101
|
Xu C, Nedergaard M, Fowell DJ, Friedl P, Ji N. Multiphoton fluorescence microscopy for in vivo imaging. Cell 2024; 187:4458-4487. [PMID: 39178829 PMCID: PMC11373887 DOI: 10.1016/j.cell.2024.07.036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 07/18/2024] [Accepted: 07/22/2024] [Indexed: 08/26/2024]
Abstract
Multiphoton fluorescence microscopy (MPFM) has been a game-changer for optical imaging, particularly for studying biological tissues deep within living organisms. MPFM overcomes the strong scattering of light in heterogeneous tissue by utilizing nonlinear excitation that confines fluorescence emission mostly to the microscope focal volume. This enables high-resolution imaging deep within intact tissue and has opened new avenues for structural and functional studies. MPFM has found widespread applications and has led to numerous scientific discoveries and insights into complex biological processes. Today, MPFM is an indispensable tool in many research communities. Its versatility and effectiveness make it a go-to technique for researchers investigating biological phenomena at the cellular and subcellular levels in their native environments. In this Review, the principles, implementations, capabilities, and limitations of MPFM are presented. Three application areas of MPFM, neuroscience, cancer biology, and immunology, are reviewed in detail and serve as examples for applying MPFM to biological research.
Collapse
Affiliation(s)
- Chris Xu
- School of Applied and Engineering Physics, Cornell University, Ithaca, NY 14850, USA
| | - Maiken Nedergaard
- Center for Translational Neuromedicine, Faculty of Health and Medical Sciences, University of Copenhagen, Nørre Alle 3B, 2200 Copenhagen, Denmark; University of Rochester Medical School, 601 Elmwood Avenue, Rochester, NY 14642, USA
| | - Deborah J Fowell
- Department of Microbiology & Immunology, Cornell University, Ithaca, NY 14853, USA
| | - Peter Friedl
- Department of Medical BioSciences, Radboud University Medical Centre, Geert Grooteplein 26-28, Nijmegen HB 6500, the Netherlands
| | - Na Ji
- Department of Neuroscience, Department of Physics, University of California Berkeley, Berkeley, CA 94720, USA.
| |
Collapse
|
102
|
Hu Y, Wang Y, Zhang L, Luo M, Wang Y. Neural Network Mechanisms Underlying General Anesthesia: Cortical and Subcortical Nuclei. Neurosci Bull 2024:10.1007/s12264-024-01286-z. [PMID: 39168960 DOI: 10.1007/s12264-024-01286-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Accepted: 06/10/2024] [Indexed: 08/23/2024] Open
Abstract
General anesthesia plays a significant role in modern medicine. However, the precise mechanism of general anesthesia remains unclear, posing a key scientific challenge in anesthesiology. Advances in neuroscience techniques have enabled targeted manipulation of specific neural circuits and the capture of brain-wide neural activity at high resolution. These advances hold promise for elucidating the intricate mechanisms of action of general anesthetics. This review aims to summarize our current understanding of the role of cortical and subcortical nuclei in modulating general anesthesia, providing new evidence of cortico-cortical and thalamocortical networks in relation to anesthesia and consciousness. These insights contribute to a comprehensive understanding of the neural network mechanisms underlying general anesthesia.
Collapse
Affiliation(s)
- Yue Hu
- Department of Anesthesiology, Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Yun Wang
- Department of Anesthesiology, Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Lingjing Zhang
- Department of Anesthesiology, Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Mengqiang Luo
- Department of Anesthesiology, Huashan Hospital, Fudan University, Shanghai, 200040, China.
| | - Yingwei Wang
- Department of Anesthesiology, Huashan Hospital, Fudan University, Shanghai, 200040, China.
| |
Collapse
|
103
|
Park K, Yeo Y, Shin K, Kwag J. Egocentric neural representation of geometric vertex in the retrosplenial cortex. Nat Commun 2024; 15:7156. [PMID: 39169030 PMCID: PMC11339352 DOI: 10.1038/s41467-024-51391-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Accepted: 08/07/2024] [Indexed: 08/23/2024] Open
Abstract
Egocentric neural representations of environmental features, such as edges and vertices, are important for constructing a geometrically detailed egocentric cognitive map for goal-directed navigation and episodic memory. While egocentric neural representations of edges like egocentric boundary/border cells exist, those that selectively represent vertices egocentrically are yet unknown. Here we report that granular retrosplenial cortex (RSC) neurons in male mice generate spatial receptive fields exclusively near the vertices of environmental geometries during free exploration, termed vertex cells. Their spatial receptive fields occurred at a specific orientation and distance relative to the heading direction of mice, indicating egocentric vector coding of vertex. Removing physical boundaries defining the environmental geometry abolished the egocentric vector coding of vertex, and goal-directed navigation strengthened the egocentric vector coding at the goal-located vertex. Our findings suggest that egocentric vector coding of vertex by granular RSC neurons helps construct an egocentric cognitive map that guides goal-directed navigation.
Collapse
Affiliation(s)
- Kyerl Park
- Department of Brain and Cognitive Sciences, Seoul National University, Seoul, 08826, Republic of Korea
- Department of Brain and Cognitive Engineering, Korea University, Seoul, 02841, Republic of Korea
| | - Yoonsoo Yeo
- Department of Brain and Cognitive Sciences, Seoul National University, Seoul, 08826, Republic of Korea
- Department of Brain and Cognitive Engineering, Korea University, Seoul, 02841, Republic of Korea
| | - Kisung Shin
- Department of Brain and Cognitive Engineering, Korea University, Seoul, 02841, Republic of Korea
| | - Jeehyun Kwag
- Department of Brain and Cognitive Sciences, Seoul National University, Seoul, 08826, Republic of Korea.
| |
Collapse
|
104
|
Liu X, Chen Y, Hang C, Cheng J, Peng D, Li Y, Jiang X. Coupling Nanoscale Precision with Multiscale Imaging: A Multifunctional Near-Infrared Dye for the Brain. ACS NANO 2024; 18:22233-22244. [PMID: 39102625 DOI: 10.1021/acsnano.4c06103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/07/2024]
Abstract
Live imaging of primary neural cells is crucial for monitoring neuronal activity, especially multiscale and multifunctional imaging that offers excellent biocompatibility. Multiscale imaging can provide insights into cellular structure and function from the nanoscale to the millimeter scale. Multifunctional imaging can monitor different activities in the brain. However, this remains a challenge because of the lack of dyes with a high signal-to-background ratio, water solubility, and multiscale and multifunctional imaging capabilities. In this study, we present a neural dye with near-infrared (NIR) emissions (>700 nm) that enables ultrafast staining (in less than 1 min) for the imaging of primary neurons. This dye not only enables multiscale neural live-cell imaging from vesicles in neurites, neural membranes, and single neurons to the whole brain but also facilitates multifunctional imaging, such as the monitoring and quantifying of synaptic vesicles and the changes in membrane potential. We also explore the potential of this NIR neural dye for staining brain slices and live brains. The NIR neural dye exhibits superior binding with neural membranes compared to commercial dyes, thereby achieving multiscale and multifunctional brain neuroimaging. In conclusion, our findings introduce a significant breakthrough in neuroimaging dyes by developing a category of small molecular dyes.
Collapse
Affiliation(s)
- Xiaoyan Liu
- Shenzhen Key Laboratory of Smart Healthcare Engineering, Guangdong Provincial Key Laboratory of Advanced Biomaterials, Department of Biomedical Engineering, Southern University of Science and Technology, No. 1088 Xueyuan Road, Nanshan District, Shenzhen 518055, China
| | - Yao Chen
- Shenzhen Key Laboratory of Smart Healthcare Engineering, Guangdong Provincial Key Laboratory of Advanced Biomaterials, Department of Biomedical Engineering, Southern University of Science and Technology, No. 1088 Xueyuan Road, Nanshan District, Shenzhen 518055, China
| | - Chen Hang
- Shenzhen Key Laboratory of Smart Healthcare Engineering, Guangdong Provincial Key Laboratory of Advanced Biomaterials, Department of Biomedical Engineering, Southern University of Science and Technology, No. 1088 Xueyuan Road, Nanshan District, Shenzhen 518055, China
| | - Jinxiong Cheng
- Shenzhen Key Laboratory of Smart Healthcare Engineering, Guangdong Provincial Key Laboratory of Advanced Biomaterials, Department of Biomedical Engineering, Southern University of Science and Technology, No. 1088 Xueyuan Road, Nanshan District, Shenzhen 518055, China
| | - Dinglu Peng
- Shenzhen Key Laboratory of Smart Healthcare Engineering, Guangdong Provincial Key Laboratory of Advanced Biomaterials, Department of Biomedical Engineering, Southern University of Science and Technology, No. 1088 Xueyuan Road, Nanshan District, Shenzhen 518055, China
| | - Ying Li
- Innovation Research Center for AIE Pharmaceutical Biology, Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou 511436, China
| | - Xingyu Jiang
- Shenzhen Key Laboratory of Smart Healthcare Engineering, Guangdong Provincial Key Laboratory of Advanced Biomaterials, Department of Biomedical Engineering, Southern University of Science and Technology, No. 1088 Xueyuan Road, Nanshan District, Shenzhen 518055, China
| |
Collapse
|
105
|
Ebina T, Sasagawa A, Hong D, Setsuie R, Obara K, Masamizu Y, Kondo M, Terada SI, Ozawa K, Uemura M, Takaji M, Watakabe A, Kobayashi K, Ohki K, Yamamori T, Murayama M, Matsuzaki M. Dynamics of directional motor tuning in the primate premotor and primary motor cortices during sensorimotor learning. Nat Commun 2024; 15:7127. [PMID: 39164245 PMCID: PMC11336224 DOI: 10.1038/s41467-024-51425-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2023] [Accepted: 08/05/2024] [Indexed: 08/22/2024] Open
Abstract
Sensorimotor learning requires reorganization of neuronal activity in the premotor cortex (PM) and primary motor cortex (M1). To reveal PM- and M1-specific reorganization in a primate, we conducted calcium imaging in common marmosets while they learned a two-target reaching (pull/push) task after mastering a one-target reaching (pull) task. Throughout learning of the two-target reaching task, the dorsorostral PM (PMdr) showed peak activity earlier than the dorsocaudal PM (PMdc) and M1. During learning, the reaction time in pull trials increased and correlated strongly with the peak timing of PMdr activity. PMdr showed decreasing representation of newly introduced (push) movement, whereas PMdc and M1 maintained high representation of pull and push movements. Many task-related neurons in PMdc and M1 exhibited a strong preference to either movement direction. PMdc neurons dynamically switched their preferred direction depending on their performance in push trials in the early learning stage, whereas M1 neurons stably retained their preferred direction and high similarity of preferred direction between neighbors. These results suggest that in primate sensorimotor learning, dynamic directional motor tuning in PMdc converts the sensorimotor association formed in PMdr to the stable and specific motor representation of M1.
Collapse
Grants
- JP19dm0207069 Japan Agency for Medical Research and Development (AMED)
- JP19dm0107150 Japan Agency for Medical Research and Development (AMED)
- JP19dm0207085 Japan Agency for Medical Research and Development (AMED)
- JP19dm0207085 Japan Agency for Medical Research and Development (AMED)
- JP15dm0207001 Japan Agency for Medical Research and Development (AMED)
- JP15dm0207001 Japan Agency for Medical Research and Development (AMED)
- 22H05160 Ministry of Education, Culture, Sports, Science and Technology (MEXT)
- 23H00388 Ministry of Education, Culture, Sports, Science and Technology (MEXT)
- 21H00302 Ministry of Education, Culture, Sports, Science and Technology (MEXT)
- 23H04977 Ministry of Education, Culture, Sports, Science and Technology (MEXT)
- 20H03546 Ministry of Education, Culture, Sports, Science and Technology (MEXT)
- 17H04982 Ministry of Education, Culture, Sports, Science and Technology (MEXT)
Collapse
Affiliation(s)
- Teppei Ebina
- Department of Physiology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Akitaka Sasagawa
- Department of Physiology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Dokyeong Hong
- Department of Physiology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Rieko Setsuie
- Brain Functional Dynamics Collaboration Laboratory, RIKEN Center for Brain Science, Saitama, Japan
| | - Keitaro Obara
- Brain Functional Dynamics Collaboration Laboratory, RIKEN Center for Brain Science, Saitama, Japan
| | - Yoshito Masamizu
- Brain Functional Dynamics Collaboration Laboratory, RIKEN Center for Brain Science, Saitama, Japan
- Laboratory of Functional Brain Circuit Construction, Graduate School of Brain Science, Doshisha University, Kyoto, Japan
| | - Masashi Kondo
- Department of Physiology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Shin-Ichiro Terada
- Department of Physiology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Katsuya Ozawa
- Brain Functional Dynamics Collaboration Laboratory, RIKEN Center for Brain Science, Saitama, Japan
| | - Masato Uemura
- Department of Physiology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Masafumi Takaji
- Laboratory for Molecular Analysis of Higher Brain Function, RIKEN Center for Brain Science, Saitama, Japan
- Laboratory for Haptic Perception and Cognitive Physiology, RIKEN Center for Brain Science, Saitama, Japan
- Institute of Innovative Research, Tokyo Institute of Technology, Kanagawa, Japan
| | - Akiya Watakabe
- Laboratory for Molecular Analysis of Higher Brain Function, RIKEN Center for Brain Science, Saitama, Japan
- Laboratory for Molecular Mechanisms of Brain Development, RIKEN Center for Brain Science, Saitama, Japan
| | - Kenta Kobayashi
- Section of Viral Vector Development, National Institute for Physiological Sciences, Aichi, Japan
| | - Kenichi Ohki
- Department of Physiology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
- International Research Center for Neurointelligence (WPI-IRCN), The University of Tokyo Institutes for Advanced Study, Tokyo, Japan
- Institute for AI and Beyond, The University of Tokyo, Tokyo, Japan
| | - Tetsuo Yamamori
- Laboratory for Molecular Analysis of Higher Brain Function, RIKEN Center for Brain Science, Saitama, Japan
- Laboratory for Haptic Perception and Cognitive Physiology, RIKEN Center for Brain Science, Saitama, Japan
- Central Institute of Experimental Animals, Kanagawa, Japan
| | - Masanori Murayama
- Laboratory for Haptic Perception and Cognitive Physiology, RIKEN Center for Brain Science, Saitama, Japan
| | - Masanori Matsuzaki
- Department of Physiology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan.
- Brain Functional Dynamics Collaboration Laboratory, RIKEN Center for Brain Science, Saitama, Japan.
- International Research Center for Neurointelligence (WPI-IRCN), The University of Tokyo Institutes for Advanced Study, Tokyo, Japan.
- Department of Biological Sciences, Graduate School of Science, The University of Tokyo, Tokyo, Japan.
| |
Collapse
|
106
|
Koyanagi M, Ogido R, Moriya A, Saigo M, Ihida S, Teranishi T, Kawada J, Katsuno T, Matsubara K, Terada T, Yamashita A, Imai S. Development of a 3-dimensional organotypic model with characteristics of peripheral sensory nerves. CELL REPORTS METHODS 2024; 4:100835. [PMID: 39116883 PMCID: PMC11384078 DOI: 10.1016/j.crmeth.2024.100835] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Revised: 05/02/2024] [Accepted: 07/16/2024] [Indexed: 08/10/2024]
Abstract
We developed a rat dorsal root ganglion (DRG)-derived sensory nerve organotypic model by culturing DRG explants on an organoid culture device. With this method, a large number of organotypic cultures can be produced simultaneously with high reproducibility simply by seeding DRG explants derived from rat embryos. Unlike previous DRG explant models, this organotypic model consists of a ganglion and an axon bundle with myelinated A fibers, unmyelinated C fibers, and stereo-myelin-forming nodes of Ranvier. The model also exhibits Ca2+ signaling in cell bodies in response to application of chemical stimuli to nerve terminals. Further, axonal transection increases the activating transcription factor 3 mRNA level in ganglia. Axons and myelin are shown to regenerate 14 days following transection. Our sensory organotypic model enables analysis of neuronal excitability in response to pain stimuli and tracking of morphological changes in the axon bundle over weeks.
Collapse
Affiliation(s)
- Madoka Koyanagi
- Department of Medical Neuropharmacology, School of Pharmaceutical Sciences, Wakayama Medical University, Wakayama 640-8156, Japan
| | - Ryosuke Ogido
- Department of Clinical Pharmacology and Therapeutics, Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto 606-8507, Japan
| | - Akari Moriya
- Department of Clinical Pharmacology and Therapeutics, Faculty of Pharmaceutical Sciences, Kyoto University, Kyoto 606-8507, Japan
| | - Mamiko Saigo
- Department of Clinical Pharmacology and Therapeutics, Kyoto University Hospital, Kyoto 606-8507, Japan
| | - Satoshi Ihida
- New Business Promotion Division, Business Development Unit, Panel Semicon Laboratories, Sharp Corporation, Tenri, Nara 632-8567, Japan
| | - Tomoko Teranishi
- New Business Promotion Division, Business Development Unit, Panel Semicon Laboratories, Sharp Corporation, Tenri, Nara 632-8567, Japan
| | - Jiro Kawada
- Jiksak Bioengineering, Inc., Kawasaki, Kanagawa 210-0821, Japan
| | - Tatsuya Katsuno
- Division of Electron Microscopic Study, Center for Anatomical Studies, Graduate School of Medicine, Kyoto University, Kyoto 606-8501, Japan
| | - Kazuo Matsubara
- School of Pharmaceutical Sciences, Wakayama Medical University, Wakayama 640-8156, Japan
| | - Tomohiro Terada
- Department of Clinical Pharmacology and Therapeutics, Kyoto University Hospital, Kyoto 606-8507, Japan
| | - Akira Yamashita
- Department of Medical Neuropharmacology, School of Pharmaceutical Sciences, Wakayama Medical University, Wakayama 640-8156, Japan
| | - Satoshi Imai
- Department of Medical Neuropharmacology, School of Pharmaceutical Sciences, Wakayama Medical University, Wakayama 640-8156, Japan; Department of Clinical Pharmacology and Therapeutics, Kyoto University Hospital, Kyoto 606-8507, Japan.
| |
Collapse
|
107
|
Zhao P, Guo C, Xie M, Chen L, Golshani P, Aharoni D. MiniXL: An open-source, large field-of-view epifluorescence miniature microscope for mice capable of single-cell resolution and multi-brain region imaging. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.16.608328. [PMID: 39229051 PMCID: PMC11370419 DOI: 10.1101/2024.08.16.608328] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 09/05/2024]
Abstract
Capturing the intricate dynamics of neural activity in freely behaving animals is essential for understanding the neural mechanisms underpinning specific behaviors. Miniaturized microscopy enables investigators to track population activity at cellular level, but the field of view (FOV) of these microscopes have been limited and does not allow multiple-brain region imaging. To fill this technological gap, we have developed the eXtra Large field-of-view Miniscope (MiniXL), a 3.5g lightweight miniaturized microscope with an FOV measuring 3.5 mm in diameter and an electrically adjustable working distance of 1.9 mm ± 200 μm. We demonstrated the capability of MiniXL recording the activity of large neuronal population in both subcortical area (hippocampal dorsal CA1) and deep brain regions (medial prefrontal cortex, mPFC and nucleus accumbens, NAc). The large FOV allows simultaneous imaging of multiple brain regions such as bilateral mPFCs or mPFC and NAc during complex social behavior and tracking cells across multiple sessions. As with all microscopes in the UCLA Miniscope ecosystem, the MiniXL is fully open-source and will be shared with the neuroscience community to lower the barriers for adoption of this technology.
Collapse
Affiliation(s)
- Pingping Zhao
- Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Changliang Guo
- Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
- State Key Laboratory of Membrane Biology, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine, National Biomedical Imaging Center, School of Future Technology, Peking University, Beijing, China
| | - Mian Xie
- School of Life Sciences, Tsinghua University, Beijing, China
| | - Liangyi Chen
- State Key Laboratory of Membrane Biology, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine, National Biomedical Imaging Center, School of Future Technology, Peking University, Beijing, China
| | - Peyman Golshani
- Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
- Greater Los Angeles Veteran Affairs Medical Center, Los Angeles, CA, USA
- Intellectual and Developmental Disabilities Research Center, University of California, Los Angeles, Los Angeles, CA, USA
| | - Daniel Aharoni
- Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| |
Collapse
|
108
|
Yu SB, Wang H, Sanchez RG, Carlson NM, Nguyen K, Zhang A, Papich ZD, Abushawish AA, Whiddon Z, Matysik W, Zhang J, Whisenant TC, Ghassemian M, Koberstein JN, Stewart ML, Myers SA, Pekkurnaz G. Neuronal activity-driven O-GlcNAcylation promotes mitochondrial plasticity. Dev Cell 2024; 59:2143-2157.e9. [PMID: 38843836 PMCID: PMC11338717 DOI: 10.1016/j.devcel.2024.05.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Revised: 02/15/2024] [Accepted: 05/09/2024] [Indexed: 06/18/2024]
Abstract
Neuronal activity is an energy-intensive process that is largely sustained by instantaneous fuel utilization and ATP synthesis. However, how neurons couple ATP synthesis rate to fuel availability is largely unknown. Here, we demonstrate that the metabolic sensor enzyme O-linked N-acetyl glucosamine (O-GlcNAc) transferase regulates neuronal activity-driven mitochondrial bioenergetics in hippocampal and cortical neurons. We show that neuronal activity upregulates O-GlcNAcylation in mitochondria. Mitochondrial O-GlcNAcylation is promoted by activity-driven glucose consumption, which allows neurons to compensate for high energy expenditure based on fuel availability. To determine the proteins that are responsible for these adjustments, we mapped the mitochondrial O-GlcNAcome of neurons. Finally, we determine that neurons fail to meet activity-driven metabolic demand when O-GlcNAcylation dynamics are prevented. Our findings suggest that O-GlcNAcylation provides a fuel-dependent feedforward control mechanism in neurons to optimize mitochondrial performance based on neuronal activity. This mechanism thereby couples neuronal metabolism to mitochondrial bioenergetics and plays a key role in sustaining energy homeostasis.
Collapse
Affiliation(s)
- Seungyoon B Yu
- Neurobiology Department, School of Biological Sciences, University of California San Diego, La Jolla, CA 92093, USA
| | - Haoming Wang
- Neurobiology Department, School of Biological Sciences, University of California San Diego, La Jolla, CA 92093, USA
| | - Richard G Sanchez
- Neurobiology Department, School of Biological Sciences, University of California San Diego, La Jolla, CA 92093, USA
| | - Natasha M Carlson
- Neurobiology Department, School of Biological Sciences, University of California San Diego, La Jolla, CA 92093, USA
| | - Khanh Nguyen
- Laboratory for Immunochemical Circuits, Center of Autoimmunity and Inflammation, and Division of Signaling and Gene Expression, La Jolla Institute for Immunology, La Jolla, CA 92093, USA
| | - Andrew Zhang
- Neurobiology Department, School of Biological Sciences, University of California San Diego, La Jolla, CA 92093, USA
| | - Zachary D Papich
- Neurobiology Department, School of Biological Sciences, University of California San Diego, La Jolla, CA 92093, USA
| | - Ahmed A Abushawish
- Neurobiology Department, School of Biological Sciences, University of California San Diego, La Jolla, CA 92093, USA
| | - Zachary Whiddon
- Neurobiology Department, School of Biological Sciences, University of California San Diego, La Jolla, CA 92093, USA
| | - Weronika Matysik
- Neurobiology Department, School of Biological Sciences, University of California San Diego, La Jolla, CA 92093, USA
| | - Jie Zhang
- Neurobiology Department, School of Biological Sciences, University of California San Diego, La Jolla, CA 92093, USA
| | - Thomas C Whisenant
- Center for Computational Biology and Bioinformatics, University of California San Diego, La Jolla, CA 92093, USA
| | - Majid Ghassemian
- Biomolecular and Proteomics Mass Spectrometry Facility, University of California San Diego, La Jolla, CA 92093, USA
| | - John N Koberstein
- Vollum Institute, Oregon Health & Science University, Portland, OR 97239, USA
| | - Melissa L Stewart
- Vollum Institute, Oregon Health & Science University, Portland, OR 97239, USA
| | - Samuel A Myers
- Laboratory for Immunochemical Circuits, Center of Autoimmunity and Inflammation, and Division of Signaling and Gene Expression, La Jolla Institute for Immunology, La Jolla, CA 92093, USA; Department of Pharmacology, Program in Immunology, and Moores Cancer Center, University of California San Diego, La Jolla, CA 92093, USA
| | - Gulcin Pekkurnaz
- Neurobiology Department, School of Biological Sciences, University of California San Diego, La Jolla, CA 92093, USA.
| |
Collapse
|
109
|
Kveim VA, Salm L, Ulmer T, Lahr M, Kandler S, Imhof F, Donato F. Divergent recruitment of developmentally defined neuronal ensembles supports memory dynamics. Science 2024; 385:eadk0997. [PMID: 39146420 DOI: 10.1126/science.adk0997] [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: 11/03/2023] [Accepted: 06/24/2024] [Indexed: 08/17/2024]
Abstract
Memories are dynamic constructs whose properties change with time and experience. The biological mechanisms underpinning these dynamics remain elusive, particularly concerning how shifts in the composition of memory-encoding neuronal ensembles influence the evolution of a memory over time. By targeting developmentally distinct subpopulations of principal neurons, we discovered that memory encoding resulted in the concurrent establishment of multiple memory traces in the mouse hippocampus. Two of these traces were instantiated in subpopulations of early- and late-born neurons and followed distinct reactivation trajectories after encoding. The divergent recruitment of these subpopulations underpinned gradual reorganization of memory ensembles and modulated memory persistence and plasticity across multiple learning episodes. Thus, our findings reveal profound and intricate relationships between ensemble dynamics and the progression of memories over time.
Collapse
Affiliation(s)
- Vilde A Kveim
- Biozentrum, Universität Basel, 4056 Basel, Switzerland
| | - Laurenz Salm
- Biozentrum, Universität Basel, 4056 Basel, Switzerland
| | - Talia Ulmer
- Biozentrum, Universität Basel, 4056 Basel, Switzerland
| | - Maria Lahr
- Biozentrum, Universität Basel, 4056 Basel, Switzerland
| | | | - Fabia Imhof
- Biozentrum, Universität Basel, 4056 Basel, Switzerland
| | - Flavio Donato
- Biozentrum, Universität Basel, 4056 Basel, Switzerland
| |
Collapse
|
110
|
Zhang Y, Guan Z, Gadient JN, Kumar S, Gunaratne G, Walseth TF, Marchant JS, Wall KA, Slama JT. Diazirine-AIOC-NAADP, a Clickable-Photoactive NAADP Analog for Sea Urchin NAADP Binding Proteins. ACS Chem Biol 2024; 19:1842-1849. [PMID: 39092791 DOI: 10.1021/acschembio.4c00425] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/04/2024]
Abstract
Calcium ions (Ca2+) play a vital role as intracellular messengers, regulating essential cellular processes. Nicotinic acid adenine dinucleotide phosphate (NAADP) serves as a potent second messenger, responsible for releasing Ca2+ in both mammals and echinoderms. Despite identification of two human NAADP receptor proteins, their counterparts in sea urchins remain elusive. Sea urchin NAADP binding proteins are important due to their unique identities and NAADP binding properties which may illuminate new signaling modalities in other species. Consequently, the development of new photoactive and clickable NAADP analogs with specificity for binding targets in sea urchin egg homogenates is a priority. We designed and synthesized diazirine-AIOC-NAADP, a photoactive and "clickable" NAADP analog, to specifically label and identify sea urchin NAADP receptors. This analog, synthesized using a chemo-enzymatic approach, induced Ca2+ release from sea urchin egg homogenates at low-micromolar concentrations. The ability of diazirine-AIOC-NAADP to mobilize Ca2+ in cultured human cells was investigated by microinjection of the probe into U2OS cells. Microinjected NAADP elicited a robust Ca2+ release, but even 6000-fold higher concentrations of diazirine-AIOC-NAADP were unable to release Ca2+. Our results indicate that our new probe is specifically recognized at low concentration by sea urchin egg NAADP receptors but not by the NAADP receptors in a human cultured cell line.
Collapse
Affiliation(s)
- Yuqing Zhang
- Department of Medicinal and Biological Chemistry, College of Pharmacy and Pharmaceutical Sciences, University of Toledo, 3000 Arlington Avenue, Toledo, Ohio 43614, United States
| | - Zhong Guan
- Department of Medicinal and Biological Chemistry, College of Pharmacy and Pharmaceutical Sciences, University of Toledo, 3000 Arlington Avenue, Toledo, Ohio 43614, United States
| | - Jennifer N Gadient
- Instrumentation Center, College of Natural Sciences and Mathematics, University of Toledo, 2801 W. Bancroft Street, Toledo, Ohio. 43606, United States
| | - Sushil Kumar
- Department of Cell Biology, Neurobiology, and Anatomy, Medical College of Wisconsin, 8701 Watertown Plank Rd, Milwaukee, Wisconsin 53226, United States
| | - Gihan Gunaratne
- Department of Cell Biology, Neurobiology, and Anatomy, Medical College of Wisconsin, 8701 Watertown Plank Rd, Milwaukee, Wisconsin 53226, United States
| | - Timothy F Walseth
- Department of Pharmacology, University of Minnesota Medical School, 312 Church Street, Minneapolis, Minnesota 55455, United States
| | - Jonathan S Marchant
- Department of Cell Biology, Neurobiology, and Anatomy, Medical College of Wisconsin, 8701 Watertown Plank Rd, Milwaukee, Wisconsin 53226, United States
| | - Katherine A Wall
- Department of Medicinal and Biological Chemistry, College of Pharmacy and Pharmaceutical Sciences, University of Toledo, 3000 Arlington Avenue, Toledo, Ohio 43614, United States
| | - James T Slama
- Department of Medicinal and Biological Chemistry, College of Pharmacy and Pharmaceutical Sciences, University of Toledo, 3000 Arlington Avenue, Toledo, Ohio 43614, United States
| |
Collapse
|
111
|
Wong VC, Houlihan PR, Liu H, Walpita D, DeSantis MC, Liu Z, O'Shea EK. Plasticity-induced actin polymerization in the dendritic shaft regulates intracellular AMPA receptor trafficking. eLife 2024; 13:e80622. [PMID: 39146380 PMCID: PMC11326776 DOI: 10.7554/elife.80622] [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: 05/27/2022] [Accepted: 07/09/2024] [Indexed: 08/17/2024] Open
Abstract
AMPA-type receptors (AMPARs) are rapidly inserted into synapses undergoing plasticity to increase synaptic transmission, but it is not fully understood if and how AMPAR-containing vesicles are selectively trafficked to these synapses. Here, we developed a strategy to label AMPAR GluA1 subunits expressed from their endogenous loci in cultured rat hippocampal neurons and characterized the motion of GluA1-containing vesicles using single-particle tracking and mathematical modeling. We find that GluA1-containing vesicles are confined and concentrated near sites of stimulation-induced structural plasticity. We show that confinement is mediated by actin polymerization, which hinders the active transport of GluA1-containing vesicles along the length of the dendritic shaft by modulating the rheological properties of the cytoplasm. Actin polymerization also facilitates myosin-mediated transport of GluA1-containing vesicles to exocytic sites. We conclude that neurons utilize F-actin to increase vesicular GluA1 reservoirs and promote exocytosis proximal to the sites of synaptic activity.
Collapse
Affiliation(s)
- Victor C Wong
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, United States
| | - Patrick R Houlihan
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, United States
| | - Hui Liu
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, United States
| | - Deepika Walpita
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, United States
| | - Michael C DeSantis
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, United States
| | - Zhe Liu
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, United States
| | - Erin K O'Shea
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, United States
| |
Collapse
|
112
|
Li ZH, Asady B, Chang L, Triana MAH, Li C, Coppens I, Moreno SN. Calcium tunneling through the ER and transfer to other organelles for optimal signaling in Toxoplasma gondii. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.15.608087. [PMID: 39185237 PMCID: PMC11343207 DOI: 10.1101/2024.08.15.608087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/27/2024]
Abstract
Ca2+ signaling in cells begins with the opening of Ca2+ channels in either the plasma membrane (PM) or the endoplasmic reticulum (ER) and results in a dramatic increase in the physiologically low (<100 nM) cytosolic Ca2+ level. The temporal and spatial Ca2+ levels are well regulated to enable precise and specific activation of critical biological processes. Ca2+ signaling regulates pathogenic features of apicomplexan parasites like Toxoplasma gondii which infects approximately one-third of the world's population. T. gondii relies on Ca2+ signals to stimulate traits of its infection cycle and several Ca2+ signaling elements play essential roles in its parasitic cycle. Active egress, an essential step for the infection cycle of T. gondii is preceded by a large increase in cytosolic Ca2+ most likely by release from intracellular stores. Intracellular parasites take up Ca2+ from the host cell during host Ca2+ signaling events to replenish intracellular stores. In this work, we investigated the mechanism by which intracellular stores are replenished with Ca2+ and demonstrated a central role for the SERCA-Ca2+-ATPase to keep not only the ER filled with Ca2+ but also acidic stores. We also show mitochondrial Ca2+ uptake, by transfer of Ca2+ from the ER most likely through membrane contact sites. We propose a central role for the ER in tunneling of calcium from the extracellular milieu through the ER to other organelles.
Collapse
Affiliation(s)
- Zhu-Hong Li
- Center for Tropical and Emerging Global Diseases, Department of Computes Science, University of Georgia, Athens, Georgia 30602
| | - Beejan Asady
- Center for Tropical and Emerging Global Diseases, Department of Computes Science, University of Georgia, Athens, Georgia 30602
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Heath, Baltimore, MD 21205
| | - Le Chang
- Center for Tropical and Emerging Global Diseases, Department of Computes Science, University of Georgia, Athens, Georgia 30602
| | - Miryam Andrea Hortua Triana
- Center for Tropical and Emerging Global Diseases, Department of Computes Science, University of Georgia, Athens, Georgia 30602
| | - Catherine Li
- Center for Tropical and Emerging Global Diseases, Department of Computes Science, University of Georgia, Athens, Georgia 30602
| | - Isabelle Coppens
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Heath, Baltimore, MD 21205
| | - Silvia N.J. Moreno
- Center for Tropical and Emerging Global Diseases, Department of Computes Science, University of Georgia, Athens, Georgia 30602
- Department of Cellular Biology, University of Georgia, Athens, Georgia 30602
| |
Collapse
|
113
|
Fischl M, Pederson A, Voglewede R, Cheng H, Drew J, Torres Cadenas L, Weisz CJC. Fast Inhibition Slows and Desynchronizes Mouse Auditory Efferent Neuron Activity. J Neurosci 2024; 44:e0382242024. [PMID: 38937103 PMCID: PMC11326868 DOI: 10.1523/jneurosci.0382-24.2024] [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: 02/27/2024] [Revised: 06/04/2024] [Accepted: 06/11/2024] [Indexed: 06/29/2024] Open
Abstract
The encoding of acoustic stimuli requires precise neuron timing. Auditory neurons in the cochlear nucleus (CN) and brainstem are well suited for accurate analysis of fast acoustic signals, given their physiological specializations of fast membrane time constants, fast axonal conduction, and reliable synaptic transmission. The medial olivocochlear (MOC) neurons that provide efferent inhibition of the cochlea reside in the ventral brainstem and participate in these fast neural circuits. However, their modulation of cochlear function occurs over time scales of a slower nature. This suggests the presence of mechanisms that reduce MOC inhibition of cochlear function. To determine how monaural excitatory and inhibitory synaptic inputs integrate to affect the timing of MOC neuron activity, we developed a novel in vitro slice preparation ("wedge-slice"). The wedge-slice maintains the ascending auditory nerve root, the entire CN and projecting axons, while preserving the ability to perform visually guided patch-clamp electrophysiology recordings from genetically identified MOC neurons. The "in vivo-like" timing of the wedge-slice demonstrates that the inhibitory pathway accelerates relative to the excitatory pathway when the ascending circuit is intact, and the CN portion of the inhibitory circuit is precise enough to compensate for reduced precision in later synapses. When combined with machine learning PSC analysis and computational modeling, we demonstrate a larger suppression of MOC neuron activity when the inhibition occurs with in vivo-like timing. This delay of MOC activity may ensure that the MOC system is only engaged by sustained background sounds, preventing a maladaptive hypersuppression of cochlear activity.
Collapse
Affiliation(s)
- Matthew Fischl
- Section on Neuronal Circuitry, National Institute on Deafness and Other Communication Disorders, NIH, Bethesda, Maryland 20892
| | - Alia Pederson
- Section on Neuronal Circuitry, National Institute on Deafness and Other Communication Disorders, NIH, Bethesda, Maryland 20892
| | - Rebecca Voglewede
- Section on Neuronal Circuitry, National Institute on Deafness and Other Communication Disorders, NIH, Bethesda, Maryland 20892
| | - Hui Cheng
- NIDCD Data Science Core, National Institute on Deafness and Other Communication Disorders, NIH, Bethesda, Maryland 20892
| | - Jordan Drew
- Section on Neuronal Circuitry, National Institute on Deafness and Other Communication Disorders, NIH, Bethesda, Maryland 20892
| | - Lester Torres Cadenas
- Section on Neuronal Circuitry, National Institute on Deafness and Other Communication Disorders, NIH, Bethesda, Maryland 20892
| | - Catherine J C Weisz
- Section on Neuronal Circuitry, National Institute on Deafness and Other Communication Disorders, NIH, Bethesda, Maryland 20892
| |
Collapse
|
114
|
Oh JY, Lee H, Jang SY, Kim H, Park G, Serikov A, Jang JH, Kim J, Yang S, Sa M, Lee SE, Han YE, Hwang TY, Jung SJ, Kim HY, Lee SE, Oh SJ, Kim J, Kim J, Kim J, McHugh TJ, Lee CJ, Nam MH, Park HJ. Central Role of Hypothalamic Circuits for Acupuncture's Anti-Parkinsonian Effects. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024:e2403245. [PMID: 39119926 DOI: 10.1002/advs.202403245] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Revised: 06/30/2024] [Indexed: 08/10/2024]
Abstract
Despite clinical data stretching over millennia, the neurobiological basis of the effectiveness of acupuncture in treating diseases of the central nervous system has remained elusive. Here, using an established model of acupuncture treatment in Parkinson's disease (PD) model mice, we show that peripheral acupuncture stimulation activates hypothalamic melanin-concentrating hormone (MCH) neurons via nerve conduction. We further identify two separate neural pathways originating from anatomically and electrophysiologically distinct MCH neuronal subpopulations, projecting to the substantia nigra and hippocampus, respectively. Through chemogenetic manipulation specifically targeting these MCH projections, their respective roles in mediating the acupuncture-induced motor recovery and memory improvements following PD onset are demonstrated, as well as the underlying mechanisms mediating recovery from dopaminergic neurodegeneration, reactive gliosis, and impaired hippocampal synaptic plasticity. Collectively, these MCH neurons constitute not only a circuit-based explanation for the therapeutic effectiveness of traditional acupuncture, but also a potential cellular target for treating both motor and non-motor PD symptoms.
Collapse
Affiliation(s)
- Ju-Young Oh
- College of Korean Medicine, Kyung Hee University, Seoul, 02447, Republic of Korea
- Studies of Translational Acupuncture Research (STAR), Kyung Hee University, Seoul, 02447, Republic of Korea
| | - Hyowon Lee
- College of Korean Medicine, Kyung Hee University, Seoul, 02447, Republic of Korea
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
| | - Sun-Young Jang
- College of Korean Medicine, Kyung Hee University, Seoul, 02447, Republic of Korea
- Studies of Translational Acupuncture Research (STAR), Kyung Hee University, Seoul, 02447, Republic of Korea
| | - Hyunjin Kim
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
- Department of KHU-KIST Convergence Science and Technology, Kyung Hee University, Seoul, 02447, Republic of Korea
| | - Geunhong Park
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
| | - Almas Serikov
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
| | - Jae-Hwan Jang
- Studies of Translational Acupuncture Research (STAR), Kyung Hee University, Seoul, 02447, Republic of Korea
| | - Junyeop Kim
- Laboratory of Stem Cells & Cell Reprogramming, Department of Chemistry, Dongguk University, Seoul, 04629, Republic of Korea
| | - Seulkee Yang
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
| | - Moonsun Sa
- Center for Cognition and Sociality, Institute for Basic Science, Daejeon, 34126, Republic of Korea
| | - Sung Eun Lee
- Center for Cognition and Sociality, Institute for Basic Science, Daejeon, 34126, Republic of Korea
| | - Young-Eun Han
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
| | - Tae-Yeon Hwang
- College of Korean Medicine, Kyung Hee University, Seoul, 02447, Republic of Korea
- Studies of Translational Acupuncture Research (STAR), Kyung Hee University, Seoul, 02447, Republic of Korea
| | - Sharon Jiyoon Jung
- Technological Convergence Center, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
| | - Hee Young Kim
- Department of Physiology, Yonsei University, Seoul, 03722, Republic of Korea
| | - Seung Eun Lee
- Center for Cognition and Sociality, Institute for Basic Science, Daejeon, 34126, Republic of Korea
- Research Animal Resource Center, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
| | - Soo-Jin Oh
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
| | - Jeongjin Kim
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
| | - Jeongyeon Kim
- Emotion, Cognition & Behavior Research Group, Korea Brain Research Institute, Daegu, 41062, Republic of Korea
| | - Jongpil Kim
- Laboratory of Stem Cells & Cell Reprogramming, Department of Chemistry, Dongguk University, Seoul, 04629, Republic of Korea
| | - Thomas J McHugh
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
- Laboratory for Circuit and Behavioral Physiology, RIKEN, Wako-shi Saitama, 351-0198, Japan
| | - C Justin Lee
- Center for Cognition and Sociality, Institute for Basic Science, Daejeon, 34126, Republic of Korea
| | - Min-Ho Nam
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
- Department of KHU-KIST Convergence Science and Technology, Kyung Hee University, Seoul, 02447, Republic of Korea
| | - Hi-Joon Park
- College of Korean Medicine, Kyung Hee University, Seoul, 02447, Republic of Korea
- Studies of Translational Acupuncture Research (STAR), Kyung Hee University, Seoul, 02447, Republic of Korea
- Department of KHU-KIST Convergence Science and Technology, Kyung Hee University, Seoul, 02447, Republic of Korea
| |
Collapse
|
115
|
Bao Y, Gong Y. Accurate neuron segmentation method for one-photon calcium imaging videos combining convolutional neural networks and clustering. Commun Biol 2024; 7:970. [PMID: 39122882 PMCID: PMC11316101 DOI: 10.1038/s42003-024-06668-7] [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: 11/07/2023] [Accepted: 08/01/2024] [Indexed: 08/12/2024] Open
Abstract
One-photon fluorescent calcium imaging helps understand brain functions by recording large-scale neural activities in freely moving animals. Automatic, fast, and accurate active neuron segmentation algorithms are essential to extract and interpret information from these videos. One-photon imaging videos' low resolution, high noise, and high background fluctuation pose significant challenges. Here, we develop a software pipeline to address the challenges of processing one-photon calcium imaging videos. We extend our previous two-photon active neuron segmentation algorithm, Shallow U-Net Neuron Segmentation (SUNS), to better suppress background fluctuations in one-photon videos. We also develop additional neuron extraction (ANE) to locate small or dim neurons missed by SUNS. To train our segmentation method, we create ground truth neurons by developing a manual labeling pipeline assisted with semi-automatic refinement. Our method is more accurate and faster than state-of-the-art techniques when processing simulated videos and multiple experimental datasets acquired over various brain regions with different imaging conditions.
Collapse
Affiliation(s)
- Yijun Bao
- Department of Biomedical Engineering, Duke University, Durham, NC, 27708, USA.
- ZJU-Hangzhou Global Scientific and Technological Innovation Center, Zhejiang University, Hangzhou, 311215, China.
| | - Yiyang Gong
- Department of Biomedical Engineering, Duke University, Durham, NC, 27708, USA.
- Department of Neurobiology, Duke University, Durham, NC, 27708, USA.
- Department of Cell Biology, University of Oklahoma Health Science Center, Oklahoma City, OK, 73104, USA.
| |
Collapse
|
116
|
Shigetomi E, Suzuki H, Hirayama YJ, Sano F, Nagai Y, Yoshihara K, Koga K, Tateoka T, Yoshioka H, Shinozaki Y, Kinouchi H, Tanaka KF, Bito H, Tsuda M, Koizumi S. Disease-relevant upregulation of P2Y 1 receptor in astrocytes enhances neuronal excitability via IGFBP2. Nat Commun 2024; 15:6525. [PMID: 39117630 PMCID: PMC11310333 DOI: 10.1038/s41467-024-50190-7] [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: 08/18/2023] [Accepted: 06/26/2024] [Indexed: 08/10/2024] Open
Abstract
Reactive astrocytes play a pivotal role in the pathogenesis of neurological diseases; however, their functional phenotype and the downstream molecules by which they modify disease pathogenesis remain unclear. Here, we genetically increase P2Y1 receptor (P2Y1R) expression, which is upregulated in reactive astrocytes in several neurological diseases, in astrocytes of male mice to explore its function and the downstream molecule. This astrocyte-specific P2Y1R overexpression causes neuronal hyperexcitability by increasing both astrocytic and neuronal Ca2+ signals. We identify insulin-like growth factor-binding protein 2 (IGFBP2) as a downstream molecule of P2Y1R in astrocytes; IGFBP2 acts as an excitatory signal to cause neuronal excitation. In neurological disease models of epilepsy and stroke, reactive astrocytes upregulate P2Y1R and increase IGFBP2. The present findings identify a mechanism underlying astrocyte-driven neuronal hyperexcitability, which is likely to be shared by several neurological disorders, providing insights that might be relevant for intervention in diverse neurological disorders.
Collapse
Affiliation(s)
- Eiji Shigetomi
- Department of Neuropharmacology, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Yamanashi, 409-3898, Japan.
- Yamanashi GLIA center, University of Yamanashi, Yamanashi, 409-3898, Japan.
| | - Hideaki Suzuki
- Department of Neuropharmacology, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Yamanashi, 409-3898, Japan
- Yamanashi GLIA center, University of Yamanashi, Yamanashi, 409-3898, Japan
| | - Yukiho J Hirayama
- Department of Neuropharmacology, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Yamanashi, 409-3898, Japan
| | - Fumikazu Sano
- Department of Neuropharmacology, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Yamanashi, 409-3898, Japan
- Yamanashi GLIA center, University of Yamanashi, Yamanashi, 409-3898, Japan
- Department of Pediatrics, Faculty of Medicine, University of Yamanashi, Yamanashi, 409-3898, Japan
| | - Yuki Nagai
- Department of Neuropharmacology, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Yamanashi, 409-3898, Japan
- Yamanashi GLIA center, University of Yamanashi, Yamanashi, 409-3898, Japan
| | - Kohei Yoshihara
- Department of Molecular and System Pharmacology, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, 812-8582, Japan
| | - Keisuke Koga
- Department of Molecular and System Pharmacology, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, 812-8582, Japan
- Department of Neurophysiology, Hyogo College of Medicine, Hyogo, 663-8501, Japan
| | - Toru Tateoka
- Department of Neurosurgery, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Yamanashi, 409-3898, Japan
| | - Hideyuki Yoshioka
- Department of Neurosurgery, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Yamanashi, 409-3898, Japan
| | - Youichi Shinozaki
- Department of Neuropharmacology, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Yamanashi, 409-3898, Japan
- Yamanashi GLIA center, University of Yamanashi, Yamanashi, 409-3898, Japan
| | - Hiroyuki Kinouchi
- Department of Neurosurgery, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Yamanashi, 409-3898, Japan
| | - Kenji F Tanaka
- Division of Brain Sciences, Institute for Advanced Medical Research, Keio University School of Medicine, Tokyo, 160-8582, Japan
| | - Haruhiko Bito
- Department of Neurochemistry, Graduate School of Medicine, The University of Tokyo, Tokyo, 113-0033, Japan
| | - Makoto Tsuda
- Department of Molecular and System Pharmacology, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, 812-8582, Japan
- Department of Life Innovation, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, 812-8582, Japan
| | - Schuichi Koizumi
- Department of Neuropharmacology, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Yamanashi, 409-3898, Japan.
- Yamanashi GLIA center, University of Yamanashi, Yamanashi, 409-3898, Japan.
| |
Collapse
|
117
|
Rutter GA, Gresch A, Delgadillo Silva L, Benninger RKP. Exploring pancreatic beta-cell subgroups and their connectivity. Nat Metab 2024:10.1038/s42255-024-01097-6. [PMID: 39117960 DOI: 10.1038/s42255-024-01097-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Accepted: 07/05/2024] [Indexed: 08/10/2024]
Abstract
Functional pancreatic islet beta cells are essential to ensure glucose homeostasis across species from zebrafish to humans. These cells show significant heterogeneity, and emerging studies have revealed that connectivity across a hierarchical network is required for normal insulin release. Here, we discuss current thinking and areas of debate around intra-islet connectivity, cellular hierarchies and potential "controlling" beta-cell populations. We focus on methodologies, including comparisons of different cell preparations as well as in vitro and in vivo approaches to imaging and controlling the activity of human and rodent islet preparations. We also discuss the analytical approaches that can be applied to live-cell data to identify and study critical subgroups of cells with a disproportionate role in control Ca2+ dynamics and thus insulin secretion (such as "first responders", "leaders" and "hubs", as defined by Ca2+ responses to glucose stimulation). Possible mechanisms by which this hierarchy is achieved, its physiological relevance and how its loss may contribute to islet failure in diabetes mellitus are also considered. A glossary of terms and links to computational resources are provided.
Collapse
Affiliation(s)
- Guy A Rutter
- CHUM Research Center and Faculty of Medicine, University of Montréal, Montréal, QC, Canada.
- Department of Metabolism, Digestion and Reproduction, Faculty of Medicine, Imperial College London, London, UK.
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore.
| | - Anne Gresch
- Department of Bioengineering, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Luis Delgadillo Silva
- CHUM Research Center and Faculty of Medicine, University of Montréal, Montréal, QC, Canada
| | - Richard K P Benninger
- Department of Bioengineering, University of Colorado Anschutz Medical Campus, Aurora, CO, USA.
| |
Collapse
|
118
|
Patel R, Bryant AS, Castelletto ML, Walsh B, Akimori D, Hallem EA. The generation of stable transgenic lines in the human-infective nematode Strongyloides stercoralis. G3 (BETHESDA, MD.) 2024; 14:jkae122. [PMID: 38839055 PMCID: PMC11304987 DOI: 10.1093/g3journal/jkae122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Revised: 05/13/2024] [Accepted: 05/23/2024] [Indexed: 06/07/2024]
Abstract
The skin-penetrating gastrointestinal parasitic nematode Strongyloides stercoralis causes strongyloidiasis, which is a neglected tropical disease that is associated with severe chronic illness and fatalities. Unlike other human-infective nematodes, S. stercoralis cycles through a single free-living generation and thus serves as a genetically tractable model organism for understanding the mechanisms that enable parasitism. Techniques such as CRISPR/Cas9-mediated mutagenesis and transgenesis are now routinely performed in S. stercoralis by introducing exogenous DNA into free-living adults and then screening their F1 progeny for transgenic or mutant larvae. However, transgenesis in S. stercoralis has been severely hindered by the inability to establish stable transgenic lines that can be propagated for multiple generations through a host; to date, studies of transgenic S. stercoralis have been limited to heterogeneous populations of transgenic F1 larvae. Here, we develop an efficient pipeline for the generation of stable transgenic lines in S. stercoralis. We also show that this approach can be used to efficiently generate stable transgenic lines in the rat-infective nematode Strongyloides ratti. The ability to generate stable transgenic lines circumvents the limitations of working with heterogeneous F1 populations, such as variable transgene expression and the inability to generate transgenics of all life stages. Our transgenesis approach will enable novel lines of inquiry into parasite biology, such as transgene-based comparisons between free-living and parasitic generations.
Collapse
Affiliation(s)
- Ruhi Patel
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Astra S Bryant
- Department of Physiology and Biophysics, University of Washington, Seattle, WA 98195, USA
| | - Michelle L Castelletto
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Breanna Walsh
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Molecular Biology Interdepartmental PhD Program, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Medical Scientist Training Program, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Damia Akimori
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Molecular Biology Interdepartmental PhD Program, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Elissa A Hallem
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA
| |
Collapse
|
119
|
Ba W, Nollet M, Yin C, Yu X, Wong S, Miao A, Beckwith EJ, Harding EC, Ma Y, Yustos R, Vyssotski AL, Wisden W, Franks NP. A REM-active basal ganglia circuit that regulates anxiety. Curr Biol 2024; 34:3301-3314.e4. [PMID: 38944034 DOI: 10.1016/j.cub.2024.06.010] [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: 06/12/2023] [Revised: 04/22/2024] [Accepted: 06/05/2024] [Indexed: 07/01/2024]
Abstract
Rapid eye movement (REM) sleep has been hypothesized to promote emotional resilience, but any neuronal circuits mediating this have not been identified. We find that in mice, somatostatin (Som) neurons in the entopeduncular nucleus (EPSom)/internal globus pallidus are predominantly active during REM sleep. This unique REM activity is both necessary and sufficient for maintaining normal REM sleep. Inhibiting or exciting EPSom neurons reduced or increased REM sleep duration, respectively. Activation of the sole downstream target of EPSom neurons, Vglut2 cells in the lateral habenula (LHb), increased sleep via the ventral tegmental area (VTA). A simple chemogenetic scheme to periodically inhibit the LHb over 4 days selectively removed a significant amount of cumulative REM sleep. Chronic, but not acute, REM reduction correlated with mice becoming anxious and more sensitive to aversive stimuli. Therefore, we suggest that cumulative REM sleep, in part generated by the EP → LHb → VTA circuit identified here, could contribute to stabilizing reactions to habitual aversive stimuli.
Collapse
Affiliation(s)
- Wei Ba
- Department of Life Sciences, Imperial College London, London SW7 2AZ, UK
| | - Mathieu Nollet
- Department of Life Sciences, Imperial College London, London SW7 2AZ, UK; UK Dementia Research Institute, Imperial College London, London SW7 2AZ, UK
| | - Chunyu Yin
- Department of Life Sciences, Imperial College London, London SW7 2AZ, UK; Department of Neonatal Medical Center, Children's Hospital of Nanjing Medical University, Nanjing 210000, China
| | - Xiao Yu
- Department of Life Sciences, Imperial College London, London SW7 2AZ, UK
| | - Sara Wong
- Department of Life Sciences, Imperial College London, London SW7 2AZ, UK; UK Dementia Research Institute, Imperial College London, London SW7 2AZ, UK
| | - Andawei Miao
- Department of Life Sciences, Imperial College London, London SW7 2AZ, UK; UK Dementia Research Institute, Imperial College London, London SW7 2AZ, UK
| | - Esteban J Beckwith
- Department of Life Sciences, Imperial College London, London SW7 2AZ, UK
| | - Edward C Harding
- Department of Life Sciences, Imperial College London, London SW7 2AZ, UK
| | - Ying Ma
- Department of Life Sciences, Imperial College London, London SW7 2AZ, UK
| | - Raquel Yustos
- Department of Life Sciences, Imperial College London, London SW7 2AZ, UK
| | - Alexei L Vyssotski
- Institute of Neuroinformatics, University of Zurich and ETH Zurich, Zurich 8057, Switzerland
| | - William Wisden
- Department of Life Sciences, Imperial College London, London SW7 2AZ, UK; UK Dementia Research Institute, Imperial College London, London SW7 2AZ, UK.
| | - Nicholas P Franks
- Department of Life Sciences, Imperial College London, London SW7 2AZ, UK; UK Dementia Research Institute, Imperial College London, London SW7 2AZ, UK.
| |
Collapse
|
120
|
Liao Z, Losonczy A. Learning, Fast and Slow: Single- and Many-Shot Learning in the Hippocampus. Annu Rev Neurosci 2024; 47:187-209. [PMID: 38663090 PMCID: PMC11519319 DOI: 10.1146/annurev-neuro-102423-100258] [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] [Indexed: 08/09/2024]
Abstract
The hippocampus is critical for memory and spatial navigation. The ability to map novel environments, as well as more abstract conceptual relationships, is fundamental to the cognitive flexibility that humans and other animals require to survive in a dynamic world. In this review, we survey recent advances in our understanding of how this flexibility is implemented anatomically and functionally by hippocampal circuitry, during both active exploration (online) and rest (offline). We discuss the advantages and limitations of spike timing-dependent plasticity and the more recently discovered behavioral timescale synaptic plasticity in supporting distinct learning modes in the hippocampus. Finally, we suggest complementary roles for these plasticity types in explaining many-shot and single-shot learning in the hippocampus and discuss how these rules could work together to support the learning of cognitive maps.
Collapse
Affiliation(s)
- Zhenrui Liao
- Department of Neuroscience and Mortimer B. Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY, USA;
| | - Attila Losonczy
- Department of Neuroscience and Mortimer B. Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY, USA;
| |
Collapse
|
121
|
Yu L, Russ AN, Algamal M, Abedin MJ, Zhao Q, Miller MR, Perle SJ, Kastanenka KV. Slow wave activity disruptions and memory impairments in a mouse model of aging. Neurobiol Aging 2024; 140:12-21. [PMID: 38701647 PMCID: PMC11188680 DOI: 10.1016/j.neurobiolaging.2024.04.006] [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: 12/12/2023] [Revised: 03/29/2024] [Accepted: 04/17/2024] [Indexed: 05/05/2024]
Abstract
The aging population suffers from memory impairments. Slow-wave activity (SWA) is composed of slow (0.5-1 Hz) and delta (1-4 Hz) oscillations, which play important roles in long-term memory and working memory function respectively. SWA disruptions might lead to memory disturbances often experienced by older adults. We conducted behavioral tests in young and older C57BL/6 J mice. SWA was monitored using wide-field imaging with voltage sensors. Cell-specific calcium imaging was used to monitor the activity of excitatory and inhibitory neurons in these mice. Older mice exhibited impairments in working memory but not memory consolidation. Voltage-sensor imaging revealed aberrant synchronization of neuronal activity in older mice. Notably, we found older mice exhibited no significant alterations in slow oscillations, whereas there was a significant increase in delta power compared to young mice. Calcium imaging revealed hypoactivity in inhibitory neurons of older mice. Combined, these results suggest that neural activity disruptions might correlate with aberrant memory performance in older mice.
Collapse
Affiliation(s)
- Lu Yu
- Department of Neurology, MassGeneral Institute of Neurodegenerative Diseases, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA 02129, USA
| | - Alyssa N Russ
- Department of Neurology, MassGeneral Institute of Neurodegenerative Diseases, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA 02129, USA
| | - Moustafa Algamal
- Department of Neurology, MassGeneral Institute of Neurodegenerative Diseases, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA 02129, USA
| | - Md Joynal Abedin
- Department of Neurology, MassGeneral Institute of Neurodegenerative Diseases, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA 02129, USA
| | - Qiuchen Zhao
- Department of Neurology, MassGeneral Institute of Neurodegenerative Diseases, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA 02129, USA
| | - Morgan R Miller
- Department of Neurology, MassGeneral Institute of Neurodegenerative Diseases, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA 02129, USA
| | - Stephen J Perle
- Department of Neurology, MassGeneral Institute of Neurodegenerative Diseases, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA 02129, USA
| | - Ksenia V Kastanenka
- Department of Neurology, MassGeneral Institute of Neurodegenerative Diseases, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA 02129, USA.
| |
Collapse
|
122
|
Lou Y, Ma J, Hu Y, Yao X, Liu Y, Wu M, Jia G, Chen Y, Chai R, Xia M, Li W. Integration of Functional Human Auditory Neural Circuits Based on a 3D Carbon Nanotube System. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2309617. [PMID: 38889308 PMCID: PMC11348147 DOI: 10.1002/advs.202309617] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/09/2023] [Revised: 04/27/2024] [Indexed: 06/20/2024]
Abstract
The physiological interactions between the peripheral and central auditory systems are crucial for auditory information transmission and perception, while reliable models for auditory neural circuits are currently lacking. To address this issue, mouse and human neural pathways are generated by utilizing a carbon nanotube nanofiber system. The super-aligned pattern of the scaffold renders the axons of the bipolar and multipolar neurons extending in a parallel direction. In addition, the electrical conductivity of the scaffold maintains the electrophysiological activity of the primary mouse auditory neurons. The mouse and human primary neurons from peripheral and central auditory units in the system are then co-cultured and showed that the two kinds of neurons form synaptic connections. Moreover, neural progenitor cells of the cochlea and auditory cortex are derived from human embryos to generate region-specific organoids and these organoids are assembled in the nanofiber-combined 3D system. Using optogenetic stimulation, calcium imaging, and electrophysiological recording, it is revealed that functional synaptic connections are formed between peripheral neurons and central neurons, as evidenced by calcium spiking and postsynaptic currents. The auditory circuit model will enable the study of the auditory neural pathway and advance the search for treatment strategies for disorders of neuronal connectivity in sensorineural hearing loss.
Collapse
Affiliation(s)
- Yiyun Lou
- ENT Institute and Otorhinolaryngology Department of Eye & ENT HospitalState Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain ScienceFudan UniversityShanghai200031China
- Institutes of Biomedical SciencesFudan UniversityShanghai200032China
| | - Jiaoyao Ma
- ENT Institute and Otorhinolaryngology Department of Eye & ENT HospitalState Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain ScienceFudan UniversityShanghai200031China
- Institutes of Biomedical SciencesFudan UniversityShanghai200032China
| | - Yangnan Hu
- State Key Laboratory of Digital Medical EngineeringDepartment of Otolaryngology Head and Neck SurgeryZhongda HospitalSchool of Life Sciences and TechnologyAdvanced Institute for Life and HealthJiangsu Province High‐Tech Key Laboratory for Bio‐Medical ResearchSoutheast UniversityNanjing210096China
- Co‐Innovation Center of NeuroregenerationNantong UniversityNantong226001China
| | - Xiaoying Yao
- Obstetrics and Gynecology HospitalFudan UniversityShanghai200011China
| | - Yaoqian Liu
- ENT Institute and Otorhinolaryngology Department of Eye & ENT HospitalState Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain ScienceFudan UniversityShanghai200031China
- Institutes of Biomedical SciencesFudan UniversityShanghai200032China
| | - Mingxuan Wu
- ENT Institute and Otorhinolaryngology Department of Eye & ENT HospitalState Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain ScienceFudan UniversityShanghai200031China
- Institutes of Biomedical SciencesFudan UniversityShanghai200032China
| | - Gaogan Jia
- ENT Institute and Otorhinolaryngology Department of Eye & ENT HospitalState Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain ScienceFudan UniversityShanghai200031China
- Institutes of Biomedical SciencesFudan UniversityShanghai200032China
| | - Yan Chen
- ENT Institute and Otorhinolaryngology Department of Eye & ENT HospitalState Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain ScienceFudan UniversityShanghai200031China
- Institutes of Biomedical SciencesFudan UniversityShanghai200032China
- NHC Key Laboratory of Hearing MedicineFudan UniversityShanghai200031China
- The Institutes of Brain Science and the Collaborative Innovation Center for Brain ScienceFudan UniversityShanghai200032China
| | - Renjie Chai
- State Key Laboratory of Digital Medical EngineeringDepartment of Otolaryngology Head and Neck SurgeryZhongda HospitalSchool of Life Sciences and TechnologyAdvanced Institute for Life and HealthJiangsu Province High‐Tech Key Laboratory for Bio‐Medical ResearchSoutheast UniversityNanjing210096China
- Co‐Innovation Center of NeuroregenerationNantong UniversityNantong226001China
| | - Mingyu Xia
- ENT Institute and Otorhinolaryngology Department of Eye & ENT HospitalState Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain ScienceFudan UniversityShanghai200031China
- Institutes of Biomedical SciencesFudan UniversityShanghai200032China
- NHC Key Laboratory of Hearing MedicineFudan UniversityShanghai200031China
- The Institutes of Brain Science and the Collaborative Innovation Center for Brain ScienceFudan UniversityShanghai200032China
| | - Wenyan Li
- ENT Institute and Otorhinolaryngology Department of Eye & ENT HospitalState Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain ScienceFudan UniversityShanghai200031China
- Institutes of Biomedical SciencesFudan UniversityShanghai200032China
- NHC Key Laboratory of Hearing MedicineFudan UniversityShanghai200031China
- The Institutes of Brain Science and the Collaborative Innovation Center for Brain ScienceFudan UniversityShanghai200032China
| |
Collapse
|
123
|
Sihn D, Chae S, Kim SP. A method to find temporal structure of neuronal coactivity patterns with across-trial correlations. J Neurosci Methods 2024; 408:110172. [PMID: 38782124 DOI: 10.1016/j.jneumeth.2024.110172] [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: 02/05/2024] [Revised: 05/08/2024] [Accepted: 05/17/2024] [Indexed: 05/25/2024]
Abstract
BACKGROUND The across-trial correlation of neurons' coactivity patterns emerges to be important for information coding, but methods for finding their temporal structures remain largely unexplored. NEW METHOD In the present study, we propose a method to find time clusters in which coactivity patterns of neurons are correlated across trials. We transform the multidimensional neural activity at each timing into a coactivity pattern of binary states, and predict the coactivity patterns at different timings. We devise a method suitable for these coactivity pattern predictions, call general event prediction. Cross-temporal prediction accuracy is then used to estimate across-trial correlations between coactivity patterns at two timings. We extract time clusters from the cross-temporal prediction accuracy by a modified k-means algorithm. RESULTS The feasibility of the proposed method is verified through simulations based on ground truth. We apply the proposed method to a calcium imaging dataset recorded from the motor cortex of mice, and demonstrate time clusters of motor cortical coactivity patterns during a motor task. COMPARISON WITH EXISTING METHODS While the existing cosine similarity method, which does not account for across-trial correlation, shows temporal structures only for contralateral neural responses, the proposed method reveals those for both contralateral and ipsilateral neural responses, demonstrating the effect of across-trial correlations. CONCLUSIONS This study introduces a novel method for measuring the temporal structure of neuronal ensemble activity.
Collapse
Affiliation(s)
- Duho Sihn
- Department of Biomedical Engineering, Ulsan National Institute of Science and Technology, Ulsan 44919, the Republic of Korea
| | - Soyoung Chae
- Department of Biomedical Engineering, Ulsan National Institute of Science and Technology, Ulsan 44919, the Republic of Korea
| | - Sung-Phil Kim
- Department of Biomedical Engineering, Ulsan National Institute of Science and Technology, Ulsan 44919, the Republic of Korea.
| |
Collapse
|
124
|
Xiang Z, He S, Chen R, Liu S, Liu M, Xu L, Zheng J, Jiang Z, Ma L, Sun Y, Qin Y, Chen Y, Li W, Wang X, Chen G, Lei W. Two-photon live imaging of direct glia-to-neuron conversion in the mouse cortex. Neural Regen Res 2024; 19:1781-1788. [PMID: 38103245 PMCID: PMC10960291 DOI: 10.4103/1673-5374.386401] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Revised: 08/23/2023] [Accepted: 09/26/2023] [Indexed: 12/18/2023] Open
Abstract
JOURNAL/nrgr/04.03/01300535-202408000-00032/figure1/v/2023-12-16T180322Z/r/image-tiff Over the past decade, a growing number of studies have reported transcription factor-based in situ reprogramming that can directly convert endogenous glial cells into functional neurons as an alternative approach for neuroregeneration in the adult mammalian central nervous system. However, many questions remain regarding how a terminally differentiated glial cell can transform into a delicate neuron that forms part of the intricate brain circuitry. In addition, concerns have recently been raised around the absence of astrocyte-to-neuron conversion in astrocytic lineage-tracing mice. In this study, we employed repetitive two-photon imaging to continuously capture the in situ astrocyte-to-neuron conversion process following ectopic expression of the neural transcription factor NeuroD1 in both proliferating reactive astrocytes and lineage-traced astrocytes in the mouse cortex. Time-lapse imaging over several weeks revealed the step-by-step transition from a typical astrocyte with numerous short, tapered branches to a typical neuron with a few long neurites and dynamic growth cones that actively explored the local environment. In addition, these lineage-converting cells were able to migrate radially or tangentially to relocate to suitable positions. Furthermore, two-photon Ca2+ imaging and patch-clamp recordings confirmed that the newly generated neurons exhibited synchronous calcium signals, repetitive action potentials, and spontaneous synaptic responses, suggesting that they had made functional synaptic connections within local neural circuits. In conclusion, we directly visualized the step-by-step lineage conversion process from astrocytes to functional neurons in vivo and unambiguously demonstrated that adult mammalian brains are highly plastic with respect to their potential for neuroregeneration and neural circuit reconstruction.
Collapse
Affiliation(s)
- Zongqin Xiang
- Guangdong-Hong Kong-Macau Institute of CNS Regeneration (GHMICR), Jinan University, Guangzhou, Guangdong Province, China
- Department of Neurosurgery, The First Affiliated Hospital, Jinan University, Guangzhou, Guangdong Province, China
- Laboratory for Neuroimmunology in Health and Diseases, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong Province, China
| | - Shu He
- Guangdong-Hong Kong-Macau Institute of CNS Regeneration (GHMICR), Jinan University, Guangzhou, Guangdong Province, China
| | - Rongjie Chen
- Guangdong-Hong Kong-Macau Institute of CNS Regeneration (GHMICR), Jinan University, Guangzhou, Guangdong Province, China
| | - Shanggong Liu
- Guangdong-Hong Kong-Macau Institute of CNS Regeneration (GHMICR), Jinan University, Guangzhou, Guangdong Province, China
| | - Minhui Liu
- VIB-KU Leuven Center for Brain & Disease Research, Leuven, Flemish Region, Belgium
| | - Liang Xu
- Guangdong-Hong Kong-Macau Institute of CNS Regeneration (GHMICR), Jinan University, Guangzhou, Guangdong Province, China
| | - Jiajun Zheng
- Department of Neurosurgery, The First Affiliated Hospital, Jinan University, Guangzhou, Guangdong Province, China
| | - Zhouquan Jiang
- Guangdong-Hong Kong-Macau Institute of CNS Regeneration (GHMICR), Jinan University, Guangzhou, Guangdong Province, China
| | - Long Ma
- Guangdong-Hong Kong-Macau Institute of CNS Regeneration (GHMICR), Jinan University, Guangzhou, Guangdong Province, China
| | - Ying Sun
- Guangdong-Hong Kong-Macau Institute of CNS Regeneration (GHMICR), Jinan University, Guangzhou, Guangdong Province, China
| | - Yongpeng Qin
- Guangdong-Hong Kong-Macau Institute of CNS Regeneration (GHMICR), Jinan University, Guangzhou, Guangdong Province, China
| | - Yi Chen
- Guangdong-Hong Kong-Macau Institute of CNS Regeneration (GHMICR), Jinan University, Guangzhou, Guangdong Province, China
| | - Wen Li
- Guangdong-Hong Kong-Macau Institute of CNS Regeneration (GHMICR), Jinan University, Guangzhou, Guangdong Province, China
| | - Xiangyu Wang
- Department of Neurosurgery, The First Affiliated Hospital, Jinan University, Guangzhou, Guangdong Province, China
| | - Gong Chen
- Guangdong-Hong Kong-Macau Institute of CNS Regeneration (GHMICR), Jinan University, Guangzhou, Guangdong Province, China
| | - Wenliang Lei
- Guangdong-Hong Kong-Macau Institute of CNS Regeneration (GHMICR), Jinan University, Guangzhou, Guangdong Province, China
| |
Collapse
|
125
|
Gautham AK, Miner LE, Franco MN, Thornquist SC, Crickmore MA. Dopamine biases decisions by limiting temporal integration. Nature 2024; 632:850-857. [PMID: 39085606 DOI: 10.1038/s41586-024-07749-7] [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: 04/21/2023] [Accepted: 06/24/2024] [Indexed: 08/02/2024]
Abstract
Motivations bias our responses to stimuli, producing behavioural outcomes that match our needs and goals. Here we describe a mechanism behind this phenomenon: adjusting the time over which stimulus-derived information is permitted to accumulate towards a decision. As a Drosophila copulation progresses, the male becomes less likely to continue mating through challenges1-3. We show that a set of copulation decision neurons (CDNs) flexibly integrates information about competing drives to mediate this decision. Early in mating, dopamine signalling restricts CDN integration time by potentiating Ca2+/calmodulin-dependent protein kinase II (CaMKII) activation in response to stimulatory inputs, imposing a high threshold for changing behaviours. Later into mating, the timescale over which the CDNs integrate termination-promoting information expands, increasing the likelihood of switching behaviours. We suggest scalable windows of temporal integration at dedicated circuit nodes as a key but underappreciated variable in state-based decision-making.
Collapse
Affiliation(s)
- Aditya K Gautham
- F. M. Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Lauren E Miner
- F. M. Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Marco N Franco
- F. M. Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Stephen C Thornquist
- F. M. Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA.
- Laboratory of Integrative Brain Function, The Rockefeller University, New York, NY, USA.
| | - Michael A Crickmore
- F. M. Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
126
|
Sheng CQ, Wu SS, Cheng YK, Wu Y, Li YM. Comprehensive review of indicators and techniques for optical mapping of intracellular calcium ions. Cereb Cortex 2024; 34:bhae346. [PMID: 39191664 DOI: 10.1093/cercor/bhae346] [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: 05/01/2024] [Revised: 07/27/2024] [Accepted: 08/09/2024] [Indexed: 08/29/2024] Open
Abstract
Calcium ions (Ca2+) play crucial roles in almost every cellular process, making the detection of changes in intracellular Ca2+ essential to understanding cell function. The fluorescence indicator method has garnered widespread application due to its exceptional sensitivity, rapid analysis, cost-effectiveness, and user-friendly nature. It has successfully delineated the spatial and temporal dynamics of Ca2+ signaling across diverse cell types. However, it is vital to understand that different indicators have varying levels of accuracy, sensitivity, and stability, making choosing the right inspection method crucial. As optical detection technologies advance, they continually broaden the horizons of scientific inquiry. This primer offers a systematic synthesis of the current fluorescence indicators and optical imaging modalities utilized for the detection of intracellular Ca2+. It elucidates their practical applications and inherent limitations, serving as an essential reference for researchers seeking to identify the most suitable detection methodologies for their calcium-centric investigations.
Collapse
Affiliation(s)
- Chu-Qiao Sheng
- Department of Pediatric Intensive Care Unit, Children's Medical Center, The First Hospital of Jilin University, Changchun, Jilin 130021, China
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, No. 2699, Qianjin Street, Changchun, Jilin 130012, China
| | - Shuang-Shuang Wu
- Department of Pediatric Hematology, Children's Medical Center, The First Hospital of Jilin University, Changchun, Jilin 130021, China
| | - Yong-Kang Cheng
- Department of Pediatric Intensive Care Unit, Children's Medical Center, The First Hospital of Jilin University, Changchun, Jilin 130021, China
| | - Yao Wu
- Department of Pediatric Intensive Care Unit, Children's Medical Center, The First Hospital of Jilin University, Changchun, Jilin 130021, China
| | - Yu-Mei Li
- Department of Pediatric Intensive Care Unit, Children's Medical Center, The First Hospital of Jilin University, Changchun, Jilin 130021, China
| |
Collapse
|
127
|
Hertzler JI, Teng J, Bernard AR, Stone MC, Kline HL, Mahata G, Kumar N, Rolls MM. Voltage-gated calcium channels act upstream of adenylyl cyclase Ac78C to promote timely initiation of dendrite regeneration. PLoS Genet 2024; 20:e1011388. [PMID: 39186815 PMCID: PMC11379402 DOI: 10.1371/journal.pgen.1011388] [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: 03/07/2024] [Revised: 09/06/2024] [Accepted: 08/06/2024] [Indexed: 08/28/2024] Open
Abstract
Most neurons are not replaced after injury and thus possess robust intrinsic mechanisms for repair after damage. Axon injury triggers a calcium wave, and calcium and cAMP can augment axon regeneration. In comparison to axon regeneration, dendrite regeneration is poorly understood. To test whether calcium and cAMP might also be involved in dendrite injury signaling, we tracked the responses of Drosophila dendritic arborization neurons to laser severing of axons and dendrites. We found that calcium and subsequently cAMP accumulate in the cell body after both dendrite and axon injury. Two voltage-gated calcium channels (VGCCs), L-Type and T-Type, are required for the calcium influx in response to dendrite injury and play a role in rapid initiation of dendrite regeneration. The AC8 family adenylyl cyclase, Ac78C, is required for cAMP production after dendrite injury and timely initiation of regeneration. Injury-induced cAMP production is sensitive to VGCC reduction, placing calcium upstream of cAMP generation. We propose that two VGCCs initiate global calcium influx in response to dendrite injury followed by production of cAMP by Ac78C. This signaling pathway promotes timely initiation of dendrite regrowth several hours after dendrite damage.
Collapse
Affiliation(s)
- J Ian Hertzler
- Biochemistry and Molecular Biology and the Huck Institutes of the Life Sciences The Pennsylvania State University, University Park, Pennsylvania, United States of America
| | - Jiajing Teng
- Biochemistry and Molecular Biology and the Huck Institutes of the Life Sciences The Pennsylvania State University, University Park, Pennsylvania, United States of America
| | - Annabelle R Bernard
- Biochemistry and Molecular Biology and the Huck Institutes of the Life Sciences The Pennsylvania State University, University Park, Pennsylvania, United States of America
| | - Michelle C Stone
- Biochemistry and Molecular Biology and the Huck Institutes of the Life Sciences The Pennsylvania State University, University Park, Pennsylvania, United States of America
| | - Hannah L Kline
- Biochemistry and Molecular Biology and the Huck Institutes of the Life Sciences The Pennsylvania State University, University Park, Pennsylvania, United States of America
| | - Gibarni Mahata
- Biochemistry and Molecular Biology and the Huck Institutes of the Life Sciences The Pennsylvania State University, University Park, Pennsylvania, United States of America
| | - Nitish Kumar
- Biochemistry and Molecular Biology and the Huck Institutes of the Life Sciences The Pennsylvania State University, University Park, Pennsylvania, United States of America
| | - Melissa M Rolls
- Biochemistry and Molecular Biology and the Huck Institutes of the Life Sciences The Pennsylvania State University, University Park, Pennsylvania, United States of America
| |
Collapse
|
128
|
Frechou MA, Martin SS, McDermott KD, Huaman EA, Gökhan Ş, Tomé WA, Coen-Cagli R, Gonçalves JT. Adult neurogenesis improves spatial information encoding in the mouse hippocampus. Nat Commun 2024; 15:6410. [PMID: 39080283 PMCID: PMC11289285 DOI: 10.1038/s41467-024-50699-x] [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: 06/24/2024] [Indexed: 08/02/2024] Open
Abstract
Adult neurogenesis is a unique form of neuronal plasticity in which newly generated neurons are integrated into the adult dentate gyrus in a process that is modulated by environmental stimuli. Adult-born neurons can contribute to spatial memory, but it is unknown whether they alter neural representations of space in the hippocampus. Using in vivo two-photon calcium imaging, we find that male and female mice previously housed in an enriched environment, which triggers an increase in neurogenesis, have increased spatial information encoding in the dentate gyrus. Ablating adult neurogenesis blocks the effect of enrichment and lowers spatial information, as does the chemogenetic silencing of adult-born neurons. Both ablating neurogenesis and silencing adult-born neurons decreases the calcium activity of dentate gyrus neurons, resulting in a decreased amplitude of place-specific responses. These findings are in contrast with previous studies that suggested a predominantly inhibitory action for adult-born neurons. We propose that adult neurogenesis improves representations of space by increasing the gain of dentate gyrus neurons and thereby improving their ability to tune to spatial features. This mechanism may mediate the beneficial effects of environmental enrichment on spatial learning and memory.
Collapse
Affiliation(s)
- M Agustina Frechou
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY, USA
- Gottesmann Institute for Stem Cell Biology and Regenerative Medicine, Albert Einstein College of Medicine, Bronx, NY, USA
- Laboratory of Neurotechnology and Biophysics, The Rockefeller University, New York, NY, USA
| | - Sunaina S Martin
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY, USA
- Gottesmann Institute for Stem Cell Biology and Regenerative Medicine, Albert Einstein College of Medicine, Bronx, NY, USA
- Department of Psychology, University of California San Diego, La Jolla, CA, USA
| | - Kelsey D McDermott
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY, USA
- Gottesmann Institute for Stem Cell Biology and Regenerative Medicine, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Evan A Huaman
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY, USA
- Gottesmann Institute for Stem Cell Biology and Regenerative Medicine, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Şölen Gökhan
- Saul R. Korey Department of Neurology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Wolfgang A Tomé
- Saul R. Korey Department of Neurology, Albert Einstein College of Medicine, Bronx, NY, USA
- Department of Radiation Oncology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Ruben Coen-Cagli
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY, USA
- Department of Systems and Computational Biology, Albert Einstein College of Medicine, Bronx, NY, USA
- Department of Ophthalmology and Visual Sciences, Albert Einstein College of Medicine, Bronx, NY, USA
| | - J Tiago Gonçalves
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY, USA.
- Gottesmann Institute for Stem Cell Biology and Regenerative Medicine, Albert Einstein College of Medicine, Bronx, NY, USA.
| |
Collapse
|
129
|
Wang T, Lee TS, Yao H, Hong J, Li Y, Jiang H, Andolina IM, Tang S. Large-scale calcium imaging reveals a systematic V4 map for encoding natural scenes. Nat Commun 2024; 15:6401. [PMID: 39080309 PMCID: PMC11289446 DOI: 10.1038/s41467-024-50821-z] [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: 11/15/2023] [Accepted: 07/22/2024] [Indexed: 08/02/2024] Open
Abstract
Biological visual systems have evolved to process natural scenes. A full understanding of visual cortical functions requires a comprehensive characterization of how neuronal populations in each visual area encode natural scenes. Here, we utilized widefield calcium imaging to record V4 cortical response to tens of thousands of natural images in male macaques. Using this large dataset, we developed a deep-learning digital twin of V4 that allowed us to map the natural image preferences of the neural population at 100-µm scale. This detailed map revealed a diverse set of functional domains in V4, each encoding distinct natural image features. We validated these model predictions using additional widefield imaging and single-cell resolution two-photon imaging. Feature attribution analysis revealed that these domains lie along a continuum from preferring spatially localized shape features to preferring spatially dispersed surface features. These results provide insights into the organizing principles that govern natural scene encoding in V4.
Collapse
Affiliation(s)
- Tianye Wang
- Peking University School of Life Sciences, Beijing, 100871, China
- Peking-Tsinghua Center for Life Sciences, Beijing, 100871, China
- IDG/McGovern Institute for Brain Research at Peking University, Beijing, 100871, China
- Key Laboratory of Machine Perception (Ministry of Education), Peking University, Beijing, 100871, China
| | - Tai Sing Lee
- Computer Science Department and Neuroscience Institute, Carnegie Mellon University, Pittsburgh, PA, 15213, USA
| | - Haoxuan Yao
- Peking University School of Life Sciences, Beijing, 100871, China
- Peking-Tsinghua Center for Life Sciences, Beijing, 100871, China
- IDG/McGovern Institute for Brain Research at Peking University, Beijing, 100871, China
- Key Laboratory of Machine Perception (Ministry of Education), Peking University, Beijing, 100871, China
| | - Jiayi Hong
- Peking University School of Life Sciences, Beijing, 100871, China
| | - Yang Li
- Peking University School of Life Sciences, Beijing, 100871, China
- Peking-Tsinghua Center for Life Sciences, Beijing, 100871, China
- IDG/McGovern Institute for Brain Research at Peking University, Beijing, 100871, China
- Key Laboratory of Machine Perception (Ministry of Education), Peking University, Beijing, 100871, China
| | - Hongfei Jiang
- Peking University School of Life Sciences, Beijing, 100871, China
- Peking-Tsinghua Center for Life Sciences, Beijing, 100871, China
- IDG/McGovern Institute for Brain Research at Peking University, Beijing, 100871, China
- Key Laboratory of Machine Perception (Ministry of Education), Peking University, Beijing, 100871, China
| | - Ian Max Andolina
- The Center for Excellence in Brain Science and Intelligence Technology, State Key Laboratory of Neuroscience, Key Laboratory of Primate Neurobiology, Institute of Neuroscience, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Shiming Tang
- Peking University School of Life Sciences, Beijing, 100871, China.
- Peking-Tsinghua Center for Life Sciences, Beijing, 100871, China.
- IDG/McGovern Institute for Brain Research at Peking University, Beijing, 100871, China.
- Key Laboratory of Machine Perception (Ministry of Education), Peking University, Beijing, 100871, China.
| |
Collapse
|
130
|
Rimoli CV, Moretti C, Soldevila F, Brémont E, Ventalon C, Gigan S. Demixing fluorescence time traces transmitted by multimode fibers. Nat Commun 2024; 15:6286. [PMID: 39060262 PMCID: PMC11282286 DOI: 10.1038/s41467-024-50306-z] [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: 06/01/2023] [Accepted: 07/05/2024] [Indexed: 07/28/2024] Open
Abstract
Optical methods based on thin multimode fibers (MMFs) are promising tools for measuring neuronal activity in deep brain regions of freely moving mice thanks to their small diameter. However, current methods are limited: while fiber photometry provides only ensemble activity, imaging techniques using of long multimode fibers are very sensitive to bending and have not been applied to unrestrained rodents yet. Here, we demonstrate the fundamentals of a new approach using a short MMF coupled to a miniscope. In proof-of-principle in vitro experiments, we disentangled spatio-temporal fluorescence signals from multiple fluorescent sources transmitted by a thin (200 µm) and short (8 mm) MMF, using a general unconstrained non-negative matrix factorization algorithm directly on the raw video data. Furthermore, we show that low-cost open-source miniscopes have sufficient sensitivity to image the same fluorescence patterns seen in our proof-of-principle experiment, suggesting a new avenue for novel minimally invasive deep brain studies using multimode fibers in freely behaving mice.
Collapse
Affiliation(s)
- Caio Vaz Rimoli
- Laboratoire Kastler Brossel, ENS-Université PSL, CNRS, Sorbonne Université, Collège de France, 24 Rue Lhomond, Paris, F-75005, France
- Institut de Biologie de l'ENS (IBENS), Département de biologie, École normale supérieure, CNRS, INSERM, Université PSL, 75005, Paris, France
| | - Claudio Moretti
- Laboratoire Kastler Brossel, ENS-Université PSL, CNRS, Sorbonne Université, Collège de France, 24 Rue Lhomond, Paris, F-75005, France
| | - Fernando Soldevila
- Laboratoire Kastler Brossel, ENS-Université PSL, CNRS, Sorbonne Université, Collège de France, 24 Rue Lhomond, Paris, F-75005, France
| | - Enora Brémont
- Institut de Biologie de l'ENS (IBENS), Département de biologie, École normale supérieure, CNRS, INSERM, Université PSL, 75005, Paris, France
| | - Cathie Ventalon
- Institut de Biologie de l'ENS (IBENS), Département de biologie, École normale supérieure, CNRS, INSERM, Université PSL, 75005, Paris, France.
| | - Sylvain Gigan
- Laboratoire Kastler Brossel, ENS-Université PSL, CNRS, Sorbonne Université, Collège de France, 24 Rue Lhomond, Paris, F-75005, France.
| |
Collapse
|
131
|
Woo MS, Mayer C, Binkle-Ladisch L, Sonner JK, Rosenkranz SC, Shaposhnykov A, Rothammer N, Tsvilovskyy V, Lorenz SM, Raich L, Bal LC, Vieira V, Wagner I, Bauer S, Glatzel M, Conrad M, Merkler D, Freichel M, Friese MA. STING orchestrates the neuronal inflammatory stress response in multiple sclerosis. Cell 2024; 187:4043-4060.e30. [PMID: 38878778 DOI: 10.1016/j.cell.2024.05.031] [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: 01/16/2024] [Revised: 04/08/2024] [Accepted: 05/16/2024] [Indexed: 07/28/2024]
Abstract
Inflammation-induced neurodegeneration is a defining feature of multiple sclerosis (MS), yet the underlying mechanisms remain unclear. By dissecting the neuronal inflammatory stress response, we discovered that neurons in MS and its mouse model induce the stimulator of interferon genes (STING). However, activation of neuronal STING requires its detachment from the stromal interaction molecule 1 (STIM1), a process triggered by glutamate excitotoxicity. This detachment initiates non-canonical STING signaling, which leads to autophagic degradation of glutathione peroxidase 4 (GPX4), essential for neuronal redox homeostasis and thereby inducing ferroptosis. Both genetic and pharmacological interventions that target STING in neurons protect against inflammation-induced neurodegeneration. Our findings position STING as a central regulator of the detrimental neuronal inflammatory stress response, integrating inflammation with glutamate signaling to cause neuronal cell death, and present it as a tractable target for treating neurodegeneration in MS.
Collapse
Affiliation(s)
- Marcel S Woo
- Institute of Neuroimmunology and Multiple Sclerosis, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Christina Mayer
- Institute of Neuroimmunology and Multiple Sclerosis, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Lars Binkle-Ladisch
- Institute of Neuroimmunology and Multiple Sclerosis, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Jana K Sonner
- Institute of Neuroimmunology and Multiple Sclerosis, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Sina C Rosenkranz
- Institute of Neuroimmunology and Multiple Sclerosis, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Artem Shaposhnykov
- Institute of Neuroimmunology and Multiple Sclerosis, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Nicola Rothammer
- Institute of Neuroimmunology and Multiple Sclerosis, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Volodymyr Tsvilovskyy
- Institute of Pharmacology, Heidelberg University, Heidelberg, Germany; DZHK (German Centre for Cardiovascular Research), partner site Heidelberg/Mannheim, Heidelberg, Germany
| | - Svenja M Lorenz
- Institute of Metabolism and Cell Death, Helmholtz Zentrum München, Neuherberg, Germany
| | - Lukas Raich
- Institute of Neuroimmunology and Multiple Sclerosis, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Lukas C Bal
- Institute of Neuroimmunology and Multiple Sclerosis, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Vanessa Vieira
- Institute of Neuroimmunology and Multiple Sclerosis, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Ingrid Wagner
- Department of Pathology and Immunology, Division of Clinical Pathology, Faculty of Medicine, University and University Hospital of Geneva, Geneva, Switzerland
| | - Simone Bauer
- Institute of Neuroimmunology and Multiple Sclerosis, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Markus Glatzel
- Institute of Neuropathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Marcus Conrad
- Institute of Metabolism and Cell Death, Helmholtz Zentrum München, Neuherberg, Germany
| | - Doron Merkler
- Department of Pathology and Immunology, Division of Clinical Pathology, Faculty of Medicine, University and University Hospital of Geneva, Geneva, Switzerland
| | - Marc Freichel
- Institute of Pharmacology, Heidelberg University, Heidelberg, Germany; DZHK (German Centre for Cardiovascular Research), partner site Heidelberg/Mannheim, Heidelberg, Germany
| | - Manuel A Friese
- Institute of Neuroimmunology and Multiple Sclerosis, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.
| |
Collapse
|
132
|
Quass GL, Rogalla MM, Ford AN, Apostolides PF. Mixed Representations of Sound and Action in the Auditory Midbrain. J Neurosci 2024; 44:e1831232024. [PMID: 38918064 PMCID: PMC11270520 DOI: 10.1523/jneurosci.1831-23.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Revised: 06/05/2024] [Accepted: 06/14/2024] [Indexed: 06/27/2024] Open
Abstract
Linking sensory input and its consequences is a fundamental brain operation. During behavior, the neural activity of neocortical and limbic systems often reflects dynamic combinations of sensory and task-dependent variables, and these "mixed representations" are suggested to be important for perception, learning, and plasticity. However, the extent to which such integrative computations might occur outside of the forebrain is less clear. Here, we conduct cellular-resolution two-photon Ca2+ imaging in the superficial "shell" layers of the inferior colliculus (IC), as head-fixed mice of either sex perform a reward-based psychometric auditory task. We find that the activity of individual shell IC neurons jointly reflects auditory cues, mice's actions, and behavioral trial outcomes, such that trajectories of neural population activity diverge depending on mice's behavioral choice. Consequently, simple classifier models trained on shell IC neuron activity can predict trial-by-trial outcomes, even when training data are restricted to neural activity occurring prior to mice's instrumental actions. Thus, in behaving mice, auditory midbrain neurons transmit a population code that reflects a joint representation of sound, actions, and task-dependent variables.
Collapse
Affiliation(s)
- Gunnar L Quass
- Department of Otolaryngology-Head & Neck Surgery, Kresge Hearing Research Institute, University of Michigan Medical School, Ann Arbor, Michigan 48109
| | - Meike M Rogalla
- Department of Otolaryngology-Head & Neck Surgery, Kresge Hearing Research Institute, University of Michigan Medical School, Ann Arbor, Michigan 48109
| | - Alexander N Ford
- Department of Otolaryngology-Head & Neck Surgery, Kresge Hearing Research Institute, University of Michigan Medical School, Ann Arbor, Michigan 48109
| | - Pierre F Apostolides
- Department of Otolaryngology-Head & Neck Surgery, Kresge Hearing Research Institute, University of Michigan Medical School, Ann Arbor, Michigan 48109
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, Michigan 48109
| |
Collapse
|
133
|
Fan Z, Karakone M, Nagarajan S, Nagy N, Mildenberger W, Petrova E, Hinte LC, Bijnen M, Häne P, Nelius E, Chen J, Ferapontova I, von Meyenn F, Trepiccione F, Berber M, Ribas DP, Eichmann A, Zennaro MC, Takeda N, Fischer JW, Spyroglou A, Reincke M, Beuschlein F, Loffing J, Greter M, Stockmann C. Macrophages preserve endothelial cell specialization in the adrenal gland to modulate aldosterone secretion and blood pressure. Cell Rep 2024; 43:114395. [PMID: 38941187 DOI: 10.1016/j.celrep.2024.114395] [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: 02/09/2023] [Revised: 04/22/2024] [Accepted: 06/07/2024] [Indexed: 06/30/2024] Open
Abstract
Macrophages play crucial roles in organ-specific functions and homeostasis. In the adrenal gland, macrophages closely associate with sinusoidal capillaries in the aldosterone-producing zona glomerulosa. We demonstrate that macrophages preserve capillary specialization and modulate aldosterone secretion. Using macrophage-specific deletion of VEGF-A, single-cell transcriptomics, and functional phenotyping, we found that the loss of VEGF-A depletes PLVAP+ fenestrated endothelial cells in the zona glomerulosa, leading to increased basement membrane collagen IV deposition and subendothelial fibrosis. This results in increased aldosterone secretion, called "haptosecretagogue" signaling. Human aldosterone-producing adenomas also show capillary rarefaction and basement membrane thickening. Mice with myeloid cell-specific VEGF-A deletion exhibit elevated serum aldosterone, hypokalemia, and hypertension, mimicking primary aldosteronism. These findings underscore macrophage-to-endothelial cell signaling as essential for endothelial cell specialization, adrenal gland function, and blood pressure regulation, with broader implications for other endocrine organs.
Collapse
Affiliation(s)
- Zheng Fan
- University of Zurich, Institute of Anatomy, 8057 Zurich, Switzerland.
| | - Mara Karakone
- University of Zurich, Institute of Anatomy, 8057 Zurich, Switzerland
| | | | - Nadine Nagy
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Wiebke Mildenberger
- University of Zurich, Institute for Experimental Immunology, 8057 Zurich, Switzerland
| | - Ekaterina Petrova
- University of Zurich, Institute for Experimental Immunology, 8057 Zurich, Switzerland
| | - Laura Catharina Hinte
- Laboratory of Nutrition and Metabolic Epigenetics, Institute for Food, Nutrition and Health, Department of Health Sciences and Technology, ETH Zurich, Zurich, Switzerland
| | - Mitchell Bijnen
- University of Zurich, Institute for Experimental Immunology, 8057 Zurich, Switzerland
| | - Philipp Häne
- University of Zurich, Institute for Experimental Immunology, 8057 Zurich, Switzerland
| | - Eric Nelius
- University of Zurich, Institute of Anatomy, 8057 Zurich, Switzerland
| | - Jing Chen
- University of Zurich, Institute of Anatomy, 8057 Zurich, Switzerland
| | - Irina Ferapontova
- University of Zurich, Institute of Anatomy, 8057 Zurich, Switzerland
| | - Ferdinand von Meyenn
- Laboratory of Nutrition and Metabolic Epigenetics, Institute for Food, Nutrition and Health, Department of Health Sciences and Technology, ETH Zurich, Zurich, Switzerland
| | - Francesco Trepiccione
- Department of Translational Medical Sciences, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Mesut Berber
- University of Zurich, Institute of Anatomy, 8057 Zurich, Switzerland
| | - David Penton Ribas
- Electrophysiology Facility (e-phac), Department of Molecular Life Sciences, University of Zurich (UZH), 8057 Zürich, Switzerland
| | - Anne Eichmann
- Cardiovascular Research Center and Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, CT, USA
| | | | - Norihiko Takeda
- Department of Cardiovascular Medicine, Graduate School of Medicine, The University of Tokyo, 7-3-1, Hongo, Bunkyo-ku, Tokyo, Japan
| | - Jens W Fischer
- Institute of Pharmacology and Clinical Pharmacology, Medical Faculty, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | - Ariadni Spyroglou
- Klinik für Endokrinologie, Diabetologie, und Klinische Ernährung, UniversitätsSpital Zürich (USZ) and UZH, Raemistrasse 100, 8091 Zurich, Switzerland; Medizinische Klinik und Poliklinik IV, Ludwig-Maximilians-University (LMU), Munich, Germany
| | - Martin Reincke
- Klinik für Endokrinologie, Diabetologie, und Klinische Ernährung, UniversitätsSpital Zürich (USZ) and UZH, Raemistrasse 100, 8091 Zurich, Switzerland
| | - Felix Beuschlein
- Klinik für Endokrinologie, Diabetologie, und Klinische Ernährung, UniversitätsSpital Zürich (USZ) and UZH, Raemistrasse 100, 8091 Zurich, Switzerland; Medizinische Klinik und Poliklinik IV, Ludwig-Maximilians-University (LMU), Munich, Germany
| | - Johannes Loffing
- University of Zurich, Institute of Anatomy, 8057 Zurich, Switzerland
| | - Melanie Greter
- University of Zurich, Institute for Experimental Immunology, 8057 Zurich, Switzerland
| | - Christian Stockmann
- University of Zurich, Institute of Anatomy, 8057 Zurich, Switzerland; INSERM U970, Paris Cardiovascular Research Center, Paris, France.
| |
Collapse
|
134
|
Apuschkin M, Burm HB, Schmidt JH, Skov LJ, Andersen RC, Bowin CF, Støier JF, Jensen KL, Posselt LP, Dmytriyeva O, Sørensen AT, Egerod KL, Holst B, Rickhag M, Schwartz TW, Gether U. An atlas of GPCRs in dopamine neurons: Identification of the free fatty acid receptor 4 as a regulator of food and water intake. Cell Rep 2024; 43:114509. [PMID: 39003735 DOI: 10.1016/j.celrep.2024.114509] [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: 05/09/2023] [Revised: 04/03/2024] [Accepted: 06/28/2024] [Indexed: 07/16/2024] Open
Abstract
Midbrain dopaminergic neurons (DANs) are subject to extensive metabotropic regulation, but the repertoire of G protein-coupled receptors (GPCRs) present in these neurons has not been mapped. Here, we isolate DANs from Dat-eGFP mice to generate a GPCR atlas by unbiased qPCR array expression analysis of 377 GPCRs. Combined with data mining of scRNA-seq databases, we identify multiple receptors in DAN subpopulations with 38 of these receptors representing the majority of transcripts. We identify 41 receptors expressed in midbrain DANs but not in non-DAN midbrain cells, including the free fatty acid receptor 4 (FFAR4). Functional expression of FFAR4 is validated by ex vivo Ca2+ imaging, and in vivo experiments support that FFAR4 negatively regulates food and water intake and bodyweight. In addition to providing a critical framework for understanding metabotropic DAN regulation, our data suggest fatty acid sensing by FFAR4 as a mechanism linking high-energy intake to the dopamine-reward pathway.
Collapse
Affiliation(s)
- Mia Apuschkin
- Molecular Neuropharmacology and Genetics Laboratory, Department of Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Hayley B Burm
- Molecular Neuropharmacology and Genetics Laboratory, Department of Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Jan H Schmidt
- Molecular Neuropharmacology and Genetics Laboratory, Department of Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Louise J Skov
- Novo Nordic Foundation Center for Basic Metabolic Research, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Rita C Andersen
- Molecular Neuropharmacology and Genetics Laboratory, Department of Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Carl-Fredrik Bowin
- Molecular Neuropharmacology and Genetics Laboratory, Department of Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark; Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Jonatan F Støier
- Molecular Neuropharmacology and Genetics Laboratory, Department of Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Kathrine L Jensen
- Molecular Neuropharmacology and Genetics Laboratory, Department of Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Leonie P Posselt
- Molecular Neuropharmacology and Genetics Laboratory, Department of Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Oksana Dmytriyeva
- Novo Nordic Foundation Center for Basic Metabolic Research, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Andreas T Sørensen
- Molecular Neuropharmacology and Genetics Laboratory, Department of Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Kristoffer L Egerod
- Novo Nordic Foundation Center for Basic Metabolic Research, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Birgitte Holst
- Department of Biomedical Sciences, Laboratory for Molecular Pharmacology and Novo Nordic Foundation Center for Basic Metabolic Research, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Mattias Rickhag
- Molecular Neuropharmacology and Genetics Laboratory, Department of Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark; Danish Research Centre for Magnetic Resonance (DRCMR), Copenhagen University Hospital Amager and Hvidovre, Hvidovre, Denmark
| | - Thue W Schwartz
- Novo Nordic Foundation Center for Basic Metabolic Research, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Ulrik Gether
- Molecular Neuropharmacology and Genetics Laboratory, Department of Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark.
| |
Collapse
|
135
|
Deister CA, Moore AI, Voigts J, Bechek S, Lichtin R, Brown TC, Moore CI. Neocortical inhibitory imbalance predicts successful sensory detection. Cell Rep 2024; 43:114233. [PMID: 38905102 DOI: 10.1016/j.celrep.2024.114233] [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: 07/16/2021] [Revised: 07/17/2023] [Accepted: 04/26/2024] [Indexed: 06/23/2024] Open
Abstract
Perceptual success depends on fast-spiking, parvalbumin-positive interneurons (FS/PVs). However, competing theories of optimal rate and correlation in pyramidal (PYR) firing make opposing predictions regarding the underlying FS/PV dynamics. We addressed this with population calcium imaging of FS/PVs and putative PYR neurons during threshold detection. In primary somatosensory and visual neocortex, a distinct PYR subset shows increased rate and spike-count correlations on detected trials ("hits"), while most show no rate change and decreased correlations. A larger fraction of FS/PVs predicts hits with either rate increases or decreases. Using computational modeling, we found that inhibitory imbalance, created by excitatory "feedback" and interactions between FS/PV pools, can account for the data. Rate-decreasing FS/PVs increase rate and correlation in a PYR subset, while rate-increasing FS/PVs reduce correlations and offset enhanced excitation in PYR neurons. These findings indicate that selection of informative PYR ensembles, through transient inhibitory imbalance, is a common motif of optimal neocortical processing.
Collapse
Affiliation(s)
- Christopher A Deister
- Department of Neuroscience and Carney Institute for Brain Sciences, Brown University, Providence, RI, USA
| | - Alexander I Moore
- Department of Neuroscience and Carney Institute for Brain Sciences, Brown University, Providence, RI, USA
| | - Jakob Voigts
- Department of Neuroscience and Carney Institute for Brain Sciences, Brown University, Providence, RI, USA; Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA, USA
| | - Sophia Bechek
- Department of Neuroscience and Carney Institute for Brain Sciences, Brown University, Providence, RI, USA
| | - Rebecca Lichtin
- Department of Neuroscience and Carney Institute for Brain Sciences, Brown University, Providence, RI, USA
| | - Tyler C Brown
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Christopher I Moore
- Department of Neuroscience and Carney Institute for Brain Sciences, Brown University, Providence, RI, USA.
| |
Collapse
|
136
|
Mim MS, Kumar N, Levis M, Unger MF, Miranda G, Gazzo D, Robinett T, Zartman JJ. Piezo regulates epithelial topology and promotes precision in organ size control. Cell Rep 2024; 43:114398. [PMID: 38935502 DOI: 10.1016/j.celrep.2024.114398] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Revised: 05/09/2024] [Accepted: 06/10/2024] [Indexed: 06/29/2024] Open
Abstract
Mechanosensitive Piezo channels regulate cell division, cell extrusion, and cell death. However, systems-level functions of Piezo in regulating organogenesis remain poorly understood. Here, we demonstrate that Piezo controls epithelial cell topology to ensure precise organ growth by integrating live-imaging experiments with pharmacological and genetic perturbations and computational modeling. Notably, the knockout or knockdown of Piezo increases bilateral asymmetry in wing size. Piezo's multifaceted functions can be deconstructed as either autonomous or non-autonomous based on a comparison between tissue-compartment-level perturbations or between genetic perturbation populations at the whole-tissue level. A computational model that posits cell proliferation and apoptosis regulation through modulation of the cutoff tension required for Piezo channel activation explains key cell and tissue phenotypes arising from perturbations of Piezo expression levels. Our findings demonstrate that Piezo promotes robustness in regulating epithelial topology and is necessary for precise organ size control.
Collapse
Affiliation(s)
- Mayesha Sahir Mim
- Department of Chemical and Biomolecular Engineering, University of Notre Dame, Notre Dame, IN 46556, USA; Bioengineering Graduate Program, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Nilay Kumar
- Department of Chemical and Biomolecular Engineering, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Megan Levis
- Department of Chemical and Biomolecular Engineering, University of Notre Dame, Notre Dame, IN 46556, USA; Bioengineering Graduate Program, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Maria F Unger
- Department of Chemical and Biomolecular Engineering, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Gabriel Miranda
- Department of Chemical and Biomolecular Engineering, University of Notre Dame, Notre Dame, IN 46556, USA
| | - David Gazzo
- Department of Chemical and Biomolecular Engineering, University of Notre Dame, Notre Dame, IN 46556, USA; Bioengineering Graduate Program, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Trent Robinett
- Department of Chemical and Biomolecular Engineering, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Jeremiah J Zartman
- Department of Chemical and Biomolecular Engineering, University of Notre Dame, Notre Dame, IN 46556, USA; Bioengineering Graduate Program, University of Notre Dame, Notre Dame, IN 46556, USA; Department of Biological Sciences, University of Notre Dame, Notre Dame, IN 46556, USA.
| |
Collapse
|
137
|
Masala N, Mittag M, Giovannetti EA, O'Neil DA, Distler FJ, Rupprecht P, Helmchen F, Yuste R, Fuhrmann M, Beck H, Wenzel M, Kelly T. Aberrant hippocampal Ca 2+ microwaves following synapsin-dependent adeno-associated viral expression of Ca 2+ indicators. eLife 2024; 13:RP93804. [PMID: 39042440 PMCID: PMC11265795 DOI: 10.7554/elife.93804] [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] [Indexed: 07/24/2024] Open
Abstract
Genetically encoded calcium indicators (GECIs) such as GCaMP are invaluable tools in neuroscience to monitor neuronal activity using optical imaging. The viral transduction of GECIs is commonly used to target expression to specific brain regions, can be conveniently used with any mouse strain of interest without the need for prior crossing with a GECI mouse line, and avoids potential hazards due to the chronic expression of GECIs during development. A key requirement for monitoring neuronal activity with an indicator is that the indicator itself minimally affects activity. Here, using common adeno-associated viral (AAV) transduction procedures, we describe spatially confined aberrant Ca2+ microwaves slowly travelling through the hippocampus following expression of GCaMP6, GCaMP7, or R-CaMP1.07 driven by the synapsin promoter with AAV-dependent gene transfer in a titre-dependent fashion. Ca2+ microwaves developed in hippocampal CA1 and CA3, but not dentate gyrus nor neocortex, were typically first observed at 4 wk after viral transduction, and persisted up to at least 8 wk. The phenomenon was robust and observed across laboratories with various experimenters and setups. Our results indicate that aberrant hippocampal Ca2+ microwaves depend on the promoter and viral titre of the GECI, density of expression, as well as the targeted brain region. We used an alternative viral transduction method of GCaMP which avoids this artefact. The results show that commonly used Ca2+-indicator AAV transduction procedures can produce artefactual Ca2+ responses. Our aim is to raise awareness in the field of these artefactual transduction-induced Ca2+ microwaves, and we provide a potential solution.
Collapse
Affiliation(s)
- Nicola Masala
- University of Bonn, Faculty of Medicine, Institute for Experimental Epileptology and Cognition Research (IEECR)BonnGermany
- University Hospital BonnBonnGermany
- Department of Epileptology, University Hospital BonnBonnGermany
| | - Manuel Mittag
- Neuroimmunology and Imaging Group, German Center for Neurodegenerative Diseases (DZNE)BonnGermany
| | | | - Darik A O'Neil
- NeuroTechnology Center, Columbia UniversityNew YorkUnited States
| | - Fabian J Distler
- University of Bonn, Faculty of Medicine, Institute for Experimental Epileptology and Cognition Research (IEECR)BonnGermany
- University Hospital BonnBonnGermany
| | - Peter Rupprecht
- Brain Research Institute, University of ZurichZurichSwitzerland
- Neuroscience Center Zurich, University of ZurichZurichSwitzerland
| | - Fritjof Helmchen
- Brain Research Institute, University of ZurichZurichSwitzerland
- Neuroscience Center Zurich, University of ZurichZurichSwitzerland
| | - Rafael Yuste
- NeuroTechnology Center, Columbia UniversityNew YorkUnited States
| | - Martin Fuhrmann
- Neuroimmunology and Imaging Group, German Center for Neurodegenerative Diseases (DZNE)BonnGermany
| | - Heinz Beck
- University of Bonn, Faculty of Medicine, Institute for Experimental Epileptology and Cognition Research (IEECR)BonnGermany
- University Hospital BonnBonnGermany
- German Center for Neurodegenerative Diseases (DZNE)BonnGermany
| | - Michael Wenzel
- University of Bonn, Faculty of Medicine, Institute for Experimental Epileptology and Cognition Research (IEECR)BonnGermany
- University Hospital BonnBonnGermany
- Department of Epileptology, University Hospital BonnBonnGermany
| | - Tony Kelly
- University of Bonn, Faculty of Medicine, Institute for Experimental Epileptology and Cognition Research (IEECR)BonnGermany
- University Hospital BonnBonnGermany
| |
Collapse
|
138
|
Kubrak O, Jørgensen AF, Koyama T, Lassen M, Nagy S, Hald J, Mazzoni G, Madsen D, Hansen JB, Larsen MR, Texada MJ, Hansen JL, Halberg KV, Rewitz K. LGR signaling mediates muscle-adipose tissue crosstalk and protects against diet-induced insulin resistance. Nat Commun 2024; 15:6126. [PMID: 39033139 PMCID: PMC11271308 DOI: 10.1038/s41467-024-50468-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Accepted: 07/04/2024] [Indexed: 07/23/2024] Open
Abstract
Obesity impairs tissue insulin sensitivity and signaling, promoting type-2 diabetes. Although improving insulin signaling is key to reversing diabetes, the multi-organ mechanisms regulating this process are poorly defined. Here, we screen the secretome and receptome in Drosophila to identify the hormonal crosstalk affecting diet-induced insulin resistance and obesity. We discover a complex interplay between muscle, neuronal, and adipose tissues, mediated by Bone Morphogenetic Protein (BMP) signaling and the hormone Bursicon, that enhances insulin signaling and sugar tolerance. Muscle-derived BMP signaling, induced by sugar, governs neuronal Bursicon signaling. Bursicon, through its receptor Rickets, a Leucine-rich-repeat-containing G-protein coupled receptor (LGR), improves insulin secretion and insulin sensitivity in adipose tissue, mitigating hyperglycemia. In mouse adipocytes, loss of the Rickets ortholog LGR4 blunts insulin responses, showing an essential role of LGR4 in adipocyte insulin sensitivity. Our findings reveal a muscle-neuronal-fat-tissue axis driving metabolic adaptation to high-sugar conditions, identifying LGR4 as a critical mediator in this regulatory network.
Collapse
Affiliation(s)
- Olga Kubrak
- Department of Biology, University of Copenhagen, 2100, Copenhagen O, Denmark
| | - Anne F Jørgensen
- Department of Biology, University of Copenhagen, 2100, Copenhagen O, Denmark
- Novo Nordisk, Novo Nordisk Park, 2760, Maaløv, Denmark
| | - Takashi Koyama
- Department of Biology, University of Copenhagen, 2100, Copenhagen O, Denmark
| | - Mette Lassen
- Department of Biology, University of Copenhagen, 2100, Copenhagen O, Denmark
| | - Stanislav Nagy
- Department of Biology, University of Copenhagen, 2100, Copenhagen O, Denmark
| | - Jacob Hald
- Novo Nordisk, Novo Nordisk Park, 2760, Maaløv, Denmark
| | | | - Dennis Madsen
- Novo Nordisk, Novo Nordisk Park, 2760, Maaløv, Denmark
| | - Jacob B Hansen
- Department of Biology, University of Copenhagen, 2100, Copenhagen O, Denmark
| | - Martin Røssel Larsen
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, 5230, Odense, Denmark
| | - Michael J Texada
- Department of Biology, University of Copenhagen, 2100, Copenhagen O, Denmark
| | | | - Kenneth V Halberg
- Department of Biology, University of Copenhagen, 2100, Copenhagen O, Denmark
| | - Kim Rewitz
- Department of Biology, University of Copenhagen, 2100, Copenhagen O, Denmark.
| |
Collapse
|
139
|
Nevelchuk S, Brawek B, Schwarz N, Valiente-Gabioud A, Wuttke TV, Kovalchuk Y, Koch H, Höllig A, Steiner F, Figarella K, Griesbeck O, Garaschuk O. Morphotype-specific calcium signaling in human microglia. J Neuroinflammation 2024; 21:175. [PMID: 39020359 PMCID: PMC11256502 DOI: 10.1186/s12974-024-03169-6] [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: 05/25/2024] [Accepted: 07/09/2024] [Indexed: 07/19/2024] Open
Abstract
BACKGROUND Key functions of Ca2+ signaling in rodent microglia include monitoring the brain state as well as the surrounding neuronal activity and sensing the danger or damage in their vicinity. Microglial Ca2+ dyshomeostasis is a disease hallmark in many mouse models of neurological disorders but the Ca2+ signal properties of human microglia remain unknown. METHODS We developed a novel genetically-encoded ratiometric Ca2+ indicator, targeting microglial cells in the freshly resected human tissue, organotypically cultured tissue slices and analyzed in situ ongoing Ca2+ signaling of decades-old microglia dwelling in their native microenvironment. RESULTS The data revealed marked compartmentalization of Ca2+ signals, with signal properties differing across the compartments and resident morphotypes. The basal Ca2+ levels were low in ramified and high in ameboid microglia. The fraction of cells with ongoing Ca2+ signaling, the fraction and the amplitude of process Ca2+ signals and the duration of somatic Ca2+ signals decreased when moving from ramified via hypertrophic to ameboid microglia. In contrast, the size of active compartments, the fraction and amplitude of somatic Ca2+ signals and the duration of process Ca2+ signals increased along this pathway.
Collapse
Affiliation(s)
- Sofia Nevelchuk
- Department of Neurophysiology, Institute of Physiology, Eberhard Karls University of Tübingen, Keplerstr. 15, 72074, Tübingen, Germany
| | - Bianca Brawek
- Department of Neurophysiology, Institute of Physiology, Eberhard Karls University of Tübingen, Keplerstr. 15, 72074, Tübingen, Germany
| | - Niklas Schwarz
- Department of Neurology and Epileptology, Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany
| | - Ariel Valiente-Gabioud
- Tools for Bio-Imaging, Max-Planck-Institute for Biological Intelligence, Martinsried, Germany
| | - Thomas V Wuttke
- Department of Neurology and Epileptology, Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany
- Department of Neurosurgery, University of Tübingen, Tübingen, Germany
| | - Yury Kovalchuk
- Department of Neurophysiology, Institute of Physiology, Eberhard Karls University of Tübingen, Keplerstr. 15, 72074, Tübingen, Germany
| | - Henner Koch
- Department of Epileptology, Neurology, RWTH Aachen University Hospital, Aachen, Germany
| | - Anke Höllig
- Department of Neurosurgery, RWTH Aachen University, Aachen, Germany
| | - Frederik Steiner
- Department of Neurophysiology, Institute of Physiology, Eberhard Karls University of Tübingen, Keplerstr. 15, 72074, Tübingen, Germany
| | - Katherine Figarella
- Department of Neurophysiology, Institute of Physiology, Eberhard Karls University of Tübingen, Keplerstr. 15, 72074, Tübingen, Germany
- Department of Anesthesiology, Critical Care and Pain Medicine, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Oliver Griesbeck
- Tools for Bio-Imaging, Max-Planck-Institute for Biological Intelligence, Martinsried, Germany
| | - Olga Garaschuk
- Department of Neurophysiology, Institute of Physiology, Eberhard Karls University of Tübingen, Keplerstr. 15, 72074, Tübingen, Germany.
| |
Collapse
|
140
|
Zhang SR, Wu DY, Luo R, Wu JL, Chen H, Li ZM, Zhuang JP, Hu NY, Li XW, Yang JM, Gao TM, Chen YH. A Prelimbic Cortex-Thalamus Circuit Bidirectionally Regulates Innate and Stress-Induced Anxiety-Like Behavior. J Neurosci 2024; 44:e2103232024. [PMID: 38886059 PMCID: PMC11255430 DOI: 10.1523/jneurosci.2103-23.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Revised: 06/05/2024] [Accepted: 06/10/2024] [Indexed: 06/20/2024] Open
Abstract
Anxiety-related disorders respond to cognitive behavioral therapies, which involved the medial prefrontal cortex (mPFC). Previous studies have suggested that subregions of the mPFC have different and even opposite roles in regulating innate anxiety. However, the specific causal targets of their descending projections in modulating innate anxiety and stress-induced anxiety have yet to be fully elucidated. Here, we found that among the various downstream pathways of the prelimbic cortex (PL), a subregion of the mPFC, PL-mediodorsal thalamic nucleus (MD) projection, and PL-ventral tegmental area (VTA) projection exhibited antagonistic effects on anxiety-like behavior, while the PL-MD projection but not PL-VTA projection was necessary for the animal to guide anxiety-related behavior. In addition, MD-projecting PL neurons bidirectionally regulated remote but not recent fear memory retrieval. Notably, restraint stress induced high-anxiety state accompanied by strengthening the excitatory inputs onto MD-projecting PL neurons, and inhibiting PL-MD pathway rescued the stress-induced anxiety. Our findings reveal that the activity of PL-MD pathway may be an essential factor to maintain certain level of anxiety, and stress increased the excitability of this pathway, leading to inappropriate emotional expression, and suggests that targeting specific PL circuits may aid the development of therapies for the treatment of stress-related disorders.
Collapse
Affiliation(s)
- Sheng-Rong Zhang
- State Key Laboratory of Organ Failure Research, Key Laboratory of Mental Health of the Ministry of Education, Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Guangdong-Hong Kong Joint Laboratory for Psychiatric Disorders, Guangdong Province Key Laboratory of Psychiatric Disorders, Guangdong Basic Research Center of Excellence for Integrated Traditional and Western Medicine for Qingzhi Diseases, Department of Neurobiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Ding-Yu Wu
- State Key Laboratory of Organ Failure Research, Key Laboratory of Mental Health of the Ministry of Education, Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Guangdong-Hong Kong Joint Laboratory for Psychiatric Disorders, Guangdong Province Key Laboratory of Psychiatric Disorders, Guangdong Basic Research Center of Excellence for Integrated Traditional and Western Medicine for Qingzhi Diseases, Department of Neurobiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Rong Luo
- State Key Laboratory of Organ Failure Research, Key Laboratory of Mental Health of the Ministry of Education, Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Guangdong-Hong Kong Joint Laboratory for Psychiatric Disorders, Guangdong Province Key Laboratory of Psychiatric Disorders, Guangdong Basic Research Center of Excellence for Integrated Traditional and Western Medicine for Qingzhi Diseases, Department of Neurobiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Jian-Lin Wu
- State Key Laboratory of Organ Failure Research, Key Laboratory of Mental Health of the Ministry of Education, Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Guangdong-Hong Kong Joint Laboratory for Psychiatric Disorders, Guangdong Province Key Laboratory of Psychiatric Disorders, Guangdong Basic Research Center of Excellence for Integrated Traditional and Western Medicine for Qingzhi Diseases, Department of Neurobiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Hao Chen
- State Key Laboratory of Organ Failure Research, Key Laboratory of Mental Health of the Ministry of Education, Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Guangdong-Hong Kong Joint Laboratory for Psychiatric Disorders, Guangdong Province Key Laboratory of Psychiatric Disorders, Guangdong Basic Research Center of Excellence for Integrated Traditional and Western Medicine for Qingzhi Diseases, Department of Neurobiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Zi-Ming Li
- State Key Laboratory of Organ Failure Research, Key Laboratory of Mental Health of the Ministry of Education, Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Guangdong-Hong Kong Joint Laboratory for Psychiatric Disorders, Guangdong Province Key Laboratory of Psychiatric Disorders, Guangdong Basic Research Center of Excellence for Integrated Traditional and Western Medicine for Qingzhi Diseases, Department of Neurobiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Jia-Pai Zhuang
- State Key Laboratory of Organ Failure Research, Key Laboratory of Mental Health of the Ministry of Education, Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Guangdong-Hong Kong Joint Laboratory for Psychiatric Disorders, Guangdong Province Key Laboratory of Psychiatric Disorders, Guangdong Basic Research Center of Excellence for Integrated Traditional and Western Medicine for Qingzhi Diseases, Department of Neurobiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Neng-Yuan Hu
- State Key Laboratory of Organ Failure Research, Key Laboratory of Mental Health of the Ministry of Education, Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Guangdong-Hong Kong Joint Laboratory for Psychiatric Disorders, Guangdong Province Key Laboratory of Psychiatric Disorders, Guangdong Basic Research Center of Excellence for Integrated Traditional and Western Medicine for Qingzhi Diseases, Department of Neurobiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Xiao-Wen Li
- State Key Laboratory of Organ Failure Research, Key Laboratory of Mental Health of the Ministry of Education, Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Guangdong-Hong Kong Joint Laboratory for Psychiatric Disorders, Guangdong Province Key Laboratory of Psychiatric Disorders, Guangdong Basic Research Center of Excellence for Integrated Traditional and Western Medicine for Qingzhi Diseases, Department of Neurobiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Jian-Ming Yang
- State Key Laboratory of Organ Failure Research, Key Laboratory of Mental Health of the Ministry of Education, Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Guangdong-Hong Kong Joint Laboratory for Psychiatric Disorders, Guangdong Province Key Laboratory of Psychiatric Disorders, Guangdong Basic Research Center of Excellence for Integrated Traditional and Western Medicine for Qingzhi Diseases, Department of Neurobiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Tian-Ming Gao
- State Key Laboratory of Organ Failure Research, Key Laboratory of Mental Health of the Ministry of Education, Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Guangdong-Hong Kong Joint Laboratory for Psychiatric Disorders, Guangdong Province Key Laboratory of Psychiatric Disorders, Guangdong Basic Research Center of Excellence for Integrated Traditional and Western Medicine for Qingzhi Diseases, Department of Neurobiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Yi-Hua Chen
- State Key Laboratory of Organ Failure Research, Key Laboratory of Mental Health of the Ministry of Education, Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Guangdong-Hong Kong Joint Laboratory for Psychiatric Disorders, Guangdong Province Key Laboratory of Psychiatric Disorders, Guangdong Basic Research Center of Excellence for Integrated Traditional and Western Medicine for Qingzhi Diseases, Department of Neurobiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| |
Collapse
|
141
|
Gauld OM, Packer AM, Russell LE, Dalgleish HWP, Iuga M, Sacadura F, Roth A, Clark BA, Häusser M. A latent pool of neurons silenced by sensory-evoked inhibition can be recruited to enhance perception. Neuron 2024; 112:2386-2403.e6. [PMID: 38729150 PMCID: PMC7616379 DOI: 10.1016/j.neuron.2024.04.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 02/12/2024] [Accepted: 04/12/2024] [Indexed: 05/12/2024]
Abstract
To investigate which activity patterns in sensory cortex are relevant for perceptual decision-making, we combined two-photon calcium imaging and targeted two-photon optogenetics to interrogate barrel cortex activity during perceptual discrimination. We trained mice to discriminate bilateral whisker deflections and report decisions by licking left or right. Two-photon calcium imaging revealed sparse coding of contralateral and ipsilateral whisker input in layer 2/3, with most neurons remaining silent during the task. Activating pyramidal neurons using two-photon holographic photostimulation evoked a perceptual bias that scaled with the number of neurons photostimulated. This effect was dominated by optogenetic activation of non-coding neurons, which did not show sensory or motor-related activity during task performance. Photostimulation also revealed potent recruitment of cortical inhibition during sensory processing, which strongly and preferentially suppressed non-coding neurons. Our results suggest that a pool of non-coding neurons, selectively suppressed by network inhibition during sensory processing, can be recruited to enhance perception.
Collapse
Affiliation(s)
- Oliver M Gauld
- Wolfson Institute for Biomedical Research, University College London, London WC1E 6BT, UK; Sainsbury Wellcome Centre, University College London, London W1T 4JG, UK.
| | - Adam M Packer
- Wolfson Institute for Biomedical Research, University College London, London WC1E 6BT, UK; Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford OX1 3PT, UK
| | - Lloyd E Russell
- Wolfson Institute for Biomedical Research, University College London, London WC1E 6BT, UK
| | - Henry W P Dalgleish
- Wolfson Institute for Biomedical Research, University College London, London WC1E 6BT, UK
| | - Maya Iuga
- Wolfson Institute for Biomedical Research, University College London, London WC1E 6BT, UK
| | - Francisco Sacadura
- Wolfson Institute for Biomedical Research, University College London, London WC1E 6BT, UK
| | - Arnd Roth
- Wolfson Institute for Biomedical Research, University College London, London WC1E 6BT, UK
| | - Beverley A Clark
- Wolfson Institute for Biomedical Research, University College London, London WC1E 6BT, UK
| | - Michael Häusser
- Wolfson Institute for Biomedical Research, University College London, London WC1E 6BT, UK.
| |
Collapse
|
142
|
Fitzpatrick MJ, Krizan J, Hsiang JC, Shen N, Kerschensteiner D. A pupillary contrast response in mice and humans: Neural mechanisms and visual functions. Neuron 2024; 112:2404-2422.e9. [PMID: 38697114 PMCID: PMC11257825 DOI: 10.1016/j.neuron.2024.04.012] [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: 04/03/2023] [Revised: 12/21/2023] [Accepted: 04/10/2024] [Indexed: 05/04/2024]
Abstract
In the pupillary light response (PLR), increases in ambient light constrict the pupil to dampen increases in retinal illuminance. Here, we report that the pupillary reflex arc implements a second input-output transformation; it senses temporal contrast to enhance spatial contrast in the retinal image and increase visual acuity. The pupillary contrast response (PCoR) is driven by rod photoreceptors via type 6 bipolar cells and M1 ganglion cells. Temporal contrast is transformed into sustained pupil constriction by the M1's conversion of excitatory input into spike output. Computational modeling explains how the PCoR shapes retinal images. Pupil constriction improves acuity in gaze stabilization and predation in mice. Humans exhibit a PCoR with similar tuning properties to mice, which interacts with eye movements to optimize the statistics of the visual input for retinal encoding. Thus, we uncover a conserved component of active vision, its cell-type-specific pathway, computational mechanisms, and optical and behavioral significance.
Collapse
Affiliation(s)
- Michael J Fitzpatrick
- Department of Ophthalmology and Visual Sciences, Washington University School of Medicine in St. Louis, St. Louis, MO 63110, USA; Graduate Program in Neuroscience, Washington University School of Medicine in St. Louis, St. Louis, MO 63110, USA; Medical Scientist Training Program, Washington University School of Medicine in St. Louis, St. Louis, MO 63110, USA
| | - Jenna Krizan
- Department of Ophthalmology and Visual Sciences, Washington University School of Medicine in St. Louis, St. Louis, MO 63110, USA; Graduate Program in Neuroscience, Washington University School of Medicine in St. Louis, St. Louis, MO 63110, USA
| | - Jen-Chun Hsiang
- Department of Ophthalmology and Visual Sciences, Washington University School of Medicine in St. Louis, St. Louis, MO 63110, USA
| | - Ning Shen
- Department of Ophthalmology and Visual Sciences, Washington University School of Medicine in St. Louis, St. Louis, MO 63110, USA
| | - Daniel Kerschensteiner
- Department of Ophthalmology and Visual Sciences, Washington University School of Medicine in St. Louis, St. Louis, MO 63110, USA; Department of Neuroscience, Washington University School of Medicine in St. Louis, St. Louis, MO 63110, USA; Department of Biomedical Engineering, Washington University School of Medicine in St. Louis, St. Louis, MO 63110, USA.
| |
Collapse
|
143
|
Leighton AH, Cheyne JE, Lohmann C. Clustered synapses develop in distinct dendritic domains in visual cortex before eye opening. eLife 2024; 12:RP93498. [PMID: 38990761 PMCID: PMC11239177 DOI: 10.7554/elife.93498] [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] [Indexed: 07/13/2024] Open
Abstract
Synaptic inputs to cortical neurons are highly structured in adult sensory systems, such that neighboring synapses along dendrites are activated by similar stimuli. This organization of synaptic inputs, called synaptic clustering, is required for high-fidelity signal processing, and clustered synapses can already be observed before eye opening. However, how clustered inputs emerge during development is unknown. Here, we employed concurrent in vivo whole-cell patch-clamp and dendritic calcium imaging to map spontaneous synaptic inputs to dendrites of layer 2/3 neurons in the mouse primary visual cortex during the second postnatal week until eye opening. We found that the number of functional synapses and the frequency of transmission events increase several fold during this developmental period. At the beginning of the second postnatal week, synapses assemble specifically in confined dendritic segments, whereas other segments are devoid of synapses. By the end of the second postnatal week, just before eye opening, dendrites are almost entirely covered by domains of co-active synapses. Finally, co-activity with their neighbor synapses correlates with synaptic stabilization and potentiation. Thus, clustered synapses form in distinct functional domains presumably to equip dendrites with computational modules for high-capacity sensory processing when the eyes open.
Collapse
Affiliation(s)
- Alexandra H Leighton
- Department of Synapse and Network Development, Netherlands Institute for NeuroscienceAmsterdamNetherlands
| | - Juliette E Cheyne
- Department of Synapse and Network Development, Netherlands Institute for NeuroscienceAmsterdamNetherlands
| | - Christian Lohmann
- Department of Synapse and Network Development, Netherlands Institute for NeuroscienceAmsterdamNetherlands
- Department of Functional Genomics, Center for Neurogenomics and Cognitive Research, VU University AmsterdamAmsterdamNetherlands
| |
Collapse
|
144
|
Manning A, Bender PTR, Boyd-Pratt H, Mendelson BZ, Hruska M, Anderson CT. Trans-synaptic Association of Vesicular Zinc Transporter 3 and Shank3 Supports Synapse-Specific Dendritic Spine Structure and Function in the Mouse Auditory Cortex. J Neurosci 2024; 44:e0619242024. [PMID: 38830758 PMCID: PMC11236586 DOI: 10.1523/jneurosci.0619-24.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 05/17/2024] [Accepted: 05/23/2024] [Indexed: 06/05/2024] Open
Abstract
Shank3 is a synaptic scaffolding protein that assists in tethering and organizing structural proteins and glutamatergic receptors in the postsynaptic density of excitatory synapses. The localization of Shank3 at excitatory synapses and the formation of stable Shank3 complexes is regulated by the binding of zinc to the C-terminal sterile-alpha-motif (SAM) domain of Shank3. Mutations in the SAM domain of Shank3 result in altered synaptic function and morphology, and disruption of zinc in synapses that express Shank3 leads to a reduction of postsynaptic proteins important for synaptic structure and function. This suggests that zinc supports the localization of postsynaptic proteins via Shank3. Many regions of the brain are highly enriched with free zinc inside glutamatergic vesicles at presynaptic terminals. At these synapses, zinc transporter 3 (ZnT3) moves zinc into vesicles where it is co-released with glutamate. Alterations in ZnT3 are implicated in multiple neurodevelopmental disorders, and ZnT3 knock-out (KO) mice-which lack synaptic zinc-show behavioral deficits associated with autism spectrum disorder and schizophrenia. Here we show that male and female ZnT3 KO mice have smaller dendritic spines and miniature excitatory postsynaptic current amplitudes than wildtype (WT) mice in the auditory cortex. Additionally, spine size deficits in ZnT3 KO mice are restricted to synapses that express Shank3. In WT mice, synapses that express both Shank3 and ZnT3 have larger spines compared to synapses that express Shank3 but not ZnT3. Together these findings suggest a mechanism whereby presynaptic ZnT3-dependent zinc supports postsynaptic structure and function via Shank3 in a synapse-specific manner.
Collapse
Affiliation(s)
- Abbey Manning
- Department of Neuroscience, West Virginia University School of Medicine, Morgantown, WV 26506
- Rockefeller Neuroscience Institute, West Virginia University School of Medicine, Morgantown, WV 26506
| | - Philip T R Bender
- Department of Neuroscience, West Virginia University School of Medicine, Morgantown, WV 26506
- Rockefeller Neuroscience Institute, West Virginia University School of Medicine, Morgantown, WV 26506
| | - Helen Boyd-Pratt
- Department of Neuroscience, West Virginia University School of Medicine, Morgantown, WV 26506
- Rockefeller Neuroscience Institute, West Virginia University School of Medicine, Morgantown, WV 26506
- Clinical and Translational Science Institute, West Virginia University School of Medicine, Morgantown, WV 26506
| | - Benjamin Z Mendelson
- Department of Neuroscience, West Virginia University School of Medicine, Morgantown, WV 26506
- Rockefeller Neuroscience Institute, West Virginia University School of Medicine, Morgantown, WV 26506
| | - Martin Hruska
- Department of Neuroscience, West Virginia University School of Medicine, Morgantown, WV 26506
- Rockefeller Neuroscience Institute, West Virginia University School of Medicine, Morgantown, WV 26506
| | - Charles T Anderson
- Department of Neuroscience, West Virginia University School of Medicine, Morgantown, WV 26506
- Rockefeller Neuroscience Institute, West Virginia University School of Medicine, Morgantown, WV 26506
| |
Collapse
|
145
|
Utashiro N, MacLaren DAA, Liu YC, Yaqubi K, Wojak B, Monyer H. Long-range inhibition from prelimbic to cingulate areas of the medial prefrontal cortex enhances network activity and response execution. Nat Commun 2024; 15:5772. [PMID: 38982042 PMCID: PMC11233578 DOI: 10.1038/s41467-024-50055-z] [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: 06/16/2023] [Accepted: 06/28/2024] [Indexed: 07/11/2024] Open
Abstract
It is well established that the medial prefrontal cortex (mPFC) exerts top-down control of many behaviors, but little is known regarding how cross-talk between distinct areas of the mPFC influences top-down signaling. We performed virus-mediated tracing and functional studies in male mice, homing in on GABAergic projections whose axons are located mainly in layer 1 and that connect two areas of the mPFC, namely the prelimbic area (PrL) with the cingulate area 1 and 2 (Cg1/2). We revealed the identity of the targeted neurons that comprise two distinct types of layer 1 GABAergic interneurons, namely single-bouquet cells (SBCs) and neurogliaform cells (NGFs), and propose that this connectivity links GABAergic projection neurons with cortical canonical circuits. In vitro electrophysiological and in vivo calcium imaging studies support the notion that the GABAergic projection neurons from the PrL to the Cg1/2 exert a crucial role in regulating the activity in the target area by disinhibiting layer 5 output neurons. Finally, we demonstrated that recruitment of these projections affects impulsivity and mechanical responsiveness, behaviors which are known to be modulated by Cg1/2 activity.
Collapse
Affiliation(s)
- Nao Utashiro
- Department of Clinical Neurobiology at the Medical Faculty of the Heidelberg University and of the German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Duncan Archibald Allan MacLaren
- Department of Clinical Neurobiology at the Medical Faculty of the Heidelberg University and of the German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Yu-Chao Liu
- Department of Clinical Neurobiology at the Medical Faculty of the Heidelberg University and of the German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Kaneschka Yaqubi
- Department of Clinical Neurobiology at the Medical Faculty of the Heidelberg University and of the German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Gastroenterology, Hepatology and Infectious Diseases, University Hospital Düsseldorf and Medical Faculty of Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Birgit Wojak
- Department of Clinical Neurobiology at the Medical Faculty of the Heidelberg University and of the German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Internal Medicine III, University Hospital Ulm, Ulm, Germany
| | - Hannah Monyer
- Department of Clinical Neurobiology at the Medical Faculty of the Heidelberg University and of the German Cancer Research Center (DKFZ), Heidelberg, Germany.
| |
Collapse
|
146
|
Subach OM, Piatkevich KD, Subach FV. NeMeHg, genetically encoded indicator for mercury ions based on mNeonGreen green fluorescent protein and merP protein from Shigella flexneri. Front Bioeng Biotechnol 2024; 12:1407874. [PMID: 39050684 PMCID: PMC11266101 DOI: 10.3389/fbioe.2024.1407874] [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: 03/27/2024] [Accepted: 05/29/2024] [Indexed: 07/27/2024] Open
Abstract
The detection of mercury ions is an important task in both environmental monitoring and cell biology research. However, existing genetically encoded sensors for mercury ions have certain limitations, such as negative fluorescence response, narrow dynamic range, or the need for cofactor supplementation. To address these limitations, we have developed novel sensors by fusing a circularly permutated version of the mNeonGreen green fluorescent protein with the merP mercury-binding protein from Gram-negative bacteria Shigella flexneri. The developed NeMeHg and iNeMeHg sensors responded to mercury ions with positive and negative fluorescence changes, respectively. We characterized their properties in vitro. Using the developed biosensors, we were able to successfully visualize changes in mercury ion concentration in mammalian cultured cells.
Collapse
Affiliation(s)
- Oksana M. Subach
- Complex of NBICS Technologies, National Research Center, Kurchatov Institute, Moscow, Russia
| | - Kiryl D. Piatkevich
- School of Life Sciences, Westlake University, Hangzhou, China
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, China
- Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou, China
| | - Fedor V. Subach
- Complex of NBICS Technologies, National Research Center, Kurchatov Institute, Moscow, Russia
| |
Collapse
|
147
|
Johnsen KA, Cruzado NA, Menard ZC, Willats AA, Charles AS, Markowitz JE, Rozell CJ. Bridging model and experiment in systems neuroscience with Cleo: the Closed-Loop, Electrophysiology, and Optophysiology simulation testbed. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.01.27.525963. [PMID: 39026717 PMCID: PMC11257437 DOI: 10.1101/2023.01.27.525963] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/20/2024]
Abstract
Systems neuroscience has experienced an explosion of new tools for reading and writing neural activity, enabling exciting new experiments such as all-optical or closed-loop control that effect powerful causal interventions. At the same time, improved computational models are capable of reproducing behavior and neural activity with increasing fidelity. Unfortunately, these advances have drastically increased the complexity of integrating different lines of research, resulting in the missed opportunities and untapped potential of suboptimal experiments. Experiment simulation can help bridge this gap, allowing model and experiment to better inform each other by providing a low-cost testbed for experiment design, model validation, and methods engineering. Specifically, this can be achieved by incorporating the simulation of the experimental interface into our models, but no existing tool integrates optogenetics, two-photon calcium imaging, electrode recording, and flexible closed-loop processing with neural population simulations. To address this need, we have developed Cleo: the Closed-Loop, Electrophysiology, and Optophysiology experiment simulation testbed. Cleo is a Python package enabling injection of recording and stimulation devices as well as closed-loop control with realistic latency into a Brian spiking neural network model. It is the only publicly available tool currently supporting two-photon and multi-opsin/wavelength optogenetics. To facilitate adoption and extension by the community, Cleo is open-source, modular, tested, and documented, and can export results to various data formats. Here we describe the design and features of Cleo, validate output of individual components and integrated experiments, and demonstrate its utility for advancing optogenetic techniques in prospective experiments using previously published systems neuroscience models.
Collapse
Affiliation(s)
- Kyle A. Johnsen
- Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | | | - Zachary C. Menard
- Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Adam A. Willats
- Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Adam S. Charles
- Department of Biomedical Engineering, The Johns Hopkins University, Baltimore, MD, USA
| | - Jeffrey E. Markowitz
- Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | | |
Collapse
|
148
|
Stempel AV, Evans DA, Arocas OP, Claudi F, Lenzi SC, Kutsarova E, Margrie TW, Branco T. Tonically active GABAergic neurons in the dorsal periaqueductal gray control instinctive escape in mice. Curr Biol 2024; 34:3031-3039.e7. [PMID: 38936364 DOI: 10.1016/j.cub.2024.05.068] [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: 04/17/2024] [Revised: 05/24/2024] [Accepted: 05/29/2024] [Indexed: 06/29/2024]
Abstract
Escape behavior is a set of locomotor actions that move an animal away from threat. While these actions can be stereotyped, it is advantageous for survival that they are flexible.1,2,3 For example, escape probability depends on predation risk and competing motivations,4,5,6,7,8,9,10,11 and flight to safety requires continuous adjustments of trajectory and must terminate at the appropriate place and time.12,13,14,15,16 This degree of flexibility suggests that modulatory components, like inhibitory networks, act on the neural circuits controlling instinctive escape.17,18,19,20,21,22 In mice, the decision to escape from imminent threats is implemented by a feedforward circuit in the midbrain, where excitatory vesicular glutamate transporter 2-positive (VGluT2+) neurons in the dorsal periaqueductal gray (dPAG) compute escape initiation and escape vigor.23,24,25 Here we tested the hypothesis that local GABAergic neurons within the dPAG control escape behavior by setting the excitability of the dPAG escape network. Using in vitro patch-clamp and in vivo neural activity recordings, we found that vesicular GABA transporter-positive (VGAT+) dPAG neurons fire action potentials tonically in the absence of synaptic inputs and are a major source of inhibition to VGluT2+ dPAG neurons. Activity in VGAT+ dPAG cells transiently decreases at escape onset and increases during escape, peaking at escape termination. Optogenetically increasing or decreasing VGAT+ dPAG activity changes the probability of escape when the stimulation is delivered at threat onset and the duration of escape when delivered after escape initiation. We conclude that the activity of tonically firing VGAT+ dPAG neurons sets a threshold for escape initiation and controls the execution of the flight action.
Collapse
Affiliation(s)
- A Vanessa Stempel
- UCL Sainsbury Wellcome Centre for Neural Circuits and Behaviour, 25 Howland St, London W1T 4JG, UK; Max Planck Institute for Brain Research, Max-von-Laue-Str. 4, 60438 Frankfurt am Main, Germany.
| | - Dominic A Evans
- UCL Sainsbury Wellcome Centre for Neural Circuits and Behaviour, 25 Howland St, London W1T 4JG, UK; Max Planck Institute for Brain Research, Max-von-Laue-Str. 4, 60438 Frankfurt am Main, Germany
| | - Oriol Pavón Arocas
- UCL Sainsbury Wellcome Centre for Neural Circuits and Behaviour, 25 Howland St, London W1T 4JG, UK
| | - Federico Claudi
- UCL Sainsbury Wellcome Centre for Neural Circuits and Behaviour, 25 Howland St, London W1T 4JG, UK
| | - Stephen C Lenzi
- UCL Sainsbury Wellcome Centre for Neural Circuits and Behaviour, 25 Howland St, London W1T 4JG, UK
| | - Elena Kutsarova
- Max Planck Institute for Brain Research, Max-von-Laue-Str. 4, 60438 Frankfurt am Main, Germany
| | - Troy W Margrie
- UCL Sainsbury Wellcome Centre for Neural Circuits and Behaviour, 25 Howland St, London W1T 4JG, UK
| | - Tiago Branco
- UCL Sainsbury Wellcome Centre for Neural Circuits and Behaviour, 25 Howland St, London W1T 4JG, UK.
| |
Collapse
|
149
|
Sharma AK, Randi F, Kumar S, Dvali S, Leifer AM. TWISP: a transgenic worm for interrogating signal propagation in Caenorhabditis elegans. Genetics 2024; 227:iyae077. [PMID: 38733622 PMCID: PMC11228852 DOI: 10.1093/genetics/iyae077] [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: 02/11/2024] [Revised: 02/11/2024] [Accepted: 04/11/2024] [Indexed: 05/13/2024] Open
Abstract
Genetically encoded optical indicators and actuators of neural activity allow for all-optical investigations of signaling in the nervous system. But commonly used indicators, actuators, and expression strategies are poorly suited for systematic measurements of signal propagation at brain scale and cellular resolution. Large-scale measurements of the brain require indicators and actuators with compatible excitation spectra to avoid optical crosstalk. They must be highly expressed in every neuron but at the same time avoid lethality and permit the animal to reach adulthood. Their expression must also be compatible with additional fluorescent labels to locate and identify neurons, such as those in the NeuroPAL cell identification system. We present TWISP, a transgenic worm for interrogating signal propagation, that addresses these needs and enables optical measurements of evoked calcium activity at brain scale and cellular resolution in the nervous system of the nematode Caenorhabditis elegans. In every neuron we express a nonconventional optical actuator, the gustatory receptor homolog GUR-3 + PRDX-2, under the control of a drug-inducible system QF + hGR, and a calcium indicator GCAMP6s, in a background with additional fluorophores from the NeuroPAL cell ID system. We show that this combination, but not others tested, avoids optical crosstalk, creates strong expression in the adult, and generates stable transgenic lines for systematic measurements of signal propagation in the worm brain.
Collapse
Affiliation(s)
- Anuj Kumar Sharma
- Department of Physics, Princeton University, Princeton, NJ 08544, USA
| | - Francesco Randi
- Department of Physics, Princeton University, Princeton, NJ 08544, USA
| | - Sandeep Kumar
- Princeton Neuroscience Institute, Princeton University, Princeton, NJ 08544, USA
| | - Sophie Dvali
- Department of Physics, Princeton University, Princeton, NJ 08544, USA
| | - Andrew M Leifer
- Department of Physics, Princeton University, Princeton, NJ 08544, USA
- Princeton Neuroscience Institute, Princeton University, Princeton, NJ 08544, USA
| |
Collapse
|
150
|
Obeng B, Bennett LJ, West BE, Wagner DJ, Fleming PJ, Tasker MN, Lorenger MK, Smith DR, Systuk T, Plummer SM, Eom J, Paine MD, Frangos CT, Wilczek MP, Shim JK, Maginnis MS, Gosse JA. Antimicrobial cetylpyridinium chloride suppresses mast cell function by targeting tyrosine phosphorylation of Syk kinase. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.04.602096. [PMID: 39026716 PMCID: PMC11257455 DOI: 10.1101/2024.07.04.602096] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/20/2024]
Abstract
Cetylpyridinium chloride (CPC) is a quaternary ammonium antimicrobial used in numerous personal care products, human food, cosmetic products, and cleaning solutions. Yet, there is minimal published data on CPC effects on eukaryotes, immune signaling, and human health. Previously, we showed that low-micromolar CPC inhibits rat mast cell function by inhibiting antigen (Ag)-stimulated Ca 2+ mobilization, microtubule polymerization, and degranulation. In this study, we extend the findings to human mast cells (LAD2) and present data indicating that CPC's mechanism of action centers on its positively-charged quaternary nitrogen in its pyridinium headgroup. CPC's inhibitory effect is independent of signaling platform receptor architecture. Tyrosine phosphorylation events are a trigger of Ca 2+ mobilization necessary for degranulation. CPC inhibits global tyrosine phosphorylation in Ag-stimulated mast cells. Specifically, CPC inhibits tyrosine phosphorylation of specific key players Syk kinase and LAT, a substrate of Syk. In contrast, CPC does not affect Lyn kinase phosphorylation. Thus, CPC's root mechanism is electrostatic disruption of particular tyrosine phosphorylation events essential for signaling. This work outlines the biochemical mechanisms underlying the effects of CPC on immune signaling and allows the prediction of CPC effects on cell types, like T cells, that share similar signaling elements.
Collapse
|