1
|
Cheng J, Koch ET, Ramandi D, Mackay JP, O'Leary TP, Rees-Jones W, Raymond LA. Synaptic modulation of glutamate in striatum of the YAC128 mouse model of Huntington disease. Neurobiol Dis 2024:106774. [PMID: 39716682 DOI: 10.1016/j.nbd.2024.106774] [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: 10/02/2024] [Revised: 12/08/2024] [Accepted: 12/18/2024] [Indexed: 12/25/2024] Open
Abstract
BACKGROUND Altered balance between striatal direct and indirect pathways contributes to early motor, cognitive and psychiatric symptoms in Huntington disease (HD). While degeneration of striatal D2-type dopamine receptor (D2)-expressing indirect pathway medium spiny neurons (iMSNs) occurs prior to that of D1-type dopamine receptor (D1)-expressing direct pathway neurons, altered corticostriatal synaptic function precedes degeneration. D2-mediated signaling on iMSNs reduces their excitability and promotes endocannabinoid (eCB) synthesis, suppressing glutamate release from cortical afferents. D2 receptors are also expressed on glutamatergic cortical terminals, cholinergic interneurons, and dopaminergic terminals from substantia nigra where they suppress release of glutamate, acetylcholine and dopamine, respectively, and these cell types may contribute to early striatal dysfunction in HD. Thus, we used corticostriatal brain slices and optogenetic probes to directly investigateneuromodulatory signaling in the transgenic YAC128 HD mouse model. RESULTS Low-dose D2 agonist quinpirole reduced cortically-evoked glutamate release in dorsal striatum of premanifest YAC128 slices but not WT, and blocking type 1 cannabinoid receptors mitigated this effect. YAC128 corticostriatal brain slices also showed increased evoked dopamine and reduced evoked eCB release compared to WT, while acetylcholine signaling patterns remained relatively intact. CONCLUSIONS These findings suggest that YAC128 corticostriatal slices show increased D2 sensitivity that is eCB-dependent, and that dopamine and eCB release are altered at an early disease stage. We provide evidence for impaired neuromodulatory signaling in early HD, guiding therapeutic efforts prior to the onset of overt motor symptoms later on.
Collapse
Affiliation(s)
- Judy Cheng
- Department of Psychiatry, Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada; Graduate Program in Neuroscience, University of British Columbia, Vancouver, BC, Canada
| | - Ellen T Koch
- Department of Psychiatry, Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada; Graduate Program in Neuroscience, University of British Columbia, Vancouver, BC, Canada
| | - Daniel Ramandi
- Department of Psychiatry, Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada; Graduate Program in Cell and Developmental Biology, University of British Columbia, Vancouver, BC, Canada
| | - James P Mackay
- Department of Psychiatry, Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada
| | - Timothy P O'Leary
- Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, BC, Canada
| | - William Rees-Jones
- Department of Psychiatry, Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada
| | - Lynn A Raymond
- Department of Psychiatry, Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada.
| |
Collapse
|
2
|
Gonzalez KC, Negrean A, Liao Z, Terada S, Zhang G, Lee S, Ócsai K, Rózsa BJ, Lin MZ, Polleux F, Losonczy A. Synaptic basis of feature selectivity in hippocampal neurons. Nature 2024:10.1038/s41586-024-08325-9. [PMID: 39695232 DOI: 10.1038/s41586-024-08325-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Accepted: 10/31/2024] [Indexed: 12/20/2024]
Abstract
A central question in neuroscience is how synaptic plasticity shapes the feature selectivity of neurons in behaving animals1. Hippocampal CA1 pyramidal neurons display one of the most striking forms of feature selectivity by forming spatially and contextually selective receptive fields called place fields, which serve as a model for studying the synaptic basis of learning and memory. Various forms of synaptic plasticity have been proposed as cellular substrates for the emergence of place fields. However, despite decades of work, our understanding of how synaptic plasticity underlies place-field formation and memory encoding remains limited, largely due to a shortage of tools and technical challenges associated with the visualization of synaptic plasticity at the single-neuron resolution in awake behaving animals. To address this, we developed an all-optical approach to monitor the spatiotemporal tuning and synaptic weight changes of dendritic spines before and after the induction of a place field in single CA1 pyramidal neurons during spatial navigation. We identified a temporally asymmetric synaptic plasticity kernel resulting from bidirectional modifications of synaptic weights around the induction of a place field. Our work identified compartment-specific differences in the magnitude and temporal expression of synaptic plasticity between basal dendrites and oblique dendrites. Our results provide experimental evidence linking synaptic plasticity to the rapid emergence of spatial selectivity in hippocampal neurons, a critical prerequisite for episodic memory.
Collapse
Affiliation(s)
- Kevin C Gonzalez
- Department of Neuroscience, Columbia University, New York, NY, USA
- Mortimer B. Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY, USA
| | - Adrian Negrean
- Department of Neuroscience, Columbia University, New York, NY, USA
- Mortimer B. Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY, USA
- Allen Brain Institute, Seattle, WA, USA
| | - Zhenrui Liao
- Department of Neuroscience, Columbia University, New York, NY, USA
- Mortimer B. Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY, USA
| | - Satoshi Terada
- Department of Neuroscience, Columbia University, New York, NY, USA
- Mortimer B. Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY, USA
| | - Guofeng Zhang
- Department of Neurobiology, Stanford University, Stanford, CA, USA
- Department of Neurosurgery, The First Affiliated Hospital, Sun Yat-sen University, Guangdong, China
| | - Sungmoo Lee
- Department of Neurobiology, Stanford University, Stanford, CA, USA
| | - Katalin Ócsai
- BrainVisionCenter, Budapest, Hungary
- Department of Algebra and Geometry, Institute of Mathematics, Budapest University of Technology and Economics, Budapest, Hungary
| | - Balázs J Rózsa
- BrainVisionCenter, Budapest, Hungary
- Laboratory of 3D Functional Network and Dendritic Imaging, Institute of Experimental Medicine, Budapest, Hungary
- Faculty of Information Technology and Bionics, Pázmány Péter University, Budapest, Hungary
| | - Michael Z Lin
- Department of Neurobiology, Stanford University, Stanford, CA, USA
- Department of Bioengineering, Stanford University, Stanford, CA, USA
| | - Franck Polleux
- Department of Neuroscience, Columbia University, New York, NY, USA.
- Mortimer B. Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY, USA.
| | - Attila Losonczy
- Department of Neuroscience, Columbia University, New York, NY, USA.
- Mortimer B. Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY, USA.
| |
Collapse
|
3
|
Kang I, Kim H, Natan R, Zhang Q, Yu SX, Ji N. Adaptive optical correction for in vivo two-photon fluorescence microscopy with neural fields. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.20.619284. [PMID: 39484436 PMCID: PMC11527033 DOI: 10.1101/2024.10.20.619284] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/03/2024]
Abstract
Adaptive optics (AO) restore ideal imaging performance in complex samples by measuring and correcting optical aberrations, but often require custom-built microscopes with carefully aligned wavefront sensing/shaping devices and can be susceptible to sample motion. Here we describe NeAT, a computational framework using neural fields for AO two-photon fluorescence microscopy. NeAT estimates wavefront aberration and recovers sample structure from a 3D image stack without requiring external datasets for training. Incorporating motion correction in learning and correcting conjugation errors commonly found in commercial microscopes, NeAT is designed for deployment in biological laboratories for in vivo imaging. We validate NeAT's performance using a custom-built microscope with a wavefront sensor under varying signal-to-noise ratios, aberration, and motion conditions. With a commercial microscope, we demonstrate real-time aberration correction for in vivo morphological and functional imaging in the living mouse brain, with NeAT improving signal and accuracy of glutamate and calcium imaging of synapses and neurons.
Collapse
|
4
|
Huang H, Lu W, Luo R, Zeng Y, Zhang Y, Su X, Zhang X, Tian B, Wang X. Astrocytic RARγ mediates hippocampal astrocytosis and neurogenesis deficits in chronic retinoic acid-induced depression. Neuropsychopharmacology 2024; 50:419-431. [PMID: 39242924 PMCID: PMC11632084 DOI: 10.1038/s41386-024-01983-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Revised: 08/10/2024] [Accepted: 08/28/2024] [Indexed: 09/09/2024]
Abstract
Accumulating clinical evidence indicates that chronic exposure to retinoic acid (RA) may lead to depressive symptoms and even increase the risk of suicidal behavior, which severely limits the clinical long-term application of RA. The exact mechanisms through which RA contributes to the onset of depression remain largely unclear. Here, we administered intraperitoneal injections of all-trans RA to male C57BL/6 J mice over a period of 21 days. Mice subjected to chronic RA exposure displayed depressive-like behaviors, accompanied by impaired hippocampal neurogenesis and heightened RA receptor gamma (RARγ) levels in the ventral hippocampus (vHip). The administration of an RARγ antagonist effectively mitigated these RA-induced neurogenesis impairments and depressive-like behaviors. Chronic exposure to RA was also observed to promote hippocampal astrocytosis and increase astrocytic Rarγ expression in the ventral dentate gyrus (vDG) of hippocampus. Notably, astrocytic RARγ in the vDG was found to be a key factor in the observed hippocampal astrocytosis and neurogenesis impairments, and depressive-like behaviors. Chronic exposure to RA resulted in increased extracellular glutamate levels in neural stem cells (NSCs), accompanied by a decrease in glutamate transporter 1 (GLT-1) expression. Enhancing astrocytic GLT-1 expression was found to alleviate both hippocampal astrocytosis and depressive-like behaviors caused by RA. These findings underscore the critical role of astrocytic RARγ-GLT-1 axis in the development of hippocampal astrocytosis, neurogenesis impairments, and depressive symptoms, suggesting that targeting RARγ-GLT-1 could potentially offer an effective therapeutic approach for depression.
Collapse
Affiliation(s)
- Huixian Huang
- Key Laboratory of Mental Health of the Ministry of Education, Guangdong-Hong Kong-Macao Greater Bay Area Centre 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, Guangdong, China
| | - Wensi Lu
- Key Laboratory of Mental Health of the Ministry of Education, Guangdong-Hong Kong-Macao Greater Bay Area Centre 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, Guangdong, China
| | - Ran Luo
- Key Laboratory of Mental Health of the Ministry of Education, Guangdong-Hong Kong-Macao Greater Bay Area Centre 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, Guangdong, China
| | - Yinyun Zeng
- Key Laboratory of Mental Health of the Ministry of Education, Guangdong-Hong Kong-Macao Greater Bay Area Centre 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, Guangdong, China
| | - Yuqin Zhang
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Xiaohong Su
- Key Laboratory of Mental Health of the Ministry of Education, Guangdong-Hong Kong-Macao Greater Bay Area Centre 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, Guangdong, China
| | - Xinyi Zhang
- Key Laboratory of Mental Health of the Ministry of Education, Guangdong-Hong Kong-Macao Greater Bay Area Centre 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, Guangdong, China
| | - Bo Tian
- Department of Neurobiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China.
| | - Xuemin Wang
- Key Laboratory of Mental Health of the Ministry of Education, Guangdong-Hong Kong-Macao Greater Bay Area Centre 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, Guangdong, China.
| |
Collapse
|
5
|
Gest AM, Sahan AZ, Zhong Y, Lin W, Mehta S, Zhang J. Molecular Spies in Action: Genetically Encoded Fluorescent Biosensors Light up Cellular Signals. Chem Rev 2024; 124:12573-12660. [PMID: 39535501 PMCID: PMC11613326 DOI: 10.1021/acs.chemrev.4c00293] [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: 04/17/2024] [Revised: 09/07/2024] [Accepted: 09/20/2024] [Indexed: 11/16/2024]
Abstract
Cellular function is controlled through intricate networks of signals, which lead to the myriad pathways governing cell fate. Fluorescent biosensors have enabled the study of these signaling pathways in living systems across temporal and spatial scales. Over the years there has been an explosion in the number of fluorescent biosensors, as they have become available for numerous targets, utilized across spectral space, and suited for various imaging techniques. To guide users through this extensive biosensor landscape, we discuss critical aspects of fluorescent proteins for consideration in biosensor development, smart tagging strategies, and the historical and recent biosensors of various types, grouped by target, and with a focus on the design and recent applications of these sensors in living systems.
Collapse
Affiliation(s)
- Anneliese
M. M. Gest
- Department
of Pharmacology, University of California,
San Diego, La Jolla, California 92093, United States
| | - Ayse Z. Sahan
- Department
of Pharmacology, University of California,
San Diego, La Jolla, California 92093, United States
- Biomedical
Sciences Graduate Program, University of
California, San Diego, La Jolla, California 92093, United States
| | - Yanghao Zhong
- Department
of Pharmacology, University of California,
San Diego, La Jolla, California 92093, United States
| | - Wei Lin
- Department
of Pharmacology, University of California,
San Diego, La Jolla, California 92093, United States
| | - Sohum Mehta
- Department
of Pharmacology, University of California,
San Diego, La Jolla, California 92093, United States
| | - Jin Zhang
- Department
of Pharmacology, University of California,
San Diego, La Jolla, California 92093, United States
- Shu
Chien-Gene Lay Department of Bioengineering, University of California, San Diego, La Jolla, California 92093, United States
- Department
of Chemistry and Biochemistry, University
of California, San Diego, La Jolla, California 92093, United States
| |
Collapse
|
6
|
Li H, Zhao Y, Dai R, Geng P, Weng D, Wu W, Yu F, Lin R, Wu Z, Li Y, Luo M. Astrocytes release ATP/ADP and glutamate in flashes via vesicular exocytosis. Mol Psychiatry 2024:10.1038/s41380-024-02851-8. [PMID: 39578520 DOI: 10.1038/s41380-024-02851-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Revised: 11/12/2024] [Accepted: 11/14/2024] [Indexed: 11/24/2024]
Abstract
Astrocytes regulate brain functions through gliotransmitters like ATP/ADP and glutamate, but their release patterns and mechanisms remain controversial. Here, we visualized ATP/ADP and glutamate response following astrocyte activation and investigated their mechanisms in vivo. Employing cOpn5-mediated optogenetic stimulation, genetically encoded fluorescent sensors, and two-photon imaging, we observed ATP/ADP released as temporally prolonged and spatially extended flashes that later converted to adenosine. This release occurs via Ca2+ and VNUT-dependent vesicular exocytosis. Additionally, astrocytes also release glutamate in flashes through TeNT-sensitive exocytosis, independent of ATP/ADP release. ATP/ADP released by astrocytes triggers further ATP/ADP release from microglia through P2Y12- and VNUT-dependent mechanisms. VNUT in astrocytes and microglia also contributes to ATP/ADP release under LPS-induced brain inflammation. These findings establish Ca2+-dependent vesicular exocytosis as a key mode of action, reveal intricate astrocyte-microglia interactions, and suggest a role for gliotransmission in brain inflammation. Furthermore, the methodologies may provide valuable tools for deciphering glial physiology and pathophysiology.
Collapse
Affiliation(s)
- Heng Li
- National Institute of Biological Sciences (NIBS), Beijing, 102206, Beijing, China
- School of Life Sciences, Tsinghua University, 100084, Beijing, China
- Tsinghua Institute of Multidisciplinary Biomedical Research (TIMBR), 102206, Beijing, China
| | - Yuqing Zhao
- National Institute of Biological Sciences (NIBS), Beijing, 102206, Beijing, China
| | - Ruicheng Dai
- Chinese Institute for Brain Research (CIBR), Beijing, 102206, Beijing, China
| | - Peiyao Geng
- National Institute of Biological Sciences (NIBS), Beijing, 102206, Beijing, China
- Tsinghua Institute of Multidisciplinary Biomedical Research (TIMBR), 102206, Beijing, China
| | - Danwei Weng
- National Institute of Biological Sciences (NIBS), Beijing, 102206, Beijing, China
| | - Wenting Wu
- National Institute of Biological Sciences (NIBS), Beijing, 102206, Beijing, China
- Tsinghua Institute of Multidisciplinary Biomedical Research (TIMBR), 102206, Beijing, China
| | - Fengting Yu
- Chinese Institute for Brain Research (CIBR), Beijing, 102206, Beijing, China
| | - Rui Lin
- National Institute of Biological Sciences (NIBS), Beijing, 102206, Beijing, China
- Tsinghua Institute of Multidisciplinary Biomedical Research (TIMBR), 102206, Beijing, China
| | - Zhaofa Wu
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, 100101, Beijing, China
| | - Yulong Li
- State Key Laboratory of Membrane Biology, School of Life Sciences, Peking University, 100871, Beijing, China
- New Cornerstone Science Laboratory, 518054, Shenzhen, China
| | - Minmin Luo
- Chinese Institute for Brain Research (CIBR), Beijing, 102206, Beijing, China.
- New Cornerstone Science Laboratory, 518054, Shenzhen, China.
- Research Unit of Medical Neurobiology, Chinese Academy of Medical Sciences, 100005, Beijing, China.
- Beijing Institute for Brain Research, Chinese Academy of Medical Sciences & Peking Union Medical College, 102206, Beijing, China.
| |
Collapse
|
7
|
Shi D, Yang Y, Tong L, Zhang L, Yang F, Tao J, Zhao M. A Novel Benzothiazole-Based Fluorescent AIE Probe for the Detection of Hydrogen Peroxide in Living Cells. Molecules 2024; 29:5181. [PMID: 39519822 PMCID: PMC11547549 DOI: 10.3390/molecules29215181] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Revised: 10/25/2024] [Accepted: 10/31/2024] [Indexed: 11/16/2024] Open
Abstract
A benzothiazole-based derivative aggregation-induced emission (AIE) fluorescent 'turn-on' probe named 2-(2-((4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)benzyl)oxy)phenyl)benzo[d]thiazole (probe BT-BO) was developed and synthesized successfully for detecting hydrogen peroxide (H2O2) in living cells. The synthesis method of probe BT-BO is facile. Probe BT-BO demonstrates a well-resolved emission peak at 604 nm and the ability to prevent the interference of reactive oxygen species (ROS), various metal ions and anion ions, and good sensitivity. Additionally, the probe boasts impressive pH range versatility, a fast response time to H2O2 and low cytotoxicity. Finally, probe BT-BO was applied successfully to image A549 and Hep G2 cells to monitor both exogenous and endogenous H2O2.
Collapse
Affiliation(s)
- Dezhi Shi
- The Cultivation Base of Shanxi Key Laboratory of Mining Area Ecological Restoration and Solid Wastes Utilization, Shanxi Institute of Technology, Yangquan 045000, China; (D.S.); (F.Y.); (J.T.)
- Yangquan Technology Innovation Center of Carbon Dioxide Capture, Utilization and Storage, Shanxi Institute of Technology, Yangquan 045000, China
| | - Yulong Yang
- Department of Veterinary Medicine, Shanxi Agricultural University, Jinzhong 030801, China; (Y.Y.); (L.T.); (L.Z.)
| | - Luan Tong
- Department of Veterinary Medicine, Shanxi Agricultural University, Jinzhong 030801, China; (Y.Y.); (L.T.); (L.Z.)
| | - Likang Zhang
- Department of Veterinary Medicine, Shanxi Agricultural University, Jinzhong 030801, China; (Y.Y.); (L.T.); (L.Z.)
| | - Fengqing Yang
- The Cultivation Base of Shanxi Key Laboratory of Mining Area Ecological Restoration and Solid Wastes Utilization, Shanxi Institute of Technology, Yangquan 045000, China; (D.S.); (F.Y.); (J.T.)
| | - Jiali Tao
- The Cultivation Base of Shanxi Key Laboratory of Mining Area Ecological Restoration and Solid Wastes Utilization, Shanxi Institute of Technology, Yangquan 045000, China; (D.S.); (F.Y.); (J.T.)
| | - Mingxia Zhao
- The Cultivation Base of Shanxi Key Laboratory of Mining Area Ecological Restoration and Solid Wastes Utilization, Shanxi Institute of Technology, Yangquan 045000, China; (D.S.); (F.Y.); (J.T.)
- Yangquan Technology Innovation Center of Carbon Dioxide Capture, Utilization and Storage, Shanxi Institute of Technology, Yangquan 045000, China
| |
Collapse
|
8
|
Morabito A, Zerlau Y, Dhanasobhon D, Berthaux E, Tzilivaki A, Moneron G, Cathala L, Poirazi P, Bacci A, DiGregorio D, Lourenço J, Rebola N. A dendritic substrate for temporal diversity of cortical inhibition. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.09.602783. [PMID: 39026855 PMCID: PMC11257522 DOI: 10.1101/2024.07.09.602783] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/20/2024]
Abstract
In the mammalian neocortex, GABAergic interneurons (INs) inhibit cortical networks in profoundly different ways. The extent to which this depends on how different INs process excitatory signals along their dendrites is poorly understood. Here, we reveal that the functional specialization of two major populations of cortical INs is determined by the unique association of different dendritic integration modes with distinct synaptic organization motifs. We found that somatostatin (SST)-INs exhibit NMDAR-dependent dendritic integration and uniform synapse density along the dendritic tree. In contrast, dendrites of parvalbumin (PV)-INs exhibit passive synaptic integration coupled with proximally enriched synaptic distributions. Theoretical analysis shows that these two dendritic configurations result in different strategies to optimize synaptic efficacy in thin dendritic structures. Yet, the two configurations lead to distinct temporal engagement of each IN during network activity. We confirmed these predictions with in vivo recordings of IN activity in the visual cortex of awake mice, revealing a rapid and linear recruitment of PV-INs as opposed to a long-lasting integrative activation of SST-INs. Our work reveals the existence of distinct dendritic strategies that confer distinct temporal representations for the two major classes of neocortical INs and thus dynamics of inhibition.
Collapse
Affiliation(s)
- Annunziato Morabito
- ICM, Paris Brain Institute, Hôpital de la Pitié-Salpêtrière, Sorbonne Université, Inserm, CNRS, Paris, 75013, France
| | - Yann Zerlau
- ICM, Paris Brain Institute, Hôpital de la Pitié-Salpêtrière, Sorbonne Université, Inserm, CNRS, Paris, 75013, France
| | - Dhanasak Dhanasobhon
- ICM, Paris Brain Institute, Hôpital de la Pitié-Salpêtrière, Sorbonne Université, Inserm, CNRS, Paris, 75013, France
| | - Emmanuelle Berthaux
- ICM, Paris Brain Institute, Hôpital de la Pitié-Salpêtrière, Sorbonne Université, Inserm, CNRS, Paris, 75013, France
| | - Alexandra Tzilivaki
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität zu Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Neuroscience Research Center, 10117 Berlin, Germany
- Einstein Center for Neurosciences, Chariteplatz 1, 10117 Berlin, Germany
- NeuroCure Cluster of Excellence, Chariteplatz 1, 10117 Berlin, Germany
| | - Gael Moneron
- Institut Pasteur, CNRS UMR3571, Synapse and Circuit Dynamics Unit, 75015 Paris, France
| | - Laurence Cathala
- ICM, Paris Brain Institute, Hôpital de la Pitié-Salpêtrière, Sorbonne Université, Inserm, CNRS, Paris, 75013, France
| | - Panayiota Poirazi
- Institute of Molecular Biology and Biotechnology (IMBB), Foundation for Research and Technology Hellas (FORTH), Heraklion, 70013, Greece
| | - Alberto Bacci
- ICM, Paris Brain Institute, Hôpital de la Pitié-Salpêtrière, Sorbonne Université, Inserm, CNRS, Paris, 75013, France
| | - David DiGregorio
- Institut Pasteur, CNRS UMR3571, Synapse and Circuit Dynamics Unit, 75015 Paris, France
| | - Joana Lourenço
- ICM, Paris Brain Institute, Hôpital de la Pitié-Salpêtrière, Sorbonne Université, Inserm, CNRS, Paris, 75013, France
| | - Nelson Rebola
- ICM, Paris Brain Institute, Hôpital de la Pitié-Salpêtrière, Sorbonne Université, Inserm, CNRS, Paris, 75013, France
| |
Collapse
|
9
|
Gür B, Ramirez L, Cornean J, Thurn F, Molina-Obando S, Ramos-Traslosheros G, Silies M. Neural pathways and computations that achieve stable contrast processing tuned to natural scenes. Nat Commun 2024; 15:8580. [PMID: 39362859 PMCID: PMC11450186 DOI: 10.1038/s41467-024-52724-5] [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: 03/28/2024] [Accepted: 09/18/2024] [Indexed: 10/05/2024] Open
Abstract
Natural scenes are highly dynamic, challenging the reliability of visual processing. Yet, humans and many animals perform accurate visual behaviors, whereas computer vision devices struggle with rapidly changing background luminance. How does animal vision achieve this? Here, we reveal the algorithms and mechanisms of rapid luminance gain control in Drosophila, resulting in stable visual processing. We identify specific transmedullary neurons as the site of luminance gain control, which pass this property to direction-selective cells. The circuitry further involves wide-field neurons, matching computational predictions that local spatial pooling drive optimal contrast processing in natural scenes when light conditions change rapidly. Experiments and theory argue that a spatially pooled luminance signal achieves luminance gain control via divisive normalization. This process relies on shunting inhibition using the glutamate-gated chloride channel GluClα. Our work describes how the fly robustly processes visual information in dynamically changing natural scenes, a common challenge of all visual systems.
Collapse
Affiliation(s)
- Burak Gür
- Institute of Developmental Biology and Neurobiology, Johannes-Gutenberg University Mainz, Mainz, Germany
- The Friedrich Miescher Institute for Biomedical Research (FMI), Basel, Switzerland
| | - Luisa Ramirez
- Institute of Developmental Biology and Neurobiology, Johannes-Gutenberg University Mainz, Mainz, Germany
| | - Jacqueline Cornean
- Institute of Developmental Biology and Neurobiology, Johannes-Gutenberg University Mainz, Mainz, Germany
| | - Freya Thurn
- Institute of Developmental Biology and Neurobiology, Johannes-Gutenberg University Mainz, Mainz, Germany
| | - Sebastian Molina-Obando
- Institute of Developmental Biology and Neurobiology, Johannes-Gutenberg University Mainz, Mainz, Germany
| | - Giordano Ramos-Traslosheros
- Institute of Developmental Biology and Neurobiology, Johannes-Gutenberg University Mainz, Mainz, Germany
- Department of Neurobiology, Harvard Medical School, Boston, MA, USA
| | - Marion Silies
- Institute of Developmental Biology and Neurobiology, Johannes-Gutenberg University Mainz, Mainz, Germany.
| |
Collapse
|
10
|
Muir J, Anguiano M, Kim CK. Neuromodulator and neuropeptide sensors and probes for precise circuit interrogation in vivo. Science 2024; 385:eadn6671. [PMID: 39325905 PMCID: PMC11488521 DOI: 10.1126/science.adn6671] [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: 03/27/2024] [Accepted: 07/01/2024] [Indexed: 09/28/2024]
Abstract
To determine how neuronal circuits encode and drive behavior, it is often necessary to measure and manipulate different aspects of neurochemical signaling in awake animals. Optogenetics and calcium sensors have paved the way for these types of studies, allowing for the perturbation and readout of spiking activity within genetically defined cell types. However, these methods lack the ability to further disentangle the roles of individual neuromodulator and neuropeptides on circuits and behavior. We review recent advances in chemical biology tools that enable precise spatiotemporal monitoring and control over individual neuroeffectors and their receptors in vivo. We also highlight discoveries enabled by such tools, revealing how these molecules signal across different timescales to drive learning, orchestrate behavioral changes, and modulate circuit activity.
Collapse
Affiliation(s)
- J. Muir
- Center for Neuroscience, University of California, Davis, Davis, CA 95618, USA
- Department of Neurology, School of Medicine, University of California, Davis, Sacramento, CA 95817, USA
| | - M. Anguiano
- Neuroscience Graduate Group, University of California, Davis, Davis, CA 95616, USA
| | - C. K. Kim
- Center for Neuroscience, University of California, Davis, Davis, CA 95618, USA
- Department of Neurology, School of Medicine, University of California, Davis, Sacramento, CA 95817, USA
| |
Collapse
|
11
|
Nomura S, Terada SI, Ebina T, Uemura M, Masamizu Y, Ohki K, Matsuzaki M. ARViS: a bleed-free multi-site automated injection robot for accurate, fast, and dense delivery of virus to mouse and marmoset cerebral cortex. Nat Commun 2024; 15:7633. [PMID: 39256380 PMCID: PMC11387507 DOI: 10.1038/s41467-024-51986-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Accepted: 08/22/2024] [Indexed: 09/12/2024] Open
Abstract
Genetically encoded fluorescent sensors continue to be developed and improved. If they could be expressed across multiple cortical areas in non-human primates, it would be possible to measure a variety of spatiotemporal dynamics of primate-specific cortical activity. Here, we develop an Automated Robotic Virus injection System (ARViS) for broad expression of a biosensor. ARViS consists of two technologies: image recognition of vasculature structures on the cortical surface to determine multiple injection sites without hitting them, and robotic control of micropipette insertion perpendicular to the cortical surface with 50 μm precision. In mouse cortex, ARViS sequentially injected virus solution into 100 sites over a duration of 100 min with a bleeding probability of only 0.1% per site. Furthermore, ARViS successfully achieved 266-site injections over the frontoparietal cortex of a female common marmoset. We demonstrate one-photon and two-photon calcium imaging in the marmoset frontoparietal cortex, illustrating the effective expression of biosensors delivered by ARViS.
Collapse
Affiliation(s)
- Shinnosuke Nomura
- Department of Physiology, Graduate School of Medicine, The University of Tokyo, Tokyo, 113-0033, Japan
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Ibaraki, 305-8575, Japan
| | - Shin-Ichiro Terada
- Department of Physiology, Graduate School of Medicine, The University of Tokyo, Tokyo, 113-0033, Japan
| | - Teppei Ebina
- Department of Physiology, Graduate School of Medicine, The University of Tokyo, Tokyo, 113-0033, Japan
| | - Masato Uemura
- Department of Physiology, Graduate School of Medicine, The University of Tokyo, Tokyo, 113-0033, Japan
| | - Yoshito Masamizu
- Brain Functional Dynamics Collaboration Laboratory, RIKEN Center for Brain Science, Saitama, 351-0198, Japan
- Laboratory of Functional Brain Circuit Construction, Graduate School of Brain Science, Doshisha University, Kyoto, 610-0394, Japan
| | - Kenichi Ohki
- Department of Physiology, Graduate School of Medicine, The University of Tokyo, Tokyo, 113-0033, Japan
- International Research Center for Neurointelligence (WPI-IRCN), The University of Tokyo Institutes for Advanced Study, Tokyo, 113-0033, Japan
- Institute for AI and Beyond, The University of Tokyo, Tokyo, 113-0033, Japan
| | - Masanori Matsuzaki
- Department of Physiology, Graduate School of Medicine, The University of Tokyo, Tokyo, 113-0033, Japan.
- Brain Functional Dynamics Collaboration Laboratory, RIKEN Center for Brain Science, Saitama, 351-0198, Japan.
- International Research Center for Neurointelligence (WPI-IRCN), The University of Tokyo Institutes for Advanced Study, Tokyo, 113-0033, Japan.
- Department of Biological Sciences, Graduate School of Science, The University of Tokyo, Tokyo, 113-0033, Japan.
| |
Collapse
|
12
|
Saha O, Melo de Farias AR, Pelletier A, Siedlecki-Wullich D, Landeira BS, Gadaut J, Carrier A, Vreulx AC, Guyot K, Shen Y, Bonnefond A, Amouyel P, Tcw J, Kilinc D, Queiroz CM, Delahaye F, Lambert JC, Costa MR. The Alzheimer's disease risk gene BIN1 regulates activity-dependent gene expression in human-induced glutamatergic neurons. Mol Psychiatry 2024; 29:2634-2646. [PMID: 38514804 PMCID: PMC11420064 DOI: 10.1038/s41380-024-02502-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2023] [Revised: 02/22/2024] [Accepted: 02/27/2024] [Indexed: 03/23/2024]
Abstract
Bridging Integrator 1 (BIN1) is the second most important Alzheimer's disease (AD) risk gene, but its physiological roles in neurons and its contribution to brain pathology remain largely elusive. In this work, we show that BIN1 plays a critical role in the regulation of calcium homeostasis, electrical activity, and gene expression of glutamatergic neurons. Using single-cell RNA-sequencing on cerebral organoids generated from isogenic BIN1 wild type (WT), heterozygous (HET) and homozygous knockout (KO) human-induced pluripotent stem cells (hiPSCs), we show that BIN1 is mainly expressed by oligodendrocytes and glutamatergic neurons, like in the human brain. Both BIN1 HET and KO cerebral organoids show specific transcriptional alterations, mainly associated with ion transport and synapses in glutamatergic neurons. We then demonstrate that BIN1 cell-autonomously regulates gene expression in glutamatergic neurons by using a novel protocol to generate pure culture of hiPSC-derived induced neurons (hiNs). Using this system, we also show that BIN1 plays a key role in the regulation of neuronal calcium transients and electrical activity via its interaction with the L-type voltage-gated calcium channel Cav1.2. BIN1 KO hiNs show reduced activity-dependent internalization and higher Cav1.2 expression compared to WT hiNs. Pharmacological blocking of this channel with clinically relevant doses of nifedipine, a calcium channel blocker, partly rescues electrical and gene expression alterations in BIN1 KO glutamatergic neurons. Further, we show that transcriptional alterations in BIN1 KO hiNs that affect biological processes related to calcium homeostasis are also present in glutamatergic neurons of the human brain at late stages of AD pathology. Together, these findings suggest that BIN1-dependent alterations in neuronal properties could contribute to AD pathophysiology and that treatment with low doses of clinically approved calcium blockers should be considered as an option to slow disease-onset and progression.
Collapse
Affiliation(s)
- Orthis Saha
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1167-RID-AGE facteurs de risque et déterminants moléculaires des maladies liées au vieillissement, DISTALZ, 1 rue du Professeur Calmette, 59019, Lille, France
| | - Ana Raquel Melo de Farias
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1167-RID-AGE facteurs de risque et déterminants moléculaires des maladies liées au vieillissement, DISTALZ, 1 rue du Professeur Calmette, 59019, Lille, France
- Brain Institute, Federal University of Rio Grande do Norte, Av. Senador Salgado Filho, 3000, Campus Universitário, Lagoa, Nova, 59078-970, Natal, Brazil
| | - Alexandre Pelletier
- Univ. Lille, Inserm, CNRS, CHU Lille, Institut Pasteur de Lille, U1283-UMR 8199 EGID, Pôle Recherche, 1 Place de Verdun, 59045, Lille, Cedex, France
- Department of Pharmacology, Physiology & Biophysics, Boston University, Chobanian & Avedisian School of Medicine, Boston, MA, 02118, USA
| | - Dolores Siedlecki-Wullich
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1167-RID-AGE facteurs de risque et déterminants moléculaires des maladies liées au vieillissement, DISTALZ, 1 rue du Professeur Calmette, 59019, Lille, France
| | - Bruna Soares Landeira
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1167-RID-AGE facteurs de risque et déterminants moléculaires des maladies liées au vieillissement, DISTALZ, 1 rue du Professeur Calmette, 59019, Lille, France
| | - Johanna Gadaut
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1167-RID-AGE facteurs de risque et déterminants moléculaires des maladies liées au vieillissement, DISTALZ, 1 rue du Professeur Calmette, 59019, Lille, France
| | - Arnaud Carrier
- Univ. Lille, Inserm, CNRS, CHU Lille, Institut Pasteur de Lille, U1283-UMR 8199 EGID, Pôle Recherche, 1 Place de Verdun, 59045, Lille, Cedex, France
| | - Anaïs-Camille Vreulx
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1167-RID-AGE facteurs de risque et déterminants moléculaires des maladies liées au vieillissement, DISTALZ, 1 rue du Professeur Calmette, 59019, Lille, France
| | - Karine Guyot
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1167-RID-AGE facteurs de risque et déterminants moléculaires des maladies liées au vieillissement, DISTALZ, 1 rue du Professeur Calmette, 59019, Lille, France
| | - Yun Shen
- Department of Pharmacology, Physiology & Biophysics, Boston University, Chobanian & Avedisian School of Medicine, Boston, MA, 02118, USA
| | - Amelie Bonnefond
- Univ. Lille, Inserm, CNRS, CHU Lille, Institut Pasteur de Lille, U1283-UMR 8199 EGID, Pôle Recherche, 1 Place de Verdun, 59045, Lille, Cedex, France
| | - Philippe Amouyel
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1167-RID-AGE facteurs de risque et déterminants moléculaires des maladies liées au vieillissement, DISTALZ, 1 rue du Professeur Calmette, 59019, Lille, France
| | - Julia Tcw
- Department of Pharmacology, Physiology & Biophysics, Boston University, Chobanian & Avedisian School of Medicine, Boston, MA, 02118, USA
- Bioinformatics Program, Faculty of Computing & Data Sciences, Boston University, Boston, MA, 02115, USA
| | - Devrim Kilinc
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1167-RID-AGE facteurs de risque et déterminants moléculaires des maladies liées au vieillissement, DISTALZ, 1 rue du Professeur Calmette, 59019, Lille, France
| | - Claudio Marcos Queiroz
- Brain Institute, Federal University of Rio Grande do Norte, Av. Senador Salgado Filho, 3000, Campus Universitário, Lagoa, Nova, 59078-970, Natal, Brazil
| | - Fabien Delahaye
- Univ. Lille, Inserm, CNRS, CHU Lille, Institut Pasteur de Lille, U1283-UMR 8199 EGID, Pôle Recherche, 1 Place de Verdun, 59045, Lille, Cedex, France
| | - Jean-Charles Lambert
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1167-RID-AGE facteurs de risque et déterminants moléculaires des maladies liées au vieillissement, DISTALZ, 1 rue du Professeur Calmette, 59019, Lille, France
| | - Marcos R Costa
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1167-RID-AGE facteurs de risque et déterminants moléculaires des maladies liées au vieillissement, DISTALZ, 1 rue du Professeur Calmette, 59019, Lille, France.
- Brain Institute, Federal University of Rio Grande do Norte, Av. Senador Salgado Filho, 3000, Campus Universitário, Lagoa, Nova, 59078-970, Natal, Brazil.
| |
Collapse
|
13
|
Karpova A, Aly AAA, Marosi EL, Mikulovic S. Fiber-based in vivo imaging: unveiling avenues for exploring mechanisms of synaptic plasticity and neuronal adaptations underlying behavior. NEUROPHOTONICS 2024; 11:S11507. [PMID: 38390518 PMCID: PMC10883581 DOI: 10.1117/1.nph.11.s1.s11507] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 01/18/2024] [Accepted: 01/23/2024] [Indexed: 02/24/2024]
Abstract
In recent decades, various subfields within neuroscience, spanning molecular, cellular, and systemic dimensions, have significantly advanced our understanding of the elaborate molecular and cellular mechanisms that underpin learning, memory, and adaptive behaviors. There have been notable advancements in imaging techniques, particularly in reaching superficial brain structures. This progress has led to their widespread adoption in numerous laboratories. However, essential physiological and cognitive processes, including sensory integration, emotional modulation of motivated behavior, motor regulation, learning, and memory consolidation, are intricately encoded within deeper brain structures. Hence, visualization techniques such as calcium imaging using miniscopes have gained popularity for studying brain activity in unrestrained animals. Despite its utility, miniscope technology is associated with substantial brain tissue damage caused by gradient refractive index lens implantation. Furthermore, its imaging capabilities are primarily confined to the neuronal somata level, thus constraining a comprehensive exploration of subcellular processes underlying adaptive behaviors. Consequently, the trajectory of neuroscience's future hinges on the development of minimally invasive optical fiber-based endo-microscopes optimized for cellular, subcellular, and molecular imaging within the intricate depths of the brain. In pursuit of this goal, select research groups have invested significant efforts in advancing this technology. In this review, we present a perspective on the potential impact of this innovation on various aspects of neuroscience, enabling the functional exploration of in vivo cellular and subcellular processes that underlie synaptic plasticity and the neuronal adaptations that govern behavior.
Collapse
Affiliation(s)
- Anna Karpova
- Leibniz Institute for Neurobiology, RG Neuroplasticity, Magdeburg, Germany
- Otto von Guericke University, Center for Behavioral Brain Sciences, Magdeburg, Germany
| | - Ahmed A. A. Aly
- Leibniz Institute for Neurobiology, RG Neuroplasticity, Magdeburg, Germany
| | - Endre Levente Marosi
- Leibniz Institute for Neurobiology, RG Cognition and Emotion, Magdeburg, Germany
| | - Sanja Mikulovic
- Otto von Guericke University, Center for Behavioral Brain Sciences, Magdeburg, Germany
- Leibniz Institute for Neurobiology, RG Cognition and Emotion, Magdeburg, Germany
- German Centre for Mental Health (DZPG), Magdeburg, Germany
| |
Collapse
|
14
|
Garcia JP, Armbruster M, Sommer M, Nunez-Beringer A, Dulla CG. Glutamate uptake is transiently compromised in the perilesional cortex following controlled cortical impact. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.28.610143. [PMID: 39257826 PMCID: PMC11383988 DOI: 10.1101/2024.08.28.610143] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/12/2024]
Abstract
Glutamate, the primary excitatory neurotransmitter in the CNS, is regulated by the excitatory amino acid transporters (EAATs) GLT-1 and GLAST. Following traumatic brain injury (TBI), extracellular glutamate levels increase, contributing to excitotoxicity, circuit dysfunction, and morbidity. Increased neuronal glutamate release and compromised astrocyte-mediated uptake contribute to elevated glutamate, but the mechanistic and spatiotemporal underpinnings of these changes are not well established. Using the controlled cortical impact (CCI) model of TBI and iGluSnFR glutamate imaging, we quantified extracellular glutamate dynamics after injury. Three days post-injury, glutamate release was increased, and glutamate uptake and GLT-1 expression were reduced. 7- and 14-days post-injury, glutamate dynamics were comparable between sham and CCI animals. Changes in peak glutamate response were unique to specific cortical layers and proximity to injury. This was likely driven by increases in glutamate release, which was spatially heterogenous, rather than reduced uptake, which was spatially uniform. The astrocyte K + channel, Kir4.1, regulates activity-dependent slowing of glutamate uptake. Surprisingly, Kir4.1 was unchanged after CCI and accordingly, activity-dependent slowing of glutamate uptake was unaltered. This dynamic glutamate dysregulation after TBI underscores a brief period in which disrupted glutamate uptake may contribute to dysfunction and highlights a potential therapeutic window to restore glutamate homeostasis.
Collapse
|
15
|
Stern MA, Dingledine R, Gross RE, Berglund K. Epilepsy insights revealed by intravital functional optical imaging. Front Neurol 2024; 15:1465232. [PMID: 39268067 PMCID: PMC11390408 DOI: 10.3389/fneur.2024.1465232] [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: 07/16/2024] [Accepted: 08/13/2024] [Indexed: 09/15/2024] Open
Abstract
Despite an abundance of pharmacologic and surgical epilepsy treatments, there remain millions of patients suffering from poorly controlled seizures. One approach to closing this treatment gap may be found through a deeper mechanistic understanding of the network alterations that underly this aberrant activity. Functional optical imaging in vertebrate models provides powerful advantages to this end, enabling the spatiotemporal acquisition of individual neuron activity patterns across multiple seizures. This coupled with the advent of genetically encoded indicators, be them for specific ions, neurotransmitters or voltage, grants researchers unparalleled access to the intact nervous system. Here, we will review how in vivo functional optical imaging in various vertebrate seizure models has advanced our knowledge of seizure dynamics, principally seizure initiation, propagation and termination.
Collapse
Affiliation(s)
- Matthew A Stern
- Department of Neurosurgery, Emory University School of Medicine, Atlanta, GA, United States
| | - Raymond Dingledine
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA, United States
| | - Robert E Gross
- Department of Neurosurgery, Emory University School of Medicine, Atlanta, GA, United States
- Department of Neurological Surgery, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, United States
| | - Ken Berglund
- Department of Neurosurgery, Emory University School of Medicine, Atlanta, GA, United States
| |
Collapse
|
16
|
Wang Q, Yang C, Chen S, Li J. Miniaturized Electrochemical Sensing Platforms for Quantitative Monitoring of Glutamate Dynamics in the Central Nervous System. Angew Chem Int Ed Engl 2024; 63:e202406867. [PMID: 38829963 DOI: 10.1002/anie.202406867] [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: 04/10/2024] [Revised: 05/30/2024] [Accepted: 06/03/2024] [Indexed: 06/05/2024]
Abstract
Glutamate is one of the most important excitatory neurotransmitters within the mammalian central nervous system. The role of glutamate in regulating neural network signaling transmission through both synaptic and extra-synaptic paths highlights the importance of the real-time and continuous monitoring of its concentration and dynamics in living organisms. Progresses in multidisciplinary research have promoted the development of electrochemical glutamate sensors through the co-design of materials, interfaces, electronic devices, and integrated systems. This review summarizes recent works reporting various electrochemical sensor designs and their applicability as miniaturized neural probes to in vivo sensing within biological environments. We start with an overview of the role and physiological significance of glutamate, the metabolic routes, and its presence in various bodily fluids. Next, we discuss the design principles, commonly employed validation models/protocols, and successful demonstrations of multifunctional, compact, and bio-integrated devices in animal models. The final section provides an outlook on the development of the next generation glutamate sensors for neuroscience and neuroengineering, with the aim of offering practical guidance for future research.
Collapse
Affiliation(s)
- Qi Wang
- Department of Materials Science and Engineering, The Ohio State University, Columbus, OH 43210, USA
| | - Chunyu Yang
- Department of Materials Science and Engineering, The Ohio State University, Columbus, OH 43210, USA
| | - Shulin Chen
- Department of Materials Science and Engineering, The Ohio State University, Columbus, OH 43210, USA
| | - Jinghua Li
- Department of Materials Science and Engineering, The Ohio State University, Columbus, OH 43210, USA
- Chronic Brain Injury Program, The Ohio State University, Columbus, OH 43210, USA
| |
Collapse
|
17
|
Hellweg L, Pfeifer M, Tarnawski M, Thing-Teoh S, Chang L, Bergner A, Kress J, Hiblot J, Wiedmer T, Superti-Furga G, Reinhardt J, Johnsson K, Leippe P. AspSnFR: A genetically encoded biosensor for real-time monitoring of aspartate in live cells. Cell Chem Biol 2024; 31:1529-1541.e12. [PMID: 38806058 DOI: 10.1016/j.chembiol.2024.05.002] [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: 01/18/2024] [Revised: 03/11/2024] [Accepted: 05/01/2024] [Indexed: 05/30/2024]
Abstract
Aspartate is crucial for nucleotide synthesis, ammonia detoxification, and maintaining redox balance via the malate-aspartate-shuttle (MAS). To disentangle these multiple roles of aspartate metabolism, tools are required that measure aspartate concentrations in real time and in live cells. We introduce AspSnFR, a genetically encoded green fluorescent biosensor for intracellular aspartate, engineered through displaying and screening biosensor libraries on mammalian cells. In live cells, AspSnFR is able to precisely and quantitatively measure cytosolic aspartate concentrations and dissect its production from glutamine. Combining high-content imaging of AspSnFR with pharmacological perturbations exposes differences in metabolic vulnerabilities of aspartate levels based on nutrient availability. Further, AspSnFR facilitates tracking of aspartate export from mitochondria through SLC25A12, the MAS' key transporter. We show that SLC25A12 is a rapidly responding and direct route to couple Ca2+ signaling with mitochondrial aspartate export. This establishes SLC25A12 as a crucial link between cellular signaling, mitochondrial respiration, and metabolism.
Collapse
Affiliation(s)
- Lars Hellweg
- Department of Chemical Biology, Max Planck Institute for Medical Research, Heidelberg, Germany; Heidelberg University, Heidelberg, Germany
| | - Martin Pfeifer
- Novartis Biomedical Research, Discovery Science, Basel, Switzerland
| | - Miroslaw Tarnawski
- Protein Expression and Characterization Facility, Max Planck Institute for Medical Research, Heidelberg, Germany
| | - Shao Thing-Teoh
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Lena Chang
- Novartis Biomedical Research, Discovery Science, Basel, Switzerland
| | - Andrea Bergner
- Department of Chemical Biology, Max Planck Institute for Medical Research, Heidelberg, Germany
| | - Jana Kress
- Department of Chemical Biology, Max Planck Institute for Medical Research, Heidelberg, Germany
| | - Julien Hiblot
- Department of Chemical Biology, Max Planck Institute for Medical Research, Heidelberg, Germany
| | - Tabea Wiedmer
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Giulio Superti-Furga
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria; Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Jürgen Reinhardt
- Novartis Biomedical Research, Discovery Science, Basel, Switzerland
| | - Kai Johnsson
- Department of Chemical Biology, Max Planck Institute for Medical Research, Heidelberg, Germany; Institute of Chemical Sciences and Engineering (ISIC), École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland.
| | - Philipp Leippe
- Department of Chemical Biology, Max Planck Institute for Medical Research, Heidelberg, Germany; CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria.
| |
Collapse
|
18
|
Duan J, Kahms M, Steinhoff A, Klingauf J. Spontaneous and evoked synaptic vesicle release arises from a single releasable pool. Cell Rep 2024; 43:114461. [PMID: 38990719 DOI: 10.1016/j.celrep.2024.114461] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Revised: 05/23/2024] [Accepted: 06/23/2024] [Indexed: 07/13/2024] Open
Abstract
The quantal content of an evoked postsynaptic response is typically determined by dividing it by the average spontaneous miniature response. However, this approach is challenged by the notion that different synaptic vesicle pools might drive spontaneous and evoked release. Here, we "silence" synaptic vesicles through pharmacological alkalinization and subsequently rescue them by optogenetic acidification. We find that such silenced synaptic vesicles, retrieved during evoked or spontaneous activity, cross-deplete the complementary release mode in a fully reversible manner. A fluorescently tagged version of the endosomal SNARE protein Vti1a, which has been suggested to identify a separate pool of spontaneously recycling synaptic vesicles, is trafficked to synaptic vesicles significantly only upon overexpression but not when endogenously tagged by CRISPR-Cas9. Thus, both release modes draw synaptic vesicles from the same readily releasable pool.
Collapse
Affiliation(s)
- Junxiu Duan
- Department of Cellular Biophysics, Institute of Medical Physics and Biophysics, University of Münster, Robert-Koch-Str. 31, 48149 Münster, Germany; Center for Soft Nanoscience SoN, University of Münster, Busso-Peus-Str.10, 48149 Münster, Germany; Cells in Motion Interfaculty Center, University of Münster, 48149 Münster, Germany; CiM Graduate School of the Cells in Motion Interfaculty Centre and the International Max Planck Research School, 48149 Münster, Germany
| | - Martin Kahms
- Department of Cellular Biophysics, Institute of Medical Physics and Biophysics, University of Münster, Robert-Koch-Str. 31, 48149 Münster, Germany; Center for Soft Nanoscience SoN, University of Münster, Busso-Peus-Str.10, 48149 Münster, Germany; Cells in Motion Interfaculty Center, University of Münster, 48149 Münster, Germany
| | - Ana Steinhoff
- Department of Cellular Biophysics, Institute of Medical Physics and Biophysics, University of Münster, Robert-Koch-Str. 31, 48149 Münster, Germany; Center for Soft Nanoscience SoN, University of Münster, Busso-Peus-Str.10, 48149 Münster, Germany; Cells in Motion Interfaculty Center, University of Münster, 48149 Münster, Germany; CiM Graduate School of the Cells in Motion Interfaculty Centre and the International Max Planck Research School, 48149 Münster, Germany
| | - Jürgen Klingauf
- Department of Cellular Biophysics, Institute of Medical Physics and Biophysics, University of Münster, Robert-Koch-Str. 31, 48149 Münster, Germany; Center for Soft Nanoscience SoN, University of Münster, Busso-Peus-Str.10, 48149 Münster, Germany; Cells in Motion Interfaculty Center, University of Münster, 48149 Münster, Germany.
| |
Collapse
|
19
|
Bouabid S, Zhang L, Vu MAT, Tang K, Graham BM, Noggle CA, Howe MW. Spatially organized striatum-wide acetylcholine dynamics for the learning and extinction of Pavlovian cues and actions. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.10.602947. [PMID: 39071401 PMCID: PMC11275942 DOI: 10.1101/2024.07.10.602947] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/30/2024]
Abstract
Striatal acetylcholine (ACh) has been linked to behavioral flexibility. A key component of flexibility is down-regulating responding as valued cues and actions become decoupled from positive outcomes. We used array fiber photometry in mice to investigate how ACh release across the striatum evolves during learning and extinction of Pavlovian associations. Changes in multi-phasic release to cues and consummatory actions were bi-directional and region-specific. Following extinction, increases in cue-evoked ACh release emerged in the anterior dorsal striatum (aDS) which preceded a down-regulation of anticipatory behavior. Silencing ACh release from cholinergic interneurons in the aDS blocked behavioral extinction. Dopamine release dipped below baseline for down-shifted cues, but glutamate input onto cholinergic interneurons did not change, suggesting an intrastriatal mechanism for the emergence of ACh increases. Our large-scale mapping of striatal ACh dynamics during learning pinpoints region-specific elevations in ACh release positioned to down-regulate behavior during extinction, a central feature of flexible behavior.
Collapse
Affiliation(s)
- Safa Bouabid
- Department of Psychological & Brain Sciences, Boston University, Boston, MA, USA
| | - Liangzhu Zhang
- Department of Psychological & Brain Sciences, Boston University, Boston, MA, USA
| | - Mai-Anh T. Vu
- Department of Psychological & Brain Sciences, Boston University, Boston, MA, USA
| | - Kylie Tang
- Department of Psychological & Brain Sciences, Boston University, Boston, MA, USA
| | - Benjamin M. Graham
- Department of Psychological & Brain Sciences, Boston University, Boston, MA, USA
| | - Christian A. Noggle
- Department of Psychological & Brain Sciences, Boston University, Boston, MA, USA
| | - Mark W. Howe
- Department of Psychological & Brain Sciences, Boston University, Boston, MA, USA
| |
Collapse
|
20
|
Zott B, Nästle L, Grienberger C, Unger F, Knauer MM, Wolf C, Keskin-Dargin A, Feuerbach A, Busche MA, Skerra A, Konnerth A. β-amyloid monomer scavenging by an anticalin protein prevents neuronal hyperactivity in mouse models of Alzheimer's Disease. Nat Commun 2024; 15:5819. [PMID: 38987287 PMCID: PMC11237084 DOI: 10.1038/s41467-024-50153-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Accepted: 07/02/2024] [Indexed: 07/12/2024] Open
Abstract
Hyperactivity mediated by synaptotoxic β-amyloid (Aβ) oligomers is one of the earliest forms of neuronal dysfunction in Alzheimer's disease. In the search for a preventive treatment strategy, we tested the effect of scavenging Aβ peptides before Aβ plaque formation. Using in vivo two-photon calcium imaging and SF-iGluSnFR-based glutamate imaging in hippocampal slices, we demonstrate that an Aβ binding anticalin protein (Aβ-anticalin) can suppress early neuronal hyperactivity and synaptic glutamate accumulation in the APP23xPS45 mouse model of β-amyloidosis. Our results suggest that the sole targeting of Aβ monomers is sufficient for the hyperactivity-suppressing effect of the Aβ-anticalin at early disease stages. Biochemical and neurophysiological analyses indicate that the Aβ-anticalin-dependent depletion of naturally secreted Aβ monomers interrupts their aggregation to neurotoxic oligomers and, thereby, reverses early neuronal and synaptic dysfunctions. Thus, our results suggest that Aβ monomer scavenging plays a key role in the repair of neuronal function at early stages of AD.
Collapse
Affiliation(s)
- Benedikt Zott
- Institute of Neuroscience, Technical University of Munich, Munich, Germany.
- Department of Neuroradiology, MRI hospital of the Technical University of Munich, Munich, Germany.
- TUM Institute for Advanced Study, Garching, Germany.
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany.
| | - Lea Nästle
- Chair of Biological Chemistry, Technical University of Munich, Freising, Germany
| | - Christine Grienberger
- Institute of Neuroscience, Technical University of Munich, Munich, Germany
- Department of Biology and Volen National Center of Complex Systems, Brandeis University, Waltham, MA, USA
| | - Felix Unger
- Institute of Neuroscience, Technical University of Munich, Munich, Germany
- Department of Neuroradiology, MRI hospital of the Technical University of Munich, Munich, Germany
- TUM Institute for Advanced Study, Garching, Germany
| | - Manuel M Knauer
- Institute of Neuroscience, Technical University of Munich, Munich, Germany
| | - Christian Wolf
- Institute of Neuroscience, Technical University of Munich, Munich, Germany
- Department of Neuroradiology, MRI hospital of the Technical University of Munich, Munich, Germany
| | | | - Anna Feuerbach
- Chair of Biological Chemistry, Technical University of Munich, Freising, Germany
| | - Marc Aurel Busche
- Institute of Neuroscience, Technical University of Munich, Munich, Germany
- UK Dementia Research Institute at UCL, University College London, London, United Kingdom
| | - Arne Skerra
- Chair of Biological Chemistry, Technical University of Munich, Freising, Germany.
| | - Arthur Konnerth
- Institute of Neuroscience, Technical University of Munich, Munich, Germany.
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany.
| |
Collapse
|
21
|
Xu C, Alameri A, Leong W, Johnson E, Chen Z, Xu B, Leong KW. Multiscale engineering of brain organoids for disease modeling. Adv Drug Deliv Rev 2024; 210:115344. [PMID: 38810702 PMCID: PMC11265575 DOI: 10.1016/j.addr.2024.115344] [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: 02/13/2024] [Revised: 04/23/2024] [Accepted: 05/25/2024] [Indexed: 05/31/2024]
Abstract
Brain organoids hold great potential for modeling human brain development and pathogenesis. They recapitulate certain aspects of the transcriptional trajectory, cellular diversity, tissue architecture and functions of the developing brain. In this review, we explore the engineering strategies to control the molecular-, cellular- and tissue-level inputs to achieve high-fidelity brain organoids. We review the application of brain organoids in neural disorder modeling and emerging bioengineering methods to improve data collection and feature extraction at multiscale. The integration of multiscale engineering strategies and analytical methods has significant potential to advance insight into neurological disorders and accelerate drug development.
Collapse
Affiliation(s)
- Cong Xu
- Department of Biomedical Engineering, Columbia University, New York, NY 10027, USA
| | - Alia Alameri
- Department of Biomedical Engineering, Columbia University, New York, NY 10027, USA
| | - Wei Leong
- Department of Biomedical Engineering, Columbia University, New York, NY 10027, USA
| | - Emily Johnson
- Department of Biomedical Engineering, Columbia University, New York, NY 10027, USA
| | - Zaozao Chen
- Department of Biomedical Engineering, Columbia University, New York, NY 10027, USA
| | - Bin Xu
- Department of Psychiatry, Columbia University, New York, NY 10032, USA.
| | - Kam W Leong
- Department of Biomedical Engineering, Columbia University, New York, NY 10027, USA.
| |
Collapse
|
22
|
Leong LM, Storace DA. Imaging different cell populations in the mouse olfactory bulb using the genetically encoded voltage indicator ArcLight. NEUROPHOTONICS 2024; 11:033402. [PMID: 38288247 PMCID: PMC10823906 DOI: 10.1117/1.nph.11.3.033402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Revised: 11/30/2023] [Accepted: 12/14/2023] [Indexed: 01/31/2024]
Abstract
Genetically encoded voltage indicators (GEVIs) are protein-based optical sensors that allow for measurements from genetically defined populations of neurons. Although in vivo imaging in the mammalian brain with early generation GEVIs was difficult due to poor membrane expression and low signal-to-noise ratio, newer and more sensitive GEVIs have begun to make them useful for answering fundamental questions in neuroscience. We discuss principles of imaging using GEVIs and genetically encoded calcium indicators, both useful tools for in vivo imaging of neuronal activity, and review some of the recent mechanistic advances that have led to GEVI improvements. We provide an overview of the mouse olfactory bulb (OB) and discuss recent studies using the GEVI ArcLight to study different cell types within the bulb using both widefield and two-photon microscopy. Specific emphasis is placed on using GEVIs to begin to study the principles of concentration coding in the OB, how to interpret the optical signals from population measurements in the in vivo brain, and future developments that will push the field forward.
Collapse
Affiliation(s)
- Lee Min Leong
- Florida State University, Department of Biological Science, Tallahassee, Florida, United States
| | - Douglas A. Storace
- Florida State University, Department of Biological Science, Tallahassee, Florida, United States
- Florida State University, Program in Neuroscience, Tallahassee, Florida, United States
- Florida State University, Institute of Molecular Biophysics, Tallahassee, Florida, United States
| |
Collapse
|
23
|
Chen W, Ge X, Zhang Q, Natan RG, Fan JL, Scanziani M, Ji N. High-throughput volumetric mapping of synaptic transmission. Nat Methods 2024; 21:1298-1305. [PMID: 38898094 DOI: 10.1038/s41592-024-02309-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Accepted: 05/15/2024] [Indexed: 06/21/2024]
Abstract
Volumetric imaging of synaptic transmission in vivo requires high spatial and high temporal resolution. Shaping the wavefront of two-photon fluorescence excitation light, we developed Bessel-droplet foci for high-contrast and high-resolution volumetric imaging of synapses. Applying our method to imaging glutamate release, we demonstrated high-throughput mapping of excitatory inputs at >1,000 synapses per volume and >500 dendritic spines per neuron in vivo and unveiled previously unseen features of functional synaptic organization in the mouse primary visual cortex.
Collapse
Affiliation(s)
- Wei Chen
- Department of Physics, University of California, Berkeley, CA, USA
- School of Mechanical Science and Engineering - Advanced Biomedical Imaging Facility, Huazhong University of Science and Technology, Wuhan, China
| | - Xinxin Ge
- Department of Physiology, University of California, San Francisco, San Francisco, CA, USA
- Howard Hughes Medical Institute, University of California, San Francisco, San Francisco, CA, USA
| | - Qinrong Zhang
- Department of Physics, University of California, Berkeley, CA, USA
| | - Ryan G Natan
- Department of Physics, University of California, Berkeley, CA, USA
- Department of Molecular and Cell Biology, University of California, Berkeley, CA, USA
| | - Jiang Lan Fan
- Joint Bioengineering Graduate Program, University of California, Berkeley, CA, USA
- Joint Bioengineering Graduate Program, University of California, San Francisco, CA, USA
| | - Massimo Scanziani
- Department of Physiology, University of California, San Francisco, San Francisco, CA, USA
- Howard Hughes Medical Institute, University of California, San Francisco, San Francisco, CA, USA
| | - Na Ji
- Department of Physics, University of California, Berkeley, CA, USA.
- Department of Molecular and Cell Biology, University of California, Berkeley, CA, USA.
- Helen Wills Neuroscience Institute, University of California, Berkeley, CA, USA.
- Molecular Biophysics and Integrated Bioimaging Division, Lawrence Berkeley National Laboratory, Berkeley, CA, USA.
| |
Collapse
|
24
|
Sekine K, Haga W, Kim S, Imayasu M, Yoshida T, Tsutsui H. Neuron-microelectrode junction induced by an engineered synapse organizer. Biochem Biophys Res Commun 2024; 712-713:149935. [PMID: 38626529 DOI: 10.1016/j.bbrc.2024.149935] [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: 03/06/2024] [Revised: 04/04/2024] [Accepted: 04/11/2024] [Indexed: 04/18/2024]
Abstract
The conventional microelectrodes for recording neuronal activities do not have innate selectivity to cell type, which is one of the critical limitations for the detailed analysis of neuronal circuits. In this study, we engineered a downsized variant of the artificial synapse organizer based on neurexin1β and a peptide-tag, fabricated gold microelectrodes functionalized with the receptor for the organizer, and performed validation experiments in primary cultured neurons. Successful inductions of synapse-like junctions were detected at the sites of contact between neurons expressing the engineered synapse organizer and functionalized microelectrodes, but not in the negative control experiment in which the electrode functionalization was omitted. Such a molecularly inducible neuron-microelectrode junction could be the basis for the next-generation electrophysiological technique enabling cell type-selective recording.
Collapse
Affiliation(s)
- Kosuke Sekine
- School of Materials Science, Japan Advanced Institute of Science and Technology, Nomi, Ishikawa, 923-1292, Japan
| | - Wataru Haga
- School of Materials Science, Japan Advanced Institute of Science and Technology, Nomi, Ishikawa, 923-1292, Japan
| | - Samyoung Kim
- School of Materials Science, Japan Advanced Institute of Science and Technology, Nomi, Ishikawa, 923-1292, Japan
| | - Mieko Imayasu
- School of Materials Science, Japan Advanced Institute of Science and Technology, Nomi, Ishikawa, 923-1292, Japan
| | - Tomoyuki Yoshida
- Department of Molecular Neuroscience, Faculty of Medicine, University of Toyama, Toyama, 930-0194, Japan; Research Center for Idling Brain Science, University of Toyama, Toyama, 930-0194, Japan
| | - Hidekazu Tsutsui
- School of Materials Science, Japan Advanced Institute of Science and Technology, Nomi, Ishikawa, 923-1292, Japan; Division of Transdisciplinary Sciences, Japan Advanced Institute of Science and Technology, Nomi, Ishikawa, 923-1292, Japan.
| |
Collapse
|
25
|
Boto T, Tomchik SM. Functional Imaging of Learning-Induced Plasticity in the Central Nervous System with Genetically Encoded Reporters in Drosophila. Cold Spring Harb Protoc 2024; 2024:pdb.top107799. [PMID: 37197830 DOI: 10.1101/pdb.top107799] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/19/2023]
Abstract
Learning and memory allow animals to adjust their behavior based on the predictive value of their past experiences. Memories often exist in complex representations, spread across numerous cells and synapses in the brain. Studying relatively simple forms of memory provides insights into the fundamental processes that underlie multiple forms of memory. Associative learning occurs when an animal learns the relationship between two previously unrelated sensory stimuli, such as when a hungry animal learns that a particular odor is followed by a tasty reward. Drosophila is a particularly powerful model to study how this type of memory works. The fundamental principles are widely shared among animals, and there is a wide range of genetic tools available to study circuit function in flies. In addition, the olfactory structures that mediate associative learning in flies, such as the mushroom body and its associated neurons, are anatomically organized, relatively well-characterized, and readily accessible to imaging. Here, we review the olfactory anatomy and physiology of the olfactory system, describe how plasticity in the olfactory pathway mediates learning and memory, and explain the general principles underlying calcium imaging approaches.
Collapse
Affiliation(s)
- Tamara Boto
- Department of Physiology, Trinity College Dublin, Dublin 2, Ireland
- Trinity College Institute of Neuroscience, Trinity College Dublin, Dublin 2, Ireland
| | - Seth M Tomchik
- Neuroscience and Pharmacology, University of Iowa Carver College of Medicine, Iowa City, Iowa 52242, USA
- Stead Family Department of Pediatrics, University of Iowa Carver College of Medicine, Iowa City, Iowa 52242, USA
- Iowa Neuroscience Institute, University of Iowa Carver College of Medicine, Iowa City, Iowa 52242, USA
| |
Collapse
|
26
|
Xiao S, Cunningham WJ, Kondabolu K, Lowet E, Moya MV, Mount RA, Ravasio C, Bortz E, Shaw D, Economo MN, Han X, Mertz J. Large-scale deep tissue voltage imaging with targeted-illumination confocal microscopy. Nat Methods 2024; 21:1094-1102. [PMID: 38840033 PMCID: PMC11500676 DOI: 10.1038/s41592-024-02275-w] [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: 07/04/2023] [Accepted: 04/09/2024] [Indexed: 06/07/2024]
Abstract
Voltage imaging with cellular specificity has been made possible by advances in genetically encoded voltage indicators. However, the kilohertz rates required for voltage imaging lead to weak signals. Moreover, out-of-focus fluorescence and tissue scattering produce background that both undermines the signal-to-noise ratio and induces crosstalk between cells, making reliable in vivo imaging in densely labeled tissue highly challenging. We describe a microscope that combines the distinct advantages of targeted illumination and confocal gating while also maximizing signal detection efficiency. The resulting benefits in signal-to-noise ratio and crosstalk reduction are quantified experimentally and theoretically. Our microscope provides a versatile solution for enabling high-fidelity in vivo voltage imaging at large scales and penetration depths, which we demonstrate across a wide range of imaging conditions and different genetically encoded voltage indicator classes.
Collapse
Affiliation(s)
- Sheng Xiao
- Department of Biomedical Engineering, Boston University, Boston, MA, USA.
| | | | | | - Eric Lowet
- Department of Biomedical Engineering, Boston University, Boston, MA, USA
- Department of Neuroscience, Erasmus MC, Rotterdam, the Netherlands
| | - Maria V Moya
- Department of Biomedical Engineering, Boston University, Boston, MA, USA
| | - Rebecca A Mount
- Department of Biomedical Engineering, Boston University, Boston, MA, USA
| | - Cara Ravasio
- Department of Biomedical Engineering, Boston University, Boston, MA, USA
| | - Emma Bortz
- Department of Biomedical Engineering, Boston University, Boston, MA, USA
| | - Dana Shaw
- Graduate Program for Neuroscience, Boston University, Boston, MA, USA
- Neurophotonics Center, Boston University, Boston, MA, USA
| | - Michael N Economo
- Department of Biomedical Engineering, Boston University, Boston, MA, USA
- Neurophotonics Center, Boston University, Boston, MA, USA
| | - Xue Han
- Department of Biomedical Engineering, Boston University, Boston, MA, USA
- Neurophotonics Center, Boston University, Boston, MA, USA
| | - Jerome Mertz
- Department of Biomedical Engineering, Boston University, Boston, MA, USA
- Neurophotonics Center, Boston University, Boston, MA, USA
| |
Collapse
|
27
|
Guo F, Fan J, Liu JM, Kong PL, Ren J, Mo JW, Lu CL, Zhong QL, Chen LY, Jiang HT, Zhang C, Wen YL, Gu TT, Li SJ, Fang YY, Pan BX, Gao TM, Cao X. Astrocytic ALKBH5 in stress response contributes to depressive-like behaviors in mice. Nat Commun 2024; 15:4347. [PMID: 38773146 PMCID: PMC11109195 DOI: 10.1038/s41467-024-48730-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Accepted: 05/07/2024] [Indexed: 05/23/2024] Open
Abstract
Epigenetic mechanisms bridge genetic and environmental factors that contribute to the pathogenesis of major depression disorder (MDD). However, the cellular specificity and sensitivity of environmental stress on brain epitranscriptomics and its impact on depression remain unclear. Here, we found that ALKBH5, an RNA demethylase of N6-methyladenosine (m6A), was increased in MDD patients' blood and depression models. ALKBH5 in astrocytes was more sensitive to stress than that in neurons and endothelial cells. Selective deletion of ALKBH5 in astrocytes, but not in neurons and endothelial cells, produced antidepressant-like behaviors. Astrocytic ALKBH5 in the mPFC regulated depression-related behaviors bidirectionally. Meanwhile, ALKBH5 modulated glutamate transporter-1 (GLT-1) m6A modification and increased the expression of GLT-1 in astrocytes. ALKBH5 astrocyte-specific knockout preserved stress-induced disruption of glutamatergic synaptic transmission, neuronal atrophy and defective Ca2+ activity. Moreover, enhanced m6A modification with S-adenosylmethionine (SAMe) produced antidepressant-like effects. Our findings indicate that astrocytic epitranscriptomics contribute to depressive-like behaviors and that astrocytic ALKBH5 may be a therapeutic target for depression.
Collapse
MESH Headings
- Animals
- Astrocytes/metabolism
- AlkB Homolog 5, RNA Demethylase/metabolism
- AlkB Homolog 5, RNA Demethylase/genetics
- Mice
- Humans
- Depressive Disorder, Major/metabolism
- Depressive Disorder, Major/genetics
- Depressive Disorder, Major/pathology
- Male
- Mice, Knockout
- Female
- Disease Models, Animal
- Mice, Inbred C57BL
- Neurons/metabolism
- Stress, Psychological/metabolism
- Adenosine/analogs & derivatives
- Adenosine/metabolism
- Excitatory Amino Acid Transporter 2/metabolism
- Excitatory Amino Acid Transporter 2/genetics
- Behavior, Animal
- Prefrontal Cortex/metabolism
- Prefrontal Cortex/pathology
- Depression/metabolism
- Depression/genetics
- Adult
- Synaptic Transmission
- Middle Aged
Collapse
Affiliation(s)
- Fang Guo
- 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, China
| | - Jun Fan
- Department of Anesthesia, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou, Guangdong, China
| | - Jin-Ming Liu
- 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, China
| | - Peng-Li Kong
- 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, China
| | - Jing Ren
- 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, China
| | - Jia-Wen Mo
- 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, China
| | - Cheng-Lin Lu
- 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, China
| | - Qiu-Ling Zhong
- 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, China
| | - Liang-Yu Chen
- 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, China
| | - Hao-Tian Jiang
- 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, China
| | - Canyuan Zhang
- 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, China
| | - You-Lu Wen
- Department of Psychology and Behavior, Guangdong 999 Brain Hospital, Institute for Brain Research and Rehabilitation, South China Normal University, Guangzhou, Guangdong, P. R. China
| | - Ting-Ting Gu
- Department of Psychology and Behavior, Guangdong 999 Brain Hospital, Institute for Brain Research and Rehabilitation, South China Normal University, Guangzhou, Guangdong, P. R. China
| | - Shu-Ji Li
- 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, China
| | - Ying-Ying Fang
- 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, China
| | - Bing-Xing Pan
- Department of Biological Science, School of Life Science, Nanchang University, Nanchang, China
| | - Tian-Ming Gao
- 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, China
| | - Xiong Cao
- 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, China.
- Department of Oncology, Nanfang Hospital, Southern Medical University Guangzhou, Guangdong, P. R. China.
- Microbiome Medicine Center, Department of Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, P. R. China.
| |
Collapse
|
28
|
Qiu GL, Peng LJ, Wang P, Yang ZL, Zhang JQ, Liu H, Zhu XN, Rao J, Liu XS. In vivo imaging reveals a synchronized correlation among neurotransmitter dynamics during propofol and sevoflurane anesthesia. Zool Res 2024; 45:679-690. [PMID: 38766749 PMCID: PMC11188615 DOI: 10.24272/j.issn.2095-8137.2023.302] [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: 11/12/2023] [Accepted: 12/25/2023] [Indexed: 05/22/2024] Open
Abstract
General anesthesia is widely applied in clinical practice. However, the precise mechanism of loss of consciousness induced by general anesthetics remains unknown. Here, we measured the dynamics of five neurotransmitters, including γ-aminobutyric acid, glutamate, norepinephrine, acetylcholine, and dopamine, in the medial prefrontal cortex and primary visual cortex of C57BL/6 mice through in vivo fiber photometry and genetically encoded neurotransmitter sensors under anesthesia to reveal the mechanism of general anesthesia from a neurotransmitter perspective. Results revealed that the concentrations of γ-aminobutyric acid, glutamate, norepinephrine, and acetylcholine increased in the cortex during propofol-induced loss of consciousness. Dopamine levels did not change following the hypnotic dose of propofol but increased significantly following surgical doses of propofol anesthesia. Notably, the concentrations of the five neurotransmitters generally decreased during sevoflurane-induced loss of consciousness. Furthermore, the neurotransmitter dynamic networks were not synchronized in the non-anesthesia groups but were highly synchronized in the anesthetic groups. These findings suggest that neurotransmitter dynamic network synchronization may cause anesthetic-induced loss of consciousness.
Collapse
Affiliation(s)
- Gao-Lin Qiu
- Department of Anesthesiology, First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, China
| | - Li-Jun Peng
- Department of Anesthesiology, First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, China
| | - Peng Wang
- Department of Anesthesiology, First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, China
| | - Zhi-Lai Yang
- Department of Anesthesiology, First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, China
| | - Ji-Qian Zhang
- Department of Anesthesiology, First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, China
| | - Hu Liu
- Department of Anesthesiology, First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, China
| | - Xiao-Na Zhu
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China. E-mail:
| | - Jin Rao
- Department of Anesthesiology, First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, China. E-mail:
| | - Xue-Sheng Liu
- Department of Anesthesiology, First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, China. E-mail:
| |
Collapse
|
29
|
Tang X, Shi J, Lin S, He Z, Cui S, Di W, Chen S, Wu J, Yuan S, Ye Q, Yang X, Shang Y, Zhang Z, Wang L, Lu L, Tang C, Xu N, Yao L. Pyramidal and parvalbumin neurons modulate the process of electroacupuncture stimulation for stroke rehabilitation. iScience 2024; 27:109695. [PMID: 38680657 PMCID: PMC11053320 DOI: 10.1016/j.isci.2024.109695] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Revised: 02/09/2024] [Accepted: 04/05/2024] [Indexed: 05/01/2024] Open
Abstract
Electroacupuncture (EA) stimulation has been shown to be beneficial in stroke rehabilitation; however, little is known about the neurological mechanism by which this peripheral stimulation approach treats for stroke. This study showed that both pyramidal and parvalbumin (PV) neuronal activity increased in the contralesional primary motor cortex forelimb motor area (M1FL) after ischemic stroke induced by focal unilateral occlusion in the M1FL. EA stimulation reduced pyramidal neuronal activity and increased PV neuronal activity. These results were obtained by a combination of fiber photometry recordings, in vivo and in vitro electrophysiological recordings, and immunofluorescence. Moreover, EA was found to regulate the expression/function of N-methyl-D-aspartate receptors (NMDARs) altered by stroke pathology. In summary, our findings suggest that EA could restore disturbed neuronal activity through the regulation of the activity of pyramidal and PV neurons. Furthermore, NMDARs we shown to play an important role in EA-mediated improvements in sensorimotor ability during stroke rehabilitation.
Collapse
Affiliation(s)
- Xiaorong Tang
- South China Research Center for Acupuncture and Moxibustion, Medical College of Acu-Moxi and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Jiahui Shi
- South China Research Center for Acupuncture and Moxibustion, Medical College of Acu-Moxi and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Shumin Lin
- South China Research Center for Acupuncture and Moxibustion, Medical College of Acu-Moxi and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Zhiyin He
- South China Research Center for Acupuncture and Moxibustion, Medical College of Acu-Moxi and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Shuai Cui
- Research Institute of Acupuncture and Meridian, Anhui University of Chinese Medicine, Hefei 230000, Anhui Province, China
- College of Acupuncture and Moxibustion, Anhui University of Chinese Medicine, Hefei 230000, Anhui Province, China
| | - Wenhui Di
- South China Research Center for Acupuncture and Moxibustion, Medical College of Acu-Moxi and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Siyun Chen
- South China Research Center for Acupuncture and Moxibustion, Medical College of Acu-Moxi and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Junshang Wu
- South China Research Center for Acupuncture and Moxibustion, Medical College of Acu-Moxi and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Si Yuan
- South China Research Center for Acupuncture and Moxibustion, Medical College of Acu-Moxi and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Qiuping Ye
- South China Research Center for Acupuncture and Moxibustion, Medical College of Acu-Moxi and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Xiaoyun Yang
- South China Research Center for Acupuncture and Moxibustion, Medical College of Acu-Moxi and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Ying Shang
- South China Research Center for Acupuncture and Moxibustion, Medical College of Acu-Moxi and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Zhaoxiang Zhang
- State Key Laboratory of Chemical Oncogenomics, Guangdong Provincial Key Laboratory of Chemical Genomics, Peking University, Shenzhen 518055, China
| | - Lin Wang
- South China Research Center for Acupuncture and Moxibustion, Medical College of Acu-Moxi and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Liming Lu
- South China Research Center for Acupuncture and Moxibustion, Medical College of Acu-Moxi and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Chunzhi Tang
- South China Research Center for Acupuncture and Moxibustion, Medical College of Acu-Moxi and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Nenggui Xu
- South China Research Center for Acupuncture and Moxibustion, Medical College of Acu-Moxi and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Lulu Yao
- South China Research Center for Acupuncture and Moxibustion, Medical College of Acu-Moxi and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| |
Collapse
|
30
|
Piantadosi SC, Lee MK, Wu M, Huynh H, Avila R, Pizzano C, Zamorano CA, Wu Y, Xavier R, Stanslaski M, Kang J, Thai S, Kim Y, Zhang J, Huang Y, Kozorovitskiy Y, Good CH, Banks AR, Rogers JA, Bruchas MR. An integrated microfluidic and fluorescence platform for probing in vivo neuropharmacology. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.14.594203. [PMID: 38798493 PMCID: PMC11118345 DOI: 10.1101/2024.05.14.594203] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
Neurotechnologies and genetic tools for dissecting neural circuit functions have advanced rapidly over the past decade, although the development of complementary pharmacological method-ologies has comparatively lagged. Understanding the precise pharmacological mechanisms of neuroactive compounds is critical for advancing basic neurobiology and neuropharmacology, as well as for developing more effective treatments for neurological and neuropsychiatric disorders. However, integrating modern tools for assessing neural activity in large-scale neural networks with spatially localized drug delivery remains a major challenge. Here, we present a dual microfluidic-photometry platform that enables simultaneous intracranial drug delivery with neural dynamics monitoring in the rodent brain. The integrated platform combines a wireless, battery-free, miniaturized fluidic microsystem with optical probes, allowing for spatially and temporally specific drug delivery while recording activity-dependent fluorescence using genetically encoded calcium indicators (GECIs), neurotransmitter sensors GRAB NE and GRAB DA , and neuropeptide sensors. We demonstrate the performance this platform for investigating neuropharmacological mechanisms in vivo and characterize its efficacy in probing precise mechanistic actions of neuroactive compounds across several rapidly evolving neuroscience domains.
Collapse
|
31
|
Gonzalez-Ramos A, Puigsasllosas-Pastor C, Arcas-Marquez A, Tornero D. Updated Toolbox for Assessing Neuronal Network Reconstruction after Cell Therapy. Bioengineering (Basel) 2024; 11:487. [PMID: 38790353 PMCID: PMC11118929 DOI: 10.3390/bioengineering11050487] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2024] [Revised: 05/02/2024] [Accepted: 05/10/2024] [Indexed: 05/26/2024] Open
Abstract
Cell therapy has proven to be a promising treatment for a range of neurological disorders, including Parkinson Disease, drug-resistant epilepsy, and stroke, by restoring function after brain damage. Nevertheless, evaluating the true effectiveness of these therapeutic interventions requires a deep understanding of the functional integration of grafted cells into existing neural networks. This review explores a powerful arsenal of molecular techniques revolutionizing our ability to unveil functional integration of grafted cells within the host brain. From precise manipulation of neuronal activity to pinpoint the functional contribution of transplanted cells by using opto- and chemo-genetics, to real-time monitoring of neuronal dynamics shedding light on functional connectivity within the reconstructed circuits by using genetically encoded (calcium) indicators in vivo. Finally, structural reconstruction and mapping communication pathways between grafted and host neurons can be achieved by monosynaptic tracing with viral vectors. The cutting-edge toolbox presented here holds immense promise for elucidating the impact of cell therapy on neural circuitry and guiding the development of more effective treatments for neurological disorders.
Collapse
Affiliation(s)
- Ana Gonzalez-Ramos
- Stanley Center for Psychiatric Research at Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Claudia Puigsasllosas-Pastor
- Laboratory of Neural Stem Cells and Brain Damage, Department of Biomedical Sciences, Institute of Neurosciences, University of Barcelona, 08036 Barcelona, Spain
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain
| | - Ainhoa Arcas-Marquez
- Laboratory of Neural Stem Cells and Brain Damage, Department of Biomedical Sciences, Institute of Neurosciences, University of Barcelona, 08036 Barcelona, Spain
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain
| | - Daniel Tornero
- Laboratory of Neural Stem Cells and Brain Damage, Department of Biomedical Sciences, Institute of Neurosciences, University of Barcelona, 08036 Barcelona, Spain
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), 28029 Madrid, Spain
| |
Collapse
|
32
|
Rae CD, Baur JA, Borges K, Dienel G, Díaz-García CM, Douglass SR, Drew K, Duarte JMN, Duran J, Kann O, Kristian T, Lee-Liu D, Lindquist BE, McNay EC, Robinson MB, Rothman DL, Rowlands BD, Ryan TA, Scafidi J, Scafidi S, Shuttleworth CW, Swanson RA, Uruk G, Vardjan N, Zorec R, McKenna MC. Brain energy metabolism: A roadmap for future research. J Neurochem 2024; 168:910-954. [PMID: 38183680 PMCID: PMC11102343 DOI: 10.1111/jnc.16032] [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: 05/27/2023] [Revised: 11/29/2023] [Accepted: 12/05/2023] [Indexed: 01/08/2024]
Abstract
Although we have learned much about how the brain fuels its functions over the last decades, there remains much still to discover in an organ that is so complex. This article lays out major gaps in our knowledge of interrelationships between brain metabolism and brain function, including biochemical, cellular, and subcellular aspects of functional metabolism and its imaging in adult brain, as well as during development, aging, and disease. The focus is on unknowns in metabolism of major brain substrates and associated transporters, the roles of insulin and of lipid droplets, the emerging role of metabolism in microglia, mysteries about the major brain cofactor and signaling molecule NAD+, as well as unsolved problems underlying brain metabolism in pathologies such as traumatic brain injury, epilepsy, and metabolic downregulation during hibernation. It describes our current level of understanding of these facets of brain energy metabolism as well as a roadmap for future research.
Collapse
Affiliation(s)
- Caroline D. Rae
- School of Psychology, The University of New South Wales, NSW 2052 & Neuroscience Research Australia, Randwick, New South Wales, Australia
| | - Joseph A. Baur
- Department of Physiology and Institute for Diabetes, Obesity and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Karin Borges
- School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, St Lucia, QLD, Australia
| | - Gerald Dienel
- Department of Neurology, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
- Department of Cell Biology and Physiology, University of New Mexico School of Medicine, Albuquerque, New Mexico, USA
| | - Carlos Manlio Díaz-García
- Department of Biochemistry and Molecular Biology, Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA
| | | | - Kelly Drew
- Center for Transformative Research in Metabolism, Institute of Arctic Biology, University of Alaska Fairbanks, Fairbanks, Alaska, USA
| | - João M. N. Duarte
- Department of Experimental Medical Science, Faculty of Medicine, Lund University, Lund, & Wallenberg Centre for Molecular Medicine, Lund University, Lund, Sweden
| | - Jordi Duran
- Institut Químic de Sarrià (IQS), Universitat Ramon Llull (URL), Barcelona, Spain
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology, Barcelona, Spain
| | - Oliver Kann
- Institute of Physiology and Pathophysiology, University of Heidelberg, D-69120; Interdisciplinary Center for Neurosciences (IZN), University of Heidelberg, Heidelberg, Germany
| | - Tibor Kristian
- Veterans Affairs Maryland Health Center System, Baltimore, Maryland, USA
- Department of Anesthesiology and the Center for Shock, Trauma, and Anesthesiology Research (S.T.A.R.), University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Dasfne Lee-Liu
- Facultad de Medicina y Ciencia, Universidad San Sebastián, Santiago, Región Metropolitana, Chile
| | - Britta E. Lindquist
- Department of Neurology, Division of Neurocritical Care, Gladstone Institute of Neurological Disease, University of California at San Francisco, San Francisco, California, USA
| | - Ewan C. McNay
- Behavioral Neuroscience, University at Albany, Albany, New York, USA
| | - Michael B. Robinson
- Departments of Pediatrics and System Pharmacology & Translational Therapeutics, Children’s Hospital of Philadelphia, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Douglas L. Rothman
- Magnetic Resonance Research Center and Departments of Radiology and Biomedical Engineering, Yale University, New Haven, Connecticut, USA
| | - Benjamin D. Rowlands
- School of Chemistry, Faculty of Science, The University of Sydney, Sydney, New South Wales, Australia
| | - Timothy A. Ryan
- Department of Biochemistry, Weill Cornell Medicine, New York, New York, USA
| | - Joseph Scafidi
- Department of Neurology, Kennedy Krieger Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Susanna Scafidi
- Anesthesiology & Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - C. William Shuttleworth
- Department of Neurosciences, University of New Mexico School of Medicine Albuquerque, Albuquerque, New Mexico, USA
| | - Raymond A. Swanson
- Department of Neurology, University of California, San Francisco, and San Francisco Veterans Affairs Medical Center, San Francisco, California, USA
| | - Gökhan Uruk
- Department of Neurology, University of California, San Francisco, and San Francisco Veterans Affairs Medical Center, San Francisco, California, USA
| | - Nina Vardjan
- Laboratory of Cell Engineering, Celica Biomedical, Ljubljana, Slovenia
- Laboratory of Neuroendocrinology—Molecular Cell Physiology, Institute of Pathophysiology, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Robert Zorec
- Laboratory of Cell Engineering, Celica Biomedical, Ljubljana, Slovenia
- Laboratory of Neuroendocrinology—Molecular Cell Physiology, Institute of Pathophysiology, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Mary C. McKenna
- Department of Pediatrics and Program in Neuroscience, University of Maryland School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
33
|
Suematsu N, Vazquez AL, Kozai TDY. Activation and depression of neural and hemodynamic responses induced by the intracortical microstimulation and visual stimulation in the mouse visual cortex. J Neural Eng 2024; 21:026033. [PMID: 38537268 PMCID: PMC11002944 DOI: 10.1088/1741-2552/ad3853] [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: 01/02/2024] [Revised: 02/28/2024] [Accepted: 03/27/2024] [Indexed: 04/09/2024]
Abstract
Objective. Intracortical microstimulation (ICMS) can be an effective method for restoring sensory perception in contemporary brain-machine interfaces. However, the mechanisms underlying better control of neuronal responses remain poorly understood, as well as the relationship between neuronal activity and other concomitant phenomena occurring around the stimulation site.Approach. Different microstimulation frequencies were investigatedin vivoon Thy1-GCaMP6s mice using widefield and two-photon imaging to evaluate the evoked excitatory neural responses across multiple spatial scales as well as the induced hemodynamic responses. Specifically, we quantified stimulation-induced neuronal activation and depression in the mouse visual cortex and measured hemodynamic oxyhemoglobin and deoxyhemoglobin signals using mesoscopic-scale widefield imaging.Main results. Our calcium imaging findings revealed a preference for lower-frequency stimulation in driving stronger neuronal activation. A depressive response following the neural activation preferred a slightly higher frequency stimulation compared to the activation. Hemodynamic signals exhibited a comparable spatial spread to neural calcium signals. Oxyhemoglobin concentration around the stimulation site remained elevated during the post-activation (depression) period. Somatic and neuropil calcium responses measured by two-photon microscopy showed similar dependence on stimulation parameters, although the magnitudes measured in soma was greater than in neuropil. Furthermore, higher-frequency stimulation induced a more pronounced activation in soma compared to neuropil, while depression was predominantly induced in soma irrespective of stimulation frequencies.Significance. These results suggest that the mechanism underlying depression differs from activation, requiring ample oxygen supply, and affecting neurons. Our findings provide a novel understanding of evoked excitatory neuronal activity induced by ICMS and offer insights into neuro-devices that utilize both activation and depression phenomena to achieve desired neural responses.
Collapse
Affiliation(s)
- Naofumi Suematsu
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, United States of America
| | - Alberto L Vazquez
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, United States of America
- Center for the Neural Basis of Cognition, University of Pittsburgh and Carnegie Mellon University, Pittsburgh, PA, United States of America
- Department of Radiology, University of Pittsburgh, Pittsburgh, PA, United States of America
- Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA, United States of America
| | - Takashi D Y Kozai
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, United States of America
- Center for the Neural Basis of Cognition, University of Pittsburgh and Carnegie Mellon University, Pittsburgh, PA, United States of America
- Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA, United States of America
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, United States of America
- NeuroTech Center, University of Pittsburgh Brain Institute, Pittsburgh, PA, United States of America
| |
Collapse
|
34
|
Yu Y, Adsit LM, Smith IT. Comprehensive software suite for functional analysis and synaptic input mapping of dendritic spines imaged in vivo. NEUROPHOTONICS 2024; 11:024307. [PMID: 38628980 PMCID: PMC11021036 DOI: 10.1117/1.nph.11.2.024307] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/17/2023] [Revised: 03/21/2024] [Accepted: 03/21/2024] [Indexed: 04/19/2024]
Abstract
Significance Advances in genetically encoded sensors and two-photon imaging have unlocked functional imaging at the level of single dendritic spines. Synaptic activity can be measured in real time in awake animals. However, tools are needed to facilitate the analysis of the large datasets acquired by the approach. Commonly available software suites for imaging calcium transients in cell bodies are ill-suited for spine imaging as dendritic spines have structural characteristics distinct from those of the cell bodies. We present an automated tuning analysis tool (AUTOTUNE), which provides analysis routines specifically developed for the extraction and analysis of signals from subcellular compartments, including dendritic subregions and spines. Aim Although the acquisition of in vivo functional synaptic imaging data is increasingly accessible, a hurdle remains in the computation-heavy analyses of the acquired data. The aim of this study is to overcome this barrier by offering a comprehensive software suite with a user-friendly interface for easy access to nonprogrammers. Approach We demonstrate the utility and effectiveness of our software with demo analyses of dendritic imaging data acquired from layer 2/3 pyramidal neurons in mouse V1 in vivo. A user manual and demo datasets are also provided. Results AUTOTUNE provides a robust workflow for analyzing functional imaging data from neuronal dendrites. Features include source image registration, segmentation of regions-of-interest and detection of structural turnover, fluorescence transient extraction and smoothing, subtraction of signals from putative backpropagating action potentials, and stimulus and behavioral parameter response tuning analyses. Conclusions AUTOTUNE is open-source and extendable for diverse functional synaptic imaging experiments. The ease of functional characterization of dendritic spine activity provided by our software can accelerate new functional studies that complement decades of morphological studies of dendrites, and further expand our understanding of neural circuits in health and in disease.
Collapse
Affiliation(s)
- Yiyi Yu
- University of California, Santa Barbara, Department of Electrical and Computer Engineering, Santa Barbara, California, United States
| | - Liam M. Adsit
- University of California, Santa Barbara, Department of Molecular, Cellular and Developmental Biology, Santa Barbara, California, United States
| | - Ikuko T. Smith
- University of California, Santa Barbara, Department of Molecular, Cellular and Developmental Biology, Santa Barbara, California, United States
- University of California, Santa Barbara, Neuroscience Research Institute, Santa Barbara, California, United States
- University of California, Santa Barbara, Department of Psychological and Brain Sciences, Santa Barbara, California, United States
| |
Collapse
|
35
|
Dunham KE, Venton BJ. Electrochemical and biosensor techniques to monitor neurotransmitter changes with depression. Anal Bioanal Chem 2024; 416:2301-2318. [PMID: 38289354 PMCID: PMC10950978 DOI: 10.1007/s00216-024-05136-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Revised: 12/29/2023] [Accepted: 01/09/2024] [Indexed: 03/21/2024]
Abstract
Depression is a common mental illness. However, its current treatments, like selective serotonin reuptake inhibitors (SSRIs) and micro-dosing ketamine, are extremely variable between patients and not well understood. Three neurotransmitters: serotonin, histamine, and glutamate, have been proposed to be key mediators of depression. This review focuses on analytical methods to quantify these neurotransmitters to better understand neurological mechanisms of depression and how they are altered during treatment. To quantitatively measure serotonin and histamine, electrochemical techniques such as chronoamperometry and fast-scan cyclic voltammetry (FSCV) have been improved to study how specific molecular targets, like transporters and receptors, change with antidepressants and inflammation. Specifically, these studies show that different SSRIs have unique effects on serotonin reuptake and release. Histamine is normally elevated during stress, and a new inflammation hypothesis of depression links histamine and cytokine release. Electrochemical measurements revealed that stress increases histamine, decreases serotonin, and leads to changes in cytokines, like interleukin-6. Biosensors can also measure non-electroactive neurotransmitters, including glutamate and cytokines. In particular, new genetic sensors have shown how glutamate changes with chronic stress, as well as with ketamine treatment. These techniques have been used to characterize how ketamine changes glutamate and serotonin, and to understand how it is different from SSRIs. This review briefly outlines how these electrochemical techniques work, but primarily highlights how they have been used to understand the mechanisms of depression. Future studies should explore multiplexing techniques and personalized medicine using biomarkers in order to investigate multi-analyte changes to antidepressants.
Collapse
Affiliation(s)
- Kelly E Dunham
- Department of Chemistry, University of Virginia, Charlottesville, VA, 22904, USA
| | - B Jill Venton
- Department of Chemistry, University of Virginia, Charlottesville, VA, 22904, USA.
| |
Collapse
|
36
|
Wu Z, Kusick GF, Berns MMM, Raychaudhuri S, Itoh K, Walter AM, Chapman ER, Watanabe S. Synaptotagmin 7 docks synaptic vesicles to support facilitation and Doc2α-triggered asynchronous release. eLife 2024; 12:RP90632. [PMID: 38536730 PMCID: PMC10972563 DOI: 10.7554/elife.90632] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/11/2024] Open
Abstract
Despite decades of intense study, the molecular basis of asynchronous neurotransmitter release remains enigmatic. Synaptotagmin (syt) 7 and Doc2 have both been proposed as Ca2+ sensors that trigger this mode of exocytosis, but conflicting findings have led to controversy. Here, we demonstrate that at excitatory mouse hippocampal synapses, Doc2α is the major Ca2+ sensor for asynchronous release, while syt7 supports this process through activity-dependent docking of synaptic vesicles. In synapses lacking Doc2α, asynchronous release after single action potentials is strongly reduced, while deleting syt7 has no effect. However, in the absence of syt7, docked vesicles cannot be replenished on millisecond timescales. Consequently, both synchronous and asynchronous release depress from the second pulse onward during repetitive activity. By contrast, synapses lacking Doc2α have normal activity-dependent docking, but continue to exhibit decreased asynchronous release after multiple stimuli. Moreover, disruption of both Ca2+ sensors is non-additive. These findings result in a new model whereby syt7 drives activity-dependent docking, thus providing synaptic vesicles for synchronous (syt1) and asynchronous (Doc2 and other unidentified sensors) release during ongoing transmission.
Collapse
Affiliation(s)
- Zhenyong Wu
- Department of Neuroscience, University of Wisconsin-MadisonMadisonUnited States
- Howard Hughes Medical InstituteMadisonUnited States
| | - Grant F Kusick
- Department of Cell Biology, Johns Hopkins University, School of MedicineBaltimoreUnited States
- Biochemistry, Cellular and Molecular Biology Graduate Program, Johns Hopkins University School of MedicineBaltimoreUnited States
| | - Manon MM Berns
- Department of Neuroscience, University of CopenhagenCopenhagenDenmark
| | - Sumana Raychaudhuri
- Department of Cell Biology, Johns Hopkins University, School of MedicineBaltimoreUnited States
| | - Kie Itoh
- Department of Cell Biology, Johns Hopkins University, School of MedicineBaltimoreUnited States
| | - Alexander M Walter
- Department of Neuroscience, University of CopenhagenCopenhagenDenmark
- Molecular and Theoretical Neuroscience, Leibniz-Institut für Molekulare Pharmakologie, FMP im CharitéCrossOverBerlinGermany
| | - Edwin R Chapman
- Department of Neuroscience, University of Wisconsin-MadisonMadisonUnited States
- Howard Hughes Medical InstituteMadisonUnited States
| | - Shigeki Watanabe
- Department of Cell Biology, Johns Hopkins University, School of MedicineBaltimoreUnited States
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of MedicineBaltimoreUnited States
| |
Collapse
|
37
|
Amo R, Uchida N, Watabe-Uchida M. Glutamate inputs send prediction error of reward, but not negative value of aversive stimuli, to dopamine neurons. Neuron 2024; 112:1001-1019.e6. [PMID: 38278147 PMCID: PMC10957320 DOI: 10.1016/j.neuron.2023.12.019] [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: 03/26/2023] [Revised: 11/10/2023] [Accepted: 12/21/2023] [Indexed: 01/28/2024]
Abstract
Midbrain dopamine neurons are thought to signal reward prediction errors (RPEs), but the mechanisms underlying RPE computation, particularly the contributions of different neurotransmitters, remain poorly understood. Here, we used a genetically encoded glutamate sensor to examine the pattern of glutamate inputs to dopamine neurons in mice. We found that glutamate inputs exhibit virtually all of the characteristics of RPE rather than conveying a specific component of RPE computation, such as reward or expectation. Notably, whereas glutamate inputs were transiently inhibited by reward omission, they were excited by aversive stimuli. Opioid analgesics altered dopamine negative responses to aversive stimuli into more positive responses, whereas excitatory responses of glutamate inputs remained unchanged. Our findings uncover previously unknown synaptic mechanisms underlying RPE computations; dopamine responses are shaped by both synergistic and competitive interactions between glutamatergic and GABAergic inputs to dopamine neurons depending on valences, with competitive interactions playing a role in responses to aversive stimuli.
Collapse
Affiliation(s)
- Ryunosuke Amo
- Department of Molecular and Cellular Biology, Center for Brain Science, Harvard University, Cambridge, MA 02138, USA
| | - Naoshige Uchida
- Department of Molecular and Cellular Biology, Center for Brain Science, Harvard University, Cambridge, MA 02138, USA
| | - Mitsuko Watabe-Uchida
- Department of Molecular and Cellular Biology, Center for Brain Science, Harvard University, Cambridge, MA 02138, USA.
| |
Collapse
|
38
|
Hedrick NG, Wright WJ, Komiyama T. Local and global predictors of synapse elimination during motor learning. SCIENCE ADVANCES 2024; 10:eadk0540. [PMID: 38489360 PMCID: PMC10942101 DOI: 10.1126/sciadv.adk0540] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Accepted: 02/12/2024] [Indexed: 03/17/2024]
Abstract
During learning, synaptic connections between excitatory neurons in the brain display considerable dynamism, with new connections being added and old connections eliminated. Synapse elimination offers an opportunity to understand the features of synapses that the brain deems dispensable. However, with limited observations of synaptic activity and plasticity in vivo, the features of synapses subjected to elimination remain poorly understood. Here, we examined the functional basis of synapse elimination in the apical dendrites of L2/3 neurons in the primary motor cortex throughout motor learning. We found no evidence that synapse elimination is facilitated by a lack of activity or other local forms of plasticity. Instead, eliminated synapses display asynchronous activity with nearby synapses, suggesting that functional synaptic clustering is a critical component of synapse survival. In addition, eliminated synapses show delayed activity timing with respect to postsynaptic output. Thus, synaptic inputs that fail to be co-active with their neighboring synapses or are mistimed with neuronal output are targeted for elimination.
Collapse
Affiliation(s)
- Nathan G. Hedrick
- Department of Neurobiology, University of California San Diego, La Jolla, CA, USA
- Center for Neural Circuits and Behavior, University of California San Diego, La Jolla, CA, USA
- Department of Neurosciences, University of California San Diego, La Jolla, CA, USA
- Halıcıoğlu Data Science Institute, University of California San Diego, La Jolla, CA, USA
| | - William J. Wright
- Department of Neurobiology, University of California San Diego, La Jolla, CA, USA
- Center for Neural Circuits and Behavior, University of California San Diego, La Jolla, CA, USA
- Department of Neurosciences, University of California San Diego, La Jolla, CA, USA
- Halıcıoğlu Data Science Institute, University of California San Diego, La Jolla, CA, USA
| | - Takaki Komiyama
- Department of Neurobiology, University of California San Diego, La Jolla, CA, USA
- Center for Neural Circuits and Behavior, University of California San Diego, La Jolla, CA, USA
- Department of Neurosciences, University of California San Diego, La Jolla, CA, USA
- Halıcıoğlu Data Science Institute, University of California San Diego, La Jolla, CA, USA
| |
Collapse
|
39
|
Simpson EH, Akam T, Patriarchi T, Blanco-Pozo M, Burgeno LM, Mohebi A, Cragg SJ, Walton ME. Lights, fiber, action! A primer on in vivo fiber photometry. Neuron 2024; 112:718-739. [PMID: 38103545 PMCID: PMC10939905 DOI: 10.1016/j.neuron.2023.11.016] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2023] [Revised: 10/16/2023] [Accepted: 11/15/2023] [Indexed: 12/19/2023]
Abstract
Fiber photometry is a key technique for characterizing brain-behavior relationships in vivo. Initially, it was primarily used to report calcium dynamics as a proxy for neural activity via genetically encoded indicators. This generated new insights into brain functions including movement, memory, and motivation at the level of defined circuits and cell types. Recently, the opportunity for discovery with fiber photometry has exploded with the development of an extensive range of fluorescent sensors for biomolecules including neuromodulators and peptides that were previously inaccessible in vivo. This critical advance, combined with the new availability of affordable "plug-and-play" recording systems, has made monitoring molecules with high spatiotemporal precision during behavior highly accessible. However, while opening exciting new avenues for research, the rapid expansion in fiber photometry applications has occurred without coordination or consensus on best practices. Here, we provide a comprehensive guide to help end-users execute, analyze, and suitably interpret fiber photometry studies.
Collapse
Affiliation(s)
- Eleanor H Simpson
- Department of Psychiatry, Columbia University Medical Center, New York, NY, USA; New York State Psychiatric Institute, New York, NY, USA.
| | - Thomas Akam
- Department of Experimental Psychology, University of Oxford, Oxford, UK; Wellcome Centre for Integrative Neuroimaging, University of Oxford, Oxford, UK.
| | - Tommaso Patriarchi
- Institute of Pharmacology and Toxicology, University of Zürich, Zürich, Switzerland; Neuroscience Center Zürich, University and ETH Zürich, Zürich, Switzerland.
| | - Marta Blanco-Pozo
- Department of Experimental Psychology, University of Oxford, Oxford, UK; Wellcome Centre for Integrative Neuroimaging, University of Oxford, Oxford, UK
| | - Lauren M Burgeno
- Department of Experimental Psychology, University of Oxford, Oxford, UK; Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK
| | - Ali Mohebi
- Department of Neurology, University of California, San Francisco, San Francisco, CA, USA
| | - Stephanie J Cragg
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK; Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| | - Mark E Walton
- Department of Experimental Psychology, University of Oxford, Oxford, UK; Wellcome Centre for Integrative Neuroimaging, University of Oxford, Oxford, UK
| |
Collapse
|
40
|
Otanuly M, Kubitschke M, Masseck OA. A Bright Future? A Perspective on Class C GPCR Based Genetically Encoded Biosensors. ACS Chem Neurosci 2024; 15:889-897. [PMID: 38380648 PMCID: PMC10921406 DOI: 10.1021/acschemneuro.3c00854] [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: 01/02/2024] [Revised: 02/11/2024] [Accepted: 02/14/2024] [Indexed: 02/22/2024] Open
Abstract
One of the major challenges in molecular neuroscience today is to accurately monitor neurotransmitters, neuromodulators, peptides, and various other biomolecules in the brain with high temporal and spatial resolution. Only a comprehensive understanding of neuromodulator dynamics, their release probability, and spatial distribution will unravel their ultimate role in cognition and behavior. This Perspective offers an overview of potential design strategies for class C GPCR-based biosensors. It briefly highlights current applications of GPCR-based biosensors, with a primary focus on class C GPCRs and their unique structural characteristics compared with other GPCR subfamilies. The discussion offers insights into plausible future design approaches for biosensor development targeting members of this specific GPCR subfamily. It is important to note that, at this stage, we are contemplating possibilities rather than presenting a concrete guide, as the pipeline is still under development.
Collapse
Affiliation(s)
- Margulan Otanuly
- Synthetische Biologie, Universität Bremen, Bremen 28359, Germany
| | | | | |
Collapse
|
41
|
Shimoda Y, Leite M, Graham RT, Marvin JS, Hasseman J, Kolb I, Looger LL, Magloire V, Kullmann DM. Extracellular glutamate and GABA transients at the transition from interictal spiking to seizures. Brain 2024; 147:1011-1024. [PMID: 37787057 PMCID: PMC10907087 DOI: 10.1093/brain/awad336] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Revised: 08/29/2023] [Accepted: 09/16/2023] [Indexed: 10/04/2023] Open
Abstract
Focal epilepsy is associated with intermittent brief population discharges (interictal spikes), which resemble sentinel spikes that often occur at the onset of seizures. Why interictal spikes self-terminate whilst seizures persist and propagate is incompletely understood. We used fluorescent glutamate and GABA sensors in an awake rodent model of neocortical seizures to resolve the spatiotemporal evolution of both neurotransmitters in the extracellular space. Interictal spikes were accompanied by brief glutamate transients which were maximal at the initiation site and rapidly propagated centrifugally. GABA transients lasted longer than glutamate transients and were maximal ∼1.5 mm from the focus where they propagated centripetally. Prior to seizure initiation GABA transients were attenuated, whilst glutamate transients increased, consistent with a progressive failure of local inhibitory restraint. As seizures increased in frequency, there was a gradual increase in the spatial extent of spike-associated glutamate transients associated with interictal spikes. Neurotransmitter imaging thus reveals a progressive collapse of an annulus of feed-forward GABA release, allowing seizures to escape from local inhibitory restraint.
Collapse
Affiliation(s)
- Yoshiteru Shimoda
- Department of Clinical & Experimental Epilepsy, UCL Queen Square Institute of Neurology, University College London, London WC1N 3BG, UK
| | - Marco Leite
- Department of Clinical & Experimental Epilepsy, UCL Queen Square Institute of Neurology, University College London, London WC1N 3BG, UK
| | - Robert T Graham
- Department of Clinical & Experimental Epilepsy, UCL Queen Square Institute of Neurology, University College London, London WC1N 3BG, UK
| | - Jonathan S Marvin
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA 20147, USA
| | - Jeremy Hasseman
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA 20147, USA
| | - Ilya Kolb
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA 20147, USA
| | - Loren L Looger
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA 20147, USA
| | - Vincent Magloire
- Department of Clinical & Experimental Epilepsy, UCL Queen Square Institute of Neurology, University College London, London WC1N 3BG, UK
| | - Dimitri M Kullmann
- Department of Clinical & Experimental Epilepsy, UCL Queen Square Institute of Neurology, University College London, London WC1N 3BG, UK
| |
Collapse
|
42
|
Davidsen K, Marvin JS, Aggarwal A, Brown TA, Sullivan LB. An engineered biosensor enables dynamic aspartate measurements in living cells. eLife 2024; 12:RP90024. [PMID: 38393319 PMCID: PMC10942590 DOI: 10.7554/elife.90024] [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: 02/25/2024] Open
Abstract
Intracellular levels of the amino acid aspartate are responsive to changes in metabolism in mammalian cells and can correspondingly alter cell function, highlighting the need for robust tools to measure aspartate abundance. However, comprehensive understanding of aspartate metabolism has been limited by the throughput, cost, and static nature of the mass spectrometry (MS)-based measurements that are typically employed to measure aspartate levels. To address these issues, we have developed a green fluorescent protein (GFP)-based sensor of aspartate (jAspSnFR3), where the fluorescence intensity corresponds to aspartate concentration. As a purified protein, the sensor has a 20-fold increase in fluorescence upon aspartate saturation, with dose-dependent fluorescence changes covering a physiologically relevant aspartate concentration range and no significant off target binding. Expressed in mammalian cell lines, sensor intensity correlated with aspartate levels measured by MS and could resolve temporal changes in intracellular aspartate from genetic, pharmacological, and nutritional manipulations. These data demonstrate the utility of jAspSnFR3 and highlight the opportunities it provides for temporally resolved and high-throughput applications of variables that affect aspartate levels.
Collapse
Affiliation(s)
- Kristian Davidsen
- Human Biology Division, Fred Hutchinson Cancer CenterSeattleUnited States
- Molecular and Cellular Biology Program, University of WashingtonSeattleUnited States
| | - Jonathan S Marvin
- Howard Hughes Medical Institute (HHMI), Janelia Research CampusAshburnUnited States
| | - Abhi Aggarwal
- Howard Hughes Medical Institute (HHMI), Janelia Research CampusAshburnUnited States
| | - Timothy A Brown
- Howard Hughes Medical Institute (HHMI), Janelia Research CampusAshburnUnited States
| | - Lucas B Sullivan
- Human Biology Division, Fred Hutchinson Cancer CenterSeattleUnited States
| |
Collapse
|
43
|
Amo R. Prediction error in dopamine neurons during associative learning. Neurosci Res 2024; 199:12-20. [PMID: 37451506 DOI: 10.1016/j.neures.2023.07.003] [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: 04/03/2023] [Revised: 06/18/2023] [Accepted: 07/07/2023] [Indexed: 07/18/2023]
Abstract
Dopamine neurons have long been thought to facilitate learning by broadcasting reward prediction error (RPE), a teaching signal used in machine learning, but more recent work has advanced alternative models of dopamine's computational role. Here, I revisit this critical issue and review new experimental evidences that tighten the link between dopamine activity and RPE. First, I introduce the recent observation of a gradual backward shift of dopamine activity that had eluded researchers for over a decade. I also discuss several other findings, such as dopamine ramping, that were initially interpreted to conflict but later found to be consistent with RPE. These findings improve our understanding of neural computation in dopamine neurons.
Collapse
Affiliation(s)
- Ryunosuke Amo
- Department of Molecular and Cellular Biology, Center for Brain Science, Harvard University, Cambridge, MA 02138, USA.
| |
Collapse
|
44
|
Kubitschke M, Masseck OA. Illuminating the brain-genetically encoded single wavelength fluorescent biosensors to unravel neurotransmitter dynamics. Biol Chem 2024; 405:55-65. [PMID: 37246368 DOI: 10.1515/hsz-2023-0175] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Accepted: 05/15/2023] [Indexed: 05/30/2023]
Abstract
Understanding how neuronal networks generate complex behavior is one of the major goals of Neuroscience. Neurotransmitter and Neuromodulators are crucial for information flow between neurons and understanding their dynamics is the key to unravel their role in behavior. To understand how the brain transmits information and how brain states arise, it is essential to visualize the dynamics of neurotransmitters, neuromodulators and neurochemicals. In the last five years, an increasing number of single-wavelength biosensors either based on periplasmic binding proteins (PBPs) or on G-protein-coupled receptors (GPCR) have been published that are able to detect neurotransmitter release in vitro and in vivo with high spatial and temporal resolution. Here we review and discuss recent progress in the development of these sensors, their limitations and future directions.
Collapse
|
45
|
Guhathakurta D, Petrušková A, Akdaş EY, Perelló-Amorós B, Frischknecht R, Anni D, Weiss EM, Walter M, Fejtová A. Hydroxynorketamine, but not ketamine, acts via α7 nicotinic acetylcholine receptor to control presynaptic function and gene expression. Transl Psychiatry 2024; 14:47. [PMID: 38253622 PMCID: PMC10803733 DOI: 10.1038/s41398-024-02744-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Revised: 12/21/2023] [Accepted: 01/08/2024] [Indexed: 01/24/2024] Open
Abstract
Ketamine is clinically used fast-acting antidepressant. Its metabolite hydroxynorketamine (HNK) shows a robust antidepressant effect in animal studies. It is unclear, how these chemically distinct compounds converge on similar neuronal effects. While KET acts mostly as N-methyl-d-aspartate receptor (NMDAR) antagonist, the molecular target of HNK remains enigmatic. Here, we show that KET and HNK converge on rapid inhibition of glutamate release by reducing the release competence of synaptic vesicles and induce nuclear translocation of pCREB that controls expression of neuroplasticity genes connected to KET- and HNK-mediated antidepressant action. Ro25-6981, a selective antagonist of GluN2B, mimics effect of KET indicating that GluN2B-containing NMDAR might mediate the presynaptic effect of KET. Selective antagonist of α7 nicotinic acetylcholine receptors (α7nAChRs) or genetic deletion of Chrna7, its pore-forming subunit, fully abolishes HNK-induced synaptic and nuclear regulations, but leaves KET-dependent cellular effects unaffected. Thus, KET or HNK-induced modulation of synaptic transmission and nuclear translocation of pCREB can be mediated by selective signaling via NMDAR or α7nAChRs, respectively. Due to the rapid metabolism of KET to HNK, it is conceivable that subsequent modulation of glutamatergic and cholinergic neurotransmission affects circuits in a cell-type-specific manner and contributes to the therapeutic potency of KET. This finding promotes further exploration of new combined medications for mood disorders.
Collapse
Affiliation(s)
- Debarpan Guhathakurta
- Department of Psychiatry and Psychotherapy, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Aneta Petrušková
- Department of Psychiatry and Psychotherapy, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
- National Institute of Mental Health, Klecany, Czech Republic
- Third Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Enes Yağız Akdaş
- Department of Psychiatry and Psychotherapy, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Bartomeu Perelló-Amorós
- Department of Biology, Animal Physiology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Renato Frischknecht
- Department of Biology, Animal Physiology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Daniela Anni
- Department of Psychiatry and Psychotherapy, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Eva-Maria Weiss
- Department of Psychiatry and Psychotherapy, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Martin Walter
- Department of Psychiatry and Psychotherapy, Universitätsklinikum Jena, Jena, Germany
| | - Anna Fejtová
- Department of Psychiatry and Psychotherapy, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany.
| |
Collapse
|
46
|
Phan TV, Mattingly HH, Vo L, Marvin JS, Looger LL, Emonet T. Direct measurement of dynamic attractant gradients reveals breakdown of the Patlak-Keller-Segel chemotaxis model. Proc Natl Acad Sci U S A 2024; 121:e2309251121. [PMID: 38194458 PMCID: PMC10801886 DOI: 10.1073/pnas.2309251121] [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: 12/07/2023] [Indexed: 01/11/2024] Open
Abstract
Chemotactic bacteria not only navigate chemical gradients, but also shape their environments by consuming and secreting attractants. Investigating how these processes influence the dynamics of bacterial populations has been challenging because of a lack of experimental methods for measuring spatial profiles of chemoattractants in real time. Here, we use a fluorescent sensor for aspartate to directly measure bacterially generated chemoattractant gradients during collective migration. Our measurements show that the standard Patlak-Keller-Segel model for collective chemotactic bacterial migration breaks down at high cell densities. To address this, we propose modifications to the model that consider the impact of cell density on bacterial chemotaxis and attractant consumption. With these changes, the model explains our experimental data across all cell densities, offering insight into chemotactic dynamics. Our findings highlight the significance of considering cell density effects on bacterial behavior, and the potential for fluorescent metabolite sensors to shed light on the complex emergent dynamics of bacterial communities.
Collapse
Affiliation(s)
- Trung V. Phan
- Department of Molecular, Cellular, and Developmental Biology, Yale University, New Haven, CT06511
- Quantitative Biology Institute, Yale University, New Haven, CT06511
| | | | - Lam Vo
- Department of Molecular, Cellular, and Developmental Biology, Yale University, New Haven, CT06511
- Quantitative Biology Institute, Yale University, New Haven, CT06511
| | - Jonathan S. Marvin
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA20147
| | - Loren L. Looger
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA20147
- HHMI, University of California, San Diego, CA92093
- Department of Neurosciences, University of California, San Diego, CA92093
| | - Thierry Emonet
- Department of Molecular, Cellular, and Developmental Biology, Yale University, New Haven, CT06511
- Quantitative Biology Institute, Yale University, New Haven, CT06511
- Department of Physics, Yale University, New Haven, CT06511
| |
Collapse
|
47
|
Suematsu N, Vazquez AL, Kozai TD. Activation and depression of neural and hemodynamic responses induced by the intracortical microstimulation and visual stimulation in the mouse visual cortex. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.01.573814. [PMID: 38260671 PMCID: PMC10802282 DOI: 10.1101/2024.01.01.573814] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2024]
Abstract
Objective . Intracortical microstimulation can be an effective method for restoring sensory perception in contemporary brain-machine interfaces. However, the mechanisms underlying better control of neuronal responses remain poorly understood, as well as the relationship between neuronal activity and other concomitant phenomena occurring around the stimulation site. Approach . Different microstimulation frequencies were investigated in vivo on Thy1-GCaMP6s mice using widefield and two-photon imaging to evaluate the evoked excitatory neural responses across multiple spatial scales as well as the induced hemodynamic responses. Specifically, we quantified stimulation-induced neuronal activation and depression in the mouse visual cortex and measured hemodynamic oxyhemoglobin and deoxyhemoglobin signals using mesoscopic-scale widefield imaging. Main results . Our calcium imaging findings revealed a preference for lower-frequency stimulation in driving stronger neuronal activation. A depressive response following the neural activation preferred a slightly higher frequency stimulation compared to the activation. Hemodynamic signals exhibited a comparable spatial spread to neural calcium signals. Oxyhemoglobin concentration around the stimulation site remained elevated during the post-activation (depression) period. Somatic and neuropil calcium responses measured by two-photon microscopy showed similar dependence on stimulation parameters, although the magnitudes measured in soma was greater than in neuropil. Furthermore, higher-frequency stimulation induced a more pronounced activation in soma compared to neuropil, while depression was predominantly induced in soma irrespective of stimulation frequencies. Significance . These results suggest that the mechanism underlying depression differs from activation, requiring ample oxygen supply, and affecting neurons. Our findings provide a novel understanding of evoked excitatory neuronal activity induced by intracortical microstimulation and offer insights into neuro-devices that utilize both activation and depression phenomena to achieve desired neural responses.
Collapse
|
48
|
Rusakov DA. Avoiding bias in fluorescence sensor readout. Nat Rev Neurosci 2024; 25:1-2. [PMID: 37950075 DOI: 10.1038/s41583-023-00768-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2023]
Affiliation(s)
- Dmitri A Rusakov
- UCL Queen Square Institute of Neurology, University College London, London, UK.
| |
Collapse
|
49
|
Miyashita T, Murakami K, Kikuchi E, Ofusa K, Mikami K, Endo K, Miyaji T, Moriyama S, Konno K, Muratani H, Moriyama Y, Watanabe M, Horiuchi J, Saitoe M. Glia transmit negative valence information during aversive learning in Drosophila. Science 2023; 382:eadf7429. [PMID: 38127757 DOI: 10.1126/science.adf7429] [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: 11/10/2022] [Accepted: 10/20/2023] [Indexed: 12/23/2023]
Abstract
During Drosophila aversive olfactory conditioning, aversive shock information needs to be transmitted to the mushroom bodies (MBs) to associate with odor information. We report that aversive information is transmitted by ensheathing glia (EG) that surround the MBs. Shock induces vesicular exocytosis of glutamate from EG. Blocking exocytosis impairs aversive learning, whereas activation of EG can replace aversive stimuli during conditioning. Glutamate released from EG binds to N-methyl-d-aspartate receptors in the MBs, but because of Mg2+ block, Ca2+ influx occurs only when flies are simultaneously exposed to an odor. Vesicular exocytosis from EG also induces shock-associated dopamine release, which plays a role in preventing formation of inappropriate associations. These results demonstrate that vesicular glutamate released from EG transmits negative valence information required for associative learning.
Collapse
Affiliation(s)
- Tomoyuki Miyashita
- Learning and Memory Project, Tokyo Metropolitan Institute of Medical Science, Tokyo 156-8506, Japan
| | - Kanako Murakami
- Learning and Memory Project, Tokyo Metropolitan Institute of Medical Science, Tokyo 156-8506, Japan
- Department of Biological Science, Graduate School of Science, Tokyo Metropolitan University, Tokyo 192-0397, Japan
| | - Emi Kikuchi
- Learning and Memory Project, Tokyo Metropolitan Institute of Medical Science, Tokyo 156-8506, Japan
| | - Kyouko Ofusa
- Learning and Memory Project, Tokyo Metropolitan Institute of Medical Science, Tokyo 156-8506, Japan
| | - Kyohei Mikami
- Center for Basic Technology Research, Tokyo Metropolitan Institute of Medical Science, Tokyo 156-8506, Japan
| | - Kentaro Endo
- Center for Basic Technology Research, Tokyo Metropolitan Institute of Medical Science, Tokyo 156-8506, Japan
| | - Takaaki Miyaji
- Department of Molecular Membrane Biology, Faculty of Pharmaceutical Sciences, Okayama University, Okayama 700-8530, Japan
- Department of Genomics and Proteomics, Advanced Science Research Center, Okayama University, Okayama 700-8530, Japan
| | - Sawako Moriyama
- Division of Endocrinology and Metabolism, Department of Internal Medicine, School of Medicine, Kurume University, Fukuoka 830-0011, Japan
| | - Kotaro Konno
- Department of Anatomy, Faculty of Medicine, Hokkaido University, Hokkaido 060-8368, Japan
| | - Hinako Muratani
- Learning and Memory Project, Tokyo Metropolitan Institute of Medical Science, Tokyo 156-8506, Japan
- Department of Engineering Science, Graduate School of Informatics and Engineering, The University of Electro-Communications, Tokyo 182-8585, Japan
| | - Yoshinori Moriyama
- Division of Endocrinology and Metabolism, Department of Internal Medicine, School of Medicine, Kurume University, Fukuoka 830-0011, Japan
| | - Masahiko Watanabe
- Department of Anatomy, Faculty of Medicine, Hokkaido University, Hokkaido 060-8368, Japan
| | - Junjiro Horiuchi
- Center for Basic Technology Research, Tokyo Metropolitan Institute of Medical Science, Tokyo 156-8506, Japan
| | - Minoru Saitoe
- Learning and Memory Project, Tokyo Metropolitan Institute of Medical Science, Tokyo 156-8506, Japan
- Center for Basic Technology Research, Tokyo Metropolitan Institute of Medical Science, Tokyo 156-8506, Japan
| |
Collapse
|
50
|
Zhou ZC, Gordon-Fennell A, Piantadosi SC, Ji N, Smith SL, Bruchas MR, Stuber GD. Deep-brain optical recording of neural dynamics during behavior. Neuron 2023; 111:3716-3738. [PMID: 37804833 PMCID: PMC10843303 DOI: 10.1016/j.neuron.2023.09.006] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Revised: 08/24/2023] [Accepted: 09/06/2023] [Indexed: 10/09/2023]
Abstract
In vivo fluorescence recording techniques have produced landmark discoveries in neuroscience, providing insight into how single cell and circuit-level computations mediate sensory processing and generate complex behaviors. While much attention has been given to recording from cortical brain regions, deep-brain fluorescence recording is more complex because it requires additional measures to gain optical access to harder to reach brain nuclei. Here we discuss detailed considerations and tradeoffs regarding deep-brain fluorescence recording techniques and provide a comprehensive guide for all major steps involved, from project planning to data analysis. The goal is to impart guidance for new and experienced investigators seeking to use in vivo deep fluorescence optical recordings in awake, behaving rodent models.
Collapse
Affiliation(s)
- Zhe Charles Zhou
- Department of Anesthesiology and Pain Medicine, University of Washington, Seattle, WA 98195, USA; Center for Neurobiology of Addiction, Pain, and Emotion, University of Washington, Seattle, WA 98195, USA
| | - Adam Gordon-Fennell
- Department of Anesthesiology and Pain Medicine, University of Washington, Seattle, WA 98195, USA; Center for Neurobiology of Addiction, Pain, and Emotion, University of Washington, Seattle, WA 98195, USA
| | - Sean C Piantadosi
- Department of Anesthesiology and Pain Medicine, University of Washington, Seattle, WA 98195, USA; Center for Neurobiology of Addiction, Pain, and Emotion, University of Washington, Seattle, WA 98195, USA
| | - Na Ji
- Department of Physics, University of California, Berkeley, Berkeley, CA 94720, USA; Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA; Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, CA 94720, USA; Molecular Biophysics and Integrated Bioimaging Division, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA
| | - Spencer LaVere Smith
- Department of Electrical and Computer Engineering, University of California Santa Barbara, Santa Barbara, CA 93106, USA
| | - Michael R Bruchas
- Department of Anesthesiology and Pain Medicine, University of Washington, Seattle, WA 98195, USA; Center for Neurobiology of Addiction, Pain, and Emotion, University of Washington, Seattle, WA 98195, USA; Department of Pharmacology, University of Washington, Seattle, WA 98195, USA; Department of Bioengineering, University of Washington, Seattle, WA 98195, USA.
| | - Garret D Stuber
- Department of Anesthesiology and Pain Medicine, University of Washington, Seattle, WA 98195, USA; Center for Neurobiology of Addiction, Pain, and Emotion, University of Washington, Seattle, WA 98195, USA; Department of Pharmacology, University of Washington, Seattle, WA 98195, USA.
| |
Collapse
|