BDNF) transcription. We have generated promoter IV mutant mice (BDNF-KIV) by inserting a GFP-STOP cassette within the Bdnf exon IV locus. This genetic manipulation results in disruption of promoter IV-mediated Bdnf expression. BDNF-KIV animals exhibited significant deficits in GABAergic interneurons in the prefrontal cortex (PFC), particularly those expressing parvalbumin, a subtype implicated in executive function and schizophrenia. Moreover, disruption of promoter IV-driven Bdnf transcription impaired inhibitory but not excitatory synaptic transmission recorded from layer V pyramidal neurons in the PFC. The attenuation of GABAergic inputs resulted in an aberrant appearance of spike-timing-dependent synaptic potentiation (STDP) in PFC slices derived from BDNF-KIV, but not wild-type littermates. These results demonstrate the importance of promoter IV-dependent Bdnf transcription in GABAergic function and reveal an unexpected regulation of STDP in the PFC by BDNF.
Collapse
|