1
|
Selten M, Bernard C, Mukherjee D, Hamid F, Hanusz-Godoy A, Oozeer F, Zimmer C, Marín O. Regulation of PV interneuron plasticity by neuropeptide-encoding genes. Nature 2025:10.1038/s41586-025-08933-z. [PMID: 40307547 DOI: 10.1038/s41586-025-08933-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Accepted: 03/24/2025] [Indexed: 05/02/2025]
Abstract
Neuronal activity must be regulated in a narrow permissive band for the proper operation of neural networks. Changes in synaptic connectivity and network activity-for example, during learning-might disturb this balance, eliciting compensatory mechanisms to maintain network function1-3. In the neocortex, excitatory pyramidal cells and inhibitory interneurons exhibit robust forms of stabilizing plasticity. However, although neuronal plasticity has been thoroughly studied in pyramidal cells4-8, little is known about how interneurons adapt to persistent changes in their activity. Here we describe a critical cellular process through which cortical parvalbumin-expressing (PV+) interneurons adapt to changes in their activity levels. We found that changes in the activity of individual PV+ interneurons drive bidirectional compensatory adjustments of the number and strength of inhibitory synapses received by these cells, specifically from other PV+ interneurons. High-throughput profiling of ribosome-associated mRNA revealed that increasing the activity of a PV+ interneuron leads to upregulation of two genes encoding multiple secreted neuropeptides: Vgf and Scg2. Functional experiments demonstrated that VGF is critically required for the activity-dependent scaling of inhibitory PV+ synapses onto PV+ interneurons. Our findings reveal an instructive role for neuropeptide-encoding genes in regulating synaptic connections among PV+ interneurons in the adult mouse neocortex.
Collapse
Affiliation(s)
- Martijn Selten
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
- Medical Research Council Centre for Neurodevelopmental Disorders, King's College London, London, UK
| | - Clémence Bernard
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
- Medical Research Council Centre for Neurodevelopmental Disorders, King's College London, London, UK
- Department of Clinical and Biomedical Sciences, Faculty of Health and Life Sciences, University of Exeter, Exeter, UK
| | - Diptendu Mukherjee
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
- Medical Research Council Centre for Neurodevelopmental Disorders, King's College London, London, UK
| | - Fursham Hamid
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
- Medical Research Council Centre for Neurodevelopmental Disorders, King's College London, London, UK
| | - Alicia Hanusz-Godoy
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
- Medical Research Council Centre for Neurodevelopmental Disorders, King's College London, London, UK
| | - Fazal Oozeer
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
- Medical Research Council Centre for Neurodevelopmental Disorders, King's College London, London, UK
| | - Christoph Zimmer
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
- Medical Research Council Centre for Neurodevelopmental Disorders, King's College London, London, UK
| | - Oscar Marín
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK.
- Medical Research Council Centre for Neurodevelopmental Disorders, King's College London, London, UK.
| |
Collapse
|
2
|
Park E, Kuljis DA, Swindell RA, Ray A, Zhu M, Christian JA, Barth AL. Somatostatin neurons detect stimulus-reward contingencies to reduce neocortical inhibition during learning. Cell Rep 2025; 44:115606. [PMID: 40257862 DOI: 10.1016/j.celrep.2025.115606] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Revised: 12/02/2024] [Accepted: 04/01/2025] [Indexed: 04/23/2025] Open
Abstract
Learning involves the association of discrete events in the world to infer causality, likely through a cascade of changes at input- and target-specific synapses. Transient or sustained disinhibition may initiate cortical circuit plasticity important for association learning, but the cellular networks involved have not been well defined. Using recordings in acute brain slices, we show that whisker-dependent sensory association learning drives a durable, target-specific reduction in inhibition from somatostatin (SST)-expressing GABAergic neurons onto pyramidal (Pyr) neurons in superficial but not deep layers of mouse somatosensory cortex. Critically, SST output was not altered when stimuli and rewards were unpaired, indicating that these neurons are sensitive to stimulus-reward contingency. Depression of SST output onto Pyr neurons could be phenocopied by chemogenetic suppression of SST activity outside of the training context. Thus, neocortical SST neuron output can undergo long-lasting modifications to selectively disinhibit superficial layers of sensory neocortex during learning.
Collapse
Affiliation(s)
- Eunsol Park
- Department of Biological Sciences, Carnegie Mellon University, 4400 Fifth Avenue, Pittsburgh, PA 15232, USA
| | - Dika A Kuljis
- Department of Biological Sciences, Carnegie Mellon University, 4400 Fifth Avenue, Pittsburgh, PA 15232, USA
| | - Rachel A Swindell
- Department of Biological Sciences, Carnegie Mellon University, 4400 Fifth Avenue, Pittsburgh, PA 15232, USA
| | - Ajit Ray
- Department of Biological Sciences, Carnegie Mellon University, 4400 Fifth Avenue, Pittsburgh, PA 15232, USA
| | - Mo Zhu
- Department of Biological Sciences, Carnegie Mellon University, 4400 Fifth Avenue, Pittsburgh, PA 15232, USA
| | - Joseph A Christian
- Department of Biological Sciences, Carnegie Mellon University, 4400 Fifth Avenue, Pittsburgh, PA 15232, USA
| | - Alison L Barth
- Department of Biological Sciences, Carnegie Mellon University, 4400 Fifth Avenue, Pittsburgh, PA 15232, USA.
| |
Collapse
|
3
|
Saenkham-Huntsinger P, Drelich AK, Huang P, Peng BH, Tseng CTK. BALB/c mice challenged with SARS-CoV-2 B.1.351 β variant cause pathophysiological and neurological changes within the lungs and brains. J Gen Virol 2024; 105:002039. [PMID: 39475775 PMCID: PMC11524415 DOI: 10.1099/jgv.0.002039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Accepted: 10/08/2024] [Indexed: 11/02/2024] Open
Abstract
Up to one-third of individuals suffering from acute SARS-CoV-2 infection with the onset of severe-to-mild diseases could develop several symptoms of neurological disorders, which could last long after resolving the infection, known as neuro-COVID. Effective therapeutic treatments for neuro-COVID remain unavailable, in part, due to the absence of animal models for studying its underlying mechanisms and developing medical countermeasures against it. Here, we explored the impact of SARS-CoV-2 infection on the well-being of respiratory and neurological functions of BALB/c mice by using a clinical isolate of β-variant, i.e. B.1.351. We found that this β-variant of SARS-CoV-2 primarily infected the lungs, causing tissue damage, profound inflammatory responses, altered respiratory functions and transient but significant hypoxia. Although live progeny viruses could not be isolated, viral RNAs were detected across many anatomical regions of the brains in most challenged mice and triggered activation of genes encoding for NF-kB, IL-6, IP-10 and RANTES and microglial cells. We noted that the significantly activated IL-6-encoded gene persisted at 4 weeks after infection. Together, these results suggest that this B.1.351/BALB/c model of SARS-CoV-2 infection warrants further studies to establish it as a desirable model for studies of neuropathogenesis and the development of effective therapeutics of neuro-COVID.
Collapse
Affiliation(s)
| | - Aleksandra K. Drelich
- Departments of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, USA
| | - Pinghan Huang
- Departments of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, USA
| | - Bi-Hung Peng
- Neurobiology, University of Texas Medical Branch, Galveston, TX, USA
| | - Chien-Te K. Tseng
- Departments of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, USA
- Neurobiology, University of Texas Medical Branch, Galveston, TX, USA
| |
Collapse
|
4
|
Ray A, Loghinov I, Ravindranath V, Barth AL. Early hippocampal hyperexcitability and synaptic reorganization in mouse models of amyloidosis. iScience 2024; 27:110629. [PMID: 39262788 PMCID: PMC11388185 DOI: 10.1016/j.isci.2024.110629] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 05/09/2024] [Accepted: 07/29/2024] [Indexed: 09/13/2024] Open
Abstract
The limited success of plaque-reducing therapies in Alzheimer's disease suggests that early treatment might be more effective in delaying or reversing memory impairments. Toward this end, it is important to establish the progression of synaptic and circuit changes before onset of plaques or cognitive deficits. Here, we used quantitative, fluorescence-based methods for synapse detection in CA1 pyramidal neurons to investigate the interaction between abnormal circuit activity, measured by Fos-immunoreactivity, and synapse reorganization in mouse models of amyloidosis. Using a genetically encoded, fluorescently labeled synaptic marker in juvenile mice (prior to sexual maturity), we find both synapse gain and loss depending on dendritic location. This progresses to broad synapse loss in aged mice. Elevated hippocampal activity in both CA3 and CA1 was present at weaning and preceded this reorganization. Thus, Aβ overproduction may initiate abnormal activity and subsequent input-specific synapse plasticity. These findings indicate that sustained amyloidosis drives heterogeneous and progressive circuit-wide abnormalities.
Collapse
Affiliation(s)
- Ajit Ray
- Department of Biological Sciences, Carnegie Mellon University, Pittsburgh, PA 15213, USA
| | - Iulia Loghinov
- Department of Biological Sciences, Carnegie Mellon University, Pittsburgh, PA 15213, USA
| | - Vijayalakshmi Ravindranath
- Centre for Neuroscience, Indian Institute of Science, Bengaluru, Karnataka 560012, India
- Centre for Brain Research, Indian Institute of Science, Bengaluru, Karnataka 560012, India
| | - Alison L. Barth
- Department of Biological Sciences, Carnegie Mellon University, Pittsburgh, PA 15213, USA
| |
Collapse
|
5
|
Yoshioka M, Takahashi M, Kershaw J, Handa M, Takada A, Takuwa H. Two-photon optogenetics-based assessment of neuronal connectivity in healthy and chronic hypoperfusion mice. NEUROPHOTONICS 2024; 11:035009. [PMID: 39345733 PMCID: PMC11436461 DOI: 10.1117/1.nph.11.3.035009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Revised: 08/16/2024] [Accepted: 08/22/2024] [Indexed: 10/01/2024]
Abstract
Significance Two-photon optogenetics and simultaneous calcium imaging can be used to visualize the response of surrounding neurons with respect to the activity of an optically stimulated target neuron, providing a direct method to assess neuronal connectivity. Aim We aim to develop a two-photon optogenetics-based method for evaluating neuronal connectivity, compare it to the existing indirect resting-state synchrony method, and investigate the application of the method to brain pathophysiology. Approach C1V1-mScarlet was introduced into GCaMP6s-expressing transgenic mice with an adeno-associated virus. Optical stimulation of a single target neuron and simultaneous calcium imaging of the target and surrounding cells were performed. Neuronal connectivity was evaluated from the correlation between the fluorescence intensity of the target and surrounding cells. Results The neuronal connectivity in the living brain was evaluated using two-photon optogenetics. However, resting-state synchrony was not always consistent with two-photon optogenetics-based connectivity. Comparison with neuronal synchrony measured during sensory stimulation suggested that the disagreement was due to external sensory input. Two-photon optogenetics-based connectivity significantly decreased in the common carotid artery occlusion model, whereas there was no significant change in the control group. Conclusions We successfully developed a direct method to evaluate neuronal connectivity in the living brain using two-photon optogenetics. The technique was successful in detecting connectivity impairment in hypoperfusion model mice.
Collapse
Affiliation(s)
- Masaki Yoshioka
- National Institutes for Quantum Science and Technology, Institute for Quantum Life Science, Quantum Neuromapping and Neuromodulation Team, Chiba, Japan
- Chiba University, Graduate School of Medicine, Department of Neurological Surgery, Chiba, Japan
| | - Manami Takahashi
- National Institutes for Quantum Science and Technology, Institute for Quantum Life Science, Quantum Neuromapping and Neuromodulation Team, Chiba, Japan
| | - Jeff Kershaw
- National Institutes for Quantum Science and Technology, Institute for Quantum Medical Science, Department of Molecular Imaging and Theranostics, Chiba, Japan
| | - Mariko Handa
- National Institutes for Quantum Science and Technology, Institute for Quantum Life Science, Quantum Neuromapping and Neuromodulation Team, Chiba, Japan
- Chiba University, Graduate School of Science, Department of Quantum Life Science, Chiba, Japan
| | - Ayaka Takada
- National Institutes for Quantum Science and Technology, Institute for Quantum Life Science, Quantum Neuromapping and Neuromodulation Team, Chiba, Japan
- Chiba University, Graduate School of Science, Department of Quantum Life Science, Chiba, Japan
| | - Hiroyuki Takuwa
- National Institutes for Quantum Science and Technology, Institute for Quantum Life Science, Quantum Neuromapping and Neuromodulation Team, Chiba, Japan
- Chiba University, Graduate School of Science, Department of Quantum Life Science, Chiba, Japan
| |
Collapse
|
6
|
Zhu M, Kuhlman SJ, Barth AL. Transient enhancement of stimulus-evoked activity in neocortex during sensory learning. Learn Mem 2024; 31:a053870. [PMID: 38955432 PMCID: PMC11261211 DOI: 10.1101/lm.053870.123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Accepted: 05/07/2024] [Indexed: 07/04/2024]
Abstract
Synaptic potentiation has been linked to learning in sensory cortex, but the connection between this potentiation and increased sensory-evoked neural activity is not clear. Here, we used longitudinal in vivo Ca2+ imaging in the barrel cortex of awake mice to test the hypothesis that increased excitatory synaptic strength during the learning of a whisker-dependent sensory-association task would be correlated with enhanced stimulus-evoked firing. To isolate stimulus-evoked responses from dynamic, task-related activity, imaging was performed outside of the training context. Although prior studies indicate that multiwhisker stimuli drive robust subthreshold activity, we observed sparse activation of L2/3 pyramidal (Pyr) neurons in both control and trained mice. Despite evidence for excitatory synaptic strengthening at thalamocortical and intracortical synapses in this brain area at the onset of learning-indeed, under our imaging conditions thalamocortical axons were robustly activated-we observed that L2/3 Pyr neurons in somatosensory (barrel) cortex displayed only modest increases in stimulus-evoked activity that were concentrated at the onset of training. Activity renormalized over longer training periods. In contrast, when stimuli and rewards were uncoupled in a pseudotraining paradigm, stimulus-evoked activity in L2/3 Pyr neurons was significantly suppressed. These findings indicate that sensory-association training but not sensory stimulation without coupled rewards may briefly enhance sensory-evoked activity, a phenomenon that might help link sensory input to behavioral outcomes at the onset of learning.
Collapse
Affiliation(s)
- Mo Zhu
- Department of Biological Sciences, Carnegie Mellon University, Pittsburgh, Pennsylvania 15213, USA
| | - Sandra J Kuhlman
- Department of Biological Sciences, Carnegie Mellon University, Pittsburgh, Pennsylvania 15213, USA
| | - Alison L Barth
- Department of Biological Sciences, Carnegie Mellon University, Pittsburgh, Pennsylvania 15213, USA
| |
Collapse
|
7
|
Voelker P, Weible AP, Niell CM, Rothbart MK, Posner MI. Molecular Mechanisms for Changing Brain Connectivity in Mice and Humans. Int J Mol Sci 2023; 24:15840. [PMID: 37958822 PMCID: PMC10648558 DOI: 10.3390/ijms242115840] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Revised: 10/26/2023] [Accepted: 10/30/2023] [Indexed: 11/15/2023] Open
Abstract
The goal of this study was to examine commonalities in the molecular basis of learning in mice and humans. In previous work we have demonstrated that the anterior cingulate cortex (ACC) and hippocampus (HC) are involved in learning a two-choice visuospatial discrimination task. Here, we began by looking for candidate genes upregulated in mouse ACC and HC with learning. We then determined which of these were also upregulated in mouse blood. Finally, we used RT-PCR to compare candidate gene expression in mouse blood with that from humans following one of two forms of learning: a working memory task (network training) or meditation (a generalized training shown to change many networks). Two genes were upregulated in mice following learning: caspase recruitment domain-containing protein 6 (Card6) and inosine monophosphate dehydrogenase 2 (Impdh2). The Impdh2 gene product catalyzes the first committed step of guanine nucleotide synthesis and is tightly linked to cell proliferation. The Card6 gene product positively modulates signal transduction. In humans, Card6 was significantly upregulated, and Impdh2 trended toward upregulation with training. These genes have been shown to regulate pathways that influence nuclear factor kappa B (NF-κB), a factor previously found to be related to enhanced synaptic function and learning.
Collapse
Affiliation(s)
- Pascale Voelker
- Department of Psychology, University of Oregon, Eugene, OR 97403, USA (M.I.P.)
| | - Aldis P. Weible
- Institute of Neuroscience, University of Oregon, Eugene, OR 97403, USA; (A.P.W.); (C.M.N.)
| | - Cristopher M. Niell
- Institute of Neuroscience, University of Oregon, Eugene, OR 97403, USA; (A.P.W.); (C.M.N.)
- Department of Biology, University of Oregon, Eugene, OR 97403, USA
| | - Mary K. Rothbart
- Department of Psychology, University of Oregon, Eugene, OR 97403, USA (M.I.P.)
| | - Michael I. Posner
- Department of Psychology, University of Oregon, Eugene, OR 97403, USA (M.I.P.)
- Institute of Neuroscience, University of Oregon, Eugene, OR 97403, USA; (A.P.W.); (C.M.N.)
| |
Collapse
|
8
|
Kononova S, Kashparov M, Xue W, Bobkova N, Leonov S, Zagorodny N. Gut Microbiome Dysbiosis as a Potential Risk Factor for Idiopathic Toe-Walking in Children: A Review. Int J Mol Sci 2023; 24:13204. [PMID: 37686011 PMCID: PMC10488280 DOI: 10.3390/ijms241713204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2023] [Revised: 08/22/2023] [Accepted: 08/23/2023] [Indexed: 09/10/2023] Open
Abstract
Idiopathic toe walking (ITW) occurs in about 5% of children. Orthopedic treatment of ITW is complicated by the lack of a known etiology. Only half of the conservative and surgical methods of treatment give a stable positive result of normalizing gait. Available data indicate that the disease is heterogeneous and multifactorial. Recently, some children with ITW have been found to have genetic variants of mutations that can lead to the development of toe walking. At the same time, some children show sensorimotor impairment, but these studies are very limited. Sensorimotor dysfunction could potentially arise from an imbalanced production of neurotransmitters that play a crucial role in motor control. Using the data obtained in the studies of several pathologies manifested by the association of sensory-motor dysfunction and intestinal dysbiosis, we attempt to substantiate the notion that malfunction of neurotransmitter production is caused by the imbalance of gut microbiota metabolites as a result of dysbiosis. This review delves into the exciting possibility of a connection between variations in the microbiome and ITW. The purpose of this review is to establish a strong theoretical foundation and highlight the benefits of further exploring the possible connection between alterations in the microbiome and TW for further studies of ITW etiology.
Collapse
Affiliation(s)
- Svetlana Kononova
- Institute of Protein Research, Russian Academy of Sciences, 142290 Pushchino, Russia
| | - Mikhail Kashparov
- Department of Traumatology and Orthopedics, Peoples’ Friendship University of Russia, 117198 Moscow, Russia; (M.K.); (N.Z.)
- Scientific and Practical Center for Child Psychoneurology, 119602 Moscow, Russia
| | - Wenyu Xue
- School of Biological and Medical Physics, Moscow Institute of Physics and Technology, 141700 Dolgoprudny, Russia; (W.X.); (S.L.)
| | - Natalia Bobkova
- Institute of Cell Biophysics, Russian Academy of Sciences, 142290 Pushchino, Russia;
| | - Sergey Leonov
- School of Biological and Medical Physics, Moscow Institute of Physics and Technology, 141700 Dolgoprudny, Russia; (W.X.); (S.L.)
- Institute of Cell Biophysics, Russian Academy of Sciences, 142290 Pushchino, Russia;
| | - Nikolaj Zagorodny
- Department of Traumatology and Orthopedics, Peoples’ Friendship University of Russia, 117198 Moscow, Russia; (M.K.); (N.Z.)
- N.N. Priorov Central Research Institute of Traumatology and Orthopedics, 127299 Moscow, Russia
| |
Collapse
|
9
|
Ray A, Christian JA, Mosso MB, Park E, Wegner W, Willig KI, Barth AL. Quantitative Fluorescence Analysis Reveals Dendrite-Specific Thalamocortical Plasticity in L5 Pyramidal Neurons during Learning. J Neurosci 2023; 43:584-600. [PMID: 36639912 PMCID: PMC9888508 DOI: 10.1523/jneurosci.1372-22.2022] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Revised: 10/28/2022] [Accepted: 11/23/2022] [Indexed: 12/14/2022] Open
Abstract
High-throughput anatomic data can stimulate and constrain new hypotheses about how neural circuits change in response to experience. Here, we use fluorescence-based reagents for presynaptic and postsynaptic labeling to monitor changes in thalamocortical synapses onto different compartments of layer 5 (L5) pyramidal (Pyr) neurons in somatosensory (barrel) cortex from mixed-sex mice during whisker-dependent learning (Audette et al., 2019). Using axonal fills and molecular-genetic tags for synapse identification in fixed tissue from Rbp4-Cre transgenic mice, we found that thalamocortical synapses from the higher-order posterior medial thalamic nucleus showed rapid morphologic changes in both presynaptic and postsynaptic structures at the earliest stages of sensory association training. Detected increases in thalamocortical synaptic size were compartment specific, occurring selectively in the proximal dendrites onto L5 Pyr and not at inputs onto their apical tufts in L1. Both axonal and dendritic changes were transient, normalizing back to baseline as animals became expert in the task. Anatomical measurements were corroborated by electrophysiological recordings at different stages of training. Thus, fluorescence-based analysis of input- and target-specific synapses can reveal compartment-specific changes in synapse properties during learning.SIGNIFICANCE STATEMENT Synaptic changes underlie the cellular basis of learning, experience, and neurologic diseases. Neuroanatomical methods to assess synaptic plasticity can provide critical spatial information necessary for building models of neuronal computations during learning and experience but are technically and fiscally intensive. Here, we describe a confocal fluorescence microscopy-based analytical method to assess input, cell type, and dendritic location-specific synaptic plasticity in a sensory learning assay. Our method not only confirms prior electrophysiological measurements but allows us to predict functional strength of synapses in a pathway-specific manner. Our findings also indicate that changes in primary sensory cortices are transient, occurring during early learning. Fluorescence-based synapse identification can be an efficient and easily adopted approach to study synaptic changes in a variety of experimental paradigms.
Collapse
Affiliation(s)
- Ajit Ray
- Department of Biological Sciences, Carnegie Mellon University, Pittsburgh Pennsylvania 15213
| | - Joseph A Christian
- Department of Biological Sciences, Carnegie Mellon University, Pittsburgh Pennsylvania 15213
| | - Matthew B Mosso
- Department of Biological Sciences, Carnegie Mellon University, Pittsburgh Pennsylvania 15213
| | - Eunsol Park
- Department of Biological Sciences, Carnegie Mellon University, Pittsburgh Pennsylvania 15213
| | - Waja Wegner
- Optical Nanoscopy in Neuroscience, Center for Nanoscale Microscopy and Molecular Physiology of the Brain, University Medical Center Göttingen, 37099 Göttingen, Germany
- Max Planck Institute for Multidisciplinary Sciences, 37077 Göttingen, Germany
| | - Katrin I Willig
- Optical Nanoscopy in Neuroscience, Center for Nanoscale Microscopy and Molecular Physiology of the Brain, University Medical Center Göttingen, 37099 Göttingen, Germany
- Max Planck Institute for Multidisciplinary Sciences, 37077 Göttingen, Germany
| | - Alison L Barth
- Department of Biological Sciences, Carnegie Mellon University, Pittsburgh Pennsylvania 15213
| |
Collapse
|
10
|
Xing Y, Zan C, Liu L. Recent advances in understanding neuronal diversity and neural circuit complexity across different brain regions using single-cell sequencing. Front Neural Circuits 2023; 17:1007755. [PMID: 37063385 PMCID: PMC10097998 DOI: 10.3389/fncir.2023.1007755] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2022] [Accepted: 02/16/2023] [Indexed: 04/18/2023] Open
Abstract
Neural circuits are characterized as interconnecting neuron networks connected by synapses. Some kinds of gene expression and/or functional changes of neurons and synaptic connections may result in aberrant neural circuits, which has been recognized as one crucial pathological mechanism for the onset of many neurological diseases. Gradual advances in single-cell sequencing approaches with strong technological advantages, as exemplified by high throughput and increased resolution for live cells, have enabled it to assist us in understanding neuronal diversity across diverse brain regions and further transformed our knowledge of cellular building blocks of neural circuits through revealing numerous molecular signatures. Currently published transcriptomic studies have elucidated various neuronal subpopulations as well as their distribution across prefrontal cortex, hippocampus, hypothalamus, and dorsal root ganglion, etc. Better characterization of brain region-specific circuits may shed light on new pathological mechanisms involved and assist in selecting potential targets for the prevention and treatment of specific neurological disorders based on their established roles. Given diverse neuronal populations across different brain regions, we aim to give a brief sketch of current progress in understanding neuronal diversity and neural circuit complexity according to their locations. With the special focus on the application of single-cell sequencing, we thereby summarize relevant region-specific findings. Considering the importance of spatial context and connectivity in neural circuits, we also discuss a few published results obtained by spatial transcriptomics. Taken together, these single-cell sequencing data may lay a mechanistic basis for functional identification of brain circuit components, which links their molecular signatures to anatomical regions, connectivity, morphology, and physiology. Furthermore, the comprehensive characterization of neuron subtypes, their distributions, and connectivity patterns via single-cell sequencing is critical for understanding neural circuit properties and how they generate region-dependent interactions in different context.
Collapse
Affiliation(s)
- Yu Xing
- Department of Neurology, Beidahuang Industry Group General Hospital, Harbin, China
| | - Chunfang Zan
- Institute for Stroke and Dementia Research (ISD), LMU Klinikum, Ludwig-Maximilian-University (LMU), Munich, Germany
| | - Lu Liu
- Munich Medical Research School (MMRS), LMU Klinikum, Ludwig-Maximilian-University (LMU), Munich, Germany
- *Correspondence: Lu Liu, ,
| |
Collapse
|
11
|
Houlihan LM, Naughton D, O'Sullivan MGJ, Lawton MT, Preul MC. Toward "bigger" data for neurosurgical anatomical research: a single centralized quantitative neurosurgical anatomy platform. Neurosurg Rev 2022; 46:22. [PMID: 36544017 DOI: 10.1007/s10143-022-01924-y] [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: 11/15/2022] [Revised: 11/16/2022] [Accepted: 11/30/2022] [Indexed: 12/24/2022]
Abstract
Quantitative neurosurgical anatomy research aims to produce surgically applicable knowledge for improving operative decision-making using measurements from anatomical dissection and tools such as stereotaxis. Although such studies attempt to answer similar research questions, there is little standardization between them, offering minimal comparability. Modern technology has been incorporated into the research methodology, but many scientific principles are lacking, and results are not broadly applicable or suitable for evaluating big-data trends. Advances in information technology and the concept of big data permit more accessible and robust means of producing valuable, standardized, reliable research. A technology project, "Inchin," is presented to address these needs for neurosurgical anatomy research. This study applies the concept of big data to neurosurgical anatomy research, specifically in quantifying surgical metrics. A remote-hosted web application was developed for computing standard neurosurgical metrics and storing measurement data. An online portal (Inchin) was developed to produce a database to facilitate and promote neurosurgical anatomical research, applying optimal scientific methodology and big-data principles to this recent and evolving field of research. Individual data sets are not insignificant, but a collective of data sets present advantages. Large data sets allow confidence in data trends that are usually obscured in smaller numbers of samples. Inchin, a single centralized software platform, can act as a global database of results of neurosurgical anatomy studies. A calculation tool ensuring standardized peer-reviewed methodology, Inchin is applied to the analysis of neurosurgical metrics and may promote efficient study collaboration within and among neurosurgical laboratories.
Collapse
Affiliation(s)
- Lena Mary Houlihan
- The Loyal and Edith Davis Neurosurgical Research Laboratory, Department of Neurosurgery, Barrow Neurological Institute, St. Joseph's Hospital and Medical Center, 350 W Thomas Rd, Phoenix, AZ, 85013, USA
| | - David Naughton
- The Loyal and Edith Davis Neurosurgical Research Laboratory, Department of Neurosurgery, Barrow Neurological Institute, St. Joseph's Hospital and Medical Center, 350 W Thomas Rd, Phoenix, AZ, 85013, USA
| | | | - Michael T Lawton
- The Loyal and Edith Davis Neurosurgical Research Laboratory, Department of Neurosurgery, Barrow Neurological Institute, St. Joseph's Hospital and Medical Center, 350 W Thomas Rd, Phoenix, AZ, 85013, USA
| | - Mark C Preul
- The Loyal and Edith Davis Neurosurgical Research Laboratory, Department of Neurosurgery, Barrow Neurological Institute, St. Joseph's Hospital and Medical Center, 350 W Thomas Rd, Phoenix, AZ, 85013, USA.
| |
Collapse
|
12
|
Park E, Barth AL. IEG expression defines SST neuron ensembles critical for motor learning. Neuron 2022; 110:3222-3224. [PMID: 36265439 DOI: 10.1016/j.neuron.2022.09.036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Neocortical interneurons have been hypothesized to be important for circuit reorganization during learning. In this issue of Neuron, Yang et al. (2022) identify a subset of Npas4-expressing somatostatin interneurons that help regulate excitatory synaptic plasticity during motor learning.
Collapse
Affiliation(s)
- Eunsol Park
- Department of Biological Sciences, Carnegie Mellon University, Pittsburgh, PA, USA
| | - Alison L Barth
- Department of Biological Sciences, Carnegie Mellon University, Pittsburgh, PA, USA.
| |
Collapse
|
13
|
Kuljis DA, Micheva KD, Ray A, Wegner W, Bowman R, Madison DV, Willig KI, Barth AL. Gephyrin-Lacking PV Synapses on Neocortical Pyramidal Neurons. Int J Mol Sci 2021; 22:ijms221810032. [PMID: 34576197 PMCID: PMC8467468 DOI: 10.3390/ijms221810032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 09/01/2021] [Accepted: 09/07/2021] [Indexed: 11/26/2022] Open
Abstract
Gephyrin has long been thought of as a master regulator for inhibitory synapses, acting as a scaffold to organize γ-aminobutyric acid type A receptors (GABAARs) at the post-synaptic density. Accordingly, gephyrin immunostaining has been used as an indicator of inhibitory synapses; despite this, the pan-synaptic localization of gephyrin to specific classes of inhibitory synapses has not been demonstrated. Genetically encoded fibronectin intrabodies generated with mRNA display (FingRs) against gephyrin (Gephyrin.FingR) reliably label endogenous gephyrin, and can be tagged with fluorophores for comprehensive synaptic quantitation and monitoring. Here we investigated input- and target-specific localization of gephyrin at a defined class of inhibitory synapse, using Gephyrin.FingR proteins tagged with EGFP in brain tissue from transgenic mice. Parvalbumin-expressing (PV) neuron presynaptic boutons labeled using Cre- dependent synaptophysin-tdTomato were aligned with postsynaptic Gephyrin.FingR puncta. We discovered that more than one-third of PV boutons adjacent to neocortical pyramidal (Pyr) cell somas lack postsynaptic gephyrin labeling. This finding was confirmed using correlative fluorescence and electron microscopy. Our findings suggest some inhibitory synapses may lack gephyrin. Gephyrin-lacking synapses may play an important role in dynamically regulating cell activity under different physiological conditions.
Collapse
Affiliation(s)
- Dika A. Kuljis
- Center for the Neural Basis of Cognition, Department of Biological Sciences, Carnegie Mellon University, Pittsburgh, PA 15213, USA; (D.A.K.); (A.R.); (R.B.)
| | - Kristina D. Micheva
- Department of Molecular and Cellular Physiology, Stanford University, Palo Alto, CA 94304, USA; (K.D.M.); (D.V.M.)
| | - Ajit Ray
- Center for the Neural Basis of Cognition, Department of Biological Sciences, Carnegie Mellon University, Pittsburgh, PA 15213, USA; (D.A.K.); (A.R.); (R.B.)
| | - Waja Wegner
- Optical Nanoscopy in Neuroscience, Center for Nanoscale Microscopy and Molecular Physiology of the Brain, University Medical Center Göttingen, 37075 Göttingen, Germany; (W.W.); (K.I.W.)
- Max Planck Institute of Experimental Medicine, 37075 Göttingen, Germany
| | - Ryan Bowman
- Center for the Neural Basis of Cognition, Department of Biological Sciences, Carnegie Mellon University, Pittsburgh, PA 15213, USA; (D.A.K.); (A.R.); (R.B.)
| | - Daniel V. Madison
- Department of Molecular and Cellular Physiology, Stanford University, Palo Alto, CA 94304, USA; (K.D.M.); (D.V.M.)
| | - Katrin I. Willig
- Optical Nanoscopy in Neuroscience, Center for Nanoscale Microscopy and Molecular Physiology of the Brain, University Medical Center Göttingen, 37075 Göttingen, Germany; (W.W.); (K.I.W.)
- Max Planck Institute of Experimental Medicine, 37075 Göttingen, Germany
| | - Alison L. Barth
- Center for the Neural Basis of Cognition, Department of Biological Sciences, Carnegie Mellon University, Pittsburgh, PA 15213, USA; (D.A.K.); (A.R.); (R.B.)
- Correspondence: ; Tel.: +1-412-268-1198
| |
Collapse
|
14
|
Zeiger WA, Marosi M, Saggi S, Noble N, Samad I, Portera-Cailliau C. Barrel cortex plasticity after photothrombotic stroke involves potentiating responses of pre-existing circuits but not functional remapping to new circuits. Nat Commun 2021; 12:3972. [PMID: 34172735 PMCID: PMC8233353 DOI: 10.1038/s41467-021-24211-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Accepted: 06/01/2021] [Indexed: 01/14/2023] Open
Abstract
Recovery after stroke is thought to be mediated by adaptive circuit plasticity, whereby surviving neurons assume the roles of those that died. However, definitive longitudinal evidence of neurons changing their response selectivity after stroke is lacking. We sought to directly test whether such functional “remapping” occurs within mouse primary somatosensory cortex after a stroke that destroys the C1 barrel. Using in vivo calcium imaging to longitudinally record sensory-evoked activity under light anesthesia, we did not find any increase in the number of C1 whisker-responsive neurons in the adjacent, spared D3 barrel after stroke. To promote plasticity after stroke, we also plucked all whiskers except C1 (forced use therapy). This led to an increase in the reliability of sensory-evoked responses in C1 whisker-responsive neurons but did not increase the number of C1 whisker-responsive neurons in spared surround barrels over baseline levels. Our results argue against remapping of functionality after barrel cortex stroke, but support a circuit-based mechanism for how rehabilitation may improve recovery. Definitive evidence for functional remapping after stroke remains lacking. Here, the authors performed in vivo intrinsic signal imaging and two-photon calcium imaging of sensory-evoked responses before and after photothrombotic stroke and found no evidence of remapping of lost functionalities to new circuits in peri-infarct cortex.
Collapse
Affiliation(s)
- William A Zeiger
- Department of Neurology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA.
| | - Máté Marosi
- Department of Neurology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA.,Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, TX, USA
| | - Satvir Saggi
- Department of Neurology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Natalie Noble
- Department of Neurology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Isa Samad
- Department of Neurology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Carlos Portera-Cailliau
- Department of Neurology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA. .,Department of Neurobiology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA.
| |
Collapse
|
15
|
Houlihan LM, Naughton D, Preul MC. Volume of Surgical Freedom: The Most Applicable Anatomical Measurement for Surgical Assessment and 3-Dimensional Modeling. Front Bioeng Biotechnol 2021; 9:628797. [PMID: 33928070 PMCID: PMC8076649 DOI: 10.3389/fbioe.2021.628797] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Accepted: 03/22/2021] [Indexed: 11/13/2022] Open
Abstract
Surgical freedom is the most important metric at the disposal of the surgeon. The volume of surgical freedom (VSF) is a new methodology that produces an optimal qualitative and quantitative representation of an access corridor and provides the surgeon with an anatomical, spatially accurate, and clinically applicable metric. In this study, illustrative dissection examples were completed using two of the most common surgical approaches, the pterional craniotomy and the supraorbital craniotomy. The VSF methodology models the surgical corridor as a cone with an irregular base. The measurement data are fitted to the cone model, and from these fitted data, the volume of the cone is calculated as a volumetric measurement of the surgical corridor. A normalized VSF compensates for inaccurate measurements that may occur as a result of dependence on probe length during data acquisition and provides a fixed reference metric that is applicable across studies. The VSF compensates for multiple inaccuracies in the practical and mathematical methods currently used for quantitative assessment, thereby enabling the production of 3-dimensional models of the surgical corridor. The VSF is therefore an improved standard for assessment of surgical freedom.
Collapse
Affiliation(s)
- Lena Mary Houlihan
- The Loyal and Edith Davis Neurosurgical Research Laboratory, Department of Neurosurgery, Barrow Neurological Institute, St. Joseph's Hospital and Medical Center, Phoenix, AZ, United States
| | - David Naughton
- The Loyal and Edith Davis Neurosurgical Research Laboratory, Department of Neurosurgery, Barrow Neurological Institute, St. Joseph's Hospital and Medical Center, Phoenix, AZ, United States
| | - Mark C Preul
- The Loyal and Edith Davis Neurosurgical Research Laboratory, Department of Neurosurgery, Barrow Neurological Institute, St. Joseph's Hospital and Medical Center, Phoenix, AZ, United States
| |
Collapse
|
16
|
Conelea CA, Jacob S, Redish AD, Ramsay IS. Considerations for Pairing Cognitive Behavioral Therapies and Non-invasive Brain Stimulation: Ignore at Your Own Risk. Front Psychiatry 2021; 12:660180. [PMID: 33912088 PMCID: PMC8072056 DOI: 10.3389/fpsyt.2021.660180] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Accepted: 03/15/2021] [Indexed: 12/30/2022] Open
Abstract
Multimodal approaches combining cognitive behavioral therapies (CBT) with non-invasive brain stimulation (NIBS) hold promise for improving the treatment of neuropsychiatric disorders. As this is a relatively new approach, it is a critical time to identify guiding principles and methodological considerations to enhance research rigor. In the current paper, we argue for a principled approach to CBT and NIBS pairings based on synergistic activation of neural circuits and identify key considerations about CBT that may influence pairing with NIBS. Careful consideration of brain-state interactions and CBT-related nuances will increase the potential for these combinations to be positively synergistic.
Collapse
Affiliation(s)
- Christine A Conelea
- Department of Psychiatry and Behavioral Sciences, University of Minnesota, Minneapolis, MN, United States
| | - Suma Jacob
- Department of Psychiatry and Behavioral Sciences, University of Minnesota, Minneapolis, MN, United States
| | - A David Redish
- Department of Neuroscience, University of Minnesota, Minneapolis, MN, United States
| | - Ian S Ramsay
- Department of Psychiatry and Behavioral Sciences, University of Minnesota, Minneapolis, MN, United States
| |
Collapse
|
17
|
Xu J, Liu J, Li Q, Mi Y, Zhou D, Meng Q, Chen G, Li N, Hou Y. Pterostilbene Alleviates Aβ 1-42 -Induced Cognitive Dysfunction via Inhibition of Oxidative Stress by Activating Nrf2 Signaling Pathway. Mol Nutr Food Res 2020; 65:e2000711. [PMID: 33280250 DOI: 10.1002/mnfr.202000711] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2020] [Revised: 11/04/2020] [Indexed: 12/19/2022]
Abstract
SCOPE In the present study, effect of pterostilbene on β-amyloid 1-42 (Aβ1-42 ) induced cognitive impairment in mice is investigated and explored its possible mechanism of action. METHODS AND RESULTS The behavior results show that pterostilbene alleviated Aβ1-42 -induces cognitive dysfunction assessed using the Y-maze test, novel object recognition task, Morris water maze test, and passive avoidance test. Pterostilbene alleviates neuron loss and accumulation of reactive oxygen species in Aβ1-42 treated mouse brain. Additionally, pterostilbene promotes nuclear factor-E2 p45-related factor 2 (Nrf2) nuclear translocation and enhance the transcription and expression of antioxidant genes such as heme oxygenase-1 and superoxide dismutase both in vivo and in vitro. Nrf2 inhibitor ML385 reverses the antioxidant function of pterostilbene in SH-SY5Y cells. Nrf2 is the master regulator of oxidative homeostasis and can be activated by substrate adaptor sequestosome-1 (also named p62). Pterostilbene promotes the binding of Kelch-like ECH-associated protein 1 and p62, which enhanced activation of Nrf2. CONCLUSION The present study reports that pterostilbene alleviated Aβ1-42 -induces cognitive dysfunction in mice. The mechanism of pterostilbene can be associated to the inhibition of oxidative stress through the Nrf2 signaling pathway.
Collapse
Affiliation(s)
- Jikai Xu
- College of Life and Health Sciences, Northeastern University, Shenyang, 110819, China.,Key Laboratory of Data Analytics and Optimization for Smart Industry, Northeastern University, Ministry of Education, Shenyang, 110819, China
| | - Jingyu Liu
- College of Life and Health Sciences, Northeastern University, Shenyang, 110819, China.,Key Laboratory of Data Analytics and Optimization for Smart Industry, Northeastern University, Ministry of Education, Shenyang, 110819, China
| | - Qing Li
- College of Life and Health Sciences, Northeastern University, Shenyang, 110819, China
| | - Yan Mi
- College of Life and Health Sciences, Northeastern University, Shenyang, 110819, China
| | - Di Zhou
- School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Qingqi Meng
- College of Life and Health Sciences, Northeastern University, Shenyang, 110819, China
| | - Gang Chen
- School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Ning Li
- School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Yue Hou
- College of Life and Health Sciences, Northeastern University, Shenyang, 110819, China.,Key Laboratory of Data Analytics and Optimization for Smart Industry, Northeastern University, Ministry of Education, Shenyang, 110819, China
| |
Collapse
|
18
|
Venkataraman L, Fair SR, McElroy CA, Hester ME, Fu H. Modeling neurodegenerative diseases with cerebral organoids and other three-dimensional culture systems: focus on Alzheimer's disease. Stem Cell Rev Rep 2020; 18:696-717. [PMID: 33180261 PMCID: PMC7658915 DOI: 10.1007/s12015-020-10068-9] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/25/2020] [Indexed: 12/11/2022]
Abstract
Many neurodegenerative diseases (NDs) such as Alzheimer’s disease, Parkinson’s disease, frontotemporal dementia, amyotrophic lateral sclerosis and Huntington’s disease, are characterized by the progressive accumulation of abnormal proteinaceous assemblies in specific cell types and regions of the brain, leading to cellular dysfunction and brain damage. Although animal- and in vitro-based studies of NDs have provided the field with an extensive understanding of some of the mechanisms underlying these diseases, findings from these studies have not yielded substantial progress in identifying treatment options for patient populations. This necessitates the development of complementary model systems that are better suited to recapitulate human-specific features of ND pathogenesis. Three-dimensional (3D) culture systems, such as cerebral organoids generated from human induced pluripotent stem cells, hold significant potential to model NDs in a complex, tissue-like environment. In this review, we discuss the advantages of 3D culture systems and 3D modeling of NDs, especially AD and FTD. We also provide an overview of the challenges and limitations of the current 3D culture systems. Finally, we propose a few potential future directions in applying state-of-the-art technologies in 3D culture systems to understand the mechanisms of NDs and to accelerate drug discovery. Graphical abstract ![]()
Collapse
Affiliation(s)
- Lalitha Venkataraman
- Department of Neuroscience, The Ohio State University Wexner Medical Center, 616 Biomedical Research Tower, 460 W. 12th Ave, Columbus, OH, 43210, USA
| | - Summer R Fair
- The Steve and Cindy Rasmussen Institute for Genomic Medicine, Abigail Wexner Research Institute at Nationwide Children's Hospital, 575 Children's Crossroad, Columbus, OH, 43215, USA
- College of Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Craig A McElroy
- College of Pharmacy, The Ohio State University, Columbus, OH, USA
| | - Mark E Hester
- Department of Neuroscience, The Ohio State University Wexner Medical Center, 616 Biomedical Research Tower, 460 W. 12th Ave, Columbus, OH, 43210, USA.
- The Steve and Cindy Rasmussen Institute for Genomic Medicine, Abigail Wexner Research Institute at Nationwide Children's Hospital, 575 Children's Crossroad, Columbus, OH, 43215, USA.
- Department of Pediatrics, The Ohio State University Wexner Medical Center, Columbus, OH, USA.
| | - Hongjun Fu
- Department of Neuroscience, The Ohio State University Wexner Medical Center, 616 Biomedical Research Tower, 460 W. 12th Ave, Columbus, OH, 43210, USA.
| |
Collapse
|
19
|
Gil R, Fernandes FF, Shemesh N. Neuroplasticity-driven timing modulations revealed by ultrafast functional magnetic resonance imaging. Neuroimage 2020; 225:117446. [PMID: 33069861 DOI: 10.1016/j.neuroimage.2020.117446] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Revised: 09/14/2020] [Accepted: 10/07/2020] [Indexed: 12/13/2022] Open
Abstract
Detecting neuroplasticity in global brain circuits in vivo is key for understanding myriad processes such as memory, learning, and recovery from injury. Functional Magnetic Resonance Imaging (fMRI) is instrumental for such in vivo mappings, yet it typically relies on mapping changes in spatial extent of activation or via signal amplitude modulations, whose interpretation can be highly ambiguous. Importantly, a central aspect of neuroplasticity involves modulation of neural activity timing properties. We thus hypothesized that this temporal dimension could serve as a new marker for neuroplasticity. To detect fMRI signals more associated with the underlying neural dynamics, we developed an ultrafast fMRI (ufMRI) approach facilitating high spatiotemporal sensitivity and resolution in distributed neural pathways. When neuroplasticity was induced in the mouse visual pathway via dark rearing, ufMRI indeed mapped temporal modulations in the entire visual pathway. Our findings therefore suggest a new dimension for exploring neuroplasticity in vivo.
Collapse
Affiliation(s)
- Rita Gil
- Champalimaud Research, Champalimaud Centre for the Unknown, Lisbon, Portugal
| | | | - Noam Shemesh
- Champalimaud Research, Champalimaud Centre for the Unknown, Lisbon, Portugal.
| |
Collapse
|