1
|
Dossi E, Zonca L, Pivonkova H, Milior G, Moulard J, Vargova L, Chever O, Holcman D, Rouach N. Astroglial gap junctions strengthen hippocampal network activity by sustaining afterhyperpolarization via KCNQ channels. Cell Rep 2024; 43:114158. [PMID: 38722742 DOI: 10.1016/j.celrep.2024.114158] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Revised: 03/01/2024] [Accepted: 04/11/2024] [Indexed: 06/01/2024] Open
Abstract
Throughout the brain, astrocytes form networks mediated by gap junction channels that promote the activity of neuronal ensembles. Although their inputs on neuronal information processing are well established, how molecular gap junction channels shape neuronal network patterns remains unclear. Here, using astroglial connexin-deficient mice, in which astrocytes are disconnected and neuronal bursting patterns are abnormal, we show that astrocyte networks strengthen bursting activity via dynamic regulation of extracellular potassium levels, independently of glutamate homeostasis or metabolic support. Using a facilitation-depression model, we identify neuronal afterhyperpolarization as the key parameter underlying bursting pattern regulation by extracellular potassium in mice with disconnected astrocytes. We confirm this prediction experimentally and reveal that astroglial network control of extracellular potassium sustains neuronal afterhyperpolarization via KCNQ voltage-gated K+ channels. Altogether, these data delineate how astroglial gap junctions mechanistically strengthen neuronal population bursts and point to approaches for controlling aberrant activity in neurological diseases.
Collapse
Affiliation(s)
- Elena Dossi
- Neuroglial Interactions in Cerebral Physiology and Pathologies, Center for Interdisciplinary Research in Biology, Collège de France, CNRS, INSERM, Labex Memolife, Université PSL, Paris, France
| | - Lou Zonca
- Group of Data Modeling and Computational Biology, Institute of Biology, Ecole Normale Superieure, CNRS, INSERM, Université PSL, Paris, France; ED386, Ecole Doctorale de Sciences Mathématiques Paris Centre, Paris, France
| | - Helena Pivonkova
- Neuroglial Interactions in Cerebral Physiology and Pathologies, Center for Interdisciplinary Research in Biology, Collège de France, CNRS, INSERM, Labex Memolife, Université PSL, Paris, France; Department of Cellular Neurophysiology, Institute of Experimental Medicine, Czech Academy of Sciences, Prague, Czech Republic; 2(nd) Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Giampaolo Milior
- Neuroglial Interactions in Cerebral Physiology and Pathologies, Center for Interdisciplinary Research in Biology, Collège de France, CNRS, INSERM, Labex Memolife, Université PSL, Paris, France
| | - Julien Moulard
- Neuroglial Interactions in Cerebral Physiology and Pathologies, Center for Interdisciplinary Research in Biology, Collège de France, CNRS, INSERM, Labex Memolife, Université PSL, Paris, France
| | - Lydia Vargova
- Department of Cellular Neurophysiology, Institute of Experimental Medicine, Czech Academy of Sciences, Prague, Czech Republic; 2(nd) Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Oana Chever
- Neuroglial Interactions in Cerebral Physiology and Pathologies, Center for Interdisciplinary Research in Biology, Collège de France, CNRS, INSERM, Labex Memolife, Université PSL, Paris, France
| | - David Holcman
- Group of Data Modeling and Computational Biology, Institute of Biology, Ecole Normale Superieure, CNRS, INSERM, Université PSL, Paris, France.
| | - Nathalie Rouach
- Neuroglial Interactions in Cerebral Physiology and Pathologies, Center for Interdisciplinary Research in Biology, Collège de France, CNRS, INSERM, Labex Memolife, Université PSL, Paris, France.
| |
Collapse
|
2
|
Yamamoto H, Spitzner FP, Takemuro T, Buendía V, Murota H, Morante C, Konno T, Sato S, Hirano-Iwata A, Levina A, Priesemann V, Muñoz MA, Zierenberg J, Soriano J. Modular architecture facilitates noise-driven control of synchrony in neuronal networks. SCIENCE ADVANCES 2023; 9:eade1755. [PMID: 37624893 PMCID: PMC10456864 DOI: 10.1126/sciadv.ade1755] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Accepted: 07/21/2023] [Indexed: 08/27/2023]
Abstract
High-level information processing in the mammalian cortex requires both segregated processing in specialized circuits and integration across multiple circuits. One possible way to implement these seemingly opposing demands is by flexibly switching between states with different levels of synchrony. However, the mechanisms behind the control of complex synchronization patterns in neuronal networks remain elusive. Here, we use precision neuroengineering to manipulate and stimulate networks of cortical neurons in vitro, in combination with an in silico model of spiking neurons and a mesoscopic model of stochastically coupled modules to show that (i) a modular architecture enhances the sensitivity of the network to noise delivered as external asynchronous stimulation and that (ii) the persistent depletion of synaptic resources in stimulated neurons is the underlying mechanism for this effect. Together, our results demonstrate that the inherent dynamical state in structured networks of excitable units is determined by both its modular architecture and the properties of the external inputs.
Collapse
Affiliation(s)
- Hideaki Yamamoto
- Research Institute of Electrical Communication (RIEC), Tohoku University, Sendai, Japan
- Graduate School of Engineering, Tohoku University, Sendai, Japan
| | - F. Paul Spitzner
- Max Planck Institute for Dynamics and Self-Organization, Göttingen, Germany
| | - Taiki Takemuro
- Research Institute of Electrical Communication (RIEC), Tohoku University, Sendai, Japan
- Graduate School of Biomedical Engineering, Tohoku University, Sendai, Japan
| | - Victor Buendía
- Max Planck Institute for Biological Cybernetics, Tübingen, Germany
- Department of Computer Science, University of Tübingen, Tübingen, Germany
- Departamento de Electromagnetismo y Física de la Materia, Universidad de Granada, Granada, Spain
| | - Hakuba Murota
- Research Institute of Electrical Communication (RIEC), Tohoku University, Sendai, Japan
- Graduate School of Engineering, Tohoku University, Sendai, Japan
| | - Carla Morante
- Departament de Física de la Matèria Condensada, Universitat de Barcelona, Barcelona, Spain
- Universitat de Barcelona Institute of Complex Systems (UBICS), Barcelona, Spain
| | - Tomohiro Konno
- Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
| | - Shigeo Sato
- Research Institute of Electrical Communication (RIEC), Tohoku University, Sendai, Japan
- Graduate School of Engineering, Tohoku University, Sendai, Japan
| | - Ayumi Hirano-Iwata
- Research Institute of Electrical Communication (RIEC), Tohoku University, Sendai, Japan
- Graduate School of Engineering, Tohoku University, Sendai, Japan
- Graduate School of Biomedical Engineering, Tohoku University, Sendai, Japan
- Advanced Institute for Materials Research (WPI-AIMR), Tohoku University, Sendai, Japan
| | - Anna Levina
- Max Planck Institute for Biological Cybernetics, Tübingen, Germany
- Department of Computer Science, University of Tübingen, Tübingen, Germany
| | - Viola Priesemann
- Max Planck Institute for Dynamics and Self-Organization, Göttingen, Germany
- Institute for the Dynamics of Complex Systems, University of Göttingen, Göttingen, Germany
| | - Miguel A. Muñoz
- Departamento de Electromagnetismo y Física de la Materia, Universidad de Granada, Granada, Spain
- Instituto Carlos I de Física Teórica y Computacional, Universidad de Granada, Granada, Spain
| | | | - Jordi Soriano
- Departament de Física de la Matèria Condensada, Universitat de Barcelona, Barcelona, Spain
- Universitat de Barcelona Institute of Complex Systems (UBICS), Barcelona, Spain
| |
Collapse
|
3
|
Adegoke MA, Teter O, Meaney DF. Flexibility of in vitro cortical circuits influences resilience from microtrauma. Front Cell Neurosci 2022; 16:991740. [PMID: 36589287 PMCID: PMC9803265 DOI: 10.3389/fncel.2022.991740] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Accepted: 11/28/2022] [Indexed: 12/23/2022] Open
Abstract
Background Small clusters comprising hundreds to thousands of neurons are an important level of brain architecture that correlates single neuronal properties to fulfill brain function, but the specific mechanisms through which this scaling occurs are not well understood. In this study, we developed an in vitro experimental platform of small neuronal circuits (islands) to probe the importance of structural properties for their development, physiology, and response to microtrauma. Methods Primary cortical neurons were plated on a substrate patterned to promote attachment in clusters of hundreds of cells (islands), transduced with GCaMP6f, allowed to mature until 10-13 days in vitro (DIV), and monitored with Ca2+ as a non-invasive proxy for electrical activity. We adjusted two structural factors-island size and cellular density-to evaluate their role in guiding spontaneous activity and network formation in neuronal islands. Results We found cellular density, but not island size, regulates of circuit activity and network function in this system. Low cellular density islands can achieve many states of activity, while high cellular density biases islands towards a limited regime characterized by low rates of activity and high synchronization, a property we summarized as "flexibility." The injury severity required for an island to lose activity in 50% of its population was significantly higher in low-density, high flexibility islands. Conclusion Together, these studies demonstrate flexible living cortical circuits are more resilient to microtrauma, providing the first evidence that initial circuit state may be a key factor to consider when evaluating the consequences of trauma to the cortex.
Collapse
Affiliation(s)
- Modupe A. Adegoke
- Department of Bioengineering, School of Engineering and Applied Sciences, University of Pennsylvania, Philadelphia, PA, United States
| | - Olivia Teter
- Department of Bioengineering, School of Engineering and Applied Sciences, University of Pennsylvania, Philadelphia, PA, United States
| | - David F. Meaney
- Department of Bioengineering, School of Engineering and Applied Sciences, University of Pennsylvania, Philadelphia, PA, United States,Department of Neurosurgery, Penn Center for Brain Injury and Repair, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States,*Correspondence: David F. Meaney,
| |
Collapse
|
4
|
Pré D, Wooten AT, Biesmans S, Hinckley S, Zhou H, Sherman SP, Kakad P, Gearhart J, Bang AG. Development of a platform to investigate long-term potentiation in human iPSC-derived neuronal networks. Stem Cell Reports 2022; 17:2141-2155. [PMID: 35985330 PMCID: PMC9481914 DOI: 10.1016/j.stemcr.2022.07.012] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Revised: 07/18/2022] [Accepted: 07/19/2022] [Indexed: 01/25/2023] Open
Abstract
Impairment of long-term potentiation (LTP) is a common feature of many pre-clinical models of neurological disorders; however, studies in humans are limited by the inaccessibility of the brain. Human induced pluripotent stem cells (hiPSCs) provide a unique opportunity to study LTP in disease-specific genetic backgrounds. Here we describe a multi-electrode array (MEA)-based assay to investigate chemically induced LTP (cLTP) across entire networks of hiPSC-derived midbrain dopaminergic (DA) and cortical neuronal populations that lasts for days, allowing studies of the late phases of LTP and enabling detection of associated molecular changes. We show that cLTP on midbrain DA neuronal networks is largely independent of the N-methyl-D-aspartate receptor (NMDAR) and partially dependent on brain-derived neurotrophic factor (BDNF). Finally, we describe activity-regulated gene expression induced by cLTP. This cLTP-MEA assay platform will enable phenotype discovery and higher-throughput analyses of synaptic plasticity on hiPSC-derived neurons.
Collapse
Affiliation(s)
- Deborah Pré
- Conrad Prebys Center for Chemical Genomics, Sanford Burnham Prebys Medical Discovery Institute, 10901 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Alexander T Wooten
- Conrad Prebys Center for Chemical Genomics, Sanford Burnham Prebys Medical Discovery Institute, 10901 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Steven Biesmans
- Conrad Prebys Center for Chemical Genomics, Sanford Burnham Prebys Medical Discovery Institute, 10901 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Sandy Hinckley
- Conrad Prebys Center for Chemical Genomics, Sanford Burnham Prebys Medical Discovery Institute, 10901 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Haowen Zhou
- Conrad Prebys Center for Chemical Genomics, Sanford Burnham Prebys Medical Discovery Institute, 10901 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Sean P Sherman
- Conrad Prebys Center for Chemical Genomics, Sanford Burnham Prebys Medical Discovery Institute, 10901 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Priyanka Kakad
- Conrad Prebys Center for Chemical Genomics, Sanford Burnham Prebys Medical Discovery Institute, 10901 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Jeffrey Gearhart
- Henry M. Jackson Foundation for the Advancement of Military Medicine on Contract to USAF School of Aerospace Medicine, Wright-Patterson AFB, Dayton, OH 45433, USA
| | - Anne G Bang
- Conrad Prebys Center for Chemical Genomics, Sanford Burnham Prebys Medical Discovery Institute, 10901 North Torrey Pines Road, La Jolla, CA 92037, USA.
| |
Collapse
|
5
|
Kulkarni AS, Burns MR, Brundin P, Wesson DW. Linking α-synuclein-induced synaptopathy and neural network dysfunction in early Parkinson's disease. Brain Commun 2022; 4:fcac165. [PMID: 35822101 PMCID: PMC9272065 DOI: 10.1093/braincomms/fcac165] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 03/11/2022] [Accepted: 06/20/2022] [Indexed: 01/18/2023] Open
Abstract
The prodromal phase of Parkinson's disease is characterized by aggregation of the misfolded pathogenic protein α-synuclein in select neural centres, co-occurring with non-motor symptoms including sensory and cognitive loss, and emotional disturbances. It is unclear whether neuronal loss is significant during the prodrome. Underlying these symptoms are synaptic impairments and aberrant neural network activity. However, the relationships between synaptic defects and network-level perturbations are not established. In experimental models, pathological α-synuclein not only impacts neurotransmission at the synaptic level, but also leads to changes in brain network-level oscillatory dynamics-both of which likely contribute to non-motor deficits observed in Parkinson's disease. Here we draw upon research from both human subjects and experimental models to propose a 'synapse to network prodrome cascade' wherein before overt cell death, pathological α-synuclein induces synaptic loss and contributes to aberrant network activity, which then gives rise to prodromal symptomology. As the disease progresses, abnormal patterns of neural activity ultimately lead to neuronal loss and clinical progression of disease. Finally, we outline goals and research needed to unravel the basis of functional impairments in Parkinson's disease and other α-synucleinopathies.
Collapse
Affiliation(s)
- Aishwarya S Kulkarni
- Department of Pharmacology & Therapeutics, University of Florida, 1200 Newell Dr, Gainesville, FL 32610, USA
| | - Matthew R Burns
- Department of Neurology, University of Florida, 1200 Newell Dr, Gainesville, FL 32610, USA
- Norman Fixel Institute for Neurological Disorders, University of Florida, 1200 Newell Dr, Gainesville, FL 32610, USA
| | - Patrik Brundin
- Pharma Research and Early Development (pRED), F. Hoffman-La Roche, Little Falls, NJ, USA
| | - Daniel W Wesson
- Department of Pharmacology & Therapeutics, University of Florida, 1200 Newell Dr, Gainesville, FL 32610, USA
- Norman Fixel Institute for Neurological Disorders, University of Florida, 1200 Newell Dr, Gainesville, FL 32610, USA
| |
Collapse
|
6
|
Zendrikov D, Paraskevov A. Emergent population activity in metric-free and metric networks of neurons with stochastic spontaneous spikes and dynamic synapses. Neurocomputing 2021. [DOI: 10.1016/j.neucom.2020.11.073] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
7
|
Moriya F, Shimba K, Kotani K, Jimbo Y. Modulation of dynamics in a pre-existing hippocampal network by neural stem cells on a microelectrode array. J Neural Eng 2021; 18. [PMID: 34380120 DOI: 10.1088/1741-2552/ac1c88] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2021] [Accepted: 08/11/2021] [Indexed: 11/12/2022]
Abstract
Objective.Neural stem cells (NSCs) are continuously produced throughout life in the hippocampus, which is a vital structure for learning and memory. NSCs in the brain incorporate into the functional hippocampal circuits and contribute to processing information. However, little is known about the mechanisms of NSCs' activity in a pre-existing neuronal network. Here, we investigate the role of NSCs in the neuronal activity of a pre-existing hippocampalin vitronetwork grown on microelectrode arrays.Approach.We assessed the change in internal dynamics of the network by additional NSCs based on spontaneous activity. We also evaluated the networks' ability to discriminate between different input patterns by measuring evoked activity in response to external inputs.Main results.Analysis of spontaneous activity revealed that additional NSCs prolonged network bursts with longer intervals, generated a lower number of initiating patterns, and decreased synchronization among neurons. Moreover, the network with NSCs showed higher synchronicity in close connections among neurons responding to external inputs and a larger difference in spike counts and cross-correlations during evoked response between two different inputs. Taken together, our results suggested that NSCs alter the internal dynamics of the pre-existing hippocampal network and produce more specific responses to external inputs, thus enhancing the ability of the network to differentiate two different inputs.Significance.We demonstrated that NSCs improve the ability to distinguish external inputs by modulating the internal dynamics of a pre-existing network in a hippocampal culture. Our results provide novel insights into the relationship between NSCs and learning and memory.
Collapse
Affiliation(s)
- Fumika Moriya
- The Department of Precision Engineering, School of Engineering, The University of Tokyo, Tokyo 113-8656, Japan.,The Japan Society for the Promotion of Science (JSPS), Tokyo, Japan
| | - Kenta Shimba
- The Department of Precision Engineering, School of Engineering, The University of Tokyo, Tokyo 113-8656, Japan
| | - Kiyoshi Kotani
- The Research Center for Advanced Science and Technology, The University of Tokyo, Tokyo 153-8904, Japan
| | - Yasuhiko Jimbo
- The Department of Precision Engineering, School of Engineering, The University of Tokyo, Tokyo 113-8656, Japan
| |
Collapse
|
8
|
Mazaud D, Capano A, Rouach N. The many ways astroglial connexins regulate neurotransmission and behavior. Glia 2021; 69:2527-2545. [PMID: 34101261 DOI: 10.1002/glia.24040] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2020] [Revised: 05/17/2021] [Accepted: 05/21/2021] [Indexed: 12/18/2022]
Abstract
Astrocytes have emerged as major players in the brain, contributing to many functions such as energy supply, neurotransmission, and behavior. They accomplish these functions in part via their capacity to form widespread intercellular networks and to release neuroactive factors, which can modulate neurotransmission at different levels, from individual synapses to neuronal networks. The extensive network communication of astrocytes is primarily mediated by gap junction channels composed of two connexins, Cx30 and Cx43, which present distinct temporal and spatial expression patterns. Yet, astroglial connexins are also involved in direct exchange with the extracellular space via hemichannels, as well as in adhesion and signaling processes via unconventional nonchannel functions. Accumulating evidence indicate that astrocytes modulate neurotransmission and behavior through these diverse connexin functions. We here review the many ways astroglial connexins regulate neuronal activity from the molecular level to behavior.
Collapse
Affiliation(s)
- David Mazaud
- Neuroglial Interactions in Cerebral Physiology and Pathologies, Center for Interdisciplinary Research in Biology, Collège de France, CNRS UMR 7241, INSERM U1050, Labex Memolife, PSL Research University, Paris, France
| | - Anna Capano
- Neuroglial Interactions in Cerebral Physiology and Pathologies, Center for Interdisciplinary Research in Biology, Collège de France, CNRS UMR 7241, INSERM U1050, Labex Memolife, PSL Research University, Paris, France.,Doctoral School N°158, Sorbonne University, Paris, France
| | - Nathalie Rouach
- Neuroglial Interactions in Cerebral Physiology and Pathologies, Center for Interdisciplinary Research in Biology, Collège de France, CNRS UMR 7241, INSERM U1050, Labex Memolife, PSL Research University, Paris, France
| |
Collapse
|
9
|
Perkins EM, Burr K, Banerjee P, Mehta AR, Dando O, Selvaraj BT, Suminaite D, Nanda J, Henstridge CM, Gillingwater TH, Hardingham GE, Wyllie DJA, Chandran S, Livesey MR. Altered network properties in C9ORF72 repeat expansion cortical neurons are due to synaptic dysfunction. Mol Neurodegener 2021; 16:13. [PMID: 33663561 PMCID: PMC7931347 DOI: 10.1186/s13024-021-00433-8] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Accepted: 02/14/2021] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Physiological disturbances in cortical network excitability and plasticity are established and widespread in amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD) patients, including those harbouring the C9ORF72 repeat expansion (C9ORF72RE) mutation - the most common genetic impairment causal to ALS and FTD. Noting that perturbations in cortical function are evidenced pre-symptomatically, and that the cortex is associated with widespread pathology, cortical dysfunction is thought to be an early driver of neurodegenerative disease progression. However, our understanding of how altered network function manifests at the cellular and molecular level is not clear. METHODS To address this we have generated cortical neurons from patient-derived iPSCs harbouring C9ORF72RE mutations, as well as from their isogenic expansion-corrected controls. We have established a model of network activity in these neurons using multi-electrode array electrophysiology. We have then mechanistically examined the physiological processes underpinning network dysfunction using a combination of patch-clamp electrophysiology, immunocytochemistry, pharmacology and transcriptomic profiling. RESULTS We find that C9ORF72RE causes elevated network burst activity, associated with enhanced synaptic input, yet lower burst duration, attributable to impaired pre-synaptic vesicle dynamics. We also show that the C9ORF72RE is associated with impaired synaptic plasticity. Moreover, RNA-seq analysis revealed dysregulated molecular pathways impacting on synaptic function. All molecular, cellular and network deficits are rescued by CRISPR/Cas9 correction of C9ORF72RE. Our study provides a mechanistic view of the early dysregulated processes that underpin cortical network dysfunction in ALS-FTD. CONCLUSION These findings suggest synaptic pathophysiology is widespread in ALS-FTD and has an early and fundamental role in driving altered network function that is thought to contribute to neurodegenerative processes in these patients. The overall importance is the identification of previously unidentified defects in pre and postsynaptic compartments affecting synaptic plasticity, synaptic vesicle stores, and network propagation, which directly impact upon cortical function.
Collapse
Affiliation(s)
- Emma M. Perkins
- Euan MacDonald Centre for MND Research, University of Edinburgh, Edinburgh, EH16 4SB UK
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, EH16 4SB UK
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, EH8 9XD UK
| | - Karen Burr
- Euan MacDonald Centre for MND Research, University of Edinburgh, Edinburgh, EH16 4SB UK
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, EH16 4SB UK
- UK Dementia Research Institute at the University of Edinburgh, Edinburgh, EH16 4SB UK
| | - Poulomi Banerjee
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, EH16 4SB UK
- UK Dementia Research Institute at the University of Edinburgh, Edinburgh, EH16 4SB UK
| | - Arpan R. Mehta
- Euan MacDonald Centre for MND Research, University of Edinburgh, Edinburgh, EH16 4SB UK
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, EH16 4SB UK
- UK Dementia Research Institute at the University of Edinburgh, Edinburgh, EH16 4SB UK
| | - Owen Dando
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, EH8 9XD UK
- UK Dementia Research Institute at the University of Edinburgh, Edinburgh, EH16 4SB UK
- Simons Initiative for the Developing Brain, University of Edinburgh, Edinburgh, EH8 9XD UK
| | - Bhuvaneish T. Selvaraj
- Euan MacDonald Centre for MND Research, University of Edinburgh, Edinburgh, EH16 4SB UK
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, EH16 4SB UK
- UK Dementia Research Institute at the University of Edinburgh, Edinburgh, EH16 4SB UK
| | - Daumante Suminaite
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, EH8 9XD UK
| | - Jyoti Nanda
- Euan MacDonald Centre for MND Research, University of Edinburgh, Edinburgh, EH16 4SB UK
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, EH16 4SB UK
- UK Dementia Research Institute at the University of Edinburgh, Edinburgh, EH16 4SB UK
| | - Christopher M. Henstridge
- Euan MacDonald Centre for MND Research, University of Edinburgh, Edinburgh, EH16 4SB UK
- Division of Systems Medicine, School of Medicine, University of Dundee, Dundee, DD1 9SY UK
| | - Thomas H. Gillingwater
- Euan MacDonald Centre for MND Research, University of Edinburgh, Edinburgh, EH16 4SB UK
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, EH8 9XD UK
| | - Giles E. Hardingham
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, EH8 9XD UK
- UK Dementia Research Institute at the University of Edinburgh, Edinburgh, EH16 4SB UK
- Simons Initiative for the Developing Brain, University of Edinburgh, Edinburgh, EH8 9XD UK
| | - David J. A. Wyllie
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, EH8 9XD UK
- Simons Initiative for the Developing Brain, University of Edinburgh, Edinburgh, EH8 9XD UK
- Centre for Brain Development and Repair, inStem, Bangalore, 560065 India
| | - Siddharthan Chandran
- Euan MacDonald Centre for MND Research, University of Edinburgh, Edinburgh, EH16 4SB UK
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, EH16 4SB UK
- UK Dementia Research Institute at the University of Edinburgh, Edinburgh, EH16 4SB UK
- Simons Initiative for the Developing Brain, University of Edinburgh, Edinburgh, EH8 9XD UK
- Centre for Brain Development and Repair, inStem, Bangalore, 560065 India
| | - Matthew R. Livesey
- Euan MacDonald Centre for MND Research, University of Edinburgh, Edinburgh, EH16 4SB UK
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, EH8 9XD UK
- Simons Initiative for the Developing Brain, University of Edinburgh, Edinburgh, EH8 9XD UK
- Sheffield Institute for Translational Neuroscience, University of Sheffield, Sheffield, S10 2HQ UK
| |
Collapse
|
10
|
Ludl AA, Soriano J. Impact of Physical Obstacles on the Structural and Effective Connectivity of in silico Neuronal Circuits. Front Comput Neurosci 2020; 14:77. [PMID: 32982710 PMCID: PMC7488194 DOI: 10.3389/fncom.2020.00077] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Accepted: 07/21/2020] [Indexed: 11/13/2022] Open
Abstract
Scaffolds and patterned substrates are among the most successful strategies to dictate the connectivity between neurons in culture. Here, we used numerical simulations to investigate the capacity of physical obstacles placed on a flat substrate to shape structural connectivity, and in turn collective dynamics and effective connectivity, in biologically-realistic neuronal networks. We considered μ-sized obstacles placed in mm-sized networks. Three main obstacle shapes were explored, namely crosses, circles and triangles of isosceles profile. They occupied either a small area fraction of the substrate or populated it entirely in a periodic manner. From the point of view of structure, all obstacles promoted short length-scale connections, shifted the in- and out-degree distributions toward lower values, and increased the modularity of the networks. The capacity of obstacles to shape distinct structural traits depended on their density and the ratio between axonal length and substrate diameter. For high densities, different features were triggered depending on obstacle shape, with crosses trapping axons in their vicinity and triangles funneling axons along the reverse direction of their tip. From the point of view of dynamics, obstacles reduced the capacity of networks to spontaneously activate, with triangles in turn strongly dictating the direction of activity propagation. Effective connectivity networks, inferred using transfer entropy, exhibited distinct modular traits, indicating that the presence of obstacles facilitated the formation of local effective microcircuits. Our study illustrates the potential of physical constraints to shape structural blueprints and remodel collective activity, and may guide investigations aimed at mimicking organizational traits of biological neuronal circuits.
Collapse
Affiliation(s)
- Adriaan-Alexander Ludl
- Computational Biology Unit, Department of Informatics, University of Bergen, Bergen, Norway.,Departament de Física de la Matèria Condensada, Universitat de Barcelona, Barcelona, Spain.,Universitat de Barcelona Institute of Complex Systems (UBICS), Barcelona, Spain
| | - Jordi Soriano
- Departament de Física de la Matèria Condensada, Universitat de Barcelona, Barcelona, Spain.,Universitat de Barcelona Institute of Complex Systems (UBICS), Barcelona, Spain
| |
Collapse
|
11
|
Dallérac G, Zapata J, Rouach N. Versatile control of synaptic circuits by astrocytes: where, when and how? Nat Rev Neurosci 2019; 19:729-743. [PMID: 30401802 DOI: 10.1038/s41583-018-0080-6] [Citation(s) in RCA: 114] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Close structural and functional interactions of astrocytes with synapses play an important role in brain function. The repertoire of ways in which astrocytes can regulate synaptic transmission is complex so that they can both promote and dampen synaptic efficacy. Such contrasting effects raise questions regarding the determinants of these divergent astroglial functions. Recent findings provide insights into where, when and how astroglial regulation of synapses takes place by revealing major molecular and functional intrinsic heterogeneity as well as switches in astrocytes occurring during development or specific patterns of neuronal activity. Astrocytes may therefore be seen as boosters or gatekeepers of synaptic circuits depending on their intrinsic and transformative properties throughout life.
Collapse
Affiliation(s)
- Glenn Dallérac
- Center for Interdisciplinary Research in Biology, Collège de France, CNRS UMR 7241, INSERM U1050, Labex Memolife, PSL Research University, Paris, France
| | - Jonathan Zapata
- Center for Interdisciplinary Research in Biology, Collège de France, CNRS UMR 7241, INSERM U1050, Labex Memolife, PSL Research University, Paris, France
| | - Nathalie Rouach
- Center for Interdisciplinary Research in Biology, Collège de France, CNRS UMR 7241, INSERM U1050, Labex Memolife, PSL Research University, Paris, France.
| |
Collapse
|
12
|
Flores JA, Ramírez-Ponce MP, Montes MÁ, Balseiro-Gómez S, Acosta J, Álvarez de Toledo G, Alés E. Proteoglycans involved in bidirectional communication between mast cells and hippocampal neurons. J Neuroinflammation 2019; 16:107. [PMID: 31109355 PMCID: PMC6528191 DOI: 10.1186/s12974-019-1504-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Accepted: 05/08/2019] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Mast cells (MCs) in the brain can respond to environmental cues and relay signals to neurons that may directly influence neuronal electrical activity, calcium signaling, and neurotransmission. MCs also express receptors for neurotransmitters and consequently can be activated by them. Here, we developed a coculture model of peritoneal MCs, incubated together with dissociated hippocampal neurons for the study of cellular mechanisms involved in the mast cell-neuron interactions. METHODS Calcium imaging was used to simultaneously record changes in intracellular calcium [Ca2+]i in neurons and MCs. To provide insight into the contribution of MCs on neurotransmitter release in rat hippocampal neurons, we used analysis of FM dye release, evoked by a cocktail of mediators from MCs stimulated by heat. RESULTS Bidirectional communication is set up between MCs and hippocampal neurons. Neuronal depolarization caused intracellular calcium [Ca2+]i oscillations in MCs that produced a quick response in neurons. Furthermore, activation of MCs with antigen or the secretagogue compound 48/80 also resulted in a neuronal [Ca2+]i response. Moreover, local application onto neurons of the MC mediator cocktail elicited Ca2+ transients and a synaptic release associated with FM dye destaining. Neuronal response was partially blocked by D-APV, a N-methyl-D-aspartate receptor (NMDAR) antagonist, and was inhibited when the cocktail was pre-digested with chondroitinase ABC, which induces enzymatic removal of proteoglycans of chondroitin sulfate (CS). CONCLUSIONS MC-hippocampal neuron interaction affects neuronal [Ca2+]i and exocytosis signaling through a NMDAR-dependent mechanism.
Collapse
Affiliation(s)
- Juan Antonio Flores
- Dpto. de Fisiología Médica y Biofísica, Facultad de Medicina, Universidad de Sevilla, Av. Sánchez Pizjuán 4, 41009 Sevilla, Spain
| | - María Pilar Ramírez-Ponce
- Dpto. de Fisiología Médica y Biofísica, Facultad de Medicina, Universidad de Sevilla, Av. Sánchez Pizjuán 4, 41009 Sevilla, Spain
| | - María Ángeles Montes
- Dpto. de Fisiología Médica y Biofísica, Facultad de Medicina, Universidad de Sevilla, Av. Sánchez Pizjuán 4, 41009 Sevilla, Spain
| | - Santiago Balseiro-Gómez
- Dpto. de Fisiología Médica y Biofísica, Facultad de Medicina, Universidad de Sevilla, Av. Sánchez Pizjuán 4, 41009 Sevilla, Spain
- Present Address: Department of Neuroscience, Yale School of Medicine, 295 Congress Avenue, New Haven, CT 06510 USA
| | - Jorge Acosta
- Dpto. de Fisiología Médica y Biofísica, Facultad de Medicina, Universidad de Sevilla, Av. Sánchez Pizjuán 4, 41009 Sevilla, Spain
| | - Guillermo Álvarez de Toledo
- Dpto. de Fisiología Médica y Biofísica, Facultad de Medicina, Universidad de Sevilla, Av. Sánchez Pizjuán 4, 41009 Sevilla, Spain
| | - Eva Alés
- Dpto. de Fisiología Médica y Biofísica, Facultad de Medicina, Universidad de Sevilla, Av. Sánchez Pizjuán 4, 41009 Sevilla, Spain
| |
Collapse
|
13
|
Valente P, Romei A, Fadda M, Sterlini B, Lonardoni D, Forte N, Fruscione F, Castroflorio E, Michetti C, Giansante G, Valtorta F, Tsai JW, Zara F, Nieus T, Corradi A, Fassio A, Baldelli P, Benfenati F. Constitutive Inactivation of the PRRT2 Gene Alters Short-Term Synaptic Plasticity and Promotes Network Hyperexcitability in Hippocampal Neurons. Cereb Cortex 2018; 29:2010-2033. [DOI: 10.1093/cercor/bhy079] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2017] [Revised: 03/13/2018] [Indexed: 12/20/2022] Open
Affiliation(s)
- Pierluigi Valente
- Department of Experimental Medicine, University of Genova, Viale Benedetto XV 3, Genova, Italy
| | - Alessandra Romei
- Department of Experimental Medicine, University of Genova, Viale Benedetto XV 3, Genova, Italy
- Center for Synaptic Neuroscience and Technology, Istituto Italiano di Tecnologia, Largo Rosanna Benzi 10, Genova, Italy
| | - Manuela Fadda
- Department of Experimental Medicine, University of Genova, Viale Benedetto XV 3, Genova, Italy
| | - Bruno Sterlini
- Department of Experimental Medicine, University of Genova, Viale Benedetto XV 3, Genova, Italy
- Center for Synaptic Neuroscience and Technology, Istituto Italiano di Tecnologia, Largo Rosanna Benzi 10, Genova, Italy
| | - Davide Lonardoni
- Department of Neuroscience and Brain Technologies, Istituto Italiano di Tecnologia, Via Morego 30, Genova, Italy
| | - Nicola Forte
- Center for Synaptic Neuroscience and Technology, Istituto Italiano di Tecnologia, Largo Rosanna Benzi 10, Genova, Italy
| | - Floriana Fruscione
- Laboratory of Neurogenetics and Neuroscience, Department Head-Neck and Neuroscience, Istituto Giannina Gaslini, Via Gerolamo Gaslini 5, Genova, Italy
| | - Enrico Castroflorio
- Department of Experimental Medicine, University of Genova, Viale Benedetto XV 3, Genova, Italy
- Center for Synaptic Neuroscience and Technology, Istituto Italiano di Tecnologia, Largo Rosanna Benzi 10, Genova, Italy
| | - Caterina Michetti
- Center for Synaptic Neuroscience and Technology, Istituto Italiano di Tecnologia, Largo Rosanna Benzi 10, Genova, Italy
| | - Giorgia Giansante
- Department of Experimental Medicine, University of Genova, Viale Benedetto XV 3, Genova, Italy
| | - Flavia Valtorta
- San Raffaele Scientific Institute and Vita Salute University, Via Olgettina 58, Milano, Italy
| | - Jin-Wu Tsai
- Institute of Brain Science, Brain Research Center, National Yang-Ming University, Taipei, Taiwan
| | - Federico Zara
- Laboratory of Neurogenetics and Neuroscience, Department Head-Neck and Neuroscience, Istituto Giannina Gaslini, Via Gerolamo Gaslini 5, Genova, Italy
| | - Thierry Nieus
- Department of Neuroscience and Brain Technologies, Istituto Italiano di Tecnologia, Via Morego 30, Genova, Italy
| | - Anna Corradi
- Department of Experimental Medicine, University of Genova, Viale Benedetto XV 3, Genova, Italy
- Center for Synaptic Neuroscience and Technology, Istituto Italiano di Tecnologia, Largo Rosanna Benzi 10, Genova, Italy
| | - Anna Fassio
- Department of Experimental Medicine, University of Genova, Viale Benedetto XV 3, Genova, Italy
- Center for Synaptic Neuroscience and Technology, Istituto Italiano di Tecnologia, Largo Rosanna Benzi 10, Genova, Italy
| | - Pietro Baldelli
- Department of Experimental Medicine, University of Genova, Viale Benedetto XV 3, Genova, Italy
- Center for Synaptic Neuroscience and Technology, Istituto Italiano di Tecnologia, Largo Rosanna Benzi 10, Genova, Italy
| | - Fabio Benfenati
- Department of Experimental Medicine, University of Genova, Viale Benedetto XV 3, Genova, Italy
- Center for Synaptic Neuroscience and Technology, Istituto Italiano di Tecnologia, Largo Rosanna Benzi 10, Genova, Italy
| |
Collapse
|
14
|
Rojas C, Tedesco M, Massobrio P, Marino A, Ciofani G, Martinoia S, Raiteri R. Acoustic stimulation can induce a selective neural network response mediated by piezoelectric nanoparticles. J Neural Eng 2018; 15:036016. [DOI: 10.1088/1741-2552/aaa140] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
|
15
|
Nieus T, D'Andrea V, Amin H, Di Marco S, Safaai H, Maccione A, Berdondini L, Panzeri S. State-dependent representation of stimulus-evoked activity in high-density recordings of neural cultures. Sci Rep 2018; 8:5578. [PMID: 29615719 PMCID: PMC5882875 DOI: 10.1038/s41598-018-23853-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2018] [Accepted: 03/21/2018] [Indexed: 01/01/2023] Open
Abstract
Neuronal responses to external stimuli vary from trial to trial partly because they depend on continuous spontaneous variations of the state of neural circuits, reflected in variations of ongoing activity prior to stimulus presentation. Understanding how post-stimulus responses relate to the pre-stimulus spontaneous activity is thus important to understand how state dependence affects information processing and neural coding, and how state variations can be discounted to better decode single-trial neural responses. Here we exploited high-resolution CMOS electrode arrays to record simultaneously from thousands of electrodes in in-vitro cultures stimulated at specific sites. We used information-theoretic analyses to study how ongoing activity affects the information that neuronal responses carry about the location of the stimuli. We found that responses exhibited state dependence on the time between the last spontaneous burst and the stimulus presentation and that the dependence could be described with a linear model. Importantly, we found that a small number of selected neurons carry most of the stimulus information and contribute to the state-dependent information gain. This suggests that a major value of large-scale recording is that it individuates the small subset of neurons that carry most information and that benefit the most from knowledge of its state dependence.
Collapse
Affiliation(s)
- Thierry Nieus
- NetS3 Laboratory, Neuroscience and Brain Technologies Department, Istituto Italiano di Tecnologia, Genova, Italy. .,Department of Biomedical and Clinical Sciences "Luigi Sacco", Università di Milano, Milano, Italy.
| | - Valeria D'Andrea
- Neural Computation Laboratory, Center for Neuroscience and Cognitive Systems @UniTn, Istituto Italiano di Tecnologia, Rovereto, Italy
| | - Hayder Amin
- NetS3 Laboratory, Neuroscience and Brain Technologies Department, Istituto Italiano di Tecnologia, Genova, Italy
| | - Stefano Di Marco
- NetS3 Laboratory, Neuroscience and Brain Technologies Department, Istituto Italiano di Tecnologia, Genova, Italy.,Scienze cliniche applicate e biotecnologiche, Università dell'Aquila, L'Aquila, Italy
| | - Houman Safaai
- Neural Computation Laboratory, Center for Neuroscience and Cognitive Systems @UniTn, Istituto Italiano di Tecnologia, Rovereto, Italy.,Department of Neurobiology, Harvard Medical School, 02115, Boston, Massachusetts, USA
| | - Alessandro Maccione
- NetS3 Laboratory, Neuroscience and Brain Technologies Department, Istituto Italiano di Tecnologia, Genova, Italy
| | - Luca Berdondini
- NetS3 Laboratory, Neuroscience and Brain Technologies Department, Istituto Italiano di Tecnologia, Genova, Italy
| | - Stefano Panzeri
- Neural Computation Laboratory, Center for Neuroscience and Cognitive Systems @UniTn, Istituto Italiano di Tecnologia, Rovereto, Italy.
| |
Collapse
|
16
|
Baertsch NA, Baertsch HC, Ramirez JM. The interdependence of excitation and inhibition for the control of dynamic breathing rhythms. Nat Commun 2018; 9:843. [PMID: 29483589 PMCID: PMC5827754 DOI: 10.1038/s41467-018-03223-x] [Citation(s) in RCA: 119] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Accepted: 01/26/2018] [Indexed: 11/09/2022] Open
Abstract
The preBötzinger Complex (preBötC), a medullary network critical for breathing, relies on excitatory interneurons to generate the inspiratory rhythm. Yet, half of preBötC neurons are inhibitory, and the role of inhibition in rhythmogenesis remains controversial. Using optogenetics and electrophysiology in vitro and in vivo, we demonstrate that the intrinsic excitability of excitatory neurons is reduced following large depolarizing inspiratory bursts. This refractory period limits the preBötC to very slow breathing frequencies. Inhibition integrated within the network is required to prevent overexcitation of preBötC neurons, thereby regulating the refractory period and allowing rapid breathing. In vivo, sensory feedback inhibition also regulates the refractory period, and in slowly breathing mice with sensory feedback removed, activity of inhibitory, but not excitatory, neurons restores breathing to physiological frequencies. We conclude that excitation and inhibition are interdependent for the breathing rhythm, because inhibition permits physiological preBötC bursting by controlling refractory properties of excitatory neurons.
Collapse
Affiliation(s)
- Nathan Andrew Baertsch
- Center for Integrative Brain Research, Seattle Children's Research Institute, 1900 9th Avenue JMB10, Seattle, WA, 98101, USA
| | - Hans Christopher Baertsch
- Center for Integrative Brain Research, Seattle Children's Research Institute, 1900 9th Avenue JMB10, Seattle, WA, 98101, USA
| | - Jan Marino Ramirez
- Center for Integrative Brain Research, Seattle Children's Research Institute, 1900 9th Avenue JMB10, Seattle, WA, 98101, USA.
- Department of Neurological Surgery, University of Washington, 1900 9th Avenue, JMB10, Seattle, WA, 98101, USA.
- Department of Pediatrics, University of Washington, 1900 9th Avenue, JMB10, Seattle, WA, 98101, USA.
| |
Collapse
|
17
|
Fardet T, Ballandras M, Bottani S, Métens S, Monceau P. Understanding the Generation of Network Bursts by Adaptive Oscillatory Neurons. Front Neurosci 2018; 12:41. [PMID: 29467607 PMCID: PMC5808224 DOI: 10.3389/fnins.2018.00041] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2017] [Accepted: 01/17/2018] [Indexed: 12/26/2022] Open
Abstract
Experimental and numerical studies have revealed that isolated populations of oscillatory neurons can spontaneously synchronize and generate periodic bursts involving the whole network. Such a behavior has notably been observed for cultured neurons in rodent's cortex or hippocampus. We show here that a sufficient condition for this network bursting is the presence of an excitatory population of oscillatory neurons which displays spike-driven adaptation. We provide an analytic model to analyze network bursts generated by coupled adaptive exponential integrate-and-fire neurons. We show that, for strong synaptic coupling, intrinsically tonic spiking neurons evolve to reach a synchronized intermittent bursting state. The presence of inhibitory neurons or plastic synapses can then modulate this dynamics in many ways but is not necessary for its appearance. Thanks to a simple self-consistent equation, our model gives an intuitive and semi-quantitative tool to understand the bursting behavior. Furthermore, it suggests that after-hyperpolarization currents are sufficient to explain bursting termination. Through a thorough mapping between the theoretical parameters and ion-channel properties, we discuss the biological mechanisms that could be involved and the relevance of the explored parameter-space. Such an insight enables us to propose experimentally-testable predictions regarding how blocking fast, medium or slow after-hyperpolarization channels would affect the firing rate and burst duration, as well as the interburst interval.
Collapse
Affiliation(s)
- Tanguy Fardet
- Laboratoire Matière et Systèmes Complexes, UMR 7057, Université Paris Diderot, USPC, Paris, France
| | - Mathieu Ballandras
- Laboratoire Matière et Systèmes Complexes, UMR 7057, Université Paris Diderot, USPC, Paris, France
| | - Samuel Bottani
- Laboratoire Matière et Systèmes Complexes, UMR 7057, Université Paris Diderot, USPC, Paris, France
| | - Stéphane Métens
- Laboratoire Matière et Systèmes Complexes, UMR 7057, Université Paris Diderot, USPC, Paris, France
| | - Pascal Monceau
- Laboratoire Matière et Systèmes Complexes, UMR 7057, Université Paris Diderot, USPC, Paris, France.,Department of Physics, Université d'Evry-Val d'Essonne, Évry, France
| |
Collapse
|
18
|
Palazzolo G, Moroni M, Soloperto A, Aletti G, Naldi G, Vassalli M, Nieus T, Difato F. Fast wide-volume functional imaging of engineered in vitro brain tissues. Sci Rep 2017; 7:8499. [PMID: 28819205 PMCID: PMC5561227 DOI: 10.1038/s41598-017-08979-8] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2017] [Accepted: 07/20/2017] [Indexed: 12/14/2022] Open
Abstract
The need for in vitro models that mimic the human brain to replace animal testing and allow high-throughput screening has driven scientists to develop new tools that reproduce tissue-like features on a chip. Three-dimensional (3D) in vitro cultures are emerging as an unmatched platform that preserves the complexity of cell-to-cell connections within a tissue, improves cell survival, and boosts neuronal differentiation. In this context, new and flexible imaging approaches are required to monitor the functional states of 3D networks. Herein, we propose an experimental model based on 3D neuronal networks in an alginate hydrogel, a tunable wide-volume imaging approach, and an efficient denoising algorithm to resolve, down to single cell resolution, the 3D activity of hundreds of neurons expressing the calcium sensor GCaMP6s. Furthermore, we implemented a 3D co-culture system mimicking the contiguous interfaces of distinct brain tissues such as the cortical-hippocampal interface. The analysis of the network activity of single and layered neuronal co-cultures revealed cell-type-specific activities and an organization of neuronal subpopulations that changed in the two culture configurations. Overall, our experimental platform represents a simple, powerful and cost-effective platform for developing and monitoring living 3D layered brain tissue on chip structures with high resolution and high throughput.
Collapse
Affiliation(s)
- G Palazzolo
- Department of Neuroscience and Brain Technologies, Fondazione Istituto Italiano di Tecnologia, Genoa, Italy
| | - M Moroni
- Department of Neuroscience and Brain Technologies, Fondazione Istituto Italiano di Tecnologia, Genoa, Italy.,Center for Neuroscience and Cognitive Systems @UniTn, Istituto Italiano di Tecnologia, Rovereto, Italy.,Center for Mind/Brain Sciences, University of Trento, Rovereto, Italy
| | - A Soloperto
- Department of Neuroscience and Brain Technologies, Fondazione Istituto Italiano di Tecnologia, Genoa, Italy
| | - G Aletti
- Dipartimento di Matematica, Università degli studi di Milano, Milano, Italy
| | - G Naldi
- Dipartimento di Matematica, Università degli studi di Milano, Milano, Italy
| | - M Vassalli
- Institute of Biophysics, National Research Council of Italy, Genoa, Italy
| | - T Nieus
- Department of Biomedical and Clinical Sciences "L. Sacco", Università degli Studi di Milano, Milano, Italy.
| | - F Difato
- Department of Neuroscience and Brain Technologies, Fondazione Istituto Italiano di Tecnologia, Genoa, Italy.
| |
Collapse
|
19
|
Orlandi JG, Casademunt J. Noise focusing in neuronal tissues: Symmetry breaking and localization in excitable networks with quenched disorder. Phys Rev E 2017; 95:052304. [PMID: 28618531 DOI: 10.1103/physreve.95.052304] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2016] [Indexed: 11/07/2022]
Abstract
We introduce a coarse-grained stochastic model for the spontaneous activity of neuronal cultures to explain the phenomenon of noise focusing, which entails localization of the noise activity in excitable networks with metric correlations. The system is modeled as a continuum excitable medium with a state-dependent spatial coupling that accounts for the dynamics of synaptic connections. The most salient feature is the emergence at the mesoscale of a vector field V(r), which acts as an advective carrier of the noise. This entails an explicit symmetry breaking of isotropy and homogeneity that stems from the amplification of the quenched fluctuations of the network by the activity avalanches, concomitant with the excitable dynamics. We discuss the microscopic interpretation of V(r) and propose an explicit construction of it. The coarse-grained model shows excellent agreement with simulations at the network level. The generic nature of the observed phenomena is discussed.
Collapse
Affiliation(s)
- Javier G Orlandi
- Departament de Física de la Matèria Condensada, Universitat de Barcelona, E-08028 Barcelona, Spain.,Complexity Science Group, Department of Physics and Astronomy, University of Calgary, Calgary, Canada T2N 1N4
| | - Jaume Casademunt
- Departament de Física de la Matèria Condensada, Universitat de Barcelona, E-08028 Barcelona, Spain.,Universitat de Barcelona Institute of Complex Systems (UBICS), Universitat de Barcelona, E-08028 Barcelona, Spain
| |
Collapse
|
20
|
Huang CH, Huang YT, Chen CC, Chan CK. Propagation and synchronization of reverberatory bursts in developing cultured networks. J Comput Neurosci 2016; 42:177-185. [PMID: 27942935 DOI: 10.1007/s10827-016-0634-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2016] [Revised: 11/14/2016] [Accepted: 11/21/2016] [Indexed: 11/30/2022]
Abstract
Developing networks of neural systems can exhibit spontaneous, synchronous activities called neural bursts, which can be important in the organization of functional neural circuits. Before the network matures, the activity level of a burst can reverberate in repeated rise-and-falls in periods of hundreds of milliseconds following an initial wave-like propagation of spiking activity, while the burst itself lasts for seconds. To investigate the spatiotemporal structure of the reverberatory bursts, we culture dissociated, rat cortical neurons on a high-density multi-electrode array to record the dynamics of neural activity over the growth and maturation of the network. We find the synchrony of the spiking significantly reduced following the initial wave and the activities become broadly distributed spatially. The synchrony recovers as the system reverberates until the end of the burst. Using a propagation model we infer the spreading speed of the spiking activity, which increases as the culture ages. We perform computer simulations of the system using a physiological model of spiking networks in two spatial dimensions and find the parameters that reproduce the observed resynchronization of spiking in the bursts. An analysis of the simulated dynamics suggests that the depletion of synaptic resources causes the resynchronization. The spatial propagation dynamics of the simulations match well with observations over the course of a burst and point to an interplay of the synaptic efficacy and the noisy neural self-activation in producing the morphology of the bursts.
Collapse
Affiliation(s)
- Chih-Hsu Huang
- Institute of Physics, Academia Sinica, Nangang, Taipei, Taiwan, 115, Republic of China
| | - Yu-Ting Huang
- Institute of Physics, Academia Sinica, Nangang, Taipei, Taiwan, 115, Republic of China.,Department of Physics and Center for Complex Systems, National Central University, Chungli, Taiwan, 320, Republic of China
| | - Chun-Chung Chen
- Institute of Physics, Academia Sinica, Nangang, Taipei, Taiwan, 115, Republic of China.
| | - C K Chan
- Institute of Physics, Academia Sinica, Nangang, Taipei, Taiwan, 115, Republic of China.,Department of Physics and Center for Complex Systems, National Central University, Chungli, Taiwan, 320, Republic of China
| |
Collapse
|
21
|
DeMarse TB, Pan L, Alagapan S, Brewer GJ, Wheeler BC. Feed-Forward Propagation of Temporal and Rate Information between Cortical Populations during Coherent Activation in Engineered In Vitro Networks. Front Neural Circuits 2016; 10:32. [PMID: 27147977 PMCID: PMC4840215 DOI: 10.3389/fncir.2016.00032] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2016] [Accepted: 04/07/2016] [Indexed: 12/28/2022] Open
Abstract
Transient propagation of information across neuronal assembles is thought to underlie many cognitive processes. However, the nature of the neural code that is embedded within these transmissions remains uncertain. Much of our understanding of how information is transmitted among these assemblies has been derived from computational models. While these models have been instrumental in understanding these processes they often make simplifying assumptions about the biophysical properties of neurons that may influence the nature and properties expressed. To address this issue we created an in vitro analog of a feed-forward network composed of two small populations (also referred to as assemblies or layers) of living dissociated rat cortical neurons. The populations were separated by, and communicated through, a microelectromechanical systems (MEMS) device containing a strip of microscale tunnels. Delayed culturing of one population in the first layer followed by the second a few days later induced the unidirectional growth of axons through the microtunnels resulting in a primarily feed-forward communication between these two small neural populations. In this study we systematically manipulated the number of tunnels that connected each layer and hence, the number of axons providing communication between those populations. We then assess the effect of reducing the number of tunnels has upon the properties of between-layer communication capacity and fidelity of neural transmission among spike trains transmitted across and within layers. We show evidence based on Victor-Purpura's and van Rossum's spike train similarity metrics supporting the presence of both rate and temporal information embedded within these transmissions whose fidelity increased during communication both between and within layers when the number of tunnels are increased. We also provide evidence reinforcing the role of synchronized activity upon transmission fidelity during the spontaneous synchronized network burst events that propagated between layers and highlight the potential applications of these MEMs devices as a tool for further investigation of structure and functional dynamics among neural populations.
Collapse
Affiliation(s)
- Thomas B DeMarse
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of FloridaGainesville, FL, USA; Department of Pediatric Neurology, University of FloridaGainesville, FL, USA
| | - Liangbin Pan
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida Gainesville, FL, USA
| | - Sankaraleengam Alagapan
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida Gainesville, FL, USA
| | - Gregory J Brewer
- Department of Bioengineering, University of California Irvine, CA, USA
| | - Bruce C Wheeler
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of FloridaGainesville, FL, USA; Department of Bioengineering, University of CaliforniaSan Diego, CA, USA
| |
Collapse
|
22
|
Alagapan S, Franca E, Pan L, Leondopulos S, Wheeler BC, DeMarse TB. Structure, Function, and Propagation of Information across Living Two, Four, and Eight Node Degree Topologies. Front Bioeng Biotechnol 2016; 4:15. [PMID: 26973833 PMCID: PMC4770194 DOI: 10.3389/fbioe.2016.00015] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2015] [Accepted: 02/04/2016] [Indexed: 11/13/2022] Open
Abstract
In this study, we created four network topologies composed of living cortical neurons and compared resultant structural-functional dynamics including the nature and quality of information transmission. Each living network was composed of living cortical neurons and were created using microstamping of adhesion promoting molecules and each was "designed" with different levels of convergence embedded within each structure. Networks were cultured over a grid of electrodes that permitted detailed measurements of neural activity at each node in the network. Of the topologies we tested, the "Random" networks in which neurons connect based on their own intrinsic properties transmitted information embedded within their spike trains with higher fidelity relative to any other topology we tested. Within our patterned topologies in which we explicitly manipulated structure, the effect of convergence on fidelity was dependent on both topology and time-scale (rate vs. temporal coding). A more detailed examination using tools from network analysis revealed that these changes in fidelity were also associated with a number of other structural properties including a node's degree, degree-degree correlations, path length, and clustering coefficients. Whereas information transmission was apparent among nodes with few connections, the greatest transmission fidelity was achieved among the few nodes possessing the highest number of connections (high degree nodes or putative hubs). These results provide a unique view into the relationship between structure and its affect on transmission fidelity, at least within these small neural populations with defined network topology. They also highlight the potential role of tools such as microstamp printing and microelectrode array recordings to construct and record from arbitrary network topologies to provide a new direction in which to advance the study of structure-function relationships.
Collapse
Affiliation(s)
- Sankaraleengam Alagapan
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida , Gainesville, FL , USA
| | - Eric Franca
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida , Gainesville, FL , USA
| | - Liangbin Pan
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida , Gainesville, FL , USA
| | - Stathis Leondopulos
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida , Gainesville, FL , USA
| | - Bruce C Wheeler
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL, USA; Department of Biomedical Engineering, University of California San Diego, San Diego, CA, USA
| | - Thomas B DeMarse
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL, USA; Department of Pediatric Neurology, University of Florida, Gainesville, FL, USA
| |
Collapse
|
23
|
Chever O, Dossi E, Pannasch U, Derangeon M, Rouach N. Astroglial networks promote neuronal coordination. Sci Signal 2016; 9:ra6. [DOI: 10.1126/scisignal.aad3066] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
|
24
|
Synaptic Depression Influences Inspiratory-Expiratory Phase Transition in Dbx1 Interneurons of the preBötzinger Complex in Neonatal Mice. J Neurosci 2015; 35:11606-11. [PMID: 26290237 DOI: 10.1523/jneurosci.0351-15.2015] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
UNLABELLED The brainstem preBötzinger complex (preBötC) generates the rhythm underlying inspiratory breathing movements and its core interneurons are derived from Dbx1-expressing precursors. Recurrent synaptic excitation is required to initiate inspiratory bursts, but whether excitatory synaptic mechanisms also contribute to inspiratory-expiratory phase transition is unknown. Here, we examined the role of short-term synaptic depression using a rhythmically active neonatal mouse brainstem slice preparation. We show that afferent axonal projections to Dbx1 preBötC neurons undergo activity-dependent depression and we identify a refractory period (∼2 s) after endogenous inspiratory bursts that precludes light-evoked bursts in channelrhodopsin-expressing Dbx1 preBötC neurons. We demonstrate that the duration of the refractory period-but neither the cycle period nor the magnitude of endogenous inspiratory bursts-is sensitive to changes in extracellular Ca(2+). Further, we show that postsynaptic factors are unlikely to explain the refractory period or its modulation by Ca(2+). Our findings are consistent with the hypothesis that short-term synaptic depression in Dbx1 preBötC neurons influences the inspiratory-expiratory phase transition during respiratory rhythmogenesis. SIGNIFICANCE STATEMENT Theories of breathing's neural origins have heretofore focused on intrinsically bursting "pacemaker" cells operating in conjunction with synaptic inhibition for phase transition and cycle timing. However, contemporary studies falsify an obligatory role for pacemaker-like neurons and synaptic inhibition, giving credence to burst-generating mechanisms based on recurrent excitation among glutamatergic interneurons of the respiratory kernel. Here, we investigated the role of short-term synaptic depression in inspiratory-expiratory phase transition. Until now, this role remained an untested prediction of mathematical models. The present data emphasize that synaptic properties of excitatory interneurons of the respiratory rhythmogenic kernel, derived from Dbx1-expressing precursors, may provide the core logic underlying the rhythm for breathing.
Collapse
|
25
|
Shaping Neuronal Network Activity by Presynaptic Mechanisms. PLoS Comput Biol 2015; 11:e1004438. [PMID: 26372048 PMCID: PMC4570815 DOI: 10.1371/journal.pcbi.1004438] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2015] [Accepted: 06/23/2015] [Indexed: 02/06/2023] Open
Abstract
Neuronal microcircuits generate oscillatory activity, which has been linked to basic functions such as sleep, learning and sensorimotor gating. Although synaptic release processes are well known for their ability to shape the interaction between neurons in microcircuits, most computational models do not simulate the synaptic transmission process directly and hence cannot explain how changes in synaptic parameters alter neuronal network activity. In this paper, we present a novel neuronal network model that incorporates presynaptic release mechanisms, such as vesicle pool dynamics and calcium-dependent release probability, to model the spontaneous activity of neuronal networks. The model, which is based on modified leaky integrate-and-fire neurons, generates spontaneous network activity patterns, which are similar to experimental data and robust under changes in the model's primary gain parameters such as excitatory postsynaptic potential and connectivity ratio. Furthermore, it reliably recreates experimental findings and provides mechanistic explanations for data obtained from microelectrode array recordings, such as network burst termination and the effects of pharmacological and genetic manipulations. The model demonstrates how elevated asynchronous release, but not spontaneous release, synchronizes neuronal network activity and reveals that asynchronous release enhances utilization of the recycling vesicle pool to induce the network effect. The model further predicts a positive correlation between vesicle priming at the single-neuron level and burst frequency at the network level; this prediction is supported by experimental findings. Thus, the model is utilized to reveal how synaptic release processes at the neuronal level govern activity patterns and synchronization at the network level. The activity of neuronal networks underlies basic neural functions such as sleep, learning and sensorimotor gating. Computational models of neuronal networks have been developed to capture the complexity of the network activity and predict how neuronal networks generate spontaneous activity. However, most computational models do not simulate the intricate synaptic release process that governs the interaction between neurons and has been shown to significantly impact neuronal network activity and animal behavior, learning and memory. Our paper demonstrates the importance of simulating the elaborate synaptic release process to understand how neuronal networks generate spontaneous activity and respond to manipulations of the release process. The model provides mechanistic explanations and predictions for experimental pharmacological and genetic manipulations. Thus, the model presents a novel computational platform to understand how mechanistic changes in the synaptic release process modulate network oscillatory activity that might impact basic neural functions.
Collapse
|
26
|
Bursting reverberation as a multiscale neuronal network process driven by synaptic depression-facilitation. PLoS One 2015; 10:e0124694. [PMID: 26017681 PMCID: PMC4446271 DOI: 10.1371/journal.pone.0124694] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2014] [Accepted: 03/06/2015] [Indexed: 11/19/2022] Open
Abstract
Neuronal networks can generate complex patterns of activity that depend on membrane properties of individual neurons as well as on functional synapses. To decipher the impact of synaptic properties and connectivity on neuronal network behavior, we investigate the responses of neuronal ensembles from small (5-30 cells in a restricted sphere) and large (acute hippocampal slice) networks to single electrical stimulation: in both cases, a single stimulus generated a synchronous long-lasting bursting activity. While an initial spike triggered a reverberating network activity that lasted 2-5 seconds for small networks, we found here that it lasted only up to 300 milliseconds in slices. To explain this phenomena present at different scales, we generalize the depression-facilitation model and extracted the network time constants. The model predicts that the reverberation time has a bell shaped relation with the synaptic density, revealing that the bursting time cannot exceed a maximum value. Furthermore, before reaching its maximum, the reverberation time increases sub-linearly with the synaptic density of the network. We conclude that synaptic dynamics and connectivity shape the mean burst duration, a property present at various scales of the networks. Thus bursting reverberation is a property of sufficiently connected neural networks, and can be generated by collective depression and facilitation of underlying functional synapses.
Collapse
|
27
|
Kaufman M, Reinartz S, Ziv NE. Adaptation to prolonged neuromodulation in cortical cultures: an invariable return to network synchrony. BMC Biol 2014; 12:83. [PMID: 25339462 PMCID: PMC4237737 DOI: 10.1186/s12915-014-0083-3] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2014] [Accepted: 09/29/2014] [Indexed: 11/17/2022] Open
Abstract
Background Prolonged neuromodulatory regimes, such as those critically involved in promoting arousal and suppressing sleep-associated synchronous activity patterns, might be expected to trigger adaptation processes and, consequently, a decline in neuromodulator-driven effects. This possibility, however, has rarely been addressed. Results Using networks of cultured cortical neurons, acetylcholine microinjections and a novel closed-loop ‘synchrony-clamp’ system, we found that acetylcholine pulses strongly suppressed network synchrony. Over the course of many hours, however, synchrony invariably reemerged, even when feedback was used to compensate for declining cholinergic efficacy. Network synchrony also reemerged following its initial suppression by noradrenaline, but this did not occlude the suppression of synchrony or its gradual reemergence following subsequent cholinergic input. Importantly, cholinergic efficacy could be restored and preserved over extended time scales by periodically withdrawing cholinergic input. Conclusions These findings indicate that the capacity of neuromodulators to suppress network synchrony is constrained by slow-acting, reactive processes. A multiplicity of neuromodulators and ultimately neuromodulator withdrawal periods might thus be necessary to cope with an inevitable reemergence of network synchrony. Electronic supplementary material The online version of this article (doi:10.1186/s12915-014-0083-3) contains supplementary material, which is available to authorized users.
Collapse
|
28
|
Orlandi JG, Stetter O, Soriano J, Geisel T, Battaglia D. Transfer entropy reconstruction and labeling of neuronal connections from simulated calcium imaging. PLoS One 2014; 9:e98842. [PMID: 24905689 PMCID: PMC4048312 DOI: 10.1371/journal.pone.0098842] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2013] [Accepted: 05/08/2014] [Indexed: 11/23/2022] Open
Abstract
Neuronal dynamics are fundamentally constrained by the underlying structural network architecture, yet much of the details of this synaptic connectivity are still unknown even in neuronal cultures in vitro. Here we extend a previous approach based on information theory, the Generalized Transfer Entropy, to the reconstruction of connectivity of simulated neuronal networks of both excitatory and inhibitory neurons. We show that, due to the model-free nature of the developed measure, both kinds of connections can be reliably inferred if the average firing rate between synchronous burst events exceeds a small minimum frequency. Furthermore, we suggest, based on systematic simulations, that even lower spontaneous inter-burst rates could be raised to meet the requirements of our reconstruction algorithm by applying a weak spatially homogeneous stimulation to the entire network. By combining multiple recordings of the same in silico network before and after pharmacologically blocking inhibitory synaptic transmission, we show then how it becomes possible to infer with high confidence the excitatory or inhibitory nature of each individual neuron.
Collapse
Affiliation(s)
- Javier G. Orlandi
- Departament d'Estructura i Consituents de la Matèria, Universitat de Barcelona, Barcelona, Spain
| | - Olav Stetter
- Max Planck Institute for Dynamics and Self-Organization, Göttingen, Germany
- Georg-August-Universität, Physics Department, Göttingen, Germany
- Bernstein Center for Computational Neuroscience, Göttingen, Germany
| | - Jordi Soriano
- Departament d'Estructura i Consituents de la Matèria, Universitat de Barcelona, Barcelona, Spain
| | - Theo Geisel
- Max Planck Institute for Dynamics and Self-Organization, Göttingen, Germany
- Georg-August-Universität, Physics Department, Göttingen, Germany
- Bernstein Center for Computational Neuroscience, Göttingen, Germany
| | - Demian Battaglia
- Max Planck Institute for Dynamics and Self-Organization, Göttingen, Germany
- Bernstein Center for Computational Neuroscience, Göttingen, Germany
- Institut de Neurosciences des Systèmes, Inserm UMR1106, Aix-Marseille Université, Marseille, France
- * E-mail:
| |
Collapse
|
29
|
Investigation of synapse formation and function in a glutamatergic-GABAergic two-neuron microcircuit. J Neurosci 2014; 34:855-68. [PMID: 24431444 DOI: 10.1523/jneurosci.0229-13.2014] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
Neural circuits are composed of mainly glutamatergic and GABAergic neurons, which communicate through synaptic connections. Many factors instruct the formation and function of these synapses; however, it is difficult to dissect the contribution of intrinsic cell programs from that of extrinsic environmental effects in an intact network. Here, we perform paired recordings from two-neuron microculture preparations of mouse hippocampal glutamatergic and GABAergic neurons to investigate how synaptic input and output of these two principal cells develop. In our reduced preparation, we found that glutamatergic neurons showed no change in synaptic output or input regardless of partner neuron cell type or neuronal activity level. In contrast, we found that glutamatergic input caused the GABAergic neuron to modify its output by way of an increase in synapse formation and a decrease in synaptic release efficiency. These findings are consistent with aspects of GABAergic synapse maturation observed in many brain regions. In addition, changes in GABAergic output are cell wide and not target-cell specific. We also found that glutamatergic neuronal activity determined the AMPA receptor properties of synapses on the partner GABAergic neuron. All modifications of GABAergic input and output required activity of the glutamatergic neuron. Because our system has reduced extrinsic factors, the changes we saw in the GABAergic neuron due to glutamatergic input may reflect initiation of maturation programs that underlie the formation and function of in vivo neural circuits.
Collapse
|
30
|
Tibau E, Valencia M, Soriano J. Identification of neuronal network properties from the spectral analysis of calcium imaging signals in neuronal cultures. Front Neural Circuits 2013; 7:199. [PMID: 24385953 PMCID: PMC3866384 DOI: 10.3389/fncir.2013.00199] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2013] [Accepted: 12/01/2013] [Indexed: 11/13/2022] Open
Abstract
Neuronal networks in vitro are prominent systems to study the development of connections in living neuronal networks and the interplay between connectivity, activity and function. These cultured networks show a rich spontaneous activity that evolves concurrently with the connectivity of the underlying network. In this work we monitor the development of neuronal cultures, and record their activity using calcium fluorescence imaging. We use spectral analysis to characterize global dynamical and structural traits of the neuronal cultures. We first observe that the power spectrum can be used as a signature of the state of the network, for instance when inhibition is active or silent, as well as a measure of the network's connectivity strength. Second, the power spectrum identifies prominent developmental changes in the network such as GABAA switch. And third, the analysis of the spatial distribution of the spectral density, in experiments with a controlled disintegration of the network through CNQX, an AMPA-glutamate receptor antagonist in excitatory neurons, reveals the existence of communities of strongly connected, highly active neurons that display synchronous oscillations. Our work illustrates the interest of spectral analysis for the study of in vitro networks, and its potential use as a network-state indicator, for instance to compare healthy and diseased neuronal networks.
Collapse
Affiliation(s)
- Elisenda Tibau
- Neurophysics Laboratory, Departament d'Estructura i Constituents de la Matèria, Universitat de Barcelona Barcelona, Spain
| | - Miguel Valencia
- Neurophysiology Laboratory, Division of Neurosciences, CIMA, Universidad de Navarra Pamplona, Spain
| | - Jordi Soriano
- Neurophysics Laboratory, Departament d'Estructura i Constituents de la Matèria, Universitat de Barcelona Barcelona, Spain
| |
Collapse
|
31
|
El Hady A, Afshar G, Bröking K, Schlüter OM, Geisel T, Stühmer W, Wolf F. Optogenetic stimulation effectively enhances intrinsically generated network synchrony. Front Neural Circuits 2013; 7:167. [PMID: 24155695 PMCID: PMC3805139 DOI: 10.3389/fncir.2013.00167] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2013] [Accepted: 09/24/2013] [Indexed: 11/20/2022] Open
Abstract
Synchronized bursting is found in many brain areas and has also been implicated in the pathophysiology of neuropsychiatric disorders such as epilepsy, Parkinson’s disease, and schizophrenia. Despite extensive studies of network burst synchronization, it is insufficiently understood how this type of network wide synchronization can be strengthened, reduced, or even abolished. We combined electrical recording using multi-electrode array with optical stimulation of cultured channelrhodopsin-2 transducted hippocampal neurons to study and manipulate network burst synchronization. We found low frequency photo-stimulation protocols that are sufficient to induce potentiation of network bursting, modifying bursting dynamics, and increasing interneuronal synchronization. Surprisingly, slowly fading-in light stimulation, which substantially delayed and reduced light-driven spiking, was at least as effective in reorganizing network dynamics as much stronger pulsed light stimulation. Our study shows that mild stimulation protocols that do not enforce particular activity patterns onto the network can be highly effective inducers of network-level plasticity.
Collapse
Affiliation(s)
- Ahmed El Hady
- Theoretical Neurophysics, Department of Non-linear Dynamics, Max Planck Institute for Dynamics and Self-Organization Göttingen, Germany ; Max Planck Institute of Experimental Medicine Göttingen, Germany ; Bernstein Focus for Neurotechnology Göttingen, Germany ; Bernstein Center for Computational Neuroscience Göttingen, Germany ; The Interdisciplinary Collaborative Research Center 889 "Cellular Mechanisms of Sensory Processing" Göttingen, Germany
| | | | | | | | | | | | | |
Collapse
|
32
|
Mendoza-Torreblanca JG, Vanoye-Carlo A, Phillips-Farfán BV, Carmona-Aparicio L, Gómez-Lira G. Synaptic vesicle protein 2A: basic facts and role in synaptic function. Eur J Neurosci 2013; 38:3529-39. [DOI: 10.1111/ejn.12360] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2013] [Revised: 08/09/2013] [Accepted: 08/17/2013] [Indexed: 10/26/2022]
Affiliation(s)
| | | | | | | | - Gisela Gómez-Lira
- Department of Pharmacobiology; Centro de Investigación y Estudios Avanzados del Instituto Politécnico Nacional; Calzada de los Tenorios 235 Col. Granjas Coapa C.P. 14330 D. F., Mexico
| |
Collapse
|
33
|
Nimmervoll B, Flucher BE, Obermair GJ. Dominance of P/Q-type calcium channels in depolarization-induced presynaptic FM dye release in cultured hippocampal neurons. Neuroscience 2013; 253:330-40. [PMID: 24012836 PMCID: PMC3824072 DOI: 10.1016/j.neuroscience.2013.08.052] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2013] [Revised: 08/09/2013] [Accepted: 08/27/2013] [Indexed: 11/26/2022]
Abstract
We analyzed depolarization-induced synaptic FM dye release in hippocampal neurons. We pharmacologically isolated the contribution of voltage-gated Ca2+ channels. 85% of synapses utilize N- and P/Q-type channels, 15% only P/Q-type channels. In both groups of synapses release kinetics are determined by P/Q-type channels. We propose a more direct coupling of P/Q-type channels to synaptic release.
Neurotransmitter release probability is related by high power to the local concentration of calcium in presynaptic terminals, which in turn is controlled by voltage-gated calcium channels. P/Q- and N-type channels trigger synaptic transmission in the majority of neurons of the central nervous system. However, whether and under which conditions both channel types act cooperatively or independently is still insufficiently understood. Previous studies suggested either a dominance of N- or P/Q-type channels, or a synergistic action of both channels, depending on the experimental paradigms. Thus, to provide insight into the properties of neurotransmitter release in cultured mouse hippocampal neurons, we used quantitative analysis of FM dye release from presynaptic boutons induced by high potassium membrane depolarization. Increasing extracellular potassium concentrations revealed a sigmoid dependence of FM dye release to the stimulation strength. Individual and combined application of the P/Q- and N-type channel-specific blockers ω-agatoxin-IVA and ω-conotoxin-GVIA, respectively, allowed us to specifically isolate the contribution of both channel types to release triggered with 40 mM KCl. Analysis of the release kinetics and the fractional release amplitude demonstrate that, whereas in only 15% of the synapses release depended exclusively on P/Q-type channels, the majority of synapses (85%) contained both N- and P/Q-type channels. Nevertheless, the kinetics of FM dye release in synapses containing both channel types was determined by the P/Q-type channels. Together, our data suggest a more direct coupling of P/Q-type channels to synaptic release compared to N-type channels, which may explain the high prevalence of neurological P/Q-type channelopathies.
Collapse
Affiliation(s)
- B Nimmervoll
- Division of Physiology, Medical University Innsbruck, Fritz-Pregl-Str. 3, 6020 Innsbruck, Austria
| | | | | |
Collapse
|
34
|
Weihberger O, Okujeni S, Mikkonen JE, Egert U. Quantitative examination of stimulus-response relations in cortical networks in vitro. J Neurophysiol 2013; 109:1764-74. [DOI: 10.1152/jn.00481.2012] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Variable responses of neuronal networks to repeated sensory or electrical stimuli reflect the interaction of the stimulus' response with ongoing activity in the brain and its modulation by adaptive mechanisms, such as cognitive context, network state, or cellular excitability and synaptic transmission capability. Here, we focus on reliability, length, delays, and variability of evoked responses with respect to their spatial distribution, interaction with spontaneous activity in the networks, and the contribution of GABAergic inhibition. We identified network-intrinsic principles that underlie the formation and modulation of spontaneous activity and stimulus-response relations with the use of state-dependent stimulation in generic neuronal networks in vitro. The duration of spontaneously recurring network-wide bursts of spikes was best predicted by the length of the preceding interval. Length, delay, and structure of responses to identical stimuli systematically depended on stimulus timing and distance to the stimulation site, which were described by a set of simple functions of spontaneous activity. Response length at proximal recording sites increased with the duration of prestimulus inactivity and was best described by a saturation function y( t) = A( 1 − e−α t). Concomitantly, the delays of polysynaptic late responses at distant sites followed an exponential decay y( t) = Be−β t + C. In addition, the speed of propagation was determined by the overall state of the network at the moment of stimulation. Disinhibition increased the number of spikes/network burst and interburst interval length at unchanged gross firing rate, whereas the response modulation by the duration of prestimulus inactivity was preserved. Our data suggest a process of network depression during bursts and subsequent recovery that limit evoked responses following distinct rules. We discuss short-term synaptic depression due to depletion of neurotransmitter vesicles as an underlying mechanism. The seemingly unreliable patterns of spontaneous activity and stimulus-response relations thus follow a predictable structure determined by the interdependencies of network structures and activity states.
Collapse
Affiliation(s)
- Oliver Weihberger
- Bernstein Center Freiburg, University of Freiburg, Freiburg, Germany
- Neurobiology and Biophysics, Faculty of Biology, University of Freiburg, Freiburg, Germany; and
- Department of Microsystems Engineering–IMTEK, University of Freiburg, Freiburg, Germany
| | - Samora Okujeni
- Bernstein Center Freiburg, University of Freiburg, Freiburg, Germany
- Neurobiology and Biophysics, Faculty of Biology, University of Freiburg, Freiburg, Germany; and
- Department of Microsystems Engineering–IMTEK, University of Freiburg, Freiburg, Germany
| | - Jarno E. Mikkonen
- Bernstein Center Freiburg, University of Freiburg, Freiburg, Germany
| | - Ulrich Egert
- Bernstein Center Freiburg, University of Freiburg, Freiburg, Germany
- Department of Microsystems Engineering–IMTEK, University of Freiburg, Freiburg, Germany
| |
Collapse
|
35
|
Lavi A, Sheinin A, Shapira R, Zelmanoff D, Ashery U. DOC2B and Munc13-1 differentially regulate neuronal network activity. ACTA ACUST UNITED AC 2013; 24:2309-23. [PMID: 23537531 DOI: 10.1093/cercor/bht081] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Alterations in the levels of synaptic proteins affect synaptic transmission and synaptic plasticity. However, the precise effects on neuronal network activity are still enigmatic. Here, we utilized microelectrode array (MEA) to elucidate how manipulation of the presynaptic release process affects the activity of neuronal networks. By combining pharmacological tools and genetic manipulation of synaptic proteins, we show that overexpression of DOC2B and Munc13-1, proteins known to promote vesicular maturation and release, elicits opposite effects on the activity of the neuronal network. Although both cause an increase in the overall number of spikes, the distribution of spikes is different. While DOC2B enhances, Munc13-1 reduces the firing rate within bursts of spikes throughout the network; however, Munc13-1 increases the rate of network bursts. DOC2B's effects were mimicked by Strontium that elevates asynchronous release but not by a DOC2B mutant that enhances spontaneous release rate. This suggests for the first time that increased asynchronous release on the single-neuron level promotes bursting activity in the network level. This innovative study demonstrates the complementary role of the network level in explaining the physiological relevance of the cellular activity of presynaptic proteins and the transformation of synaptic release manipulation from the neuron to the network level.
Collapse
Affiliation(s)
- Ayal Lavi
- Department of Neurobiology, The George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv 69978, Israel Sagol School of Neuroscience, Tel Aviv University, Tel Aviv 69978, Israel
| | - Anton Sheinin
- Department of Neurobiology, The George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv 69978, Israel Sagol School of Neuroscience, Tel Aviv University, Tel Aviv 69978, Israel
| | - Ronit Shapira
- Department of Neurobiology, The George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv 69978, Israel
| | - Daniel Zelmanoff
- Department of Neurobiology, The George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv 69978, Israel
| | - Uri Ashery
- Department of Neurobiology, The George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv 69978, Israel Sagol School of Neuroscience, Tel Aviv University, Tel Aviv 69978, Israel
| |
Collapse
|
36
|
Stetter O, Battaglia D, Soriano J, Geisel T. Model-free reconstruction of excitatory neuronal connectivity from calcium imaging signals. PLoS Comput Biol 2012; 8:e1002653. [PMID: 22927808 PMCID: PMC3426566 DOI: 10.1371/journal.pcbi.1002653] [Citation(s) in RCA: 145] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2012] [Accepted: 07/01/2012] [Indexed: 12/13/2022] Open
Abstract
A systematic assessment of global neural network connectivity through direct electrophysiological assays has remained technically infeasible, even in simpler systems like dissociated neuronal cultures. We introduce an improved algorithmic approach based on Transfer Entropy to reconstruct structural connectivity from network activity monitored through calcium imaging. We focus in this study on the inference of excitatory synaptic links. Based on information theory, our method requires no prior assumptions on the statistics of neuronal firing and neuronal connections. The performance of our algorithm is benchmarked on surrogate time series of calcium fluorescence generated by the simulated dynamics of a network with known ground-truth topology. We find that the functional network topology revealed by Transfer Entropy depends qualitatively on the time-dependent dynamic state of the network (bursting or non-bursting). Thus by conditioning with respect to the global mean activity, we improve the performance of our method. This allows us to focus the analysis to specific dynamical regimes of the network in which the inferred functional connectivity is shaped by monosynaptic excitatory connections, rather than by collective synchrony. Our method can discriminate between actual causal influences between neurons and spurious non-causal correlations due to light scattering artifacts, which inherently affect the quality of fluorescence imaging. Compared to other reconstruction strategies such as cross-correlation or Granger Causality methods, our method based on improved Transfer Entropy is remarkably more accurate. In particular, it provides a good estimation of the excitatory network clustering coefficient, allowing for discrimination between weakly and strongly clustered topologies. Finally, we demonstrate the applicability of our method to analyses of real recordings of in vitro disinhibited cortical cultures where we suggest that excitatory connections are characterized by an elevated level of clustering compared to a random graph (although not extreme) and can be markedly non-local. Unraveling the general organizing principles of connectivity in neural circuits is a crucial step towards understanding brain function. However, even the simpler task of assessing the global excitatory connectivity of a culture in vitro, where neurons form self-organized networks in absence of external stimuli, remains challenging. Neuronal cultures undergo spontaneous switching between episodes of synchronous bursting and quieter inter-burst periods. We introduce here a novel algorithm which aims at inferring the connectivity of neuronal cultures from calcium fluorescence recordings of their network dynamics. To achieve this goal, we develop a suitable generalization of Transfer Entropy, an information-theoretic measure of causal influences between time series. Unlike previous algorithmic approaches to reconstruction, Transfer Entropy is data-driven and does not rely on specific assumptions about neuronal firing statistics or network topology. We generate simulated calcium signals from networks with controlled ground-truth topology and purely excitatory interactions and show that, by restricting the analysis to inter-bursts periods, Transfer Entropy robustly achieves a good reconstruction performance for disparate network connectivities. Finally, we apply our method to real data and find evidence of non-random features in cultured networks, such as the existence of highly connected hub excitatory neurons and of an elevated (but not extreme) level of clustering.
Collapse
Affiliation(s)
- Olav Stetter
- Max Planck Institute for Dynamics and Self-Organization, Göttingen, Germany
- Georg August University, Physics Department, Göttingen, Germany
- Bernstein Center for Computational Neuroscience, Göttingen, Germany
| | - Demian Battaglia
- Max Planck Institute for Dynamics and Self-Organization, Göttingen, Germany
- Bernstein Center for Computational Neuroscience, Göttingen, Germany
- * E-mail:
| | - Jordi Soriano
- Departament d'ECM , Facultat de F?sica, Universitat de Barcelona, Barcelona, Spain
| | - Theo Geisel
- Max Planck Institute for Dynamics and Self-Organization, Göttingen, Germany
- Georg August University, Physics Department, Göttingen, Germany
- Bernstein Center for Computational Neuroscience, Göttingen, Germany
| |
Collapse
|
37
|
Niedringhaus M, Chen X, Dzakpasu R, Conant K. MMPs and soluble ICAM-5 increase neuronal excitability within in vitro networks of hippocampal neurons. PLoS One 2012; 7:e42631. [PMID: 22912716 PMCID: PMC3418258 DOI: 10.1371/journal.pone.0042631] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2012] [Accepted: 07/09/2012] [Indexed: 12/16/2022] Open
Abstract
Matrix metalloproteinases (MMPs) are zinc-dependent endopeptidases that are released from neurons in an activity dependent manner. Published studies suggest their activity is important to varied forms of learning and memory. At least one MMP can stimulate an increase in the size of dendritic spines, structures which represent the post synaptic component for a large number of glutamatergic synapses. This change may be associated with increased synaptic glutamate receptor incorporation, and an increased amplitude and/or frequency of α-amino-3-hydroxyl-5-methyl-4-isoxazole-propionate (AMPA) mini excitatory post-synaptic currents (EPSCs). An associated increase in the probability of action potential occurrence would be expected. While the mechanism(s) by which MMPs may influence synaptic structure and function are not completely understood, MMP dependent shedding of specific cell adhesion molecules (CAMs) could play an important role. CAMs are ideally positioned to be cleaved by synaptically released MMPs, and shed N terminal domains could potentially interact with previously unengaged integrins to stimulate dendritic actin polymerization with spine expansion. In the present study, we have used multielectrode arrays (MEAs) to investigate MMP and soluble CAM dependent changes in neuronal activity recorded from hippocampal cultures. We have focused on intercellular adhesion molecule-5 (ICAM-5) in particular, as this CAM is expressed on glutamatergic dendrites and shed in an MMP dependent manner. We show that chemical long-term potentiation (cLTP) evoked changes in recorded activity, and the dynamics of action potential bursts in particular, are altered by MMP inhibition. A blocking antibody to β1 integrins has a similar effect. We also show that the ectodomain of ICAM-5 can stimulate β1 integrin dependent increases in spike counts and burst number. These results support a growing body of literature suggesting that MMPs have important effects on neuronal excitability. They also support the possibility that MMP dependent shedding of specific synaptic CAMs can contribute to these effects.
Collapse
Affiliation(s)
- Mark Niedringhaus
- Interdisciplinary Program in Neuroscience, Georgetown University Medical Center, Washington, District of Columbia, United States of America
| | - Xin Chen
- Department of Physics, Georgetown University, Washington, District of Columbia, United States of America
| | - Rhonda Dzakpasu
- Interdisciplinary Program in Neuroscience, Georgetown University Medical Center, Washington, District of Columbia, United States of America
- Department of Physics, Georgetown University, Washington, District of Columbia, United States of America
- Department of Pharmacology and Physiology, Georgetown University Medical Center, Washington, District of Columbia, United States of America
- * E-mail: (KC); (RD)
| | - Katherine Conant
- Interdisciplinary Program in Neuroscience, Georgetown University Medical Center, Washington, District of Columbia, United States of America
- Department of Neuroscience, Georgetown University Medical Center, Washington, District of Columbia, United States of America
- * E-mail: (KC); (RD)
| |
Collapse
|
38
|
Gavello D, Rojo-Ruiz J, Marcantoni A, Franchino C, Carbone E, Carabelli V. Leptin counteracts the hypoxia-induced inhibition of spontaneously firing hippocampal neurons: a microelectrode array study. PLoS One 2012; 7:e41530. [PMID: 22848520 PMCID: PMC3405131 DOI: 10.1371/journal.pone.0041530] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2012] [Accepted: 06/26/2012] [Indexed: 01/24/2023] Open
Abstract
Besides regulating energy balance and reducing body-weight, the adipokine leptin has been recently shown to be neuroprotective and antiapoptotic by promoting neuronal survival after excitotoxic and oxidative insults. Here, we investigated the firing properties of mouse hippocampal neurons and the effects of leptin pretreatment on hypoxic damage (2 hours, 3% O(2)). Experiments were carried out by means of the microelectrode array (MEA) technology, monitoring hippocampal neurons activity from 11 to 18 days in vitro (DIV). Under normoxic conditions, hippocampal neurons were spontaneously firing, either with prevailing isolated and randomly distributed spikes (11 DIV), or with patterns characterized by synchronized bursts (18 DIV). Exposure to hypoxia severely impaired the spontaneous activity of hippocampal neurons, reducing their firing frequency by 54% and 69%, at 11 and 18 DIV respectively, and synchronized their firing activity. Pretreatment with 50 nM leptin reduced the firing frequency of normoxic neurons and contrasted the hypoxia-induced depressive action, either by limiting the firing frequency reduction (at both ages) or by increasing it to 126% (in younger neurons). In order to find out whether leptin exerts its effect by activating large conductance Ca(2+)-activated K(+) channels (BK), as shown on rat hippocampal neurons, we applied the BK channel blocker paxilline (1 µM). Our data show that paxilline reversed the effects of leptin, both on normoxic and hypoxic neurons, suggesting that the adipokine counteracts hypoxia through BK channels activation in mouse hippocampal neurons.
Collapse
Affiliation(s)
- Daniela Gavello
- Department of Drug Science and Technology, NIS Center, CNISM, University of Torino, Torino, Italy
| | - Jonathan Rojo-Ruiz
- Department of Drug Science and Technology, NIS Center, CNISM, University of Torino, Torino, Italy
| | - Andrea Marcantoni
- Department of Drug Science and Technology, NIS Center, CNISM, University of Torino, Torino, Italy
| | - Claudio Franchino
- Department of Drug Science and Technology, NIS Center, CNISM, University of Torino, Torino, Italy
| | - Emilio Carbone
- Department of Drug Science and Technology, NIS Center, CNISM, University of Torino, Torino, Italy
| | - Valentina Carabelli
- Department of Drug Science and Technology, NIS Center, CNISM, University of Torino, Torino, Italy
- * E-mail:
| |
Collapse
|