1
|
He Y, Wei Z, Xu J, Jin F, Li T, Qian L, Ma J, Zheng W, Javanmardi N, Wang T, Sun K, Feng ZQ. Genetics-Based Targeting Strategies for Precise Neuromodulation. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025:e13817. [PMID: 40387259 DOI: 10.1002/advs.202413817] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Revised: 01/10/2025] [Indexed: 05/20/2025]
Abstract
Genetics-based neuromodulation schemes are capable of selectively manipulating the activity of defined cell populations with high temporal-spatial resolution, providing unprecedented opportunities for probing cellular biological mechanisms, resolving neuronal projection pathways, mapping neural profiles, and precisely treating neurological and psychiatric disorders. Multimodal implementation schemes, which involve the use of exogenous stimuli such as light, heat, mechanical force, chemicals, electricity, and magnetic stimulation in combination with specific genetically engineered effectors, greatly expand their application space and scenarios. In particular, advanced wireless stimulation schemes have enabled low-invasive targeted neuromodulation through local delivery of navigable micro- and nanosized stimulators. In this review, the fundamental principles and implementation protocols of genetics-based precision neuromodulation are first introduced.The implementation schemes are systematically summarized, including optical, thermal, force, chemical, electrical, and magnetic stimulation, with an emphasis on those wireless and low-invasive strategies. Representative studies are dissected and analyzed for their advantages and disadvantages. Finally, the significance of genetics-based precision neuromodulation is emphasized and the open challenges and future perspectives are concluded.
Collapse
Affiliation(s)
- Yuyuan He
- School of Chemistry and Chemical Engineering, Nanjing University of Science and Technology, Nanjing, 210094, P.R. China
| | - Zhidong Wei
- School of Chemistry and Chemical Engineering, Nanjing University of Science and Technology, Nanjing, 210094, P.R. China
| | - Jianda Xu
- Department of Orthopedics, Changzhou Hospital of Traditional Chinese Medicine, Changzhou Hospital Affiliated to Nanjing University of Chinese Medicine, Changzhou, 213003, P. R. China
| | - Fei Jin
- School of Chemistry and Chemical Engineering, Nanjing University of Science and Technology, Nanjing, 210094, P.R. China
| | - Tong Li
- School of Chemistry and Chemical Engineering, Nanjing University of Science and Technology, Nanjing, 210094, P.R. China
| | - Lili Qian
- School of Chemistry and Chemical Engineering, Nanjing University of Science and Technology, Nanjing, 210094, P.R. China
| | - Juan Ma
- School of Chemistry and Chemical Engineering, Nanjing University of Science and Technology, Nanjing, 210094, P.R. China
| | - Weiying Zheng
- School of Chemistry and Chemical Engineering, Nanjing University of Science and Technology, Nanjing, 210094, P.R. China
| | - Negar Javanmardi
- School of Chemistry and Chemical Engineering, Nanjing University of Science and Technology, Nanjing, 210094, P.R. China
| | - Ting Wang
- State Key Laboratory of Digital Medical Engineering, Southeast University, Nanjing, 210096, P.R. China
| | - Kangjian Sun
- The Fourth Affiliated Hospital of Nanjing Medical University, Nanjing, 210031, P. R. China
| | - Zhang-Qi Feng
- School of Chemistry and Chemical Engineering, Nanjing University of Science and Technology, Nanjing, 210094, P.R. China
| |
Collapse
|
2
|
Beau M, Herzfeld DJ, Naveros F, Hemelt ME, D'Agostino F, Oostland M, Sánchez-López A, Chung YY, Maibach M, Kyranakis S, Stabb HN, Martínez Lopera MG, Lajko A, Zedler M, Ohmae S, Hall NJ, Clark BA, Cohen D, Lisberger SG, Kostadinov D, Hull C, Häusser M, Medina JF. A deep learning strategy to identify cell types across species from high-density extracellular recordings. Cell 2025; 188:2218-2234.e22. [PMID: 40023155 DOI: 10.1016/j.cell.2025.01.041] [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: 05/16/2024] [Revised: 11/20/2024] [Accepted: 01/28/2025] [Indexed: 03/04/2025]
Abstract
High-density probes allow electrophysiological recordings from many neurons simultaneously across entire brain circuits but fail to reveal cell type. Here, we develop a strategy to identify cell types from extracellular recordings in awake animals and reveal the computational roles of neurons with distinct functional, molecular, and anatomical properties. We combine optogenetics and pharmacology using the cerebellum as a testbed to generate a curated ground-truth library of electrophysiological properties for Purkinje cells, molecular layer interneurons, Golgi cells, and mossy fibers. We train a semi-supervised deep learning classifier that predicts cell types with greater than 95% accuracy based on the waveform, discharge statistics, and layer of the recorded neuron. The classifier's predictions agree with expert classification on recordings using different probes, in different laboratories, from functionally distinct cerebellar regions, and across species. Our classifier extends the power of modern dynamical systems analyses by revealing the unique contributions of simultaneously recorded cell types during behavior.
Collapse
Affiliation(s)
- Maxime Beau
- Wolfson Institute for Biomedical Research, University College London, London, UK
| | - David J Herzfeld
- Department of Neurobiology, Duke University School of Medicine, Durham, NC, USA
| | - Francisco Naveros
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA; Department of Computer Engineering, Automation and Robotics, Research Centre for Information and Communication Technologies, University of Granada, Granada, Spain
| | - Marie E Hemelt
- Department of Neurobiology, Duke University School of Medicine, Durham, NC, USA
| | - Federico D'Agostino
- Wolfson Institute for Biomedical Research, University College London, London, UK
| | - Marlies Oostland
- Wolfson Institute for Biomedical Research, University College London, London, UK; Swammerdam Institute for Life Sciences, University of Amsterdam, Amsterdam, the Netherlands
| | | | - Young Yoon Chung
- Wolfson Institute for Biomedical Research, University College London, London, UK
| | - Michael Maibach
- Wolfson Institute for Biomedical Research, University College London, London, UK
| | - Stephen Kyranakis
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA
| | - Hannah N Stabb
- Wolfson Institute for Biomedical Research, University College London, London, UK
| | | | - Agoston Lajko
- Wolfson Institute for Biomedical Research, University College London, London, UK
| | - Marie Zedler
- Wolfson Institute for Biomedical Research, University College London, London, UK
| | - Shogo Ohmae
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA
| | - Nathan J Hall
- Department of Neurobiology, Duke University School of Medicine, Durham, NC, USA
| | - Beverley A Clark
- Wolfson Institute for Biomedical Research, University College London, London, UK
| | - Dana Cohen
- The Leslie and Susan Gonda Multidisciplinary Brain Research Center, Bar-Ilan University, Ramat-Gan, Israel
| | - Stephen G Lisberger
- Department of Neurobiology, Duke University School of Medicine, Durham, NC, USA
| | - Dimitar Kostadinov
- Wolfson Institute for Biomedical Research, University College London, London, UK; Centre for Developmental Neurobiology, King's College London, London, UK
| | - Court Hull
- Department of Neurobiology, Duke University School of Medicine, Durham, NC, USA
| | - Michael Häusser
- Wolfson Institute for Biomedical Research, University College London, London, UK; School of Biomedical Sciences, The University of Hong Kong, Hong Kong, China
| | - Javier F Medina
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA.
| |
Collapse
|
3
|
Cording KR, Tu EM, Wang H, Agopyan-Miu AHCW, Bateup HS. Cntnap2 loss drives striatal neuron hyperexcitability and behavioral inflexibility. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.05.09.593387. [PMID: 38766169 PMCID: PMC11100810 DOI: 10.1101/2024.05.09.593387] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2024]
Abstract
Autism spectrum disorder (ASD) is a neurodevelopmental disorder characterized by two major diagnostic criteria - persistent deficits in social communication and interaction, and the presence of restricted, repetitive patterns of behavior (RRBs). Evidence from both human and animal model studies of ASD suggest that alteration of striatal circuits, which mediate motor learning, action selection, and habit formation, may contribute to the manifestation of RRBs. CNTNAP2 is a syndromic ASD risk gene, and loss of function of Cntnap2 in mice is associated with RRBs. How loss of Cntnap2 impacts striatal neuron function is largely unknown. In this study, we utilized Cntnap2 -/- mice to test whether altered striatal neuron activity contributes to aberrant motor behaviors relevant to ASD. We find that Cntnap2 -/- mice exhibit increased cortical drive of direct pathway striatal projection neurons (dSPNs). This enhanced drive is likely due to increased intrinsic excitability of dSPNs, which make them more responsive to cortical inputs. We find that Cntnap2 -/- mice exhibit spontaneous repetitive behaviors, increased motor routine learning, perseveration, and cognitive inflexibility. Increased corticostriatal drive of the direct pathway may therefore contribute to the acquisition of repetitive, inflexible behaviors in Cntnap2 mice.
Collapse
Affiliation(s)
- Katherine R. Cording
- Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, CA USA
- Department of Neuroscience, University of California, Berkeley, Berkeley, CA USA
| | - Emilie M. Tu
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA USA
| | - Hongli Wang
- Department of Neuroscience, University of California, Berkeley, Berkeley, CA USA
| | | | - Helen S. Bateup
- Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, CA USA
- Department of Neuroscience, University of California, Berkeley, Berkeley, CA USA
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA USA
| |
Collapse
|
4
|
Zhou W, Jia L, Yue L, Hu L. Advances and applications of peripheral optogenetics in animal models. Neuroscience 2025; 567:163-171. [PMID: 39765287 DOI: 10.1016/j.neuroscience.2025.01.009] [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: 08/19/2024] [Revised: 12/27/2024] [Accepted: 01/04/2025] [Indexed: 01/13/2025]
Abstract
Peripheral optogenetics is an emerging neuromodulation technique that regulates the activity of the nervous system outside the brain through the expression of photosensitive proteins and the application of photic stimulation. This article reviews recent advances in applying optogenetics to the spinal cord and peripheral nerves, offering a comprehensive understanding of the functions and regulatory mechanisms of the peripheral nervous system through the modulation of specific neuronal activities. By showcasing novel opportunities for disease treatment, this technique opens new avenues in psychophysiological research and neural regulation therapy. Despite current challenges, such as operability, effectiveness, and selective neuron targeting, peripheral optogenetics holds significant potential for advancing neuromodulation. Continued research and technological innovations will further expand its role, offering new possibilities for understanding and treating disorders involving the peripheral nervous system.
Collapse
Affiliation(s)
- Wenqian Zhou
- Department of Psychology, Shandong Second Medical University, Weifang, China
| | - Liping Jia
- Department of Psychology, Shandong Second Medical University, Weifang, China
| | - Lupeng Yue
- State Key Laboratory of Cognitive Science and Mental Health, Institute of Psychology, Chinese Academy of Sciences, Beijing, China; Department of Psychology, University of Chinese Academy of Sciences, Beijing, China.
| | - Li Hu
- State Key Laboratory of Cognitive Science and Mental Health, Institute of Psychology, Chinese Academy of Sciences, Beijing, China; Department of Psychology, University of Chinese Academy of Sciences, Beijing, China.
| |
Collapse
|
5
|
Frumento D, Ţălu Ș. Light-based technologies in immunotherapy: advances, mechanisms and applications. Immunotherapy 2025; 17:123-131. [PMID: 40032620 PMCID: PMC11901425 DOI: 10.1080/1750743x.2025.2470111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Accepted: 02/18/2025] [Indexed: 03/05/2025] Open
Abstract
Light-based immunotherapy uses specific wavelengths of light to activate or modulate immune responses. It primarily employs two mechanisms: direct activation of immune cells and indirect modulation of the tumor microenvironment (TME). Several light-based technologies are under investigation or clinical use in immunotherapy, including photodynamic immunotherapy (PDIT) and photothermal therapy (PTT). Optogenetic tools have the potential to precisely control T-cell receptor activation, cytokine release, or the activity of other immune effector cells. Light-based technologies present innovative opportunities within the realm of immunotherapy. The ability to precisely regulate immune cell activation via optogenetics, alongside the improved targeting of cancer cells through photoimmunotherapy, signifies a transformative shift in our strategies for immune modulation. Although many of these technologies remain in the experimental stage for various applications, initial findings are encouraging, especially concerning cancer treatment and immune modulation. Continued research and clinical trials are essential to fully harness the capabilities of light technology in the context of immune cell therapy.
Collapse
Affiliation(s)
| | - Ștefan Ţălu
- The Directorate of Research, Development and Innovation Management (DMCDI), The Technical University of Cluj-Napoca, Cluj-Napoca, Romania
| |
Collapse
|
6
|
Vaissiere T, Michaelson SD, Creson T, Goins J, Fürth D, Balazsfi D, Rojas C, Golovin R, Meletis K, Miller CA, O'Connor D, Fontolan L, Rumbaugh G. Syngap1 promotes cognitive function through regulation of cortical sensorimotor dynamics. Nat Commun 2025; 16:812. [PMID: 39827187 PMCID: PMC11743135 DOI: 10.1038/s41467-025-56125-0] [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: 10/30/2023] [Accepted: 01/07/2025] [Indexed: 01/22/2025] Open
Abstract
Perception, a cognitive construct, emerges through sensorimotor integration (SMI). The genetic mechanisms that shape SMI required for perception are unknown. Here, we demonstrate in mice that expression of the autism/intellectual disability gene, Syngap1, in cortical excitatory neurons is required for the formation of somatomotor networks that promote SMI-mediated perception. Cortical Syngap1 expression was necessary and sufficient for setting tactile sensitivity, sustaining tactile object exploration, and promoting tactile learning. Mice with deficient Syngap1 expression exhibited impaired neural dynamics induced by exploratory touches within a cortical-thalamic network that promotes attention and perception. Disrupted neuronal dynamics were associated with circuit-specific long-range synaptic connectivity abnormalities. Our data support a model where autonomous Syngap1 expression in cortical excitatory neurons promotes cognitive abilities through the assembly of long-range circuits that integrate temporally-overlapping sensory and motor signals, a process that promotes perception and attention. These data provide systems-level insights into the robust association between Syngap1 expression and cognitive ability.
Collapse
Affiliation(s)
- Thomas Vaissiere
- Department of Neuroscience, The Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology, Jupiter, FL, USA
| | - Sheldon D Michaelson
- Department of Neuroscience, The Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology, Jupiter, FL, USA
| | - Thomas Creson
- Department of Neuroscience, The Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology, Jupiter, FL, USA
| | - Jessie Goins
- Department of Neuroscience, The Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology, Jupiter, FL, USA
| | - Daniel Fürth
- SciLifeLab, Department of Immunology, Genetics & Pathology, Uppsala University, Uppsala, Sweden
| | - Diana Balazsfi
- Department of Neuroscience, The Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology, Jupiter, FL, USA
| | - Camilo Rojas
- Department of Neuroscience, The Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology, Jupiter, FL, USA
| | - Randall Golovin
- Department of Neuroscience, The Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology, Jupiter, FL, USA
| | | | - Courtney A Miller
- Department of Neuroscience, The Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology, Jupiter, FL, USA
- Department of Molecular Medicine, UF Scripps Biomedical Research, Jupiter, FL, USA
| | - Daniel O'Connor
- Department of Neuroscience, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Lorenzo Fontolan
- Aix-Marseille Université, INSERM, INMED, Turing Centre for Living Systems, Marseille, France
| | - Gavin Rumbaugh
- Department of Neuroscience, The Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology, Jupiter, FL, USA.
| |
Collapse
|
7
|
Sundqvist N, Podéus H, Sten S, Engström M, Dura-Bernal S, Cedersund G. A Model-Driven Meta-Analysis Supports the Emerging Consensus View that Inhibitory Neurons Dominate BOLD-fMRI Responses. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.15.618416. [PMID: 39464088 PMCID: PMC11507712 DOI: 10.1101/2024.10.15.618416] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/29/2024]
Abstract
Functional magnetic resonance imaging (fMRI) is a pivotal tool for mapping neuronal activity in the brain. Traditionally, the observed hemodynamic changes are assumed to reflect the activity of the most common neuronal type: excitatory neurons. In contrast, recent experiments, using optogenetic techniques, suggest that the fMRI-signal instead reflects the activity of inhibitory interneurons. However, these data paint a complex picture, with numerous regulatory interactions, and where the different experiments display many qualitative differences. It is therefore not trivial how to quantify the relative contributions of the different cell types and to combine all observations into a unified theory. To address this, we present a new model-driven meta-analysis, which provides a unified and quantitative explanation for all data. This model-driven analysis allows for quantification of the relative contribution of different cell types: the contribution to the BOLD-signal from the excitatory cells is <20 % and 50-80 % comes from the interneurons. Our analysis also provides a mechanistic explanation for the observed experiment-to-experiment differences, e.g. a biphasic vascular response dependent on different stimulation intensities and an emerging secondary post-stimulation peak during longer stimulations. In summary, our study provides a new, emerging consensus-view supporting the larger role of interneurons in fMRI.
Collapse
Affiliation(s)
- Nicolas Sundqvist
- Department of Biomedical Engineering, Linköping University, Linköping, Sweden
| | - Henrik Podéus
- Department of Biomedical Engineering, Linköping University, Linköping, Sweden
| | - Sebastian Sten
- Drug Metabolism and Pharmacokinetics, Research and Early Development, Cardiovascular, Renal and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Maria Engström
- Department of Health, Medicine and Caring Sciences, Linköping University, Linköping, Sweden
- Center for Medical Image Science and Visualization (CMIV), Linköping University, Linköping, Sweden
| | - Salvador Dura-Bernal
- Department of Physiology and Pharmacology, State University of New York (SUNY) Downstate Health Sciences University, Brooklyn, NY, USA
- Center for Biomedical Imaging and Neuromodulation, Nathan Kline Institute for Psychiatric Research, Orangeburg, NY, USA
| | - Gunnar Cedersund
- Department of Biomedical Engineering, Linköping University, Linköping, Sweden
- Center for Medical Image Science and Visualization (CMIV), Linköping University, Linköping, Sweden
- School of Medical Sciences and Inflammatory Response and Infection Susceptibility Centre (iRiSC), Faculty of Medicine and Health, Örebro University, Örebro, Sweden
| |
Collapse
|
8
|
Sugimoto K, Chung DY, Fischer P, Takizawa T, Qin T, Yaseen MA, Sakadžić S, Ayata C. Optogenetic Functional Activation Is Detrimental During Acute Ischemic Stroke in Mice. Stroke 2024; 55:2502-2509. [PMID: 39234742 PMCID: PMC11421960 DOI: 10.1161/strokeaha.124.048032] [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: 03/09/2024] [Revised: 07/03/2024] [Accepted: 08/12/2024] [Indexed: 09/06/2024]
Abstract
BACKGROUND Functional activation of the focal ischemic brain has been reported to improve outcomes by augmenting collateral blood flow. However, functional activation also increases metabolic demand and might thereby worsen outcomes. Indeed, preclinical and clinical reports have been conflicting. Here, we tested the effect of functional activation during acute ischemic stroke using distal middle cerebral artery occlusion in anesthetized mice. METHODS Using transgenic mice expressing channelrhodopsin-2 in neurons, we delivered functional activation using physiological levels of transcranial optogenetic stimulation of the moderately ischemic cortex (ie, penumbra), identified using real-time full-field laser speckle perfusion imaging during a 1-hour distal microvascular clip of the middle cerebral artery. Neuronal activation was confirmed using evoked field potentials, and infarct volumes were measured in tissue slices 48 hours later. RESULTS Optogenetic stimulation of the penumbra was associated with more than 2-fold larger infarcts than stimulation of the contralateral homotopic region and the sham stimulation group (n=10, 7, and 9; 11.0±5.6 versus 5.1±4.3 versus 4.1±3.7 mm3; P=0.008, 1-way ANOVA). Identical stimulation in wild-type mice that do not express channelrhodopsin-2 did not have an effect. Optogenetic stimulation was associated with a small increase in penumbral perfusion that did not explain enlarged infarcts. CONCLUSIONS Our data suggest that increased neuronal activity during acute focal arterial occlusions can be detrimental, presumably due to increased metabolic demand, and may have implications for the clinical management of hyperacute stroke patients.
Collapse
Affiliation(s)
- Kazutaka Sugimoto
- Department of Radiology, Massachusetts General Hospital, Harvard Medical School, USA
- Department of Neurosurgery, Yamaguchi University School of Medicine, Japan
| | - David Y. Chung
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, USA
| | - Paul Fischer
- Department of Radiology, Massachusetts General Hospital, Harvard Medical School, USA
- Department of Neurology with Experimental Neurology Charité-Universitätsmedizin Berlin and Berlin Institute of Health at Charité – Universitätsmedizin Berlin,BIH Biomedical Innovation Academy, BIH Charité Junior Clinician Scientist Program, Berlin Germany
| | - Tsubasa Takizawa
- Department of Radiology, Massachusetts General Hospital, Harvard Medical School, USA
- Department of Neurology, Keio University School of Medicine, Tokyo, Japan
| | - Tao Qin
- Department of Radiology, Massachusetts General Hospital, Harvard Medical School, USA
| | - Mohammad A. Yaseen
- Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown, MA, USA
- Bioengineering Department, Northeastern University, Boston, MA, USA
| | - Sava Sakadžić
- Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown, MA, USA
| | - Cenk Ayata
- Department of Radiology, Massachusetts General Hospital, Harvard Medical School, USA
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, USA
| |
Collapse
|
9
|
Vaissiere T, Michaelson SD, Creson T, Goins J, Fürth D, Balazsfi D, Rojas C, Golovin R, Meletis K, Miller CA, O’Connor D, Fontolan L, Rumbaugh G. Syngap1 Promotes Cognitive Function through Regulation of Cortical Sensorimotor Dynamics. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.09.27.559787. [PMID: 37808765 PMCID: PMC10557642 DOI: 10.1101/2023.09.27.559787] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/10/2023]
Abstract
Perception, a cognitive construct, emerges through sensorimotor integration (SMI). The genetic mechanisms that shape SMI required for perception are unknown. Here, we demonstrate in mice that expression of the autism/intellectual disability gene, Syngap1, in cortical excitatory neurons is required for formation of somatomotor networks that promote SMI-mediated perception. Cortical Syngap1 expression was necessary and sufficient for setting tactile sensitivity, sustaining tactile object exploration, and promoting tactile learning. Mice with deficient Syngap1 expression exhibited impaired neural dynamics induced by exploratory touches within a cortical-thalamic network known to promote attention and perception. Disrupted neuronal dynamics were associated with circuit-specific long-range synaptic connectivity abnormalities. Our data support a model where autonomous Syngap1 expression in cortical excitatory neurons promotes cognitive abilities through assembly of circuits that integrate temporally-overlapping sensory and motor signals, a process that promotes perception and attention. These data provide systems-level insights into the robust association between Syngap1 expression and cognitive ability.
Collapse
Affiliation(s)
- Thomas Vaissiere
- Department of Neuroscience, The Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology, Jupiter, FL, USA
| | - Sheldon D. Michaelson
- Department of Neuroscience, The Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology, Jupiter, FL, USA
| | - Thomas Creson
- Department of Neuroscience, The Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology, Jupiter, FL, USA
| | - Jessie Goins
- Department of Neuroscience, The Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology, Jupiter, FL, USA
| | - Daniel Fürth
- SciLifeLab, Department of Immunology, Genetics & Pathology, Uppsala University, Uppsala, Sweden
| | - Diana Balazsfi
- Department of Neuroscience, The Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology, Jupiter, FL, USA
| | - Camilo Rojas
- Department of Neuroscience, The Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology, Jupiter, FL, USA
| | - Randall Golovin
- Department of Neuroscience, The Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology, Jupiter, FL, USA
| | | | - Courtney A. Miller
- Department of Neuroscience, The Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology, Jupiter, FL, USA
- Department of Molecular Medicine, UF Scripps Biomedical Research, Jupiter, FL, USA
| | - Daniel O’Connor
- Department of Neuroscience, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Lorenzo Fontolan
- Aix-Marseille Université, INSERM, INMED, Turing Centre for Living Systems, Marseille, 13009, France
| | - Gavin Rumbaugh
- Department of Neuroscience, The Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology, Jupiter, FL, USA
| |
Collapse
|
10
|
Barykina NV, Carey EM, Oliinyk OS, Nimmerjahn A, Verkhusha VV. Destabilized near-infrared fluorescent nanobodies enable background-free targeting of GFP-based biosensors for imaging and manipulation. Nat Commun 2024; 15:7788. [PMID: 39242569 PMCID: PMC11379940 DOI: 10.1038/s41467-024-51857-x] [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: 12/19/2023] [Accepted: 08/19/2024] [Indexed: 09/09/2024] Open
Abstract
Near-infrared (NIR) probes are highly sought after as fluorescent tags for multicolor cellular and in vivo imaging. Here we develop small NIR fluorescent nanobodies, termed NIR-FbLAG16 and NIR-FbLAG30, enabling background-free visualization of various GFP-derived probes and biosensors. We also design a red-shifted variant, NIR-Fb(718), to simultaneously target several antigens within the NIR spectral range. Leveraging the antigen-stabilizing property of the developed NIR-Fbs, we then create two modular systems for precise control of gene expression in GFP-labeled cells. Applying the NIR-Fbs in vivo, we target cells expressing GFP and the calcium biosensor GCaMP6 in the somatosensory cortex of transgenic mice. Simultaneously tracking calcium activity and the reference signal from NIR-FbLAGs bound to GCaMP6 enables ratiometric deep-brain in vivo imaging. Altogether, NIR-FbLAGs present a promising approach for imaging and manipulating various processes in live cells and behaving animals expressing GFP-based probes.
Collapse
Affiliation(s)
- Natalia V Barykina
- Department of Genetics, and Gruss-Lipper Biophotonics Center, Albert Einstein College of Medicine, Bronx, New York, NY, 10461, USA
| | - Erin M Carey
- Waitt Advanced Biophotonics Center, Salk Institute for Biological Studies, La Jolla, CA, 92037, USA
| | - Olena S Oliinyk
- Medicum, Faculty of Medicine, University of Helsinki, Helsinki, 00290, Finland
| | - Axel Nimmerjahn
- Waitt Advanced Biophotonics Center, Salk Institute for Biological Studies, La Jolla, CA, 92037, USA
| | - Vladislav V Verkhusha
- Department of Genetics, and Gruss-Lipper Biophotonics Center, Albert Einstein College of Medicine, Bronx, New York, NY, 10461, USA.
- Medicum, Faculty of Medicine, University of Helsinki, Helsinki, 00290, Finland.
| |
Collapse
|
11
|
Cheng X, Nareddula S, Gao HC, Chen Y, Xiao T, Nadew YY, Xu F, Edens PA, Quinn CJ, Kimbrough A, Huang F, Chubykin AA. Impaired Experience-Dependent Theta Oscillation Synchronization and Inter-Areal Synaptic Connectivity in the Visual Cortex of Fmr1 KO Mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.23.601989. [PMID: 39211264 PMCID: PMC11360911 DOI: 10.1101/2024.07.23.601989] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
Fragile X syndrome (FX) is the most prevalent inheritable form of autism spectrum disorder (ASD), characterized by hypersensitivity, difficulty in habituating to new sensory stimuli, and intellectual disability. Individuals with FX often experience visual perception and learning deficits. Visual experience leads to the emergence of the familiarity-evoked theta band oscillations in the primary visual cortex (V1) and the lateromedial area (LM) of mice. These theta oscillations in V1 and LM are synchronized with each other, providing a mechanism of sensory multi-areal binding. However, how this multi-areal binding and the corresponding theta oscillations are altered in FX is not known. Using iDISCO whole brain clearing with light-sheet microscopy, we quantified immediate early gene Fos expression in V1 and LM, identifying deficits in experience-dependent neural activity in FX mice. We performed simultaneous in vivo recordings with silicon probes in V1 and LM of awake mice and channelrhodopsin-2-assisted circuit mapping (CRACM) in acute brain slices to examine the neural activity and strength of long-range synaptic connections between V1 and LM in both wildtype (WT) and Fmr1 knockout (KO) mice, the model of FX, before and after visual experience. Our findings reveal synchronized familiarity-evoked theta oscillations in V1 and LM, the increased strength of V1→LM functional and synaptic connections, which correlated with the corresponding changes of presynaptic short-term plasticity in WT mice. The LM oscillations were attenuated in FX mice and correlated with impaired functional and synaptic connectivity and short-term plasticity in the feedforward (FF) V1→LM and feedback (FB) LM→V1 pathways. Finally, using 4Pi single-molecule localization microscopy (SMLM) in thick brain tissue, we identified experience-dependent changes in the density and shape of dendritic spines in layer 5 pyramidal cells of WT mice, which correlated with the functional synaptic measurements. Interestingly, there was an increased dendritic spine density and length in naïve FX mice that failed to respond to experience. Our study provides the first comprehensive characterization of the role of visual experience in triggering inter-areal neural synchrony and shaping synaptic connectivity in WT and FX mice.
Collapse
|
12
|
Kim RG, Cho J, Park JY, Kim YR, Lee MC, Kim HI. Neuron type-specific optogenetic stimulation for differential stroke recovery in chronic capsular infarct. Exp Mol Med 2024; 56:1439-1449. [PMID: 38825647 PMCID: PMC11263592 DOI: 10.1038/s12276-024-01253-8] [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: 10/29/2023] [Revised: 02/28/2024] [Accepted: 03/18/2024] [Indexed: 06/04/2024] Open
Abstract
Cortical neuromodulation (CNM) is widely used to promote recovery after stroke. Despite the beneficial results of CNM, the roles played by different neuron types in the effects of current CNM techniques are unable to be differentiated. Our aim was to use selective optogenetic cortical stimulation to explore how different subpopulations of neuronal cells contribute to poststroke recovery. We transduced the sensory-parietal cortex (SPC) of rats with CamKII-ChR2 (pyramidal neurons), PV-ChR2 (parvalbumin-expressing inhibitory neurons), or hSyn-ChR2 (pan-neuronal population) before inducing photothrombotic capsular infarct lesions. We found that selective stimulation of inhibitory neurons resulted in significantly greater motor recovery than stimulation of excitatory neurons or the pan-neuronal population. Furthermore, 2-deoxy-2-[18F] fluoro-D-glucose microPET (FDG-microPET) imaging revealed a significant reduction in cortical diaschisis and activation of the corticostriatal neural circuit, which were correlated with behavioral recovery in the PV-ChR2 group. The spatial pattern of brain-derived neurotrophic factor (BDNF) expression was evident in the stimulated cortex and underlying cortico-subcortical circuit. Our results indicate that the plasticity of inhibitory neurons is crucial for functional recovery after capsular infarct. Modifying CNM parameters to potentiate the stimulation of inhibitory neurons could improve poststroke outcomes.
Collapse
Affiliation(s)
- Ra Gyung Kim
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology, 123 Choemdangwagi-ro, Buk-gu, Gwangju, 61005, Republic of Korea
- Research Headquarter, Korea Brain Research Institute, 61 Cheomdan-ro, Dong-gu, Daegu, 41062, Republic of Korea
| | - Jongwook Cho
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology, 123 Choemdangwagi-ro, Buk-gu, Gwangju, 61005, Republic of Korea
| | - Ji-Young Park
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology, 123 Choemdangwagi-ro, Buk-gu, Gwangju, 61005, Republic of Korea
| | - Young Ro Kim
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown, MA, 02129, USA
| | - Min-Cheol Lee
- Pathology Center, Seegene Medical Foundation, 320 Cheonho-Daero, Seongdong-gu, Seoul, 04805, Republic of Korea
| | - Hyoung-Ihl Kim
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology, 123 Choemdangwagi-ro, Buk-gu, Gwangju, 61005, Republic of Korea.
- Department of Neurosurgery, Presbyterian Medical Center, 365 Seowon-ro, Wansan-gu, Jeonju-si, Jeollabuk-do, 54987, Republic of Korea.
| |
Collapse
|
13
|
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
|
14
|
Osetrova M, Tkachev A, Mair W, Guijarro Larraz P, Efimova O, Kurochkin I, Stekolshchikova E, Anikanov N, Foo JC, Cazenave-Gassiot A, Mitina A, Ogurtsova P, Guo S, Potashnikova DM, Gulin AA, Vasin AA, Sarycheva A, Vladimirov G, Fedorova M, Kostyukevich Y, Nikolaev E, Wenk MR, Khrameeva EE, Khaitovich P. Lipidome atlas of the adult human brain. Nat Commun 2024; 15:4455. [PMID: 38796479 PMCID: PMC11127996 DOI: 10.1038/s41467-024-48734-y] [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: 12/15/2022] [Accepted: 05/07/2024] [Indexed: 05/28/2024] Open
Abstract
Lipids are the most abundant but poorly explored components of the human brain. Here, we present a lipidome map of the human brain comprising 75 regions, including 52 neocortical ones. The lipidome composition varies greatly among the brain regions, affecting 93% of the 419 analyzed lipids. These differences reflect the brain's structural characteristics, such as myelin content (345 lipids) and cell type composition (353 lipids), but also functional traits: functional connectivity (76 lipids) and information processing hierarchy (60 lipids). Combining lipid composition and mRNA expression data further enhances functional connectivity association. Biochemically, lipids linked with structural and functional brain features display distinct lipid class distribution, unsaturation extent, and prevalence of omega-3 and omega-6 fatty acid residues. We verified our conclusions by parallel analysis of three adult macaque brains, targeted analysis of 216 lipids, mass spectrometry imaging, and lipidome assessment of sorted murine neurons.
Collapse
Affiliation(s)
- Maria Osetrova
- Skolkovo Institute of Science and Technology, Moscow, Russia
| | - Anna Tkachev
- Skolkovo Institute of Science and Technology, Moscow, Russia
| | - Waltraud Mair
- Skolkovo Institute of Science and Technology, Moscow, Russia
| | | | - Olga Efimova
- Skolkovo Institute of Science and Technology, Moscow, Russia
| | - Ilia Kurochkin
- Skolkovo Institute of Science and Technology, Moscow, Russia
| | | | | | - Juat Chin Foo
- Singapore Lipidomics Incubator, Life Sciences Institute and Precision Medicine Translational Research Program, Department of Biochemistry, Yong Loo Lin School of Medicine; National University of Singapore, Singapore, Singapore
| | - Amaury Cazenave-Gassiot
- Singapore Lipidomics Incubator, Life Sciences Institute and Precision Medicine Translational Research Program, Department of Biochemistry, Yong Loo Lin School of Medicine; National University of Singapore, Singapore, Singapore
| | | | | | - Song Guo
- Skolkovo Institute of Science and Technology, Moscow, Russia
| | - Daria M Potashnikova
- Department of Cell Biology and Histology, Faculty of Biology, Lomonosov Moscow State University, Moscow, Russia
| | - Alexander A Gulin
- N. N. Semenov Federal Research Center for Chemical Physics Russian Academy of Sciences, Moscow, Russia
| | - Alexander A Vasin
- N. N. Semenov Federal Research Center for Chemical Physics Russian Academy of Sciences, Moscow, Russia
- Department of Chemistry, Lomonosov Moscow State University, Moscow, Russia
| | | | - Gleb Vladimirov
- Skolkovo Institute of Science and Technology, Moscow, Russia
| | | | | | - Evgeny Nikolaev
- Skolkovo Institute of Science and Technology, Moscow, Russia
| | - Markus R Wenk
- Singapore Lipidomics Incubator, Life Sciences Institute and Precision Medicine Translational Research Program, Department of Biochemistry, Yong Loo Lin School of Medicine; National University of Singapore, Singapore, Singapore.
| | | | | |
Collapse
|
15
|
Beau M, Herzfeld DJ, Naveros F, Hemelt ME, D’Agostino F, Oostland M, Sánchez-López A, Chung YY, Michael Maibach, Kyranakis S, Stabb HN, Martínez Lopera MG, Lajko A, Zedler M, Ohmae S, Hall NJ, Clark BA, Cohen D, Lisberger SG, Kostadinov D, Hull C, Häusser M, Medina JF. A deep-learning strategy to identify cell types across species from high-density extracellular recordings. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.30.577845. [PMID: 38352514 PMCID: PMC10862837 DOI: 10.1101/2024.01.30.577845] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/23/2024]
Abstract
High-density probes allow electrophysiological recordings from many neurons simultaneously across entire brain circuits but don't reveal cell type. Here, we develop a strategy to identify cell types from extracellular recordings in awake animals, revealing the computational roles of neurons with distinct functional, molecular, and anatomical properties. We combine optogenetic activation and pharmacology using the cerebellum as a testbed to generate a curated ground-truth library of electrophysiological properties for Purkinje cells, molecular layer interneurons, Golgi cells, and mossy fibers. We train a semi-supervised deep-learning classifier that predicts cell types with greater than 95% accuracy based on waveform, discharge statistics, and layer of the recorded neuron. The classifier's predictions agree with expert classification on recordings using different probes, in different laboratories, from functionally distinct cerebellar regions, and across animal species. Our classifier extends the power of modern dynamical systems analyses by revealing the unique contributions of simultaneously-recorded cell types during behavior.
Collapse
Affiliation(s)
- Maxime Beau
- Wolfson Institute for Biomedical Research, University College London, London, UK
| | - David J. Herzfeld
- Department of Neurobiology, Duke University School of Medicine, Durham, NC, USA
| | - Francisco Naveros
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA
- Department of Computer Engineering, Automation and Robotics, Research Centre for Information and Communication Technologies, University of Granada, Granada, Spain
| | - Marie E. Hemelt
- Department of Neurobiology, Duke University School of Medicine, Durham, NC, USA
| | - Federico D’Agostino
- Wolfson Institute for Biomedical Research, University College London, London, UK
| | - Marlies Oostland
- Wolfson Institute for Biomedical Research, University College London, London, UK
- Swammerdam Institute for Life Sciences, University of Amsterdam, Amsterdam, the Netherlands
| | | | - Young Yoon Chung
- Wolfson Institute for Biomedical Research, University College London, London, UK
| | - Michael Maibach
- Wolfson Institute for Biomedical Research, University College London, London, UK
| | - Stephen Kyranakis
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA
| | - Hannah N. Stabb
- Wolfson Institute for Biomedical Research, University College London, London, UK
| | | | - Agoston Lajko
- Wolfson Institute for Biomedical Research, University College London, London, UK
| | - Marie Zedler
- Wolfson Institute for Biomedical Research, University College London, London, UK
| | - Shogo Ohmae
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA
| | - Nathan J. Hall
- Department of Neurobiology, Duke University School of Medicine, Durham, NC, USA
| | - Beverley A. Clark
- Wolfson Institute for Biomedical Research, University College London, London, UK
| | - Dana Cohen
- The Leslie and Susan Gonda Multidisciplinary Brain Research Center, Bar-Ilan University, Ramat-Gan, Israel
| | | | - Dimitar Kostadinov
- Wolfson Institute for Biomedical Research, University College London, London, UK
- Centre for Developmental Neurobiology, King’s College London, London, UK
| | - Court Hull
- Department of Neurobiology, Duke University School of Medicine, Durham, NC, USA
| | - Michael Häusser
- Wolfson Institute for Biomedical Research, University College London, London, UK
| | - Javier F. Medina
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA
| |
Collapse
|
16
|
Dmitrieva E, Malkov A. Optogenetic stimulation of medial septal glutamatergic neurons modulates theta-gamma coupling in the hippocampus. Neurobiol Learn Mem 2024; 211:107929. [PMID: 38685526 DOI: 10.1016/j.nlm.2024.107929] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Revised: 04/08/2024] [Accepted: 04/24/2024] [Indexed: 05/02/2024]
Abstract
Hippocampal cross-frequency theta-gamma coupling (TGC) is a basic mechanism for information processing, retrieval, and consolidation of long-term and working memory. While the role of entorhinal afferents in the modulation of hippocampal TGC is widely accepted, the influence of other main input to the hippocampus, from the medial septal area (MSA, the pacemaker of the hippocampal theta rhythm) is poorly understood. Optogenetics allows us to explore how different neuronal populations of septohippocampal circuits control neuronal oscillations in vivo. Rhythmic activation of septal glutamatergic neurons has been shown to drive hippocampal theta oscillations, but the role of these neuronal populations in information processing during theta activation has remained unclear. Here we investigated the influence of phasic activation of MSA glutamatergic neurons expressing channelrhodopsin II on theta-gamma coupling in the hippocampus. During the experiment, local field potentials of MSA and hippocampus of freely behaving mice were modulated by 470 nm light flashes with theta frequency (2-10) Hz. It was shown that both the power and the strength of modulation of gamma rhythm nested on hippocampal theta waves depend on the frequency of stimulation. The modulation of the amplitude of slow gamma rhythm (30-50 Hz) prevailed over modulation of fast gamma (55-100 Hz) during flash trains and the observed effects were specific for theta stimulation of MSA. We discuss the possibility that phasic depolarization of septal glutamatergic neurons controls theta-gamma coupling in the hippocampus and plays a role in memory retrieval and consolidation.
Collapse
Affiliation(s)
- Elena Dmitrieva
- Institute of Theoretical and Experimental Biophysics Russian Academy of Sciences, Pushchino, Russia
| | - Anton Malkov
- Institute of Theoretical and Experimental Biophysics Russian Academy of Sciences, Pushchino, Russia.
| |
Collapse
|
17
|
Fiala A, Kaun KR. What do the mushroom bodies do for the insect brain? Twenty-five years of progress. Learn Mem 2024; 31:a053827. [PMID: 38862175 PMCID: PMC11199942 DOI: 10.1101/lm.053827.123] [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: 04/21/2024] [Accepted: 04/22/2024] [Indexed: 06/13/2024]
Abstract
In 1998, a special edition of Learning & Memory was published with a discrete focus of synthesizing the state of the field to provide an overview of the function of the insect mushroom body. While molecular neuroscience and optical imaging of larger brain areas were advancing, understanding the basic functioning of neuronal circuits, particularly in the context of the mushroom body, was rudimentary. In the past 25 years, technological innovations have allowed researchers to map and understand the in vivo function of the neuronal circuits of the mushroom body system, making it an ideal model for investigating the circuit basis of sensory encoding, memory formation, and behavioral decisions. Collaborative efforts within the community have played a crucial role, leading to an interactive connectome of the mushroom body and accessible genetic tools for studying mushroom body circuit function. Looking ahead, continued technological innovation and collaborative efforts are likely to further advance our understanding of the mushroom body and its role in behavior and cognition, providing insights that generalize to other brain structures and species.
Collapse
Affiliation(s)
- André Fiala
- Department of Molecular Neurobiology of Behaviour, University of Göttingen, Göttingen 37077, Germany
| | - Karla R Kaun
- Department of Neuroscience, Brown University, Providence, Rhode Island 02806, USA
| |
Collapse
|
18
|
Silva NT, Ramírez-Buriticá J, Pritchett DL, Carey MR. Climbing fibers provide essential instructive signals for associative learning. Nat Neurosci 2024; 27:940-951. [PMID: 38565684 PMCID: PMC11088996 DOI: 10.1038/s41593-024-01594-7] [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: 04/18/2022] [Accepted: 02/05/2024] [Indexed: 04/04/2024]
Abstract
Supervised learning depends on instructive signals that shape the output of neural circuits to support learned changes in behavior. Climbing fiber (CF) inputs to the cerebellar cortex represent one of the strongest candidates in the vertebrate brain for conveying neural instructive signals. However, recent studies have shown that Purkinje cell stimulation can also drive cerebellar learning and the relative importance of these two neuron types in providing instructive signals for cerebellum-dependent behaviors remains unresolved. In the present study we used cell-type-specific perturbations of various cerebellar circuit elements to systematically evaluate their contributions to delay eyeblink conditioning in mice. Our findings reveal that, although optogenetic stimulation of either CFs or Purkinje cells can drive learning under some conditions, even subtle reductions in CF signaling completely block learning to natural stimuli. We conclude that CFs and corresponding Purkinje cell complex spike events provide essential instructive signals for associative cerebellar learning.
Collapse
Affiliation(s)
- N Tatiana Silva
- Neuroscience Program, Champalimaud Center for the Unknown, Lisbon, Portugal
| | | | - Dominique L Pritchett
- Neuroscience Program, Champalimaud Center for the Unknown, Lisbon, Portugal.
- Biology Department, Howard University, Washington, DC, USA.
| | - Megan R Carey
- Neuroscience Program, Champalimaud Center for the Unknown, Lisbon, Portugal.
| |
Collapse
|
19
|
El Hajj R, Al Sagheer T, Ballout N. Optogenetics in chronic neurodegenerative diseases, controlling the brain with light: A systematic review. J Neurosci Res 2024; 102:e25321. [PMID: 38588013 DOI: 10.1002/jnr.25321] [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: 10/24/2023] [Revised: 02/20/2024] [Accepted: 03/09/2024] [Indexed: 04/10/2024]
Abstract
Neurodegenerative diseases are progressive disorders characterized by synaptic loss and neuronal death. Optogenetics combines optical and genetic methods to control the activity of specific cell types. The efficacy of this approach in neurodegenerative diseases has been investigated in many reviews, however, none of them tackled it systematically. Our study aimed to review systematically the findings of optogenetics and its potential applications in animal models of chronic neurodegenerative diseases and compare it with deep brain stimulation and designer receptors exclusively activated by designer drugs techniques. The search strategy was performed based on the PRISMA guidelines and the risk of bias was assessed following the Systematic Review Centre for Laboratory Animal Experimentation tool. A total of 247 articles were found, of which 53 were suitable for the qualitative analysis. Our data revealed that optogenetic manipulation of distinct neurons in the brain is efficient in rescuing memory impairment, alleviating neuroinflammation, and reducing plaque pathology in Alzheimer's disease. Similarly, this technique shows an advanced understanding of the contribution of various neurons involved in the basal ganglia pathways with Parkinson's disease motor symptoms and pathology. However, the optogenetic application using animal models of Huntington's disease, multiple sclerosis, and amyotrophic lateral sclerosis was limited. Optogenetics is a promising technique that enhanced our knowledge in the research of neurodegenerative diseases and addressed potential therapeutic solutions for managing these diseases' symptoms and delaying their progression. Nevertheless, advanced investigations should be considered to improve optogenetic tools' efficacy and safety to pave the way for their translatability to the clinic.
Collapse
Affiliation(s)
- Rojine El Hajj
- Neuroscience Research Center, Faculty of Medical Sciences, Lebanese University, Beirut, Lebanon
| | - Tareq Al Sagheer
- Neuroscience Research Center, Faculty of Medical Sciences, Lebanese University, Beirut, Lebanon
| | - Nissrine Ballout
- Neuroscience Research Center, Faculty of Medical Sciences, Lebanese University, Beirut, Lebanon
| |
Collapse
|
20
|
Mochizuki T, Manita S, Shimura H, Kira S, Sawada N, Bito H, Sakimura K, Augustine GJ, Mitsui T, Takeda M, Kitamura K. Optogenetic stimulation of neurons in the anterior cingulate cortex induces changes in intravesical bladder pressure and the micturition reflex. Sci Rep 2024; 14:6367. [PMID: 38493201 PMCID: PMC10944464 DOI: 10.1038/s41598-024-56806-8] [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: 05/12/2023] [Accepted: 03/11/2024] [Indexed: 03/18/2024] Open
Abstract
Lower urinary tract (LUT) function is controlled by the central nervous system, including higher-order cognitive brain regions. The anterior cingulate cortex (ACC) is one of these regions, but the role of its activity in LUT function remains poorly understood. In the present study, we conducted optogenetic experiments to manipulate neural activity in mouse ACC while monitoring bladder pressure to elucidate how the activity of ACC regulates LUT function. Selective optogenetic stimulation of excitatory neurons in ACC induced a sharp increase in bladder pressure, whereas activation of inhibitory neurons in ACC prolonged the interval between bladder contractions. Pharmacological manipulation of ACC also altered bladder contractions, consistent with those observed in optogenetic experiments. Optogenetic mapping of the cortical area responsible for eliciting the increase in bladder pressure revealed that stimulation to ACC showed more potent effects than the neighboring motor cortical areas. These results suggest that ACC plays a crucial role in initiating the bladder pressure change and the micturition reflex. Thus, the balance between excitation and inhibition in ACC may regulate the reflex bidirectionally.
Collapse
Affiliation(s)
- Takanori Mochizuki
- Department of Urology, Faculty of Medicine, University of Yamanashi, Chuo, Yamanashi, Japan
| | - Satoshi Manita
- Department of Neurophysiology, Faculty of Medicine, University of Yamanashi, Chuo, Yamanashi, Japan
| | - Hiroshi Shimura
- Department of Urology, Faculty of Medicine, University of Yamanashi, Chuo, Yamanashi, Japan
| | - Satoru Kira
- Department of Urology, Faculty of Medicine, University of Yamanashi, Chuo, Yamanashi, Japan
| | - Norifumi Sawada
- Department of Urology, Faculty of Medicine, University of Yamanashi, Chuo, Yamanashi, Japan
| | - Haruhiko Bito
- Department of Neurochemistry, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Kenji Sakimura
- Department of Cellular Neurobiology, Brain Research Institute, Niigata University, Niigata, Japan
| | | | - Takahiko Mitsui
- Department of Urology, Faculty of Medicine, University of Yamanashi, Chuo, Yamanashi, Japan
| | - Masayuki Takeda
- Department of Urology, Faculty of Medicine, University of Yamanashi, Chuo, Yamanashi, Japan
| | - Kazuo Kitamura
- Department of Neurophysiology, Faculty of Medicine, University of Yamanashi, Chuo, Yamanashi, Japan.
| |
Collapse
|
21
|
Kaspar C, Ivanenko A, Lehrich J, Klingauf J, Pernice WHP. Biohybrid Photonic Platform for Subcellular Stimulation and Readout of In Vitro Neurons. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2304561. [PMID: 38164885 DOI: 10.1002/advs.202304561] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Revised: 12/20/2023] [Indexed: 01/03/2024]
Abstract
Targeted manipulation of neural activity via light has become an indispensable tool for gaining insights into the intricate processes governing single neurons and complex neural networks. To shed light onto the underlying interaction mechanisms, it is crucial to achieve precise control of individual neural activity, as well as a spatial read-out resolution on the nanoscale. Here, a versatile photonic platform with subcellular resolution for stimulation and monitoring of in-vitro neurons is demonstrated. Low-loss photonic waveguides are fabricated on glass substrates using nanoimprint lithography and featuring a loss of only -0.9 ± 0.2 dB cm-1 at 489 nm and are combined with optical fiber-based waveguide-access and backside total internal reflection fluorescence microscopy. Neurons are grown on the bio-functionalized photonic chip surface and, expressing the light-sensitive ion channel Channelrhodopsin-2, are stimulated within the evanescent field penetration depth of 57 nm of the biocompatible waveguides. The versatility and cost-efficiency of the platform, along with the possible subcellular resolution, enable tailor-made investigations of neural interaction dynamics with defined spatial control and high throughput.
Collapse
Affiliation(s)
- Corinna Kaspar
- Institute of Physics, University of Muenster, Heisenbergstr. 11, 48149, Muenster, Germany
- Center for Soft Nanoscience, University of Muenster, Busso-Peuss-Str. 10, 48149, Muenster, Germany
| | - Alexander Ivanenko
- Center for Soft Nanoscience, University of Muenster, Busso-Peuss-Str. 10, 48149, Muenster, Germany
- Institute of Medical Physics and Biophysics, University of Muenster, Robert-Koch-Str. 31, 48149, Muenster, Germany
| | - Julia Lehrich
- Center for Soft Nanoscience, University of Muenster, Busso-Peuss-Str. 10, 48149, Muenster, Germany
- Institute of Medical Physics and Biophysics, University of Muenster, Robert-Koch-Str. 31, 48149, Muenster, Germany
| | - Jürgen Klingauf
- Center for Soft Nanoscience, University of Muenster, Busso-Peuss-Str. 10, 48149, Muenster, Germany
- Institute of Medical Physics and Biophysics, University of Muenster, Robert-Koch-Str. 31, 48149, Muenster, Germany
| | - Wolfram H P Pernice
- Institute of Physics, University of Muenster, Heisenbergstr. 11, 48149, Muenster, Germany
- Center for Soft Nanoscience, University of Muenster, Busso-Peuss-Str. 10, 48149, Muenster, Germany
- Kirchhoff-Institut for Physics, Heidelberg University, Im Neuenheimer Feld 227, 69120, Heidelberg, Germany
| |
Collapse
|
22
|
Loonen ICM, Voskuyl RA, Schenke M, van Heiningen SH, van den Maagdenberg AMJM, Tolner EA. Spontaneous and optogenetically induced cortical spreading depolarization in familial hemiplegic migraine type 1 mutant mice. Neurobiol Dis 2024; 192:106405. [PMID: 38211710 DOI: 10.1016/j.nbd.2024.106405] [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: 09/30/2023] [Revised: 12/15/2023] [Accepted: 01/04/2024] [Indexed: 01/13/2024] Open
Abstract
Mechanisms underlying the migraine aura are incompletely understood, which to large extent is related to a lack of models in which cortical spreading depolarization (CSD), the correlate of the aura, occurs spontaneously. Here, we investigated electrophysiological and behavioural CSD features in freely behaving mice expressing mutant CaV2.1 Ca2+ channels, either with the milder R192Q or the severer S218L missense mutation in the α1 subunit, known to cause familial hemiplegic migraine type 1 (FHM1) in patients. Very rarely, spontaneous CSDs were observed in mutant but never in wildtype mice. In homozygous Cacna1aR192Q mice exclusively single-wave CSDs were observed whereas heterozygous Cacna1aS218L mice displayed multiple-wave events, seemingly in line with the more severe clinical phenotype associated with the S218L mutation. Spontaneous CSDs were associated with body stretching, one-directional slow head turning, and rotating movement of the body. Spontaneous CSD events were compared with those induced in a controlled manner using minimally invasive optogenetics. Also in the optogenetic experiments single-wave CSDs were observed in Cacna1aR192Q and Cacna1aS218L mice (whereas the latter also showed multiple-wave events) with movements similar to those observed with spontaneous events. Compared to wildtype mice, FHM1 mutant mice exhibited a reduced threshold and an increased propagation speed for optogenetically induced CSD with a more profound CSD-associated dysfunction, as indicated by a prolonged suppression of transcallosal evoked potentials and a reduction of unilateral forepaw grip performance. When induced during sleep, the optogenetic CSD threshold was particularly lowered, which may explain why spontaneous CSD events predominantly occurred during sleep. In conclusion, our data show that key neurophysiological and behavioural features of optogenetically induced CSDs mimic those of rare spontaneous events in FHM1 R192Q and S218L mutant mice with differences in severity in line with FHM1 clinical phenotypes seen with these mutations.
Collapse
Affiliation(s)
- Inge C M Loonen
- Department of Human Genetics, Leiden University Medical Center, Leiden 2333 RC, the Netherlands
| | - Rob A Voskuyl
- Department of Human Genetics, Leiden University Medical Center, Leiden 2333 RC, the Netherlands
| | - Maarten Schenke
- Department of Human Genetics, Leiden University Medical Center, Leiden 2333 RC, the Netherlands
| | - Sandra H van Heiningen
- Department of Human Genetics, Leiden University Medical Center, Leiden 2333 RC, the Netherlands
| | - Arn M J M van den Maagdenberg
- Department of Human Genetics, Leiden University Medical Center, Leiden 2333 RC, the Netherlands; Department of Neurology, Leiden University Medical Center, Leiden 2333 RC, the Netherlands
| | - Else A Tolner
- Department of Human Genetics, Leiden University Medical Center, Leiden 2333 RC, the Netherlands; Department of Neurology, Leiden University Medical Center, Leiden 2333 RC, the Netherlands.
| |
Collapse
|
23
|
Mester JR, Rozak MW, Dorr A, Goubran M, Sled JG, Stefanovic B. Network response of brain microvasculature to neuronal stimulation. Neuroimage 2024; 287:120512. [PMID: 38199427 DOI: 10.1016/j.neuroimage.2024.120512] [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: 09/08/2023] [Revised: 12/21/2023] [Accepted: 01/04/2024] [Indexed: 01/12/2024] Open
Abstract
Neurovascular coupling (NVC), or the adjustment of blood flow in response to local increases in neuronal activity is a hallmark of healthy brain function, and the physiological foundation for functional magnetic resonance imaging (fMRI). However, it remains only partly understood due to the high complexity of the structure and function of the cerebrovascular network. Here we set out to understand NVC at the network level, i.e. map cerebrovascular network reactivity to activation of neighbouring neurons within a 500×500×500 μm3 cortical volume (∼30 high-resolution 3-nL fMRI voxels). Using 3D two-photon fluorescence microscopy data, we quantified blood volume and flow changes in the brain vessels in response to spatially targeted optogenetic activation of cortical pyramidal neurons. We registered the vessels in a series of image stacks acquired before and after stimulations and applied a deep learning pipeline to segment the microvascular network from each time frame acquired. We then performed image analysis to extract the microvascular graphs, and graph analysis to identify the branch order of each vessel in the network, enabling the stratification of vessels by their branch order, designating branches 1-3 as precapillary arterioles and branches 4+ as capillaries. Forty-five percent of all vessels showed significant calibre changes; with 85 % of responses being dilations. The largest absolute CBV change was in the capillaries; the smallest, in the venules. Capillary CBV change was also the largest fraction of the total CBV change, but normalized to the baseline volume, arterioles and precapillary arterioles showed the biggest relative CBV change. From linescans along arteriole-venule microvascular paths, we measured red blood cell velocities and hematocrit, allowing for estimation of pressure and local resistance along these paths. While diameter changes following neuronal activation gradually declined along the paths; the pressure drops from arterioles to venules increased despite decreasing resistance: blood flow thus increased more than local resistance decreases would predict. By leveraging functional volumetric imaging and high throughput deep learning-based analysis, our study revealed distinct hemodynamic responses across the vessel types comprising the microvascular network. Our findings underscore the need for large, dense sampling of brain vessels for characterization of neurovascular coupling at the network level in health and disease.
Collapse
Affiliation(s)
- James R Mester
- University of Toronto, Department of Medical Biophysics, Toronto, Ontario, Canada; Physical Sciences, Sunnybrook Research Institute, Toronto, Ontario, Canada
| | - Matthew W Rozak
- University of Toronto, Department of Medical Biophysics, Toronto, Ontario, Canada; Physical Sciences, Sunnybrook Research Institute, Toronto, Ontario, Canada
| | - Adrienne Dorr
- Physical Sciences, Sunnybrook Research Institute, Toronto, Ontario, Canada
| | - Maged Goubran
- University of Toronto, Department of Medical Biophysics, Toronto, Ontario, Canada; Physical Sciences, Sunnybrook Research Institute, Toronto, Ontario, Canada
| | - John G Sled
- University of Toronto, Department of Medical Biophysics, Toronto, Ontario, Canada; Mouse Imaging Centre, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Bojana Stefanovic
- University of Toronto, Department of Medical Biophysics, Toronto, Ontario, Canada; Physical Sciences, Sunnybrook Research Institute, Toronto, Ontario, Canada.
| |
Collapse
|
24
|
Ma J, Egodawaththa NM, Guruge C, Márquez OAV, Likes M, Nesnas N. Blue and Green Light Responsive Caged Glutamate. J Photochem Photobiol A Chem 2024; 447:115183. [PMID: 37928883 PMCID: PMC10621743 DOI: 10.1016/j.jphotochem.2023.115183] [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] [Indexed: 11/07/2023]
Abstract
Glutamate (Glu) is an excitatory neurotransmitter that plays a critical role in memory. Brain mapping activities of such pathways relied heavily on the ability to release Glu with spatiotemporal precision. Several photo-protecting groups (PPGs), referred to as photocages or cages, were designed to accomplish the release of Glu upon irradiation. Previously reported Glu cages responded to UV upon irradiation with single photons, which limited their use in vivo experiments due to cytotoxicity. Other caged designs suffered from lower quantum efficiency (QE) of release necessitating higher concentrations and/or longer photoirradiation times. There have been limited examples of cages that respond to visible light with single photon irradiation. Herein, we report the efficient preparation of 11 caged Glu examples that respond to two visible wavelengths, 467 nm (thiocoumarin based) and 515-540 nm (BODIPY based). The kinetics of photouncaging were studied for all caged designs, and we report all quantum efficiencies, i.e., quantum yields (Φ), that ranged from 0.0001-0.65. Two of the BODIPY cages are reported here for the first time, and one, Me-BODIPY-Br-Glu, shows the most efficient Glu release with a QE of 0.65. Similar caged designs can be extended to the inhibitory neurotransmitter, GABA. This would enable the use of two visible wavelengths to modulate the release of excitatory and inhibitory neurotransmitters upon demand via optical control.
Collapse
Affiliation(s)
| | | | - Charitha Guruge
- Department of Biomedical and Chemical Engineering and Sciences, Florida Institute of Technology, 150 West University Blvd., Melbourne, FL 32901, United States
| | - Oriana A. Valladares Márquez
- Department of Biomedical and Chemical Engineering and Sciences, Florida Institute of Technology, 150 West University Blvd., Melbourne, FL 32901, United States
| | - Molly Likes
- Department of Biomedical and Chemical Engineering and Sciences, Florida Institute of Technology, 150 West University Blvd., Melbourne, FL 32901, United States
| | - Nasri Nesnas
- Department of Biomedical and Chemical Engineering and Sciences, Florida Institute of Technology, 150 West University Blvd., Melbourne, FL 32901, United States
| |
Collapse
|
25
|
Kinsky NR, Vöröslakos M, Lopez Ruiz JR, Watkins de Jong L, Slager N, McKenzie S, Yoon E, Diba K. Simultaneous electrophysiology and optogenetic perturbation of the same neurons in chronically implanted animals using μLED silicon probes. STAR Protoc 2023; 4:102570. [PMID: 37729059 PMCID: PMC10510336 DOI: 10.1016/j.xpro.2023.102570] [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: 04/24/2023] [Revised: 07/14/2023] [Accepted: 08/21/2023] [Indexed: 09/22/2023] Open
Abstract
Micro-light-emitting-diode (μLED) silicon probes feature independently controllable miniature light-emitting-diodes (LEDs) embedded at several positions in each shank of a multi-shank probe, enabling temporally and spatially precise optogenetic neural circuit interrogation. Here, we present a protocol for performing causal and reproducible neural circuit manipulations in chronically implanted, freely moving animals. We describe steps for introducing optogenetic constructs, preparing and implanting a μLED probe, performing simultaneous in vivo electrophysiology with focal optogenetic perturbation, and recovering a probe following termination of an experiment. For complete details on the use and execution of this protocol, please refer to Watkins de Jong et al. (2023).1.
Collapse
Affiliation(s)
- Nathaniel R Kinsky
- Department of Anesthesiology and Neuroscience Graduate Program, University of Michigan Medical School, Ann Arbor, MI 48109, USA.
| | - Mihály Vöröslakos
- Department of Electrical Engineering and Computer Science, University of Michigan, Ann Arbor, MI 48109, USA; Neuroscience Institute, Langone Medical Center, New York University, New York, NY 10016, USA
| | - Jose Roberto Lopez Ruiz
- Department of Electrical Engineering and Computer Science, University of Michigan, Ann Arbor, MI 48109, USA; Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
| | - Laurel Watkins de Jong
- Department of Anesthesiology and Neuroscience Graduate Program, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Nathan Slager
- Department of Electrical Engineering and Computer Science, University of Michigan, Ann Arbor, MI 48109, USA; Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
| | - Sam McKenzie
- Department of Neuroscience, University of New Mexico, Albuquerque, NM 87131, USA
| | - Euisik Yoon
- Department of Electrical Engineering and Computer Science, University of Michigan, Ann Arbor, MI 48109, USA; Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, USA; Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI 48109, USA; Center for Nanomedicine, Institute for Basic Science (IBS) and Graduate Program of Nano Biomedical Engineering (Nano BME), Advanced Science Institute, Yonsei University, Seoul 03722, South Korea
| | - Kamran Diba
- Department of Anesthesiology and Neuroscience Graduate Program, University of Michigan Medical School, Ann Arbor, MI 48109, USA.
| |
Collapse
|
26
|
Dell’Orco M, Weisend JE, Perrone-Bizzozero NI, Carlson AP, Morton RA, Linsenbardt DN, Shuttleworth CW. Repetitive spreading depolarization induces gene expression changes related to synaptic plasticity and neuroprotective pathways. Front Cell Neurosci 2023; 17:1292661. [PMID: 38162001 PMCID: PMC10757627 DOI: 10.3389/fncel.2023.1292661] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Accepted: 11/17/2023] [Indexed: 01/03/2024] Open
Abstract
Spreading depolarization (SD) is a slowly propagating wave of profound depolarization that sweeps through cortical tissue. While much emphasis has been placed on the damaging consequences of SD, there is uncertainty surrounding the potential activation of beneficial pathways such as cell survival and plasticity. The present study used unbiased assessments of gene expression to evaluate that compensatory and repair mechanisms could be recruited following SD, regardless of the induction method, which prior to this work had not been assessed. We also tested assumptions of appropriate controls and the spatial extent of expression changes that are important for in vivo SD models. SD clusters were induced with either KCl focal application or optogenetic stimulation in healthy mice. Cortical RNA was extracted and sequenced to identify differentially expressed genes (DEGs). SDs using both induction methods significantly upregulated 16 genes (vs. sham animals) that included the cell proliferation-related genes FOS, JUN, and DUSP6, the plasticity-related genes ARC and HOMER1, and the inflammation-related genes PTGS2, EGR2, and NR4A1. The contralateral hemisphere is commonly used as control tissue for DEG studies, but its activity could be modified by near-global disruption of activity in the adjacent brain. We found 21 upregulated genes when comparing SD-involved cortex vs. tissue from the contralateral hemisphere of the same animals. Interestingly, there was almost complete overlap (21/16) with the DEGs identified using sham controls. Neuronal activity also differs in SD initiation zones, where sustained global depolarization is required to initiate propagating events. We found that gene expression varied as a function of the distance from the SD initiation site, with greater expression differences observed in regions further away. Functional and pathway enrichment analyses identified axonogenesis, branching, neuritogenesis, and dendritic growth as significantly enriched in overlapping DEGs. Increased expression of SD-induced genes was also associated with predicted inhibition of pathways associated with cell death, and apoptosis. These results identify novel biological pathways that could be involved in plasticity and/or circuit modification in brain tissue impacted by SD. These results also identify novel functional targets that could be tested to determine potential roles in the recovery and survival of peri-infarct tissues.
Collapse
Affiliation(s)
- Michela Dell’Orco
- Department of Neurosciences, The University of New Mexico School of Medicine, Albuquerque, NM, United States
| | - Jordan E. Weisend
- Department of Neurosciences, The University of New Mexico School of Medicine, Albuquerque, NM, United States
| | - Nora I. Perrone-Bizzozero
- Department of Neurosciences, The University of New Mexico School of Medicine, Albuquerque, NM, United States
| | - Andrew P. Carlson
- Department of Neurosurgery, The University of New Mexico School of Medicine, Albuquerque, NM, United States
| | - Russell A. Morton
- Department of Neurosciences, The University of New Mexico School of Medicine, Albuquerque, NM, United States
| | - David N. Linsenbardt
- Department of Neurosciences, The University of New Mexico School of Medicine, Albuquerque, NM, United States
| | - C. William Shuttleworth
- Department of Neurosciences, The University of New Mexico School of Medicine, Albuquerque, NM, United States
| |
Collapse
|
27
|
Kaya Z, Belder N, Sever-Bahcekapili M, Donmez-Demir B, Erdener ŞE, Bozbeyoglu N, Bagci C, Eren-Kocak E, Yemisci M, Karatas H, Erdemli E, Gursel I, Dalkara T. Vesicular HMGB1 release from neurons stressed with spreading depolarization enables confined inflammatory signaling to astrocytes. J Neuroinflammation 2023; 20:295. [PMID: 38082296 PMCID: PMC10712196 DOI: 10.1186/s12974-023-02977-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Accepted: 11/28/2023] [Indexed: 12/18/2023] Open
Abstract
The role of high mobility group box 1 (HMGB1) in inflammation is well characterized in the immune system and in response to tissue injury. More recently, HMGB1 was also shown to initiate an "inflammatory signaling cascade" in the brain parenchyma after a mild and brief disturbance, such as cortical spreading depolarization (CSD), leading to headache. Despite substantial evidence implying a role for inflammatory signaling in prevalent neuropsychiatric disorders such as migraine and depression, how HMGB1 is released from healthy neurons and how inflammatory signaling is initiated in the absence of apparent cell injury are not well characterized. We triggered a single cortical spreading depolarization by optogenetic stimulation or pinprick in naïve Swiss albino or transgenic Thy1-ChR2-YFP and hGFAP-GFP adult mice. We evaluated HMGB1 release in brain tissue sections prepared from these mice by immunofluorescent labeling and immunoelectron microscopy. EzColocalization and Costes thresholding algorithms were used to assess the colocalization of small extracellular vesicles (sEVs) carrying HMGB1 with astrocyte or microglia processes. sEVs were also isolated from the brain after CSD, and neuron-derived sEVs were captured by CD171 (L1CAM). sEVs were characterized with flow cytometry, scanning electron microscopy, nanoparticle tracking analysis, and Western blotting. We found that HMGB1 is released mainly within sEVs from the soma of stressed neurons, which are taken up by surrounding astrocyte processes. This creates conditions for selective communication between neurons and astrocytes bypassing microglia, as evidenced by activation of the proinflammatory transcription factor NF-ĸB p65 in astrocytes but not in microglia. Transmission immunoelectron microscopy data illustrated that HMGB1 was incorporated into sEVs through endosomal mechanisms. In conclusion, proinflammatory mediators released within sEVs can induce cell-specific inflammatory signaling in the brain without activating transmembrane receptors on other cells and causing overt inflammation.
Collapse
Affiliation(s)
- Zeynep Kaya
- Institute of Neurological Sciences and Psychiatry, Hacettepe University, Sıhhiye, Ankara, Turkey
| | - Nevin Belder
- Institute of Neurological Sciences and Psychiatry, Hacettepe University, Sıhhiye, Ankara, Turkey
- Biotechnology Institute, Ankara University, Ankara, Turkey
| | - Melike Sever-Bahcekapili
- Institute of Neurological Sciences and Psychiatry, Hacettepe University, Sıhhiye, Ankara, Turkey
| | - Buket Donmez-Demir
- Institute of Neurological Sciences and Psychiatry, Hacettepe University, Sıhhiye, Ankara, Turkey
| | - Şefik Evren Erdener
- Institute of Neurological Sciences and Psychiatry, Hacettepe University, Sıhhiye, Ankara, Turkey
| | - Naz Bozbeyoglu
- Department of Molecular Biology and Genetics, Science Faculty, Bilkent University, Ankara, Turkey
| | - Canan Bagci
- Institute of Neurological Sciences and Psychiatry, Hacettepe University, Sıhhiye, Ankara, Turkey
- Department of Biomedical Engineering, Faculty of Engineering and Natural Sciences, Bahçeşehir University, Istanbul, Turkey
| | - Emine Eren-Kocak
- Institute of Neurological Sciences and Psychiatry, Hacettepe University, Sıhhiye, Ankara, Turkey
| | - Muge Yemisci
- Institute of Neurological Sciences and Psychiatry, Hacettepe University, Sıhhiye, Ankara, Turkey
| | - Hulya Karatas
- Institute of Neurological Sciences and Psychiatry, Hacettepe University, Sıhhiye, Ankara, Turkey
| | - Esra Erdemli
- Department of Histology and Embryology, Faculty of Medicine, Ankara University, Ankara, Turkey
| | - Ihsan Gursel
- Department of Molecular Biology and Genetics, Science Faculty, Bilkent University, Ankara, Turkey
- Izmir Biomedicine and Genome Center, Dokuz Eylul University, İzmir, Turkey
| | - Turgay Dalkara
- Institute of Neurological Sciences and Psychiatry, Hacettepe University, Sıhhiye, Ankara, Turkey.
| |
Collapse
|
28
|
Jiang S, Wu X, Yang F, Rommelfanger NJ, Hong G. Activation of mechanoluminescent nanotransducers by focused ultrasound enables light delivery to deep-seated tissue in vivo. Nat Protoc 2023; 18:3787-3820. [PMID: 37914782 PMCID: PMC11405139 DOI: 10.1038/s41596-023-00895-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2023] [Accepted: 07/27/2023] [Indexed: 11/03/2023]
Abstract
Light is used extensively in biological and medical research for optogenetic neuromodulation, fluorescence imaging, photoactivatable gene editing and light-based therapies. The major challenge to the in vivo implementation of light-based methods in deep-seated structures of the brain or of internal organs is the limited penetration of photons in biological tissue. The presence of light scattering and absorption has resulted in the development of invasive techniques such as the implantation of optical fibers, the insertion of endoscopes and the surgical removal of overlying tissues to overcome light attenuation and deliver it deep into the body. However, these procedures are highly invasive and make it difficult to reposition and adjust the illuminated area in each animal. Here, we detail a noninvasive approach to deliver light (termed 'deLight') in deep tissue via systemically injected mechanoluminescent nanotransducers that can be gated by using focused ultrasound. This approach achieves localized light emission with sub-millimeter resolution and millisecond response times in any vascularized organ of living mice without requiring invasive implantation of light-emitting devices. For example, deLight enables optogenetic neuromodulation in live mice without a craniotomy or brain implants. deLight provides a generalized method for applications that require a light source in deep tissues in vivo, such as deep-brain fluorescence imaging and photoactivatable genome editing. The implementation of the entire protocol for an in vivo application takes ~1-2 weeks.
Collapse
Affiliation(s)
- Shan Jiang
- Department of Materials Science and Engineering, Stanford University, Stanford, CA, USA
- Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA, USA
| | - Xiang Wu
- Department of Materials Science and Engineering, Stanford University, Stanford, CA, USA
- Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA, USA
| | - Fan Yang
- Department of Materials Science and Engineering, Stanford University, Stanford, CA, USA
- Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA, USA
| | - Nicholas J Rommelfanger
- Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA, USA
- Department of Applied Physics, Stanford University, Stanford, CA, USA
| | - Guosong Hong
- Department of Materials Science and Engineering, Stanford University, Stanford, CA, USA.
- Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA, USA.
| |
Collapse
|
29
|
Wang J, Lin J, Chen Y, Liu J, Zheng Q, Deng M, Wang R, Zhang Y, Feng S, Xu Z, Ye W, Hu Y, Duan J, Lin Y, Dai J, Chen Y, Li Y, Luo T, Chen Q, Lu Z. An ultra-compact promoter drives widespread neuronal expression in mouse and monkey brains. Cell Rep 2023; 42:113348. [PMID: 37910509 DOI: 10.1016/j.celrep.2023.113348] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Revised: 07/11/2023] [Accepted: 10/10/2023] [Indexed: 11/03/2023] Open
Abstract
Promoters are essential tools for basic and translational neuroscience research. An ideal promoter should possess the shortest possible DNA sequence with cell-type selectivity. However, whether ultra-compact promoters can offer neuron-specific expression is unclear. Here, we report the development of an extremely short promoter that enables selective gene expression in neurons, but not glial cells, in the brain. The promoter sequence originates from the human CALM1 gene and is only 120 bp in size. The CALM1 promoter (pCALM1) embedded in an adeno-associated virus (AAV) genome directed broad reporter expression in excitatory and inhibitory neurons in mouse and monkey brains. Moreover, pCALM1, when inserted into an all-in-one AAV vector expressing SpCas9 and sgRNA, drives constitutive and conditional in vivo gene editing in neurons and elicits functional alterations. These data demonstrate the ability of pCALM1 to conduct restricted neuronal gene expression, illustrating the feasibility of ultra-miniature promoters for targeting brain-cell subtypes.
Collapse
Affiliation(s)
- Jingyi Wang
- Department of Anesthesiology, Peking University Shenzhen Hospital, Shenzhen 518034, China; Shenzhen Technological Research Center for Primate Translational Medicine, Shenzhen Key Laboratory for Molecular Biology of Neural Development, Shenzhen-Hong Kong Institute of Brain Science, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jianbang Lin
- Shenzhen Technological Research Center for Primate Translational Medicine, Shenzhen Key Laboratory for Molecular Biology of Neural Development, Shenzhen-Hong Kong Institute of Brain Science, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yefei Chen
- Shenzhen Technological Research Center for Primate Translational Medicine, Shenzhen Key Laboratory for Molecular Biology of Neural Development, Shenzhen-Hong Kong Institute of Brain Science, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Jing Liu
- Shenzhen Technological Research Center for Primate Translational Medicine, Shenzhen Key Laboratory for Molecular Biology of Neural Development, Shenzhen-Hong Kong Institute of Brain Science, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China; Department of Anesthesiology, Affiliated Shenzhen Maternity & Child Healthcare Hospital, Southern Medical University, Shenzhen 518027, China
| | - Qiongping Zheng
- Shenzhen Technological Research Center for Primate Translational Medicine, Shenzhen Key Laboratory for Molecular Biology of Neural Development, Shenzhen-Hong Kong Institute of Brain Science, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Mao Deng
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan 528400, China
| | - Ruiqi Wang
- Shenzhen Technological Research Center for Primate Translational Medicine, Shenzhen Key Laboratory for Molecular Biology of Neural Development, Shenzhen-Hong Kong Institute of Brain Science, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Yujing Zhang
- Shenzhen Technological Research Center for Primate Translational Medicine, Shenzhen Key Laboratory for Molecular Biology of Neural Development, Shenzhen-Hong Kong Institute of Brain Science, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Shijing Feng
- Shenzhen Technological Research Center for Primate Translational Medicine, Shenzhen Key Laboratory for Molecular Biology of Neural Development, Shenzhen-Hong Kong Institute of Brain Science, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Zhenyan Xu
- Shenzhen Technological Research Center for Primate Translational Medicine, Shenzhen Key Laboratory for Molecular Biology of Neural Development, Shenzhen-Hong Kong Institute of Brain Science, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Weiyi Ye
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan 528400, China
| | - Yu Hu
- Shenzhen Technological Research Center for Primate Translational Medicine, Shenzhen Key Laboratory for Molecular Biology of Neural Development, Shenzhen-Hong Kong Institute of Brain Science, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Jiamei Duan
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan 528400, China; School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Yunping Lin
- Shenzhen Technological Research Center for Primate Translational Medicine, Shenzhen Key Laboratory for Molecular Biology of Neural Development, Shenzhen-Hong Kong Institute of Brain Science, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Ji Dai
- Shenzhen Technological Research Center for Primate Translational Medicine, Shenzhen Key Laboratory for Molecular Biology of Neural Development, Shenzhen-Hong Kong Institute of Brain Science, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China; University of Chinese Academy of Sciences, Beijing 100049, China; Guangdong Provincial Key Laboratory of Brain Connectome and Behavior, CAS Key Laboratory of Brain Connectome and Manipulation, The Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Yu Chen
- Shenzhen Technological Research Center for Primate Translational Medicine, Shenzhen Key Laboratory for Molecular Biology of Neural Development, Shenzhen-Hong Kong Institute of Brain Science, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China; University of Chinese Academy of Sciences, Beijing 100049, China; Guangdong Provincial Key Laboratory of Brain Connectome and Behavior, CAS Key Laboratory of Brain Connectome and Manipulation, The Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Yuantao Li
- Department of Anesthesiology, Affiliated Shenzhen Maternity & Child Healthcare Hospital, Southern Medical University, Shenzhen 518027, China; Biomedical Research Institute, Hubei University of Medicine, Shiyan 442000, China
| | - Tao Luo
- Department of Anesthesiology, Peking University Shenzhen Hospital, Shenzhen 518034, China
| | - Qian Chen
- University of Chinese Academy of Sciences, Beijing 100049, China; Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan 528400, China; School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China; Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China.
| | - Zhonghua Lu
- Shenzhen Technological Research Center for Primate Translational Medicine, Shenzhen Key Laboratory for Molecular Biology of Neural Development, Shenzhen-Hong Kong Institute of Brain Science, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China; University of Chinese Academy of Sciences, Beijing 100049, China; Guangdong Provincial Key Laboratory of Brain Connectome and Behavior, CAS Key Laboratory of Brain Connectome and Manipulation, The Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China; Biomedical Imaging Science and System Key Laboratory, Chinese Academy of Sciences, Shenzhen 518055, China.
| |
Collapse
|
30
|
Broersen R, Albergaria C, Carulli D, Carey MR, Canto CB, De Zeeuw CI. Synaptic mechanisms for associative learning in the cerebellar nuclei. Nat Commun 2023; 14:7459. [PMID: 37985778 PMCID: PMC10662440 DOI: 10.1038/s41467-023-43227-w] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Accepted: 11/03/2023] [Indexed: 11/22/2023] Open
Abstract
Associative learning during delay eyeblink conditioning (EBC) depends on an intact cerebellum. However, the relative contribution of changes in the cerebellar nuclei to learning remains a subject of ongoing debate. In particular, little is known about the changes in synaptic inputs to cerebellar nuclei neurons that take place during EBC and how they shape the membrane potential of these neurons. Here, we probed the ability of these inputs to support associative learning in mice, and investigated structural and cell-physiological changes within the cerebellar nuclei during learning. We find that optogenetic stimulation of mossy fiber afferents to the anterior interposed nucleus (AIP) can substitute for a conditioned stimulus and is sufficient to elicit conditioned responses (CRs) that are adaptively well-timed. Further, EBC induces structural changes in mossy fiber and inhibitory inputs, but not in climbing fiber inputs, and it leads to changes in subthreshold processing of AIP neurons that correlate with conditioned eyelid movements. The changes in synaptic and spiking activity that precede the CRs allow for a decoder to distinguish trials with a CR. Our data reveal how structural and physiological modifications of synaptic inputs to cerebellar nuclei neurons can facilitate learning.
Collapse
Affiliation(s)
- Robin Broersen
- Department of Cerebellar Coordination and Cognition, Netherlands Institute for Neuroscience, Royal Netherlands Academy of Arts and Sciences, Amsterdam, The Netherlands
- Department of Neuroscience, Erasmus MC, Rotterdam, The Netherlands
| | - Catarina Albergaria
- Neuroscience Program, Champalimaud Center for the Unknown, Lisbon, Portugal
- University College London, Sainsbury Wellcome Centre, London, UK
| | - Daniela Carulli
- Laboratory for Neuroregeneration, Netherlands Institute for Neuroscience, Royal Netherlands Academy of Arts and Sciences, Amsterdam, The Netherlands
| | - Megan R Carey
- Neuroscience Program, Champalimaud Center for the Unknown, Lisbon, Portugal.
| | - Cathrin B Canto
- Department of Cerebellar Coordination and Cognition, Netherlands Institute for Neuroscience, Royal Netherlands Academy of Arts and Sciences, Amsterdam, The Netherlands.
- Department of Neuroscience, Erasmus MC, Rotterdam, The Netherlands.
| | - Chris I De Zeeuw
- Department of Cerebellar Coordination and Cognition, Netherlands Institute for Neuroscience, Royal Netherlands Academy of Arts and Sciences, Amsterdam, The Netherlands.
- Department of Neuroscience, Erasmus MC, Rotterdam, The Netherlands.
| |
Collapse
|
31
|
Xiao D, Yan Y, Murphy TH. Mesotrode chronic simultaneous mesoscale cortical imaging and subcortical or peripheral nerve spiking activity recording in mice. eLife 2023; 12:RP87691. [PMID: 37962180 PMCID: PMC10645427 DOI: 10.7554/elife.87691] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2023] Open
Abstract
Brain function originates from hierarchical spatial-temporal neural dynamics distributed across cortical and subcortical networks. However, techniques available to assess large-scale brain network activity with single-neuron resolution in behaving animals remain limited. Here, we present Mesotrode that integrates chronic wide-field mesoscale cortical imaging and compact multi-site cortical/subcortical cellular electrophysiology in head-fixed mice that undergo self-initiated running or orofacial movements. Specifically, we harnessed the flexibility of chronic multi-site tetrode recordings to monitor single-neuron activity in multiple subcortical structures while simultaneously imaging the mesoscale activity of the entire dorsal cortex. A mesoscale spike-triggered averaging procedure allowed the identification of cortical activity motifs preferentially associated with single-neuron spiking. Using this approach, we were able to characterize chronic single-neuron-related functional connectivity maps for up to 60 days post-implantation. Neurons recorded from distinct subcortical structures display diverse but segregated cortical maps, suggesting that neurons of different origins participate in distinct cortico-subcortical pathways. We extended the capability of Mesotrode by implanting the micro-electrode at the facial motor nerve and found that facial nerve spiking is functionally associated with the PTA, RSP, and M2 network, and optogenetic inhibition of the PTA area significantly reduced the facial movement of the mice. These findings demonstrate that Mesotrode can be used to sample different combinations of cortico-subcortical networks over prolonged periods, generating multimodal and multi-scale network activity from a single implant, offering new insights into the neural mechanisms underlying specific behaviors.
Collapse
Affiliation(s)
- Dongsheng Xiao
- University of British Columbia, Department of Psychiatry, Kinsmen Laboratory of Neurological ResearchVancouverCanada
- Djavad Mowafaghian Centre for Brain Health, University of British ColumbiaVancouverCanada
| | - Yuhao Yan
- University of British Columbia, Department of Psychiatry, Kinsmen Laboratory of Neurological ResearchVancouverCanada
- Djavad Mowafaghian Centre for Brain Health, University of British ColumbiaVancouverCanada
| | - Timothy H Murphy
- University of British Columbia, Department of Psychiatry, Kinsmen Laboratory of Neurological ResearchVancouverCanada
- Djavad Mowafaghian Centre for Brain Health, University of British ColumbiaVancouverCanada
| |
Collapse
|
32
|
Quimby AE, Wei K, Adewole D, Eliades S, Cullen DK, Brant JA. Signal processing and stimulation potential within the ascending auditory pathway: a review. Front Neurosci 2023; 17:1277627. [PMID: 38027521 PMCID: PMC10658786 DOI: 10.3389/fnins.2023.1277627] [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: 08/14/2023] [Accepted: 10/12/2023] [Indexed: 12/01/2023] Open
Abstract
The human auditory system encodes sound with a high degree of temporal and spectral resolution. When hearing fails, existing neuroprosthetics such as cochlear implants may partially restore hearing through stimulation of auditory neurons at the level of the cochlea, though not without limitations inherent to electrical stimulation. Novel approaches to hearing restoration, such as optogenetics, offer the potential of improved performance. We review signal processing in the ascending auditory pathway and the current state of conventional and emerging neural stimulation strategies at various levels of the auditory system.
Collapse
Affiliation(s)
- Alexandra E. Quimby
- Department of Otolaryngology and Communication Sciences, SUNY Upstate Medical University, Syracuse, NY, United States
| | - Kimberly Wei
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Dayo Adewole
- Corporal Michael J. Crescenz Veterans Affairs Medical Center, Philadelphia, PA, United States
| | - Steven Eliades
- Department of Head and Neck Surgery and Communication Sciences, Duke University, Durham, NC, United States
| | - D. Kacy Cullen
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
- Corporal Michael J. Crescenz Veterans Affairs Medical Center, Philadelphia, PA, United States
| | - Jason A. Brant
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
- Department of Otorhinolaryngology – Head and Neck Surgery, University of Pennsylvania, Philadelphia, PA, United States
| |
Collapse
|
33
|
Zhang P, Ma D, Cheng X, Tsai AP, Tang Y, Gao HC, Fang L, Bi C, Landreth GE, Chubykin AA, Huang F. Deep learning-driven adaptive optics for single-molecule localization microscopy. Nat Methods 2023; 20:1748-1758. [PMID: 37770712 PMCID: PMC10630144 DOI: 10.1038/s41592-023-02029-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Accepted: 08/23/2023] [Indexed: 09/30/2023]
Abstract
The inhomogeneous refractive indices of biological tissues blur and distort single-molecule emission patterns generating image artifacts and decreasing the achievable resolution of single-molecule localization microscopy (SMLM). Conventional sensorless adaptive optics methods rely on iterative mirror changes and image-quality metrics. However, these metrics result in inconsistent metric responses and thus fundamentally limit their efficacy for aberration correction in tissues. To bypass iterative trial-then-evaluate processes, we developed deep learning-driven adaptive optics for SMLM to allow direct inference of wavefront distortion and near real-time compensation. Our trained deep neural network monitors the individual emission patterns from single-molecule experiments, infers their shared wavefront distortion, feeds the estimates through a dynamic filter and drives a deformable mirror to compensate sample-induced aberrations. We demonstrated that our method simultaneously estimates and compensates 28 wavefront deformation shapes and improves the resolution and fidelity of three-dimensional SMLM through >130-µm-thick brain tissue specimens.
Collapse
Affiliation(s)
- Peiyi Zhang
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, USA
| | - Donghan Ma
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, USA
- Davidson School of Chemical Engineering, Purdue University, West Lafayette, IN, USA
| | - Xi Cheng
- Department of Biological Sciences, Purdue University, West Lafayette, IN, USA
- Purdue Institute for Integrative Neuroscience, Purdue University, West Lafayette, IN, USA
| | - Andy P Tsai
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Yu Tang
- Department of Biological Sciences, Purdue University, West Lafayette, IN, USA
- Purdue Institute for Integrative Neuroscience, Purdue University, West Lafayette, IN, USA
| | - Hao-Cheng Gao
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, USA
| | - Li Fang
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, USA
| | - Cheng Bi
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, USA
| | - Gary E Landreth
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, USA.
- Department of Anatomy, Cell Biology and Physiology, Indiana University School of Medicine, Indianapolis, IN, USA.
| | - Alexander A Chubykin
- Department of Biological Sciences, Purdue University, West Lafayette, IN, USA.
- Purdue Institute for Integrative Neuroscience, Purdue University, West Lafayette, IN, USA.
| | - Fang Huang
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, USA.
- Purdue Institute for Integrative Neuroscience, Purdue University, West Lafayette, IN, USA.
- Purdue Institute of Inflammation, Immunology and Infectious Disease, Purdue University, West Lafayette, IN, USA.
| |
Collapse
|
34
|
Andersen MA, Schouenborg J. Polydimethylsiloxane as a more biocompatible alternative to glass in optogenetics. Sci Rep 2023; 13:16090. [PMID: 37752160 PMCID: PMC10522705 DOI: 10.1038/s41598-023-43297-2] [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: 05/12/2023] [Accepted: 09/21/2023] [Indexed: 09/28/2023] Open
Abstract
Optogenetics is highly useful to stimulate or inhibit defined neuronal populations and is often used together with electrophysiological recordings. Due to poor penetration of light in tissue, there is a need for biocompatible wave guides. Glass wave guides are relatively stiff and known to cause glia reaction that likely influence the activity in the remaining neurons. We developed highly flexible micro wave guides for optogenetics that can be used in combination with long-lasting electrophysiological recordings. We designed and evaluated polydimethylsiloxane (PDMS) mono-fibers, which use the tissue as cladding, with a diameter of 71 ± 10 µm and 126 ± 5 µm. We showed that micro PDMS fibers transmitted 9-33 mW/mm2 light energy enough to activate channelrhodopsin. This was confirmed in acute extracellular recordings in vivo in which optogenetic stimulation through the PDMS fibers generated action potentials in rat hippocampus with a short onset latency. PDMS fibers had significantly less microglia and astrocytic activation in the zone nearest to the implant as compared to glass. There was no obvious difference in number of adjacent neurons between size matched wave guides. Micro PDMS wave guide demonstrates in vivo functionality and improved biocompatibility as compared to glass. This enables the delivery of light with less tissue damage.
Collapse
Affiliation(s)
- Michael Aagaard Andersen
- Neuronano Research Center, Department of Experimental Medicine, Lund University, Lund, Sweden.
- Department of Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.
| | - Jens Schouenborg
- Neuronano Research Center, Department of Experimental Medicine, Lund University, Lund, Sweden
| |
Collapse
|
35
|
Dell’Orco M, Weisend JE, Perrone-Bizzozero NI, Carlson AP, Morton RA, Linsenbardt DN, Shuttleworth CW. Repetitive Spreading Depolarization induces gene expression changes related to synaptic plasticity and neuroprotective pathways. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.02.27.530317. [PMID: 36909568 PMCID: PMC10002705 DOI: 10.1101/2023.02.27.530317] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/06/2023]
Abstract
Spreading depolarization (SD) is a slowly propagating wave of profound depolarization that sweeps through cortical tissue. While much emphasis has been placed on the damaging consequences of SD, there is uncertainty surrounding the potential activation of beneficial pathways such as cell survival and plasticity. The present study used unbiased assessments of gene expression to evaluate that compensatory and repair mechanisms could be recruited following SD, regardless of the induction method, which prior to this work had not been assessed. We also tested assumptions of appropriate controls and the spatial extent of expression changes that are important for in vivo SD models. SD clusters were induced with either KCl focal application or optogenetic stimulation in healthy mice. Cortical RNA was extracted and sequenced to identify differentially expressed genes (DEGs). SDs using both induction methods significantly upregulated 16 genes (versus sham animals) that included the cell proliferation-related genes FOS, JUN, and DUSP6, the plasticity-related genes ARC and HOMER1, and the inflammation-related genes PTGS2, EGR2, and NR4A1. The contralateral hemisphere is commonly used as control tissue for DEG studies, but its activity could be modified by near-global disruption of activity in the adjacent brain. We found 21 upregulated genes when comparing SD-involved cortex versus tissue from the contralateral hemisphere of the same animals. Interestingly, there was almost complete overlap (21/16) with the DEGs identified using sham controls. Neuronal activity also differs in SD initiation zones, where sustained global depolarization is required to initiate propagating events. We found that gene expression varied as a function of the distance from the SD initiation site, with greater expression differences observed in regions further away. Functional and pathway enrichment analyses identified axonogenesis, branching, neuritogenesis, and dendritic growth as significantly enriched in overlapping DEGs. Increased expression of SD-induced genes was also associated with predicted inhibition of pathways associated with cell death, and apoptosis. These results identify novel biological pathways that could be involved in plasticity and/or circuit modification in brain tissue impacted by SD. These results also identify novel functional targets that could be tested to determine potential roles in recovery and survival of peri-infarct tissues.
Collapse
Affiliation(s)
- Michela Dell’Orco
- Department of Neurosciences, University of New Mexico School of Medicine, Albuquerque, New Mexico, 87131, USA
| | - Jordan E. Weisend
- Department of Neurosciences, University of New Mexico School of Medicine, Albuquerque, New Mexico, 87131, USA
| | - Nora I. Perrone-Bizzozero
- Department of Neurosciences, University of New Mexico School of Medicine, Albuquerque, New Mexico, 87131, USA
| | - Andrew P. Carlson
- Department of Neurosurgery, University of New Mexico School of Medicine, Albuquerque, New Mexico, 87131, USA
| | - Russell A. Morton
- Department of Neurosciences, University of New Mexico School of Medicine, Albuquerque, New Mexico, 87131, USA
| | - David N Linsenbardt
- Department of Neurosciences, University of New Mexico School of Medicine, Albuquerque, New Mexico, 87131, USA
| | - C. William Shuttleworth
- Department of Neurosciences, University of New Mexico School of Medicine, Albuquerque, New Mexico, 87131, USA
| |
Collapse
|
36
|
Sha MFR, Koga Y, Murata Y, Taniguchi M, Yamaguchi M. Learning-dependent structural plasticity of intracortical and sensory connections to functional domains of the olfactory tubercle. Front Neurosci 2023; 17:1247375. [PMID: 37680965 PMCID: PMC10480507 DOI: 10.3389/fnins.2023.1247375] [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: 06/26/2023] [Accepted: 08/09/2023] [Indexed: 09/09/2023] Open
Abstract
The olfactory tubercle (OT), which is a component of the olfactory cortex and ventral striatum, has functional domains that play a role in odor-guided motivated behaviors. Learning odor-guided attractive and aversive behavior activates the anteromedial (am) and lateral (l) domains of the OT, respectively. However, the mechanism driving learning-dependent activation of specific OT domains remains unknown. We hypothesized that the neuronal connectivity of OT domains is plastically altered through olfactory experience. To examine the plastic potential of synaptic connections to OT domains, we optogenetically stimulated intracortical inputs from the piriform cortex or sensory inputs from the olfactory bulb to the OT in mice in association with a food reward for attractive learning and electrical foot shock for aversive learning. For both intracortical and sensory connections, axon boutons that terminated in the OT domains were larger in the amOT than in the lOT for mice exhibiting attractive learning and larger in the lOT than in the amOT for mice exhibiting aversive learning. These results indicate that both intracortical and sensory connections to the OT domains have learning-dependent plastic potential, suggesting that this plasticity underlies learning-dependent activation of specific OT domains and the acquisition of appropriate motivated behaviors.
Collapse
Affiliation(s)
| | | | | | | | - Masahiro Yamaguchi
- Department of Physiology, Kochi Medical School, Kochi University, Kochi, Japan
| |
Collapse
|
37
|
Kwan WC, Brunton EK, Begeng JM, Richardson RT, Ibbotson MR, Tong W. Timing is Everything: Stochastic Optogenetic Stimulation Reduces Adaptation in Retinal Ganglion Cells. ANNUAL INTERNATIONAL CONFERENCE OF THE IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. ANNUAL INTERNATIONAL CONFERENCE 2023; 2023:1-4. [PMID: 38083106 DOI: 10.1109/embc40787.2023.10340849] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2023]
Abstract
Optogenetics gives us unprecedented power to investigate brain connectivity. The ability to activate neural circuits with single cell resolution and its ease of application has provided a wealth of knowledge in brain function. More recently, optogenetics has shown tremendous utility in prosthetics applications, including vision restoration for patients with retinitis pigmentosa. One of the disadvantages of optogenetics, however, is its poor temporal bandwidth, i.e. the cell's inability to fire at a rate that matches the optical stimulation rate at high frequencies (>30 Hz). This research proposes a new strategy to overcome the temporal limits of optogenetic stimulation. Using whole-cell current clamp recordings in mouse retinal ganglion cells expressing channelrhodopsin-2 (H134R variant), we observed that randomizing inter-pulse intervals can significantly increase a retinal ganglion cell's temporal response to high frequency stimulation.Clinical Relevance- A significant disadvantage of optogenetic stimulation is its poor temporal dynamics which prohibit its widespread use in retinal prosthetics. We have shown that randomizing the interval between stimulation pulses reduces adaptation in retinal ganglion cells. This stimulation strategy may contribute to new levels of functional restoration in therapeutics which incorporate optogenetics.
Collapse
|
38
|
Kim S, Moon HS, Vo TT, Kim CH, Im GH, Lee S, Choi M, Kim SG. Whole-brain mapping of effective connectivity by fMRI with cortex-wide patterned optogenetics. Neuron 2023; 111:1732-1747.e6. [PMID: 37001524 DOI: 10.1016/j.neuron.2023.03.002] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Revised: 11/23/2022] [Accepted: 03/02/2023] [Indexed: 04/03/2023]
Abstract
Functional magnetic resonance imaging (fMRI) with optogenetic neural manipulation is a powerful tool that enables brain-wide mapping of effective functional networks. To achieve flexible manipulation of neural excitation throughout the mouse cortex, we incorporated spatiotemporal programmable optogenetic stimuli generated by a digital micromirror device into an MRI scanner via an optical fiber bundle. This approach offered versatility in space and time in planning the photostimulation pattern, combined with in situ optical imaging and cell-type-specific or circuit-specific genetic targeting in individual mice. Brain-wide effective connectivity obtained by fMRI with optogenetic stimulation of atlas-based cortical regions is generally congruent with anatomically defined axonal tracing data but is affected by the types of anesthetics that act selectively on specific connections. fMRI combined with flexible optogenetics opens a new path to investigate dynamic changes in functional brain states in the same animal through high-throughput brain-wide effective connectivity mapping.
Collapse
Affiliation(s)
- Seonghoon Kim
- Center for Neuroscience Imaging Research, Institute for Basic Science, Suwon, Republic of Korea; School of Biological Sciences, Seoul National University, Seoul, Republic of Korea
| | - Hyun Seok Moon
- Center for Neuroscience Imaging Research, Institute for Basic Science, Suwon, Republic of Korea; Department of Biomedical Engineering, Sungkyunkwan University, Suwon, Republic of Korea; Department of Intelligent Precision Healthcare Convergence, Sungkyunkwan University, Suwon, Republic of Korea
| | - Thanh Tan Vo
- Center for Neuroscience Imaging Research, Institute for Basic Science, Suwon, Republic of Korea; Department of Biomedical Engineering, Sungkyunkwan University, Suwon, Republic of Korea; Department of Intelligent Precision Healthcare Convergence, Sungkyunkwan University, Suwon, Republic of Korea
| | - Chang-Ho Kim
- School of Biological Sciences, Seoul National University, Seoul, Republic of Korea; Institute of Molecular Biology and Genetics, Seoul National University, Seoul, Republic of Korea
| | - Geun Ho Im
- Center for Neuroscience Imaging Research, Institute for Basic Science, Suwon, Republic of Korea
| | - Sungho Lee
- School of Biological Sciences, Seoul National University, Seoul, Republic of Korea
| | - Myunghwan Choi
- Center for Neuroscience Imaging Research, Institute for Basic Science, Suwon, Republic of Korea; School of Biological Sciences, Seoul National University, Seoul, Republic of Korea; Institute of Molecular Biology and Genetics, Seoul National University, Seoul, Republic of Korea.
| | - Seong-Gi Kim
- Center for Neuroscience Imaging Research, Institute for Basic Science, Suwon, Republic of Korea; Department of Biomedical Engineering, Sungkyunkwan University, Suwon, Republic of Korea; Department of Intelligent Precision Healthcare Convergence, Sungkyunkwan University, Suwon, Republic of Korea.
| |
Collapse
|
39
|
Martín-Cortecero J, Isaías-Camacho EU, Boztepe B, Ziegler K, Mease RA, Groh A. Monosynaptic trans-collicular pathways link mouse whisker circuits to integrate somatosensory and motor cortical signals. PLoS Biol 2023; 21:e3002126. [PMID: 37205722 DOI: 10.1371/journal.pbio.3002126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2023] [Accepted: 04/14/2023] [Indexed: 05/21/2023] Open
Abstract
The superior colliculus (SC), a conserved midbrain node with extensive long-range connectivity throughout the brain, is a key structure for innate behaviors. Descending cortical pathways are increasingly recognized as central control points for SC-mediated behaviors, but how cortico-collicular pathways coordinate SC activity at the cellular level is poorly understood. Moreover, despite the known role of the SC as a multisensory integrator, the involvement of the SC in the somatosensory system is largely unexplored in comparison to its involvement in the visual and auditory systems. Here, we mapped the connectivity of the whisker-sensitive region of the SC in mice with trans-synaptic and intersectional tracing tools and in vivo electrophysiology. The results reveal a novel trans-collicular connectivity motif in which neurons in motor- and somatosensory cortices impinge onto the brainstem-SC-brainstem sensory-motor arc and onto SC-midbrain output pathways via only one synapse in the SC. Intersectional approaches and optogenetically assisted connectivity quantifications in vivo reveal convergence of motor and somatosensory cortical input on individual SC neurons, providing a new framework for sensory-motor integration in the SC. More than a third of the cortical recipient neurons in the whisker SC are GABAergic neurons, which include a hitherto unknown population of GABAergic projection neurons targeting thalamic nuclei and the zona incerta. These results pinpoint a whisker region in the SC of mice as a node for the integration of somatosensory and motor cortical signals via parallel excitatory and inhibitory trans-collicular pathways, which link cortical and subcortical whisker circuits for somato-motor integration.
Collapse
Affiliation(s)
- Jesús Martín-Cortecero
- Medical Biophysics, Institute for Physiology and Pathophysiology, Heidelberg University, Germany
| | | | - Berin Boztepe
- Medical Biophysics, Institute for Physiology and Pathophysiology, Heidelberg University, Germany
| | - Katharina Ziegler
- Medical Biophysics, Institute for Physiology and Pathophysiology, Heidelberg University, Germany
| | - Rebecca Audrey Mease
- Medical Biophysics, Institute for Physiology and Pathophysiology, Heidelberg University, Germany
| | - Alexander Groh
- Medical Biophysics, Institute for Physiology and Pathophysiology, Heidelberg University, Germany
| |
Collapse
|
40
|
Sugimoto K, Yang J, Fischer P, Takizawa T, Mulder I, Qin T, Erdogan TD, Yaseen MA, Sakadžić S, Chung DY, Ayata C. Optogenetic Spreading Depolarizations Do Not Worsen Acute Ischemic Stroke Outcome. Stroke 2023; 54:1110-1119. [PMID: 36876481 PMCID: PMC10050120 DOI: 10.1161/strokeaha.122.041351] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2022] [Accepted: 02/01/2023] [Indexed: 03/07/2023]
Abstract
BACKGROUND Spreading depolarizations (SDs) are believed to contribute to injury progression and worsen outcomes in focal cerebral ischemia because exogenously induced SDs have been associated with enlarged infarct volumes. However, previous studies used highly invasive methods to trigger SDs that can directly cause tissue injury (eg, topical KCl) and confound the interpretation. Here, we tested whether SDs indeed enlarge infarcts when induced via a novel, noninjurious method using optogenetics. METHODS Using transgenic mice expressing channelrhodopsin-2 in neurons (Thy1-ChR2-YFP), we induced 8 optogenetic SDs to trigger SDs noninvasively at a remote cortical location in a noninjurious manner during 1-hour distal microvascular clip or proximal an endovascular filament occlusion of the middle cerebral artery. Laser speckle imaging was used to monitor cerebral blood flow. Infarct volumes were then quantified at 24 or 48 hours. RESULTS Infarct volumes in the optogenetic SD arm did not differ from the control arm in either distal or proximal middle cerebral artery occlusion, despite a 6-fold and 4-fold higher number of SDs, respectively. Identical optogenetic illumination in wild-type mice did not affect the infarct volume. Full-field laser speckle imaging showed that optogenetic stimulation did not affect the perfusion in the peri-infarct cortex. CONCLUSIONS Altogether, these data show that SDs induced noninvasively using optogenetics do not worsen tissue outcomes. Our findings compel a careful reexamination of the notion that SDs are causally linked to infarct expansion.
Collapse
Affiliation(s)
- Kazutaka Sugimoto
- Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02129
- Department of Neurosurgery, Yamaguchi University School of Medicine, Ube, Yamaguchi 7558505, Japan
| | - Joanna Yang
- Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02129
| | - Paul Fischer
- Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02129
| | - Tsubasa Takizawa
- Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02129
| | - Inge Mulder
- Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02129
| | - Tao Qin
- Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02129
| | - Taylan D. Erdogan
- Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02129
| | - Mohammad A. Yaseen
- Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown, MA 02129
| | - Sava Sakadžić
- Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown, MA 02129
| | - David Y. Chung
- Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02129
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114
| | - Cenk Ayata
- Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02129
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114
| |
Collapse
|
41
|
Rodrigues AF, Rebelo C, Reis T, Simões S, Bernardino L, Peça J, Ferreira L. Engineering optical tools for remotely controlled brain stimulation and regeneration. Biomater Sci 2023; 11:3034-3050. [PMID: 36947145 DOI: 10.1039/d2bm02059a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/20/2023]
Abstract
Neurological disorders are one of the world's leading medical and societal challenges due to the lack of efficacy of the first line treatment. Although pharmacological and non-pharmacological interventions have been employed with the aim of regulating neuronal activity and survival, they have failed to avoid symptom relapse and disease progression in the vast majority of patients. In the last 5 years, advanced drug delivery systems delivering bioactive molecules and neuromodulation strategies have been developed to promote tissue regeneration and remodel neuronal circuitry. However, both approaches still have limited spatial and temporal precision over the desired target regions. While external stimuli such as electromagnetic fields and ultrasound have been employed in the clinic for non-invasive neuromodulation, they do not have the capability of offering single-cell spatial resolution as light stimulation. Herein, we review the latest progress in this area of study and discuss the prospects of using light-responsive nanomaterials to achieve on-demand delivery of drugs and neuromodulation, with the aim of achieving brain stimulation and regeneration.
Collapse
Affiliation(s)
- Artur Filipe Rodrigues
- Center for Neurosciences and Cell Biology, Center for Innovative Biomedicine and Biotechnology, University of Coimbra, 3000-517 Coimbra, Portugal.
- Institute of Interdisciplinary Research, University of Coimbra, 3000-354 Coimbra, Portugal
| | - Catarina Rebelo
- Center for Neurosciences and Cell Biology, Center for Innovative Biomedicine and Biotechnology, University of Coimbra, 3000-517 Coimbra, Portugal.
- Institute of Interdisciplinary Research, University of Coimbra, 3000-354 Coimbra, Portugal
- Faculty of Medicine, Pólo das Ciências da Saúde, Unidade Central, University of Coimbra, 3000-354 Coimbra, Portugal.
| | - Tiago Reis
- Center for Neurosciences and Cell Biology, Center for Innovative Biomedicine and Biotechnology, University of Coimbra, 3000-517 Coimbra, Portugal.
- Institute of Interdisciplinary Research, University of Coimbra, 3000-354 Coimbra, Portugal
- Faculty of Medicine, Pólo das Ciências da Saúde, Unidade Central, University of Coimbra, 3000-354 Coimbra, Portugal.
| | - Susana Simões
- Center for Neurosciences and Cell Biology, Center for Innovative Biomedicine and Biotechnology, University of Coimbra, 3000-517 Coimbra, Portugal.
- Institute of Interdisciplinary Research, University of Coimbra, 3000-354 Coimbra, Portugal
- Faculty of Medicine, Pólo das Ciências da Saúde, Unidade Central, University of Coimbra, 3000-354 Coimbra, Portugal.
| | - Liliana Bernardino
- Health Sciences Research Centre, Faculty of Health Sciences, University of Beira Interior, 6201-506 Covilhã, Portugal
| | - João Peça
- Center for Neurosciences and Cell Biology, Center for Innovative Biomedicine and Biotechnology, University of Coimbra, 3000-517 Coimbra, Portugal.
- Institute of Interdisciplinary Research, University of Coimbra, 3000-354 Coimbra, Portugal
- Faculty of Medicine, Pólo das Ciências da Saúde, Unidade Central, University of Coimbra, 3000-354 Coimbra, Portugal.
| | - Lino Ferreira
- Center for Neurosciences and Cell Biology, Center for Innovative Biomedicine and Biotechnology, University of Coimbra, 3000-517 Coimbra, Portugal.
- Institute of Interdisciplinary Research, University of Coimbra, 3000-354 Coimbra, Portugal
- Faculty of Medicine, Pólo das Ciências da Saúde, Unidade Central, University of Coimbra, 3000-354 Coimbra, Portugal.
| |
Collapse
|
42
|
Pollmann EH, Yin H, Uguz I, Dubey A, Wingel KE, Choi JS, Moazeni S, Gilhotra Y, Pavlovsky VA, Banees A, Boominathan V, Robinson J, Veeraraghavan A, Pieribone VA, Pesaran B, Shepard KL. Subdural CMOS optical probe (SCOPe) for bidirectional neural interfacing. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.02.07.527500. [PMID: 36798295 PMCID: PMC9934536 DOI: 10.1101/2023.02.07.527500] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/11/2023]
Abstract
Optical neurotechnologies use light to interface with neurons and can monitor and manipulate neural activity with high spatial-temporal precision over large cortical extents. While there has been significant progress in miniaturizing microscope for head-mounted configurations, these existing devices are still very bulky and could never be fully implanted. Any viable translation of these technologies to human use will require a much more noninvasive, fully implantable form factor. Here, we leverage advances in microelectronics and heterogeneous optoelectronic packaging to develop a transformative, ultrathin, miniaturized device for bidirectional optical stimulation and recording: the subdural CMOS Optical Probe (SCOPe). By being thin enough to lie entirely within the subdural space of the primate brain, SCOPe defines a path for the eventual human translation of a new generation of brain-machine interfaces based on light.
Collapse
|
43
|
Alberto GE, Klorig DC, Goldstein AT, Godwin DW. Alcohol withdrawal produces changes in excitability, population discharge probability, and seizure threshold. ALCOHOL, CLINICAL & EXPERIMENTAL RESEARCH 2023; 47:211-218. [PMID: 36543333 PMCID: PMC10197957 DOI: 10.1111/acer.15004] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 12/15/2022] [Accepted: 12/19/2022] [Indexed: 12/24/2022]
Abstract
BACKGROUND Alcohol withdrawal syndrome (AWS) results from the sudden cessation of chronic alcohol use and is associated with high morbidity and mortality. Alcohol withdrawal-induced central nervous system (CNS) hyperexcitability results from complex, compensatory changes in synaptic efficacy and intrinsic excitability. These changes in excitability counteract the depressing effects of chronic ethanol on neural transmission and underlie symptoms of AWS, which range from mild anxiety to seizures and death. The development of targeted pharmacotherapies for treating AWS has been slow, due in part to the lack of available animal models that capture the key features of human AWS. Using a unique optogenetic method of probing network excitability, we examined electrophysiologic correlates of hyperexcitability sensitive to early changes in CNS excitability. This method is sensitive to pharmacologic treatments that reduce excitability and may represent a platform for AWS drug development. METHODS We applied a newly developed method, the optogenetic population discharge threshold (oPDT), which uses light intensity response curves to measure network excitability in chronically implanted mice. Excitability was tracked using the oPDT before, during, and after the chronic intermittent exposure (CIE) model of alcohol withdrawal (WD). RESULTS Alcohol withdrawal produced a dose-dependent leftward shift in the oPDT curve (denoting increased excitability), which was detectable in as few as three exposure cycles. This shift in excitability mirrored an increase in the number of spontaneous interictal spikes during withdrawal. In addition, Withdrawal lowered seizure thresholds and increased seizure severity in optogenetically kindled mice. CONCLUSION We demonstrate that the oPDT provides a sensitive measure of alcohol withdrawal-induced hyperexcitability. The ability to actively probe the progression of excitability without eliciting potentially confounding seizures promises to be a useful tool in the preclinical development of next-generation pharmacotherapies for AWS.
Collapse
Affiliation(s)
- Gregory E. Alberto
- Wake Forest School of Medicine; Department of Neurobiology and Anatomy; Winston-Salem, NC, USA
- Wake Forest School of Medicine; Department of Neurology; Winston-Salem, NC, USA
- Dartmouth-Hitchcock Medical Center; Department of Psychiatry; Lebanon, NH, USA
| | - David C. Klorig
- Wake Forest School of Medicine; Department of Neurobiology and Anatomy; Winston-Salem, NC, USA
- Wake Forest School of Medicine; Department of Neurology; Winston-Salem, NC, USA
- Wake Forest School of Medicine; Department of Physiology and Pharmacology; Winston-Salem, NC, USA
| | - Allison T. Goldstein
- Wake Forest School of Medicine; Department of Neurobiology and Anatomy; Winston-Salem, NC, USA
| | - Dwayne W. Godwin
- Wake Forest School of Medicine; Department of Neurobiology and Anatomy; Winston-Salem, NC, USA
- Wake Forest School of Medicine; Department of Neurology; Winston-Salem, NC, USA
- Research and Education Department, W.G. (Bill) Hefner Veterans Affairs Medical Center; Salisbury, NC, USA
- Wake Forest School of Medicine; Department of Physiology and Pharmacology; Winston-Salem, NC, USA
| |
Collapse
|
44
|
Gerfen CR. Segregation of D1 and D2 dopamine receptors in the striatal direct and indirect pathways: An historical perspective. Front Synaptic Neurosci 2023; 14:1002960. [PMID: 36741471 PMCID: PMC9892636 DOI: 10.3389/fnsyn.2022.1002960] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Accepted: 12/05/2022] [Indexed: 01/20/2023] Open
Abstract
The direct and indirect striatal pathways form a cornerstone of the circuits of the basal ganglia. Dopamine has opponent affects on the function of these pathways due to the segregation of the D1- and D2-dopamine receptors in the spiny projection neurons giving rise to the direct and indirect pathways. An historical perspective is provided on the discovery of dopamine receptor segregation leading to models of how the direct and indirect affect motor behavior.
Collapse
|
45
|
Wang W, Wu X, Kevin Tang KW, Pyatnitskiy I, Taniguchi R, Lin P, Zhou R, Capocyan SLC, Hong G, Wang H. Ultrasound-Triggered In Situ Photon Emission for Noninvasive Optogenetics. J Am Chem Soc 2023; 145:1097-1107. [PMID: 36606703 DOI: 10.1021/jacs.2c10666] [Citation(s) in RCA: 35] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Optogenetics has revolutionized neuroscience understanding by allowing spatiotemporal control over cell-type specific neurons in neural circuits. However, the sluggish development of noninvasive photon delivery in the brain has limited the clinical application of optogenetics. Focused ultrasound (FUS)-derived mechanoluminescence has emerged as a promising tool for in situ photon emission, but there is not yet a biocompatible liquid-phase mechanoluminescence system for spatiotemporal optogenetics. To achieve noninvasive optogenetics with a high temporal resolution and desirable biocompatibility, we have developed liposome (Lipo@IR780/L012) nanoparticles for FUS-triggered mechanoluminescence in brain photon delivery. Synchronized and stable blue light emission was generated in solution under FUS irradiation due to the cascade reactions in liposomes. In vitro tests revealed that Lipo@IR780/L012 could be triggered by FUS for light emission at different stimulation frequencies, resulting in activation of opsin-expressing spiking HEK cells under the FUS irradiation. In vivo optogenetic stimulation further demonstrated that motor cortex neurons could be noninvasively and reversibly activated under the repetitive FUS irradiation after intravenous injection of lipid nanoparticles to achieve limb movements.
Collapse
Affiliation(s)
- Wenliang Wang
- Biomedical Engineering Cockrell School of Engineering, The University of Texas at Austin, Austin, Texas 78712, United States
| | - Xiang Wu
- Department of Materials Science and Engineering, Stanford University, Stanford, California 94305, United States.,Wu Tsai Neurosciences Institute, Stanford University, Stanford, California 94305, United States
| | - Kai Wing Kevin Tang
- Biomedical Engineering Cockrell School of Engineering, The University of Texas at Austin, Austin, Texas 78712, United States
| | - Ilya Pyatnitskiy
- Biomedical Engineering Cockrell School of Engineering, The University of Texas at Austin, Austin, Texas 78712, United States
| | - Rayna Taniguchi
- Biomedical Engineering Cockrell School of Engineering, The University of Texas at Austin, Austin, Texas 78712, United States
| | - Peter Lin
- Biomedical Engineering Cockrell School of Engineering, The University of Texas at Austin, Austin, Texas 78712, United States
| | - Richard Zhou
- Biomedical Engineering Cockrell School of Engineering, The University of Texas at Austin, Austin, Texas 78712, United States
| | - Sam Lander C Capocyan
- Biomedical Engineering Cockrell School of Engineering, The University of Texas at Austin, Austin, Texas 78712, United States
| | - Guosong Hong
- Department of Materials Science and Engineering, Stanford University, Stanford, California 94305, United States.,Wu Tsai Neurosciences Institute, Stanford University, Stanford, California 94305, United States
| | - Huiliang Wang
- Biomedical Engineering Cockrell School of Engineering, The University of Texas at Austin, Austin, Texas 78712, United States
| |
Collapse
|
46
|
Hu R, Shankar J, Dong GZ, Villar PS, Araneda RC. α 2-Adrenergic modulation of I h in adult-born granule cells in the olfactory bulb. Front Cell Neurosci 2023; 16:1055569. [PMID: 36687519 PMCID: PMC9853206 DOI: 10.3389/fncel.2022.1055569] [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: 09/28/2022] [Accepted: 11/29/2022] [Indexed: 01/09/2023] Open
Abstract
In the olfactory bulb (OB), a large population of axon-less inhibitory interneurons, the granule cells (GCs), coordinate network activity and tune the output of principal neurons, the mitral and tufted cells (MCs), through dendrodendritic interactions. Furthermore, GCs undergo neurogenesis throughout life, providing a source of plasticity to the neural network of the OB. The function and integration of GCs in the OB are regulated by several afferent neuromodulatory signals, including noradrenaline (NA), a state-dependent neuromodulator that plays a crucial role in the regulation of cortical function and task-specific decision processes. However, the mechanisms by which NA regulates GC function are not fully understood. Here, we show that NA modulates hyperpolarization-activated currents (Ih) via the activation of α2-adrenergic receptors (ARs) in adult-born GCs (abGCs), thus directly acting on channels that play essential roles in regulating neuronal excitability and network oscillations in the brain. This modulation affects the dendrodendritic output of GCs leading to an enhancement of lateral inhibition onto the MCs. Furthermore, we show that NA modulates subthreshold resonance in GCs, which could affect the temporal integration of abGCs. Together, these results provide a novel mechanism by which a state-dependent neuromodulator acting on Ih can regulate GC function in the OB.
Collapse
|
47
|
Innate Immunity in Cardiovascular Diseases-Identification of Novel Molecular Players and Targets. J Clin Med 2023; 12:jcm12010335. [PMID: 36615135 PMCID: PMC9821340 DOI: 10.3390/jcm12010335] [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: 11/01/2022] [Revised: 12/20/2022] [Accepted: 12/25/2022] [Indexed: 01/03/2023] Open
Abstract
During the past few years, unexpected developments have driven studies in the field of clinical immunology. One driver of immense impact was the outbreak of a pandemic caused by the novel virus SARS-CoV-2. Excellent recent reviews address diverse aspects of immunological re-search into cardiovascular diseases. Here, we specifically focus on selected studies taking advantage of advanced state-of-the-art molecular genetic methods ranging from genome-wide epi/transcriptome mapping and variant scanning to optogenetics and chemogenetics. First, we discuss the emerging clinical relevance of advanced diagnostics for cardiovascular diseases, including those associated with COVID-19-with a focus on the role of inflammation in cardiomyopathies and arrhythmias. Second, we consider newly identified immunological interactions at organ and system levels which affect cardiovascular pathogenesis. Thus, studies into immune influences arising from the intestinal system are moving towards therapeutic exploitation. Further, powerful new research tools have enabled novel insight into brain-immune system interactions at unprecedented resolution. This latter line of investigation emphasizes the strength of influence of emotional stress-acting through defined brain regions-upon viral and cardiovascular disorders. Several challenges need to be overcome before the full impact of these far-reaching new findings will hit the clinical arena.
Collapse
|
48
|
Hádinger N, Bősz E, Tóth B, Vantomme G, Lüthi A, Acsády L. Region-selective control of the thalamic reticular nucleus via cortical layer 5 pyramidal cells. Nat Neurosci 2023; 26:116-130. [PMID: 36550291 PMCID: PMC9829539 DOI: 10.1038/s41593-022-01217-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Accepted: 10/26/2022] [Indexed: 12/24/2022]
Abstract
Corticothalamic pathways, responsible for the top-down control of the thalamus, have a canonical organization such that every cortical region sends output from both layer 6 (L6) and layer 5 (L5) to the thalamus. Here we demonstrate a qualitative, region-specific difference in the organization of mouse corticothalamic pathways. Specifically, L5 pyramidal cells of the frontal cortex, but not other cortical regions, establish monosynaptic connections with the inhibitory thalamic reticular nucleus (TRN). The frontal L5-TRN pathway parallels the L6-TRN projection but has distinct morphological and physiological features. The exact spike output of the L5-contacted TRN cells correlated with the level of cortical synchrony. Optogenetic perturbation of the L5-TRN connection disrupted the tight link between cortical and TRN activity. L5-driven TRN cells innervated thalamic nuclei involved in the control of frontal cortex activity. Our data show that frontal cortex functions require a highly specialized cortical control over intrathalamic inhibitory processes.
Collapse
Affiliation(s)
- Nóra Hádinger
- Laboratory of Thalamus Research, Institute of Experimental Medicine, Budapest, Hungary.
| | - Emília Bősz
- Laboratory of Thalamus Research, Institute of Experimental Medicine, Budapest, Hungary
- János Szentágothai Doctoral School of Neurosciences, Semmelweis University, Budapest, Hungary
| | - Boglárka Tóth
- Laboratory of Thalamus Research, Institute of Experimental Medicine, Budapest, Hungary
| | - Gil Vantomme
- Department of Fundamental Neurosciences, University of Lausanne, Lausanne, Switzerland
| | - Anita Lüthi
- Department of Fundamental Neurosciences, University of Lausanne, Lausanne, Switzerland
| | - László Acsády
- Laboratory of Thalamus Research, Institute of Experimental Medicine, Budapest, Hungary.
| |
Collapse
|
49
|
Bi X, Beck C, Gong Y. A kinetic-optimized CoChR variant with enhanced high-frequency spiking fidelity. Biophys J 2022; 121:4166-4178. [PMID: 36151721 PMCID: PMC9675021 DOI: 10.1016/j.bpj.2022.09.024] [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: 01/19/2022] [Revised: 07/09/2022] [Accepted: 09/21/2022] [Indexed: 11/22/2022] Open
Abstract
Channelrhodopsins are a promising toolset for noninvasive optical manipulation of genetically identifiable neuron populations. Existing channelrhodopsins have generally suffered from a trade-off between two desired properties: fast channel kinetics and large photocurrent. Such a trade-off hinders spatiotemporally precise optogenetic activation during both one-photon and two-photon photostimulation. Furthermore, the simultaneous use of spectrally separated genetically encoded indicators and channelrhodopsins has generally suffered from non-negligible crosstalk in photocurrent or fluorescence. These limitations have hindered crosstalk-free dual-channel experiments needed to establish relationships between multiple neural populations. Recent large-scale transcriptome sequencing revealed one potent optogenetic actuator, the channelrhodopsin from species Chloromonas oogama (CoChR), which possessed high cyan light-driven photocurrent but slow channel kinetics. We rationally designed and engineered a kinetic-optimized CoChR variant that was faster than native CoChR while maintaining large photocurrent amplitude. When expressed in cultured hippocampal pyramidal neurons, our CoChR variant improved high-frequency spiking fidelity under one-photon illumination. Our CoChR variant's blue-shifted excitation spectrum enabled simultaneous cyan photostimulation and red calcium imaging with negligible photocurrent crosstalk.
Collapse
Affiliation(s)
- Xiaoke Bi
- Department of Biomedical Engineering, Duke University, Durham, North Carolina.
| | - Connor Beck
- Department of Biomedical Engineering, Duke University, Durham, North Carolina
| | - Yiyang Gong
- Department of Biomedical Engineering, Duke University, Durham, North Carolina.
| |
Collapse
|
50
|
Pekarek BT, Kochukov M, Lozzi B, Wu T, Hunt PJ, Tepe B, Hanson Moss E, Tantry EK, Swanson JL, Dooling SW, Patel M, Belfort BDW, Romero JM, Bao S, Hill MC, Arenkiel BR. Oxytocin signaling is necessary for synaptic maturation of adult-born neurons. Genes Dev 2022; 36:1100-1118. [PMID: 36617877 PMCID: PMC9851403 DOI: 10.1101/gad.349930.122] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Accepted: 11/14/2022] [Indexed: 12/13/2022]
Abstract
Neural circuit plasticity and sensory response dynamics depend on forming new synaptic connections. Despite recent advances toward understanding the consequences of circuit plasticity, the mechanisms driving circuit plasticity are unknown. Adult-born neurons within the olfactory bulb have proven to be a powerful model for studying circuit plasticity, providing a broad and accessible avenue into neuron development, migration, and circuit integration. We and others have shown that efficient adult-born neuron circuit integration hinges on presynaptic activity in the form of diverse signaling peptides. Here, we demonstrate a novel oxytocin-dependent mechanism of adult-born neuron synaptic maturation and circuit integration. We reveal spatial and temporal enrichment of oxytocin receptor expression within adult-born neurons in the murine olfactory bulb, with oxytocin receptor expression peaking during activity-dependent integration. Using viral labeling, confocal microscopy, and cell type-specific RNA-seq, we demonstrate that oxytocin receptor signaling promotes synaptic maturation of newly integrating adult-born neurons by regulating their morphological development and expression of mature synaptic AMPARs and other structural proteins.
Collapse
Affiliation(s)
- Brandon T Pekarek
- Genetics and Genomics Graduate Program, Baylor College of Medicine, Houston, Texas 77030, USA
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas 77030, USA
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, Texas 77030, USA
| | - Mikhail Kochukov
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, Texas 77030, USA
- Department of Anesthesiology, Baylor College of Medicine, Houston, Texas 77030, USA
| | - Brittney Lozzi
- Genetics and Genomics Graduate Program, Baylor College of Medicine, Houston, Texas 77030, USA
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas 77030, USA
| | - Timothy Wu
- Genetics and Genomics Graduate Program, Baylor College of Medicine, Houston, Texas 77030, USA
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas 77030, USA
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, Texas 77030, USA
- Medical Scientist Training Program, Baylor College of Medicine, Houston, Texas 77030, USA
| | - Patrick J Hunt
- Genetics and Genomics Graduate Program, Baylor College of Medicine, Houston, Texas 77030, USA
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas 77030, USA
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, Texas 77030, USA
- Medical Scientist Training Program, Baylor College of Medicine, Houston, Texas 77030, USA
| | - Burak Tepe
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas 77030, USA
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, Texas 77030, USA
| | - Elizabeth Hanson Moss
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas 77030, USA
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, Texas 77030, USA
| | - Evelyne K Tantry
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas 77030, USA
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, Texas 77030, USA
| | - Jessica L Swanson
- Genetics and Genomics Graduate Program, Baylor College of Medicine, Houston, Texas 77030, USA
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas 77030, USA
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, Texas 77030, USA
| | - Sean W Dooling
- Genetics and Genomics Graduate Program, Baylor College of Medicine, Houston, Texas 77030, USA
| | - Mayuri Patel
- Development, Disease Models, and Therapeutics Graduate Program, Baylor College of Medicine, Houston, Texas 77030, USA
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, Texas 77030, USA
| | - Benjamin D W Belfort
- Genetics and Genomics Graduate Program, Baylor College of Medicine, Houston, Texas 77030, USA
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas 77030, USA
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, Texas 77030, USA
- Medical Scientist Training Program, Baylor College of Medicine, Houston, Texas 77030, USA
| | - Juan M Romero
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, Texas 77030, USA
- Medical Scientist Training Program, Baylor College of Medicine, Houston, Texas 77030, USA
- Department of Neuroscience, Baylor College of Medicine, Houston, Texas 77030, USA
| | - Suyang Bao
- Development, Disease Models, and Therapeutics Graduate Program, Baylor College of Medicine, Houston, Texas 77030, USA
| | - Matthew C Hill
- Development, Disease Models, and Therapeutics Graduate Program, Baylor College of Medicine, Houston, Texas 77030, USA
| | - Benjamin R Arenkiel
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas 77030, USA
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, Texas 77030, USA
- Department of Neuroscience, Baylor College of Medicine, Houston, Texas 77030, USA
| |
Collapse
|