1
|
Qiu Y, Zhu L, Cai W, Zhu L. Research Progress on BDNF and Depression. ACS Chem Neurosci 2025. [PMID: 40359301 DOI: 10.1021/acschemneuro.5c00193] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/15/2025] Open
Abstract
Depression is a potentially life-threatening psychiatric disorder that affects the physical and mental health of millions of individuals worldwide. It can manifest at any stage of life, inducing profound emotional despondency, negative cognitions, and, in severe cases, suicidal ideation, often accompanied by physical symptoms, bringing a significant burden on both families and society. Brain-derived neurotrophic factor (BDNF), a member of the neurotrophic factor family, is widely expressed in the central nervous system (CNS), particularly in regions, such as the hippocampus and cortex. Numerous studies have shown that an imbalance or inadequate conversion of pro-brain-derived neurotrophic factor (proBDNF) into its mature form, mature BDNF (mBDNF), may impair neuronal plasticity, which is crucial to the pathogenesis of depression. This paper provides a comprehensive review of the neurotrophic mechanisms implicated in depression, covering the location, expression, and release of BDNF; the relationship between proBDNF, mBDNF, and depression; and the downstream signaling pathways triggered by BNDF binding to its receptors. This review aims to provide a theoretical foundation for understanding the pathogenesis and clinical treatment of depression.
Collapse
Affiliation(s)
- Yahong Qiu
- The Key Laboratory of Developmental Genes and Human Disease, Department of Histology and Embryology, School of Medicine, Southeast University, Nanjing, Jiangsu 210009, China
| | - Lixia Zhu
- Patent Examination Cooperation (JIANGSU) Center of the Patent Office, China National Intellectual Property Administration (CNIPA), Suzhou, Jiangsu 215163, China
| | - Wenyan Cai
- The Key Laboratory of Developmental Genes and Human Disease, Department of Histology and Embryology, School of Medicine, Southeast University, Nanjing, Jiangsu 210009, China
| | - Lijuan Zhu
- The Key Laboratory of Developmental Genes and Human Disease, Department of Histology and Embryology, School of Medicine, Southeast University, Nanjing, Jiangsu 210009, China
| |
Collapse
|
2
|
Turkistani A, Al-kuraishy HM, Al-Gareeb AI, Albuhadily AK, Elhussieny O, AL-Farga A, Aqlan F, Saad HM, Batiha GES. The functional and molecular roles of p75 neurotrophin receptor (p75 NTR) in epilepsy. J Cent Nerv Syst Dis 2024; 16:11795735241247810. [PMID: 38655152 PMCID: PMC11036928 DOI: 10.1177/11795735241247810] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Accepted: 03/14/2024] [Indexed: 04/26/2024] Open
Abstract
Epilepsy is a chronic neurological disorder manifested by recurring unprovoked seizures resulting from an imbalance in the inhibitory and excitatory neurotransmitters in the brain. The process of epileptogenesis involves a complex interplay between the reduction of inhibitory gamma-aminobutyric acid (GABA) and the enhancement of excitatory glutamate. Pro-BDNF/p75NTR expression is augmented in both glial cells and neurons following epileptic seizures and status epileptics (SE). Over-expression of p75NTR is linked with the pathogenesis of epilepsy, and augmentation of pro-BDNF/p75NTR is implicated in the pathogenesis of epilepsy. However, the precise mechanistic function of p75NTR in epilepsy has not been completely elucidated. Therefore, this review aimed to revise the mechanistic pathway of p75NTR in epilepsy.
Collapse
Affiliation(s)
- Areej Turkistani
- Department of pharmacology and toxicology, Collage of Medicine, Taif University, Taif, Kingdom of Saudi
| | - Hayder M. Al-kuraishy
- Professor in department of clinical pharmacology and medicine, college of medicine, Mustansiriyah University, Baghdad, Iraq
| | - Ali I. Al-Gareeb
- Professor in department of clinical pharmacology and medicine, college of medicine, Mustansiriyah University, Baghdad, Iraq
| | - Ali K. Albuhadily
- Professor in department of clinical pharmacology and medicine, college of medicine, Mustansiriyah University, Baghdad, Iraq
| | - Omnya Elhussieny
- Department of Histology and Cytology, Faculty of Veterinary Medicine, Matrouh University, Marsa Matruh, Egypt
| | - Ammar AL-Farga
- Biochemistry Department, College of Sciences, University of Jeddah, Jeddah, Saudia Arbia
| | - Faisal Aqlan
- Department of Chemistry, College of Sciences, Ibb University, Ibb Governorate, Yemen
| | - Hebatallah M. Saad
- Department of Pathology, Faculty of Veterinary Medicine, Matrouh University, Matrouh, Egypt
| | - Gaber El-Saber Batiha
- Department of Pharmacology and Therapeutics, Faculty of Veterinary Medicine, Damanhour University, Damanhour, Egypt
| |
Collapse
|
3
|
Wan J, Ma L, Jiao X, Dong W, Lin J, Qiu Y, Wu W, Liu Q, Chen C, Huang H, Li S, Zheng H, Wu Y. Impaired synaptic plasticity and decreased excitability of hippocampal glutamatergic neurons mediated by BDNF downregulation contribute to cognitive dysfunction in mice induced by repeated neonatal exposure to ketamine. CNS Neurosci Ther 2024; 30:e14604. [PMID: 38332635 PMCID: PMC10853651 DOI: 10.1111/cns.14604] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2023] [Revised: 12/22/2023] [Accepted: 01/03/2024] [Indexed: 02/10/2024] Open
Abstract
AIM Repeated exposure to ketamine during the neonatal period in mice leads to cognitive impairments in adulthood. These impairments are likely caused by synaptic plasticity and excitability damage. We investigated the precise role of brain-derived neurotrophic factor (BDNF) in the cognitive impairments induced by repeated ketamine exposure during the neonatal period. METHODS We evaluated the cognitive function of mice using the Morris water maze test and novel object recognition test. Western blotting and immunofluorescence were used to detect the protein levels of BDNF. Western blotting, Golgi-Cox staining, transmission electron microscopy, and long-term potentiation (LTP) recordings were used to assess synaptic plasticity in the hippocampus. The excitability of neurons was evaluated using c-Fos. In the intervention experiment, pAdeno-CaMKIIα-BDNF-mNeuronGreen was injected into the hippocampal CA1 region of mice to increase the level of BDNF. The excitability of neurons was enhanced using a chemogenetic approach. RESULTS Our findings suggest that cognitive impairments in mice repeatedly exposed to ketamine during the neonatal period are associated with downregulated BDNF protein level, synaptic plasticity damage, and decreased excitability of glutamatergic neurons in the hippocampal CA1 region. Furthermore, the specific upregulation of BDNF in glutamatergic neurons of the hippocampal CA1 region and the enhancement of excitability can improve impaired synaptic plasticity and cognitive function in mice. CONCLUSION BDNF downregulation mediates synaptic plasticity and excitability damage, leading to cognitive impairments in adulthood following repeated ketamine exposure during the neonatal period.
Collapse
Affiliation(s)
- Jie Wan
- Jiangsu Province Key Laboratory of Anesthesiology, Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic DrugsXuzhou Medical UniversityXuzhouChina
| | - Linhui Ma
- Jiangsu Province Key Laboratory of Anesthesiology, Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic DrugsXuzhou Medical UniversityXuzhouChina
- Department of Anesthesiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Xinhao Jiao
- Jiangsu Province Key Laboratory of Anesthesiology, Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic DrugsXuzhou Medical UniversityXuzhouChina
| | - Wei Dong
- Jiangsu Province Key Laboratory of Anesthesiology, Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic DrugsXuzhou Medical UniversityXuzhouChina
| | - Jiatao Lin
- Jiangsu Province Key Laboratory of Anesthesiology, Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic DrugsXuzhou Medical UniversityXuzhouChina
| | - Yongkang Qiu
- Jiangsu Province Key Laboratory of Anesthesiology, Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic DrugsXuzhou Medical UniversityXuzhouChina
| | - Weifeng Wu
- Jiangsu Province Key Laboratory of Anesthesiology, Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic DrugsXuzhou Medical UniversityXuzhouChina
| | - Qiang Liu
- Jiangsu Province Key Laboratory of Anesthesiology, Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic DrugsXuzhou Medical UniversityXuzhouChina
| | - Chen Chen
- Jiangsu Province Key Laboratory of Anesthesiology, Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic DrugsXuzhou Medical UniversityXuzhouChina
| | - He Huang
- Jiangsu Province Key Laboratory of Anesthesiology, Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic DrugsXuzhou Medical UniversityXuzhouChina
| | - Shuai Li
- Department of Anesthesiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Hui Zheng
- Department of Anesthesiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Yuqing Wu
- Jiangsu Province Key Laboratory of Anesthesiology, Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic DrugsXuzhou Medical UniversityXuzhouChina
| |
Collapse
|
4
|
Gao C, Cao Y, He M, Zhang X, Zhong Q, Tang L, Chen T, Zhang Z. SAG treatment ameliorates memory impairment related to sleep loss by upregulating synaptic plasticity in adolescent mice. Behav Brain Res 2023; 450:114468. [PMID: 37148913 DOI: 10.1016/j.bbr.2023.114468] [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: 01/28/2023] [Revised: 03/17/2023] [Accepted: 03/18/2023] [Indexed: 05/08/2023]
Abstract
Adequate sleep during the developmental stage can promote learning and memory functions because synaptic protein synthesis at primed synapses during sleep profoundly affects neurological function. The Sonic hedgehog (Shh) signaling pathway affects neuroplasticity in the hippocampus during the development of the central nervous system. In this study, the changes in synaptic morphology and function induced by sleep deprivation and the potential therapeutic effect of a Shh agonist (SAG) on these changes were investigated in adolescent mice. Adolescent mice were subjected to sleep deprivation for 20 hrs (2pm to 10 am the next day) and were free to sleep for the remaining 4 hrs per day for 10 consecutive days. Sleep-deprived mice were injected with SAG (10mg/kg body weight, i.p.) or saline (i.p.) every day 5min before the onset of the 20h sleep deprivation period. Chronic sleep deprivation impaired recognition and spatial memory, decreased the number of dendritic spines and mEPSCs of hippocampal CA1 pyramidal neurons, decreased the postsynaptic density, and reduced Shh and glioma-associated oncogene homolog 1 (Gli1) expression. SAG significantly protected against sleep deprivation-induced memory dysfunction, increased the CA1 pyramidal neuronal dendritic spine number and mEPSC frequency, and increased Gli1 expression. In conclusion, sleep deprivation induces memory impairment in adolescent mice, and SAG treatment prevents this impairment, probably by enhancing synaptic function in the hippocampal CA1 region.
Collapse
Affiliation(s)
- Chenyi Gao
- Department of Anesthesiology, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Yue Cao
- Department of Anesthesiology, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Mengying He
- Department of Anesthesiology, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Xuemin Zhang
- Department of Anesthesiology, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Qi Zhong
- Department of Anesthesiology, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Lijuan Tang
- Department of Anesthesiology, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Ting Chen
- Department of Anesthesiology, Zhongnan Hospital of Wuhan University, Wuhan 430071, China.
| | - Zongze Zhang
- Department of Anesthesiology, Zhongnan Hospital of Wuhan University, Wuhan 430071, China.
| |
Collapse
|
5
|
Jiang F, Bello ST, Gao Q, Lai Y, Li X, He L. Advances in the Electrophysiological Recordings of Long-Term Potentiation. Int J Mol Sci 2023; 24:ijms24087134. [PMID: 37108295 PMCID: PMC10138642 DOI: 10.3390/ijms24087134] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Revised: 04/01/2023] [Accepted: 04/06/2023] [Indexed: 04/29/2023] Open
Abstract
Understanding neuronal firing patterns and long-term potentiation (LTP) induction in studying learning, memory, and neurological diseases is critical. However, recently, despite the rapid advancement in neuroscience, we are still constrained by the experimental design, detection tools for exploring the mechanisms and pathways involved in LTP induction, and detection ability of neuronal action potentiation signals. This review will reiterate LTP-related electrophysiological recordings in the mammalian brain for nearly 50 years and explain how excitatory and inhibitory neural LTP results have been detected and described by field- and single-cell potentials, respectively. Furthermore, we focus on describing the classic model of LTP of inhibition and discuss the inhibitory neuron activity when excitatory neurons are activated to induce LTP. Finally, we propose recording excitatory and inhibitory neurons under the same experimental conditions by combining various electrophysiological technologies and novel design suggestions for future research. We discussed different types of synaptic plasticity, and the potential of astrocytes to induce LTP also deserves to be explored in the future.
Collapse
Affiliation(s)
- Feixu Jiang
- Department of Neuroscience, City University of Hong Kong, Kowloon, Hong Kong
| | | | - Qianqian Gao
- Department of Neuroscience, City University of Hong Kong, Kowloon, Hong Kong
| | - Yuanying Lai
- Department of Neuroscience, City University of Hong Kong, Kowloon, Hong Kong
| | - Xiao Li
- Department of Neuroscience, City University of Hong Kong, Kowloon, Hong Kong
- Research Institute of City University of Hong Kong, Shenzhen 518057, China
| | - Ling He
- Department of Neuroscience, City University of Hong Kong, Kowloon, Hong Kong
- Research Institute of City University of Hong Kong, Shenzhen 518057, China
| |
Collapse
|
6
|
Hui KK, Chater TE, Goda Y, Tanaka M. How Staying Negative Is Good for the (Adult) Brain: Maintaining Chloride Homeostasis and the GABA-Shift in Neurological Disorders. Front Mol Neurosci 2022; 15:893111. [PMID: 35875665 PMCID: PMC9305173 DOI: 10.3389/fnmol.2022.893111] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Accepted: 06/10/2022] [Indexed: 01/27/2023] Open
Abstract
Excitatory-inhibitory (E-I) imbalance has been shown to contribute to the pathogenesis of a wide range of neurodevelopmental disorders including autism spectrum disorders, epilepsy, and schizophrenia. GABA neurotransmission, the principal inhibitory signal in the mature brain, is critically coupled to proper regulation of chloride homeostasis. During brain maturation, changes in the transport of chloride ions across neuronal cell membranes act to gradually change the majority of GABA signaling from excitatory to inhibitory for neuronal activation, and dysregulation of this GABA-shift likely contributes to multiple neurodevelopmental abnormalities that are associated with circuit dysfunction. Whilst traditionally viewed as a phenomenon which occurs during brain development, recent evidence suggests that this GABA-shift may also be involved in neuropsychiatric disorders due to the "dematuration" of affected neurons. In this review, we will discuss the cell signaling and regulatory mechanisms underlying the GABA-shift phenomenon in the context of the latest findings in the field, in particular the role of chloride cotransporters NKCC1 and KCC2, and furthermore how these regulatory processes are altered in neurodevelopmental and neuropsychiatric disorders. We will also explore the interactions between GABAergic interneurons and other cell types in the developing brain that may influence the GABA-shift. Finally, with a greater understanding of how the GABA-shift is altered in pathological conditions, we will briefly outline recent progress on targeting NKCC1 and KCC2 as a therapeutic strategy against neurodevelopmental and neuropsychiatric disorders associated with improper chloride homeostasis and GABA-shift abnormalities.
Collapse
Affiliation(s)
- Kelvin K. Hui
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY, United States
- Institute for Aging Research, Albert Einstein College of Medicine, Bronx, NY, United States
| | - Thomas E. Chater
- Laboratory for Synaptic Plasticity and Connectivity, RIKEN Center for Brain Science, Wako, Japan
| | - Yukiko Goda
- Laboratory for Synaptic Plasticity and Connectivity, RIKEN Center for Brain Science, Wako, Japan
- Synapse Biology Unit, Okinawa Institute for Science and Technology Graduate University, Onna, Japan
| | - Motomasa Tanaka
- Laboratory for Protein Conformation Diseases, RIKEN Center for Brain Science, Wako, Japan
| |
Collapse
|
7
|
Llorca A, Deogracias R. Origin, Development, and Synaptogenesis of Cortical Interneurons. Front Neurosci 2022; 16:929469. [PMID: 35833090 PMCID: PMC9272671 DOI: 10.3389/fnins.2022.929469] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Accepted: 06/01/2022] [Indexed: 11/13/2022] Open
Abstract
The mammalian cerebral cortex represents one of the most recent and astonishing inventions of nature, responsible of a large diversity of functions that range from sensory processing to high-order cognitive abilities, such as logical reasoning or language. Decades of dedicated study have contributed to our current understanding of this structure, both at structural and functional levels. A key feature of the neocortex is its outstanding richness in cell diversity, composed by multiple types of long-range projecting neurons and locally connecting interneurons. In this review, we will describe the great diversity of interneurons that constitute local neocortical circuits and summarize the mechanisms underlying their development and their assembly into functional networks.
Collapse
Affiliation(s)
- Alfredo Llorca
- Visual Neuroscience Laboratory, Centre for Discovery Brain Sciences, School of Biomedical Sciences, University of Edinburgh, Edinburg, United Kingdom
- *Correspondence: Alfredo Llorca
| | - Ruben Deogracias
- Neuronal Circuits Formation and Brain Disorders Laboratory, Institute of Neurosciences of Castilla y León (INCyL), University of Salamanca, Salamanca, Spain
- Institute of Biomedical Research of Salamanca, Salamanca, Spain
- Department of Cell Biology and Pathology, School of Medicine, University of Salamanca, Salamanca, Spain
- Ruben Deogracias
| |
Collapse
|
8
|
Prowse N, Hayley S. Microglia and BDNF at the crossroads of stressor related disorders: Towards a unique trophic phenotype. Neurosci Biobehav Rev 2021; 131:135-163. [PMID: 34537262 DOI: 10.1016/j.neubiorev.2021.09.018] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 09/08/2021] [Accepted: 09/08/2021] [Indexed: 12/16/2022]
Abstract
Stressors ranging from psychogenic/social to neurogenic/injury to systemic/microbial can impact microglial inflammatory processes, but less is known regarding their effects on trophic properties of microglia. Recent studies do suggest that microglia can modulate neuronal plasticity, possibly through brain derived neurotrophic factor (BDNF). This is particularly important given the link between BDNF and neuropsychiatric and neurodegenerative pathology. We posit that certain activated states of microglia play a role in maintaining the delicate balance of BDNF release onto neuronal synapses. This focused review will address how different "activators" influence the expression and release of microglial BDNF and address the question of tropomyosin receptor kinase B (TrkB) expression on microglia. We will then assess sex-based differences in microglial function and BDNF expression, and how microglia are involved in the stress response and related disorders such as depression. Drawing on research from a variety of other disorders, we will highlight challenges and opportunities for modulators that can shift microglia to a "trophic" phenotype with a view to potential therapeutics relevant for stressor-related disorders.
Collapse
Affiliation(s)
- Natalie Prowse
- Department of Neuroscience, Carleton University, Ottawa, ON K1S 5B6, Canada.
| | - Shawn Hayley
- Department of Neuroscience, Carleton University, Ottawa, ON K1S 5B6, Canada.
| |
Collapse
|
9
|
CCL2/CCR2 Contributes to the Altered Excitatory-inhibitory Synaptic Balance in the Nucleus Accumbens Shell Following Peripheral Nerve Injury-induced Neuropathic Pain. Neurosci Bull 2021; 37:921-933. [PMID: 34003466 DOI: 10.1007/s12264-021-00697-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Accepted: 02/10/2021] [Indexed: 12/12/2022] Open
Abstract
The medium spiny neurons (MSNs) in the nucleus accumbens (NAc) integrate excitatory and inhibitory synaptic inputs and gate motivational and emotional behavior output. Here we report that the relative intensity of excitatory and inhibitory synaptic inputs to MSNs of the NAc shell was decreased in mice with neuropathic pain induced by spinal nerve ligation (SNL). SNL increased the frequency, but not the amplitude of spontaneous inhibitory postsynaptic currents (sIPSCs), and decreased both the frequency and amplitude of spontaneous excitatory postsynaptic currents (sEPSCs) in the MSNs. SNL also decreased the paired-pulse ratio (PPR) of evoked IPSCs but increased the PPR of evoked EPSCs. Moreover, acute bath application of C-C motif chemokine ligand 2 (CCL2) increased the frequency and amplitude of sIPSCs and sEPSCs in the MSNs, and especially strengthened the amplitude of N-methyl-D-aspartate receptor (NMDAR)-mediated miniature EPSCs. Further Ccl2 overexpression in the NAc in vivo decreased the peak amplitude of the sEPSC/sIPSC ratio. Finally, Ccr2 knock-down improved the impaired induction of NMDAR-dependent long-term depression (LTD) in the NAc after SNL. These results suggest that CCL2/CCR2 signaling plays a role in the integration of excitatory/inhibitory synaptic transmission and leads to an increase of the LTD induction threshold at the synapses of MSNs during neuropathic pain.
Collapse
|
10
|
Abstract
The neurotrophic factor BDNF is an important regulator for the development of brain circuits, for synaptic and neuronal network plasticity, as well as for neuroregeneration and neuroprotection. Up- and downregulations of BDNF levels in human blood and tissue are associated with, e.g., neurodegenerative, neurological, or even cardiovascular diseases. The changes in BDNF concentration are caused by altered dynamics in BDNF expression and release. To understand the relevance of major variations of BDNF levels, detailed knowledge regarding physiological and pathophysiological stimuli affecting intra- and extracellular BDNF concentration is important. Most work addressing the molecular and cellular regulation of BDNF expression and release have been performed in neuronal preparations. Therefore, this review will summarize the stimuli inducing release of BDNF, as well as molecular mechanisms regulating the efficacy of BDNF release, with a focus on cells originating from the brain. Further, we will discuss the current knowledge about the distinct stimuli eliciting regulated release of BDNF under physiological conditions.
Collapse
Affiliation(s)
- Tanja Brigadski
- Department of Informatics and Microsystem Technology, University of Applied Sciences Kaiserslautern, D-66482, Zweibrücken, Germany.
| | - Volkmar Leßmann
- Institute of Physiology, Otto-von-Guericke University, D-39120, Magdeburg, Germany.
- Center for Behavioral Brain Sciences, Magdeburg, Germany.
| |
Collapse
|
11
|
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: 20] [Impact Index Per Article: 4.0] [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
|
12
|
Delmotte Q, Diabira D, Belaidouni Y, Hamze M, Kochmann M, Montheil A, Gaiarsa JL, Porcher C, Belgacem YH. Sonic Hedgehog Signaling Agonist (SAG) Triggers BDNF Secretion and Promotes the Maturation of GABAergic Networks in the Postnatal Rat Hippocampus. Front Cell Neurosci 2020; 14:98. [PMID: 32425757 PMCID: PMC7212340 DOI: 10.3389/fncel.2020.00098] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Accepted: 03/31/2020] [Indexed: 12/13/2022] Open
Abstract
Sonic hedgehog (Shh) signaling plays critical roles during early central nervous system development, such as neural cell proliferation, patterning of the neural tube and neuronal differentiation. While Shh signaling is still present in the postnatal brain, the roles it may play are, however, largely unknown. In particular, Shh signaling components are found at the synaptic junction in the maturing hippocampus during the first two postnatal weeks. This period is characterized by the presence of ongoing spontaneous synaptic activity at the cellular and network levels thought to play important roles in the onset of neuronal circuit formation and synaptic plasticity. Here, we demonstrate that non-canonical Shh signaling increases the frequency of the synchronized electrical activity called Giant Depolarizing Potentials (GDP) and enhances spontaneous GABA post-synaptic currents in the rodent hippocampus during the early postnatal period. This effect is mediated specifically through the Shh co-receptor Smoothened via intracellular Ca2+ signal and the activation of the BDNF-TrkB signaling pathway. Given the importance of these spontaneous events on neuronal network maturation and refinement, this study opens new perspectives for Shh signaling on the control of early stages of postnatal brain maturation and physiology.
Collapse
Affiliation(s)
- Quentin Delmotte
- Aix-Marseille Univ, Marseille, France.,INSERM (Institut National de la Santé et de la Recherche Médicale) Unité 1249, Marseille, France.,INMED (Institut de Neurobiologie de la Méditerranée), Parc Scientifique de Luminy, Marseille, France
| | - Diabe Diabira
- INSERM (Institut National de la Santé et de la Recherche Médicale) Unité 1249, Marseille, France.,INMED (Institut de Neurobiologie de la Méditerranée), Parc Scientifique de Luminy, Marseille, France
| | - Yasmine Belaidouni
- Aix-Marseille Univ, Marseille, France.,INSERM (Institut National de la Santé et de la Recherche Médicale) Unité 1249, Marseille, France.,INMED (Institut de Neurobiologie de la Méditerranée), Parc Scientifique de Luminy, Marseille, France
| | - Mira Hamze
- Aix-Marseille Univ, Marseille, France.,INSERM (Institut National de la Santé et de la Recherche Médicale) Unité 1249, Marseille, France.,INMED (Institut de Neurobiologie de la Méditerranée), Parc Scientifique de Luminy, Marseille, France
| | - Marine Kochmann
- Aix-Marseille Univ, Marseille, France.,Institut des Neurosciences de La Timone, Marseille, France
| | - Aurélie Montheil
- Aix-Marseille Univ, Marseille, France.,INSERM (Institut National de la Santé et de la Recherche Médicale) Unité 1249, Marseille, France.,INMED (Institut de Neurobiologie de la Méditerranée), Parc Scientifique de Luminy, Marseille, France
| | - Jean-Luc Gaiarsa
- Aix-Marseille Univ, Marseille, France.,INSERM (Institut National de la Santé et de la Recherche Médicale) Unité 1249, Marseille, France.,INMED (Institut de Neurobiologie de la Méditerranée), Parc Scientifique de Luminy, Marseille, France
| | - Christophe Porcher
- Aix-Marseille Univ, Marseille, France.,INSERM (Institut National de la Santé et de la Recherche Médicale) Unité 1249, Marseille, France.,INMED (Institut de Neurobiologie de la Méditerranée), Parc Scientifique de Luminy, Marseille, France
| | - Yesser H Belgacem
- INSERM (Institut National de la Santé et de la Recherche Médicale) Unité 1249, Marseille, France.,INMED (Institut de Neurobiologie de la Méditerranée), Parc Scientifique de Luminy, Marseille, France
| |
Collapse
|
13
|
Chronic exercise buffers the cognitive dysfunction and decreases the susceptibility to seizures in PTZ-treated rats. Epilepsy Behav 2019; 98:173-187. [PMID: 31377659 DOI: 10.1016/j.yebeh.2019.07.032] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Revised: 07/02/2019] [Accepted: 07/08/2019] [Indexed: 02/02/2023]
Abstract
Epilepsy is a serious neurological disorder posing a severe burden to our society. Cognitive deficits are very common comorbidities of epilepsy. It is known that enhanced cognition has been demonstrated as an indicator for successful treatment of epilepsy. Physical exercise shows a positive consequence on cognition in healthy individuals and improves health and life conditions in people with epilepsy. However, there is no direct evidence to determine the role and the potential mechanism of physical exercise on the cognitive impairment and the relationship of susceptibility to seizures. The goal of the current investigation was to explore whether sustained physical exercise improves the cognitive dysfunction and simultaneously decreases the susceptibility to seizures in rats with epilepsy. Rats were treated with pentylenetetrazole (PTZ) (35 mg/kg, i.p. [intraperitoneally]) for 36 days to induce chronic epilepsy. During the induction period, rats were exposed to voluntary wheel running or forced swimming 30 min prior to each PTZ injection from the 16th day. The cognition of rats was evaluated by object recognition test and passive avoidance test. The susceptibility to seizures was evaluated by seizure frequency and duration. The levels of synaptic-related proteins including PSD95 (postsynaptic density 95), Synapsin, GluA1, and BDNF (brain-derived neurotrophic factor) were measured to evaluate the hippocampal synaptic plasticity. Furthermore, the GAD67 (glutamic acid decarboxylase) levels and GABA (γ-aminobutyric acid)ergic function in PTZ-treated rats were also determined. Finally, antagonist of GABAAR (GABAA receptors) bicuculline was used to explore the reversal effects of physical activity on seizures and cognition. The results showed that rats subjected to voluntary wheel running or forced swimming showed a significant reduction of seizure frequency and duration in PTZ-treated group relative to rats without running or swimming. In addition, both running and swimming improved cognitive function as measured by enhanced performance in object recognition test and passive avoidance test. Furthermore, the reduced levels of synaptic-related proteins and GABAergic function were reversed by exercise compared with rats without exercise. Moreover, antagonism of hippocampal CA3 (cornu ammonis 3) GABAergic neurons blocks the reversal effects of physical activity on seizures and cognition in PTZ-treated rats. These data showed that chronic physical exercise reduced the frequency of seizures and improved the cognitive function in a rat model of chronic epilepsy through normalization of CA3 synaptic plasticity and GABAergic function. Our findings suggest that chronic physical exercise has beneficial effects on controlling seizure through enhancement of cognition and highlights the possibility to translate into reduced seizure recurrence in people with epilepsy.
Collapse
|
14
|
Medlej Y, Salah H, Wadi L, Saad S, Bashir B, Allam J, Atoui Z, Darwish N, Karnib N, Darwish H, Kobeissy F, Wang KKW, Hamade E, Obeid M. Lestaurtinib (CEP-701) modulates the effects of early life hypoxic seizures on cognitive and emotional behaviors in immature rats. Epilepsy Behav 2019; 92:332-340. [PMID: 30769278 DOI: 10.1016/j.yebeh.2019.01.023] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Revised: 01/17/2019] [Accepted: 01/18/2019] [Indexed: 11/30/2022]
Abstract
Hypoxic encephalopathy of the newborn is a major cause of long-term neurological sequelae. We have previously shown that CEP-701 (lestaurtinib), a drug with an established safety profile in children, attenuates short-term hyperexcitability and tropomyosin-related kinase B (TrkB) receptor activation in a well-established rat model of early life hypoxic seizures (HS). Here, we investigated the potential long-term neuroprotective effects of a post-HS transient CEP-701 treatment. Following exposure to global hypoxia, 10 day old male Sprague-Dawley pups received CEP-701 or its vehicle and were sequentially subjected to the light-dark box test (LDT), forced swim test (FST), open field test (OFT), Morris water maze (MWM), and the modified active avoidance (MAAV) test between postnatal days 24 and 44 (P24-44). Spontaneous seizure activity was assessed by epidural cortical electroencephalography (EEG) between P50 and 100. Neuronal density and glial fibrillary acidic protein (GFAP) levels were evaluated on histological sections in the hippocampus, amygdala, and prefrontal cortex at P100. Vehicle-treated hypoxic rats exhibited significantly increased immobility in the FST compared with controls, and post-HS CEP-701 administration reversed this HS-induced depressive-like behavior (p < 0.05). In the MAAV test, CEP-701-treated hypoxic rats were slower at learning both context-cued and tone-signaled shock-avoidance behaviors (p < 0.05). All other behavioral outcomes were comparable, and no recurrent seizures, neuronal loss, or increase in GFAP levels were detected in any of the groups. We showed that early life HS predispose to long-lasting depressive-like behaviors, and that these are prevented by CEP-701, likely via TrkB modulation. Future mechanistically more specific studies will further investigate the potential role of TrkB signaling pathway modulation in achieving neuroprotection against neonatal HS, without causing neurodevelopmental adverse effects.
Collapse
Affiliation(s)
- Yasser Medlej
- Department of Anatomy, Cell biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Houssein Salah
- Department of Anatomy, Cell biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Lara Wadi
- Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Sarah Saad
- Faculty of Arts and Sciences, American University of Beirut, Lebanon
| | - Bashir Bashir
- Faculty of Arts and Sciences, American University of Beirut, Lebanon
| | - Jad Allam
- Faculty of Arts and Sciences, American University of Beirut, Lebanon
| | - Zahraa Atoui
- Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Nora Darwish
- Faculty of Arts and Sciences, American University of Beirut, Lebanon
| | - Nabil Karnib
- Department of Anatomy, Cell biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut, Lebanon; Department of Natural Sciences, School of Arts and Sciences, Lebanese American University, Lebanon
| | - Hala Darwish
- Department of Anatomy, Cell biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut, Lebanon; Rafic Hariri School of Nursing, American University of Beirut, Beirut, Lebanon
| | - Firas Kobeissy
- Department of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, Beirut, Lebanon; Program for Neurotrauma, Neuroproteomics, Department of Emergency Medicine, Department of Chemistry, Department of Neuroscience, and Department of Psychiatry, McKnight Brain Institute, University of Florida, Gainesville, FL, USA
| | - Kevin K W Wang
- Program for Neurotrauma, Neuroproteomics, Department of Emergency Medicine, Department of Chemistry, Department of Neuroscience, and Department of Psychiatry, McKnight Brain Institute, University of Florida, Gainesville, FL, USA
| | - Eva Hamade
- Department of Biochemistry, Faculty of Science, Lebanese University, Lebanon
| | - Makram Obeid
- Department of Anatomy, Cell biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut, Lebanon; Division of Child Neurology, Department of Pediatric and Adolescent Medicine, American University of Beirut Medical Center, Beirut, Lebanon.
| |
Collapse
|
15
|
Brain-Derived Neurotrophic Factor Is Required for the Neuroprotective Effect of Mifepristone on Immature Purkinje Cells in Cerebellar Slice Culture. Int J Mol Sci 2019; 20:ijms20020285. [PMID: 30642045 PMCID: PMC6359295 DOI: 10.3390/ijms20020285] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2018] [Revised: 01/08/2019] [Accepted: 01/10/2019] [Indexed: 12/02/2022] Open
Abstract
Endogenous γ-aminobutyric acid (GABA)-dependent activity induces death of developing Purkinje neurons in mouse organotypic cerebellar cultures and the synthetic steroid mifepristone blocks this effect. Here, using brain-derived neurotrophic factor (BDNF) heterozygous mice, we show that BDNF plays no role in immature Purkinje cell death. However, interestingly, BDNF haploinsufficiency impairs neuronal survival induced by mifepristone and GABAA-receptors antagonist (bicuculline) treatments, indicating that the underlying neuroprotective mechanism requires the neurotrophin full expression.
Collapse
|
16
|
Porcher C, Medina I, Gaiarsa JL. Mechanism of BDNF Modulation in GABAergic Synaptic Transmission in Healthy and Disease Brains. Front Cell Neurosci 2018; 12:273. [PMID: 30210299 PMCID: PMC6121065 DOI: 10.3389/fncel.2018.00273] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Accepted: 08/06/2018] [Indexed: 12/18/2022] Open
Abstract
In the mature healthy mammalian neuronal networks, γ-aminobutyric acid (GABA) mediates synaptic inhibition by acting on GABAA and GABAB receptors (GABAAR, GABABR). In immature networks and during numerous pathological conditions the strength of GABAergic synaptic inhibition is much less pronounced. In these neurons the activation of GABAAR produces paradoxical depolarizing action that favors neuronal network excitation. The depolarizing action of GABAAR is a consequence of deregulated chloride ion homeostasis. In addition to depolarizing action of GABAAR, the GABABR mediated inhibition is also less efficient. One of the key molecules regulating the GABAergic synaptic transmission is the brain derived neurotrophic factor (BDNF). BDNF and its precursor proBDNF, can be released in an activity-dependent manner. Mature BDNF operates via its cognate receptors tropomyosin related kinase B (TrkB) whereas proBDNF binds the p75 neurotrophin receptor (p75NTR). In this review article, we discuss recent finding illuminating how mBDNF-TrkB and proBDNF-p75NTR signaling pathways regulate GABA related neurotransmission under physiological conditions and during epilepsy.
Collapse
Affiliation(s)
- Christophe Porcher
- Aix Marseille University, Marseille, France.,Institut National de la Santé et de la Recherche Médicale (INSERM) U901, Marseille, France.,Institut de Neurobiologie de la Méditerranée (INMED), Marseille, France
| | - Igor Medina
- Aix Marseille University, Marseille, France.,Institut National de la Santé et de la Recherche Médicale (INSERM) U901, Marseille, France.,Institut de Neurobiologie de la Méditerranée (INMED), Marseille, France
| | - Jean-Luc Gaiarsa
- Aix Marseille University, Marseille, France.,Institut National de la Santé et de la Recherche Médicale (INSERM) U901, Marseille, France.,Institut de Neurobiologie de la Méditerranée (INMED), Marseille, France
| |
Collapse
|
17
|
LTP or LTD? Modeling the Influence of Stress on Synaptic Plasticity. eNeuro 2018; 5:eN-TNC-0242-17. [PMID: 29662939 PMCID: PMC5898787 DOI: 10.1523/eneuro.0242-17.2018] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2017] [Revised: 01/15/2018] [Accepted: 01/21/2018] [Indexed: 01/03/2023] Open
Abstract
In cognitive memory, long-term potentiation (LTP) has been shown to occur when presynaptic and postsynaptic activities are highly correlated and glucocorticoid concentrations are in an optimal (i.e., low normal) range. In all other conditions, LTP is attenuated or even long-term depression (LTD) occurs. In this paper, we focus on NMDA receptor (NMDA-R)-dependent LTP and LTD, two processes involving various molecular mechanisms. To understand which of these mechanisms are indispensable for explaining the experimental evidence reported in the literature, we here propose a parsimonious model of NMDA-R-dependent synaptic plasticity. Central to this model are two processes. First, AMPA receptor-subunit trafficking; and second, glucocorticoid-dependent modifications of the brain-derived neurotrophic factor (BDNF)-receptor system. In 2008, we have published a core model, which contained the first process, while in the current paper we present an extended model, which also includes the second process. Using the extended model, we could show that stress attenuates LTP, while it enhances LTD. These simulation results are in agreement with experimental findings from other labs. In 2013, surprising experimental evidence showed that the GluA1 C-tail is unnecessary for LTP. When using our core model in its original form, our simulations already predicted that there would be no requirement for the GluA1 C-tail for LTP, allowing to eliminate a redundant mechanism from our model. In summary, we present a mathematical model that displays reduced complexity and is useful for explaining when and how LTP or LTD occurs at synapses during cognitive memory formation.
Collapse
|
18
|
BDNF at the synapse: why location matters. Mol Psychiatry 2017; 22:1370-1375. [PMID: 28937692 PMCID: PMC5646361 DOI: 10.1038/mp.2017.144] [Citation(s) in RCA: 150] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2017] [Revised: 04/13/2017] [Accepted: 05/26/2017] [Indexed: 12/14/2022]
Abstract
Neurotrophic factors, a family of secreted proteins that support the growth, survival and differentiation of neurons, have been intensively studied for decades due to the powerful and diverse effects on neuronal physiology, as well as their therapeutic potential. Such efforts have led to a detailed understanding on the molecular mechanisms of neurotrophic factor signaling. One member, brain-derived neurotrophic factor (BDNF) has drawn much attention due to its pleiotropic roles in the central nervous system and implications in various brain disorders. In addition, recent advances linking the rapid-acting antidepressant, ketamine, to BDNF translation and BDNF-dependent signaling, has re-emphasized the importance of understanding the precise details of BDNF biology at the synapse. Although substantial knowledge related to the genetic, epigenetic, cell biological and biochemical aspects of BDNF biology has now been established, certain aspects related to the precise localization and release of BDNF at the synapse have remained obscure. A recent series of genetic and cell biological studies have shed light on the question-the site of BDNF release at the synapse. In this Perspectives article, these new insights will be placed in the context of previously unresolved issues related to BDNF biology, as well as how BDNF may function as a downstream mediator of newer pharmacological agents currently under investigation for treating psychiatric disorders.
Collapse
|
19
|
Liao Q, Zhang R, Wang X, Nian W, Ke L, Ouyang W, Zhang Z. Effect of fluoride exposure on mRNA expression of cav1.2 and calcium signal pathway apoptosis regulators in PC12 cells. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2017; 54:74-79. [PMID: 28697452 DOI: 10.1016/j.etap.2017.06.018] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/09/2017] [Revised: 06/08/2017] [Accepted: 06/20/2017] [Indexed: 06/07/2023]
Abstract
This study investigated the effects of fluoride exposure on the mRNA expression of Cav1.2 calcium signaling pathway and apoptosis regulatory molecules in PC12 cells. The viability of PC12 cell receiving high fluoride (5.0mM) and low fluoride (0.5mM) alone or fluoride combined with L-type calcium channel (LTCC) agonist/inhibitor (5umol/L FPL6417/2umol/L nifedipine) was detected using cell counting kit-8 at different time points (2, 4, 6, 8, 12, 10, and 24h). Changes in the cell configuration were observed after exposing the cells to fluoride for 24h. The expression levels of molecules related to the LTCC were examined, particularly, Cav1.2, c-fos, CAMK II, Bax, and Bcl-2. Fluoride poisoning induced severe cell injuries, such as decreased PC12 cell activity, enhanced cell apoptosis, high c-fos, CAMKII, and Bax mRNA expression levels. Bcl-2 expression level was also reduced. Meanwhile, high fluoride, high fluoride with FPL64176, and low fluoride with FPL64176 enhanced the Cav1.2 expression level. In contrast, low fluoride, high fluoride with nifedipine, and low fluoride with nifedipine reduced the Cav1.2 expression level. Thus, Cav1.2 may be an important molecular target for the fluorosis treatment, and the LTCC inhibitor nifedipine may be an effective drug for fluorosis.
Collapse
Affiliation(s)
- Qiuxia Liao
- College of Chemistry and Life Science at Zhejiang Normal University, Jinhua, Zhejiang, 321004, PR China
| | - Rui Zhang
- College of Chemistry and Life Science at Zhejiang Normal University, Jinhua, Zhejiang, 321004, PR China
| | - Xiaoyu Wang
- College of Chemistry and Life Science at Zhejiang Normal University, Jinhua, Zhejiang, 321004, PR China
| | - Weiwei Nian
- College of Chemistry and Life Science at Zhejiang Normal University, Jinhua, Zhejiang, 321004, PR China
| | - Lulu Ke
- College of Chemistry and Life Science at Zhejiang Normal University, Jinhua, Zhejiang, 321004, PR China
| | - Wei Ouyang
- College of Sports and Health Science, Zhejiang Normal University, Jinhua, Zhejiang, 321004, PR China
| | - Zigui Zhang
- College of Xing Zhi, Zhejiang Normal University, Jinhua, Zhejiang, 321004, PR China.
| |
Collapse
|
20
|
Griguoli M, Cherubini E. Early Correlated Network Activity in the Hippocampus: Its Putative Role in Shaping Neuronal Circuits. Front Cell Neurosci 2017; 11:255. [PMID: 28878628 PMCID: PMC5572250 DOI: 10.3389/fncel.2017.00255] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2017] [Accepted: 08/08/2017] [Indexed: 12/02/2022] Open
Abstract
Synchronized neuronal activity occurring at different developmental stages in various brain structures represents a hallmark of developmental circuits. This activity, which differs in its specific patterns among animal species may play a crucial role in de novo formation and in shaping neuronal networks. In the rodent hippocampus in vitro, the so-called giant depolarizing potentials (GDPs) constitute a primordial form of neuronal synchrony preceding more organized forms of activity such as oscillations in the theta and gamma frequency range. GDPs are generated at the network level by the interaction of the neurotransmitters glutamate and GABA which, immediately after birth, exert both a depolarizing and excitatory action on their targets. GDPs are triggered by GABAergic interneurons, which in virtue of their extensive axonal branching operate as functional hubs to synchronize large ensembles of cells. Intrinsic bursting activity, driven by a persistent sodium conductance and facilitated by the low expression of Kv7.2 and Kv7.3 channel subunits, responsible for IM, exerts a permissive role in GDP generation. Here, we discuss how GDPs are generated in a probabilistic way when neuronal excitability within a local circuit reaches a certain threshold and how GDP-associated calcium transients act as coincident detectors for enhancing synaptic strength at emerging GABAergic and glutamatergic synapses. We discuss the possible in vivo correlate of this activity. Finally, we debate recent data showing how, in several animal models of neuropsychiatric disorders including autism, a GDPs dysfunction is associated to morphological alterations of neuronal circuits and behavioral deficits reminiscent of those observed in patients.
Collapse
Affiliation(s)
- Marilena Griguoli
- European Brain Research Institute (EBRI) "Fondazione Rita Levi-Montalcini"Rome, Italy
| | - Enrico Cherubini
- European Brain Research Institute (EBRI) "Fondazione Rita Levi-Montalcini"Rome, Italy.,Department of Neuroscience, International School for Advanced StudiesTrieste, Italy
| |
Collapse
|
21
|
Meunier CNJ, Chameau P, Fossier PM. Modulation of Synaptic Plasticity in the Cortex Needs to Understand All the Players. Front Synaptic Neurosci 2017; 9:2. [PMID: 28203201 PMCID: PMC5285384 DOI: 10.3389/fnsyn.2017.00002] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2016] [Accepted: 01/13/2017] [Indexed: 12/19/2022] Open
Abstract
The prefrontal cortex (PFC) is involved in cognitive tasks such as working memory, decision making, risk assessment and regulation of attention. These functions performed by the PFC are supposed to rely on rhythmic electrical activity generated by neuronal network oscillations determined by a precise balance between excitation and inhibition balance (E/I balance) resulting from the coordinated activities of recurrent excitation and feedback and feedforward inhibition. Functional alterations in PFC functions have been associated with cognitive deficits in several pathologies such as major depression, anxiety and schizophrenia. These pathological situations are correlated with alterations of different neurotransmitter systems (i.e., serotonin (5-HT), dopamine (DA), acetylcholine…) that result in alterations of the E/I balance. The aim of this review article is to cover the basic aspects of the regulation of the E/I balance as well as to highlight the importance of the complementarity role of several neurotransmitters in the modulation of the plasticity of excitatory and inhibitory synapses. We illustrate our purpose by recent findings that demonstrate that 5-HT and DA cooperate to regulate the plasticity of excitatory and inhibitory synapses targeting layer 5 pyramidal neurons (L5PyNs) of the PFC and to fine tune the E/I balance. Using a method based on the decomposition of the synaptic conductance into its excitatory and inhibitory components, we show that concomitant activation of D1-like receptors (D1Rs) and 5-HT1ARs, through a modulation of NMDA receptors, favors long term potentiation (LTP) of both excitation and inhibition and consequently does not modify the E/I balance. We also demonstrate that activation of D2-receptors requires functional 5-HT1ARs to shift the E-I balance towards more inhibition and to favor long term depression (LTD) of excitatory synapses through the activation of glycogen synthase kinase 3β (GSK3β). This cooperation between different neurotransmitters is particularly relevant in view of pathological situations in which alterations of one neurotransmitter system will also have consequences on the regulation of synaptic efficacy by other neurotransmitters. This opens up new perspectives in the development of therapeutic strategies for the pharmacological treatment of neuronal disorders.
Collapse
Affiliation(s)
- Claire N J Meunier
- Institut de Neurosciences Paris-Saclay (NeuroPSI), UMR 91197 CNRS-Université Paris-Saclay Paris, France
| | - Pascal Chameau
- Swammerdam Institute for Life Sciences, Center for NeuroScience, University of Amsterdam Amsterdam, Netherlands
| | - Philippe M Fossier
- Institut de Neurosciences Paris-Saclay (NeuroPSI), UMR 91197 CNRS-Université Paris-Saclay Paris, France
| |
Collapse
|
22
|
Morton RA, Valenzuela CF. Further characterization of the effect of ethanol on voltage-gated Ca(2+) channel function in developing CA3 hippocampal pyramidal neurons. Brain Res 2015; 1633:19-26. [PMID: 26711851 DOI: 10.1016/j.brainres.2015.12.023] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2015] [Revised: 11/16/2015] [Accepted: 12/14/2015] [Indexed: 10/22/2022]
Abstract
Developmental ethanol exposure damages the hippocampus, a brain region involved in learning and memory. Alterations in synaptic transmission and plasticity may play a role in this effect of ethanol. We previously reported that acute and repeated exposure to ethanol during the third trimester-equivalent inhibits long-term potentiation of GABAA receptor-dependent synaptic currents in CA3 pyramidal neurons through a mechanism that depends on retrograde release of brain-derived neurotrophic factor driven by activation of voltage-gated Ca(2+) channels (Zucca and Valenzuela, 2010). We found evidence indicating that voltage-gated Ca(2+) channels are inhibited in the presence of ethanol, an effect that may play a role in its mechanism of action. Here, we further investigated the acute effect of ethanol on the function of voltage-gated Ca(2+) channels in CA3 pyramidal neurons using Ca(2+) imaging techniques. These experiments revealed that acute ethanol exposure inhibits voltage-gated Ca(2+) channels both in somatic and proximal dendritic compartments. To investigate the long-term consequences of ethanol on voltage-gated Ca(2+) channels, we used patch-clamp electrophysiological techniques to assess the function of L-type voltage-gated Ca(2+) channels during and following ten days of vapor ethanol exposure. During ethanol withdrawal periods, the function of these channels was not significantly affected by vapor chamber exposure. Taken together with our previous findings, our results suggest that 3(rd) trimester-equivalent ethanol exposure transiently inhibits L-type voltage-gated Ca(2+) channel function in CA3 pyramidal neurons and that compensatory mechanisms restore their function during ethanol withdrawal. Transient inhibition of these channels by ethanol may be, in part, responsible for the hippocampal abnormalities associated with developmental exposure to this agent.
Collapse
Affiliation(s)
- Russell A Morton
- Department of Neurosciences, School of Medicine, University of New Mexico Health Sciences Center, Albuquerque, NM, USA
| | - C Fernando Valenzuela
- Department of Neurosciences, School of Medicine, University of New Mexico Health Sciences Center, Albuquerque, NM, USA.
| |
Collapse
|
23
|
Baculis BC, Valenzuela CF. Ethanol exposure during the third trimester equivalent does not affect GABAA or AMPA receptor-mediated spontaneous synaptic transmission in rat CA3 pyramidal neurons. J Negat Results Biomed 2015; 14:19. [PMID: 26627643 PMCID: PMC4667401 DOI: 10.1186/s12952-015-0041-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2015] [Accepted: 11/24/2015] [Indexed: 11/17/2022] Open
Abstract
Background Ethanol exposure during the rodent equivalent to the 3rd trimester of human pregnancy (i.e., first 1–2 weeks of neonatal life) has been shown to produce structural and functional alterations in the CA3 hippocampal sub-region, which is involved in associative memory. Synaptic plasticity mechanisms dependent on retrograde release of brain-derived neurotrophic factor (BDNF) driven by activation of L-type voltage-gated Ca2+ channels (L-VGCCs) are thought to play a role in stabilization of both GABAergic and glutamatergic synapses in CA3 pyramidal neurons. We previously showed that ethanol exposure during the first week of life blocks BDNF/L-VGCC-dependent long-term potentiation of GABAA receptor-mediated synaptic transmission in these neurons. Here, we tested whether this effect is associated with lasting alterations in GABAergic and glutamatergic transmission. Methods Rats were exposed to air or ethanol for 3 h/day between postnatal days three and five in vapor inhalation chambers, a paradigm that produces peak serum ethanol levels near 0.3 g/dl. Whole-cell patch-clamp electrophysiological recordings of spontaneous inhibitory and excitatory postsynaptic currents (sIPSCs and sEPSCs, respectively) were obtained from CA3 pyramidal neurons in coronal brain slices prepared at postnatal days 13–17. Results Ethanol exposure did not significantly affect the frequency, amplitude, rise-time and half-width of either sIPSCs or sEPSCs. Conclusions We show that an ethanol exposure paradigm known to inhibit synaptic plasticity mechanisms that may participate in the stabilization of GABAergic and glutamatergic synapses in CA3 pyramidal neurons does not produce lasting functional alterations in these synapses, suggesting that compensatory mechanisms restored the balance of excitatory and inhibitory synaptic transmission.
Collapse
Affiliation(s)
- Brian Charles Baculis
- Department of Neurosciences, School of Medicine, University of New Mexico Health Sciences Center, Albuquerque, NM, 87131, USA.
| | - Carlos Fernando Valenzuela
- Department of Neurosciences, School of Medicine, University of New Mexico Health Sciences Center, Albuquerque, NM, 87131, USA.
| |
Collapse
|
24
|
Abstract
The endogenous cannabinoid (endocannabinoid) system is an important regulator of synaptic function. Endocannabinoids acutely modulate inhibitory and excitatory transmission, and also mediate long-term depression at GABAergic and glutamatergic synapses. Typically, endocannabinoid synthesis and release is stimulated by depolarization-induced calcium influx and/or activation of phospholipase-C (PLC) signaling triggered by mGluR activation. Recently it has been shown that brain-derived neurotrophic factor (BDNF) can also induce endocannabinoid release. Although there is growing evidence for cross-talk between BDNF and endocannabinoid signaling, little is known about the functional relevance of these interactions. In the present studies, we examined BDNF - endocannabinoid interactions in regulating activity-dependent long-term depression at inhibitory synapses (iLTD). We found that theta burst stimulation (TBS) in layer 2/3 of mouse somatosensory cortical slices can induce a form of endocannabinoid-mediated iLTD that is independent of metabotropic glutamate receptor (mGluR) activation. This endocannabinoid-dependent iLTD, however, requires endogenous BDNF-trkB signaling, as it is blocked by a trk tyrosine kinase inhibitor and by a trkB receptor antagonist, and also requires activation of diacylglycerol lipase (DAG-lipase, DGL). In addition, endocannabinoid-mediated iLTD can be induced by combining a subthreshold concentration of exogenous BDNF with weak TBS stimulation that by itself is insufficient to induce iLTD. Taken together, our results suggest that TBS can induce the release of endogenous BDNF, which triggers DGL-dependent endocannabinoid mobilization and cannabinoid receptor-dependent iLTD at layer 2/3 cortical synapses.
Collapse
|
25
|
Pro-brain-derived neurotrophic factor inhibits GABAergic neurotransmission by activating endocytosis and repression of GABAA receptors. J Neurosci 2015; 34:13516-34. [PMID: 25274828 DOI: 10.1523/jneurosci.2069-14.2014] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
GABA is the canonical inhibitory neurotransmitter in the CNS. This inhibitory action is largely mediated by GABA type A receptors (GABAARs). Among the many factors controlling GABAergic transmission, brain-derived neurotrophic factor (BDNF) appears to play a major role in regulating synaptic inhibition. Recent findings have demonstrated that BDNF can be released as a precursor (proBDNF). Although the role of mature BDNF on GABAergic synaptogenesis and maintenance has been well studied, an important question still unanswered is whether secreted proBDNF might affect GABAergic neurotransmission. Here, we have used 14 d in vitro primary culture of hippocampal neurons and ex vivo preparations from rats to study the function of proBDNF in regulation of GABAAR trafficking and activity. We demonstrate that proBDNF impairs GABAergic transmission by the activation of two distinct pathways: (1) a RhoA-Rock-PTEN pathway that decreases the phosphorylation levels of GABAAR, thus affecting receptor function and triggering endocytosis and degradation of internalized receptors, and (2) a JAK-STAT-ICER pathway leading to the repression of GABAARs synthesis. These effects lead to the diminution of GABAergic synapses and are correlated with a decrease in GABAergic synaptic currents. These results revealed new functions for proBDNF-p75 neurotrophin receptor signaling pathway in the control of the efficacy of GABAergic synaptic activity by regulating the trafficking and synthesis of GABAARs at inhibitory synapses.
Collapse
|
26
|
|
27
|
Characterization of L-type voltage-gated Ca(2+) channel expression and function in developing CA3 pyramidal neurons. Neuroscience 2013; 238:59-70. [PMID: 23415785 DOI: 10.1016/j.neuroscience.2013.02.008] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2012] [Revised: 01/28/2013] [Accepted: 02/05/2013] [Indexed: 11/23/2022]
Abstract
Voltage-gated calcium channels (VGCCs) play a major role during the development of the central nervous system (CNS). Ca(2+) influx via VGCCs regulates axonal growth and neuronal migration as well as synaptic plasticity. Specifically, L-type VGCCs have been well characterized to be involved in the formation and refinement of the connections within the CA3 region of the hippocampus. The majority of the growth, formation, and refinement in the CNS occurs during the third trimester of human pregnancy. An equivalent developmental time period in rodents occurs during the first 2weeks of post-natal life, and the expression pattern of L-type VGCCs during this time period has not been well characterized. In this study, we show that Cav1.2 channels are more highly expressed during this developmental period compared to adolescence (post-natal day 30) and that L-type VGCCs significantly contribute to the overall Ca(2+) currents. These findings suggest that L-type VGCCs are functionally expressed during the crucial developmental period.
Collapse
|
28
|
Selective kinase inhibitors as tools for neuroscience research. Neuropharmacology 2012; 63:1227-37. [DOI: 10.1016/j.neuropharm.2012.07.024] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2012] [Revised: 07/06/2012] [Accepted: 07/11/2012] [Indexed: 01/02/2023]
|
29
|
Laudes T, Meis S, Munsch T, Lessmann V. Impaired transmission at corticothalamic excitatory inputs and intrathalamic GABAergic synapses in the ventrobasal thalamus of heterozygous BDNF knockout mice. Neuroscience 2012; 222:215-27. [DOI: 10.1016/j.neuroscience.2012.07.005] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2012] [Revised: 06/27/2012] [Accepted: 07/02/2012] [Indexed: 01/08/2023]
|
30
|
|
31
|
Everett JC, Licón-Muñoz Y, Valenzuela CF. Effects of third trimester-equivalent ethanol exposure on Cl(-) co-transporter expression, network activity, and GABAergic transmission in the CA3 hippocampal region of neonatal rats. Alcohol 2012; 46:595-601. [PMID: 22703993 PMCID: PMC3411872 DOI: 10.1016/j.alcohol.2012.04.003] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2011] [Revised: 04/26/2012] [Accepted: 04/26/2012] [Indexed: 12/30/2022]
Abstract
Fetal alcohol spectrum disorders are often associated with structural and functional hippocampal abnormalities, leading to long-lasting learning and memory deficits. The mechanisms underlying these abnormalities are not fully understood. Here, we investigated whether ethanol exposure during the 3rd trimester-equivalent period alters spontaneous network activity that is involved in neuronal circuit development in the CA3 hippocampal region. This activity is driven by GABA(A) receptors, which can have excitatory actions in developing neurons as a consequence of greater expression of the Cl(-) importer, NKCC1, with respect to expression of the Cl(-) exporter, KCC2, resulting in high [Cl(-)](i). Rat pups were exposed to ethanol vapor from postnatal day (P) 2-16 (4 h/day). Weight gain was significantly reduced in pups exposed to ethanol compared to control at P15 and 16. Brain slices were prepared immediately after the end of the 4-h exposure on P4-16 and experiments were also performed under ethanol-free conditions at the end of the exposure paradigm (P17-22). Ethanol exposure did not significantly affect expression of KCC2 or NKCC1, nor did it affect network activity in the CA3 hippocampal region. Ethanol exposure significantly decreased the frequency (at P9-11) and increased the amplitude (at P5-8 and P17-21) of GABA(A) receptor-mediated miniature postsynaptic currents. These data suggest that repeated in vivo exposure to ethanol during the 3rd trimester-equivalent period alters GABAergic transmission in the CA3 hippocampal region, an effect that could lead to abnormal circuit maturation and perhaps contribute to the pathophysiology of fetal alcohol spectrum disorders.
Collapse
Affiliation(s)
- Julie C Everett
- Department of Neurosciences, School of Medicine, University of New Mexico Health Sciences Center, Albuquerque, NM 87131, USA
| | | | | |
Collapse
|
32
|
Ben-Ari Y, Khalilov I, Kahle KT, Cherubini E. The GABA excitatory/inhibitory shift in brain maturation and neurological disorders. Neuroscientist 2012; 18:467-86. [PMID: 22547529 DOI: 10.1177/1073858412438697] [Citation(s) in RCA: 434] [Impact Index Per Article: 33.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Ionic currents and the network-driven patterns they generate differ in immature and adult neurons: The developing brain is not a "small adult brain." One of the most investigated examples is the developmentally regulated shift of actions of the transmitter GABA that inhibit adult neurons but excite immature ones because of an initially higher intracellular chloride concentration [Cl(-)](i), leading to depolarizing and often excitatory actions of GABA instead of hyperpolarizing and inhibitory actions. The levels of [Cl(-)](i) are also highly labile, being readily altered transiently or persistently by enhanced episodes of activity in relation to synaptic plasticity or a variety of pathological conditions, including seizures and brain insults. Among the plethora of channels, transporters, and other devices involved in controlling [Cl(-)](i), two have emerged as playing a particularly important role: the chloride importer NKCC1 and the chloride exporter KCC2. Here, the authors stress the importance of determining how [Cl(-)](i) is dynamically regulated and how this affects brain operation in health and disease. In a clinical perspective, agents that control [Cl(-)](i) and reinstate inhibitory actions of GABA open novel therapeutic perspectives in many neurological disorders, including infantile epilepsies, autism spectrum disorders, and other developmental disorders.
Collapse
|
33
|
Langlois A, Diabira D, Ferrand N, Porcher C, Gaiarsa JL. NMDA-dependent switch of proBDNF actions on developing GABAergic synapses. Cereb Cortex 2012; 23:1085-96. [PMID: 22510533 DOI: 10.1093/cercor/bhs071] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
The brain-derived neurotrophic factor (BDNF) has emerged as an important messenger for activity-dependent development of neuronal network. Recent findings have suggested that a significant proportion of BDNF can be secreted as a precursor (proBDNF) and cleaved by extracellular proteases to yield the mature form. While the actions of proBDNF on maturation and plasticity of excitatory synapses have been studied, the effect of the precursor on developing GABAergic synapses remains largely unknown. Here, we show that regulated secretion of proBDNF exerts a bidirectional control of GABAergic synaptic activity with NMDA receptors driving the polarity of the plasticity. When NMDA receptors are activated during ongoing synaptic activity, regulated Ca(2+)-dependent secretion of proBDNF signals via p75(NTR) to depress GABAergic synaptic activity, while in the absence of NMDA receptors activation, secreted proBDNF induces a p75(NTR)-dependent potentiation of GABAergic synaptic activity. These results revealed a new function for proBDNF-p75(NTR) signaling in synaptic plasticity and a novel mechanism by which synaptic activity can modulate the development of GABAergic synaptic connections.
Collapse
Affiliation(s)
- Anais Langlois
- Institut National de la Santé et de la Recherche Médicale Unité 901, Marseille 13009, France
| | | | | | | | | |
Collapse
|
34
|
Castillo PE. Presynaptic LTP and LTD of excitatory and inhibitory synapses. Cold Spring Harb Perspect Biol 2012; 4:cshperspect.a005728. [PMID: 22147943 DOI: 10.1101/cshperspect.a005728] [Citation(s) in RCA: 111] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Ubiquitous forms of long-term potentiation (LTP) and depression (LTD) are caused by enduring increases or decreases in neurotransmitter release. Such forms or presynaptic plasticity are equally observed at excitatory and inhibitory synapses and the list of locations expressing presynaptic LTP and LTD continues to grow. In addition to the mechanistically distinct forms of postsynaptic plasticity, presynaptic plasticity offers a powerful means to modify neural circuits. A wide range of induction mechanisms has been identified, some of which occur entirely in the presynaptic terminal, whereas others require retrograde signaling from the postsynaptic to presynaptic terminals. In spite of this diversity of induction mechanisms, some common induction rules can be identified across synapses. Although the precise molecular mechanism underlying long-term changes in transmitter release in most cases remains unclear, increasing evidence indicates that presynaptic LTP and LTD can occur in vivo and likely mediate some forms of learning.
Collapse
Affiliation(s)
- Pablo E Castillo
- Dominick P. Purpura Department of Neuroscience Albert Einstein College of Medicine, Bronx, New York 10461, USA.
| |
Collapse
|
35
|
Contribution of metabotropic GABA(B) receptors to neuronal network construction. Pharmacol Ther 2011; 132:170-9. [PMID: 21718720 DOI: 10.1016/j.pharmthera.2011.06.004] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2011] [Accepted: 06/08/2011] [Indexed: 01/05/2023]
Abstract
In the 1980s, Bowery and colleagues discovered the presence of a novel, bicuculline-resistant and baclofen-sensitive type of GABA receptor on peripheral nerve terminals, the GABA(B) receptor. Since this pioneering work, GABA(B) receptors have been identified in the Central Nervous System (CNS), where they provide an important inhibitory control of postsynaptic excitability and presynaptic transmitter release. GABA(B) receptors have been implicated in a number of important processes in the adult brain such as the regulation of synaptic plasticity and modulation of rhythmic activity. As a result of these studies, several potential therapeutic applications of GABA(B) receptor ligands have been identified. Recent advances have further shown that GABA(B) receptors play more than a classical inhibitory role in adult neurotransmission, and can in fact function as an important developmental signal early in life. Here we summarize current knowledge on the contribution of GABA(B) receptors to the construction and function of developing neuronal networks.
Collapse
|
36
|
Kuczewski N, Fuchs C, Ferrand N, Jovanovic JN, Gaiarsa JL, Porcher C. Mechanism of GABAB receptor-induced BDNF secretion and promotion of GABAA receptor membrane expression. J Neurochem 2011; 118:533-45. [PMID: 21255015 DOI: 10.1111/j.1471-4159.2011.07192.x] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Recent studies have shown that GABA(B) receptors play more than a classical inhibitory role and can function as an important synaptic maturation signal early in life. In a previous study, we reported that GABA(B) receptor activation triggers secretion of brain-derived neurotrophic factor (BDNF) and promotes the functional maturation of GABAergic synapses in the developing rat hippocampus. To identify the signalling pathway linking GABA(B) receptor activation to BDNF secretion in these cells, we have now used the phosphorylated form of the cAMP response element-binding protein as a biological sensor for endogenous BDNF release. In the present study, we show that GABA(B) receptor-induced secretion of BDNF relies on the activation of phospholipase C, followed by the formation of diacylglycerol, activation of protein kinase C, and the opening of L-type voltage-dependent Ca(2+) channels. We further show that once released by GABA(B) receptor activation, BDNF increases the membrane expression of β(2/3) -containing GABA(A) receptors in neuronal cultures. These results reveal a novel function of GABA(B) receptors in regulating the expression of GABA(A) receptor through BDNF-tropomyosin-related kinase B receptor dependent signalling pathway.
Collapse
Affiliation(s)
- Nicola Kuczewski
- INSERM (Institut National de la Santé et de la Recherche Médicale) Unité 901, Marseille, France.
| | | | | | | | | | | |
Collapse
|
37
|
Long-term plasticity at inhibitory synapses. Curr Opin Neurobiol 2011; 21:328-38. [PMID: 21334194 DOI: 10.1016/j.conb.2011.01.006] [Citation(s) in RCA: 165] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2010] [Accepted: 01/25/2011] [Indexed: 12/18/2022]
Abstract
Experience-dependent modifications of neural circuits and function are believed to heavily depend on changes in synaptic efficacy such as LTP/LTD. Hence, much effort has been devoted to elucidating the mechanisms underlying these forms of synaptic plasticity. Although most of this work has focused on excitatory synapses, it is now clear that diverse mechanisms of long-term inhibitory plasticity have evolved to provide additional flexibility to neural circuits. By changing the excitatory/inhibitory balance, GABAergic plasticity can regulate excitability, neural circuit function and ultimately, contribute to learning and memory, and neural circuit refinement. Here we discuss recent advancements in our understanding of the mechanisms and functional relevance of GABAergic inhibitory synaptic plasticity.
Collapse
|
38
|
Kuczewski N, Porcher C, Gaiarsa JL. Activity-dependent dendritic secretion of brain-derived neurotrophic factor modulates synaptic plasticity. Eur J Neurosci 2010; 32:1239-44. [DOI: 10.1111/j.1460-9568.2010.07378.x] [Citation(s) in RCA: 67] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
39
|
Gao HL, Xu H, Xin N, Zheng W, Chi ZH, Wang ZY. Disruption of the CaMKII/CREB signaling is associated with zinc deficiency-induced learning and memory impairments. Neurotox Res 2010; 19:584-91. [PMID: 20593259 DOI: 10.1007/s12640-010-9206-y] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2010] [Revised: 06/12/2010] [Accepted: 06/21/2010] [Indexed: 12/28/2022]
Abstract
Many studies have shown that zinc deficiency not only retards growth, but also affects several brain functions, including learning and memory. However, the underlying mechanism of impaired hippocampus-dependent learning and memory under zinc deficiency is poorly understood. In this study, young mice were fed a zinc-deficient diet (0.85 ppm) for 5 weeks. Morris water maze result showed that zinc deficiency results in spatial learning impairment. We then examined whether zinc depletion-induced learning and memory defects are associated with changes in signaling molecules essential for the expression of long-term potentiation. Immunoblot results showed that the protein levels of calmodulin (CaM), phosphorylated CaM-dependent protein kinase II (CaMKII), and phosphorylated cAMP-responsive element binding protein (CREB) were significantly reduced, whereas the total protein levels of CaMKII and CREB did not change in the zinc-deficient hippocampus. Thus, we provide a previously unrecognized mechanism whereby zinc deficiency impairs hippocampal learning and memory, at least in part, through disruption of the CaM/CaMKII/CREB signaling pathway.
Collapse
Affiliation(s)
- Hui-Ling Gao
- Key Lab of Cell Biology of Ministry of Education of China, China Medical University, Bei-Er Road 92, Shenyang 110001, People's Republic of China
| | | | | | | | | | | |
Collapse
|
40
|
Lamsa KP, Kullmann DM, Woodin MA. Spike-timing dependent plasticity in inhibitory circuits. Front Synaptic Neurosci 2010; 2:8. [PMID: 21423494 PMCID: PMC3059674 DOI: 10.3389/fnsyn.2010.00008] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2010] [Accepted: 05/17/2010] [Indexed: 01/21/2023] Open
Abstract
Inhibitory circuits in the brain rely on GABA-releasing interneurons. For long, inhibitory circuits were considered weakly plastic in the face of patterns of neuronal activity that trigger long-term changes in the synapses between excitatory principal cells. Recent studies however have shown that GABAergic circuits undergo various forms of long-term plasticity. For the purpose of this review, we identify three major long-term plasticity expression sites. The first locus is the glutamatergic synapses that excite GABAergic inhibitory cells and drive their activity. Such synapses, on many but not all inhibitory interneurons, exhibit long-term potentiation (LTP) and depression (LTD). Second, GABAergic synapses themselves can undergo changes in GABA release probability or postsynaptic GABA receptors. The third site of plasticity is in the postsynaptic anion gradient of GABAergic synapses; coincident firing of GABAergic axons and postsynaptic neurons can cause a long-lasting change in the reversal potential of GABAA receptors mediating fast inhibitory postsynaptic potentials. We review the recent literature on these forms of plasticity by asking how they may be triggered by specific patterns of pre- and postsynaptic action potentials, although very few studies have directly examined spike-timing dependent plasticity (STDP) protocols in inhibitory circuits. Plasticity of interneuron recruitment and of GABAergic signaling provides for a rich flexibility in inhibition that may be central to many aspects of brain function. We do not consider plasticity at glutamatergic synapses on Purkinje cells and other GABAergic principal cells.
Collapse
Affiliation(s)
- Karri P Lamsa
- Department of Pharmacology, Oxford University Oxford, UK
| | | | | |
Collapse
|
41
|
Low concentrations of alcohol inhibit BDNF-dependent GABAergic plasticity via L-type Ca2+ channel inhibition in developing CA3 hippocampal pyramidal neurons. J Neurosci 2010; 30:6776-81. [PMID: 20463239 DOI: 10.1523/jneurosci.5405-09.2010] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Fetal alcohol spectrum disorder (FASD) is associated with learning and memory alterations that could be, in part, a consequence of hippocampal damage. The CA3 hippocampal subfield is one of the regions affected by ethanol (EtOH), including exposure during the third trimester-equivalent (i.e., neonatal period in rats). However, the mechanism of action of EtOH is poorly understood. In CA3 pyramidal neurons from neonatal rats, dendritic BDNF release causes long-term potentiation of the frequency of GABAA receptor-mediated spontaneous postsynaptic currents (LTP-GABAA) and this mechanism is thought to play a role in GABAergic synapse maturation. Here, we show that short- and long-term exposure of neonatal male rats to low EtOH concentrations abolishes LTP-GABAA by inhibiting L-type voltage-gated Ca2+ channels. These findings support the recommendation that even light drinking should be avoided during pregnancy.
Collapse
|
42
|
Normal hearing is required for the emergence of long-lasting inhibitory potentiation in cortex. J Neurosci 2010; 30:331-41. [PMID: 20053914 DOI: 10.1523/jneurosci.4554-09.2010] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Long-term synaptic plasticity is a putative mechanism for learning in adults. However, there is little understanding of how synaptic plasticity mechanisms develop or whether their maturation depends on experience. Since inhibitory synapses are particularly malleable to sensory stimulation, long-lasting potentiation of inhibitory synapses was characterized in auditory thalamocortical slices. Intracortical high-frequency electrical stimulation led to a 67% increase in inhibitory synaptic currents. In the absence of stimulation, inhibitory potentiation was induced by a brief exposure to exogenous brain-derived neurotrophic factor (BDNF). BDNF exposure occluded any additional potentiation by high-frequency afferent stimulation, suggesting that BDNF signaling is sufficient to account for inhibitory potentiation. Moreover, inhibitory potentiation was reduced significantly by extracellular application of a BDNF scavenger or by intracellular blockade of BDNF receptor [tropomyosin-related kinase B (TrkB)] signaling. In contrast, glutamatergic or GABAergic antagonists did not prevent the induction of inhibitory potentiation. Since BDNF and TrkB expression are influenced strongly by activity, we predicted that inhibitory potentiation would be diminished by manipulations that decrease central auditory activity, such as hearing loss. Two forms of hearing loss were examined: conductive hearing loss in which the cochleae are not damaged or sensorineural hearing loss in which both cochleae are removed. Both forms of hearing loss were found to reduce significantly the magnitude of inhibitory potentiation. These data indicate that early experience is necessary for the normal development of BDNF-mediated long-lasting inhibitory potentiation, which may be associated with perceptual deficits at later ages.
Collapse
|
43
|
BDNF signaling in the formation, maturation and plasticity of glutamatergic and GABAergic synapses. Exp Brain Res 2009; 199:203-34. [PMID: 19777221 DOI: 10.1007/s00221-009-1994-z] [Citation(s) in RCA: 230] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2009] [Accepted: 08/12/2009] [Indexed: 01/17/2023]
Abstract
In the past 15 years numerous reports provided strong evidence that brain-derived neurotrophic factor (BDNF) is one of the most important modulators of glutamatergic and GABAergic synapses. Remarkable progress regarding localization, kinetics, and molecular mechanisms of BDNF secretion has been achieved, and a large number of studies provided evidence that continuous extracellular supply of BDNF is important for the proper formation and functional maturation of glutamatergic and GABAergic synapses. BDNF can play a permissive role in shaping synaptic networks, making them more susceptible for the occurrence of plastic changes. In addition, BDNF appears to be also an instructive factor for activity-dependent long-term synaptic plasticity. BDNF release just in response to synaptic stimulation might be a molecular trigger to convert high-frequency synaptic activity into long-term synaptic memories. This review attempts to summarize the current knowledge in synaptic secretion and synaptic action of BDNF, including both permissive and instructive effects of BDNF in synaptic plasticity.
Collapse
|
44
|
Mechanisms, locations, and kinetics of synaptic BDNF secretion: an update. Neurosci Res 2009; 65:11-22. [PMID: 19523993 DOI: 10.1016/j.neures.2009.06.004] [Citation(s) in RCA: 224] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2009] [Revised: 06/01/2009] [Accepted: 06/03/2009] [Indexed: 01/12/2023]
Abstract
Brain-derived neurotrophic factor (BDNF) and other members of the protein family of neurotrophins have been implicated in a multitude of processes that are important for neuronal development and synaptic plasticity in the rodent central nervous system. In comparison to the wealth of information available with respect to the biological functions of neurotrophins, our knowledge regarding the processes that govern synaptic secretion of neurotrophins is scarce. Using live cell imaging of GFP-tagged neurotrophins in primary neurons, immunocytochemical detection of endogenous BDNF in fixed cells, and by blocking the action of endogenously released BDNF by means of TrkB receptor bodies in living neurons, several studies in recent years have allowed to better understand the time course and the mechanisms of synaptic secretion of neurotrophins. This review will summarize the current knowledge regarding the intracellular processing of proneurotrophins, the targeting of neurotrophin vesicles to axons and dendrites, and the mechanisms of activity-dependent secretion of BDNF at synapses. Since these processes are known to be cell type dependent, special emphasis is given to observations gained from experiments in primary neurons.
Collapse
|
45
|
Abstract
The age of an experimental animal can be a critical variable, yet age matters are often overlooked within neuroscience. Many studies make use of young animals, without considering possible differences between immature and mature subjects. This is especially problematic when attempting to model traits or diseases that do not emerge until adulthood. In this commentary we discuss the reasons for this apparent bias in age of experimental animals, and illustrate the problem with a systematic review of published articles on long-term potentiation. Additionally, we review the developmental stages of a rat and discuss the difficulty of using the weight of an animal as a predictor of its age. Finally, we provide original data from our laboratory and review published data to emphasize that development is an ongoing process that does not end with puberty. Developmental changes can be quantitative in nature, involving gradual changes, rapid switches, or inverted U-shaped curves. Changes can also be qualitative. Thus, phenomena that appear to be unitary may be governed by different mechanisms at different ages. We conclude that selection of the age of the animals may be critically important in the design and interpretation of neurobiological studies.
Collapse
Affiliation(s)
- James Edgar McCutcheon
- Department of Cellular and Molecular Pharmacology, Rosalind Franklin University of Medicine and Science, The Chicago Medical School, 3333 Green Bay Road, North Chicago, IL 60064, USA
| | | |
Collapse
|
46
|
Trzaska KA, King CC, Li KY, Kuzhikandathil EV, Nowycky MC, Ye JH, Rameshwar P. Brain-derived neurotrophic factor facilitates maturation of mesenchymal stem cell-derived dopamine progenitors to functional neurons. J Neurochem 2009; 110:1058-69. [PMID: 19493166 DOI: 10.1111/j.1471-4159.2009.06201.x] [Citation(s) in RCA: 88] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The generation of dopamine (DA) neurons from stem cells holds great promise in the treatment of Parkinson's disease and other neural disease associated with dysfunction of DA neurons. Mesenchymal stem cells (MSCs) derived from the adult bone marrow show plasticity with regards to generating cells of other germ layers. In addition to reduced ethical concerns, MSCs could be transplanted across allogeneic barriers, making them desirable stem cells for clinical applications. We have reported on the generation of DA cells from human MSCs using sonic hedgehog (SHH), fibroblast growth factor 8 and basic fibroblast growth factor. Despite the secretion of DA, the cells did not show evidence of functional neurons, and were therefore designated DA progenitors. Here, we report on the role of brain-derived neurotrophic factor (BDNF) in the maturation of the MSC-derived DA progenitors. 9-day induced MSCs show significant tropomyosin-receptor-kinase B expression, which correlate with its ligand, BDNF, being able to induce functional maturation. The latter was based on Ca2+ imaging analyses and electrophysiology. BDNF-treated cells showed the following: increases in intracellular Ca2+ upon depolarization and after stimulation with the neurotransmitters acetylcholine and GABA and, post-synaptic currents by electrophysiological analyses. In addition, BDNF induced increased DA release upon depolarization. Taken together, these results demonstrate the crucial role for BDNF in the functional maturation of MSC-derived DA progenitors.
Collapse
Affiliation(s)
- Katarzyna A Trzaska
- Department of Medicine-Hematology/Oncology, New Jersey Medical School, University of Medicine and Dentistry of New Jersey, Newark, New Jersey, USA
| | | | | | | | | | | | | |
Collapse
|
47
|
At immature mossy-fiber-CA3 synapses, correlated presynaptic and postsynaptic activity persistently enhances GABA release and network excitability via BDNF and cAMP-dependent PKA. J Neurosci 2009; 29:2637-47. [PMID: 19244539 DOI: 10.1523/jneurosci.5019-08.2009] [Citation(s) in RCA: 71] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
In the adult rat hippocampus, the axons of granule cells in the dentate gyrus, the mossy fibers (MF), form excitatory glutamatergic synapses with CA3 principal cells. In neonates, MF release into their targets mainly GABA, which at this developmental stage is depolarizing. Here we tested the hypothesis that, at immature MF-CA3 synapses, correlated presynaptic [single fiber-evoked GABA(A)-mediated postsynaptic potentials (GPSPs)] and postsynaptic activity (back propagating action potentials) may exert a critical control on synaptic efficacy. This form of plasticity, called spike-timing-dependent plasticity (STDP), is a Hebbian type form of learning extensively studied at the level of glutamatergic synapses. Depending on the relative timing, pairing postsynaptic spiking and single MF-GPSPs induced bidirectional changes in synaptic efficacy. In case of positive pairing, spike-timing-dependent-long-term potentiation (STD-LTP) was associated with a persistent increase in GPSP slope and in the probability of cell firing. The transduction pathway involved a rise of calcium in the postsynaptic cell and the combined activity of cAMP-dependent PKA (protein kinase A) and brain-derived neurotrophic factor (BDNF). Retrograde signaling via BDNF and presynaptic TrkB receptors led to a persistent increase in GABA release. In "presynaptically" silent neurons, the enhanced probability of GABA release induced by the pairing protocol, unsilenced these synapses. Shifting E(GABA) from the depolarizing to the hyperpolarizing direction with bumetanide failed to modify synaptic strength. Thus, STD-LTP of GPSPs provides a reliable way to convey information from granule cells to the CA3 associative network at a time when glutamatergic synapses are still poorly developed.
Collapse
|
48
|
Activity-dependent dendritic release of BDNF and biological consequences. Mol Neurobiol 2009; 39:37-49. [PMID: 19156544 DOI: 10.1007/s12035-009-8050-7] [Citation(s) in RCA: 109] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2008] [Accepted: 01/07/2009] [Indexed: 01/08/2023]
Abstract
Network construction and reorganization is modulated by the level and pattern of synaptic activity generated in the nervous system. During the past decades, neurotrophins, and in particular brain-derived neurotrophic factor (BDNF), have emerged as attractive candidates for linking synaptic activity and brain plasticity. Thus, neurotrophin expression and secretion are under the control of activity-dependent mechanisms and, besides their classical role in supporting neuronal survival neurotrophins, modulate nearly all key steps of network construction from neuronal migration to experience-dependent refinement of local connections. In this paper, we provide an overview of recent findings showing that BDNF can serve as a target-derived messenger for activity-dependent synaptic plasticity and development at the single cell level.
Collapse
|
49
|
Kuczewski N, Langlois A, Fiorentino H, Bonnet S, Marissal T, Diabira D, Ferrand N, Porcher C, Gaiarsa JL. Spontaneous glutamatergic activity induces a BDNF-dependent potentiation of GABAergic synapses in the newborn rat hippocampus. J Physiol 2008; 586:5119-28. [PMID: 18772203 DOI: 10.1113/jphysiol.2008.158550] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
Spontaneous ongoing synaptic activity is thought to play an instructive role in the maturation of the neuronal circuits. However the type of synaptic activity involved and how this activity is translated into structural and functional changes is not fully understood. Here we show that ongoing glutamatergic synaptic activity triggers a long-lasting potentiation of gamma-aminobutyric acid (GABA) mediated synaptic activity (LLP(GABA-A)) in the developing rat hippocampus. LLP(GABA-A) induction requires (i) the activation of AMPA receptors and L-type voltage-dependent calcium channels, (ii) the release of endogenous brain-derived neurotrophic factor (BDNF), and (iii) the activation of postsynaptic tropomyosin-related kinase receptors B (TrkB). We found that spontaneous glutamatergic activity is required to maintain a high level of native BDNF in the newborn rat hippocampus and that application of exogenous BDNF induced LLP(GABA-A) in the absence of glutamatergic activity. These results suggest that ongoing glutamatergic synaptic activity plays a pivotal role in the functional maturation of hippocampal GABAergic synapses by means of a cascade involving BDNF release and downstream signalling through postsynaptic TrkB receptor activation.
Collapse
Affiliation(s)
- Nicola Kuczewski
- INSERM U901 (Institut National de la Santé et de la Recherche Médicale Unité 901), Marseille 13009, France
| | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Backpropagating action potentials trigger dendritic release of BDNF during spontaneous network activity. J Neurosci 2008; 28:7013-23. [PMID: 18596175 DOI: 10.1523/jneurosci.1673-08.2008] [Citation(s) in RCA: 93] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Brain-derived neurotrophic factor (BDNF) is a major regulator of activity-dependent synapse development and plasticity. Because BDNF is a secreted protein, it has been proposed that BDNF is released from target neurons in an activity-dependent manner. However, direct evidence for postsynaptic release of BDNF triggered by ongoing network activity is still lacking. Here we transfected cultures of dissociated hippocampal neurons with green fluorescent protein (GFP)-tagged BDNF and combined whole-cell recording, time-lapse fluorescent imaging, and immunostaining to monitor activity-dependent dendritic release of BDNF. We found that spontaneous backpropagating action potentials, but not synaptic activity alone, led to a Ca2+-dependent dendritic release of BDNF-GFP. Moreover, we provide evidence that endogenous BDNF released from a single neuron can phosphorylate CREB (cAMP response element-binding protein) in neighboring neurons, an important step of immediate early gene activation. Therefore, together, our results support the hypothesis that BDNF might act as a target-derived messenger of activity-dependent synaptic plasticity and development.
Collapse
|