1
|
Parnami K, Surana A, Choudhary V, Bhattacharyya A. Deprivation of visual input alters specific subset of inhibitory neurons and affect thalamic afferent terminals in V1 of rd1 mouse. Front Cell Neurosci 2024; 18:1422613. [PMID: 39444393 PMCID: PMC11496165 DOI: 10.3389/fncel.2024.1422613] [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: 04/24/2024] [Accepted: 09/19/2024] [Indexed: 10/25/2024] Open
Abstract
Retinitis Pigmentosa (RP) is a heterogenous group of inherited disorder, and its progression not only affects the retina but also the primary visual cortex. This manifests imbalances in the excitatory and inhibitory neurotransmission. Here, we investigated if changes in cortical functioning is linked to alterations in GABAergic population of neurons and its two important subsets, somatostatin (SST) and parvalbumin (PV) neuron in rd1 model of retinal degeneration (RD). We demonstrate marked decrease in the proportion of SST neurons in different layers of cortex whereas PV neurons were less affected. Moreover, we found reduced expression of glutamatergic thalamic afferents (VGLUT2) due to lack of visual activity. These results suggest PV neurons are likely recruited by the cortical circuitry to increase the inhibitory drive and compensate the disrupted inhibition-excitation balance. However, reduced SST expression perhaps results in weakening of stimulus selectivity. Delineating their functional role during RD will provide insights for acquisition of high-resolution vision thereby improving current state of vision restoration.
Collapse
Affiliation(s)
- Kashish Parnami
- Amity Institute of Neuropsychology and Neurosciences, Amity University Noida, Noida, India
| | - Anushka Surana
- Amity Institute of Neuropsychology and Neurosciences, Amity University Noida, Noida, India
| | - Vineet Choudhary
- Department of Biotechnology, All India Institute of Medical Sciences (AIIMS), New Delhi, India
| | - Anwesha Bhattacharyya
- Amity Institute of Neuropsychology and Neurosciences, Amity University Noida, Noida, India
| |
Collapse
|
2
|
Luna R, Li J, Bauer R, van Leeuwen C. Retinal waves in adaptive rewiring networks orchestrate convergence and divergence in the visual system. Netw Neurosci 2024; 8:653-672. [PMID: 39355440 PMCID: PMC11340993 DOI: 10.1162/netn_a_00370] [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: 10/16/2023] [Accepted: 02/26/2024] [Indexed: 10/03/2024] Open
Abstract
Spontaneous retinal wave activity shaping the visual system is a complex neurodevelopmental phenomenon. Retinal ganglion cells are the hubs through which activity diverges throughout the visual system. We consider how these divergent hubs emerge, using an adaptively rewiring neural network model. Adaptive rewiring models show in a principled way how brains could achieve their complex topologies. Modular small-world structures with rich-club effects and circuits of convergent-divergent units emerge as networks evolve, driven by their own spontaneous activity. Arbitrary nodes of an initially random model network were designated as retinal ganglion cells. They were intermittently exposed to the retinal waveform, as the network evolved through adaptive rewiring. A significant proportion of these nodes developed into divergent hubs within the characteristic complex network architecture. The proportion depends parametrically on the wave incidence rate. Higher rates increase the likelihood of hub formation, while increasing the potential of ganglion cell death. In addition, direct neighbors of designated ganglion cells differentiate like amacrine cells. The divergence observed in ganglion cells resulted in enhanced convergence downstream, suggesting that retinal waves control the formation of convergence in the lateral geniculate nuclei. We conclude that retinal waves stochastically control the distribution of converging and diverging activity in evolving complex networks.
Collapse
Affiliation(s)
- Raúl Luna
- Department of Psychobiology and Methodology in Behavioural Sciences, Faculty of Psychology, Universidad Complutense de Madrid, Madrid, Spain
- Institute of Optics, Spanish National Research Council (CSIC), Madrid, Spain
- KU Leuven, Brain and Cognition, Leuven, Belgium
| | - Jia Li
- KU Leuven, Brain and Cognition, Leuven, Belgium
| | - Roman Bauer
- NICE Research Group, Computer Science Research Centre, University of Surrey, Guildford, UK
| | - Cees van Leeuwen
- KU Leuven, Brain and Cognition, Leuven, Belgium
- RPTU Kaiserslautern, Cognitive Science, Kaiserslautern, Germany
| |
Collapse
|
3
|
Yoon D, Oh SM, Na HS, Choi BR, Kim KW, Lee YS, Lee DR, Lee DY. Metabolomics study to reveal cognitive improvement with treatment of Scrophularia buergeriana. Sci Rep 2024; 14:17007. [PMID: 39043762 PMCID: PMC11266482 DOI: 10.1038/s41598-024-66371-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Accepted: 07/01/2024] [Indexed: 07/25/2024] Open
Abstract
Population aging around the world is rapidly progressing; as a result, cognitive decline developing into dementia is becoming a social problem. There is no drug that can cure dementia, and though drugs that alleviate the symptoms of dementia have been developed, they also have side effects. Therefore, we conducted a study on improving cognitive function using natural products that have secured safety. We confirmed the effect of an extract of Scrophularia buergeriana on scopolamine-induced cognitive impairment through mouse behavioral experiments, and we observed metabolic changes in the cortex and hippocampus via brain tissue dissection after the behavioral experiment. Mitigating effects of S. buergeriana on cognitive impairment caused by scopolamine were observed in passive avoidance and Morris water maze tests. A metabolic analysis revealed biomarkers related to the alleviating effect of cognitive impairment. Niacinamide, tyrosine, uridine, and valine in the cortex and GABA, choline, creatine, formate, fumarate, hypoxanthine, leucine, myo-inositol, pyroglutamate, and taurine in the hippocampus were identified as biomarker candidates for recovering cognitive impairment. In addition to behavioral experiments, this metabolomics study using specific regions of the brain may be helpful in understanding the effects of cognitive improvement.
Collapse
Affiliation(s)
- Dahye Yoon
- Department of Herbal Crop Research, National Institute of Horticultural and Herbal Science, RDA, Eumseong, 27709, Republic of Korea
| | - Seon Min Oh
- Natural Product Research Center and Natural Product Central Bank, KRIBB, Ochang, 28116, Republic of Korea
| | - Hyeon Seon Na
- Department of Herbal Crop Research, National Institute of Horticultural and Herbal Science, RDA, Eumseong, 27709, Republic of Korea
| | - Bo-Ram Choi
- Department of Herbal Crop Research, National Institute of Horticultural and Herbal Science, RDA, Eumseong, 27709, Republic of Korea
| | - Kwan-Woo Kim
- Department of Herbal Crop Research, National Institute of Horticultural and Herbal Science, RDA, Eumseong, 27709, Republic of Korea
| | - Young-Seob Lee
- Department of Herbal Crop Research, National Institute of Horticultural and Herbal Science, RDA, Eumseong, 27709, Republic of Korea
| | | | - Dae Young Lee
- BK21 FOUR KNU Creative BioResearch Group, School of Life Sciences, Kyungpook National University, Daegu, 41566, Republic of Korea.
| |
Collapse
|
4
|
Sarkar S, Martinez Reyes C, Jensen CM, Gavornik JP. M2 receptors are required for spatiotemporal sequence learning in mouse primary visual cortex. J Neurophysiol 2024; 131:1213-1225. [PMID: 38629848 PMCID: PMC11381118 DOI: 10.1152/jn.00016.2024] [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: 01/09/2024] [Revised: 03/08/2024] [Accepted: 04/16/2024] [Indexed: 06/09/2024] Open
Abstract
Acetylcholine is a neurotransmitter that plays a variety of roles in the central nervous system. It was previously shown that blocking muscarinic receptors with a nonselective antagonist prevents a form of experience-dependent plasticity termed "spatiotemporal sequence learning" in the mouse primary visual cortex (V1). Muscarinic signaling is a complex process involving the combined activities of five different G protein-coupled receptors, M1-M5, all of which are expressed in the murine brain but differ from each other functionally and in anatomical localization. Here we present electrophysiological evidence that M2, but not M1, receptors are required for spatiotemporal sequence learning in mouse V1. We show in male mice that M2 is highly expressed in the neuropil in V1, especially in thalamorecipient layer 4, and colocalizes with the soma in a subset of somatostatin-expressing neurons in deep layers. We also show that expression of M2 receptors is higher in the monocular region of V1 than it is in the binocular region but that the amount of experience-dependent sequence potentiation is similar in both regions and that blocking muscarinic signaling after visual stimulation does not prevent plasticity. This work establishes a new functional role for M2-type receptors in processing temporal information and demonstrates that monocular circuits are modified by experience in a manner similar to binocular circuits.NEW & NOTEWORTHY Muscarinic acetylcholine receptors are required for multiple forms of plasticity in the brain and support perceptual functions, but the precise role of the five subtypes (M1-M5) are unclear. Here we show that the M2 receptor is specifically required to encode experience-dependent representations of spatiotemporal relationships in both monocular and binocular regions of mouse V1. This work identifies a novel functional role for M2 receptors in coding temporal information into cortical circuits.
Collapse
Affiliation(s)
- Susrita Sarkar
- Center for Systems Neuroscience, Department of Biology, Boston University, Boston, Massachusetts, United States
| | - Catalina Martinez Reyes
- Center for Systems Neuroscience, Department of Biology, Boston University, Boston, Massachusetts, United States
| | - Cambria M Jensen
- Center for Systems Neuroscience, Department of Biology, Boston University, Boston, Massachusetts, United States
| | - Jeffrey P Gavornik
- Center for Systems Neuroscience, Department of Biology, Boston University, Boston, Massachusetts, United States
| |
Collapse
|
5
|
Marrero K, Aruljothi K, Delgadillo C, Kabbara S, Swatch L, Zagha E. Goal-Directed Learning is Multidimensional and Accompanied by Diverse and Widespread Changes in Neocortical Signaling. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.02.13.528412. [PMID: 36824924 PMCID: PMC9948952 DOI: 10.1101/2023.02.13.528412] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/17/2023]
Abstract
New tasks are often learned in stages with each stage reflecting a different learning challenge. Accordingly, each learning stage is likely mediated by distinct neuronal processes. And yet, most rodent studies of the neuronal correlates of goal-directed learning focus on individual outcome measures and individual brain regions. Here, we longitudinally studied mice from naïve to expert performance in a head-fixed, operant conditioning whisker discrimination task. In addition to tracking the primary behavioral outcome of stimulus discrimination, we tracked and compared an array of object-based and temporal-based behavioral measures. These behavioral analyses identify multiple, partially overlapping learning stages in this task, consistent with initial response implementation, early stimulus-response generalization, and late response inhibition. To begin to understand the neuronal foundations of these learning processes, we performed widefield Ca2+ imaging of dorsal neocortex throughout learning and correlated behavioral measures with neuronal activity. We found distinct and widespread correlations between neocortical activation patterns and various behavioral measures. For example, improvements in sensory discrimination correlated with target stimulus evoked activations of licking-related cortices along with distractor stimulus evoked global cortical suppression. Our study reveals multidimensional learning for a simple goal-directed learning task and generates hypotheses for the neuronal modulations underlying these various learning processes.
Collapse
Affiliation(s)
- Krista Marrero
- Neuroscience Graduate Program, University of California, Riverside 900 University Avenue, Riverside CA 92521 USA
| | - Krithiga Aruljothi
- Department of Psychology, University of California, Riverside 900 University Avenue, Riverside CA 92521 USA
| | - Christian Delgadillo
- Division of Biomedical Sciences, University of California, Riverside 900 University Avenue, Riverside CA 92521 USA
| | - Sarah Kabbara
- Neuroscience Graduate Program, University of California, Riverside 900 University Avenue, Riverside CA 92521 USA
| | - Lovleen Swatch
- College of Natural & Agricultural Sciences, University of California, Riverside 900 University Avenue, Riverside CA 92521 USA
| | - Edward Zagha
- Neuroscience Graduate Program, University of California, Riverside 900 University Avenue, Riverside CA 92521 USA
- Department of Psychology, University of California, Riverside 900 University Avenue, Riverside CA 92521 USA
| |
Collapse
|
6
|
Riedemann T, Sutor B. Cell-Type-Specific Effects of Somatostatin on Synaptic Transmission in the Anterior Cingulate Cortex. J Neurosci 2024; 44:e0598232024. [PMID: 38378274 PMCID: PMC10977029 DOI: 10.1523/jneurosci.0598-23.2024] [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: 03/30/2023] [Revised: 01/19/2024] [Accepted: 01/24/2024] [Indexed: 02/22/2024] Open
Abstract
Inhibitory modulation of glutamatergic information processing is a prerequisite for proper network function. Among the many groups of interneurons (INs), somatostatin-expressing interneurons (SOM-INs) play an important role in the maintenance of physiological brain activity. We have previously shown that somatostatin (SOM) causes a reduction in pyramidal cell (PC) excitability. However, the mechanisms of action of the peptide on cortical synaptic circuits are still unclear. To understand the effects of the neuropeptide SOM on cortical synaptic circuits, we performed a detailed side-by-side comparison of its postsynaptic effects on PCs, SOM-INs, and layer 1 interneurons (L1-INs) in the anterior cingulate cortex of male and female mice and found that SOM produced pronounced postsynaptic effects in PCs while having little to no effect on either IN type. This comparison allowed us to link the observed postsynaptic effects to SOM-induced modulations of glutamatergic and GABAergic synaptic transmission and to trace the impact of the neuropeptide on the neuronal circuitry between these three cell types. We show here that SOM depresses glutamatergic synaptic transmission via a presynaptic mechanism while exerting a differential impact on GABAA receptor- and GABAB receptor-mediated transmission at the pre- and postsynaptic level resulting in a shift of inhibition in L2/3 PCs from L1-INs to SOM-INs. In summary, this study unravels a novel aspect by which SOM modulates synaptic signaling between PCs, L1-INs, and SOM-INs.
Collapse
Affiliation(s)
- Therese Riedemann
- Department of Physiological Genomics, Institute of Physiology, Biomedical Center, Ludwig-Maximilians-Universität München, Planegg-Martinsried 82152, Germany
- Center of Physiology, Pathophysiology and Biophysics, Institute of Physiology and Pathophysiology, Paracelsus Medical University, Salzburg 5020, Austria
| | - Bernd Sutor
- Department of Physiological Genomics, Institute of Physiology, Biomedical Center, Ludwig-Maximilians-Universität München, Planegg-Martinsried 82152, Germany
| |
Collapse
|
7
|
Kalafut NC, Huang X, Wang D. Joint variational autoencoders for multimodal imputation and embedding. NAT MACH INTELL 2023; 5:631-642. [PMID: 39175596 PMCID: PMC11340721 DOI: 10.1038/s42256-023-00663-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2022] [Accepted: 04/21/2023] [Indexed: 08/24/2024]
Abstract
Single-cell multimodal datasets have measured various characteristics of individual cells, enabling a deep understanding of cellular and molecular mechanisms. However, multimodal data generation remains costly and challenging, and missing modalities happen frequently. Recently, machine learning approaches have been developed for data imputation but typically require fully matched multimodalities to learn common latent embeddings that potentially lack modality specificity. To address these issues, we developed an open-source machine learning model, Joint Variational Autoencoders for multimodal Imputation and Embedding (JAMIE). JAMIE takes single-cell multimodal data that can have partially matched samples across modalities. Variational autoencoders learn the latent embeddings of each modality. Then, embeddings from matched samples across modalities are aggregated to identify joint cross-modal latent embeddings before reconstruction. To perform cross-modal imputation, the latent embeddings of one modality can be used with the decoder of the other modality. For interpretability, Shapley values are used to prioritize input features for cross-modal imputation and known sample labels. We applied JAMIE to both simulation data and emerging single-cell multimodal data including gene expression, chromatin accessibility, and electrophysiology in human and mouse brains. JAMIE significantly outperforms existing state-of-the-art methods in general and prioritized multimodal features for imputation, providing potentially novel mechanistic insights at cellular resolution.
Collapse
Affiliation(s)
- Noah Cohen Kalafut
- Department of Computer Sciences, Wisconsin, US
- Waisman Center, University of Wisconsin-Madison, Wisconsin, US
| | - Xiang Huang
- Waisman Center, University of Wisconsin-Madison, Wisconsin, US
| | - Daifeng Wang
- Department of Computer Sciences, Wisconsin, US
- Waisman Center, University of Wisconsin-Madison, Wisconsin, US
- Department of Biostatistics and Medical Informatics, Wisconsin, US
| |
Collapse
|
8
|
Chen KJ, Yoshimura R, Edmundo CA, Truong TM, Civelli O, Alachkar A, Abbott GW. Behavioral and neuro-functional consequences of eliminating the KCNQ3 GABA binding site in mice. Front Mol Neurosci 2023; 16:1192628. [PMID: 37305551 PMCID: PMC10248464 DOI: 10.3389/fnmol.2023.1192628] [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: 03/23/2023] [Accepted: 05/09/2023] [Indexed: 06/13/2023] Open
Abstract
Voltage-gated potassium (Kv) channels formed by α subunits KCNQ2-5 are important in regulating neuronal excitability. We previously found that GABA directly binds to and activates channels containing KCNQ3, challenging the traditional understanding of inhibitory neurotransmission. To investigate the functional significance and behavioral role of this direct interaction, mice with a mutated KCNQ3 GABA binding site (Kcnq3-W266L) were generated and subjected to behavioral studies. Kcnq3-W266L mice exhibited distinctive behavioral phenotypes, of which reduced nociceptive and stress responses were profound and sex-specific. In female Kcnq3-W266L mice, the phenotype was shifted towards more nociceptive effects, while in male Kcnq3-W266L mice, it was shifted towards the stress response. In addition, female Kcnq3-W266L mice exhibited lower motor activity and reduced working spatial memory. The neuronal activity in the lateral habenula and visual cortex was altered in the female Kcnq3-W266L mice, suggesting that GABAergic activation of KCNQ3 in these regions may play a role in the regulation of the responses. Given the known overlap between the nociceptive and stress brain circuits, our data provide new insights into a sex-dependent role of KCNQ3 in regulating neural circuits involved in nociception and stress, via its GABA binding site. These findings identify new targets for effective treatments for neurological and psychiatric conditions such as pain and anxiety.
Collapse
Affiliation(s)
- Kiki J. Chen
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University of California, Irvine, Irvine, CA, United States
| | - Ryan Yoshimura
- Bioelectricity Laboratory, Department of Physiology and Biophysics, School of Medicine, University of California, Irvine, Irvine, CA, United States
| | - Clarissa Adriana Edmundo
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University of California, Irvine, Irvine, CA, United States
| | - Tri Minh Truong
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University of California, Irvine, Irvine, CA, United States
| | - Olivier Civelli
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University of California, Irvine, Irvine, CA, United States
| | - Amal Alachkar
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University of California, Irvine, Irvine, CA, United States
- UC Irvine Center for the Neurobiology of Learning and Memory, University of California, Irvine, Irvine, CA, United States
- Institute for Genomics and Bioinformatics, School of Information and Computer Sciences, University of California, Irvine, Irvine, CA, United States
| | - Geoffrey W. Abbott
- Bioelectricity Laboratory, Department of Physiology and Biophysics, School of Medicine, University of California, Irvine, Irvine, CA, United States
| |
Collapse
|
9
|
Zhang L, Zhang Y, Jiang X, Mao L, Xia Y, Fan Y, Li N, Jiang Z, Qin X, Jiang Y, Liu G, Qiu F, Zhang J, Zou Z, Chen C. Disruption of the lung-gut-brain axis is responsible for cortex damage induced by pulmonary exposure to zinc oxide nanoparticles. Toxicology 2023; 485:153390. [PMID: 36535435 DOI: 10.1016/j.tox.2022.153390] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Revised: 12/02/2022] [Accepted: 12/03/2022] [Indexed: 12/23/2022]
Abstract
Increasing evidence shows that gut microbiota is important for host health in response to metal nanomaterials exposure. However, the effect of gut microbiota on the cortex damage caused by pulmonary exposure to zinc oxide nanoparticles (ZnONPs) remains mainly unknown. In this study, a total of 48 adult C57BL/6J mice were intratracheally instilled with 0.6 mg/kg ZnONPs in the presence or absence of antibiotics (ABX) treatment. Besides, 24 mice were treated with or without fecal microbiota transplantation (FMT) after the intraperitoneal administration of ABX. Our results demonstrated for the first time that dysbiosis induced by ABX treatment significantly aggravated cortex damage induced by pulmonary exposure to ZnONPs. Such damage might highly occur through the induction of oxidative stress, manifested by the enhancement of antioxidative enzymes and products of lipid peroxidation. However, ferroptosis was not involved in this process. Interestingly, our data revealed that ABX treatment exacerbated the alterations of gut-brain peptides (including Sst, Sstr2, and Htr4) induced by ZnONPs in both gut and cortex tissues. Moreover, fecal microbiota transplantation (FMT) was able to alleviate cerebral cortex damage, oxidative stress, and alterations of gut-brain peptides induced by pulmonary exposure to ZnONPs. The results together indicate that pulmonary exposure to ZnONPs causes cerebral cortex damage possibly via the disruption of the lung-gut-brain axis. These findings not only propose valuable insights into the mechanism of ZnONPs neurotoxicity but also provide a potential therapeutic method against brain disorders induced by pulmonary exposure to ZnONPs. AVAILABILITY OF DATA AND MATERIALS: The datasets used and/or analyzed during the current study are available from the The corresponding author on reasonable request.
Collapse
Affiliation(s)
- Lingbing Zhang
- Department of Pharmacy, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, PR China
| | - Yandan Zhang
- Molecular Biology Laboratory of Respiratory Disease, Institute of Life Sciences, Chongqing Medical University, Chongqing 400016, PR China
| | - Xuejun Jiang
- Research Center for Environment and Human Health, School of Public Health, Chongqing Medical University, Chongqing, 400016, PR China; Center of Experimental Teaching for Public Health, Experimental Teaching and Management Center, Chongqing Medical University, Chongqing 400016, PR China
| | - Lejiao Mao
- Molecular Biology Laboratory of Respiratory Disease, Institute of Life Sciences, Chongqing Medical University, Chongqing 400016, PR China; Research Center for Environment and Human Health, School of Public Health, Chongqing Medical University, Chongqing, 400016, PR China
| | - Yinyin Xia
- Department of Occupational and Environmental Health, School of Public Health, Chongqing Medical University, Chongqing 400016, PR China
| | - Yinzhen Fan
- Molecular Biology Laboratory of Respiratory Disease, Institute of Life Sciences, Chongqing Medical University, Chongqing 400016, PR China
| | - Na Li
- Molecular Biology Laboratory of Respiratory Disease, Institute of Life Sciences, Chongqing Medical University, Chongqing 400016, PR China
| | - Ziqi Jiang
- Molecular Biology Laboratory of Respiratory Disease, Institute of Life Sciences, Chongqing Medical University, Chongqing 400016, PR China
| | - Xia Qin
- Department of Pharmacy, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, PR China
| | - Yu Jiang
- Department of Respiratory Medicine, The University‑Town Affiliated Hospital of Chongqing Medical University, Chongqing 401331, PR China
| | - Gang Liu
- Department of Emergency, The University‑Town Affiliated Hospital of Chongqing Medical University, Chongqing 401331, PR China
| | - Feng Qiu
- Department of Pharmacy, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, PR China
| | - Jun Zhang
- Molecular Biology Laboratory of Respiratory Disease, Institute of Life Sciences, Chongqing Medical University, Chongqing 400016, PR China; Research Center for Environment and Human Health, School of Public Health, Chongqing Medical University, Chongqing, 400016, PR China.
| | - Zhen Zou
- Molecular Biology Laboratory of Respiratory Disease, Institute of Life Sciences, Chongqing Medical University, Chongqing 400016, PR China; Research Center for Environment and Human Health, School of Public Health, Chongqing Medical University, Chongqing, 400016, PR China.
| | - Chengzhi Chen
- Research Center for Environment and Human Health, School of Public Health, Chongqing Medical University, Chongqing, 400016, PR China; Department of Occupational and Environmental Health, School of Public Health, Chongqing Medical University, Chongqing 400016, PR China.
| |
Collapse
|
10
|
Zhou W, Ke S, Li W, Yuan J, Li X, Jin R, Jia X, Jiang T, Dai Z, He G, Fang Z, Shi L, Zhang Q, Gong H, Luo Q, Sun W, Li A, Li P. Mapping the Function of Whole-Brain Projection at the Single Neuron Level. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2202553. [PMID: 36228099 PMCID: PMC9685445 DOI: 10.1002/advs.202202553] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Revised: 09/18/2022] [Indexed: 06/16/2023]
Abstract
Axonal projection conveys neural information. The divergent and diverse projections of individual neurons imply the complexity of information flow. It is necessary to investigate the relationship between the projection and functional information at the single neuron level for understanding the rules of neural circuit assembly, but a gap remains due to a lack of methods to map the function to whole-brain projection. Here an approach is developed to bridge two-photon calcium imaging in vivo with high-resolution whole-brain imaging based on sparse labeling with the genetically encoded calcium indicator GCaMP6. Reliable whole-brain projections are captured by the high-definition fluorescent micro-optical sectioning tomography (HD-fMOST). A cross-modality cell matching is performed and the functional annotation of whole-brain projection at the single-neuron level (FAWPS) is obtained. Applying it to the layer 2/3 (L2/3) neurons in mouse visual cortex, the relationship is investigated between functional preferences and axonal projection features. The functional preference of projection motifs and the correlation between axonal length in MOs and neuronal orientation selectivity, suggest that projection motif-defined neurons form a functionally specific information flow, and the projection strength in specific targets relates to the information clarity. This pipeline provides a new way to understand the principle of neuronal information transmission.
Collapse
Affiliation(s)
- Wei Zhou
- Britton Chance Center and MoE Key Laboratory for Biomedical PhotonicsWuhan National Laboratory for OptoelectronicsHuazhong University of Science and TechnologyWuhan430074China
- Research Unit of Multimodal Cross Scale Neural Signal Detection and ImagingChinese Academy of Medical SciencesHUST‐Suzhou Institute for BrainsmaticsJITRISuzhou215100China
| | - Shanshan Ke
- Britton Chance Center and MoE Key Laboratory for Biomedical PhotonicsWuhan National Laboratory for OptoelectronicsHuazhong University of Science and TechnologyWuhan430074China
- Research Unit of Multimodal Cross Scale Neural Signal Detection and ImagingChinese Academy of Medical SciencesHUST‐Suzhou Institute for BrainsmaticsJITRISuzhou215100China
| | - Wenwei Li
- Britton Chance Center and MoE Key Laboratory for Biomedical PhotonicsWuhan National Laboratory for OptoelectronicsHuazhong University of Science and TechnologyWuhan430074China
- Research Unit of Multimodal Cross Scale Neural Signal Detection and ImagingChinese Academy of Medical SciencesHUST‐Suzhou Institute for BrainsmaticsJITRISuzhou215100China
| | - Jing Yuan
- Britton Chance Center and MoE Key Laboratory for Biomedical PhotonicsWuhan National Laboratory for OptoelectronicsHuazhong University of Science and TechnologyWuhan430074China
- Research Unit of Multimodal Cross Scale Neural Signal Detection and ImagingChinese Academy of Medical SciencesHUST‐Suzhou Institute for BrainsmaticsJITRISuzhou215100China
| | - Xiangning Li
- Britton Chance Center and MoE Key Laboratory for Biomedical PhotonicsWuhan National Laboratory for OptoelectronicsHuazhong University of Science and TechnologyWuhan430074China
- Research Unit of Multimodal Cross Scale Neural Signal Detection and ImagingChinese Academy of Medical SciencesHUST‐Suzhou Institute for BrainsmaticsJITRISuzhou215100China
| | - Rui Jin
- Britton Chance Center and MoE Key Laboratory for Biomedical PhotonicsWuhan National Laboratory for OptoelectronicsHuazhong University of Science and TechnologyWuhan430074China
- Research Unit of Multimodal Cross Scale Neural Signal Detection and ImagingChinese Academy of Medical SciencesHUST‐Suzhou Institute for BrainsmaticsJITRISuzhou215100China
| | - Xueyan Jia
- Research Unit of Multimodal Cross Scale Neural Signal Detection and ImagingChinese Academy of Medical SciencesHUST‐Suzhou Institute for BrainsmaticsJITRISuzhou215100China
| | - Tao Jiang
- Research Unit of Multimodal Cross Scale Neural Signal Detection and ImagingChinese Academy of Medical SciencesHUST‐Suzhou Institute for BrainsmaticsJITRISuzhou215100China
| | - Zimin Dai
- Britton Chance Center and MoE Key Laboratory for Biomedical PhotonicsWuhan National Laboratory for OptoelectronicsHuazhong University of Science and TechnologyWuhan430074China
- Research Unit of Multimodal Cross Scale Neural Signal Detection and ImagingChinese Academy of Medical SciencesHUST‐Suzhou Institute for BrainsmaticsJITRISuzhou215100China
| | - Guannan He
- Britton Chance Center and MoE Key Laboratory for Biomedical PhotonicsWuhan National Laboratory for OptoelectronicsHuazhong University of Science and TechnologyWuhan430074China
- Research Unit of Multimodal Cross Scale Neural Signal Detection and ImagingChinese Academy of Medical SciencesHUST‐Suzhou Institute for BrainsmaticsJITRISuzhou215100China
| | - Zhiwei Fang
- Britton Chance Center and MoE Key Laboratory for Biomedical PhotonicsWuhan National Laboratory for OptoelectronicsHuazhong University of Science and TechnologyWuhan430074China
- Research Unit of Multimodal Cross Scale Neural Signal Detection and ImagingChinese Academy of Medical SciencesHUST‐Suzhou Institute for BrainsmaticsJITRISuzhou215100China
| | - Liang Shi
- Britton Chance Center and MoE Key Laboratory for Biomedical PhotonicsWuhan National Laboratory for OptoelectronicsHuazhong University of Science and TechnologyWuhan430074China
- Research Unit of Multimodal Cross Scale Neural Signal Detection and ImagingChinese Academy of Medical SciencesHUST‐Suzhou Institute for BrainsmaticsJITRISuzhou215100China
| | - Qi Zhang
- Britton Chance Center and MoE Key Laboratory for Biomedical PhotonicsWuhan National Laboratory for OptoelectronicsHuazhong University of Science and TechnologyWuhan430074China
| | - Hui Gong
- Britton Chance Center and MoE Key Laboratory for Biomedical PhotonicsWuhan National Laboratory for OptoelectronicsHuazhong University of Science and TechnologyWuhan430074China
- Research Unit of Multimodal Cross Scale Neural Signal Detection and ImagingChinese Academy of Medical SciencesHUST‐Suzhou Institute for BrainsmaticsJITRISuzhou215100China
| | - Qingming Luo
- Key Laboratory of Biomedical Engineering of Hainan Province, School of Biomedical EngineeringHainan UniversityHaikou570228China
| | - Wenzhi Sun
- Chinese Institute for Brain ResearchBeijing102206China
- School of Basic Medical SciencesCapital Medical UniversityBeijing100069China
| | - Anan Li
- Britton Chance Center and MoE Key Laboratory for Biomedical PhotonicsWuhan National Laboratory for OptoelectronicsHuazhong University of Science and TechnologyWuhan430074China
- Research Unit of Multimodal Cross Scale Neural Signal Detection and ImagingChinese Academy of Medical SciencesHUST‐Suzhou Institute for BrainsmaticsJITRISuzhou215100China
| | - Pengcheng Li
- Britton Chance Center and MoE Key Laboratory for Biomedical PhotonicsWuhan National Laboratory for OptoelectronicsHuazhong University of Science and TechnologyWuhan430074China
- Research Unit of Multimodal Cross Scale Neural Signal Detection and ImagingChinese Academy of Medical SciencesHUST‐Suzhou Institute for BrainsmaticsJITRISuzhou215100China
| |
Collapse
|
11
|
Casello SM, Flores RJ, Yarur HE, Wang H, Awanyai M, Arenivar MA, Jaime-Lara RB, Bravo-Rivera H, Tejeda HA. Neuropeptide System Regulation of Prefrontal Cortex Circuitry: Implications for Neuropsychiatric Disorders. Front Neural Circuits 2022; 16:796443. [PMID: 35800635 PMCID: PMC9255232 DOI: 10.3389/fncir.2022.796443] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2021] [Accepted: 04/27/2022] [Indexed: 01/08/2023] Open
Abstract
Neuropeptides, a diverse class of signaling molecules in the nervous system, modulate various biological effects including membrane excitability, synaptic transmission and synaptogenesis, gene expression, and glial cell architecture and function. To date, most of what is known about neuropeptide action is limited to subcortical brain structures and tissue outside of the central nervous system. Thus, there is a knowledge gap in our understanding of neuropeptide function within cortical circuits. In this review, we provide a comprehensive overview of various families of neuropeptides and their cognate receptors that are expressed in the prefrontal cortex (PFC). Specifically, we highlight dynorphin, enkephalin, corticotropin-releasing factor, cholecystokinin, somatostatin, neuropeptide Y, and vasoactive intestinal peptide. Further, we review the implication of neuropeptide signaling in prefrontal cortical circuit function and use as potential therapeutic targets. Together, this review summarizes established knowledge and highlights unknowns of neuropeptide modulation of neural function underlying various biological effects while offering insights for future research. An increased emphasis in this area of study is necessary to elucidate basic principles of the diverse signaling molecules used in cortical circuits beyond fast excitatory and inhibitory transmitters as well as consider components of neuropeptide action in the PFC as a potential therapeutic target for neurological disorders. Therefore, this review not only sheds light on the importance of cortical neuropeptide studies, but also provides a comprehensive overview of neuropeptide action in the PFC to serve as a roadmap for future studies in this field.
Collapse
Affiliation(s)
- Sanne M. Casello
- Unit on Neuromodulation and Synaptic Integration, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, United States
| | - Rodolfo J. Flores
- Unit on Neuromodulation and Synaptic Integration, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, United States
| | - Hector E. Yarur
- Unit on Neuromodulation and Synaptic Integration, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, United States
| | - Huikun Wang
- Unit on Neuromodulation and Synaptic Integration, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, United States
| | - Monique Awanyai
- Unit on Neuromodulation and Synaptic Integration, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, United States
| | - Miguel A. Arenivar
- Unit on Neuromodulation and Synaptic Integration, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, United States
| | - Rosario B. Jaime-Lara
- Unit on Neuromodulation and Synaptic Integration, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, United States
- National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD, United States
| | - Hector Bravo-Rivera
- Unit on Neuromodulation and Synaptic Integration, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, United States
| | - Hugo A. Tejeda
- Unit on Neuromodulation and Synaptic Integration, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, United States
- *Correspondence: Hugo A. Tejeda,
| |
Collapse
|
12
|
Heo Y, Yoon E, Jeon YE, Yun JH, Ishimoto N, Woo H, Park SY, Song JJ, Lee W. Cryo-EM structure of the human somatostatin receptor 2 complex with its agonist somatostatin delineates the ligand binding specificity. eLife 2022; 11:76823. [PMID: 35446253 PMCID: PMC9054131 DOI: 10.7554/elife.76823] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Accepted: 04/20/2022] [Indexed: 11/18/2022] Open
Abstract
Somatostatin is a peptide hormone that regulates endocrine systems by binding to G-protein-coupled somatostatin receptors. Somatostatin receptor 2 (SSTR2) is a human somatostatin receptor and is highly implicated in hormone disorders, cancers, and neurological diseases. Here, we report the high-resolution cryo-EM structure of full-length human SSTR2 bound to the agonist somatostatin (SST-14) in complex with inhibitory G (Gi) proteins. Our structural and mutagenesis analyses show that seven transmembrane helices form a deep pocket for ligand binding and that SSTR2 recognizes the highly conserved Trp-Lys motif of SST-14 at the bottom of the pocket. Furthermore, our sequence analysis combined with AlphaFold modeled structures of other SSTR isoforms provide a structural basis for the mechanism by which SSTR family proteins specifically interact with their cognate ligands. This work provides the first glimpse into the molecular recognition mechanism of somatostatin receptors and a crucial resource to develop therapeutics targeting somatostatin receptors.
Collapse
Affiliation(s)
- Yunseok Heo
- Department of Biochemistry, Yonsei University, Seoul, Republic of Korea
| | - Eojin Yoon
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon, Republic of Korea
| | - Ye-Eun Jeon
- Department of Biochemistry, Yonsei University, Seoul, Republic of Korea
| | - Ji-Hye Yun
- Department of Biochemistry, Yonsei University, Seoul, Republic of Korea
| | - Naito Ishimoto
- Drug Design Laboratory, Yokohama City University, Yokohama, Japan
| | - Hyeonuk Woo
- Department of Chemistry, Seoul National University, Seoul, Republic of Korea
| | - Sam-Yong Park
- Drug Design Laboratory, Yokohama City University, Yokohama, Japan
| | - Ji-Joon Song
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon, Republic of Korea
| | - Weontae Lee
- Department of Biochemistry, Yonsei University, Seoul, Republic of Korea
| |
Collapse
|
13
|
Astrocytes Modulate Somatostatin Interneuron Signaling in the Visual Cortex. Cells 2022; 11:cells11091400. [PMID: 35563706 PMCID: PMC9102536 DOI: 10.3390/cells11091400] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Revised: 04/14/2022] [Accepted: 04/18/2022] [Indexed: 02/05/2023] Open
Abstract
At glutamatergic synapses, astrocytes respond to the neurotransmitter glutamate with intracellular Ca2+ elevations and the release of gliotransmitters that modulate synaptic transmission. While the functional interactions between neurons and astrocytes have been intensively studied at glutamatergic synapses, the role of astrocytes at GABAergic synapses has been less investigated. In the present study, we combine optogenetics with 2-photon Ca2+ imaging experiments and patch-clamp recording techniques to investigate the signaling between Somatostatin (SST)-releasing GABAergic interneurons and astrocytes in brain slice preparations from the visual cortex (VCx). We found that an intense stimulation of SST interneurons evokes Ca2+ elevations in astrocytes that fundamentally depend on GABAB receptor (GABABR) activation, and that this astrocyte response is modulated by the neuropeptide somatostatin. After episodes of SST interneuron hyperactivity, we also observed a long-lasting reduction of the inhibitory postsynaptic current (IPSC) amplitude onto pyramidal neurons (PNs). This reduction of inhibitory tone (i.e., disinhibition) is counterbalanced by the activation of astrocytes that upregulate SST interneuron-evoked IPSC amplitude by releasing ATP that, after conversion to adenosine, activates A1Rs. Our results describe a hitherto unidentified modulatory mechanism of inhibitory transmission to VCx layer II/III PNs that involves the functional recruitment of astrocytes by SST interneuron signaling.
Collapse
|
14
|
Urrutia-Piñones J, Morales-Moraga C, Sanguinetti-González N, Escobar AP, Chiu CQ. Long-Range GABAergic Projections of Cortical Origin in Brain Function. Front Syst Neurosci 2022; 16:841869. [PMID: 35392440 PMCID: PMC8981584 DOI: 10.3389/fnsys.2022.841869] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Accepted: 02/10/2022] [Indexed: 12/12/2022] Open
Abstract
The study of long-range GABAergic projections has traditionally been focused on those with subcortical origin. In the last few years, cortical GABAergic neurons have been shown to not only mediate local inhibition, but also extend long-range axons to remote cortical and subcortical areas. In this review, we delineate the different types of long-range GABAergic neurons (LRGNs) that have been reported to arise from the hippocampus and neocortex, paying attention to the anatomical and functional circuits they form to understand their role in behavior. Although cortical LRGNs are similar to their interneuron and subcortical counterparts, they comprise distinct populations that show specific patterns of cortico-cortical and cortico-fugal connectivity. Functionally, cortical LRGNs likely induce timed disinhibition in target regions to synchronize network activity. Thus, LRGNs are emerging as a new element of cortical output, acting in concert with long-range excitatory projections to shape brain function in health and disease.
Collapse
Affiliation(s)
- Jocelyn Urrutia-Piñones
- Ph.D. Program in Neuroscience, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile
- Facultad de Ciencias, Instituto de Neurociencia, Universidad de Valparaíso, Valparaíso, Chile
| | - Camila Morales-Moraga
- Facultad de Ciencias, Instituto de Neurociencia, Universidad de Valparaíso, Valparaíso, Chile
- Centro Interdisciplinario de Neurociencia de Valparaíso, Universidad de Valparaíso, Valparaíso, Chile
| | - Nicole Sanguinetti-González
- Ph.D. Program in Neuroscience, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile
- Facultad de Ciencias, Instituto de Neurociencia, Universidad de Valparaíso, Valparaíso, Chile
| | - Angelica P. Escobar
- Instituto de Fisiología, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile
- Centro de Neurobiología y Fisiopatología Integrativa, Universidad de Valparaíso, Valparaíso, Chile
| | - Chiayu Q. Chiu
- Facultad de Ciencias, Instituto de Neurociencia, Universidad de Valparaíso, Valparaíso, Chile
- Centro Interdisciplinario de Neurociencia de Valparaíso, Universidad de Valparaíso, Valparaíso, Chile
| |
Collapse
|
15
|
Weiler S, Guggiana Nilo D, Bonhoeffer T, Hübener M, Rose T, Scheuss V. Orientation and direction tuning align with dendritic morphology and spatial connectivity in mouse visual cortex. Curr Biol 2022; 32:1743-1753.e7. [PMID: 35276098 DOI: 10.1016/j.cub.2022.02.048] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Revised: 12/06/2021] [Accepted: 02/15/2022] [Indexed: 01/21/2023]
Abstract
The functional properties of neocortical pyramidal cells (PCs), such as direction and orientation selectivity in visual cortex, predominantly derive from their excitatory and inhibitory inputs. For layer 2/3 (L2/3) PCs, the detailed relationship between their functional properties and how they sample and integrate information across cortical space is not fully understood. Here, we study this relationship by combining functional in vivo two-photon calcium imaging, in vitro functional circuit mapping, and dendritic reconstruction of the same L2/3 PCs in mouse visual cortex. Our work reveals direct correlations between dendritic morphology and functional input connectivity and the orientation as well as direction tuning of L2/3 PCs. First, the apical dendritic tree is elongated along the postsynaptic preferred orientation, considering the representation of visual space in the cortex as determined by its retinotopic organization. Additionally, sharply orientation-tuned cells show a less complex apical tree compared with broadly tuned cells. Second, in direction-selective L2/3 PCs, the spatial distribution of presynaptic partners is offset from the soma opposite to the preferred direction. Importantly, although the presynaptic excitatory and inhibitory input distributions spatially overlap on average, the excitatory input distribution is spatially skewed along the preferred direction, in contrast to the inhibitory distribution. Finally, the degree of asymmetry is positively correlated with the direction selectivity of the postsynaptic L2/3 PC. These results show that the dendritic architecture and the spatial arrangement of excitatory and inhibitory presynaptic cells of L2/3 PCs play important roles in shaping their orientation and direction tuning.
Collapse
Affiliation(s)
- Simon Weiler
- Max Planck Institute of Neurobiology, Martinsried, Germany; Graduate School of Systemic Neurosciences, Ludwig-Maximilians-Universität München, Großhaderner Str. 2, 82152 Planegg, Germany
| | | | | | - Mark Hübener
- Max Planck Institute of Neurobiology, Martinsried, Germany
| | - Tobias Rose
- Max Planck Institute of Neurobiology, Martinsried, Germany
| | - Volker Scheuss
- Max Planck Institute of Neurobiology, Martinsried, Germany; Department of Psychiatry, Ludwig-Maximilians-Universität München, Nussbaumstr. 7, 80336 München, Germany.
| |
Collapse
|
16
|
Somatostatin and Somatostatin-Containing Interneurons—From Plasticity to Pathology. Biomolecules 2022; 12:biom12020312. [PMID: 35204812 PMCID: PMC8869243 DOI: 10.3390/biom12020312] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 01/27/2022] [Accepted: 02/11/2022] [Indexed: 01/27/2023] Open
Abstract
Despite the obvious differences in the pathophysiology of distinct neuropsychiatric diseases or neurodegenerative disorders, some of them share some general but pivotal mechanisms, one of which is the disruption of excitation/inhibition balance. Such an imbalance can be generated by changes in the inhibitory system, very often mediated by somatostatin-containing interneurons (SOM-INs). In physiology, this group of inhibitory interneurons, as well as somatostatin itself, profoundly shapes the brain activity, thus influencing the behavior and plasticity; however, the changes in the number, density and activity of SOM-INs or levels of somatostatin are found throughout many neuropsychiatric and neurological conditions, both in patients and animal models. Here, we (1) briefly describe the brain somatostatinergic system, characterizing the neuropeptide somatostatin itself, its receptors and functions, as well the physiology and circuitry of SOM-INs; and (2) summarize the effects of the activity of somatostatin and SOM-INs in both physiological brain processes and pathological brain conditions, focusing primarily on learning-induced plasticity and encompassing selected neuropsychological and neurodegenerative disorders, respectively. The presented data indicate the somatostatinergic-system-mediated inhibition as a substantial factor in the mechanisms of neuroplasticity, often disrupted in a plethora of brain pathologies.
Collapse
|
17
|
Kwon KM, Lee MJ, Chung HS, Pak JH, Jeon CJ. The Organization of Somatostatin-Immunoreactive Cells in the Visual Cortex of the Gerbil. Biomedicines 2022; 10:biomedicines10010092. [PMID: 35052772 PMCID: PMC8773527 DOI: 10.3390/biomedicines10010092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Revised: 12/29/2021] [Accepted: 12/31/2021] [Indexed: 11/16/2022] Open
Abstract
Somatostatin (SST) is widely expressed in the brain and plays various, vital roles involved in neuromodulation. The purpose of this study is to characterize the organization of SST neurons in the Mongolian gerbil visual cortex (VC) using immunocytochemistry, quantitative analysis, and confocal microscopy. As a diurnal animal, the Mongolian gerbil provides us with a different perspective to other commonly used nocturnal rodent models. In this study, SST neurons were located in all layers of the VC except in layer I; they were most common in layer V. Most SST neurons were multipolar round/oval or stellate cells. No pyramidal neurons were found. Moreover, 2-color immunofluorescence revealed that only 33.50%, 24.05%, 16.73%, 0%, and 64.57% of SST neurons contained gamma-aminobutyric acid, calbindin-D28K, calretinin, parvalbumin, and calcium/calmodulin-dependent protein kinase II, respectively. In contrast, neuropeptide Y and nitric oxide synthase were abundantly expressed, with 80.07% and 75.41% in SST neurons, respectively. Our immunocytochemical analyses of SST with D1 and D2 dopamine receptors and choline acetyltransferase, α7 and β2 nicotinic acetylcholine receptors suggest that dopaminergic and cholinergic fibers contact some SST neurons. The results showed some distinguishable features of SST neurons and provided some insight into their afferent circuitry in the gerbil VC. These findings may support future studies investigating the role of SST neurons in visual processing.
Collapse
Affiliation(s)
- Kyung-Min Kwon
- Department of Biology, School of Life Sciences, BK21 FOUR KNU Creative Bio-Research Group, College of Natural Sciences, Brain Science and Engineering Institute, Kyungpook National University, Daegu 41566, Korea; (K.-M.K.); (M.-J.L.)
- Research Institute for Dok-do and Ulleung-do Island, Department of Biology, School of Life Sciences, Kyungpook National University, 80 Daehak-ro, Buk-gu, Daegu 41566, Korea;
| | - Myung-Jun Lee
- Department of Biology, School of Life Sciences, BK21 FOUR KNU Creative Bio-Research Group, College of Natural Sciences, Brain Science and Engineering Institute, Kyungpook National University, Daegu 41566, Korea; (K.-M.K.); (M.-J.L.)
| | - Han-Saem Chung
- Department of Biology, School of Life Sciences, College of Natural Sciences, Kyungpook National University, Daegu 41566, Korea;
| | - Jae-Hong Pak
- Research Institute for Dok-do and Ulleung-do Island, Department of Biology, School of Life Sciences, Kyungpook National University, 80 Daehak-ro, Buk-gu, Daegu 41566, Korea;
- Department of Biology, School of Life Sciences, College of Natural Sciences, Kyungpook National University, Daegu 41566, Korea;
| | - Chang-Jin Jeon
- Department of Biology, School of Life Sciences, BK21 FOUR KNU Creative Bio-Research Group, College of Natural Sciences, Brain Science and Engineering Institute, Kyungpook National University, Daegu 41566, Korea; (K.-M.K.); (M.-J.L.)
- Research Institute for Dok-do and Ulleung-do Island, Department of Biology, School of Life Sciences, Kyungpook National University, 80 Daehak-ro, Buk-gu, Daegu 41566, Korea;
- Correspondence:
| |
Collapse
|
18
|
Rikhye RV, Yildirim M, Hu M, Breton-Provencher V, Sur M. Reliable Sensory Processing in Mouse Visual Cortex through Cooperative Interactions between Somatostatin and Parvalbumin Interneurons. J Neurosci 2021; 41:8761-8778. [PMID: 34493543 PMCID: PMC8528503 DOI: 10.1523/jneurosci.3176-20.2021] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 08/16/2021] [Accepted: 08/23/2021] [Indexed: 11/21/2022] Open
Abstract
Intrinsic neuronal variability significantly limits information encoding in the primary visual cortex (V1). However, under certain conditions, neurons can respond reliably with highly precise responses to the same visual stimuli from trial to trial. This suggests that there exists intrinsic neural circuit mechanisms that dynamically modulate the intertrial variability of visual cortical neurons. Here, we sought to elucidate the role of different inhibitory interneurons (INs) in reliable coding in mouse V1. To study the interactions between somatostatin-expressing interneurons (SST-INs) and parvalbumin-expressing interneurons (PV-INs), we used a dual-color calcium imaging technique that allowed us to simultaneously monitor these two neural ensembles while awake mice, of both sexes, passively viewed natural movies. SST neurons were more active during epochs of reliable pyramidal neuron firing, whereas PV neurons were more active during epochs of unreliable firing. SST neuron activity lagged that of PV neurons, consistent with a feedback inhibitory SST→PV circuit. To dissect the role of this circuit in pyramidal neuron activity, we used temporally limited optogenetic activation and inactivation of SST and PV interneurons during periods of reliable and unreliable pyramidal cell firing. Transient firing of SST neurons increased pyramidal neuron reliability by actively suppressing PV neurons, a proposal that was supported by a rate-based model of V1 neurons. These results identify a cooperative functional role for the SST→PV circuit in modulating the reliability of pyramidal neuron activity.SIGNIFICANCE STATEMENT Cortical neurons often respond to identical sensory stimuli with large variability. However, under certain conditions, the same neurons can also respond highly reliably. The circuit mechanisms that contribute to this modulation remain unknown. Here, we used novel dual-wavelength calcium imaging and temporally selective optical perturbation to identify an inhibitory neural circuit in visual cortex that can modulate the reliability of pyramidal neurons to naturalistic visual stimuli. Our results, supported by computational models, suggest that somatostatin interneurons increase pyramidal neuron reliability by suppressing parvalbumin interneurons via the inhibitory SST→PV circuit. These findings reveal a novel role of the SST→PV circuit in modulating the fidelity of neural coding critical for visual perception.
Collapse
Affiliation(s)
- Rajeev V Rikhye
- Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139
- Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139
| | - Murat Yildirim
- Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139
- Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139
| | - Ming Hu
- Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139
- Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139
| | - Vincent Breton-Provencher
- Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139
- Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139
| | - Mriganka Sur
- Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139
- Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139
| |
Collapse
|
19
|
Noise-induced properties of active dendrites. Proc Natl Acad Sci U S A 2021; 118:2023381118. [PMID: 34413187 DOI: 10.1073/pnas.2023381118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Dendrites play an essential role in the integration of highly fluctuating input in vivo into neurons across all nervous systems. Yet, they are often studied under conditions where inputs to dendrites are sparse. The dynamic properties of active dendrites facing in vivo-like fluctuating input thus remain elusive. In this paper, we uncover dynamics in a canonical model of a dendritic compartment with active calcium channels, receiving in vivo-like fluctuating input. In a single-compartment model of the active dendrite with fast calcium activation, we show noise-induced nonmonotonic behavior in the relationship of the membrane potential output, and mean input emerges. In contrast, noise can induce bistability in the input-output relation in the system with slowly activating calcium channels. Both phenomena are absent in a noiseless condition. Furthermore, we show that timescales of the emerging stochastic bistable dynamics extend far beyond a deterministic system due to stochastic switching between the solutions. A numerical simulation of a multicompartment model neuron shows that in the presence of in vivo-like synaptic input, the bistability uncovered in our analysis persists. Our results reveal that realistic synaptic input contributes to sustained dendritic nonlinearities, and synaptic noise is a significant component of dendritic input integration.
Collapse
|
20
|
Wiring of higher-order cortical areas: Spatiotemporal development of cortical hierarchy. Semin Cell Dev Biol 2021; 118:35-49. [PMID: 34034988 DOI: 10.1016/j.semcdb.2021.05.010] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Revised: 04/27/2021] [Accepted: 05/08/2021] [Indexed: 01/04/2023]
Abstract
A hierarchical development of cortical areas was suggested over a century ago, but the diversity and complexity of cortical hierarchy properties have so far prevented a formal demonstration. The aim of this review is to clarify the similarities and differences in the developmental processes underlying cortical development of primary and higher-order areas. We start by recapitulating the historical and recent advances underlying the biological principle of cortical hierarchy in adults. We then revisit the arguments for a hierarchical maturation of cortical areas, and further integrate the principles of cortical areas specification during embryonic and postnatal development. We highlight how the dramatic expansion in cortical size might have contributed to the increased number of association areas sustaining cognitive complexification in evolution. Finally, we summarize the recent observations of an alteration of cortical hierarchy in neuropsychiatric disorders and discuss their potential developmental origins.
Collapse
|
21
|
Song YH, Yoon J, Lee SH. The role of neuropeptide somatostatin in the brain and its application in treating neurological disorders. Exp Mol Med 2021; 53:328-338. [PMID: 33742131 PMCID: PMC8080805 DOI: 10.1038/s12276-021-00580-4] [Citation(s) in RCA: 52] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Revised: 01/23/2021] [Accepted: 01/25/2021] [Indexed: 02/07/2023] Open
Abstract
Somatostatin (SST) is a well-known neuropeptide that is expressed throughout the brain. In the cortex, SST is expressed in a subset of GABAergic neurons and is known as a protein marker of inhibitory interneurons. Recent studies have identified the key functions of SST in modulating cortical circuits in the brain and cognitive function. Furthermore, reduced expression of SST is a hallmark of various neurological disorders, including Alzheimer's disease and depression. In this review, we summarize the current knowledge on SST expression and function in the brain. In particular, we describe the physiological roles of SST-positive interneurons in the cortex. We further describe the causal relationship between pathophysiological changes in SST function and various neurological disorders, such as Alzheimer's disease. Finally, we discuss potential treatments and possibility of novel drug developments for neurological disorders based on the current knowledge on the function of SST and SST analogs in the brain derived from experimental and clinical studies.
Collapse
Affiliation(s)
- You-Hyang Song
- grid.37172.300000 0001 2292 0500Department of Biological Sciences, Korea Advanced Institute of Science and Technology, 291 Daehak-ro, Yuseong-gu, Daejeon 34141 Republic of Korea
| | - Jiwon Yoon
- grid.37172.300000 0001 2292 0500Department of Biological Sciences, Korea Advanced Institute of Science and Technology, 291 Daehak-ro, Yuseong-gu, Daejeon 34141 Republic of Korea
| | - Seung-Hee Lee
- grid.37172.300000 0001 2292 0500Department of Biological Sciences, Korea Advanced Institute of Science and Technology, 291 Daehak-ro, Yuseong-gu, Daejeon 34141 Republic of Korea
| |
Collapse
|
22
|
Fontinha BM, Zekoll T, Al-Rawi M, Gallach M, Reithofer F, Barker AJ, Hofbauer M, Fischer RM, von Haeseler A, Baier H, Tessmar-Raible K. TMT-Opsins differentially modulate medaka brain function in a context-dependent manner. PLoS Biol 2021; 19:e3001012. [PMID: 33411725 PMCID: PMC7837489 DOI: 10.1371/journal.pbio.3001012] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2019] [Revised: 01/26/2021] [Accepted: 12/10/2020] [Indexed: 12/13/2022] Open
Abstract
Vertebrate behavior is strongly influenced by light. Light receptors, encoded by functional opsin proteins, are present inside the vertebrate brain and peripheral tissues. This expression feature is present from fishes to human and appears to be particularly prominent in diurnal vertebrates. Despite their conserved widespread occurrence, the nonvisual functions of opsins are still largely enigmatic. This is even more apparent when considering the high number of opsins. Teleosts possess around 40 opsin genes, present from young developmental stages to adulthood. Many of these opsins have been shown to function as light receptors. This raises the question of whether this large number might mainly reflect functional redundancy or rather maximally enables teleosts to optimally use the complex light information present under water. We focus on tmt-opsin1b and tmt-opsin2, c-opsins with ancestral-type sequence features, conserved across several vertebrate phyla, expressed with partly similar expression in non-rod, non-cone, non-retinal-ganglion-cell brain tissues and with a similar spectral sensitivity. The characterization of the single mutants revealed age- and light-dependent behavioral changes, as well as an impact on the levels of the preprohormone sst1b and the voltage-gated sodium channel subunit scn12aa. The amount of daytime rest is affected independently of the eyes, pineal organ, and circadian clock in tmt-opsin1b mutants. We further focused on daytime behavior and the molecular changes in tmt-opsin1b/2 double mutants, and found that-despite their similar expression and spectral features-these opsins interact in part nonadditively. Specifically, double mutants complement molecular and behavioral phenotypes observed in single mutants in a partly age-dependent fashion. Our work provides a starting point to disentangle the highly complex interactions of vertebrate nonvisual opsins, suggesting that tmt-opsin-expressing cells together with other visual and nonvisual opsins provide detailed light information to the organism for behavioral fine-tuning. This work also provides a stepping stone to unravel how vertebrate species with conserved opsins, but living in different ecological niches, respond to similar light cues and how human-generated artificial light might impact on behavioral processes in natural environments.
Collapse
Affiliation(s)
- Bruno M. Fontinha
- Max F. Perutz Laboratories, University of Vienna, Vienna, Austria
- Research Platform ‘‘Rhythms of Life,” University of Vienna, Vienna, Austria
| | - Theresa Zekoll
- Max F. Perutz Laboratories, University of Vienna, Vienna, Austria
- Research Platform ‘‘Rhythms of Life,” University of Vienna, Vienna, Austria
| | - Mariam Al-Rawi
- Max F. Perutz Laboratories, University of Vienna, Vienna, Austria
- Research Platform ‘‘Rhythms of Life,” University of Vienna, Vienna, Austria
| | - Miguel Gallach
- Center for Integrative Bioinformatics Vienna, Max F. Perutz Laboratories, University of Vienna and Medical University of Vienna, Vienna, Austria
| | - Florian Reithofer
- Max F. Perutz Laboratories, University of Vienna, Vienna, Austria
- Research Platform ‘‘Rhythms of Life,” University of Vienna, Vienna, Austria
| | | | - Maximilian Hofbauer
- Max F. Perutz Laboratories, University of Vienna, Vienna, Austria
- Research Platform ‘‘Rhythms of Life,” University of Vienna, Vienna, Austria
- loopbio, Vienna, Austria
| | - Ruth M. Fischer
- Max F. Perutz Laboratories, University of Vienna, Vienna, Austria
| | - Arndt von Haeseler
- Max F. Perutz Laboratories, University of Vienna, Vienna, Austria
- Research Platform ‘‘Rhythms of Life,” University of Vienna, Vienna, Austria
- Center for Integrative Bioinformatics Vienna, Max F. Perutz Laboratories, University of Vienna and Medical University of Vienna, Vienna, Austria
- Bioinformatics and Computational Biology, Faculty of Computer Science, University of Vienna, Vienna, Austria
| | - Herwig Baier
- Max Planck Institute of Neurobiology, Martinsried, Germany
| | - Kristin Tessmar-Raible
- Max F. Perutz Laboratories, University of Vienna, Vienna, Austria
- Research Platform ‘‘Rhythms of Life,” University of Vienna, Vienna, Austria
- FENS-Kavli Network of Excellence, Brussels, Belgium
| |
Collapse
|
23
|
Dienel SJ, Ciesielski AJ, Bazmi HH, Profozich EA, Fish KN, Lewis DA. Distinct Laminar and Cellular Patterns of GABA Neuron Transcript Expression in Monkey Prefrontal and Visual Cortices. Cereb Cortex 2020; 31:2345-2363. [PMID: 33338196 DOI: 10.1093/cercor/bhaa341] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Revised: 10/21/2020] [Accepted: 10/21/2020] [Indexed: 12/25/2022] Open
Abstract
The functional output of a cortical region is shaped by its complement of GABA neuron subtypes. GABA-related transcript expression differs substantially between the primate dorsolateral prefrontal cortex (DLPFC) and primary visual (V1) cortices in gray matter homogenates, but the laminar and cellular bases for these differences are unknown. Quantification of levels of GABA-related transcripts in layers 2 and 4 of monkey DLPFC and V1 revealed three distinct expression patterns: 1) transcripts with higher levels in DLPFC and layer 2 [e.g., somatostatin (SST)]; 2) transcripts with higher levels in V1 and layer 4 [e.g., parvalbumin (PV)], and 3) transcripts with similar levels across layers and regions [e.g., glutamic acid decarboxylase (GAD67)]. At the cellular level, these patterns reflected transcript- and cell type-specific differences: the SST pattern primarily reflected differences in the relative proportions of SST mRNA-positive neurons, the PV pattern primarily reflected differences in PV mRNA expression per neuron, and the GAD67 pattern reflected opposed patterns in the relative proportions of GAD67 mRNA-positive neurons and in GAD67 mRNA expression per neuron. These findings suggest that differences in the complement of GABA neuron subtypes and in gene expression levels per neuron contribute to the specialization of inhibitory neurotransmission across cortical circuits.
Collapse
Affiliation(s)
- Samuel J Dienel
- Medical Scientist Training Program, University of Pittsburgh, Pittsburgh, PA 15213, USA.,Translational Neuroscience Program, Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15213, USA.,Center for the Neural Basis of Cognition, Carnegie Mellon University, Pittsburgh, PA 15213, USA.,Department of Neuroscience, Dietrich School of Arts and Sciences, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Andrew J Ciesielski
- Translational Neuroscience Program, Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Holly H Bazmi
- Translational Neuroscience Program, Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Elizabeth A Profozich
- Department of Neuroscience, Dietrich School of Arts and Sciences, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Kenneth N Fish
- Translational Neuroscience Program, Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - David A Lewis
- Translational Neuroscience Program, Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15213, USA.,Center for the Neural Basis of Cognition, Carnegie Mellon University, Pittsburgh, PA 15213, USA.,Department of Neuroscience, Dietrich School of Arts and Sciences, University of Pittsburgh, Pittsburgh, PA 15213, USA
| |
Collapse
|
24
|
Gallerani N, Au E. Loss of Clustered Protocadherin Diversity Alters the Spatial Distribution of Cortical Interneurons in Mice. Cereb Cortex Commun 2020; 1:tgaa089. [PMID: 34296145 PMCID: PMC8152951 DOI: 10.1093/texcom/tgaa089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Revised: 11/13/2020] [Accepted: 11/19/2020] [Indexed: 11/13/2022] Open
Abstract
Cortical interneurons (cINs) are locally projecting inhibitory neurons that are distributed throughout the cortex. Due to their relatively limited range of influence, their arrangement in the cortex is critical to their function. cINs achieve this arrangement through a process of tangential and radial migration and apoptosis during development. In this study, we investigated the role of clustered protocadherins (cPcdhs) in establishing the spatial patterning of cINs through the use of genetic cPcdh knockout mice. cPcdhs are expressed in cINs and are known to play key functions in cell spacing and cell survival, but their role in cINs is poorly understood. Using spatial statistical analysis, we found that the 2 main subclasses of cINs, parvalbumin-expressing and somatostatin-expressing (SST) cINs, are nonrandomly spaced within subclass but randomly with respect to each other. We also found that the relative laminar distribution of each subclass was distinctly altered in whole α- or β-cluster mutants. Examination of perinatal time points revealed that the mutant phenotypes emerged relatively late, suggesting that cPcdhs may be acting during cIN morphological elaboration and synaptogenesis. We then analyzed an isoform-specific knockout for pcdh-αc2 and found that it recapitulated the α-cluster knockout but only in SST cells, suggesting that subtype-specific expression of cPcdh isoforms may help govern subtype-specific spatial distribution.
Collapse
Affiliation(s)
- Nicholas Gallerani
- Department of Pathology & Cell Biology, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Edmund Au
- Department of Pathology & Cell Biology, Columbia University Irving Medical Center, New York, NY 10032, USA.,Department of Rehabilitative Medicine and Regeneration, Columbia University Irving Medical Center, New York, NY 10032, USA.,Columbia University Irving Medical Center, New York NY, 10032, USA
| |
Collapse
|