1
|
Mesa H, Zhang EY, Wang Y, Zhang Q. Human neurons lacking amyloid precursor protein exhibit cholesterol-associated developmental and presynaptic deficits. J Cell Physiol 2024; 239:e30999. [PMID: 36966431 DOI: 10.1002/jcp.30999] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 01/29/2023] [Accepted: 03/06/2023] [Indexed: 03/27/2023]
Abstract
Amyloid precursor protein (APP) produces aggregable β-amyloid peptides and its mutations are associated with familial Alzheimer's disease (AD), which makes it one of the most studied proteins. However, APP's role in the human brain remains unclear despite years of investigation. One problem is that most studies on APP have been carried out in cell lines or model organisms, which are physiologically different from human neurons in the brain. Recently, human-induced neurons (hiNs) derived from induced pluripotent stem cells (iPSCs) provide a practical platform for studying the human brain in vitro. Here, we generated APP-null iPSCs using CRISPR/Cas9 genome editing technology and differentiate them into matured human neurons with functional synapses using a two-step procedure. During hiN differentiation and maturation, APP-null cells exhibited less neurite growth and reduced synaptogenesis in serum-free but not serum-containing media. We have found that cholesterol (Chol) remedies those developmental defects in APP-null cells, consistent with Chol's role in neurodevelopment and synaptogenesis. The phenotypic rescue was also achieved by coculturing those cells with wild-type mouse astrocytes, suggesting that APP's developmental role is likely astrocytic. Next, we examined matured hiNs using patch-clamp recording and detected reduced synaptic transmission in APP-null cells. This change was largely due to decreased synaptic vesicle (SV) release and retrieval, which was confirmed by live-cell imaging using two SV-specific fluorescent reporters. Adding Chol shortly before stimulation mitigated the SV deficits in APP-null iNs, indicating that APP facilitates presynaptic membrane Chol turnover during the SV exo-/endocytosis cycle. Taken together, our study in hiNs supports the notion that APP contributes to neurodevelopment, synaptogenesis, and neurotransmission via maintaining brain Chol homeostasis. Given the vital role of Chol in the central nervous system, the functional connection between APP and Chol bears important implications in the pathogenesis of AD.
Collapse
Affiliation(s)
- Haylee Mesa
- Stiles-Nicholson Brain Institute, Florida Atlantic University, Jupiter, Florida, USA
| | - Elaine Y Zhang
- Stiles-Nicholson Brain Institute, Florida Atlantic University, Jupiter, Florida, USA
- Brentwood High School, Brentwood, Tennessee, USA
| | - Yingcai Wang
- Department of Biomedical Sciences, Florida Atlantic University, Boca Raton, Florida, USA
| | - Qi Zhang
- Stiles-Nicholson Brain Institute, Florida Atlantic University, Jupiter, Florida, USA
- Department of Chemistry and Biochemistry, Florida Atlantic University, Boca Raton, Florida, USA
| |
Collapse
|
2
|
Zhou H, Bi GQ, Liu G. Intracellular magnesium optimizes transmission efficiency and plasticity of hippocampal synapses by reconfiguring their connectivity. Nat Commun 2024; 15:3406. [PMID: 38649706 PMCID: PMC11035601 DOI: 10.1038/s41467-024-47571-3] [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: 07/24/2023] [Accepted: 04/02/2024] [Indexed: 04/25/2024] Open
Abstract
Synapses at dendritic branches exhibit specific properties for information processing. However, how the synapses are orchestrated to dynamically modify their properties, thus optimizing information processing, remains elusive. Here, we observed at hippocampal dendritic branches diverse configurations of synaptic connectivity, two extremes of which are characterized by low transmission efficiency, high plasticity and coding capacity, or inversely. The former favors information encoding, pertinent to learning, while the latter prefers information storage, relevant to memory. Presynaptic intracellular Mg2+ crucially mediates the dynamic transition continuously between the two extreme configurations. Consequently, varying intracellular Mg2+ levels endow individual branches with diverse synaptic computations, thus modulating their ability to process information. Notably, elevating brain Mg2+ levels in aging animals restores synaptic configuration resembling that of young animals, coincident with improved learning and memory. These findings establish intracellular Mg2+ as a crucial factor reconfiguring synaptic connectivity at dendrites, thus optimizing their branch-specific properties in information processing.
Collapse
Affiliation(s)
- Hang Zhou
- Faculty of Life and Health Sciences, Shenzhen University of Advanced Technology, Shenzhen, 518107, China.
- Interdisciplinary Center for Brain Information, Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China.
| | - Guo-Qiang Bi
- Faculty of Life and Health Sciences, Shenzhen University of Advanced Technology, Shenzhen, 518107, China
- Interdisciplinary Center for Brain Information, Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
- Shenzhen-Hong Kong Institute of Brain Science, Shenzhen, 518055, China
- Hefei National Laboratory for Physical Sciences at the Microscale, and School of Life Sciences, University of Science and Technology of China, Hefei, 230031, China
| | - Guosong Liu
- School of Medicine, Tsinghua University, Beijing, 100084, China.
- NeuroCentria Inc., Walnut Creek, CA, 94596, USA.
| |
Collapse
|
3
|
D'Aloia A, Pastori V, Blasa S, Campioni G, Peri F, Sacco E, Ceriani M, Lecchi M, Costa B. A new advanced cellular model of functional cholinergic-like neurons developed by reprogramming the human SH-SY5Y neuroblastoma cell line. Cell Death Discov 2024; 10:24. [PMID: 38216593 PMCID: PMC10786877 DOI: 10.1038/s41420-023-01790-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Revised: 12/11/2023] [Accepted: 12/20/2023] [Indexed: 01/14/2024] Open
Abstract
Modeling human neuronal properties in physiological and pathological conditions is essential to identify novel potential drugs and to explore pathological mechanisms of neurological diseases. For this purpose, we generated a three-dimensional (3D) neuronal culture, by employing the readily available human neuroblastoma SH-SY5Y cell line, and a new differentiation protocol. The entire differentiation process occurred in a matrix and lasted 47 days, with 7 days of pre-differentiation phase and 40 days of differentiation, and allowed the development of a 3D culture in conditions consistent with the physiological environment. Neurons in the culture were electrically active, were able to establish functional networks, and showed features of cholinergic neurons. Hence here we provide an easily accessible, reproducible, and suitable culture method that might empower studies on synaptic function, vesicle trafficking, and metabolism, which sustain neuronal activity and cerebral circuits. Moreover, this novel differentiation protocol could represent a promising cellular tool to study physiological cellular processes, such as migration, differentiation, maturation, and to develop novel therapeutic approaches.
Collapse
Affiliation(s)
- Alessia D'Aloia
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Piazza della Scienza 2, 20126, Milano, Italy.
| | - Valentina Pastori
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Piazza della Scienza 2, 20126, Milano, Italy
| | - Stefania Blasa
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Piazza della Scienza 2, 20126, Milano, Italy
- Milan Center for Neuroscience (NeuroMI), University of Milano-Bicocca, Piazza dell'Ateneo Nuovo 1, 20126, Milano, Italy
| | - Gloria Campioni
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Piazza della Scienza 2, 20126, Milano, Italy
- SYSBIO-ISBE-IT, Europe, 20126, Milano, Italy
| | - Francesco Peri
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Piazza della Scienza 2, 20126, Milano, Italy
| | - Elena Sacco
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Piazza della Scienza 2, 20126, Milano, Italy
- Milan Center for Neuroscience (NeuroMI), University of Milano-Bicocca, Piazza dell'Ateneo Nuovo 1, 20126, Milano, Italy
- SYSBIO-ISBE-IT, Europe, 20126, Milano, Italy
- Inter-University Center for the Promotion of the 3Rs Principles in Teaching & Research, Pisa, Italy
| | - Michela Ceriani
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Piazza della Scienza 2, 20126, Milano, Italy
- Milan Center for Neuroscience (NeuroMI), University of Milano-Bicocca, Piazza dell'Ateneo Nuovo 1, 20126, Milano, Italy
| | - Marzia Lecchi
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Piazza della Scienza 2, 20126, Milano, Italy
- Milan Center for Neuroscience (NeuroMI), University of Milano-Bicocca, Piazza dell'Ateneo Nuovo 1, 20126, Milano, Italy
- Inter-University Center for the Promotion of the 3Rs Principles in Teaching & Research, Pisa, Italy
| | - Barbara Costa
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Piazza della Scienza 2, 20126, Milano, Italy
- Milan Center for Neuroscience (NeuroMI), University of Milano-Bicocca, Piazza dell'Ateneo Nuovo 1, 20126, Milano, Italy
| |
Collapse
|
4
|
Neher E. Interpretation of presynaptic phenotypes of synaptic plasticity in terms of a two-step priming process. J Gen Physiol 2024; 156:e202313454. [PMID: 38112713 PMCID: PMC10730358 DOI: 10.1085/jgp.202313454] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2023] Open
Abstract
Studies on synaptic proteins involved in neurotransmitter release often aim at distinguishing between their roles in vesicle priming (the docking of synaptic vesicles to the plasma membrane and the assembly of a release machinery) as opposed to the process of vesicle fusion. This has traditionally been done by estimating two parameters, the size of the pool of fusion-competent vesicles (the readily releasable pool, RRP) and the probability that such vesicles are released by an action potential, with the aim of determining how these parameters are affected by molecular perturbations. Here, it is argued that the assumption of a homogeneous RRP may be too simplistic and may blur the distinction between vesicle priming and fusion. Rather, considering priming as a dynamic and reversible multistep process allows alternative interpretations of mutagenesis-induced changes in synaptic transmission and suggests mechanisms for variability in synaptic strength and short-term plasticity among synapses, as well as for interactions between short- and long-term plasticity. In many cases, assigned roles of proteins or causes for observed phenotypes are shifted from fusion- to priming-related when considering multistep priming. Activity-dependent enhancement of priming is an essential element in this alternative view and its variation among synapse types can explain why some synapses show depression and others show facilitation at low to intermediate stimulation frequencies. Multistep priming also suggests a mechanism for frequency invariance of steady-state release, which can be observed in some synapses involved in sensory processing.
Collapse
Affiliation(s)
- Erwin Neher
- Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| |
Collapse
|
5
|
Longfield SF, Mollazade M, Wallis TP, Gormal RS, Joensuu M, Wark JR, van Waardenberg AJ, Small C, Graham ME, Meunier FA, Martínez-Mármol R. Tau forms synaptic nano-biomolecular condensates controlling the dynamic clustering of recycling synaptic vesicles. Nat Commun 2023; 14:7277. [PMID: 37949856 PMCID: PMC10638352 DOI: 10.1038/s41467-023-43130-4] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2022] [Accepted: 11/01/2023] [Indexed: 11/12/2023] Open
Abstract
Neuronal communication relies on the release of neurotransmitters from various populations of synaptic vesicles. Despite displaying vastly different release probabilities and mobilities, the reserve and recycling pool of vesicles co-exist within a single cluster suggesting that small synaptic biomolecular condensates could regulate their nanoscale distribution. Here, we performed a large-scale activity-dependent phosphoproteome analysis of hippocampal neurons in vitro and identified Tau as a highly phosphorylated and disordered candidate protein. Single-molecule super-resolution microscopy revealed that Tau undergoes liquid-liquid phase separation to generate presynaptic nanoclusters whose density and number are regulated by activity. This activity-dependent diffusion process allows Tau to translocate into the presynapse where it forms biomolecular condensates, to selectively control the mobility of recycling vesicles. Tau, therefore, forms presynaptic nano-biomolecular condensates that regulate the nanoscale organization of synaptic vesicles in an activity-dependent manner.
Collapse
Affiliation(s)
- Shanley F Longfield
- Clem Jones Centre for Ageing Dementia Research (CJCADR), Queensland Brain Institute (QBI), The University of Queensland; St Lucia Campus, Brisbane, QLD, 4072, Australia
| | - Mahdie Mollazade
- Clem Jones Centre for Ageing Dementia Research (CJCADR), Queensland Brain Institute (QBI), The University of Queensland; St Lucia Campus, Brisbane, QLD, 4072, Australia
| | - Tristan P Wallis
- Clem Jones Centre for Ageing Dementia Research (CJCADR), Queensland Brain Institute (QBI), The University of Queensland; St Lucia Campus, Brisbane, QLD, 4072, Australia
| | - Rachel S Gormal
- Clem Jones Centre for Ageing Dementia Research (CJCADR), Queensland Brain Institute (QBI), The University of Queensland; St Lucia Campus, Brisbane, QLD, 4072, Australia
| | - Merja Joensuu
- Clem Jones Centre for Ageing Dementia Research (CJCADR), Queensland Brain Institute (QBI), The University of Queensland; St Lucia Campus, Brisbane, QLD, 4072, Australia
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland; St Lucia Campus, Brisbane, QLD, 4072, Australia
| | - Jesse R Wark
- Synapse Proteomics, Children's Medical Research Institute (CMRI), The University of Sydney, 214 Hawkesbury Road, Westmead, NSW, 2145, Australia
| | | | - Christopher Small
- Clem Jones Centre for Ageing Dementia Research (CJCADR), Queensland Brain Institute (QBI), The University of Queensland; St Lucia Campus, Brisbane, QLD, 4072, Australia
| | - Mark E Graham
- Synapse Proteomics, Children's Medical Research Institute (CMRI), The University of Sydney, 214 Hawkesbury Road, Westmead, NSW, 2145, Australia
| | - Frédéric A Meunier
- Clem Jones Centre for Ageing Dementia Research (CJCADR), Queensland Brain Institute (QBI), The University of Queensland; St Lucia Campus, Brisbane, QLD, 4072, Australia.
- School of Biomedical Science, The University of Queensland; St Lucia Campus, Brisbane, QLD, 4072, Australia.
| | - Ramón Martínez-Mármol
- Clem Jones Centre for Ageing Dementia Research (CJCADR), Queensland Brain Institute (QBI), The University of Queensland; St Lucia Campus, Brisbane, QLD, 4072, Australia.
| |
Collapse
|
6
|
Zhong C, Akmentin W, Role LW, Talmage DA. Axonal α7* nicotinic acetylcholine receptors modulate glutamatergic signaling and synaptic vesicle organization in ventral hippocampal projections. Front Neural Circuits 2022; 16:978837. [PMID: 36213206 PMCID: PMC9537472 DOI: 10.3389/fncir.2022.978837] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2022] [Accepted: 09/01/2022] [Indexed: 11/13/2022] Open
Abstract
Modulation of the release of glutamate by activation of presynaptic nicotinic acetylcholine receptors (nAChRs) is one of the most prevalent mechanism of nicotinic facilitation of glutamatergic transmission in cortico-limbic circuits. By imaging gene chimeric co-cultures from mouse, we examined the role of α7* nAChRs mediated cholinergic modulation of glutamate release and synaptic vesicle organization in ventral hippocampal projections. We directly visualized exogenous and endogenous cholinergic facilitation of glutamate release in this specialized preparation of circuits in vitro. Disrupting α7* nAChRs mediated cholinergic signaling genetically or pharmacologically diminished cholinergic facilitation of glutamate release at presynaptic terminals. Alteration of α7* nAChRs mediated cholinergic signaling along glutamatergic axons also decreased functional synaptic vesicle clustering to presynaptic terminals. These findings suggest that presynaptic α7* nAChRs contribute to cholinergic modulation of glutamate release and synaptic vesicle organization.
Collapse
Affiliation(s)
- Chongbo Zhong
- National Institute of Neurological Disorders and Stroke, National Institutes of Health (NIH), Bethesda, MD, United States
| | - Wendy Akmentin
- Department of Neurobiology and Behavior, Center for Nervous System Disorders, Stony Brook University, Stony Brook, NY, United States
| | - Lorna W. Role
- National Institute of Neurological Disorders and Stroke, National Institutes of Health (NIH), Bethesda, MD, United States
| | - David A. Talmage
- National Institute of Neurological Disorders and Stroke, National Institutes of Health (NIH), Bethesda, MD, United States
| |
Collapse
|
7
|
Cary BA, Turrigiano GG. Stability of neocortical synapses across sleep and wake states during the critical period in rats. eLife 2021; 10:66304. [PMID: 34151775 PMCID: PMC8275129 DOI: 10.7554/elife.66304] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Accepted: 06/20/2021] [Indexed: 12/02/2022] Open
Abstract
Sleep is important for brain plasticity, but its exact function remains mysterious. An influential but controversial idea is that a crucial function of sleep is to drive widespread downscaling of excitatory synaptic strengths. Here, we used real-time sleep classification, ex vivo measurements of postsynaptic strength, and in vivo optogenetic monitoring of thalamocortical synaptic efficacy to ask whether sleep and wake states can constitutively drive changes in synaptic strength within the neocortex of juvenile rats. We found that miniature excitatory postsynaptic current amplitudes onto L4 and L2/3 pyramidal neurons were stable across sleep- and wake-dense epochs in both primary visual (V1) and prefrontal cortex (PFC). Further, chronic monitoring of thalamocortical synaptic efficacy in V1 of freely behaving animals revealed stable responses across even prolonged periods of natural sleep and wake. Together, these data demonstrate that sleep does not drive widespread downscaling of synaptic strengths during the highly plastic critical period in juvenile animals. Whether this remarkable stability across sleep and wake generalizes to the fully mature nervous system remains to be seen.
Collapse
Affiliation(s)
- Brian A Cary
- Department of Biology, Brandeis University, Waltham, United States
| | | |
Collapse
|
8
|
Thomas D, Rubio V, Iragavarapu V, Guzman E, Pelletier OB, Alamgir S, Zhang Q, Stawikowski MJ. Solvatochromic and pH-Sensitive Fluorescent Membrane Probes for Imaging of Live Cells. ACS Chem Neurosci 2021; 12:719-734. [PMID: 33508202 DOI: 10.1021/acschemneuro.0c00732] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Membrane trafficking is essential for all cells, and visualizing it is particularly useful for studying neuronal functions. Here we report the synthesis, characterization, and application of several membrane- and pH-sensitive probes suitable for live-cell fluorescence imaging. These probes are based on a 1,8-naphthalimide fluorophore scaffold. They exhibit a solvatochromic effect, and one of them, ND6, shows a substantial fluorescence difference between pH 6 and 7. The solvatochromic effect and pH-sensitivity of those probes are explained using quantum chemical calculations, and molecular dynamics simulation confirms their integration and interaction with membrane lipids. For live-cell fluorescence imaging, we tested those probes in a cancer cell line (MCF7), cancer spheroids (MDA-MB-468), and cultured hippocampal neurons. Confocal imaging showed an excellent signal-to-noise ratio from 400:1 to about 1300:1 for cell membrane labeling. We applied ND6 during stimulation to label nerve terminals via dye uptake during evoked synaptic vesicle turnover. By ND6 imaging, we revealed cholesterol's multifaced role in replenishing synaptic vesicle pools. Our results demonstrate these fluorescent probes' great potential in studying membrane dynamic and synaptic functions in neurons and other secretory cells and tissues.
Collapse
Affiliation(s)
- Deborah Thomas
- Department of Chemistry and Biochemistry, Florida Atlantic University, Boca Raton, Florida, United States
| | - Vicente Rubio
- Department of Chemistry and Biochemistry, Florida Atlantic University, Boca Raton, Florida, United States
| | - Vijaya Iragavarapu
- Charles E. Schmidt College of Medicine, Florida Atlantic University, Boca Raton, Florida, United States
| | - Esther Guzman
- Harbor Branch Oceanographic Institute, Florida Atlantic University, Fort Pierce, Florida, United States
| | - Oliver B. Pelletier
- Department of Biomedical Sciences, Charles E. Schmidt College of Medicine, Florida Atlantic University, Boca Raton, Florida, United States
| | - Shahriar Alamgir
- Department of Biomedical Sciences, Charles E. Schmidt College of Medicine, Florida Atlantic University, Boca Raton, Florida, United States
- The Brain Institute, Florida Atlantic University, 5353 Parkside Drive, Jupiter, Florida, United States
| | - Qi Zhang
- Department of Biomedical Sciences, Charles E. Schmidt College of Medicine, Florida Atlantic University, Boca Raton, Florida, United States
- The Brain Institute, Florida Atlantic University, 5353 Parkside Drive, Jupiter, Florida, United States
| | - Maciej J. Stawikowski
- Department of Chemistry and Biochemistry, Florida Atlantic University, Boca Raton, Florida, United States
| |
Collapse
|
9
|
Nagaraja C. Ventral root evoked entrainment of disinhibited bursts across early postnatal development in mice. IBRO Rep 2020; 9:310-318. [PMID: 33294722 PMCID: PMC7689330 DOI: 10.1016/j.ibror.2020.10.005] [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: 07/17/2020] [Accepted: 10/25/2020] [Indexed: 12/03/2022] Open
Abstract
Ventral root evoked entrainment of disinhibited bursts can be elicited in P24 spinal cord preparations. Disinhibited bursting and dorsal root evoked entrainment can be elicited even at P39. Ventral root evoked entrainment shows a decline from P0−15, and the coefficient of variation increases during this period. Ventral root evoked entrainment decays after a trial and shows some recovery after long periods following a trial. Dopamine D2 receptor antagonists and mGluR1 agonists can enhance ventral root evoked entrainment.
Early in the postnatal period, motoneuron axon stimulation can excite motor networks in the spinal cord. Here we tested if these excitatory effects changed across early postnatal development up to postnatal day (P) 24 by when mice are capable of weight-bearing locomotion and locomotor networks are considered functionally mature. This was accomplished in the isolated spinal cord preparation using ventral root evoked entrainment of disinhibited bursts. Ventral root evoked entrainment was defined and characterized over the first 2 weeks of postnatal development, and was found to decline over this period, but entrainment could still be detected in mice as old as P24. Disinhibited bursting could be elicited, and dorsal root evoked entrainment could be recorded as late as P39 and remained unchanged in effectiveness, suggesting that poor tissue viability may not be the cause of the decline in ventral root evoked entrainment. Pharmacological experiments performed on younger animals established that dopamine D2 receptor antagonists and mGluR1 agonists both enhanced ventral root evoked entrainment. In conclusion, the motoneuronal inputs to spinal motor networks via the excitatory pathway is modulated by dopamine and metabotropic glutamate receptors and may be under powerful inhibitory control, which may explain why there is a developmental decline in entrainment.
Collapse
Affiliation(s)
- Chetan Nagaraja
- Developmental Neurobiology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, 20892, United States
| |
Collapse
|
10
|
Genç Ö, An JY, Fetter RD, Kulik Y, Zunino G, Sanders SJ, Davis GW. Homeostatic plasticity fails at the intersection of autism-gene mutations and a novel class of common genetic modifiers. eLife 2020; 9:55775. [PMID: 32609087 PMCID: PMC7394548 DOI: 10.7554/elife.55775] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Accepted: 06/07/2020] [Indexed: 01/08/2023] Open
Abstract
We identify a set of common phenotypic modifiers that interact with five independent autism gene orthologs (RIMS1, CHD8, CHD2, WDFY3, ASH1L) causing a common failure of presynaptic homeostatic plasticity (PHP) in Drosophila. Heterozygous null mutations in each autism gene are demonstrated to have normal baseline neurotransmission and PHP. However, PHP is sensitized and rendered prone to failure. A subsequent electrophysiology-based genetic screen identifies the first known heterozygous mutations that commonly genetically interact with multiple ASD gene orthologs, causing PHP to fail. Two phenotypic modifiers identified in the screen, PDPK1 and PPP2R5D, are characterized. Finally, transcriptomic, ultrastructural and electrophysiological analyses define one mechanism by which PHP fails; an unexpected, maladaptive up-regulation of CREG, a conserved, neuronally expressed, stress response gene and a novel repressor of PHP. Thus, we define a novel genetic landscape by which diverse, unrelated autism risk genes may converge to commonly affect the robustness of synaptic transmission.
Collapse
Affiliation(s)
- Özgür Genç
- Department of Biochemistry and Biophysics Kavli Institute for Fundamental Neuroscience University of California, San Francisco, San Francisco, United States
| | - Joon-Yong An
- Department of Psychiatry UCSF Weill Institute for Neurosciences University of California, San Francisco, San Francisco, United States.,School of Biosystem and Biomedical Science, College of Health Science, Korea University, Seoul, Republic of Korea
| | - Richard D Fetter
- Department of Biochemistry and Biophysics Kavli Institute for Fundamental Neuroscience University of California, San Francisco, San Francisco, United States
| | - Yelena Kulik
- Department of Biochemistry and Biophysics Kavli Institute for Fundamental Neuroscience University of California, San Francisco, San Francisco, United States
| | - Giulia Zunino
- Department of Biochemistry and Biophysics Kavli Institute for Fundamental Neuroscience University of California, San Francisco, San Francisco, United States
| | - Stephan J Sanders
- Department of Psychiatry UCSF Weill Institute for Neurosciences University of California, San Francisco, San Francisco, United States
| | - Graeme W Davis
- Department of Biochemistry and Biophysics Kavli Institute for Fundamental Neuroscience University of California, San Francisco, San Francisco, United States
| |
Collapse
|
11
|
A Slow Short-Term Depression at Purkinje to Deep Cerebellar Nuclear Neuron Synapses Supports Gain-Control and Linear Encoding over Second-Long Time Windows. J Neurosci 2020; 40:5937-5953. [PMID: 32554551 DOI: 10.1523/jneurosci.2078-19.2020] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Revised: 04/21/2020] [Accepted: 05/23/2020] [Indexed: 11/21/2022] Open
Abstract
Modifications in the sensitivity of neural elements allow the brain to adapt its functions to varying demands. Frequency-dependent short-term synaptic depression (STD) provides a dynamic gain-control mechanism enabling adaptation to different background conditions alongside enhanced sensitivity to input-driven changes in activity. In contrast, synapses displaying frequency-invariant transmission can faithfully transfer ongoing presynaptic rates enabling linear processing, deemed critical for many functions. However, rigid frequency-invariant transmission may lead to runaway dynamics and low sensitivity to changes in rate. Here, I investigated the Purkinje cell to deep cerebellar nuclei neuron synapses (PC_DCNs), which display frequency invariance, and yet, PCs maintain background activity at disparate rates, even at rest. Using protracted PC_DCN activation (120 s) to mimic background activity in cerebellar slices from mature mice of both sexes, I identified a previously unrecognized, frequency-dependent, slow STD (S-STD), adapting IPSC amplitudes in tens of seconds to minutes. However, after changes in activation rates, over a behavior-relevant second-long time window, S-STD enabled scaled linear encoding of PC rates in synaptic charge transfer and DCN spiking activity. Combined electrophysiology, optogenetics, and statistical analysis suggested that S-STD mechanism is input-specific, involving decreased ready-to-release quanta, and distinct from faster short-term plasticity (f-STP). Accordingly, an S-STD component with a scaling effect (i.e., activity-dependent release sites inactivation), extending a model explaining PC_DCN release on shorter timescales using balanced f-STP, reproduced the experimental results. Thus, these results elucidates a novel slow gain-control mechanism able to support linear transfer of behavior-driven/learned PC rates concurrently with background activity adaptation, and furthermore, provides an alternative pathway to refine PC output.SIGNIFICANCE STATEMENT The brain can adapt to varying demands by dynamically changing the gain of its synapses; however, some tasks require ongoing linear transfer of presynaptic rates, seemingly incompatible with nonlinear gain adaptation. Here, I report a novel slow gain-control mechanism enabling scaled linear encoding of presynaptic rates over behavior-relevant time windows, and adaptation to background activity at the Purkinje to deep cerebellar nuclear neurons synapses (PC_DCNs). A previously unrecognized PC_DCNs slow and frequency-dependent short-term synaptic depression (S-STD) mediates this process. Experimental evidence and simulations suggested that scaled linear encoding emerges from the combination of S-STD slow dynamics and frequency-invariant transmission at faster timescales. These results demonstrate a mechanism reconciling rate code with background activity adaptation and suitable for flexibly tuning PCs output via background activity modulation.
Collapse
|
12
|
Ge D, Noakes PG, Lavidis NA. What are Neurotransmitter Release Sites and Do They Interact? Neuroscience 2020; 425:157-168. [DOI: 10.1016/j.neuroscience.2019.11.017] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2019] [Revised: 11/10/2019] [Accepted: 11/11/2019] [Indexed: 12/22/2022]
|
13
|
Yamashita M, Kawaguchi SY, Hori T, Takahashi T. Vesicular GABA Uptake Can Be Rate Limiting for Recovery of IPSCs from Synaptic Depression. Cell Rep 2019; 22:3134-3141. [PMID: 29562170 DOI: 10.1016/j.celrep.2018.02.080] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2016] [Revised: 01/05/2018] [Accepted: 02/21/2018] [Indexed: 11/16/2022] Open
Abstract
Synaptic efficacy plays crucial roles in neuronal circuit operation and synaptic plasticity. Presynaptic determinants of synaptic efficacy are neurotransmitter content in synaptic vesicles and the number of vesicles undergoing exocytosis at a time. Bursts of presynaptic firings depress synaptic efficacy, mainly due to depletion of releasable vesicles, whereas recovery from strong depression is initiated by endocytic vesicle retrieval followed by refilling of vesicles with neurotransmitter. We washed out presynaptic cytosolic GABA to induce a rundown of IPSCs at cerebellar inhibitory cell pairs in slices from rats and then allowed fast recovery by elevating GABA concentration using photo-uncaging. The time course of this recovery coincided with that of IPSCs from activity-dependent depression induced by a train of high-frequency stimulation. We conclude that vesicular GABA uptake can be a limiting step for the recovery of inhibitory neurotransmission from synaptic depression.
Collapse
Affiliation(s)
- Manami Yamashita
- Laboratory of Molecular Synaptic Function, Graduate School of Brain Science, Doshisha University, Kyoto 610-0394, Japan; Department of Physiology, Faculty of Medicine, Osaka Medical College, Osaka 569-8686, Japan
| | - Shin-Ya Kawaguchi
- Society-Academia Collaboration for Innovation, Kyoto University, Kyoto 606-8501, Japan
| | - Tetsuya Hori
- Department of Neurophysiology, Graduate School of Life and Medical Sciences, Doshisha University, Kyoto 610-0394, Japan.
| | - Tomoyuki Takahashi
- Cellular and Molecular Synaptic Function Unit, Okinawa Institute of Science and Technology (OIST) Graduate University, Okinawa 904-0495, Japan.
| |
Collapse
|
14
|
Frere S, Slutsky I. Alzheimer's Disease: From Firing Instability to Homeostasis Network Collapse. Neuron 2019; 97:32-58. [PMID: 29301104 DOI: 10.1016/j.neuron.2017.11.028] [Citation(s) in RCA: 172] [Impact Index Per Article: 28.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2017] [Revised: 11/14/2017] [Accepted: 11/17/2017] [Indexed: 12/22/2022]
Abstract
Alzheimer's disease (AD) starts from pure cognitive impairments and gradually progresses into degeneration of specific brain circuits. Although numerous factors initiating AD have been extensively studied, the common principles underlying the transition from cognitive deficits to neuronal loss remain unknown. Here we describe an evolutionarily conserved, integrated homeostatic network (IHN) that enables functional stability of central neural circuits and safeguards from neurodegeneration. We identify the critical modules comprising the IHN and propose a central role of neural firing in controlling the complex homeostatic network at different spatial scales. We hypothesize that firing instability and impaired synaptic plasticity at early AD stages trigger a vicious cycle, leading to dysregulation of the whole IHN. According to this hypothesis, the IHN collapse represents the major driving force of the transition from early memory impairments to neurodegeneration. Understanding the core elements of homeostatic control machinery, the reciprocal connections between distinct IHN modules, and the role of firing homeostasis in this hierarchy has important implications for physiology and should offer novel conceptual approaches for AD and other neurodegenerative disorders.
Collapse
Affiliation(s)
- Samuel Frere
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, 69978 Tel Aviv, Israel
| | - Inna Slutsky
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, 69978 Tel Aviv, Israel; Sagol School of Neuroscience, Tel Aviv University, 69978 Tel Aviv, Israel.
| |
Collapse
|
15
|
Toth AB, Hori K, Novakovic MM, Bernstein NG, Lambot L, Prakriya M. CRAC channels regulate astrocyte Ca 2+ signaling and gliotransmitter release to modulate hippocampal GABAergic transmission. Sci Signal 2019; 12:12/582/eaaw5450. [PMID: 31113852 DOI: 10.1126/scisignal.aaw5450] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Astrocytes are the major glial subtype in the brain and mediate numerous functions ranging from metabolic support to gliotransmitter release through signaling mechanisms controlled by Ca2+ Despite intense interest, the Ca2+ influx pathways in astrocytes remain obscure, hindering mechanistic insights into how Ca2+ signaling is coupled to downstream astrocyte-mediated effector functions. Here, we identified store-operated Ca2+ release-activated Ca2+ (CRAC) channels encoded by Orai1 and STIM1 as a major route of Ca2+ entry for driving sustained and oscillatory Ca2+ signals in astrocytes after stimulation of metabotropic purinergic and protease-activated receptors. Using synaptopHluorin as an optical reporter, we showed that the opening of astrocyte CRAC channels stimulated vesicular exocytosis to mediate the release of gliotransmitters, including ATP. Furthermore, slice electrophysiological recordings showed that activation of astrocytes by protease-activated receptors stimulated interneurons in the CA1 hippocampus to increase inhibitory postsynaptic currents on CA1 pyramidal cells. These results reveal a central role for CRAC channels as regulators of astrocyte Ca2+ signaling, gliotransmitter release, and astrocyte-mediated tonic inhibition of CA1 pyramidal neurons.
Collapse
Affiliation(s)
- Anna B Toth
- Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Kotaro Hori
- Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Michaela M Novakovic
- Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Natalie G Bernstein
- Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Laurie Lambot
- Department of Physiology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Murali Prakriya
- Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA.
| |
Collapse
|
16
|
Ortega JM, Genç Ö, Davis GW. Molecular mechanisms that stabilize short term synaptic plasticity during presynaptic homeostatic plasticity. eLife 2018; 7:40385. [PMID: 30422113 PMCID: PMC6250423 DOI: 10.7554/elife.40385] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2018] [Accepted: 11/12/2018] [Indexed: 11/13/2022] Open
Abstract
Presynaptic homeostatic plasticity (PHP) compensates for impaired postsynaptic neurotransmitter receptor function through a rapid, persistent adjustment of neurotransmitter release, an effect that can exceed 200%. An unexplained property of PHP is the preservation of short-term plasticity (STP), thereby stabilizing activity-dependent synaptic information transfer. We demonstrate that the dramatic potentiation of presynaptic release during PHP is achieved while simultaneously maintaining a constant ratio of primed to super-primed synaptic vesicles, thereby preserving STP. Mechanistically, genetic, biochemical and electrophysiological evidence argue that a constant ratio of primed to super-primed synaptic vesicles is achieved by the concerted action of three proteins: Unc18, Syntaxin1A and RIM. Our data support a model based on the regulated availability of Unc18 at the presynaptic active zone, a process that is restrained by Syntaxin1A and facilitated by RIM. As such, regulated vesicle priming/super-priming enables PHP to stabilize both synaptic gain and the activity-dependent transfer of information at a synapse.
Collapse
Affiliation(s)
- Jennifer M Ortega
- Department of Biochemistry and Biophysics, Kavli Institute for Fundamental Neuroscience, University of California San Francisco, San Francisco, California
| | - Özgür Genç
- Department of Biochemistry and Biophysics, Kavli Institute for Fundamental Neuroscience, University of California San Francisco, San Francisco, California
| | - Graeme W Davis
- Department of Biochemistry and Biophysics, Kavli Institute for Fundamental Neuroscience, University of California San Francisco, San Francisco, California
| |
Collapse
|
17
|
Ghelani T, Sigrist SJ. Coupling the Structural and Functional Assembly of Synaptic Release Sites. Front Neuroanat 2018; 12:81. [PMID: 30386217 PMCID: PMC6198076 DOI: 10.3389/fnana.2018.00081] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Accepted: 09/18/2018] [Indexed: 01/04/2023] Open
Abstract
Information processing in our brains depends on the exact timing of calcium (Ca2+)-activated exocytosis of synaptic vesicles (SVs) from unique release sites embedded within the presynaptic active zones (AZs). While AZ scaffolding proteins obviously provide an efficient environment for release site function, the molecular design creating such release sites had remained unknown for a long time. Recent advances in visualizing the ultrastructure and topology of presynaptic protein architectures have started to elucidate how scaffold proteins establish “nanodomains” that connect voltage-gated Ca2+ channels (VGCCs) physically and functionally with release-ready SVs. Scaffold proteins here seem to operate as “molecular rulers or spacers,” regulating SV-VGCC physical distances within tens of nanometers and, thus, influence the probability and plasticity of SV release. A number of recent studies at Drosophila and mammalian synapses show that the stable positioning of discrete clusters of obligate release factor (M)Unc13 defines the position of SV release sites, and the differential expression of (M)Unc13 isoforms at synapses can regulate SV-VGCC coupling. We here review the organization of matured AZ scaffolds concerning their intrinsic organization and role for release site formation. Moreover, we also discuss insights into the developmental sequence of AZ assembly, which often entails a tightening between VGCCs and SV release sites. The findings discussed here are retrieved from vertebrate and invertebrate preparations and include a spectrum of methods ranging from cell biology, super-resolution light and electron microscopy to biophysical and electrophysiological analysis. Our understanding of how the structural and functional organization of presynaptic AZs are coupled has matured, as these processes are crucial for the understanding of synapse maturation and plasticity, and, thus, accurate information transfer and storage at chemical synapses.
Collapse
Affiliation(s)
- Tina Ghelani
- Faculty of Biology, Chemistry, Pharmacy, Freie Universität Berlin, Berlin, Germany
| | - Stephan J Sigrist
- Faculty of Biology, Chemistry, Pharmacy, Freie Universität Berlin, Berlin, Germany.,NeuroCure Cluster of Excellence, Charité Universitätsmedizin Berlin, Berlin, Germany
| |
Collapse
|
18
|
Styr B, Slutsky I. Imbalance between firing homeostasis and synaptic plasticity drives early-phase Alzheimer's disease. Nat Neurosci 2018; 21:463-473. [PMID: 29403035 DOI: 10.1038/s41593-018-0080-x] [Citation(s) in RCA: 219] [Impact Index Per Article: 31.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2017] [Accepted: 01/08/2018] [Indexed: 12/18/2022]
Abstract
During recent years, the preclinical stage of Alzheimer's disease (AD) has become a major focus of research. Continued failures in clinical trials and the realization that early intervention may offer better therapeutic outcome triggered a conceptual shift from late-stage AD pathology to early-stage pathophysiology. While much effort has been directed at understanding the factors initiating AD, little is known about the principle basis underlying the disease progression at its early stages. In this Perspective, we suggest a hypothesis to explain the transition from 'silent' signatures of aberrant neural circuit activity to clinically evident memory impairments. Namely, we propose that failures in firing homeostasis and imbalance between firing stability and synaptic plasticity in cortico-hippocampal circuits represent the driving force of early disease progression. We analyze the main types of possible homeostatic failures and provide the essential conceptual framework for examining the causal link between dysregulation of firing homeostasis, aberrant neural circuit activity and memory-related plasticity impairments associated with early AD.
Collapse
Affiliation(s)
- Boaz Styr
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Inna Slutsky
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel. .,Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel.
| |
Collapse
|
19
|
Hauswirth AG, Ford KJ, Wang T, Fetter RD, Tong A, Davis GW. A postsynaptic PI3K-cII dependent signaling controller for presynaptic homeostatic plasticity. eLife 2018; 7:31535. [PMID: 29303480 PMCID: PMC5773188 DOI: 10.7554/elife.31535] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2017] [Accepted: 01/04/2018] [Indexed: 01/29/2023] Open
Abstract
Presynaptic homeostatic plasticity stabilizes information transfer at synaptic connections in organisms ranging from insect to human. By analogy with principles of engineering and control theory, the molecular implementation of PHP is thought to require postsynaptic signaling modules that encode homeostatic sensors, a set point, and a controller that regulates transsynaptic negative feedback. The molecular basis for these postsynaptic, homeostatic signaling elements remains unknown. Here, an electrophysiology-based screen of the Drosophila kinome and phosphatome defines a postsynaptic signaling platform that includes a required function for PI3K-cII, PI3K-cIII and the small GTPase Rab11 during the rapid and sustained expression of PHP. We present evidence that PI3K-cII localizes to Golgi-derived, clathrin-positive vesicles and is necessary to generate an endosomal pool of PI(3)P that recruits Rab11 to recycling endosomal membranes. A morphologically distinct subdivision of this platform concentrates postsynaptically where we propose it functions as a homeostatic controller for retrograde, trans-synaptic signaling.
Collapse
Affiliation(s)
- Anna G Hauswirth
- Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, United States.,Kavli Institute for Fundamental Neuroscience, University of California, San Francisco, San Francisco, United States
| | - Kevin J Ford
- Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, United States.,Kavli Institute for Fundamental Neuroscience, University of California, San Francisco, San Francisco, United States
| | - Tingting Wang
- Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, United States.,Kavli Institute for Fundamental Neuroscience, University of California, San Francisco, San Francisco, United States
| | - Richard D Fetter
- Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, United States.,Kavli Institute for Fundamental Neuroscience, University of California, San Francisco, San Francisco, United States
| | - Amy Tong
- Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, United States.,Kavli Institute for Fundamental Neuroscience, University of California, San Francisco, San Francisco, United States
| | - Graeme W Davis
- Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, United States.,Kavli Institute for Fundamental Neuroscience, University of California, San Francisco, San Francisco, United States
| |
Collapse
|
20
|
Flick B, Schneider S, Melching-Kollmuss S, Fussell KC, Gröters S, Buesen R, Strauss V, van Ravenzwaay B. Investigations of putative reproductive toxicity of low-dose exposures to vinclozolin in Wistar rats. Arch Toxicol 2016; 91:1941-1956. [PMID: 27612472 DOI: 10.1007/s00204-016-1811-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2016] [Accepted: 08/04/2016] [Indexed: 01/11/2023]
Abstract
The current investigation examines whether the fungicide vinclozolin, which has an anti-androgenic mode of action, is capable of disrupting endocrine homeostasis at very low doses. The data generated clarify whether a non-monotonic dose-response relationship exists to enhance the current debate about the regulation of endocrine disruptors. Moreover, it is part of a series of investigations assessing the dose-response relationship of single and combined administration of anti-androgenic substances. A pre-postnatal in vivo study design was chosen which was compliant with regulatory testing protocols. The test design was improved by additional endpoints addressing hormone levels, morphology and histopathological examinations. Doses were chosen to represent an effect level (20 mg/kg bw/d), the current NOAEL (4 mg/kg bw/d), and a dose close to the "ADI" (0.005 mg/kg bw/d) for the detection of a possible non-monotonic dose-response curve. Anti-androgenic changes were observable at the effect level but not at lower exposures. Nipple/areola counts appeared to be the most sensitive measure of effect, followed by male sex organ weights at sexual maturation, and finally gross and histopathological findings. The results indicate the absence of evidence for effects at low or very low dose levels. A non-monotonic dose-response relationship was not evident.
Collapse
Affiliation(s)
- Burkhard Flick
- Experimental Toxicology and Ecology, BASF SE, 67056, Ludwigshafen, Germany
| | - Steffen Schneider
- Experimental Toxicology and Ecology, BASF SE, 67056, Ludwigshafen, Germany
| | | | | | - Sibylle Gröters
- Experimental Toxicology and Ecology, BASF SE, 67056, Ludwigshafen, Germany
| | - Roland Buesen
- Experimental Toxicology and Ecology, BASF SE, 67056, Ludwigshafen, Germany
| | - Volker Strauss
- Experimental Toxicology and Ecology, BASF SE, 67056, Ludwigshafen, Germany
| | | |
Collapse
|
21
|
Sun Q, Weinger JG, Mao F, Liu G. Regulation of structural and functional synapse density by L-threonate through modulation of intraneuronal magnesium concentration. Neuropharmacology 2016; 108:426-39. [PMID: 27178134 DOI: 10.1016/j.neuropharm.2016.05.006] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2015] [Revised: 04/18/2016] [Accepted: 05/09/2016] [Indexed: 12/25/2022]
Abstract
Oral administration of the combination of L-threonate (threonate) and magnesium (Mg(2+)) in the form of L-Threonic acid Magnesium salt (L-TAMS) can enhance learning and memory in young rats and prevent memory decline in aging rats and in Alzheimer's disease model mice. Recent results from a human clinical trial demonstrate the efficacy of L-TAMS in restoring global cognitive abilities of older adults. Previously, we reported that neuronal intracellular Mg(2+) serves as a critical signaling molecule for controlling synapse density, a key factor that determines cognitive ability. The elevation of brain Mg(2+) by oral administration of L-TAMS in intact animals plays a significant role in mediating the therapeutic effects of L-TAMS. The current study sought to elucidate the unique role of threonate. We aimed to understand if threonate acts directly to elevate intraneuronal Mg(2+), and why Mg(2+) given without threonate is ineffective for enhancing learning and memory ability. We discovered that threonate is naturally present in cerebrospinal fluid (CSF) and oral treatment with L-TAMS elevated CSF threonate. In cultured hippocampal neurons, threonate treatment directly induced an increase in intracellular Mg(2+) concentration. Functionally, elevating threonate upregulated expression of NR2B-containing NMDAR, boosted mitochondrial membrane potential (ΔΨm), and increased functional synapse density in neuronal cultures. These effects are unique to threonate, as other common Mg(2+) anions failed to have the same results. Mechanistically, threonate's effects were specifically mediated through glucose transporters (GLUTs). We also evaluated the effects of threonate in human neural stem cell-derived neurons, and found it was equally effective at upregulating synapse density. The current study provides an explanation for why threonate is an essential component of L-TAMS and supports the use of L-TAMS to promote cognitive abilities in human.
Collapse
Affiliation(s)
- Qifeng Sun
- School of Medicine, Tsinghua University, Beijing, 100084, China
| | | | - Fei Mao
- Neurocentria, Inc., Fremont, CA 94538, USA
| | - Guosong Liu
- School of Medicine, Tsinghua University, Beijing, 100084, China; Neurocentria, Inc., Fremont, CA 94538, USA.
| |
Collapse
|
22
|
Abstract
UNLABELLED Hippocampal activity is fundamental for episodic memory formation and consolidation. During phases of rest and sleep, it exhibits sharp-wave/ripple (SPW/R) complexes, which are short episodes of increased activity with superimposed high-frequency oscillations. Simultaneously, spike sequences reflecting previous behavior, such as traversed trajectories in space, are replayed. Whereas these phenomena are thought to be crucial for the formation and consolidation of episodic memory, their neurophysiological mechanisms are not well understood. Here we present a unified model showing how experience may be stored and thereafter replayed in association with SPW/Rs. We propose that replay and SPW/Rs are tightly interconnected as they mutually generate and support each other. The underlying mechanism is based on the nonlinear dendritic computation attributable to dendritic sodium spikes that have been prominently found in the hippocampal regions CA1 and CA3, where SPW/Rs and replay are also generated. Besides assigning SPW/Rs a crucial role for replay and thus memory processing, the proposed mechanism also explains their characteristic features, such as the oscillation frequency and the overall wave form. The results shed a new light on the dynamical aspects of hippocampal circuit learning. SIGNIFICANCE STATEMENT During phases of rest and sleep, the hippocampus, the "memory center" of the brain, generates intermittent patterns of strongly increased overall activity with high-frequency oscillations, the so-called sharp-wave/ripples. We investigate their role in learning and memory processing. They occur together with replay of activity sequences reflecting previous behavior. Developing a unifying computational model, we propose that both phenomena are tightly linked, by mutually generating and supporting each other. The underlying mechanism depends on nonlinear amplification of synchronous inputs that has been prominently found in the hippocampus. Besides assigning sharp-wave/ripples a crucial role for replay generation and thus memory processing, the proposed mechanism also explains their characteristic features, such as the oscillation frequency and the overall wave form.
Collapse
|
23
|
Scalability of Asynchronous Networks Is Limited by One-to-One Mapping between Effective Connectivity and Correlations. PLoS Comput Biol 2015; 11:e1004490. [PMID: 26325661 PMCID: PMC4556689 DOI: 10.1371/journal.pcbi.1004490] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2014] [Accepted: 08/05/2015] [Indexed: 11/19/2022] Open
Abstract
Network models are routinely downscaled compared to nature in terms of numbers of nodes or edges because of a lack of computational resources, often without explicit mention of the limitations this entails. While reliable methods have long existed to adjust parameters such that the first-order statistics of network dynamics are conserved, here we show that limitations already arise if also second-order statistics are to be maintained. The temporal structure of pairwise averaged correlations in the activity of recurrent networks is determined by the effective population-level connectivity. We first show that in general the converse is also true and explicitly mention degenerate cases when this one-to-one relationship does not hold. The one-to-one correspondence between effective connectivity and the temporal structure of pairwise averaged correlations implies that network scalings should preserve the effective connectivity if pairwise averaged correlations are to be held constant. Changes in effective connectivity can even push a network from a linearly stable to an unstable, oscillatory regime and vice versa. On this basis, we derive conditions for the preservation of both mean population-averaged activities and pairwise averaged correlations under a change in numbers of neurons or synapses in the asynchronous regime typical of cortical networks. We find that mean activities and correlation structure can be maintained by an appropriate scaling of the synaptic weights, but only over a range of numbers of synapses that is limited by the variance of external inputs to the network. Our results therefore show that the reducibility of asynchronous networks is fundamentally limited.
Collapse
|
24
|
Bachiller S, Rybkina T, Porras-García E, Pérez-Villegas E, Tabares L, Armengol JA, Carrión AM, Ruiz R. The HERC1 E3 Ubiquitin Ligase is essential for normal development and for neurotransmission at the mouse neuromuscular junction. Cell Mol Life Sci 2015; 72:2961-71. [PMID: 25746226 PMCID: PMC11113414 DOI: 10.1007/s00018-015-1878-2] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2014] [Revised: 02/15/2015] [Accepted: 02/27/2015] [Indexed: 12/14/2022]
Abstract
The ubiquitin-proteasome system (UPS) plays a fundamental role in protein degradation in neurons, and there is strong evidence that it fulfills a key role in synaptic transmission. The aim of the present work was to study the implication of one component of the UPS, the HERC1 E3 Ubiquitin Ligase, in motor function and neuromuscular transmission. The tambaleante (tbl) mutant mouse carries a spontaneous mutation in HERC1 E3 Ubiquitin Ligase, provoking an ataxic phenotype that develops in the second month of life. Our results show that motor performance in mutant mice is altered at postnatal day 30, before the cerebellar neurodegeneration takes place. This defect is associated with by: (a) a reduction of the motor end-plate area, (b) less efficient neuromuscular activity in vivo, and (c) an impaired evoked neurotransmitter release. Together, these data suggest that the HERC1 E3 Ubiquitin Ligase is fundamental for normal muscle function and that it is essential for neurotransmitter release at the mouse neuromuscular junction.
Collapse
Affiliation(s)
- S. Bachiller
- Department of Physiology, Anatomy and Cellular Biology, University of Pablo de Olavide, Carretera de Utrera Km 1, 41013 Seville, Spain
| | - T. Rybkina
- Department of Physiology, Anatomy and Cellular Biology, University of Pablo de Olavide, Carretera de Utrera Km 1, 41013 Seville, Spain
| | - E. Porras-García
- Department of Physiology, Anatomy and Cellular Biology, University of Pablo de Olavide, Carretera de Utrera Km 1, 41013 Seville, Spain
| | - E. Pérez-Villegas
- Department of Physiology, Anatomy and Cellular Biology, University of Pablo de Olavide, Carretera de Utrera Km 1, 41013 Seville, Spain
| | - L. Tabares
- Department of Medical Physiology and Biophysics, University of Seville, Seville, Spain
| | - J. A. Armengol
- Department of Physiology, Anatomy and Cellular Biology, University of Pablo de Olavide, Carretera de Utrera Km 1, 41013 Seville, Spain
- School of Medicine, University of Cartagena de Indias, Cartagena, Colombia
| | - A. M. Carrión
- Department of Physiology, Anatomy and Cellular Biology, University of Pablo de Olavide, Carretera de Utrera Km 1, 41013 Seville, Spain
| | - R. Ruiz
- Department of Physiology, Anatomy and Cellular Biology, University of Pablo de Olavide, Carretera de Utrera Km 1, 41013 Seville, Spain
| |
Collapse
|
25
|
García-Pérez E, Mahfooz K, Covita J, Zandueta A, Wesseling JF. Levetiracetam accelerates the onset of supply rate depression in synaptic vesicle trafficking. Epilepsia 2015; 56:535-45. [PMID: 25684406 DOI: 10.1111/epi.12930] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/31/2014] [Indexed: 11/28/2022]
Abstract
OBJECTIVE To determine if levetiracetam (LEV) enhances the impact in excitatory presynaptic terminals of a rate-limiting mechanism in vesicle trafficking termed supply rate depression that emerges to limit synaptic transmission during heavy, epileptiform use. METHODS The effect of LEV was measured with electrophysiologic assays of monosynaptic connections in ex vivo hippocampal slices from wild-type and synapsin knockout mice, and in primary cell culture neurons from wild-type and synaptic vesicle glycoprotein 2a (SV2a) knockout mice. RESULTS LEV enhanced the impact of supply rate depression at Schaffer collateral synapses by shortening the time course for induction. The LEV effect was selective for supply rate depression because other presynaptic vesicle trafficking mechanisms were not affected. The half maximal effective concentration (EC50 ) was ~50 μm. The maximal effect was ~15% and occurred at 100 μm, which is a clinically relevant concentration. An experimental protocol is established for distinguishing atypical antiepileptic drugs (AEDs) that affect supply rate depression, such as LEV, from typical AEDs, such as carbamazepine, that affect upstream mechanisms. The LEV effect was abolished at synapses from knockout mice lacking SV2a and from synapses lacking synapsin 1 and 2. SIGNIFICANCE The findings are consistent with the new hypothesis that LEV acts to treat epilepsy by accelerating the induction of supply rate depression at excitatory synapses during incipient epileptic activity. The absence of the effect in the knockouts confirms that presynaptic function is the target. More specifically, the absence in SV2a knockouts is consistent with previous binding studies suggesting that SV2a is the target for LEV. The absence in synapsin knockouts indicates that the phenotypic target intersects with the biochemical pathway that is altered in synapsin knockouts. The results from synapsin knockouts additionally suggest that development of functional analogs with increased potency might be possible because induction of supply rate depression is faster in synapsin knockouts compared to wild-type synapses treated with LEV.
Collapse
|
26
|
TAKAHASHI T. Strength and precision of neurotransmission at mammalian presynaptic terminals. PROCEEDINGS OF THE JAPAN ACADEMY. SERIES B, PHYSICAL AND BIOLOGICAL SCIENCES 2015; 91:305-320. [PMID: 26194855 PMCID: PMC4631896 DOI: 10.2183/pjab.91.305] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/26/2015] [Accepted: 05/06/2015] [Indexed: 05/30/2023]
Abstract
Classically, the basic concept of chemical synaptic transmission was established at the frog neuromuscular junction, and direct intracellular recordings from presynaptic terminals at the squid giant presynaptic terminal have further clarified principles of neurotransmitter release. More recently, whole-cell patch-camp recordings from the calyx of Held in rodent brainstem slices have extended the classical concept to mammalian synapses providing new insights into the mechanisms underlying strength and precision of neurotransmission and developmental changes therein. This review summarizes findings from our laboratory and others on these subjects, mainly at the calyx of Held, with a particular focus on precise, high-fidelity, fast neurotransmission. The mechanisms by which presynaptic terminals acquire strong, precise neurotransmission during postnatal development are also discussed.
Collapse
Affiliation(s)
- Tomoyuki TAKAHASHI
- Cellular and Molecular Synaptic Function Unit, Okinawa Institute of Science and Technology Graduate University, Okinawa, Japan
| |
Collapse
|
27
|
Jahnke S, Memmesheimer RM, Timme M. Oscillation-induced signal transmission and gating in neural circuits. PLoS Comput Biol 2014; 10:e1003940. [PMID: 25503492 PMCID: PMC4263355 DOI: 10.1371/journal.pcbi.1003940] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2014] [Accepted: 09/26/2014] [Indexed: 11/19/2022] Open
Abstract
Reliable signal transmission constitutes a key requirement for neural circuit function. The propagation of synchronous pulse packets through recurrent circuits is hypothesized to be one robust form of signal transmission and has been extensively studied in computational and theoretical works. Yet, although external or internally generated oscillations are ubiquitous across neural systems, their influence on such signal propagation is unclear. Here we systematically investigate the impact of oscillations on propagating synchrony. We find that for standard, additive couplings and a net excitatory effect of oscillations, robust propagation of synchrony is enabled in less prominent feed-forward structures than in systems without oscillations. In the presence of non-additive coupling (as mediated by fast dendritic spikes), even balanced oscillatory inputs may enable robust propagation. Here, emerging resonances create complex locking patterns between oscillations and spike synchrony. Interestingly, these resonances make the circuits capable of selecting specific pathways for signal transmission. Oscillations may thus promote reliable transmission and, in co-action with dendritic nonlinearities, provide a mechanism for information processing by selectively gating and routing of signals. Our results are of particular interest for the interpretation of sharp wave/ripple complexes in the hippocampus, where previously learned spike patterns are replayed in conjunction with global high-frequency oscillations. We suggest that the oscillations may serve to stabilize the replay. Rhythmic activity in the brain is ubiquitous, its functions are debated. Here we show that it may contribute to the reliable transmission of information within brain areas. We find that its effect is particularly strong if we take nonlinear coupling into account. This experimentally found neuronal property implies that inputs which arrive nearly simultaneously can have a much stronger impact than expected from the sum of their individuals strengths. In such systems, rhythmic activity supports information transmission even if its positive and negative part exactly cancels all the time. Further, the information transmission can adapt to the oscillation frequency to optimally benefit from it. Finally, we show that rhythms with different frequencies may enable or disable communication channels, and are thus suitable for the steering of information flow.
Collapse
Affiliation(s)
- Sven Jahnke
- Network Dynamics, Max Planck Institute for Dynamics and Self-Organization (MPIDS), Göttingen, Germany
- Bernstein Center for Computational Neuroscience (BCCN), Göttingen, Germany
- Institute for Nonlinear Dynamics, Fakultät für Physik, Georg-August Universität Göttingen, Göttingen Germany
- * E-mail:
| | | | - Marc Timme
- Network Dynamics, Max Planck Institute for Dynamics and Self-Organization (MPIDS), Göttingen, Germany
- Bernstein Center for Computational Neuroscience (BCCN), Göttingen, Germany
- Institute for Nonlinear Dynamics, Fakultät für Physik, Georg-August Universität Göttingen, Göttingen Germany
| |
Collapse
|
28
|
Abstract
It is well established that the active properties of nerve and muscle cells are stabilized by homeostatic signaling systems. In organisms ranging from Drosophila to humans, neurons restore baseline function in the continued presence of destabilizing perturbations by rebalancing ion channel expression, modifying neurotransmitter receptor surface expression and trafficking, and modulating neurotransmitter release. This review focuses on the homeostatic modulation of presynaptic neurotransmitter release, termed presynaptic homeostasis. First, we highlight criteria that can be used to define a process as being under homeostatic control. Next, we review the remarkable conservation of presynaptic homeostasis at the Drosophila, mouse, and human neuromuscular junctions and emerging parallels at synaptic connections in the mammalian central nervous system. We then highlight recent progress identifying cellular and molecular mechanisms. We conclude by reviewing emerging parallels between the mechanisms of homeostatic signaling and genetic links to neurological disease.
Collapse
Affiliation(s)
- Graeme W Davis
- Department of Biochemistry and Biophysics, University of California, San Francisco, California 94158;
| | | |
Collapse
|
29
|
de Jong APH, Fioravante D. Translating neuronal activity at the synapse: presynaptic calcium sensors in short-term plasticity. Front Cell Neurosci 2014; 8:356. [PMID: 25400547 PMCID: PMC4212674 DOI: 10.3389/fncel.2014.00356] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2014] [Accepted: 10/09/2014] [Indexed: 01/03/2023] Open
Abstract
The complex manner in which patterns of presynaptic neural activity are translated into short-term plasticity (STP) suggests the existence of multiple presynaptic calcium (Ca(2+)) sensors, which regulate the amplitude and time-course of STP and are the focus of this review. We describe two canonical Ca(2+)-binding protein domains (C2 domains and EF-hands) and define criteria that need to be met for a protein to qualify as a Ca(2+) sensor mediating STP. With these criteria in mind, we discuss various forms of STP and identify established and putative Ca(2+) sensors. We find that despite the multitude of proposed sensors, only three are well established in STP: Munc13, protein kinase C (PKC) and synaptotagmin-7. For putative sensors, we pinpoint open questions and potential pitfalls. Finally, we discuss how the molecular properties and modes of action of Ca(2+) sensors can explain their differential involvement in STP and shape net synaptic output.
Collapse
Affiliation(s)
| | - Diasynou Fioravante
- Department of Neurobiology, Physiology and Behavior, Center for Neuroscience, University of California Davis Davis, CA, USA
| |
Collapse
|
30
|
Wang T, Hauswirth AG, Tong A, Dickman DK, Davis GW. Endostatin is a trans-synaptic signal for homeostatic synaptic plasticity. Neuron 2014; 83:616-29. [PMID: 25066085 DOI: 10.1016/j.neuron.2014.07.003] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/16/2014] [Indexed: 10/25/2022]
Abstract
At synapses in organisms ranging from fly to human, a decrease in postsynaptic neurotransmitter receptor function elicits a homeostatic increase in presynaptic release that restores baseline synaptic efficacy. This process, termed presynaptic homeostasis, requires a retrograde, trans-synaptic signal of unknown identity. In a forward genetic screen for homeostatic plasticity genes, we identified multiplexin. Multiplexin is the Drosophila homolog of Collagen XV/XVIII, a matrix protein that can be proteolytically cleaved to release Endostatin, an antiangiogenesis signaling factor. Here we demonstrate that Multiplexin is required for normal calcium channel abundance, presynaptic calcium influx, and neurotransmitter release. Remarkably, Endostatin has a specific activity, independent of baseline synapse development, that is required for the homeostatic modulation of presynaptic calcium influx and neurotransmitter release. Our data support a model in which proteolytic release of Endostatin signals trans-synaptically, acting in concert with the presynaptic CaV2.1 calcium channel, to promote presynaptic homeostasis.
Collapse
Affiliation(s)
- Tingting Wang
- Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Anna G Hauswirth
- Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Amy Tong
- Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Dion K Dickman
- Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Graeme W Davis
- Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA 94158, USA.
| |
Collapse
|
31
|
Schikorski T. Readily releasable vesicles recycle at the active zone of hippocampal synapses. Proc Natl Acad Sci U S A 2014; 111:5415-20. [PMID: 24706824 PMCID: PMC3986142 DOI: 10.1073/pnas.1321541111] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
During the synaptic vesicle cycle, synaptic vesicles fuse with the plasma membrane and recycle for repeated exo/endocytic events. By using activity-dependent N-(3-triethylammoniumpropyl)-4-(4-(dibutylamino) styryl) pyridinium dibromide dye uptake combined with fast (<1 s) microwave-assisted fixation followed by photoconversion and ultrastructural 3D analysis, we tracked endocytic vesicles over time, "frame by frame." The first retrieved synaptic vesicles appeared 4 s after stimulation, and these endocytic vesicles were located just above the active zone. Second, the retrieved vesicles did not show any sign of a protein coat, and coated pits were not detected. Between 10 and 30 s, large labeled vesicles appeared that had up to 5 times the size of an individual synaptic vesicle. Starting at around 20 s, these large labeled vesicles decreased in number in favor of labeled synaptic vesicles, and after 30 s, labeled vesicles redocked at the active zone. The data suggest that readily releasable vesicles are retrieved as noncoated vesicles at the active zone.
Collapse
Affiliation(s)
- Thomas Schikorski
- Department of Neuroscience, Universidad Central del Caribe, Bayamon, PR 00956
| |
Collapse
|
32
|
Alyea RA, Gollapudi BB, Rasoulpour RJ. Are we ready to consider transgenerational epigenetic effects in human health risk assessment? ENVIRONMENTAL AND MOLECULAR MUTAGENESIS 2014; 55:292-298. [PMID: 24259352 DOI: 10.1002/em.21831] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/31/2013] [Revised: 10/25/2013] [Accepted: 10/25/2013] [Indexed: 06/02/2023]
Abstract
Recently, there has been a growing concern that chemically or nutritionally mediated epigenetic changes might lead to adverse health outcomes. The natural question is whether the existing chemical safety assessment paradigm is or is not protective of epigenetic-mediated effects, and if there is a need to incorporate new endpoints to specifically address epigenetics. Of particular interest are transgenerational epigenetic effects, which can be passed on through multiple generations. To investigate these questions, a comparison was performed between OECD guideline rat toxicology studies versus several rat transgenerational epigenetic studies. This analysis focused on vinclozolin owing to the availability of a comprehensive suite of dose-response data (NOAEL, reference dose, and human exposure estimates) for both conventional and epigenetic endpoints. This analysis revealed that vinclozolin transgenerational effects were demonstrated at a dose level (100 mg/kg/day) that was: (1) ∼40-fold higher than the overall lowest-observed-adverse-effect level (LOAEL) from rat guideline studies, (2) ∼80-fold higher than the lowest NOAEL from rat guideline studies, (3) ∼80,000-fold higher than the reference dose for the molecule, and (4) ∼1.2-million fold above human exposure estimates. Through this analysis, we conclude that additional research across a spectrum of doses is necessary to elucidate the interplay between epigenetics and apical endpoints before considering epigenetics in human health risk assessment. Therefore, we recommend focusing future research toward (1) examining for potential causal relationships between epigenetic alterations and adverse apical endpoints, and (2) understanding the dose-response relationship of these causal epigenetic alterations when compared with those of the apical endpoints.
Collapse
Affiliation(s)
- Rebecca A Alyea
- Toxicology and Environmental Research and Consulting, The Dow Chemical Company, Midland, Michigan
| | | | | |
Collapse
|
33
|
Abstract
The brain is astonishing in its complexity and capacity for change. This has fascinated scientists for more than a century, filling the pages of this journal for the past 25 years. But a paradigm shift is underway. It seems likely that the plasticity that drives our ability to learn and remember can only be meaningful in the context of otherwise stable, reproducible, and predictable baseline neural function. Without the existence of potent mechanisms that stabilize neural function, our capacity to learn and remember would be lost in the chaos of daily experiential change. This underscores two great mysteries in neuroscience. How are the functional properties of individual neurons and neural circuits stably maintained throughout life? And, in the face of potent stabilizing mechanisms, how can neural circuitry be modified during neural development, learning, and memory? Answers are emerging in the rapidly developing field of homeostatic plasticity.
Collapse
Affiliation(s)
- Graeme W Davis
- Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA 94158, USA.
| |
Collapse
|
34
|
Low-frequency neuromuscular depression is a consequence of a reduction in nerve terminal Ca2+ currents at mammalian motor nerve endings. Anesthesiology 2013; 119:326-34. [PMID: 23535502 DOI: 10.1097/aln.0b013e31829083d8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND The decline in voluntary muscle contraction during low-frequency nerve stimulation is used clinically to assess the type and degree of neuromuscular block. The mechanism underlying this depression is unknown. METHODS Simultaneous electrophysiological measurements of neurotransmitter release and prejunctional Ca currents were made at mouse neuromuscular junctions to evaluate the hypothesis that decreases in nerve terminal Ca currents are responsible for low-frequency depression. RESULTS Under conditions generally used to measure Ca currents at the neuromuscular junction, increasing the frequency of nerve stimulation briefly from 0.017 to 0.1-1 Hz caused a simultaneous reduction in the release of the neurotransmitter acetylcholine to 52.2 ± 4.4% of control and the Ca current peak to 75.4 ± 2.0% of control (P < 0.001, n = 5 experiments for both measurements, mean ± SEM for all data). In conditions used for train-of-four monitoring (4 stimuli, 2 Hz), neurotransmitter release declined to 42.0 ± 1.0% of control and the Ca current peak declined to 75.8 ± 3.3% of control between the first and fourth stimulus (P < 0.001, n = 7 experiments for both measurements). Depression in acetylcholine release during train-of-four protocols also occurred in the absence of neuromuscular-blocking drugs. DISCUSSION The results demonstrate that neuromuscular depression during train-of-four monitoring is due to a decline in nerve terminal Ca currents, hence reducing the release of acetylcholine. As similar processes may come into play at higher stimulation frequencies, agents that antagonize the decline in Ca currents could be used to treat conditions in which neuromuscular depression can be debilitating.
Collapse
|
35
|
Lee KJ, Queenan BN, Rozeboom AM, Bellmore R, Lim ST, Vicini S, Pak DTS. Mossy fiber-CA3 synapses mediate homeostatic plasticity in mature hippocampal neurons. Neuron 2013; 77:99-114. [PMID: 23312519 DOI: 10.1016/j.neuron.2012.10.033] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/31/2012] [Indexed: 11/19/2022]
Abstract
Network activity homeostatically alters synaptic efficacy to constrain neuronal output. However, it is unclear how such compensatory adaptations coexist with synaptic information storage, especially in established networks. Here, we report that in mature hippocampal neurons in vitro, network activity preferentially regulated excitatory synapses within the proximal dendrites of CA3 neurons. These homeostatic synapses exhibited morphological, functional, and molecular signatures of the specialized contacts between mossy fibers of dentate granule cells and thorny excrescences (TEs) of CA3 pyramidal neurons. In vivo TEs were also selectively and bidirectionally altered by chronic activity changes. TE formation required presynaptic synaptoporin and was suppressed by the activity-inducible kinase, Plk2. These results implicate the mossy fiber-TE synapse as an independently tunable gain control locus that permits efficacious homeostatic adjustment of mossy fiber-CA3 synapses, while preserving synaptic weights that may encode information elsewhere within the mature hippocampal circuit.
Collapse
Affiliation(s)
- Kea Joo Lee
- Department of Pharmacology & Physiology, Georgetown University Medical Center, Washington, DC 20057, USA
| | | | | | | | | | | | | |
Collapse
|
36
|
Abstract
After releasing neurotransmitter, synaptic vesicles are retrieved by endocytosis and recycled via fast and slow pathways to be reused for synaptic transmission. To maintain the synaptic efficacy, vesicles must be refilled with neurotransmitter during recycling. However, the refilling speed estimated in isolated or reconstructed vesicles is, thus far, too slow to fill up vesicles within the period of recycling. Here, we re-examined the vesicle refilling rate directly at central glutamatergic synapses in slices, using simultaneous presynaptic and postsynaptic whole-cell recording combined with caged glutamate photolysis. After washing out vesicular glutamate, refilling of vesicles with uncaged glutamate caused a recovery of EPSCs with a time constant of 15 s that varied depending upon temperature, age, and cytosolic Cl(-) concentrations. This time constant is faster than that of the slow recycling pathway (∼30 s) after clathrin-mediated endocytosis but is much too slow to fill up vesicles replenished through fast recycling pathways (<1 s).
Collapse
|
37
|
Schneider S, Marxfeld H, Gröters S, Buesen R, van Ravenzwaay B. Vinclozolin--no transgenerational inheritance of anti-androgenic effects after maternal exposure during organogenesis via the intraperitoneal route. Reprod Toxicol 2013; 37:6-14. [PMID: 23313085 DOI: 10.1016/j.reprotox.2012.12.003] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2011] [Revised: 12/19/2012] [Accepted: 12/20/2012] [Indexed: 10/27/2022]
Abstract
The goal of this study was to examine the potential transgenerational inheritance of anti-androgenic effects induced by Vinclozolin administered intraperitoneally to pregnant Wistar rats (Crl:WI[Han]). Dams were dosed with Vinclozolin at 0, 4 or 100mg/kg bw/d on gestation days 6-15. Male offspring of F1-F3 generations were bred with untreated females to yield F2-F4 offspring. No evident anti-androgenic effects were observed at 4mg/kg bw/d, but a case of hypospadias as well as delayed sexual maturation in F1 male offspring was observed as a sign of anti-androgenicity at 100mg/kg bw/d. However, F1-F3 males developed normally to sexual maturity and were able to mate and to generate healthy progeny. Sperm count, morphology and motility were not affected in F1-F4 generation male offspring. In conclusion, transgenerational inheritance of Vinclozolin's anti-androgenic effects was not evident in outbred Wistar rats.
Collapse
Affiliation(s)
- Steffen Schneider
- Experimental Toxicology and Ecotoxicology, BASF SE, 67056 Ludwigshafen, Germany
| | | | | | | | | |
Collapse
|
38
|
Hong T, Lazarenko RM, Colvin DC, Flores RL, Zhang Q, Xu YQ. Effect of Competitive Surface Functionalization on Dual-Modality Fluorescence and Magnetic Resonance Imaging of Single-Walled Carbon Nanotubes. THE JOURNAL OF PHYSICAL CHEMISTRY. C, NANOMATERIALS AND INTERFACES 2012; 116:16319-16324. [PMID: 26644813 PMCID: PMC4670557 DOI: 10.1021/jp305372z] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/24/2023]
Abstract
It is well-known that ionic surfactant coated single-walled carbon nanotubes (SWNTs) possess higher near-infrared fluorescence (NIRF) quantum yield than nonionic polymer functionalized SWNTs. However, the influence of surface functionalization on the magnetic properties of SWNTs for T2-weighted magnetic resonance imaging (MRI) has not been reported. Here, we demonstrate that SWNTs functionalized by nonionic polymers display superior T2 relaxivity for MRI as compared to those coated by ionic surfactants. This difference may indicate that micelle structures formed by ionic surfactants are sufficiently tight to partially exclude water protons from the iron catalysts attached to the ends of SWNTs. On the basis of the different effects of the two types of suspension agents on NIRF and MRI of functionalized SWNTs, we further explore the competitive surface functionalization between ionic surfactants and nonionic polymers by stepwise replacing ionic surfactant molecules in a nanotube suspension with nonionic polymers. The superior NIRF of ionic surfactant coated SWNTs gradually quenches whereas no improvement on T2 relaxivity is observed during this replacement process. This result may indicate that nonionic polymers wrap around the outside of micelle structures to form small nanotube bundles rather than replacing ionic surfactants in the micelle structures to directly interact with the SWNT surface. Finally, we demonstrate the feasibility of dual-modality NIRF and MRI of nonionic polymer functionalized SWNTs in brain cells.
Collapse
Affiliation(s)
- Tu Hong
- Department of Electrical Engineering and Computer Science, Vanderbilt University, Nashville, Tennessee 37235, United States
| | - Roman M. Lazarenko
- Department of Pharmacology, Vanderbilt University Medical Center, Nashville, Tennessee 37232, United States
| | - Daniel C. Colvin
- Department of Radiology and Radiological Sciences, Vanderbilt University Medical Center, Nashville, Tennessee 37232, United States
| | - Roel L. Flores
- Interdisciplinary Program in Materials Science, Vanderbilt University, Nashville, Tennessee 37235, United States
| | - Qi Zhang
- Department of Pharmacology, Vanderbilt University Medical Center, Nashville, Tennessee 37232, United States
| | - Ya-Qiong Xu
- Department of Electrical Engineering and Computer Science, Vanderbilt University, Nashville, Tennessee 37235, United States
- Interdisciplinary Program in Materials Science, Vanderbilt University, Nashville, Tennessee 37235, United States
- Department of Physics and Astronomy, Vanderbilt University, Nashville, Tennessee 37235, United States
| |
Collapse
|
39
|
Abstract
Synaptic vesicles release neurotransmitter at chemical synapses, thus initiating the flow of information in neural networks. To achieve this, vesicles undergo a dynamic cycle of fusion and retrieval to maintain the structural and functional integrity of the presynaptic terminals in which they reside. Moreover, compelling evidence indicates these vesicles differ in their availability for release and mobilization in response to stimuli, prompting classification into at least three different functional pools. Ongoing studies of the molecular and cellular bases for this heterogeneity attempt to link structure to physiology and clarify how regulation of vesicle pools influences synaptic strength and presynaptic plasticity. We discuss prevailing perspectives on vesicle pools, the role they play in shaping synaptic transmission, and the open questions that challenge current understanding.
Collapse
Affiliation(s)
- AbdulRasheed A Alabi
- Department of Molecular and Cellular Physiology, Stanford Institute for Neuro-Innovation and Translational Neurosciences, Stanford Medical School, Stanford, California 94305, USA
| | | |
Collapse
|
40
|
Feng L, Sametsky EA, Gusev AG, Uteshev VV. Responsiveness to nicotine of neurons of the caudal nucleus of the solitary tract correlates with the neuronal projection target. J Neurophysiol 2012; 108:1884-94. [PMID: 22815399 DOI: 10.1152/jn.00296.2012] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The caudal nucleus of the solitary tract (NTS) is the key integrating center of visceral sensory-motor signaling supporting autonomic homeostasis. Two key projections of this nucleus are the parabrachial nucleus (PbN) and the dorsal motor nucleus of the vagus (DMV). The PbN integrates and relays viscerosensory information primarily to the forebrain, supporting behavioral, emotional, and endocrine responses to visceral events, while the DMV contains parasympathetic preganglionic cholinergic motoneurons that support primarily gastrointestinal reflexes. Subsets of caudal NTS neurons express presynaptic and somatodendritic nicotinic acetylcholine receptors (nAChRs). However, the anatomical identification of nicotine-responsive caudal NTS neurons has not been determined. This study used in vivo and ex vivo fluorescent tracing and slice patch-clamp electrophysiological recordings from anatomically identified caudal NTS neurons to test the hypothesis that the responsiveness of these cells to nicotine correlates with the target of their axonal projections. The results demonstrate that the majority of glutamatergic terminals that synapse on PbN-projecting caudal NTS neurons are unaffected by nicotine. Moreover, only a fraction of these cells express somatodendritic nAChRs. In contrast, the majority of DMV-projecting caudal NTS neurons exhibit robust presynaptic and somatodendritic responsiveness to nicotine. However, PbN-projecting neurons also exhibit significantly lower background frequencies of glutamatergic miniature postsynaptic currents than DMV-projecting neurons. Therefore, presynaptic unresponsiveness to nicotine may result from deficient glutamatergic innervation of PbN-projecting neurons. Nevertheless, the caudal NTS contains function-specific subsets of cells with target-specific responsiveness to nicotine. These results may support development of therapeutic strategies for selective targeting of specific autonomic pathways and impaired autonomic homeostasis.
Collapse
Affiliation(s)
- Lin Feng
- Department of Pharmacology, Southern Illinois University School of Medicine, Springfield, Illinois, USA
| | | | | | | |
Collapse
|
41
|
Nauen DW, Bi GQ. Measuring action potential-evoked transmission at individual synaptic contacts. J Neural Eng 2012; 9:036014. [PMID: 22626987 DOI: 10.1088/1741-2560/9/3/036014] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
In the neuronal culture experimental system, the total synaptic connection between two neurons can consist of large numbers of synaptic sites, each behaving probabilistically. Studies of synaptic function with paired recordings typically consider the summed response across all of these sites and from this infer the average response. Understanding of synaptic transmission and plasticity could be improved by examination of activity at as few synaptic sites as possible. To this end, we develop a system for recording responses from individual contacts. It relies on a precisely regulated pneumatic/hydrostatic pressure system to create a microenvironment within which individual synapses are active, and an acoustic signature method to monitor the stability of this microenvironment noninvasively. With this method we are able to record action potential-evoked postsynaptic currents consistent with individual quanta. The approach does not distort synaptic current waveforms and permits stable recording for several hours. The method is applied to address mechanisms of short-term plasticity, the variability of latency at individual synaptic sites and, in a preliminary experiment, the independence of nearby synapses on the same axon.
Collapse
Affiliation(s)
- David W Nauen
- Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA.
| | | |
Collapse
|
42
|
Wherefore art thou, homeo(stasis)? Functional diversity in homeostatic synaptic plasticity. Neural Plast 2012; 2012:718203. [PMID: 22685679 PMCID: PMC3362963 DOI: 10.1155/2012/718203] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2011] [Revised: 01/06/2012] [Accepted: 01/09/2012] [Indexed: 11/18/2022] Open
Abstract
Homeostatic plasticity has emerged as a fundamental regulatory principle that strives to maintain neuronal activity within optimal ranges by altering diverse aspects of neuronal function. Adaptation to network activity is often viewed as an essential negative feedback restraint that prevents runaway excitation or inhibition. However, the precise importance of these homeostatic functions is often theoretical rather than empirically derived. Moreover, a remarkable multiplicity of homeostatic adaptations has been observed. To clarify these issues, it may prove useful to ask: why do homeostatic mechanisms exist, what advantages do these adaptive responses confer on a given cell population, and why are there so many seemingly divergent effects? Here, we approach these questions by applying the principles of control theory to homeostatic synaptic plasticity of mammalian neurons and suggest that the varied responses observed may represent distinct functional classes of control mechanisms directed toward disparate physiological goals.
Collapse
|
43
|
The role of metaplasticity mechanisms in regulating memory destabilization and reconsolidation. Neurosci Biobehav Rev 2012; 36:1667-707. [PMID: 22484475 DOI: 10.1016/j.neubiorev.2012.03.008] [Citation(s) in RCA: 144] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2011] [Revised: 03/09/2012] [Accepted: 03/21/2012] [Indexed: 12/13/2022]
Abstract
Memory allows organisms to predict future events based on prior experiences. This requires encoded information to persist once important predictors are extracted, while also being modifiable in response to changes within the environment. Memory reconsolidation may allow stored information to be modified in response to related experience. However, there are many boundary conditions beyond which reconsolidation may not occur. One interpretation of these findings is that the event triggering memory retrieval must contain new information about a familiar stimulus in order to induce reconsolidation. Presently, the mechanisms that affect the likelihood of reconsolidation occurring under these conditions are not well understood. Here we speculate on a number of systems that may play a role in protecting memory from being destabilized during retrieval. We conclude that few memories may enter a state in which they cannot be modified. Rather, metaplasticity mechanisms may serve to alter the specific reactivation cues necessary to destabilize a memory. This might imply that destabilization mechanisms can differ depending on learning conditions.
Collapse
|
44
|
Kolle SN, Melching-Kollmuss S, Krennrich G, Landsiedel R, van Ravenzwaay B. Assessment of combinations of antiandrogenic compounds vinclozolin and flutamide in a yeast based reporter assay. Regul Toxicol Pharmacol 2011; 60:373-80. [DOI: 10.1016/j.yrtph.2011.05.005] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2011] [Revised: 05/06/2011] [Accepted: 05/11/2011] [Indexed: 11/27/2022]
|
45
|
Ribrault C, Sekimoto K, Triller A. From the stochasticity of molecular processes to the variability of synaptic transmission. Nat Rev Neurosci 2011; 12:375-87. [PMID: 21685931 DOI: 10.1038/nrn3025] [Citation(s) in RCA: 122] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The variability of the postsynaptic response following a single action potential arises from two sources: the neurotransmitter release is probabilistic, and the postsynaptic response to neurotransmitter release has variable timing and amplitude. At individual synapses, the number of molecules of a given type that are involved in these processes is small enough that the stochastic (random) properties of molecular events cannot be neglected. How the stochasticity of molecular processes contributes to the variability of synaptic transmission, its sensitivity and its robustness to molecular fluctuations has important implications for our understanding of the mechanistic basis of synaptic transmission and of synaptic plasticity.
Collapse
Affiliation(s)
- Claire Ribrault
- Laboratoire Matières et Systèmes Complexes, CNRS-UMR7057, Université Paris 7, F-75205 Paris cedex 13, France
| | | | | |
Collapse
|
46
|
Schneider S, Kaufmann W, Strauss V, van Ravenzwaay B. Vinclozolin: A feasibility and sensitivity study of the ILSI-HESI F1-extended one-generation rat reproduction protocol. Regul Toxicol Pharmacol 2011; 59:91-100. [DOI: 10.1016/j.yrtph.2010.09.010] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2010] [Revised: 09/20/2010] [Accepted: 09/22/2010] [Indexed: 12/31/2022]
|
47
|
Wang ZW. Origin of quantal size variation and high-frequency miniature postsynaptic currents at the Caenorhabditis elegans neuromuscular junction. J Neurosci Res 2010; 88:3425-32. [PMID: 20722072 PMCID: PMC3058485 DOI: 10.1002/jnr.22468] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2010] [Revised: 04/26/2010] [Accepted: 06/20/2010] [Indexed: 11/08/2022]
Abstract
The neuromuscular junction (NMJ) of Caenorhabditis elegans has proved to be a very useful model synapse for investigating molecular mechanisms of synaptic transmission. Intriguingly, miniature postsynaptic currents (minis) at this synapse occur at an unusually high frequency (50-90 Hz in wild-type worms) and show large variation in quantal size (from <10 pA to >200 pA). It is important to understand the cellular and molecular bases for these properties of minis in order to interpret electrophysiological data from this synapse properly. Existing data suggest that several factors may contribute to the high frequency and quantal size variation, including 1) the establishment of multiple NMJs with each body-wall muscle cell, 2) diversity of postsynaptic receptors (two acetylcholine receptors and one GABA receptor), 3) association of one presynaptic site with several body-wall muscle cells, 4) effects of Ca(2+) at the presynaptic site, and 5) a possibly elevated (less negative) resting membrane potential in motoneurons. Neither the frequency nor the quantal size of minis is affected by electrical coupling of body-wall muscle cells. Furthermore, quantal size variation is not due to synchronized multivesicular release. Analyses of the C. elegans NMJ may lead to a better understanding of the mechanisms controlling the frequency and quantal size of minis of other synapses as well.
Collapse
Affiliation(s)
- Zhao-Wen Wang
- Department of Neuroscience, University of Connecticut Health Center, Farmington, CT 06030-3401, USA.
| |
Collapse
|
48
|
Postsynaptic GluA1 enables acute retrograde enhancement of presynaptic function to coordinate adaptation to synaptic inactivity. Proc Natl Acad Sci U S A 2010; 107:21806-11. [PMID: 21098665 DOI: 10.1073/pnas.1016399107] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Prolonged blockade of AMPA-type glutamate receptors in hippocampal neuron cultures leads to homeostatic enhancements of pre- and postsynaptic function that appear correlated at individual synapses, suggesting some form of transsynaptic coordination. The respective modifications are important for overall synaptic strength but their interrelationship, dynamics, and molecular underpinnings are unclear. Here we demonstrate that adaptation begins postsynaptically but is ultimately communicated to presynaptic terminals and expressed as an accelerated turnover of synaptic vesicles. Critical postsynaptic modifications occur over hours, but enable retrograde communication within minutes once AMPA receptor (AMPAR) blockade is removed, causing elevation of both spontaneous and evoked vesicle fusion. The retrograde signaling does not require spiking activity and can be interrupted by NBQX, philanthotoxin, postsynaptic BAPTA, or external sequestration of BDNF, consistent with the acute release of retrograde messenger, triggered by postsynaptic Ca(2+) elevation via Ca(2+)-permeable AMPARs.
Collapse
|
49
|
Miyawaki H, Hirano T. Different correlations among physiological and morphological properties at single glutamatergic synapses in the rat hippocampus and the cerebellum. Synapse 2010; 65:412-23. [PMID: 20812293 DOI: 10.1002/syn.20860] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2010] [Accepted: 08/15/2010] [Indexed: 11/10/2022]
Abstract
Synapses in the mammalian central nervous system show substantial diversity in their physiological and morphological properties. However, the correlations among them have remained elusive. Here, we tried to clarify the correlations by establishing a method to record excitatory postsynaptic currents (EPSCs) at individual synapses and also to observe the morphology at the same time. A pair of pre- and postsynaptic neurons were labeled with different fluorescent dyes, and a presynaptic varicosity was selectively stimulated with a θ-tube glass electrode under conditions in which action potential generation was suppressed. Two representative types of excitatory glutamatergic synapses, one on hippocampal pyramidal neurons and the other on cerebellar Purkinje neurons, were studied. The correlations between the properties of quantal EPSCs (qEPSCs) and those of synaptic morphology were analyzed in rat primary culture preparations. The amplitude and the decay time of qEPSC were correlated with the size of the postsynaptic spine only at hippocampal synapses. In contrast, the size of the presynaptic varicosity was correlated with the size of the postsynaptic spine and the quantal content of evoked EPSCs only at granule neuron-Purkinje neuron synapses in the cerebellum. These results suggest that the interaction between pre- and postsynaptic structures and the coupling of postsynaptic responsiveness and the spine morphology differ between cerebellar and hippocampal glutamatergic synapses.
Collapse
Affiliation(s)
- Hiroyuki Miyawaki
- Department of Biophysics, Graduate School of Science, Kyoto University, Kitashirakawa-Oiwake-Cho, Sakyo-Ku, Kyoto, Japan
| | | |
Collapse
|
50
|
Nauen DW. Methods of measuring activity at individual synapses: a review of techniques and the findings they have made possible. J Neurosci Methods 2010; 194:195-205. [PMID: 20888362 DOI: 10.1016/j.jneumeth.2010.09.011] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2010] [Revised: 09/20/2010] [Accepted: 09/21/2010] [Indexed: 10/19/2022]
Abstract
Neurons in the brain are often linked by single synaptic contacts (Gulyás et al., 1993) and the probabilistic character of synaptic activity makes it desirable to increase the resolution of physiological experiments by observing the function of the smallest possible number of synaptic terminals, ideally, one. Because they are critically important and technically difficult to resolve, several of the core questions investigated in singe-site experiments have been under study for decades (Auger and Marty, 2000). Many approaches have been taken toward the goal of measuring activity at few synapses, and consideration of the capabilities and limitations of each of these methods permits a review of the contributions each has made possible to present understanding of synaptic function. A number of methodological advances in recent years have increased resolving power. New techniques often build on previous developments and many effective approaches combine components of existing specialized methods with new technology. One theme that emerges is that synaptic properties vary among regions, reducing the utility of general questions such as whether synaptic glutamate saturates receptors or how rapidly synaptic vesicle pools are depleted. For several core questions, multiple studies using different methods have reached similar conclusions, suggesting that consensus may be emerging for some anatomic synapses.
Collapse
Affiliation(s)
- David W Nauen
- Department of Neurobiology, University of Pittsburgh School of Medicine, W1401 BST, 200 Lothrop Street, Pittsburgh, PA 15261, United States.
| |
Collapse
|