1
|
Ma H, Khaled HG, Wang X, Mandelberg NJ, Cohen SM, He X, Tsien RW. Excitation-transcription coupling, neuronal gene expression and synaptic plasticity. Nat Rev Neurosci 2023; 24:672-692. [PMID: 37773070 DOI: 10.1038/s41583-023-00742-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/24/2023] [Indexed: 09/30/2023]
Abstract
Excitation-transcription coupling (E-TC) links synaptic and cellular activity to nuclear gene transcription. It is generally accepted that E-TC makes a crucial contribution to learning and memory through its role in underpinning long-lasting synaptic enhancement in late-phase long-term potentiation and has more recently been linked to late-phase long-term depression: both processes require de novo gene transcription, mRNA translation and protein synthesis. E-TC begins with the activation of glutamate-gated N-methyl-D-aspartate-type receptors and voltage-gated L-type Ca2+ channels at the membrane and culminates in the activation of transcription factors in the nucleus. These receptors and ion channels mediate E-TC through mechanisms that include long-range signalling from the synapse to the nucleus and local interactions within dendritic spines, among other possibilities. Growing experimental evidence links these E-TC mechanisms to late-phase long-term potentiation and learning and memory. These advances in our understanding of the molecular mechanisms of E-TC mean that future efforts can focus on understanding its mesoscale functions and how it regulates neuronal network activity and behaviour in physiological and pathological conditions.
Collapse
Affiliation(s)
- Huan Ma
- Department of Neurobiology, Affiliated Mental Health Center and Hangzhou Seventh People's Hospital, Zhejiang University School of Medicine, Hangzhou, China.
- Liangzhu Laboratory, MOE Frontier Science Center for Brain Science and Brain-Machine Integration, State Key Laboratory of Brain-Machine Intelligence, Zhejiang University, Hangzhou, China.
- NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University, Hangzhou, China.
- Research Units for Emotion and Emotional Disorders, Chinese Academy of Medical Sciences, Beijing, China.
| | - Houda G Khaled
- NYU Neuroscience Institute and Department of Neuroscience and Physiology, NYU Langone Medical Center, New York, NY, USA
- Center for Neural Science, New York University, New York, NY, USA
| | - Xiaohan Wang
- NYU Neuroscience Institute and Department of Neuroscience and Physiology, NYU Langone Medical Center, New York, NY, USA
| | - Nataniel J Mandelberg
- NYU Neuroscience Institute and Department of Neuroscience and Physiology, NYU Langone Medical Center, New York, NY, USA
| | - Samuel M Cohen
- NYU Neuroscience Institute and Department of Neuroscience and Physiology, NYU Langone Medical Center, New York, NY, USA
| | - Xingzhi He
- Department of Neurobiology, Affiliated Mental Health Center and Hangzhou Seventh People's Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Liangzhu Laboratory, MOE Frontier Science Center for Brain Science and Brain-Machine Integration, State Key Laboratory of Brain-Machine Intelligence, Zhejiang University, Hangzhou, China
- NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University, Hangzhou, China
- Research Units for Emotion and Emotional Disorders, Chinese Academy of Medical Sciences, Beijing, China
| | - Richard W Tsien
- NYU Neuroscience Institute and Department of Neuroscience and Physiology, NYU Langone Medical Center, New York, NY, USA.
- Center for Neural Science, New York University, New York, NY, USA.
| |
Collapse
|
2
|
Cheung SKK, Kwok J, Or PMY, Wong CW, Feng B, Choy KW, Chang RCC, Burbach JPH, Cheng ASL, Chan AM. Neuropathological signatures revealed by transcriptomic and proteomic analysis in Pten-deficient mouse models. Sci Rep 2023; 13:6763. [PMID: 37185447 PMCID: PMC10130134 DOI: 10.1038/s41598-023-33869-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2022] [Accepted: 04/20/2023] [Indexed: 05/17/2023] Open
Abstract
PTEN hamartoma tumour syndrome is characterised by mutations in the human PTEN gene. We performed transcriptomic and proteomic analyses of neural tissues and primary cultures from heterozygous and homozygous Pten-knockout mice. The somatosensory cortex of heterozygous Pten-knockout mice was enriched in immune response and oligodendrocyte development Gene Ontology (GO) terms. Parallel proteomic analysis revealed differentially expressed proteins (DEPs) related to dendritic spine development, keratinisation and hamartoma signatures. However, primary astrocytes (ASTs) from heterozygous Pten-knockout mice were enriched in the extracellular matrix GO term, while primary cortical neurons (PCNs) were enriched in immediate-early genes. In ASTs from homozygous Pten-knockout mice, cilium-related activity was enriched, while PCNs exhibited downregulation of forebrain neuron generation and differentiation, implying an altered excitatory/inhibitory balance. By integrating DEPs with pre-filtered differentially expressed genes, we identified the enrichment of traits of intelligence, cognitive function and schizophrenia, while DEPs in ASTs were significantly associated with intelligence and depression.
Collapse
Affiliation(s)
- Stanley K K Cheung
- School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, SAR, China
| | - Jacinda Kwok
- School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, SAR, China
- Department of Pharmaceutical Sciences, University of Toronto, Toronto, Canada
| | - Penelope M Y Or
- School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, SAR, China
| | - Chi Wai Wong
- School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, SAR, China
- Louvain Institute of Biomolecular Science and Technology, Université catholique de Louvain, Louvain-la-Neuve, Belgium
| | - Bo Feng
- School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, SAR, China
| | - Kwong Wai Choy
- Department of Obstetrics and Gynaecology, The Chinese University of Hong Kong, Hong Kong, SAR, China
| | - Raymond C C Chang
- Laboratory of Neurodegenerative Diseases, School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, SAR, China
| | - J Peter H Burbach
- Department of Translational Neuroscience, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Alfred S L Cheng
- School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, SAR, China
| | - Andrew M Chan
- School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, SAR, China.
- Brain and Mind Institute, The Chinese University of Hong Kong, 4/F, Hui Yeung Shing Building, Hong Kong, SAR, China.
| |
Collapse
|
3
|
Chen Y, Li S, Zhang T, Yang F, Lu B. Corticosterone antagonist or TrkB agonist attenuates schizophrenia-like behavior in a mouse model combining Bdnf-e6 deficiency and developmental stress. iScience 2022; 25:104609. [PMID: 35789832 PMCID: PMC9250029 DOI: 10.1016/j.isci.2022.104609] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Revised: 05/16/2022] [Accepted: 06/08/2022] [Indexed: 12/17/2022] Open
Affiliation(s)
- Yanhui Chen
- School of Pharmaceutical Sciences, IDG/McGovern Institute for Brain Research, Tsinghua-Peking Joint Center for Life Sciences, Tsinghua University, Beijing 100084, China
| | - Shangjin Li
- School of Pharmaceutical Sciences, IDG/McGovern Institute for Brain Research, Tsinghua-Peking Joint Center for Life Sciences, Tsinghua University, Beijing 100084, China
| | - Tianyi Zhang
- School of Pharmaceutical Sciences, IDG/McGovern Institute for Brain Research, Tsinghua-Peking Joint Center for Life Sciences, Tsinghua University, Beijing 100084, China
| | - Feng Yang
- China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing 100084, China
- Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing 100070, China
| | - Bai Lu
- School of Pharmaceutical Sciences, IDG/McGovern Institute for Brain Research, Tsinghua-Peking Joint Center for Life Sciences, Tsinghua University, Beijing 100084, China
- Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing 100070, China
- Corresponding author
| |
Collapse
|
4
|
Wollet M, Kim JH. Brain-Derived Neurotrophic Factor Is Involved in Activity-Dependent Tonotopic Refinement of MNTB Neurons. Front Neural Circuits 2022; 16:784396. [PMID: 35185479 PMCID: PMC8850952 DOI: 10.3389/fncir.2022.784396] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Accepted: 01/07/2022] [Indexed: 01/21/2023] Open
Abstract
In the mammalian brain, auditory brainstem nuclei are arranged topographically according to acoustic frequency responsiveness. During postnatal development, the axon initial segment (AIS) of principal neurons undergoes structural refinement depending on location along the tonotopic axis within the medial nucleus of the trapezoid body (MNTB). However, the molecular mechanisms underlying the structural refinement of the AIS along the tonotopic axis in the auditory brainstem have not been explored. We tested the hypothesis that brain-derived neurotrophic factor (BDNF) is a molecular mediator of the structural development of the MNTB in an activity-dependent manner. Using BDNF heterozygous mutant (BDNF+/- ) mice, we examined the impact of global BDNF reduction on structural and functional development of MNTB neurons by assessing AIS structure and associated intrinsic neuronal properties. BDNF reduction inhibits the structural and functional differentiation of principal neurons along the tonotopic axis in the MNTB. Augmented sound input during the critical period of development has been shown to enhance the structural refinement of the AIS of MNTB neurons. However, in BDNF +/- mice, MNTB neurons did not show this activity-dependent structural modification of the AIS following repeated sound stimulation. In addition, BDNF+/- mice lacked a defined isofrequency band of neuronal activity following exposure to 16 kHz sound, suggesting degradation of tonotopy. Taken together, structural development and functional refinement of auditory brainstem neurons require physiological levels of BDNF to establish proper tonotopic gradients.
Collapse
Affiliation(s)
| | - Jun Hee Kim
- Department of Cellular and Integrative Physiology, UT Health San Antonio, San Antonio, TX, United States
| |
Collapse
|
5
|
Long-lasting Postnatal Sensory Deprivation Alters Dendritic Morphology of Pyramidal Neurons in the Rat Hippocampus: Behavioral Correlates. Neuroscience 2022; 480:79-96. [PMID: 34785272 DOI: 10.1016/j.neuroscience.2021.11.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2021] [Revised: 11/04/2021] [Accepted: 11/05/2021] [Indexed: 11/20/2022]
Abstract
The role of normal sensory inputs in the development of sensory cortices is well known, however, their impacts on the hippocampus, an integrator of sensory modalities with important roles in cognitive functions, has received much less attention. Here, we applied a long-term sensory deprivation paradigm by trimming the rats' whiskers bilaterally, from postnatal day 3 to 59. Female sensory-deprived (SD) rats showed more on-wall rearing and visits to the center of the open-field box, shorter periods of grooming, less defecation and less anxiety-like behaviors in the elevated plus-maze compared to controls, who had their intact whiskers brushed. Passive avoidance memory retention was sex-dependently impaired in the female SD rats. In the radial arm maze, however, reference spatial memory was impaired only in the male SD rats. Nonetheless, working memory errors increased in both sexes of SD rats. Besides depletion of CA1 and CA3 pyramidal neurons in SD rats, Sholl analysis of Golgi-Cox stained neurons revealed that prolonged sensory deprivation has retracted the arborization of CA1 basal dendrites in SD group, while solely female SD rats had diminished CA1 apical dendrites. Sholl analysis of CA3 neurons in SD animals also disclosed significantly more branched apical dendrites in males and basal dendrites in females. Sensory deprivation also led to a considerable spine loss and variation of different spine types in a sex-dependent manner. Our findings suggest that experience-dependent structural plasticity is capable of spreading far beyond the manipulated sensory zones and the inevitable functional alterations can be expressed in a multifactorial sex-dependent manner.
Collapse
|
6
|
Prestigio C, Ferrante D, Marte A, Romei A, Lignani G, Onofri F, Valente P, Benfenati F, Baldelli P. REST/NRSF drives homeostatic plasticity of inhibitory synapses in a target-dependent fashion. eLife 2021; 10:e69058. [PMID: 34855580 PMCID: PMC8639147 DOI: 10.7554/elife.69058] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Accepted: 11/22/2021] [Indexed: 12/31/2022] Open
Abstract
The repressor-element 1-silencing transcription/neuron-restrictive silencer factor (REST/NRSF) controls hundreds of neuron-specific genes. We showed that REST/NRSF downregulates glutamatergic transmission in response to hyperactivity, thus contributing to neuronal homeostasis. However, whether GABAergic transmission is also implicated in the homeostatic action of REST/NRSF is unknown. Here, we show that hyperactivity-induced REST/NRSF activation, triggers a homeostatic rearrangement of GABAergic inhibition, with increased frequency of miniature inhibitory postsynaptic currents (IPSCs) and amplitude of evoked IPSCs in mouse cultured hippocampal neurons. Notably, this effect is limited to inhibitory-onto-excitatory neuron synapses, whose density increases at somatic level and decreases in dendritic regions, demonstrating a complex target- and area-selectivity. The upscaling of perisomatic inhibition was occluded by TrkB receptor inhibition and resulted from a coordinated and sequential activation of the Npas4 and Bdnf gene programs. On the opposite, the downscaling of dendritic inhibition was REST-dependent, but BDNF-independent. The findings highlight the central role of REST/NRSF in the complex transcriptional responses aimed at rescuing physiological levels of network activity in front of the ever-changing environment.
Collapse
Affiliation(s)
- Cosimo Prestigio
- Department of Experimental Medicine, University of GenovaGenovaItaly
- Center for Synaptic Neuroscience and Technology, Istituto Italiano di TecnologiaGenovaItaly
| | - Daniele Ferrante
- Department of Experimental Medicine, University of GenovaGenovaItaly
- Center for Synaptic Neuroscience and Technology, Istituto Italiano di TecnologiaGenovaItaly
| | - Antonella Marte
- Department of Experimental Medicine, University of GenovaGenovaItaly
| | - Alessandra Romei
- Center for Synaptic Neuroscience and Technology, Istituto Italiano di TecnologiaGenovaItaly
| | - Gabriele Lignani
- Department of Clinical and Experimental Epilepsy, UCL Queen Square Institute of Neurology, Queen Square HouseLondonUnited Kingdom
| | - Franco Onofri
- Department of Experimental Medicine, University of GenovaGenovaItaly
- IRCCS, Ospedale Policlinico San MartinoGenovaItaly
| | - Pierluigi Valente
- Department of Experimental Medicine, University of GenovaGenovaItaly
- IRCCS, Ospedale Policlinico San MartinoGenovaItaly
| | - Fabio Benfenati
- Center for Synaptic Neuroscience and Technology, Istituto Italiano di TecnologiaGenovaItaly
- IRCCS, Ospedale Policlinico San MartinoGenovaItaly
| | - Pietro Baldelli
- Department of Experimental Medicine, University of GenovaGenovaItaly
- IRCCS, Ospedale Policlinico San MartinoGenovaItaly
| |
Collapse
|
7
|
Woodward EM, Coutellier L. Age- and sex-specific effects of stress on parvalbumin interneurons in preclinical models: Relevance to sex differences in clinical neuropsychiatric and neurodevelopmental disorders. Neurosci Biobehav Rev 2021; 131:1228-1242. [PMID: 34718048 PMCID: PMC8642301 DOI: 10.1016/j.neubiorev.2021.10.031] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 10/06/2021] [Accepted: 10/23/2021] [Indexed: 01/06/2023]
Abstract
Stress is a major risk factor for neurodevelopmental and neuropsychiatric disorders, with the capacity to impact susceptibility to disease as well as long-term neurobiological and behavioral outcomes. Parvalbumin (PV) interneurons, the most prominent subtype of GABAergic interneurons in the cortex, are uniquely responsive to stress due to their protracted development throughout the highly plastic neonatal period and into puberty and adolescence. Additionally, PV + interneurons appear to respond to stress in a sex-specific manner. This review aims to discuss existing preclinical studies that support our overall hypothesis that the sex-and age-specific impacts of stress on PV + interneurons contribute to differences in individual vulnerability to stress across the lifespan, particularly in regard to sex differences in the diagnostic rate of neurodevelopmental and neuropsychiatric diseases in clinical populations. We also emphasize the importance of studying sex as a biological variable to fully understand the mechanistic and behavioral differences between males and females in models of neuropsychiatric disease.
Collapse
Affiliation(s)
- Emma M Woodward
- Department of Neuroscience, Ohio State University, 255 Institute for Behavioral Medicine Research Building, 460 Medical Center Drive, Columbus, OH, 43210, United States
| | - Laurence Coutellier
- Department of Neuroscience, Ohio State University, 255 Institute for Behavioral Medicine Research Building, 460 Medical Center Drive, Columbus, OH, 43210, United States; Department of Psychology, Ohio State University, 53 Psychology Building, 1835 Neil Avenue, Columbus, OH, 43210, United States.
| |
Collapse
|
8
|
Postnatal Sox6 Regulates Synaptic Function of Cortical Parvalbumin-Expressing Neurons. J Neurosci 2021; 41:8876-8886. [PMID: 34503995 PMCID: PMC8549537 DOI: 10.1523/jneurosci.0021-21.2021] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Revised: 08/12/2021] [Accepted: 08/16/2021] [Indexed: 11/29/2022] Open
Abstract
Cortical parvalbumin-expressing (Pvalb+) neurons provide robust inhibition to neighboring pyramidal neurons, crucial for the proper functioning of cortical networks. This class of inhibitory neurons undergoes extensive synaptic formation and maturation during the first weeks after birth and continue to dynamically maintain their synaptic output throughout adulthood. While several transcription factors, such as Nkx2-1, Lhx6, and Sox6, are known to be necessary for the differentiation of progenitors into Pvalb+ neurons, which transcriptional programs underlie the postnatal maturation and maintenance of Pvalb+ neurons' innervation and synaptic function remains largely unknown. Because Sox6 is continuously expressed in Pvalb+ neurons until adulthood, we used conditional knock-out strategies to investigate its putative role in the postnatal maturation and synaptic function of cortical Pvalb+ neurons in mice of both sexes. We found that early postnatal loss of Sox6 in Pvalb+ neurons leads to failure of synaptic bouton growth, whereas later removal in mature Pvalb+ neurons in the adult causes shrinkage of already established synaptic boutons. Paired recordings between Pvalb+ neurons and pyramidal neurons revealed reduced release probability and increased failure rate of Pvalb+ neurons' synaptic output. Furthermore, Pvalb+ neurons lacking Sox6 display reduced expression of full-length tropomyosin-receptor kinase B (TrkB), a key modulator of GABAergic transmission. Once re-expressed in neurons lacking Sox6, TrkB was sufficient to rescue the morphologic synaptic phenotype. Finally, we showed that Sox6 mRNA levels were increased by motor training. Our data thus suggest a constitutive role for Sox6 in the maintenance of synaptic output from Pvalb+ neurons into adulthood. SIGNIFICANCE STATEMENT Cortical parvalbumin-expressing (Pvalb+) inhibitory neurons provide robust inhibition to neighboring pyramidal neurons, crucial for the proper functioning of cortical networks. These inhibitory neurons undergo extensive synaptic formation and maturation during the first weeks after birth and continue to dynamically maintain their synaptic output throughout adulthood. However, it remains largely unknown which transcriptional programs underlie the postnatal maturation and maintenance of Pvalb+ neurons. Here, we show that the transcription factor Sox6 cell-autonomously regulates the synaptic maintenance and output of Pvalb+ neurons until adulthood, leaving unaffected other maturational features of this neuronal population.
Collapse
|
9
|
Lau CG, Zhang H, Murthy VN. Deletion of TrkB in parvalbumin interneurons alters cortical neural dynamics. J Cell Physiol 2021; 237:949-964. [PMID: 34491578 PMCID: PMC8810709 DOI: 10.1002/jcp.30571] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 08/20/2021] [Accepted: 08/23/2021] [Indexed: 11/25/2022]
Abstract
Signaling by neurotrophins such as the brain‐derived neurotrophic factor (BDNF) is known to modulate development of interneurons, but the circuit effects of this modulation remain unclear. Here, we examined the impact of deleting TrkB, a BDNF receptor, in parvalbumin‐expressing (PV) interneurons on the balance of excitation and inhibition (E‐I) in cortical circuits. In the mouse olfactory cortex, TrkB deletion impairs multiple aspects of PV neuronal function including synaptic excitation, intrinsic excitability, and the innervation pattern of principal neurons. Impaired PV cell function resulted in aberrant spiking patterns in principal neurons in response to stimulation of sensory inputs. Surprisingly, dampened PV neuronal function leads to a paradoxical decrease in overall excitability in cortical circuits. Our study demonstrates that, by modulating PV circuit plasticity and development, TrkB plays a critical role in shaping the evoked pattern of activity in a cortical network.
Collapse
Affiliation(s)
- Chunyue Geoffrey Lau
- Department of Neuroscience, City University of Hong Kong, Hong Kong, China.,Department of Molecular and Cellular Biology and Center for Brain Science, Harvard University, Cambridge, Massachusetts, USA
| | - Huiqi Zhang
- Department of Neuroscience, City University of Hong Kong, Hong Kong, China
| | - Venkatesh N Murthy
- Department of Molecular and Cellular Biology and Center for Brain Science, Harvard University, Cambridge, Massachusetts, USA
| |
Collapse
|
10
|
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
|
11
|
Inhibitory neurotransmission drives endocannabinoid degradation to promote memory consolidation. Nat Commun 2020; 11:6407. [PMID: 33335094 PMCID: PMC7747732 DOI: 10.1038/s41467-020-20121-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2018] [Accepted: 11/12/2020] [Indexed: 11/08/2022] Open
Abstract
Endocannabinoids retrogradely regulate synaptic transmission and their abundance is controlled by the fine balance between endocannabinoid synthesis and degradation. While the common assumption is that “on-demand” release determines endocannabinoid signaling, their rapid degradation is expected to control the temporal profile of endocannabinoid action and may impact neuronal signaling. Here we show that memory formation through fear conditioning selectively accelerates the degradation of endocannabinoids in the cerebellum. Learning induced a lasting increase in GABA release and this was responsible for driving the change in endocannabinoid degradation. Conversely, Gq-DREADD activation of cerebellar Purkinje cells enhanced endocannabinoid signaling and impaired memory consolidation. Our findings identify a previously unappreciated reciprocal interaction between GABA and the endocannabinoid system in which GABA signaling accelerates endocannabinoid degradation, and triggers a form of learning-induced metaplasticity. Endocannabinoid levels are controlled by the fine balance between their synthesis and degradation. Here, the authors show that memory formation through fear conditioning selectively accelerates the degradation of endocannabinoids in the cerebellum via a lasting increase in GABA release.
Collapse
|
12
|
Charles James J, Funke K. Repetitive transcranial magnetic stimulation reverses reduced excitability of rat visual cortex induced by dark rearing during early critical period. Dev Neurobiol 2020; 80:399-410. [PMID: 33006265 DOI: 10.1002/dneu.22785] [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: 07/27/2020] [Revised: 09/14/2020] [Accepted: 09/24/2020] [Indexed: 01/20/2023]
Abstract
Early critical period of visual cortex is characterized by enhanced activity-driven neuronal plasticity establishing the specificity of neuronal connections required for optimal processing of sensory signals. Deprivation from visual input by dark rearing (DR) during this period leads to a lasting impairment of visual performance. Previously, we demonstrated that repetitive transcranial magnetic stimulation (rTMS) applied with intermittent theta-burst (iTBS) pattern during the critical period improved the visual performance of the DR rats. In this study, we describe that the excitability of the binocular part of the visual cortex (V1b), as measured in acute brain slices by input-output ratios of field excitatory synaptic potentials (fEPSPs), is lowered in DR rats compared to normal controls. Verum rTMS applied with the iTBS pattern during DR reversed this DR effect, while no rTMS effect was evident in the non-DR (nDR) rats. In addition, verum rTMS reduced the number of neurons expressing the 67 kD isoform of glutamic acid decarboxylase (GAD67), the calcium-binding protein calbindin (CB) and the zinc-finger transcription factor zif268/EGR1, as determined via immunohistochemistry, only in DR rats but not in nDR rats. Moreover, rTMS reduced the number of neurons expressing the calcium-binding protein parvalbumin (PV) only in nDR rats which showed more PV+ neurons compared to DR rats. This study confirms that iTBS-rTMS may be able to prevent or reverse the effects of DR on visual cortex physiology, likely through a modulation of the activity of inhibitory interneurons.
Collapse
Affiliation(s)
| | - Klaus Funke
- Department of Neurophysiology, Medical Faculty, Ruhr-University Bochum, Bochum, Germany
| |
Collapse
|
13
|
Huang S, Kirkwood A. Endocannabinoid Signaling Contributes to Experience-Induced Increase of Synaptic Release Sites From Parvalbumin Interneurons in Mouse Visual Cortex. Front Cell Neurosci 2020; 14:571133. [PMID: 33192316 PMCID: PMC7556304 DOI: 10.3389/fncel.2020.571133] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Accepted: 08/28/2020] [Indexed: 11/13/2022] Open
Abstract
During postnatal development of the visual cortex between eye-opening to puberty, visual experience promotes a gradual increase in the strength of inhibitory synaptic connections from parvalbumin-positive interneurons (PV-INs) onto layer 2/3 pyramidal cells. However, the detailed connectivity properties and molecular mechanisms underlying these developmental changes are not well understood. Using dual-patch clamp in brain slices from G42 mice, we revealed that both connection probability and the number of synaptic release sites contributed to the enhancement of synaptic strength. The increase of release site number was hindered by dark rearing from eye-opening and rescued by 3-days re-exposure to the normal visual environment. The effect of light re-exposure on restoring synaptic release sites in dark reared mice was mimicked by the agonist of cannabinoid-1 (CB1) receptors and blocked by an antagonist of these receptors, suggesting a role for endocannabinoid signaling in light-induced maturation of inhibitory connectivity from PV-INs to pyramidal cells during postnatal development.
Collapse
Affiliation(s)
- Shiyong Huang
- Program in Neuroscience, Hussman Institute for Autism, Baltimore, MD, United States.,The Mind/Brain Institute, Johns Hopkins University, Baltimore, MD, United States
| | - Alfredo Kirkwood
- The Mind/Brain Institute, Johns Hopkins University, Baltimore, MD, United States.,Department of Neuroscience, Johns Hopkins University, Baltimore, MD, United States
| |
Collapse
|
14
|
Laliberté G, Othman R, Vaucher E. Mesoscopic Mapping of Stimulus-Selective Response Plasticity in the Visual Pathways Modulated by the Cholinergic System. Front Neural Circuits 2020; 14:38. [PMID: 32719589 PMCID: PMC7350895 DOI: 10.3389/fncir.2020.00038] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Accepted: 05/22/2020] [Indexed: 11/13/2022] Open
Abstract
The cholinergic potentiation of visual conditioning enhances visual acuity and discrimination of the trained stimulus. To determine if this also induces long-term plastic changes on cortical maps and connectivity in the visual cortex and higher associative areas, mesoscopic calcium imaging was performed in head-fixed awake GCaMP6s adult mice before and after conditioning. The conditioned stimulus (0.03 cpd, 30°, 100% contrast, 1 Hz-drifting gratings) was presented 10 min daily for a week. Saline or Donepezil (DPZ, 0.3 mg/kg, s.c.), a cholinesterase inhibitor that potentiates cholinergic transmission, were injected prior to each conditioning session and compared to a sham-conditioned group. Cortical maps of resting state and evoked response to the monocular presentation of conditioned or non-conditioned stimulus (30°, 50 and 75% contrast; 90°, 50, 75, and 100% contrast) were established. Amplitude, duration, and latency of the peak response, as well as size of activation were measured in the primary visual cortex (V1), secondary visual areas (AL, A, AM, PM, LM, RL), retrosplenial cortex (RSC), and higher cortical areas. Visual stimulation increased calcium signaling in all primary and secondary visual areas, the RSC, but no other cortices. There were no significant effects of sham-conditioning or conditioning alone, but DPZ treatment during conditioning significantly decreased the integrated neuronal activity of superficial layers evoked by the conditioned stimulus in V1, AL, PM, and LM. The activity of downstream cortical areas was not changed. The size of the activated area was decreased in V1 and PM, and the signal-to-noise ratio was decreased in AL and PM. Interestingly, signal correlation was seen only between V1, the ventral visual pathway, and the RSC, and was decreased by DPZ administration. The resting state activity was slightly correlated and rarely affected by treatments, except between binocular and monocular V1 in both hemispheres. In conclusion, cholinergic potentiation of visual conditioning induced change in visual processing in the superficial cortical layers. This effect might be a key mechanism in the establishment of the fine cortical tuning in response to the conditioned visual stimulus.
Collapse
Affiliation(s)
- Guillaume Laliberté
- Laboratoire de Neurobiologie de la Cognition Visuelle, École d'Optométrie, Université de Montréal, Montréal, QC, Canada
| | - Rahmeh Othman
- Laboratoire de Neurobiologie de la Cognition Visuelle, École d'Optométrie, Université de Montréal, Montréal, QC, Canada.,Départment de Pharmacologie et Physiologie, Faculté de Médecine, Université de Montréal, Montréal, QC, Canada
| | - Elvire Vaucher
- Laboratoire de Neurobiologie de la Cognition Visuelle, École d'Optométrie, Université de Montréal, Montréal, QC, Canada
| |
Collapse
|
15
|
Capogna M, Castillo PE, Maffei A. The ins and outs of inhibitory synaptic plasticity: Neuron types, molecular mechanisms and functional roles. Eur J Neurosci 2020; 54:6882-6901. [PMID: 32663353 DOI: 10.1111/ejn.14907] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Revised: 06/30/2020] [Accepted: 07/08/2020] [Indexed: 01/05/2023]
Abstract
GABAergic interneurons are highly diverse, and their synaptic outputs express various forms of plasticity. Compelling evidence indicates that activity-dependent changes of inhibitory synaptic transmission play a significant role in regulating neural circuits critically involved in learning and memory and circuit refinement. Here, we provide an updated overview of inhibitory synaptic plasticity with a focus on the hippocampus and neocortex. To illustrate the diversity of inhibitory interneurons, we discuss the case of two highly divergent interneuron types, parvalbumin-expressing basket cells and neurogliaform cells, which support unique roles on circuit dynamics. We also present recent progress on the molecular mechanisms underlying long-term, activity-dependent plasticity of fast inhibitory transmission. Lastly, we discuss the role of inhibitory synaptic plasticity in neuronal circuits' function. The emerging picture is that inhibitory synaptic transmission in the CNS is extremely diverse, undergoes various mechanistically distinct forms of plasticity and contributes to a much more refined computational role than initially thought. Both the remarkable diversity of inhibitory interneurons and the various forms of plasticity expressed by GABAergic synapses provide an amazingly rich inhibitory repertoire that is central to a variety of complex neural circuit functions, including memory.
Collapse
Affiliation(s)
- Marco Capogna
- Department of Biomedicine, Danish National Research Foundation Center of Excellence PROMEMO, Aarhus University, Aarhus, Denmark
| | - Pablo E Castillo
- Dominck P Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY, USA.,Department of Psychiatry and Behavioral Sciences, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Arianna Maffei
- Center for Neural Circuit Dynamics and Department of Neurobiology and Behavior, Stony Brook University, Stony Brook, NY, USA
| |
Collapse
|
16
|
Cisneros-Franco JM, Voss P, Thomas ME, de Villers-Sidani E. Critical periods of brain development. HANDBOOK OF CLINICAL NEUROLOGY 2020; 173:75-88. [PMID: 32958196 DOI: 10.1016/b978-0-444-64150-2.00009-5] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Brain plasticity is maximal at specific time windows during early development known as critical periods (CPs), during which sensory experience is necessary to establish optimal cortical representations of the surrounding environment. After CP closure, a range of functional and structural elements prevent passive experience from eliciting significant plastic changes in the brain. The transition from a plastic to a more fixed state is advantageous as it allows for the sequential consolidation and retention of new and more complex perceptual, motor, and cognitive functions. However, the formation of stable neural representations may pose limitations on future revisions to the circuitry. If sensory experience is abnormal or absent during this time, it can have profound effects on sensory representations in adulthood, resulting in quasi-permanent adaptations that can make it nearly impossible to learn certain skills or process certain stimulus properties later on in life. This chapter begins with a brief introduction to experience-dependent plasticity throughout the lifespan (Section Introduction). Next, we define what constitutes a CP (Section What Are Critical Periods?) and review some of the key CPs in the visual and auditory systems (Section Key Critical Periods of Sensory Systems). We then discuss the mechanisms whereby cortical plasticity is regulated both locally and through neuromodulatory systems (Section How Are Critical Periods Regulated?). Finally, we highlight studies showing that CPs can be extended beyond their normal epochs, closed prematurely, or reopened during adult life by merely altering sensory inputs (Section Timing of Critical Periods: Can CP Plasticity Be Extended, Limited, or Reactivated?).
Collapse
Affiliation(s)
- J Miguel Cisneros-Franco
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, QC, Canada; Centre for Research on Brain, Language and Music, McGill University, Montreal, QC, Canada
| | - Patrice Voss
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, QC, Canada; Centre for Research on Brain, Language and Music, McGill University, Montreal, QC, Canada
| | - Maryse E Thomas
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, QC, Canada; Centre for Research on Brain, Language and Music, McGill University, Montreal, QC, Canada
| | - Etienne de Villers-Sidani
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, QC, Canada; Centre for Research on Brain, Language and Music, McGill University, Montreal, QC, Canada.
| |
Collapse
|
17
|
Chiu CQ, Barberis A, Higley MJ. Preserving the balance: diverse forms of long-term GABAergic synaptic plasticity. Nat Rev Neurosci 2019; 20:272-281. [PMID: 30837689 DOI: 10.1038/s41583-019-0141-5] [Citation(s) in RCA: 70] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Cellular mechanisms that regulate the interplay of synaptic excitation and inhibition are thought to be central to the functional stability of healthy neuronal circuits. A growing body of literature demonstrates the capacity for inhibitory GABAergic synapses to exhibit long-term plasticity in response to changes in neuronal activity. Here, we review this expanding field of research, focusing on the diversity of mechanisms that link glutamatergic signalling, postsynaptic action potentials and inhibitory synaptic strength. Several lines of evidence indicate that multiple, parallel forms of plasticity serve to regulate activity at both the input and output domains of individual neurons. Overall, these varied phenomena serve to promote both stability and flexibility over the life of the organism.
Collapse
Affiliation(s)
- Chiayu Q Chiu
- Centro Interdisciplinario de Neurociencia de Valparaiso, Universidad de Valparaiso, Valparaiso, Chile
| | | | - Michael J Higley
- Department of Neuroscience, Yale University, New Haven, CT, USA.
| |
Collapse
|
18
|
Modol L, Bollmann Y, Tressard T, Baude A, Che A, Duan ZRS, Babij R, De Marco García NV, Cossart R. Assemblies of Perisomatic GABAergic Neurons in the Developing Barrel Cortex. Neuron 2019; 105:93-105.e4. [PMID: 31780328 DOI: 10.1016/j.neuron.2019.10.007] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Revised: 07/23/2019] [Accepted: 10/01/2019] [Indexed: 02/07/2023]
Abstract
The developmental journey of cortical interneurons encounters several activity-dependent milestones. During the early postnatal period in developing mice, GABAergic neurons are transient preferential recipients of thalamic inputs and undergo activity-dependent migration arrest, wiring, and programmed cell-death. Despite their importance for the emergence of sensory experience and the role of activity in their integration into cortical networks, the collective dynamics of GABAergic neurons during that neonatal period remain unknown. Here, we study coordinated activity in GABAergic cells of the mouse barrel cortex using in vivo calcium imaging. We uncover a transient structure in GABAergic population dynamics that disappears in a sensory-dependent process. Its building blocks are anatomically clustered GABAergic assemblies mostly composed by prospective parvalbumin-expressing cells. These progressively widen their territories until forming a uniform perisomatic GABAergic network. Such transient patterning of GABAergic activity is a functional scaffold that links the cortex to the external world prior to active exploration. VIDEO ABSTRACT.
Collapse
Affiliation(s)
- Laura Modol
- Aix Marseille Univ, INSERM, INMED, Turing Center for Living Systems, Marseille, France
| | - Yannick Bollmann
- Aix Marseille Univ, INSERM, INMED, Turing Center for Living Systems, Marseille, France
| | - Thomas Tressard
- Aix Marseille Univ, INSERM, INMED, Turing Center for Living Systems, Marseille, France
| | - Agnès Baude
- Aix Marseille Univ, INSERM, INMED, Turing Center for Living Systems, Marseille, France
| | - Alicia Che
- Brain and Mind Research Institute, Weill Cornell Medical College, New York, New York, USA
| | - Zhe Ran S Duan
- Brain and Mind Research Institute, Weill Cornell Medical College, New York, New York, USA; Weill Cornell/Rockefeller/Sloan Kettering Tri-Institutional MD-PhD Program, New York, NY 10021, USA
| | - Rachel Babij
- Brain and Mind Research Institute, Weill Cornell Medical College, New York, New York, USA; Weill Cornell/Rockefeller/Sloan Kettering Tri-Institutional MD-PhD Program, New York, NY 10021, USA
| | | | - Rosa Cossart
- Aix Marseille Univ, INSERM, INMED, Turing Center for Living Systems, Marseille, France.
| |
Collapse
|
19
|
You H, Chu P, Guo W, Lu B. A subpopulation of Bdnf-e1-expressing glutamatergic neurons in the lateral hypothalamus critical for thermogenesis control. Mol Metab 2019; 31:109-123. [PMID: 31918913 PMCID: PMC6920260 DOI: 10.1016/j.molmet.2019.11.013] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Revised: 11/16/2019] [Accepted: 11/17/2019] [Indexed: 12/14/2022] Open
Abstract
Objective Brown adipose tissue (BAT)–mediated thermogenesis plays a key role in energy homeostasis and the maintenance of body temperature. Previous work suggests that brain-derived neurotrophic factor (BDNF) is involved in BAT thermogenesis, but the underlying neural circuits and molecular mechanism remain largely unknown. This is in part due to the difficulties in manipulating BDNF expression in different brain regions through different promoters and the lack of tools to identify neurons in the brain specifically involved in BAT thermogenesis. Methods We have created several lines of mutant mice in which BDNF transcription from a specific promoter was selectively disrupted by replacing Bdnf with green fluorescent protein (GFP; Bdnf-e1, -e4, and -e6−/− mice). As such, cells expressing Bdnf-e1, -e4, or -e6 were labeled with GFP. To identify BAT-connected thermogenesis neurons in brain, we applied the retrograde pseudorabies virus labeling method from BAT. We also used chemogenetic tools to manipulate specific neurons coupled with BAT temperature recording. Moreover, we developed a new TrkB agonist antibody to rescue the BAT thermogenesis deficits. Results We show that selective disruption of Bdnf expression from promoter 1 (Bdnf-e1) resulted in severe obesity and deficits of BAT-mediated thermogenesis. Body temperature response to cold was impaired in Bdnf-e1−/− mice. BAT expression of Ucp1 and Pcg1a, genes known to regulate thermogenesis, was also reduced, accompanying a decrease in the sympathetic activity of BAT. Staining of cells expressing Bdnf-e1 transcript, combined with transsynaptic, retrograde-tracing labeling of BAT-connected neurons, identified a group of excitatory neurons in lateral hypothalamus (LH) critical for thermogenesis regulation. Moreover, an adaptive thermogenesis defect in Bdnf-e1−/− mice was rescued by injecting an agonistic antibody for TrkB, the BDNF receptor, into LH. Remarkably, activation of the excitatory neurons (VGLUT2+) in LH through chemogenetic tools resulted in a rise of BAT temperature. Conclusions These results reveal a specific role of BDNF promoter I in thermogenesis regulation and define a small subset of neurons in LH that contribute to such regulation. Only Bdnf-e1−/−, but not Bdnf-e4−/− or Bdnf-e6−/−, mutant mice exhibited deficiencies of BAT thermogenesis. Neurons that are both Bdnf-e1 expressing and BAT-connected were found only in LH. BAT-connected neurons in LH are glutamatergic. Activation of the LH glutamatergic neurons resulted in an increase in BAT temperature. Administration of TrkB agonist antibody in LH rescued thermogenesis deficits.
Collapse
Affiliation(s)
- He You
- School of Pharmaceutical Sciences, Tsinghua University, Beijing, 100084, China; School of Life Sciences, Tsinghua University, Beijing, 100084, China; Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Pengcheng Chu
- School of Pharmaceutical Sciences, Tsinghua University, Beijing, 100084, China; School of Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Wei Guo
- School of Pharmaceutical Sciences, Tsinghua University, Beijing, 100084, China
| | - Bai Lu
- School of Pharmaceutical Sciences, Tsinghua University, Beijing, 100084, China.
| |
Collapse
|
20
|
Chowdhury TG, Wable GS, Chen YW, Tateyama K, Yu I, Wang JY, Reyes AD, Aoki C. Voluntary Wheel Running Exercise Evoked by Food-Restriction Stress Exacerbates Weight Loss of Adolescent Female Rats But Also Promotes Resilience by Enhancing GABAergic Inhibition of Pyramidal Neurons in the Dorsal Hippocampus. Cereb Cortex 2019; 29:4035-4049. [PMID: 30462186 PMCID: PMC6931273 DOI: 10.1093/cercor/bhy283] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2018] [Revised: 10/11/2018] [Accepted: 10/17/2018] [Indexed: 11/13/2022] Open
Abstract
Adolescence is marked by increased vulnerability to mental disorders and maladaptive behaviors, including anorexia nervosa. Food-restriction (FR) stress evokes foraging, which translates to increased wheel running exercise (EX) for caged rodents, a maladaptive behavior, since it does not improve food access and exacerbates weight loss. While almost all adolescent rodents increase EX following FR, some then become resilient by suppressing EX by the second-fourth FR day, which minimizes weight loss. We asked whether GABAergic plasticity in the hippocampus may underlie this gain in resilience. In vitro slice physiology revealed doubling of pyramidal neurons' GABA response in the dorsal hippocampus of food-restricted animals with wheel access (FR + EX for 4 days), but without increase of mIPSC amplitudes. mIPSC frequency increased by 46%, but electron microscopy revealed no increase in axosomatic GABAergic synapse number onto pyramidal cells and only a modest increase (26%) of GABAergic synapse lengths. These changes suggest increase of vesicular release probability and extrasynaptic GABAA receptors and unsilencing of GABAergic synapses. GABAergic synapse lengths correlated with individual's suppression of wheel running and weight loss. These analyses indicate that EX can have dual roles-exacerbate weight loss but also promote resilience to some by dampening hippocampal excitability.
Collapse
Affiliation(s)
| | | | - Yi-Wen Chen
- Center for Neural Science, NYU, New York, NY, USA
| | - Kei Tateyama
- Center for Neural Science, NYU, New York, NY, USA
| | - Irene Yu
- Center for Neural Science, NYU, New York, NY, USA
| | - Jia-Yi Wang
- Center for Neural Science, NYU, New York, NY, USA
| | - Alex D Reyes
- Center for Neural Science, NYU, New York, NY, USA
| | - Chiye Aoki
- Center for Neural Science, NYU, New York, NY, USA
- The Neuroscience Institute, NYU Langone Medical Center, New York, NY, USA
| |
Collapse
|
21
|
Meis S, Endres T, Munsch T, Lessmann V. Impact of Chronic BDNF Depletion on GABAergic Synaptic Transmission in the Lateral Amygdala. Int J Mol Sci 2019; 20:ijms20174310. [PMID: 31484392 PMCID: PMC6747405 DOI: 10.3390/ijms20174310] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Revised: 08/30/2019] [Accepted: 09/01/2019] [Indexed: 01/14/2023] Open
Abstract
Brain-derived neurotrophic factor (BDNF) has previously been shown to play an important role in glutamatergic synaptic plasticity in the amygdala, correlating with cued fear learning. While glutamatergic neurotransmission is facilitated by BDNF signaling in the amygdala, its mechanism of action at inhibitory synapses in this nucleus is far less understood. We therefore analyzed the impact of chronic BDNF depletion on GABAA-mediated synaptic transmission in BDNF heterozygous knockout mice (BDNF+/−). Analysis of miniature and evoked inhibitory postsynaptic currents (IPSCs) in the lateral amygdala (LA) revealed neither pre- nor postsynaptic differences in BDNF+/− mice compared to wild-type littermates. In addition, long-term potentiation (LTP) of IPSCs was similar in both genotypes. In contrast, facilitation of spontaneous IPSCs (sIPSCs) by norepinephrine (NE) was significantly reduced in BDNF+/− mice. These results argue against a generally impaired efficacy and plasticity at GABAergic synapses due to a chronic BDNF deficit. Importantly, the increase in GABAergic tone mediated by NE is reduced in BDNF+/− mice. As release of NE is elevated during aversive behavioral states in the amygdala, effects of a chronic BDNF deficit on GABAergic inhibition may become evident in response to states of high arousal, leading to amygdala hyper-excitability and impaired amygdala function.
Collapse
Affiliation(s)
- Susanne Meis
- Institut für Physiologie, Otto-von-Guericke-Universität, D-39120 Magdeburg, Germany.
- Center for Behavioral Brain Sciences, D-39106 Magdeburg, Germany.
| | - Thomas Endres
- Institut für Physiologie, Otto-von-Guericke-Universität, D-39120 Magdeburg, Germany.
| | - Thomas Munsch
- Institut für Physiologie, Otto-von-Guericke-Universität, D-39120 Magdeburg, Germany.
- Center for Behavioral Brain Sciences, D-39106 Magdeburg, Germany.
| | - Volkmar Lessmann
- Institut für Physiologie, Otto-von-Guericke-Universität, D-39120 Magdeburg, Germany.
- Center for Behavioral Brain Sciences, D-39106 Magdeburg, Germany.
| |
Collapse
|
22
|
Kloosterboer E, Funke K. Repetitive transcranial magnetic stimulation recovers cortical map plasticity induced by sensory deprivation due to deafferentiation. J Physiol 2019; 597:4025-4051. [PMID: 31145483 PMCID: PMC6852264 DOI: 10.1113/jp277507] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2018] [Accepted: 05/17/2019] [Indexed: 12/14/2022] Open
Abstract
KEY POINTS Partial sensory deprivation (deafferentation) by removing whiskers from the rat snout resulted in a reduced responsiveness of related cortical representations. Repetitive transcranial magnetic stimulation (three blocks of intermittent theta-burst) applied for 5 days in combination with sensory exploration restored the normal responsiveness level of the deafferented barrel cortex. However, intracortical inhibition (lateral and recurrent) appeared to be reduced after repetitive transcranial magnetic stimulation, probably as the cause of improved responsiveness. Repetitive transcranial magnetic stimulation also reduced the asymmetry of the lateral spread of sensory activity. ABSTRACT Repetitive transcranial magnetic stimulation (rTMS) modulates human cortical excitability. It has the potential to support recovery to normal cortical function when the excitation-inhibition balance is altered (e.g. after a stroke or loss of sensory input). We tested cortical map plasticity on the basis of sensory responses (local field potentials, LFPs) and expression of neuronal activity marker proteins within the barrel cortex of rats receiving either active or sham rTMS after selective unilateral deafferentation by whiskers plucking. Rats received daily rTMS [intermittent theta-burst (iTBS), active or sham] for 5 days before exploring an enriched environment. Our previous studies indicated a disinhibitory effect of iTBS on cortical activity. Therefore, we also expected disinhibitory effects if deafferentation causes depression of sensory responses. Deafferentation resulted in an acute general reduction of sensory responsiveness and enhanced expression of inhibitory activity markers (GAD67, parvalbumin) in the deafferented hemisphere. Active but not sham-iTBS-rTMS normalized these measures. The stronger caudal-to-frontal horizontal spread of activity across barrels was reduced after deafferentation but not restored after active iTBS, despite generally increased responses. Fitting the LFP data with a computational model of different strengths and types of excitatory and inhibitory connections further revealed an iTBS-induced reduction of lateral and recurrent inhibition as the most probable scenario. Whether the disinhibitory effect of iTBS for the restoration of normal cortical function in the acute phase of depression after deafferentiation is also beneficial in humans remains to be demonstrated. As recently discussed, disinhibition appears to be required to open a window for neuronal plasticity.
Collapse
Affiliation(s)
- Ellen Kloosterboer
- Department of Neurophysiology, Medical Faculty, Ruhr-University Bochum, Bochum, Germany
| | - Klaus Funke
- Department of Neurophysiology, Medical Faculty, Ruhr-University Bochum, Bochum, Germany
| |
Collapse
|
23
|
Shinoda Y, Sadakata T, Yagishita K, Kinameri E, Katoh-Semba R, Sano Y, Furuichi T. Aspects of excitatory/inhibitory synapses in multiple brain regions are correlated with levels of brain-derived neurotrophic factor/neurotrophin-3. Biochem Biophys Res Commun 2018; 509:429-434. [PMID: 30594389 DOI: 10.1016/j.bbrc.2018.12.100] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Accepted: 12/14/2018] [Indexed: 12/29/2022]
Abstract
Appropriate synapse formation during development is necessary for normal brain function, and synapse impairment is often associated with brain dysfunction. Brain-derived neurotrophic factor (BDNF) and neurotrophin-3 (NT-3) are key factors in regulating synaptic development. We previously reported that BDNF/NT-3 secretion was enhanced by calcium-dependent activator protein for secretion 2 (CADPS2). Although BDNF/NT-3 and CADPS2 are co-expressed in various brain regions, the effect of Cadps2-deficiency on brain region-specific BDNF/NT-3 levels and synaptic development remains elusive. Here, we show developmental changes of BDNF/NT-3 levels and we assess disruption of excitatory/inhibitory synapses in multiple brain regions (cerebellum, hypothalamus, striatum, hippocampus, parietal cortex and prefrontal cortex) of Cadps2 knockout (KO) mice compared with wild-type (WT) mice. Compared with WT, BDNF levels in KO mice were reduced in young/adult hippocampus, but increased in young hypothalamus, while NT-3 levels were reduced in adult cerebellum and young hippocampus, but increased in adult parietal cortex. Immunofluorescence of vGluT1, an excitatory synapse marker, and vGAT, an inhibitory synapse marker, in adult KO showed that vGluT1 was higher in the cerebellum and parietal cortex but lower in the hippocampus, whereas vGAT was lower in the hippocampus and parietal cortex compared with WT. Immunolabeling for both vGluT1 and vGAT was increased in the parietal cortex but vGAT was decreased in the cerebellum in adult KO compared with WT. These data suggest that CADPS2-mediated secretion of BDNF/NT-3 may be involved in development and maturation of synapses and in the balance between inhibitory and excitatory synapses.
Collapse
Affiliation(s)
- Yo Shinoda
- Department of Environmental Health, School of Pharmacy, Tokyo University of Pharmacy and Life Sciences, Hachioji, Tokyo, 192-0392, Japan; Department of Applied Biological Science, Faculty of Science and Technology, Tokyo University of Science, Noda, Chiba, 278-8510, Japan; Laboratory for Molecular Neurogenesis, RIKEN Brain Science Institute, Wako, Saitama, 351-0198, Japan.
| | - Tetsushi Sadakata
- Laboratory for Molecular Neurogenesis, RIKEN Brain Science Institute, Wako, Saitama, 351-0198, Japan; Education and Research Support Center, Gunma University Graduate School of Medicine, Maebashi, Gunma, 371-8511, Japan
| | - Kaori Yagishita
- Department of Applied Biological Science, Faculty of Science and Technology, Tokyo University of Science, Noda, Chiba, 278-8510, Japan
| | - Emi Kinameri
- Laboratory for Molecular Neurogenesis, RIKEN Brain Science Institute, Wako, Saitama, 351-0198, Japan
| | - Ritsuko Katoh-Semba
- Department of Applied Biological Science, Faculty of Science and Technology, Tokyo University of Science, Noda, Chiba, 278-8510, Japan; Laboratory for Molecular Neurogenesis, RIKEN Brain Science Institute, Wako, Saitama, 351-0198, Japan
| | - Yoshitake Sano
- Department of Applied Biological Science, Faculty of Science and Technology, Tokyo University of Science, Noda, Chiba, 278-8510, Japan
| | - Teiichi Furuichi
- Department of Applied Biological Science, Faculty of Science and Technology, Tokyo University of Science, Noda, Chiba, 278-8510, Japan; Laboratory for Molecular Neurogenesis, RIKEN Brain Science Institute, Wako, Saitama, 351-0198, Japan.
| |
Collapse
|
24
|
Ueno H, Suemitsu S, Murakami S, Kitamura N, Wani K, Matsumoto Y, Okamoto M, Aoki S, Ishihara T. Juvenile stress induces behavioral change and affects perineuronal net formation in juvenile mice. BMC Neurosci 2018; 19:41. [PMID: 30012101 PMCID: PMC6048828 DOI: 10.1186/s12868-018-0442-z] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2018] [Accepted: 07/11/2018] [Indexed: 12/16/2022] Open
Abstract
Background Many neuropsychiatric disorders develop in early life. Although the mechanisms involved have not been elucidated, it is possible that functional abnormalities of parvalbumin-positive interneurons (PV neurons) are present. Several previous studies have shown that juvenile stress is implicated in the development of neuropsychiatric disorders. We aimed to clarify the effects of juvenile stress on behavior and on the central nervous system. We investigated behavioral abnormalities of chronically-stressed mice during juvenilehood and the effect of juvenile stress on PV neurons and WFA-positive perineuronal nets (PNNs), which are associated with vulnerability and plasticity in the mouse brain. Results Due to juvenile stress, mice showed neurodevelopmental disorder-like behavior. Juvenile stressed mice did not show depressive-like behaviors, but on the contrary, they showed increased activity and decreased anxiety-like behavior. In the central nervous system of juvenile stressed mice, the fluorescence intensity of WFA-positive PNNs decreased, which may signify increased vulnerability. Conclusion This study suggested that juvenile stressed mice showed behavioral abnormalities, resembling those seen in neuropsychiatric disorders, and increased brain vulnerability.
Collapse
Affiliation(s)
- Hiroshi Ueno
- Department of Medical Technology, Kawasaki University of Medical Welfare, 288, Matsushima, Kurashiki, Okayama, 701-0193, Japan. .,Department of Medical Technology, Graduate School of Health Sciences, Okayama University, Okayama, 700-8558, Japan.
| | - Shunsuke Suemitsu
- Department of Psychiatry, Kawasaki Medical School, Kurashiki, 701-0192, Japan
| | - Shinji Murakami
- Department of Psychiatry, Kawasaki Medical School, Kurashiki, 701-0192, Japan
| | - Naoya Kitamura
- Department of Psychiatry, Kawasaki Medical School, Kurashiki, 701-0192, Japan
| | - Kenta Wani
- Department of Psychiatry, Kawasaki Medical School, Kurashiki, 701-0192, Japan
| | - Yosuke Matsumoto
- Department of Neuropsychiatry, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, 700-8558, Japan
| | - Motoi Okamoto
- Department of Medical Technology, Graduate School of Health Sciences, Okayama University, Okayama, 700-8558, Japan
| | - Shozo Aoki
- Department of Psychiatry, Kawasaki Medical School, Kurashiki, 701-0192, Japan
| | - Takeshi Ishihara
- Department of Psychiatry, Kawasaki Medical School, Kurashiki, 701-0192, Japan
| |
Collapse
|
25
|
Gu F, Parada I, Yang T, Longo FM, Prince DA. Partial TrkB receptor activation suppresses cortical epileptogenesis through actions on parvalbumin interneurons. Neurobiol Dis 2018; 113:45-58. [PMID: 29408225 DOI: 10.1016/j.nbd.2018.01.018] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2017] [Revised: 01/21/2018] [Accepted: 01/24/2018] [Indexed: 01/17/2023] Open
Abstract
Post-traumatic epilepsy is one of the most common and difficult to treat forms of acquired epilepsy worldwide. Currently, there is no effective way to prevent post-traumatic epileptogenesis. It is known that abnormalities of interneurons, particularly parvalbumin-containing interneurons, play a critical role in epileptogenesis following traumatic brain injury. Thus, enhancing the function of existing parvalbumin interneurons might provide a logical therapeutic approach to prevention of post-traumatic epilepsy. The known positive effects of brain-derived neurotrophic factor on interneuronal growth and function through activation of its receptor tropomyosin receptor kinase B, and its decrease after traumatic brain injury, led us to hypothesize that enhancing trophic support might improve parvalbumin interneuronal function and decrease epileptogenesis. To test this hypothesis, we used the partial neocortical isolation ('undercut', UC) model of posttraumatic epileptogenesis in mature rats that were treated for 2 weeks, beginning on the day of injury, with LM22A-4, a newly designed partial agonist at the tropomyosin receptor kinase B. Effects of treatment were assessed with Western blots to measure pAKT/AKT; immunocytochemistry and whole cell patch clamp recordings to examine functional and structural properties of GABAergic interneurons; field potential recordings of epileptiform discharges in vitro; and video-EEG recordings of PTZ-induced seizures in vivo. Results showed that LM22A-4 treatment 1) increased pyramidal cell perisomatic immunoreactivity for VGAT, GAD65 and parvalbumin; 2) increased the density of close appositions of VGAT/gephyrin immunoreactive puncta (putative inhibitory synapses) on pyramidal cell somata; 3) increased the frequency of mIPSCs in pyramidal cells; and 4) decreased the incidence of spontaneous and evoked epileptiform discharges in vitro. 5) Treatment of rats with PTX BD4-3, another partial TrkB receptor agonist, reduced the incidence of bicuculline-induced ictal episodes in vitro and PTZ induced electrographic and behavioral ictal episodes in vivo. 6) Inactivation of TrkB receptors in undercut TrkBF616A mice with 1NMPP1 abolished both LM22A-4-induced effects on mIPSCs and on increased perisomatic VGAT-IR. Results indicate that chronic activation of the tropomyosin receptor kinase B by a partial agonist after cortical injury can enhance structural and functional measures of GABAergic inhibition and suppress posttraumatic epileptogenesis. Although the full agonist effects of brain-derived neurotrophic factor and tropomyosin receptor kinase B activation in epilepsy models have been controversial, the present results indicate that such trophic activation by a partial agonist may potentially serve as an effective therapeutic option for prophylactic treatment of posttraumatic epileptogenesis, and treatment of other neurological and psychiatric disorders whose pathogenesis involves impaired parvalbumin interneuronal function.
Collapse
Affiliation(s)
- Feng Gu
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, United States
| | - Isabel Parada
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, United States
| | - Tao Yang
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, United States
| | - Frank M Longo
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, United States
| | - David A Prince
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, United States.
| |
Collapse
|
26
|
Gainey MA, Feldman DE. Multiple shared mechanisms for homeostatic plasticity in rodent somatosensory and visual cortex. Philos Trans R Soc Lond B Biol Sci 2017; 372:rstb.2016.0157. [PMID: 28093551 DOI: 10.1098/rstb.2016.0157] [Citation(s) in RCA: 63] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/19/2016] [Indexed: 12/17/2022] Open
Abstract
We compare the circuit and cellular mechanisms for homeostatic plasticity that have been discovered in rodent somatosensory (S1) and visual (V1) cortex. Both areas use similar mechanisms to restore mean firing rate after sensory deprivation. Two time scales of homeostasis are evident, with distinct mechanisms. Slow homeostasis occurs over several days, and is mediated by homeostatic synaptic scaling in excitatory networks and, in some cases, homeostatic adjustment of pyramidal cell intrinsic excitability. Fast homeostasis occurs within less than 1 day, and is mediated by rapid disinhibition, implemented by activity-dependent plasticity in parvalbumin interneuron circuits. These processes interact with Hebbian synaptic plasticity to maintain cortical firing rates during learned adjustments in sensory representations.This article is part of the themed issue 'Integrating Hebbian and homeostatic plasticity'.
Collapse
Affiliation(s)
- Melanie A Gainey
- Department of Molecular and Cell Biology, Helen Wills Neuroscience Institute, University of California, Berkeley, CA 94720-3200, USA
| | - Daniel E Feldman
- Department of Molecular and Cell Biology, Helen Wills Neuroscience Institute, University of California, Berkeley, CA 94720-3200, USA
| |
Collapse
|
27
|
Gu F, Parada I, Shen F, Li J, Bacci A, Graber K, Taghavi RM, Scalise K, Schwartzkroin P, Wenzel J, Prince DA. Structural alterations in fast-spiking GABAergic interneurons in a model of posttraumatic neocortical epileptogenesis. Neurobiol Dis 2017; 108:100-114. [PMID: 28823934 DOI: 10.1016/j.nbd.2017.08.008] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2017] [Revised: 08/11/2017] [Accepted: 08/16/2017] [Indexed: 01/22/2023] Open
Abstract
Electrophysiological experiments in the partial cortical isolation ("undercut" or "UC") model of injury-induced neocortical epileptogenesis have shown alterations in GABAergic synaptic transmission attributable to abnormalities in presynaptic terminals. To determine whether the decreased inhibition was associated with structural abnormalities in GABAergic interneurons, we used immunocytochemical techniques, confocal microscopy and EM in UC and control sensorimotor rat cortex to analyze structural alterations in fast-spiking parvalbumin-containing interneurons and pyramidal (Pyr) cells of layer V. Principle findings were: 1) there were no decreases in counts of parvalbumin (PV)- or GABA-immunoreactive interneurons in UC cortex, however there were significant reductions in expression of VGAT and GAD-65 and -67 in halos of GABAergic terminals around Pyr somata in layer V. 2) Consistent with previous results, somatic size and density of Pyr cells was decreased in infragranular layers of UC cortex. 3) Dendrites of biocytin-filled FS interneurons were significantly decreased in volume. 4) There were decreases in the size and VGAT content of GABAergic boutons in axons of biocytin-filled FS cells in the UC, together with a decrease in colocalization with postsynaptic gephyrin, suggesting a reduction in GABAergic synapses. Quantitative EM of layer V Pyr somata confirmed the reduction in inhibitory synapses. 5) There were marked and lasting reductions in brain derived neurotrophic factor (BDNF)-IR and -mRNA in Pyr cells and decreased TrkB-IR on PV cells in UC cortex. 6) Results lead to the hypothesis that reduction in trophic support by BDNF derived from Pyr cells may contribute to the regressive changes in axonal terminals and dendrites of FS cells in the UC cortex and decreased GABAergic inhibition. SIGNIFICANCE Injury to cortical structures is a major cause of epilepsy, accounting for about 20% of cases in the general population, with an incidence as high as ~50% among brain-injured personnel in wartime. Loss of GABAergic inhibitory interneurons is a significant pathophysiological factor associated with epileptogenesis following brain trauma and other etiologies. Results of these experiments show that the largest population of cortical interneurons, the parvalbumin-containing fast-spiking (FS) interneurons, are preserved in the partial neocortical isolation model of partial epilepsy. However, axonal terminals of these cells are structurally abnormal, have decreased content of GABA synthetic enzymes and vesicular GABA transporter and make fewer synapses onto pyramidal neurons. These structural abnormalities underlie defects in GABAergic neurotransmission that are a key pathophysiological factor in epileptogenesis found in electrophysiological experiments. BDNF, and its TrkB receptor, key factors for maintenance of interneurons and pyramidal neurons, are decreased in the injured cortex. Results suggest that supplying BDNF to the injured epileptogenic brain may reverse the structural and functional abnormalities in the parvalbumin FS interneurons and provide an antiepileptogenic therapy.
Collapse
Affiliation(s)
- Feng Gu
- Epilepsy Research Laboratories, Stanford Univ. Sch. of Medicine, United States
| | - Isabel Parada
- Epilepsy Research Laboratories, Stanford Univ. Sch. of Medicine, United States
| | - Fran Shen
- Epilepsy Research Laboratories, Stanford Univ. Sch. of Medicine, United States
| | - Judith Li
- Epilepsy Research Laboratories, Stanford Univ. Sch. of Medicine, United States
| | - Alberto Bacci
- ICM - Hôpital Pitié Salpêtrière, 7, bd de l'hôpital, 75013 Paris, France
| | - Kevin Graber
- Epilepsy Research Laboratories, Stanford Univ. Sch. of Medicine, United States
| | - Reza Moein Taghavi
- Epilepsy Research Laboratories, Stanford Univ. Sch. of Medicine, United States
| | - Karina Scalise
- Epilepsy Research Laboratories, Stanford Univ. Sch. of Medicine, United States
| | - Philip Schwartzkroin
- Department of Neurological Surgery, University of California, Davis, United States
| | - Jurgen Wenzel
- Department of Neurological Surgery, University of California, Davis, United States
| | - David A Prince
- Epilepsy Research Laboratories, Stanford Univ. Sch. of Medicine, United States.
| |
Collapse
|
28
|
Chen YW, Surgent O, Rana BS, Lee F, Aoki C. Variant BDNF-Val66Met Polymorphism is Associated with Layer-Specific Alterations in GABAergic Innervation of Pyramidal Neurons, Elevated Anxiety and Reduced Vulnerability of Adolescent Male Mice to Activity-Based Anorexia. Cereb Cortex 2017; 27:3980-3993. [PMID: 27578497 PMCID: PMC6248700 DOI: 10.1093/cercor/bhw210] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2016] [Accepted: 06/09/2016] [Indexed: 02/05/2023] Open
Abstract
Previously, we determined that rodents' vulnerability to food restriction (FR)-evoked wheel running during adolescence (activity-based anorexia, ABA) is associated with failures to increase GABAergic innervation of hippocampal and medial prefrontal pyramidal neurons. Since brain-derived neurotrophic factor (BDNF) promotes GABAergic synaptogenesis, we hypothesized that individual differences in this vulnerability may arise from differences in the link between BDNF bioavailability and FR-evoked wheel running. We tested this hypothesis in male BDNF-Val66Met knock-in mice (BDNFMet/Met), known for reduction in the activity-dependent BDNF secretion and elevated anxiety-like behaviors. We found that 1) in the absence of FR or a wheel (i.e., control), BDNFMet/Met mice are more anxious than wild-type (WT) littermates, 2) electron microscopically verified GABAergic innervations of pyramidal neurons of BDNFMet/Met mice are reduced at distal dendrites in hippocampal CA1 and medial prefrontal cortex, 3) following ABA, WT mice exhibit anxiety equal to those of the BDNFMet/Met mice and have lost GABAergic innervation along distal dendrites, 4) BDNFMet/Met mice show blunted ABA vulnerability, and 5) unexpectedly, GABAergic innervation is higher at somata of BDNFMet/Met mice than of WT. We conclude that lamina-specific GABAergic inhibition is important for regulating anxiety, whether arising from environmental stress, such as food deprivation, or genetically, such as BDNFMet/Met single nucleotide polymorphism.
Collapse
Affiliation(s)
- Yi-Wen Chen
- Center for Neural Science, New York University, New York, NY 10003, USA
| | - Olivia Surgent
- Center for Neural Science, New York University, New York, NY 10003, USA
| | - Barkha S Rana
- Center for Neural Science, New York University, New York, NY 10003, USA
| | - Francis Lee
- Department of Psychiatry, Weill Cornell Medical College, New York, NY 10065, USA
| | - Chiye Aoki
- Center for Neural Science, New York University, New York, NY 10003, USA
| |
Collapse
|
29
|
Aoki C, Chowdhury TG, Wable GS, Chen YW. Synaptic changes in the hippocampus of adolescent female rodents associated with resilience to anxiety and suppression of food restriction-evoked hyperactivity in an animal model for anorexia nervosa. Brain Res 2017; 1654:102-115. [PMID: 26779892 PMCID: PMC4947030 DOI: 10.1016/j.brainres.2016.01.019] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2015] [Accepted: 01/10/2016] [Indexed: 12/30/2022]
Abstract
Anorexia nervosa is a mental illness that emerges primarily during early adolescence, with mortality rate that is 200 times higher than that of suicide. The illness is characterized by intense fear of gaining weight, heightened anxiety, obstinate food restriction, often accompanied by excessive exercise, in spite of mounting hunger. The illness affects females nine times more often than males, suggesting an endocrine role in its etiology. Its relapse rate exceeds 25%, yet there are no accepted pharmacological treatments to prevent this. Here, we summarize studies from this laboratory that have used adolescent female rodents in activity-based anorexia (ABA), an animal model of anorexia nervosa, with the goal of identifying neurobiological underpinnings of this disease. We put forth a hypothesis that a GABAergic mechanism within the hippocampus is central to regulating an individual׳s anxiety which, in turn, strongly influences the individual׳s resilience/vulnerability to ABA. In particular, we propose that ionotropic GABAA receptors containing the subunits alpha4 and delta, are at play for exerting shunting inhibition upon hippocampal pyramidal neurons that become more excitable during ABA. Since these receptors confer insensitivity to benzodiazepines, this pharmacological profile of ABA fits with lack of report indicating efficacy of benzodiazepines in reducing the anxiety experienced by individuals with anorexia nervosa. The idea that the GABAergic system of the hippocampus regulates resilience/vulnerability to anorexia nervosa complements current opinions about the important roles of the prefrontal cortex, amygdala, striatum, gustatory pathways and feeding centers of the hypothalamus and of the neuromodulators, serotonin and dopamine, in the etiology of the disease. This article is part of a Special Issue entitled SI: Adolescent plasticity.
Collapse
Affiliation(s)
- Chiye Aoki
- Center for Neural Science, 4 Washington Place, Room 809, New York, NY 10003, United States.
| | - Tara G Chowdhury
- Center for Neural Science, 4 Washington Place, Room 809, New York, NY 10003, United States
| | - Gauri S Wable
- Center for Neural Science, 4 Washington Place, Room 809, New York, NY 10003, United States
| | - Yi-Wen Chen
- Center for Neural Science, 4 Washington Place, Room 809, New York, NY 10003, United States
| |
Collapse
|
30
|
Koh DXP, Sng JCG. HDAC1 negatively regulates Bdnf and Pvalb required for parvalbumin interneuron maturation in an experience-dependent manner. J Neurochem 2016; 139:369-380. [PMID: 27534825 DOI: 10.1111/jnc.13773] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2016] [Revised: 06/24/2016] [Accepted: 08/10/2016] [Indexed: 01/29/2023]
Abstract
During early postnatal development, neuronal circuits are sculpted by sensory experience provided by the external environment. This experience-dependent regulation of circuitry development consolidates the balance of excitatory-inhibitory (E/I) neurons in the brain. The cortical barrel-column that innervates a single principal whisker is used to provide a clear reference frame for studying the consolidation of E/I circuitry. Sensory deprivation of S1 at birth disrupts the consolidation of excitatory-inhibitory balance by decreasing inhibitory transmission of parvalbumin interneurons. The molecular mechanisms underlying this decrease in inhibition are not completely understood. Our findings show that epigenetic mechanisms, in particular histone deacetylation by histone deacetylases, negatively regulate the expression of brain-derived neurotrophic factor (Bdnf) and parvalbumin (Pvalb) genes during development, which are required for the maturation of parvalbumin interneurons. After whisker deprivation, increased histone deacetylase 1 expression and activity led to increased histone deacetylase 1 binding and decreased histone acetylation at Bdnf promoters I-IV and Pvalb promoter, resulting in the repression of Bdnf and Pvalb gene transcription. The decrease in Bdnf expression further affected parvalbumin interneuron maturation at layer II/III in S1, demonstrated by decreased parvalbumin expression, a marker for parvalbumin interneuron maturation. Knockdown of HDAC1 recovered Bdnf and Pvalb gene transcription and also prevented the decrease of inhibitory synapses accompanying whisker deprivation.
Collapse
Affiliation(s)
- Dawn X P Koh
- National University of Singapore, Graduate School of Integrative Sciences and Engineering, Singapore, Singapore.,Singapore Institute of Clinical Sciences (SICS), A*STAR, Brenner Centre for Molecular Medicine, Singapore, Singapore.,Department of Pharmacology, National University of Singapore, Yong Loo Lin School of Medicine, Singapore, Singapore
| | - Judy C G Sng
- Singapore Institute of Clinical Sciences (SICS), A*STAR, Brenner Centre for Molecular Medicine, Singapore, Singapore. .,Department of Pharmacology, National University of Singapore, Yong Loo Lin School of Medicine, Singapore, Singapore.
| |
Collapse
|
31
|
GABAergic Function as a Limiting Factor for Prefrontal Maturation during Adolescence. Trends Neurosci 2016; 39:441-448. [PMID: 27233681 DOI: 10.1016/j.tins.2016.04.010] [Citation(s) in RCA: 113] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2016] [Revised: 04/22/2016] [Accepted: 04/25/2016] [Indexed: 11/24/2022]
Abstract
Adolescence is a vulnerable period for the onset of mental illnesses including schizophrenia and affective disorders, yet the neurodevelopmental processes underlying this vulnerability remain poorly understood. The prefrontal cortex (PFC) and its local GABAergic system are thought to contribute to the core of cognitive deficits associated with such disorders. However, clinical and preclinical end-point analyses performed in adults are likely to give limited insight into the cellular mechanisms that are altered during adolescence but are only manifested in adulthood. This perspective summarizes work regarding the developmental trajectories of the GABAergic system in the PFC during adolescence to provide an insight into the increased susceptibility to psychiatric disorders during this critical developmental period.
Collapse
|
32
|
Excitation-Transcription Coupling in Parvalbumin-Positive Interneurons Employs a Novel CaM Kinase-Dependent Pathway Distinct from Excitatory Neurons. Neuron 2016; 90:292-307. [PMID: 27041500 DOI: 10.1016/j.neuron.2016.03.001] [Citation(s) in RCA: 66] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2015] [Revised: 11/23/2015] [Accepted: 02/24/2016] [Indexed: 11/20/2022]
Abstract
Properly functional CNS circuits depend on inhibitory interneurons that in turn rely upon activity-dependent gene expression for morphological development, connectivity, and excitatory-inhibitory coordination. Despite its importance, excitation-transcription coupling in inhibitory interneurons is poorly understood. We report that PV+ interneurons employ a novel CaMK-dependent pathway to trigger CREB phosphorylation and gene expression. As in excitatory neurons, voltage-gated Ca(2+) influx through CaV1 channels triggers CaM nuclear translocation via local Ca(2+) signaling. However, PV+ interneurons are distinct in that nuclear signaling is mediated by γCaMKI, not γCaMKII. CREB phosphorylation also proceeds with slow, sigmoid kinetics, rate-limited by paucity of CaMKIV, protecting against saturation of phospho-CREB in the face of higher firing rates and bigger Ca(2+) transients. Our findings support the generality of CaM shuttling to drive nuclear CaMK activity, and they are relevant to disease pathophysiology, insofar as dysfunction of PV+ interneurons and molecules underpinning their excitation-transcription coupling both relate to neuropsychiatric disease.
Collapse
|
33
|
Hoppenrath K, Härtig W, Funke K. Intermittent Theta-Burst Transcranial Magnetic Stimulation Alters Electrical Properties of Fast-Spiking Neocortical Interneurons in an Age-Dependent Fashion. Front Neural Circuits 2016; 10:22. [PMID: 27065812 PMCID: PMC4811908 DOI: 10.3389/fncir.2016.00022] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2015] [Accepted: 03/13/2016] [Indexed: 11/13/2022] Open
Abstract
Modulation of human cortical excitability by repetitive transcranial magnetic stimulation (rTMS) appears to be in part related to changed activity of inhibitory systems. Our own studies showed that intermittent theta-burst stimulation (iTBS) applied via rTMS to rat cortex primarily affects the parvalbumin-expressing (PV) fast-spiking interneurons (FSIs), evident via a strongly reduced PV expression. We further found the iTBS effect on PV to be age-dependent since no reduction in PV could be induced before the perineuronal nets (PNNs) of FSIs start to grow around postnatal day (PD) 30. To elucidate possible iTBS-induced changes in the electrical properties of FSIs and cortical network activity during cortical critical period, we performed ex vivo-in vitro whole-cell patch clamp recordings from pre-labeled FSIs in the current study. FSIs of verum iTBS-treated rats displayed a higher excitability than sham-treated controls at PD29-38, evident as higher rates of induced action potential firing at low current injections (100-200 pA) and a more depolarized resting membrane potential. This effect was absent in younger (PD26-28) and older animals (PD40-62). Slices of verum iTBS-treated rats further showed higher rates of spontaneous excitatory postsynaptic currents (sEPSCs). Based on these and previous findings we conclude that FSIs are particularly sensitive to TBS during early cortical development, when FSIs show an activity-driven step of maturation which is paralleled by intense growth of the PNNs and subsequent closure of the cortical critical period. Although to be proven further, rTMS may be a possible early intervention to compensate for hypo-activity related mal-development of cortical neuronal circuits.
Collapse
Affiliation(s)
- Kathrin Hoppenrath
- Department of Neurophysiology, Medical Faculty, Ruhr-University BochumBochum, Germany; Rottendorf Pharma GmbHEnnigerloh, Germany
| | - Wolfgang Härtig
- Pathophysiology of Neuroglia, Paul Flechsig Institute for Brain Research, University of Leipzig Leipzig, Germany
| | - Klaus Funke
- Department of Neurophysiology, Medical Faculty, Ruhr-University Bochum Bochum, Germany
| |
Collapse
|
34
|
Whisker Deprivation Drives Two Phases of Inhibitory Synapse Weakening in Layer 4 of Rat Somatosensory Cortex. PLoS One 2016; 11:e0148227. [PMID: 26840956 PMCID: PMC4740487 DOI: 10.1371/journal.pone.0148227] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2015] [Accepted: 01/14/2016] [Indexed: 01/15/2023] Open
Abstract
Inhibitory synapse development in sensory neocortex is experience-dependent, with sustained sensory deprivation yielding fewer and weaker inhibitory synapses. Whether this represents arrest of synapse maturation, or a more complex set of processes, is unclear. To test this, we measured the dynamics of inhibitory synapse development in layer 4 of rat somatosensory cortex (S1) during continuous whisker deprivation from postnatal day 7, and in age-matched controls. In deprived columns, spontaneous miniature inhibitory postsynaptic currents (mIPSCs) and evoked IPSCs developed normally until P15, when IPSC amplitude transiently decreased, recovering by P16 despite ongoing deprivation. IPSCs remained normal until P22, when a second, sustained phase of weakening began. Delaying deprivation onset by 5 days prevented the P15 weakening. Both early and late phase weakening involved measurable reduction in IPSC amplitude relative to prior time points. Thus, deprivation appears to drive two distinct phases of active IPSC weakening, rather than simple arrest of synapse maturation.
Collapse
|
35
|
Singer W, Geisler HS, Panford-Walsh R, Knipper M. Detection of Excitatory and Inhibitory Synapses in the Auditory System Using Fluorescence Immunohistochemistry and High-Resolution Fluorescence Microscopy. Methods Mol Biol 2016; 1427:263-76. [PMID: 27259932 DOI: 10.1007/978-1-4939-3615-1_15] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
In sensory systems, a balanced excitatory and inhibitory circuit along the ascending pathway is not only important for the establishment of topographically ordered connections from the periphery to the cortex but also for temporal precision of signal processing. The accomplishment of spatial and temporal cortical resolution in the central nervous system is a process that is likely initiated by the first sensory experiences that drive a period of increased intracortical inhibition. In the auditory system, the time of first sensory experience is also the period in which a reorganization of cochlear efferent and afferent fibers occurs leading to the mature innervation of inner and outer hair cells. This mature hair cell innervation is the basis of accurate sound processing along the ascending pathway up to the auditory cortex. We describe here, a protocol for detecting excitatory and inhibitory marker proteins along the ascending auditory pathway, which could be a useful tool for detecting changes in auditory signal processing during various forms of hearing disorders. Our protocol uses fluorescence immunohistochemistry in combination with high-resolution fluorescence microscopy in cochlear and brain tissue.
Collapse
Affiliation(s)
- Wibke Singer
- Department of Otolaryngology, Hearing Research Centre Tübingen (THRC), Molecular Physiology of Hearing, ENT Clinic, University of Tübingen, Elfriede-Aulhorn-Str. 5, Tübingen, 72076, Germany
| | - Hyun-Soon Geisler
- Department of Otolaryngology, Hearing Research Centre Tübingen (THRC), Molecular Physiology of Hearing, ENT Clinic, University of Tübingen, Elfriede-Aulhorn-Str. 5, Tübingen, 72076, Germany
| | - Rama Panford-Walsh
- Department of Otolaryngology, Hearing Research Centre Tübingen (THRC), Molecular Physiology of Hearing, ENT Clinic, University of Tübingen, Elfriede-Aulhorn-Str. 5, Tübingen, 72076, Germany.,DNA Genotek Inc., Ottawa, ON, Canada
| | - Marlies Knipper
- Department of Otolaryngology, Hearing Research Centre Tübingen (THRC), Molecular Physiology of Hearing, ENT Clinic, University of Tübingen, Elfriede-Aulhorn-Str. 5, Tübingen, 72076, Germany.
| |
Collapse
|
36
|
Nelson SB, Valakh V. Excitatory/Inhibitory Balance and Circuit Homeostasis in Autism Spectrum Disorders. Neuron 2015; 87:684-98. [PMID: 26291155 DOI: 10.1016/j.neuron.2015.07.033] [Citation(s) in RCA: 696] [Impact Index Per Article: 77.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Autism spectrum disorders (ASDs) and related neurological disorders are associated with mutations in many genes affecting the ratio between neuronal excitation and inhibition. However, understanding the impact of these mutations on network activity is complicated by the plasticity of these networks, making it difficult in many cases to separate initial deficits from homeostatic compensation. Here we explore the contrasting evidence for primary defects in inhibition or excitation in ASDs and attempt to integrate the findings in terms of the brain's ability to maintain functional homeostasis.
Collapse
Affiliation(s)
- Sacha B Nelson
- Department of Biology and Center for Behavioral Genomics, Brandeis University, 415 South Street, Waltham, MA 02454, USA.
| | - Vera Valakh
- Department of Biology and Center for Behavioral Genomics, Brandeis University, 415 South Street, Waltham, MA 02454, USA
| |
Collapse
|
37
|
Ehrlich DE, Josselyn SA. Plasticity-related genes in brain development and amygdala-dependent learning. GENES BRAIN AND BEHAVIOR 2015; 15:125-43. [PMID: 26419764 DOI: 10.1111/gbb.12255] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/07/2015] [Revised: 09/12/2015] [Accepted: 09/14/2015] [Indexed: 12/31/2022]
Abstract
Learning about motivationally important stimuli involves plasticity in the amygdala, a temporal lobe structure. Amygdala-dependent learning involves a growing number of plasticity-related signaling pathways also implicated in brain development, suggesting that learning-related signaling in juveniles may simultaneously influence development. Here, we review the pleiotropic functions in nervous system development and amygdala-dependent learning of a signaling pathway that includes brain-derived neurotrophic factor (BDNF), extracellular signaling-related kinases (ERKs) and cyclic AMP-response element binding protein (CREB). Using these canonical, plasticity-related genes as an example, we discuss the intersection of learning-related and developmental plasticity in the immature amygdala, when aversive and appetitive learning may influence the developmental trajectory of amygdala function. We propose that learning-dependent activation of BDNF, ERK and CREB signaling in the immature amygdala exaggerates and accelerates neural development, promoting amygdala excitability and environmental sensitivity later in life.
Collapse
Affiliation(s)
- D E Ehrlich
- Department of Neuroscience and Physiology, Neuroscience Institute, NYU Langone Medical Center, New York, NY, USA.,Department of Otolaryngology, NYU Langone School of Medicine, New York, NY, USA
| | - S A Josselyn
- Program in Neurosciences & Mental Health, Hospital for Sick Children, Toronto, ON, Canada.,Department of Psychology, University of Toronto, Toronto, ON, Canada.,Institute of Medical Sciences, University of Toronto, Toronto, ON, Canada.,Department of Physiology, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
38
|
Chumak T, Rüttiger L, Lee SC, Campanelli D, Zuccotti A, Singer W, Popelář J, Gutsche K, Geisler HS, Schraven SP, Jaumann M, Panford-Walsh R, Hu J, Schimmang T, Zimmermann U, Syka J, Knipper M. BDNF in Lower Brain Parts Modifies Auditory Fiber Activity to Gain Fidelity but Increases the Risk for Generation of Central Noise After Injury. Mol Neurobiol 2015; 53:5607-27. [PMID: 26476841 PMCID: PMC5012152 DOI: 10.1007/s12035-015-9474-x] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2015] [Accepted: 10/05/2015] [Indexed: 11/24/2022]
Abstract
For all sensory organs, the establishment of spatial and temporal cortical resolution is assumed to be initiated by the first sensory experience and a BDNF-dependent increase in intracortical inhibition. To address the potential of cortical BDNF for sound processing, we used mice with a conditional deletion of BDNF in which Cre expression was under the control of the Pax2 or TrkC promoter. BDNF deletion profiles between these mice differ in the organ of Corti (BDNFPax2-KO) versus the auditory cortex and hippocampus (BDNFTrkC-KO). We demonstrate that BDNFPax2-KO but not BDNFTrkC-KO mice exhibit reduced sound-evoked suprathreshold ABR waves at the level of the auditory nerve (wave I) and inferior colliculus (IC) (wave IV), indicating that BDNF in lower brain regions but not in the auditory cortex improves sound sensitivity during hearing onset. Extracellular recording of IC neurons of BDNFPax2 mutant mice revealed that the reduced sensitivity of auditory fibers in these mice went hand in hand with elevated thresholds, reduced dynamic range, prolonged latency, and increased inhibitory strength in IC neurons. Reduced parvalbumin-positive contacts were found in the ascending auditory circuit, including the auditory cortex and hippocampus of BDNFPax2-KO, but not of BDNFTrkC-KO mice. Also, BDNFPax2-WT but not BDNFPax2-KO mice did lose basal inhibitory strength in IC neurons after acoustic trauma. These findings suggest that BDNF in the lower parts of the auditory system drives auditory fidelity along the entire ascending pathway up to the cortex by increasing inhibitory strength in behaviorally relevant frequency regions. Fidelity and inhibitory strength can be lost following auditory nerve injury leading to diminished sensory outcome and increased central noise.
Collapse
Affiliation(s)
- Tetyana Chumak
- Department of Auditory Neuroscience, Institute of Experimental Medicine, Academy of Sciences of the Czech Republic, Vídeňská 1083, 142 20, Prague, Czech Republic
| | - Lukas Rüttiger
- Department of Otolaryngology, Hearing Research Centre Tübingen, Molecular Physiology of Hearing, University of Tübingen, Elfriede-Aulhorn-Str. 5, 72076, Tübingen, Germany
| | - Sze Chim Lee
- Department of Otolaryngology, Hearing Research Centre Tübingen, Molecular Physiology of Hearing, University of Tübingen, Elfriede-Aulhorn-Str. 5, 72076, Tübingen, Germany
| | - Dario Campanelli
- Department of Otolaryngology, Hearing Research Centre Tübingen, Molecular Physiology of Hearing, University of Tübingen, Elfriede-Aulhorn-Str. 5, 72076, Tübingen, Germany
| | - Annalisa Zuccotti
- Department of Otolaryngology, Hearing Research Centre Tübingen, Molecular Physiology of Hearing, University of Tübingen, Elfriede-Aulhorn-Str. 5, 72076, Tübingen, Germany.,Department of Clinical Neurobiology, University Hospital and DKFZ Heidelberg, In Neuenheimer Feld 280, 69120, Heidelberg, Germany
| | - Wibke Singer
- Department of Otolaryngology, Hearing Research Centre Tübingen, Molecular Physiology of Hearing, University of Tübingen, Elfriede-Aulhorn-Str. 5, 72076, Tübingen, Germany
| | - Jiří Popelář
- Department of Auditory Neuroscience, Institute of Experimental Medicine, Academy of Sciences of the Czech Republic, Vídeňská 1083, 142 20, Prague, Czech Republic
| | - Katja Gutsche
- Instituto de Biologíay Genética Molecular, Universidad de Valladolid y Consejo Superior de Investigaciones Científicas, E-47003, Valladolid, Spain
| | - Hyun-Soon Geisler
- Department of Otolaryngology, Hearing Research Centre Tübingen, Molecular Physiology of Hearing, University of Tübingen, Elfriede-Aulhorn-Str. 5, 72076, Tübingen, Germany
| | - Sebastian Philipp Schraven
- Department of Otolaryngology, Plastic, Aesthetic and Reconstructive Head and Neck Surgery, Comprehensive Hearing Center, University of Würzburg, Josef-Schneider-Straße 11, 97080, Würzburg, Germany
| | - Mirko Jaumann
- Department of Otolaryngology, Hearing Research Centre Tübingen, Molecular Physiology of Hearing, University of Tübingen, Elfriede-Aulhorn-Str. 5, 72076, Tübingen, Germany
| | | | - Jing Hu
- Centre for Integrative Neuroscience, University of Tübingen, Otfried-Müller-Straße 25, 72076, Tübingen, Germany
| | - Thomas Schimmang
- Instituto de Biologíay Genética Molecular, Universidad de Valladolid y Consejo Superior de Investigaciones Científicas, E-47003, Valladolid, Spain
| | - Ulrike Zimmermann
- Department of Otolaryngology, Hearing Research Centre Tübingen, Molecular Physiology of Hearing, University of Tübingen, Elfriede-Aulhorn-Str. 5, 72076, Tübingen, Germany
| | - Josef Syka
- Department of Auditory Neuroscience, Institute of Experimental Medicine, Academy of Sciences of the Czech Republic, Vídeňská 1083, 142 20, Prague, Czech Republic
| | - Marlies Knipper
- Department of Otolaryngology, Hearing Research Centre Tübingen, Molecular Physiology of Hearing, University of Tübingen, Elfriede-Aulhorn-Str. 5, 72076, Tübingen, Germany.
| |
Collapse
|
39
|
Tong CW, Wang ZL, Li P, Zhu H, Chen CY, Hua TM. Effects of senescence on the expression of BDNF and TrkB receptor in the lateral geniculate nucleus of cats. DONG WU XUE YAN JIU = ZOOLOGICAL RESEARCH 2015; 36:48-53. [PMID: 25730461 DOI: 10.13918/j.issn.2095-8137.2015.1.48] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
To explore the neural mechanisms mediating aging-related visual function declines, we compared the expressions of brain-derived neurotrophic factor (BDNF) and its high affinity receptor-tyrosine kinase B (TrkB) between young and old adult cats. Nissl staining was used to display neurons in each layer of the lateral geniculate nucleus (LGN). The BDNF- and TrkB receptor-immunoreactive neurons were labeled immunohistochemically, observed under optical microscope and photographed. Their neuronal density and immunoreactive intensity were measured. Results showed that the mean density of the Nissl stained neurons in each LGN layer were comparable between old and young adult cats, and their BDNF and TrkB proteins were widely expressed in all LGN layers. However, compared with young adult cats, both the density and optical absorbance intensity of BDNF- and TrkB-immunoreactive cells in each LGN layer in old cats were significantly decreased. These findings indicate that the decreased expressions of BDNF and TrkB proteins in the LGN may be an important factor inducing the compromised inhibition in the central visual nucleus and the functional visual decline in senescent individuals.
Collapse
Affiliation(s)
- Chuan-Wang Tong
- College of Life Science, Anhui Normal University, Wuhu 241000, China;College of Biological Engineering, Wuhu Institute of Technology, Wuhu 241000, China
| | - Zi-Lu Wang
- College of Life Science, Anhui Normal University, Wuhu 241000, China
| | - Peng Li
- College of Life Science, Anhui Normal University, Wuhu 241000, China
| | - Hui Zhu
- College of Life Science, Anhui Normal University, Wuhu 241000, China
| | - Cui-Yun Chen
- College of Life Science, Anhui Normal University, Wuhu 241000, China
| | - Tian-Miao Hua
- College of Life Science, Anhui Normal University, Wuhu 241000, China.
| |
Collapse
|
40
|
Knipper M, Panford-Walsh R, Singer W, Rüttiger L, Zimmermann U. Specific synaptopathies diversify brain responses and hearing disorders: you lose the gain from early life. Cell Tissue Res 2015; 361:77-93. [PMID: 25843689 PMCID: PMC4487345 DOI: 10.1007/s00441-015-2168-x] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2015] [Accepted: 03/05/2015] [Indexed: 01/08/2023]
Abstract
Before hearing onset, inner hair cell (IHC) maturation proceeds under the influence of spontaneous Ca(2+) action potentials (APs). The temporal signature of the IHC Ca(2+) AP is modified through an efferent cholinergic feedback from the medial olivocochlear bundle (MOC) and drives the IHC pre- and post-synapse phenotype towards low spontaneous (spike) rate (SR), high-threshold characteristics. With sensory experience, the IHC pre- and post-synapse phenotype matures towards the instruction of low-SR, high-threshold and of high-SR, low-threshold auditory fiber characteristics. Corticosteroid feedback together with local brain-derived nerve growth factor (BDNF) and catecholaminergic neurotransmitters (dopamine) might be essential for this developmental step. In this review, we address the question of whether the control of low-SR and high-SR fiber characteristics is linked to various degrees of vulnerability of auditory fibers in the mature system. In particular, we examine several IHC synaptopathies in the context of various hearing disorders and exemplified shortfalls before and after hearing onset.
Collapse
Affiliation(s)
- Marlies Knipper
- Department of Otolaryngology, Head and Neck Surgery, Tübingen Hearing Research Center (THRC), Molecular Physiology of Hearing, University of Tübingen, Elfriede-Aulhorn-Straße 5, 72076 Tübingen, Germany
| | | | - Wibke Singer
- Department of Otolaryngology, Head and Neck Surgery, Tübingen Hearing Research Center (THRC), Molecular Physiology of Hearing, University of Tübingen, Elfriede-Aulhorn-Straße 5, 72076 Tübingen, Germany
| | - Lukas Rüttiger
- Department of Otolaryngology, Head and Neck Surgery, Tübingen Hearing Research Center (THRC), Molecular Physiology of Hearing, University of Tübingen, Elfriede-Aulhorn-Straße 5, 72076 Tübingen, Germany
| | - Ulrike Zimmermann
- Department of Otolaryngology, Head and Neck Surgery, Tübingen Hearing Research Center (THRC), Molecular Physiology of Hearing, University of Tübingen, Elfriede-Aulhorn-Straße 5, 72076 Tübingen, Germany
| |
Collapse
|
41
|
Castillo-Padilla DV, Funke K. Effects of chronic iTBS-rTMS and enriched environment on visual cortex early critical period and visual pattern discrimination in dark-reared rats. Dev Neurobiol 2015; 76:19-33. [DOI: 10.1002/dneu.22296] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2015] [Revised: 04/14/2015] [Accepted: 04/14/2015] [Indexed: 11/11/2022]
Affiliation(s)
- Diana V. Castillo-Padilla
- Clinical Research Subdivision; National Institute of Psychiatry Ramón de la Fuente Muñiz; México D.F 14370 México
- Department of Neurophysiology; Medical Faculty; Ruhr-University Bochum; 44780 Bochum Germany
| | - Klaus Funke
- Department of Neurophysiology; Medical Faculty; Ruhr-University Bochum; 44780 Bochum Germany
| |
Collapse
|
42
|
Chen YW, Wable GS, Chowdhury TG, Aoki C. Enlargement of Axo-Somatic Contacts Formed by GAD-Immunoreactive Axon Terminals onto Layer V Pyramidal Neurons in the Medial Prefrontal Cortex of Adolescent Female Mice Is Associated with Suppression of Food Restriction-Evoked Hyperactivity and Resilience to Activity-Based Anorexia. Cereb Cortex 2015; 26:2574-89. [PMID: 25979087 DOI: 10.1093/cercor/bhv087] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Many, but not all, adolescent female mice that are exposed to a running wheel while food restricted (FR) become excessive wheel runners, choosing to run even during the hours of food availability, to the point of death. This phenomenon is called activity-based anorexia (ABA). We used electron microscopic immunocytochemistry to ask whether individual differences in ABA resilience may correlate with the lengths of axo-somatic contacts made by GABAergic axon terminals onto layer 5 pyramidal neurons (L5P) in the prefrontal cortex. Contact lengths were, on average, 40% greater for the ABA-induced mice, relative to controls. Correspondingly, the proportion of L5P perikaryal plasma membrane contacted by GABAergic terminals was 45% greater for the ABA mice. Contact lengths in the anterior cingulate cortex correlated negatively and strongly with the overall wheel activity after FR (R = -0.87, P < 0.01), whereas those in the prelimbic cortex correlated negatively with wheel running specifically during the hours of food availability of the FR days (R = -0.84, P < 0.05). These negative correlations support the idea that increases in the glutamic acid decarboxylase (GAD) terminal contact lengths onto L5P contribute toward ABA resilience through suppression of wheel running, a behavior that is intrinsically rewarding and helpful for foraging but maladaptive within a cage.
Collapse
Affiliation(s)
- Yi-Wen Chen
- Center for Neural Science, New York University, New York, NY 10003, USA
| | | | | | - Chiye Aoki
- Center for Neural Science, New York University, New York, NY 10003, USA
| |
Collapse
|
43
|
Nosheny RL, Belichenko PV, Busse BL, Weissmiller AM, Dang V, Das D, Fahimi A, Salehi A, Smith SJ, Mobley WC. Increased cortical synaptic activation of TrkB and downstream signaling markers in a mouse model of Down Syndrome. Neurobiol Dis 2015; 77:173-90. [PMID: 25753471 DOI: 10.1016/j.nbd.2015.02.022] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2014] [Revised: 02/15/2015] [Accepted: 02/25/2015] [Indexed: 01/01/2023] Open
Abstract
Down Syndrome (DS), trisomy 21, is characterized by synaptic abnormalities and cognitive deficits throughout the lifespan and with development of Alzheimer's disease (AD) neuropathology and progressive cognitive decline in adults. Synaptic abnormalities are also present in the Ts65Dn mouse model of DS, but which synapses are affected and the mechanisms underlying synaptic dysfunction are unknown. Here we show marked increases in the levels and activation status of TrkB and associated signaling proteins in cortical synapses in Ts65Dn mice. Proteomic analysis at the single synapse level of resolution using array tomography (AT) uncovered increased colocalization of activated TrkB with signaling endosome related proteins, and demonstrated increased TrkB signaling. The extent of increases in TrkB signaling differed in each of the cortical layers examined and with respect to the type of synapse, with the most marked increases seen in inhibitory synapses. These findings are evidence of markedly abnormal TrkB-mediated signaling in synapses. They raise the possibility that dysregulated TrkB signaling contributes to synaptic dysfunction and cognitive deficits in DS.
Collapse
Affiliation(s)
- R L Nosheny
- Department of Neurosciences, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA; Department of Molecular and Cellular Physiology, Stanford University, 279 Campus Drive, Stanford, CA 94305, USA.
| | - P V Belichenko
- Department of Neurosciences, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | - B L Busse
- Department of Molecular and Cellular Physiology, Stanford University, 279 Campus Drive, Stanford, CA 94305, USA
| | - A M Weissmiller
- Department of Neurosciences, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | - V Dang
- Department of Psychiatry & Behavioral Sciences, Stanford Medical School, VA Palo Alto Health Care System, 3801 Miranda Avenue, Palo Alto, CA 94304, USA
| | - D Das
- Department of Psychiatry & Behavioral Sciences, Stanford Medical School, VA Palo Alto Health Care System, 3801 Miranda Avenue, Palo Alto, CA 94304, USA
| | - A Fahimi
- Department of Psychiatry & Behavioral Sciences, Stanford Medical School, VA Palo Alto Health Care System, 3801 Miranda Avenue, Palo Alto, CA 94304, USA
| | - A Salehi
- Department of Psychiatry & Behavioral Sciences, Stanford Medical School, VA Palo Alto Health Care System, 3801 Miranda Avenue, Palo Alto, CA 94304, USA
| | - S J Smith
- Department of Molecular and Cellular Physiology, Stanford University, 279 Campus Drive, Stanford, CA 94305, USA
| | - W C Mobley
- Department of Neurosciences, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
| |
Collapse
|
44
|
Npas4 expression in two experimental models of the barrel cortex plasticity. Neural Plast 2015; 2015:175701. [PMID: 25785202 PMCID: PMC4345254 DOI: 10.1155/2015/175701] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2014] [Accepted: 01/29/2015] [Indexed: 12/11/2022] Open
Abstract
Npas4 has recently been identified as an important factor in brain plasticity, particularly in mechanisms of inhibitory control. Little is known about Npas4 expression in terms of cortical plasticity. In the present study expressions of Npas4 and the archetypal immediate early gene (IEG) c-Fos were investigated in the barrel cortex of mice after sensory deprivation (sparing one row of whiskers for 7 days) or sensory conditioning (pairing stimulation of one row of whiskers with aversive stimulus). Laser microdissection of individual barrel rows allowed for analysis of IEGs expression precisely in deprived and nondeprived barrels (in deprivation study) or stimulated and nonstimulated barrels (in conditioning study). Cortex activation by sensory conditioning was found to upregulate the expression of both Npas4 and c-Fos. Reorganization of cortical circuits triggered by removal of selected rows of whiskers strongly affected c-Fos but not Npas4 expression. We hypothesize that increased inhibitory synaptogenesis observed previously after conditioning may be mediated by Npas4 expression.
Collapse
|
45
|
Itahashi M, Abe H, Tanaka T, Mizukami S, Kikuchihara Y, Yoshida T, Shibutani M. Maternal exposure to 3,3’-iminodipropionitrile targets late-stage differentiation of hippocampal granule cell lineages to affect brain-derived neurotrophic factor signaling and interneuron subpopulations in rat offspring. J Appl Toxicol 2014; 35:884-94. [DOI: 10.1002/jat.3086] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2014] [Revised: 09/23/2014] [Accepted: 09/24/2014] [Indexed: 01/13/2023]
Affiliation(s)
- Megu Itahashi
- Laboratory of Veterinary Pathology; Tokyo University of Agriculture and Technology; 3-5-8 Saiwai-cho, Fuchu-shi Tokyo 183-8509 Japan
- Pathogenetic Veterinary Science, United Graduate School of Veterinary Sciences; Gifu University; 1-1 Yanagido, Gifu-shi Gifu 501-1193 Japan
| | - Hajime Abe
- Laboratory of Veterinary Pathology; Tokyo University of Agriculture and Technology; 3-5-8 Saiwai-cho, Fuchu-shi Tokyo 183-8509 Japan
- Pathogenetic Veterinary Science, United Graduate School of Veterinary Sciences; Gifu University; 1-1 Yanagido, Gifu-shi Gifu 501-1193 Japan
| | - Takeshi Tanaka
- Laboratory of Veterinary Pathology; Tokyo University of Agriculture and Technology; 3-5-8 Saiwai-cho, Fuchu-shi Tokyo 183-8509 Japan
- Pathogenetic Veterinary Science, United Graduate School of Veterinary Sciences; Gifu University; 1-1 Yanagido, Gifu-shi Gifu 501-1193 Japan
| | - Sayaka Mizukami
- Laboratory of Veterinary Pathology; Tokyo University of Agriculture and Technology; 3-5-8 Saiwai-cho, Fuchu-shi Tokyo 183-8509 Japan
- Pathogenetic Veterinary Science, United Graduate School of Veterinary Sciences; Gifu University; 1-1 Yanagido, Gifu-shi Gifu 501-1193 Japan
| | - Yoh Kikuchihara
- Laboratory of Veterinary Pathology; Tokyo University of Agriculture and Technology; 3-5-8 Saiwai-cho, Fuchu-shi Tokyo 183-8509 Japan
| | - Toshinori Yoshida
- Laboratory of Veterinary Pathology; Tokyo University of Agriculture and Technology; 3-5-8 Saiwai-cho, Fuchu-shi Tokyo 183-8509 Japan
| | - Makoto Shibutani
- Laboratory of Veterinary Pathology; Tokyo University of Agriculture and Technology; 3-5-8 Saiwai-cho, Fuchu-shi Tokyo 183-8509 Japan
| |
Collapse
|
46
|
Sun QQ, Zhang Z, Sun J, Nair AS, Petrus DP, Zhang C. Functional and structural specific roles of activity-driven BDNF within circuits formed by single spiny stellate neurons of the barrel cortex. Front Cell Neurosci 2014; 8:372. [PMID: 25414642 PMCID: PMC4222225 DOI: 10.3389/fncel.2014.00372] [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: 09/27/2014] [Accepted: 10/21/2014] [Indexed: 11/13/2022] Open
Abstract
Brain derived neurotrophic factor (BDNF) plays key roles in several neurodevelopmental disorders and actions of pharmacological treatments. However, it is unclear how specific BDNF’s effects are on different circuit components. Current studies have largely focused on the role of BDNF in modification of synaptic development. The precise roles of BDNF in the refinement of a functional circuit in vivo remain unclear. Val66Met polymorphism of BDNF may be associated with increased risk for cognitive impairments and is mediated at least in part by activity-dependent trafficking and/or secretion of BDNF. Using mutant mice that lacked activity-driven BDNF expression (bdnf-KIV), we previously reported that experience regulation of the cortical GABAergic network is mediated by activity-driven BDNF expression. Here, we demonstrate that activity-driven BDNF’s effects on circuits formed by the layer IV spiny stellate cells are highly specific. Structurally, dendritic but not axonal morphology was altered in the mutant. Physiologically, GABAergic but not glutamatergic synapses were severely affected. The effects on GABA transmission occurs via presynaptic alteration of calcium-dependent release probability. These results suggest that neuronal activity through activity-driven BDNF expression, can selectively regulate specific features of layer IV circuits in vivo. We postulate that the role of activity-dependent BDNF is to modulate the computational ability of circuits that relate to the gain control (i.e., feed-forward inhibition); whereas the basic wiring of circuits relevant to the sensory pathway is spared. Gain control modulation within cortical circuits has broad impact on cognitive processing and brain state-transitions. Cognitive behavior and mode is determined by brain states, thus the studying of circuit alteration by endogenous BDNF provides insights into the cellular and molecular mechanisms of diseases mediated by BDNF.
Collapse
Affiliation(s)
- Qian-Quan Sun
- Department of Zoology and Physiology, University of Wyoming, Laramie, WY USA
| | - Zhi Zhang
- Department of Zoology and Physiology, University of Wyoming, Laramie, WY USA
| | - June Sun
- Department of Zoology and Physiology, University of Wyoming, Laramie, WY USA
| | - Anand S Nair
- Department of Zoology and Physiology, University of Wyoming, Laramie, WY USA
| | - Dan P Petrus
- Department of Zoology and Physiology, University of Wyoming, Laramie, WY USA
| | - Chunzhao Zhang
- Department of Zoology and Physiology, University of Wyoming, Laramie, WY USA
| |
Collapse
|
47
|
Rebound potentiation of inhibition in juvenile visual cortex requires vision-induced BDNF expression. J Neurosci 2014; 34:10770-9. [PMID: 25100608 DOI: 10.1523/jneurosci.5454-13.2014] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
The developmental increase in the strength of inhibitory synaptic circuits defines the time window of the critical period for plasticity in sensory cortices. Conceptually, plasticity of inhibitory synapses is an attractive mechanism to allow for homeostatic adaptation to the sensory environment. However, a brief duration of visual deprivation that causes maximal change in excitatory synapses produces minimal change in inhibitory synaptic transmission. Here we examined developmental and experience-dependent changes in inhibition by measuring miniature IPSCs (mIPSCs) in layer 2/3 pyramidal neurons of mouse visual cortex. During development from postnatal day 21 (P21) to P35, GABAA receptor function changed from fewer higher-conductance channels to more numerous lower-conductance channels without altering the average mIPSC amplitude. Although a week of visual deprivation did not alter the average mIPSC amplitude, a subsequent 2 h exposure to light produced a rapid rebound potentiation. This form of plasticity is restricted to a critical period before the developmental change in GABAergic synaptic properties is completed, and hence is absent by P35. Visual experience-dependent rebound potentiation of mIPSCs is accompanied by an increase in the open channel number and requires activity-dependent transcription of brain-derived neurotrophic factor (BDNF). Mice lacking BDNF transcription through promoter IV did not show developmental changes in inhibition and lacked rebound potentiation. Our results suggest that sensory experience may have distinct functional consequences in normal versus deprived sensory cortices, and that experience-dependent BDNF expression controls the plasticity of inhibitory synaptic transmission particularly when recovering vision during the critical period.
Collapse
|
48
|
Is there a relationship between brain-derived neurotrophic factor for driving neuronal auditory circuits with onset of auditory function and the changes following cochlear injury or during aging? Neuroscience 2014; 283:26-43. [PMID: 25064058 DOI: 10.1016/j.neuroscience.2014.07.025] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2014] [Revised: 07/15/2014] [Accepted: 07/17/2014] [Indexed: 01/06/2023]
Abstract
Brain-derived neurotrophic factor, BDNF, is one of the most important neurotrophic factors acting in the peripheral and central nervous system. In the auditory system its function was initially defined by using constitutive knockout mouse mutants and shown to be essential for survival of neurons and afferent innervation of hair cells in the peripheral auditory system. Further examination of BDNF null mutants also revealed a more complex requirement during re-innervation processes involving the efferent system of the cochlea. Using adult mouse mutants defective in BDNF signaling, it could be shown that a tonotopical gradient of BDNF expression within cochlear neurons is required for maintenance of a specific spatial innervation pattern of outer hair cells and inner hair cells. Additionally, BDNF is required for maintenance of voltage-gated potassium channels (KV) in cochlear neurons, which may form part of a maturation step within the ascending auditory pathway with onset of hearing and might be essential for cortical acuity of sound-processing and experience-dependent plasticity. A presumptive harmful role of BDNF during acoustic trauma and consequences of a loss of cochlear BDNF during aging are discussed in the context of a partial reversion of this maturation step. We compare the potentially beneficial and harmful roles of BDNF for the mature auditory system with those BDNF functions known in other sensory circuits, such as the vestibular, visual, olfactory, or somatosensory system.
Collapse
|
49
|
Branchi I, Cirulli F. Early experiences: Building up the tools to face the challenges of adult life. Dev Psychobiol 2014; 56:1661-74. [DOI: 10.1002/dev.21235] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2013] [Accepted: 06/06/2014] [Indexed: 12/17/2022]
Affiliation(s)
- Igor Branchi
- Section of Behavioural Neurosciences; Department of Cell Biology and Neurosciences; Istituto Superiore di Sanità, Viale Regina Elena 299 00161 Rome Italy
- Institute of Anatomy; University of Zurich; 190 Winterthurestrasse 8057 Zurich Switzerland
| | - Francesca Cirulli
- Section of Behavioural Neurosciences; Department of Cell Biology and Neurosciences; Istituto Superiore di Sanità, Viale Regina Elena 299 00161 Rome Italy
| |
Collapse
|
50
|
Li J, Tang Y, Wang Y, Tang R, Jiang W, Yang GY, Gao WQ. Neurovascular recovery via co-transplanted neural and vascular progenitors leads to improved functional restoration after ischemic stroke in rats. Stem Cell Reports 2014; 3:101-14. [PMID: 25068125 PMCID: PMC4110773 DOI: 10.1016/j.stemcr.2014.05.012] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2013] [Revised: 05/15/2014] [Accepted: 05/15/2014] [Indexed: 12/23/2022] Open
Abstract
The concept of the “neurovascular unit,” emphasizing the interactions between neural and vascular components in the brain, raised the notion that neural progenitor cell (NPC) transplantation therapy aimed at neural repair may be insufficient for the treatment of ischemic stroke. Here, we demonstrate that enhanced neurovascular recovery via cotransplantation of NPCs and embryonic stem cell-derived vascular progenitor cells (VPCs) in a rat stroke model is correlated with improved functional recovery after stroke. We found that cotransplantation promoted the survival, migration, differentiation, and maturation of neuronal and vascular cells derived from the cotransplanted progenitors. Furthermore, it triggered an increased generation of VEGF-, BDNF-, and IGF1-expressing neural cells derived from the grafted NPCs. Consistently, compared with transplantation of NPCs alone, cotransplantation more effectively improved the neurobehavioral deficits and attenuated the infarct volume. Thus, cotransplantation of NPCs and VPCs represents a more effective therapeutic strategy for the treatment of stroke than transplantation of NPCs alone. Neural and vascular progenitor cell cotransplantation therapy for ischemic stroke Better neurovascular recovery by cotransplanted progenitor cells in the infarct area Better infarction reduction and functional restoration by cotransplantation Neurovascular recovery likely mediated by neural production of growth/trophic factors
Collapse
Affiliation(s)
- Jia Li
- State Key Laboratory of Oncogenes and Related Genes, Renji-Med X Clinical Stem Cell Research Center, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China ; School of Biomedical Engineering and Med-X Research Institute, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Yaohui Tang
- School of Biomedical Engineering and Med-X Research Institute, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Yongting Wang
- School of Biomedical Engineering and Med-X Research Institute, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Rongbiao Tang
- Department of Radiology, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Weifang Jiang
- Institute of Neuroscience, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200030, China
| | - Guo-Yuan Yang
- School of Biomedical Engineering and Med-X Research Institute, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Wei-Qiang Gao
- State Key Laboratory of Oncogenes and Related Genes, Renji-Med X Clinical Stem Cell Research Center, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China ; School of Biomedical Engineering and Med-X Research Institute, Shanghai Jiao Tong University, Shanghai 200030, China
| |
Collapse
|