1
|
Hamilos AE, Wijsman IC, Ding Q, Assawaphadungsit P, Ozcan Z, Assad JA. A mechanism linking dopamine's roles in reinforcement, movement and motivation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.04.04.647288. [PMID: 40236124 PMCID: PMC11996583 DOI: 10.1101/2025.04.04.647288] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/17/2025]
Abstract
Dopamine neurons (DANs) play seemingly distinct roles in reinforcement, 1-3 motivation, 4,5 and movement, 6,7 and DA-modulating therapies relieve symptoms across a puzzling spectrum of neurologic and psychiatric symptoms. 8 Yet, the mechanistic relationship among these roles is unknown. Here, we show DA's tripartite roles are causally linked by a process in which phasic striatal DA rapidly and persistently recalibrates the propensity to move, a measure of vigor. Using a self-timed movement task, we found that single exposures to reward-related DA transients (both endogenous and exogenously-induced) exerted one-shot updates to movement timing-but in a surprising fashion. Rather than reinforce specific movement times, DA transients quantitatively changed movement timing on the next trial, with larger transients leading to earlier movements (and smaller to later), consistent with a stochastic search process that calibrates the frequency of movement. Both abrupt and gradual changes in external and internal contingencies-such as timing criterion, reward content, and satiety state-caused changes to the amplitude of DA transients that causally altered movement timing. The rapidity and bidirectionality of the one-shot effects are difficult to reconcile with gradual synaptic plasticity, and instead point to more flexible cellular mechanisms, such as DA-dependent modulation of neuronal excitability. Our findings shed light on how natural reinforcement, as well as DA-related disorders such as Parkinson's disease, could affect behavioral vigor.
Collapse
|
2
|
Morita T, Lyn NG, von Heynitz RK, Goldman OV, Sorrells TR, DeGennaro M, Matthews BJ, Houri-Zeevi L, Vosshall LB. Cross-modal sensory compensation increases mosquito attraction to humans. SCIENCE ADVANCES 2025; 11:eadn5758. [PMID: 39742477 DOI: 10.1126/sciadv.adn5758] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/16/2023] [Accepted: 11/20/2024] [Indexed: 01/03/2025]
Abstract
Sensory compensation occurs when loss of one sense leads to enhanced perception by another sense. We have identified a previously undescribed mechanism of sensory compensation in female Aedes aegypti mosquitoes. Odorant receptor co-receptor (Orco) mutants show enhanced attraction to human skin temperature and increased heat-evoked neuronal activity in foreleg sensory neurons. Ir140, a foreleg-enriched member of the ionotropic receptor (IR) superfamily of sensory receptors, is up-regulated in Orco mutant legs. Ir140, Orco double mutants do not show the enhanced heat seeking seen in Orco single mutants, suggesting that up-regulation of Ir140 in the foreleg is a key mechanism underlying sensory compensation in Orco mutants. Because Orco expression is sparse in legs, this sensory compensation requires an indirect, long-range mechanism. Our findings highlight how female Aedes aegypti mosquitoes, despite suffering olfactory sensory loss, maintain the overall effectiveness of their host-seeking behavior by up-regulating attraction to human skin temperature, further enhancing their status as the most dangerous predator of humans.
Collapse
Affiliation(s)
- Takeshi Morita
- Laboratory of Neurogenetics and Behavior, The Rockefeller University, New York, NY 10065, USA
- Howard Hughes Medical Institute, New York, NY 10065, USA
| | - Nia G Lyn
- Laboratory of Neurogenetics and Behavior, The Rockefeller University, New York, NY 10065, USA
- Howard Hughes Medical Institute, New York, NY 10065, USA
| | - Ricarda K von Heynitz
- Laboratory of Neurogenetics and Behavior, The Rockefeller University, New York, NY 10065, USA
| | - Olivia V Goldman
- Laboratory of Neurogenetics and Behavior, The Rockefeller University, New York, NY 10065, USA
- Kavli Neural Systems Institute, New York, NY 10065, USA
| | - Trevor R Sorrells
- Laboratory of Neurogenetics and Behavior, The Rockefeller University, New York, NY 10065, USA
- Howard Hughes Medical Institute, New York, NY 10065, USA
- Kavli Neural Systems Institute, New York, NY 10065, USA
| | - Matthew DeGennaro
- Laboratory of Neurogenetics and Behavior, The Rockefeller University, New York, NY 10065, USA
- Howard Hughes Medical Institute, New York, NY 10065, USA
| | - Benjamin J Matthews
- Laboratory of Neurogenetics and Behavior, The Rockefeller University, New York, NY 10065, USA
- Howard Hughes Medical Institute, New York, NY 10065, USA
| | - Leah Houri-Zeevi
- Laboratory of Neurogenetics and Behavior, The Rockefeller University, New York, NY 10065, USA
- Howard Hughes Medical Institute, New York, NY 10065, USA
| | - Leslie B Vosshall
- Laboratory of Neurogenetics and Behavior, The Rockefeller University, New York, NY 10065, USA
- Howard Hughes Medical Institute, New York, NY 10065, USA
- Kavli Neural Systems Institute, New York, NY 10065, USA
| |
Collapse
|
3
|
Sun C, Wei J, Gu X, Wu M, Li M, Liu Y, An N, Wu K, Wu S, Wu J, Xu M, Wu JC, Wang YL, Chao DY, Zhang Y, Wu S. Different multicellular trichome types coordinate herbivore mechanosensing and defense in tomato. THE PLANT CELL 2024; 36:koae269. [PMID: 39404780 PMCID: PMC11638769 DOI: 10.1093/plcell/koae269] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Revised: 07/12/2024] [Accepted: 10/03/2024] [Indexed: 12/15/2024]
Abstract
Herbivore-induced wounding can elicit a defense response in plants. However, whether plants possess a surveillance system capable of detecting herbivore threats and initiating preparatory defenses before wounding occurs remains unclear. In this study, we reveal that tomato (Solanum lycopersicum) trichomes can detect and respond to the mechanical stimuli generated by herbivores. Mechanical stimuli are preferentially perceived by long trichomes, and this mechanosensation is transduced via intra-trichome communication. This communication presumably involves calcium waves, and the transduced signals activate the jasmonic acid (JA) signaling pathway in short glandular trichomes, resulting in the upregulation of the Woolly (Wo)-SlMYC1 regulatory module for terpene biosynthesis. This induced defense mechanism provides plants with an early warning system against the threat of herbivore invasion. Our findings represent a perspective on the role of multicellular trichomes in plant defense and the underlying intra-trichome communication.
Collapse
Affiliation(s)
- Chao Sun
- College of Life Sciences, College of Horticulture, Fujian Agriculture and Forestry University, Fuzhou 350002, China
| | - JinBo Wei
- College of Life Sciences, College of Horticulture, Fujian Agriculture and Forestry University, Fuzhou 350002, China
| | - XinYun Gu
- College of Life Sciences, College of Horticulture, Fujian Agriculture and Forestry University, Fuzhou 350002, China
| | - MinLiang Wu
- College of Life Sciences, College of Horticulture, Fujian Agriculture and Forestry University, Fuzhou 350002, China
| | - Meng Li
- College of Life Sciences, College of Horticulture, Fujian Agriculture and Forestry University, Fuzhou 350002, China
| | - YiXi Liu
- College of Life Sciences, College of Horticulture, Fujian Agriculture and Forestry University, Fuzhou 350002, China
| | - NingKai An
- College of Life Sciences, College of Horticulture, Fujian Agriculture and Forestry University, Fuzhou 350002, China
| | - KeMeng Wu
- College of Life Sciences, College of Horticulture, Fujian Agriculture and Forestry University, Fuzhou 350002, China
| | - ShaSha Wu
- College of Life Sciences, College of Horticulture, Fujian Agriculture and Forestry University, Fuzhou 350002, China
| | - JunQing Wu
- College of Life Sciences, College of Horticulture, Fujian Agriculture and Forestry University, Fuzhou 350002, China
| | - MeiZhi Xu
- College of Life Sciences, College of Horticulture, Fujian Agriculture and Forestry University, Fuzhou 350002, China
| | - Jia-Chen Wu
- CAS Center for Excellence in Molecular Plant Sciences, Shanghai Institute of Plant Physiology and Ecology, Chinese Academy of Sciences, Shanghai 200032, China
| | - Ya-Ling Wang
- CAS Center for Excellence in Molecular Plant Sciences, Shanghai Institute of Plant Physiology and Ecology, Chinese Academy of Sciences, Shanghai 200032, China
| | - Dai-Yin Chao
- CAS Center for Excellence in Molecular Plant Sciences, Shanghai Institute of Plant Physiology and Ecology, Chinese Academy of Sciences, Shanghai 200032, China
| | - YouJun Zhang
- Department of Plant Protection, Institute of Vegetables and Flowers, Chinese Academy of Agricultural Sciences, Beijing 100081, China
| | - Shuang Wu
- College of Life Sciences, College of Horticulture, Fujian Agriculture and Forestry University, Fuzhou 350002, China
| |
Collapse
|
4
|
Talei Franzesi G, Gupta I, Hu M, Piatkveich K, Yildirim M, Zhao JP, Eom M, Han S, Park D, Andaraarachchi H, Li Z, Greenhagen J, Islam AM, Vashishtha P, Yaqoob Z, Pak N, Wissner-Gross AD, Martin-Alarcon D, Veinot J, So PT, Kortshagen U, Yoon YG, Sur M, Boyden ES. In Vivo Optical Clearing of Mammalian Brain. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.05.611421. [PMID: 39282466 PMCID: PMC11398509 DOI: 10.1101/2024.09.05.611421] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/16/2025]
Abstract
Established methods for imaging the living mammalian brain have, to date, taken optical properties of the tissue as fixed; we here demonstrate that it is possible to modify the optical properties of the brain itself to significantly enhance at-depth imaging while preserving native physiology. Using a small amount of any of several biocompatible materials to raise the refractive index of solutions superfusing the brain prior to imaging, we could increase several-fold the signals from the deepest cells normally visible and, under both one-photon and two-photon imaging, visualize cells previously too dim to see. The enhancement was observed for both anatomical and functional fluorescent reporters across a broad range of emission wavelengths. Importantly, visual tuning properties of cortical neurons in awake mice, and electrophysiological properties of neurons assessed ex vivo, were not altered by this procedure.
Collapse
|
5
|
Tournissac M, Chaigneau E, Pfister S, Aydin AK, Goulam Houssen Y, O'Herron P, Filosa J, Collot M, Joutel A, Charpak S. Neurovascular coupling and CO 2 interrogate distinct vascular regulations. Nat Commun 2024; 15:7635. [PMID: 39223128 PMCID: PMC11369082 DOI: 10.1038/s41467-024-49698-9] [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: 10/23/2023] [Accepted: 06/12/2024] [Indexed: 09/04/2024] Open
Abstract
Neurovascular coupling (NVC), which mediates rapid increases in cerebral blood flow in response to neuronal activation, is commonly used to map brain activation or dysfunction. Here we tested the reemerging hypothesis that CO2 generated by neuronal metabolism contributes to NVC. We combined functional ultrasound and two-photon imaging in the mouse barrel cortex to specifically examine the onsets of local changes in vessel diameter, blood flow dynamics, vascular/perivascular/intracellular pH, and intracellular calcium signals along the vascular arbor in response to a short and strong CO2 challenge (10 s, 20%) and whisker stimulation. We report that the brief hypercapnia reversibly acidifies all cells of the arteriole wall and the periarteriolar space 3-4 s prior to the arteriole dilation. During this prolonged lag period, NVC triggered by whisker stimulation is not affected by the acidification of the entire neurovascular unit. As it also persists under condition of continuous inflow of CO2, we conclude that CO2 is not involved in NVC.
Collapse
Affiliation(s)
- Marine Tournissac
- Sorbonne Université, Inserm U968, Vision Institute, Paris, France.
- Université Paris Cité, Institute of Psychiatry and Neuroscience of Paris (IPNP), INSERM U1266, 75014, Paris, France.
| | | | - Sonia Pfister
- Chemistry of Photoresponsive Systems, Laboratoire de Chémo-Biologie Synthétique et Thérapeutique (CBST) UMR 7199, CNRS, Université de Strasbourg, Strasbourg, F-67400, Illkirch, France
| | - Ali-Kemal Aydin
- Sorbonne Université, Inserm U968, Vision Institute, Paris, France
| | | | - Philip O'Herron
- Department of Physiology, Augusta University, Augusta, GA, USA
| | - Jessica Filosa
- Department of Physiology, Augusta University, Augusta, GA, USA
| | - Mayeul Collot
- Chemistry of Photoresponsive Systems, Laboratoire de Chémo-Biologie Synthétique et Thérapeutique (CBST) UMR 7199, CNRS, Université de Strasbourg, Strasbourg, F-67400, Illkirch, France
| | - Anne Joutel
- Université Paris Cité, Institute of Psychiatry and Neuroscience of Paris (IPNP), INSERM U1266, 75014, Paris, France
| | - Serge Charpak
- Sorbonne Université, Inserm U968, Vision Institute, Paris, France.
| |
Collapse
|
6
|
Ayala DA, Matarazzo A, Seaberg BL, Patel M, Tijerina E, Matthews C, Bizi G, Brown A, Ta A, Rimer M, Srinivasan R. Heterogeneous brain region-specific responses to astrocytic mitochondrial DNA damage in mice. Sci Rep 2024; 14:18586. [PMID: 39127716 PMCID: PMC11316820 DOI: 10.1038/s41598-024-69499-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Accepted: 08/06/2024] [Indexed: 08/12/2024] Open
Abstract
Astrocytes display context-specific diversity in their functions and respond to noxious stimuli between brain regions. Astrocytic mitochondria have emerged as key players in governing astrocytic functional heterogeneity, given their ability to dynamically adapt their morphology to regional demands on ATP generation and Ca2+ buffering functions. Although there is reciprocal regulation between mitochondrial dynamics and mitochondrial Ca2+ signaling in astrocytes, the extent of this regulation in astrocytes from different brain regions remains unexplored. Brain-wide, experimentally induced mitochondrial DNA (mtDNA) loss in astrocytes showed that mtDNA integrity is critical for astrocyte function, however, possible diverse responses to this noxious stimulus between brain areas were not reported in these experiments. To selectively damage mtDNA in astrocytes in a brain-region-specific manner, we developed a novel adeno-associated virus (AAV)-based tool, Mito-PstI expressing the restriction enzyme PstI, specifically in astrocytic mitochondria. Here, we applied Mito-PstI to two brain regions, the dorsolateral striatum and dentate gyrus, and we show that Mito-PstI induces astrocytic mtDNA loss in vivo, but with remarkable brain-region-dependent differences on mitochondrial dynamics, Ca2+ fluxes, and astrocytic and microglial reactivity. Thus, AAV-Mito-PstI is a novel tool to explore the relationship between astrocytic mitochondrial network dynamics and astrocytic mitochondrial Ca2+ signaling in a brain-region-selective manner.
Collapse
Affiliation(s)
- Daniela A Ayala
- Department of Neuroscience and Experimental Therapeutics, College of Medicine, Texas A&M University, Bryan, TX, 77843, USA
- Graduate Program in Medical Sciences, Texas A&M University, Bryan, TX, 77843, USA
| | - Anthony Matarazzo
- Department of Neuroscience and Experimental Therapeutics, College of Medicine, Texas A&M University, Bryan, TX, 77843, USA
- Graduate Program in Genetics and Genomics, Texas A&M University, Bryan, TX, 77843, USA
| | - Bonnie L Seaberg
- Department of Neuroscience and Experimental Therapeutics, College of Medicine, Texas A&M University, Bryan, TX, 77843, USA
| | - Misha Patel
- Department of Neuroscience and Experimental Therapeutics, College of Medicine, Texas A&M University, Bryan, TX, 77843, USA
| | - Eliana Tijerina
- Department of Neuroscience and Experimental Therapeutics, College of Medicine, Texas A&M University, Bryan, TX, 77843, USA
| | - Camryn Matthews
- Department of Neuroscience and Experimental Therapeutics, College of Medicine, Texas A&M University, Bryan, TX, 77843, USA
| | - Gabriel Bizi
- Department of Neuroscience and Experimental Therapeutics, College of Medicine, Texas A&M University, Bryan, TX, 77843, USA
| | - Ashton Brown
- Department of Neuroscience and Experimental Therapeutics, College of Medicine, Texas A&M University, Bryan, TX, 77843, USA
| | - Alan Ta
- Department of Neuroscience and Experimental Therapeutics, College of Medicine, Texas A&M University, Bryan, TX, 77843, USA
| | - Mendell Rimer
- Department of Neuroscience and Experimental Therapeutics, College of Medicine, Texas A&M University, Bryan, TX, 77843, USA.
- Graduate Program in Genetics and Genomics, Texas A&M University, Bryan, TX, 77843, USA.
- Texas A&M Institute for Neuroscience, Texas A&M University, Bryan, TX, 77843, USA.
| | - Rahul Srinivasan
- Department of Neuroscience and Experimental Therapeutics, College of Medicine, Texas A&M University, Bryan, TX, 77843, USA.
- Graduate Program in Medical Sciences, Texas A&M University, Bryan, TX, 77843, USA.
- Texas A&M Institute for Neuroscience, Texas A&M University, Bryan, TX, 77843, USA.
| |
Collapse
|
7
|
Ayala DA, Matarazzo A, Seaberg BL, Patel M, Tijerina E, Matthews C, Bizi G, Brown A, Ta A, Rimer M, Srinivasan R. Heterogeneous brain region-specific responses to astrocytic mitochondrial DNA damage in mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.29.596517. [PMID: 38853966 PMCID: PMC11160752 DOI: 10.1101/2024.05.29.596517] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2024]
Abstract
Astrocytes use Ca 2+ signals to regulate multiple aspects of normal and pathological brain function. Astrocytes display context-specific diversity in their functions, and in their response to noxious stimuli between brain regions. Indeed, astrocytic mitochondria have emerged as key players in governing astrocytic functional heterogeneity, given their ability to dynamically adapt their morphology to regional demands on their ATP generation and Ca 2+ buffering functions. Although there is reciprocal regulation between mitochondrial dynamics and mitochondrial Ca 2+ signaling in astrocytes, the extent of this regulation into the rich diversity of astrocytes in different brain regions remains largely unexplored. Brain-wide, experimentally induced mitochondrial DNA (mtDNA) loss in astrocytes showed that mtDNA integrity is critical for proper astrocyte function, however, few insights into possible diverse responses to this noxious stimulus from astrocytes in different brain areas were reported in these experiments. To selectively damage mtDNA in astrocytes in a brain-region-specific manner, we developed a novel adeno-associated virus (AAV)-based tool, Mito-PstI, which expresses the restriction enzyme PstI, specifically in astrocytic mitochondria. Here, we applied Mito-PstI to two distinct brain regions, the dorsolateral striatum, and the hippocampal dentate gyrus, and we show that Mito-PstI can induce astrocytic mtDNA loss in vivo , but with remarkable brain-region-dependent differences on mitochondrial dynamics, spontaneous Ca 2+ fluxes and astrocytic as well as microglial reactivity. Thus, AAV-Mito-PstI is a novel tool to explore the relationship between astrocytic mitochondrial network dynamics and astrocytic mitochondrial Ca 2+ signaling in a brain-region-selective manner.
Collapse
|
8
|
Zhang Y, Looger LL. Fast and sensitive GCaMP calcium indicators for neuronal imaging. J Physiol 2024; 602:1595-1604. [PMID: 36811153 DOI: 10.1113/jp283832] [Citation(s) in RCA: 27] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2022] [Accepted: 02/15/2023] [Indexed: 02/24/2023] Open
Abstract
We review the principles of development and deployment of genetically encoded calcium indicators (GECIs) for the detection of neural activity. Our focus is on the popular GCaMP family of green GECIs, culminating in the recent release of the jGCaMP8 sensors, with dramatically improved kinetics relative to previous generations. We summarize the properties of GECIs in multiple colour channels (blue, cyan, green, yellow, red, far-red) and highlight areas for further improvement. With their low-millisecond rise-times, the jGCaMP8 indicators allow new classes of experiments following neural activity in time frames approaching the underlying computations.
Collapse
Affiliation(s)
- Yan Zhang
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA, USA
| | - Loren L Looger
- Department of Neurosciences, Howard Hughes Medical Institute, University of California, San Diego, La Jolla, CA, USA
| |
Collapse
|
9
|
Saito K, Shigetomi E, Shinozaki Y, Kobayashi K, Parajuli B, Kubota Y, Sakai K, Miyakawa M, Horiuchi H, Nabekura J, Koizumi S. Microglia sense astrocyte dysfunction and prevent disease progression in an Alexander disease model. Brain 2024; 147:698-716. [PMID: 37955589 PMCID: PMC10834242 DOI: 10.1093/brain/awad358] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Revised: 09/28/2023] [Accepted: 10/06/2023] [Indexed: 11/14/2023] Open
Abstract
Alexander disease (AxD) is an intractable neurodegenerative disorder caused by GFAP mutations. It is a primary astrocyte disease with a pathological hallmark of Rosenthal fibres within astrocytes. AxD astrocytes show several abnormal phenotypes. Our previous study showed that AxD astrocytes in model mice exhibit aberrant Ca2+ signals that induce AxD aetiology. Here, we show that microglia have unique phenotypes with morphological and functional alterations, which are related to the pathogenesis of AxD. Immunohistochemical studies of 60TM mice (AxD model) showed that AxD microglia exhibited highly ramified morphology. Functional changes in microglia were assessed by Ca2+ imaging using hippocampal brain slices from Iba1-GCaMP6-60TM mice and two-photon microscopy. We found that AxD microglia showed aberrant Ca2+ signals, with high frequency Ca2+ signals in both the processes and cell bodies. These microglial Ca2+ signals were inhibited by pharmacological blockade or genetic knockdown of P2Y12 receptors but not by tetrodotoxin, indicating that these signals are independent of neuronal activity but dependent on extracellular ATP from non-neuronal cells. Our single-cell RNA sequencing data showed that the expression level of Entpd2, an astrocyte-specific gene encoding the ATP-degrading enzyme NTPDase2, was lower in AxD astrocytes than in wild-type astrocytes. In situ ATP imaging using the adeno-associated virus vector GfaABC1D ATP1.0 showed that exogenously applied ATP was present longer in 60TM mice than in wild-type mice. Thus, the increased ATP level caused by the decrease in its metabolizing enzyme in astrocytes could be responsible for the enhancement of microglial Ca2+ signals. To determine whether these P2Y12 receptor-mediated Ca2+ signals in AxD microglia play a significant role in the pathological mechanism, a P2Y12 receptor antagonist, clopidogrel, was administered. Clopidogrel significantly exacerbated pathological markers in AxD model mice and attenuated the morphological features of microglia, suggesting that microglia play a protective role against AxD pathology via P2Y12 receptors. Taken together, we demonstrated that microglia sense AxD astrocyte dysfunction via P2Y12 receptors as an increase in extracellular ATP and alter their morphology and Ca2+ signalling, thereby protecting against AxD pathology. Although AxD is a primary astrocyte disease, our study may facilitate understanding of the role of microglia as a disease modifier, which may contribute to the clinical diversity of AxD.
Collapse
Affiliation(s)
- Kozo Saito
- Department of Neuropharmacology, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Yamanashi 409-3898, Japan
- GLIA Center, University of Yamanashi, Yamanashi 409-3898, Japan
| | - Eiji Shigetomi
- Department of Neuropharmacology, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Yamanashi 409-3898, Japan
- GLIA Center, University of Yamanashi, Yamanashi 409-3898, Japan
| | - Youichi Shinozaki
- Department of Neuropharmacology, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Yamanashi 409-3898, Japan
- GLIA Center, University of Yamanashi, Yamanashi 409-3898, Japan
| | - Kenji Kobayashi
- Department of Neuropharmacology, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Yamanashi 409-3898, Japan
| | - Bijay Parajuli
- Department of Neuropharmacology, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Yamanashi 409-3898, Japan
- GLIA Center, University of Yamanashi, Yamanashi 409-3898, Japan
| | - Yuto Kubota
- Department of Neuropharmacology, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Yamanashi 409-3898, Japan
| | - Kent Sakai
- Department of Neuropharmacology, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Yamanashi 409-3898, Japan
- GLIA Center, University of Yamanashi, Yamanashi 409-3898, Japan
| | - Miho Miyakawa
- Department of Neuropharmacology, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Yamanashi 409-3898, Japan
- GLIA Center, University of Yamanashi, Yamanashi 409-3898, Japan
| | - Hiroshi Horiuchi
- Division of Homeostatic Development, National Institute for Physiological Sciences, National Institutes of Natural Sciences, Aichi 444-8585, Japan
| | - Junichi Nabekura
- Division of Homeostatic Development, National Institute for Physiological Sciences, National Institutes of Natural Sciences, Aichi 444-8585, Japan
| | - Schuichi Koizumi
- Department of Neuropharmacology, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Yamanashi 409-3898, Japan
- GLIA Center, University of Yamanashi, Yamanashi 409-3898, Japan
| |
Collapse
|
10
|
Haffner V, Nourian Z, Boerman EM, Lambert MD, Hanft LM, Krenz M, Baines CP, Duan D, McDonald KS, Domeier TL. Calcium handling dysfunction and cardiac damage following acute ventricular preload challenge in the dystrophin-deficient mouse heart. Am J Physiol Heart Circ Physiol 2023; 325:H1168-H1177. [PMID: 37737731 PMCID: PMC10907071 DOI: 10.1152/ajpheart.00265.2023] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Revised: 09/13/2023] [Accepted: 09/15/2023] [Indexed: 09/23/2023]
Abstract
Duchenne muscular dystrophy (DMD) is the most common muscular dystrophy and is caused by mutations in the dystrophin gene. Dystrophin deficiency is associated with structural and functional changes of the muscle cell sarcolemma and/or stretch-induced ion channel activation. In this investigation, we use mice with transgenic cardiomyocyte-specific expression of the GCaMP6f Ca2+ indicator to test the hypothesis that dystrophin deficiency leads to cardiomyocyte Ca2+ handling abnormalities following preload challenge. α-MHC-MerCreMer-GCaMP6f transgenic mice were developed on both a wild-type (WT) or dystrophic (Dmdmdx-4Cv) background. Isolated hearts of 3-7-mo male mice were perfused in unloaded Langendorff mode (0 mmHg) and working heart mode (preload = 20 mmHg). Following a 30-min preload challenge, hearts were perfused in unloaded Langendorff mode with 40 μM blebbistatin, and GCaMP6f was imaged using confocal fluorescence microscopy. Incidence of premature ventricular complexes (PVCs) was monitored before and following preload elevation at 20 mmHg. Hearts of both wild-type and dystrophic mice exhibited similar left ventricular contractile function. Following preload challenge, dystrophic hearts exhibited a reduction in GCaMP6f-positive cardiomyocytes and an increase in number of cardiomyocytes exhibiting Ca2+ waves/overload. Incidence of cardiac arrhythmias was low in both wild-type and dystrophic hearts during unloaded Langendorff mode. However, after preload elevation to 20-mmHg hearts of dystrophic mice exhibited an increased incidence of PVCs compared with hearts of wild-type mice. In conclusion, these data indicate susceptibility to preload-induced Ca2+ overload, ventricular damage, and ventricular dysfunction in male Dmdmdx-4Cv hearts. Our data support the hypothesis that cardiomyocyte Ca2+ overload underlies cardiac dysfunction in muscular dystrophy.NEW & NOTEWORTHY The mechanisms of cardiac disease progression in muscular dystrophy are complex and poorly understood. Using a transgenic mouse model with cardiomyocyte-specific expression of the GCaMP6f Ca2+ indicator, the present study provides further support for the Ca2+-overload hypothesis of disease progression and ventricular arrhythmogenesis in muscular dystrophy.
Collapse
Affiliation(s)
- Vivian Haffner
- Department of Medical Pharmacology and Physiology, School of Medicine, University of Missouri, Columbia, Missouri, United States
| | - Zahra Nourian
- Department of Medical Pharmacology and Physiology, School of Medicine, University of Missouri, Columbia, Missouri, United States
| | - Erika M Boerman
- Department of Medical Pharmacology and Physiology, School of Medicine, University of Missouri, Columbia, Missouri, United States
| | - Michelle D Lambert
- Department of Medical Pharmacology and Physiology, School of Medicine, University of Missouri, Columbia, Missouri, United States
| | - Laurin M Hanft
- Department of Medical Pharmacology and Physiology, School of Medicine, University of Missouri, Columbia, Missouri, United States
| | - Maike Krenz
- Department of Medical Pharmacology and Physiology, School of Medicine, University of Missouri, Columbia, Missouri, United States
- The Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri, United States
| | - Christopher P Baines
- Department of Biomedical Sciences, College of Veterinary Medicine, University of Missouri, Columbia, Missouri, United States
- The Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri, United States
| | - Dongsheng Duan
- Department of Molecular Microbiology and Immunology, School of Medicine, University of Missouri, Columbia, Missouri, United States
- Department of Neurology, School of Medicine, University of Missouri, Columbia, Missouri, United States
| | - Kerry S McDonald
- Department of Medical Pharmacology and Physiology, School of Medicine, University of Missouri, Columbia, Missouri, United States
| | - Timothy L Domeier
- Department of Medical Pharmacology and Physiology, School of Medicine, University of Missouri, Columbia, Missouri, United States
- Department of Medicine, School of Medicine, University of Missouri, Columbia, Missouri, United States
| |
Collapse
|
11
|
Matityahu L, Gilin N, Sarpong GA, Atamna Y, Tiroshi L, Tritsch NX, Wickens JR, Goldberg JA. Acetylcholine waves and dopamine release in the striatum. Nat Commun 2023; 14:6852. [PMID: 37891198 PMCID: PMC10611775 DOI: 10.1038/s41467-023-42311-5] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Accepted: 10/06/2023] [Indexed: 10/29/2023] Open
Abstract
Striatal dopamine encodes reward, with recent work showing that dopamine release occurs in spatiotemporal waves. However, the mechanism of dopamine waves is unknown. Here we report that acetylcholine release in mouse striatum also exhibits wave activity, and that the spatial scale of striatal dopamine release is extended by nicotinic acetylcholine receptors. Based on these findings, and on our demonstration that single cholinergic interneurons can induce dopamine release, we hypothesized that the local reciprocal interaction between cholinergic interneurons and dopamine axons suffices to drive endogenous traveling waves. We show that the morphological and physiological properties of cholinergic interneuron - dopamine axon interactions can be modeled as a reaction-diffusion system that gives rise to traveling waves. Analytically-tractable versions of the model show that the structure and the nature of propagation of acetylcholine and dopamine traveling waves depend on their coupling, and that traveling waves can give rise to empirically observed correlations between these signals. Thus, our study provides evidence for striatal acetylcholine waves in vivo, and proposes a testable theoretical framework that predicts that the observed dopamine and acetylcholine waves are strongly coupled phenomena.
Collapse
Affiliation(s)
- Lior Matityahu
- Department of Medical Neurobiology, Institute of Medical Research Israel - Canada, The Faculty of Medicine, The Hebrew University of Jerusalem, 9112102, Jerusalem, Israel
| | - Naomi Gilin
- Department of Medical Neurobiology, Institute of Medical Research Israel - Canada, The Faculty of Medicine, The Hebrew University of Jerusalem, 9112102, Jerusalem, Israel
| | - Gideon A Sarpong
- Okinawa Institute of Science and Technology Graduate University, Okinawa, Japan
| | - Yara Atamna
- Department of Medical Neurobiology, Institute of Medical Research Israel - Canada, The Faculty of Medicine, The Hebrew University of Jerusalem, 9112102, Jerusalem, Israel
| | - Lior Tiroshi
- Department of Medical Neurobiology, Institute of Medical Research Israel - Canada, The Faculty of Medicine, The Hebrew University of Jerusalem, 9112102, Jerusalem, Israel
| | - Nicolas X Tritsch
- Neuroscience Institute, New York University Grossman School of Medicine, New York, NY, 10016, USA
| | - Jeffery R Wickens
- Okinawa Institute of Science and Technology Graduate University, Okinawa, Japan
| | - Joshua A Goldberg
- Department of Medical Neurobiology, Institute of Medical Research Israel - Canada, The Faculty of Medicine, The Hebrew University of Jerusalem, 9112102, Jerusalem, Israel.
| |
Collapse
|
12
|
Yaganoglu S, Kalyviotis K, Vagena-Pantoula C, Jülich D, Gaub BM, Welling M, Lopes T, Lachowski D, Tang SS, Del Rio Hernandez A, Salem V, Müller DJ, Holley SA, Vermot J, Shi J, Helassa N, Török K, Pantazis P. Highly specific and non-invasive imaging of Piezo1-dependent activity across scales using GenEPi. Nat Commun 2023; 14:4352. [PMID: 37468521 PMCID: PMC10356793 DOI: 10.1038/s41467-023-40134-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Accepted: 07/11/2023] [Indexed: 07/21/2023] Open
Abstract
Mechanosensing is a ubiquitous process to translate external mechanical stimuli into biological responses. Piezo1 ion channels are directly gated by mechanical forces and play an essential role in cellular mechanotransduction. However, readouts of Piezo1 activity are mainly examined by invasive or indirect techniques, such as electrophysiological analyses and cytosolic calcium imaging. Here, we introduce GenEPi, a genetically-encoded fluorescent reporter for non-invasive optical monitoring of Piezo1-dependent activity. We demonstrate that GenEPi has high spatiotemporal resolution for Piezo1-dependent stimuli from the single-cell level to that of the entire organism. GenEPi reveals transient, local mechanical stimuli in the plasma membrane of single cells, resolves repetitive contraction-triggered stimulation of beating cardiomyocytes within microtissues, and allows for robust and reliable monitoring of Piezo1-dependent activity in vivo. GenEPi will enable non-invasive optical monitoring of Piezo1 activity in mechanochemical feedback loops during development, homeostatic regulation, and disease.
Collapse
Affiliation(s)
- Sine Yaganoglu
- Department of Biosystems Science and Engineering, Eidgenössische Technische Hochschule (ETH) Zurich, Basel, Switzerland
| | | | | | - Dörthe Jülich
- Department of Molecular, Cellular and Developmental Biology, Yale University, New Haven, CT, USA
| | - Benjamin M Gaub
- Department of Biosystems Science and Engineering, Eidgenössische Technische Hochschule (ETH) Zurich, Basel, Switzerland
| | - Maaike Welling
- Department of Biosystems Science and Engineering, Eidgenössische Technische Hochschule (ETH) Zurich, Basel, Switzerland
- Department of Bioengineering, Imperial College London, London, UK
| | - Tatiana Lopes
- Section of Investigative Medicine, Department of Metabolism, Digestion, and Reproduction, Imperial College London, London, UK
| | | | - See Swee Tang
- Department of Bioengineering, Imperial College London, London, UK
| | | | - Victoria Salem
- Department of Bioengineering, Imperial College London, London, UK
| | - Daniel J Müller
- Department of Biosystems Science and Engineering, Eidgenössische Technische Hochschule (ETH) Zurich, Basel, Switzerland
| | - Scott A Holley
- Department of Molecular, Cellular and Developmental Biology, Yale University, New Haven, CT, USA
| | - Julien Vermot
- Department of Bioengineering, Imperial College London, London, UK
| | - Jian Shi
- Leeds Institute of Cardiovascular and Metabolic Medicine, LIGHT Laboratories, University of Leeds, Leeds, UK
| | - Nordine Helassa
- Molecular and Clinical Sciences Research Institute, St. George's, University of London, London, UK
- Department of Biochemistry, Cell and Systems Biology, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, UK
| | - Katalin Török
- Molecular and Clinical Sciences Research Institute, St. George's, University of London, London, UK
| | - Periklis Pantazis
- Department of Biosystems Science and Engineering, Eidgenössische Technische Hochschule (ETH) Zurich, Basel, Switzerland.
- Department of Bioengineering, Imperial College London, London, UK.
| |
Collapse
|
13
|
Evans SW, Shi DQ, Chavarha M, Plitt MH, Taxidis J, Madruga B, Fan JL, Hwang FJ, van Keulen SC, Suomivuori CM, Pang MM, Su S, Lee S, Hao YA, Zhang G, Jiang D, Pradhan L, Roth RH, Liu Y, Dorian CC, Reese AL, Negrean A, Losonczy A, Makinson CD, Wang S, Clandinin TR, Dror RO, Ding JB, Ji N, Golshani P, Giocomo LM, Bi GQ, Lin MZ. A positively tuned voltage indicator for extended electrical recordings in the brain. Nat Methods 2023; 20:1104-1113. [PMID: 37429962 PMCID: PMC10627146 DOI: 10.1038/s41592-023-01913-z] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Accepted: 05/15/2023] [Indexed: 07/12/2023]
Abstract
Genetically encoded voltage indicators (GEVIs) enable optical recording of electrical signals in the brain, providing subthreshold sensitivity and temporal resolution not possible with calcium indicators. However, one- and two-photon voltage imaging over prolonged periods with the same GEVI has not yet been demonstrated. Here, we report engineering of ASAP family GEVIs to enhance photostability by inversion of the fluorescence-voltage relationship. Two of the resulting GEVIs, ASAP4b and ASAP4e, respond to 100-mV depolarizations with ≥180% fluorescence increases, compared with the 50% fluorescence decrease of the parental ASAP3. With standard microscopy equipment, ASAP4e enables single-trial detection of spikes in mice over the course of minutes. Unlike GEVIs previously used for one-photon voltage recordings, ASAP4b and ASAP4e also perform well under two-photon illumination. By imaging voltage and calcium simultaneously, we show that ASAP4b and ASAP4e can identify place cells and detect voltage spikes with better temporal resolution than commonly used calcium indicators. Thus, ASAP4b and ASAP4e extend the capabilities of voltage imaging to standard one- and two-photon microscopes while improving the duration of voltage recordings.
Collapse
Affiliation(s)
- S Wenceslao Evans
- Department of Neurobiology, Stanford University Medical Center, Stanford, CA, USA
| | - Dong-Qing Shi
- Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Mariya Chavarha
- Department of Bioengineering, Stanford University, Stanford, CA, USA
| | - Mark H Plitt
- Department of Neurobiology, Stanford University Medical Center, Stanford, CA, USA
| | - Jiannis Taxidis
- Department of Neurology, UCLA David Geffen School of Medicine, Los Angeles, CA, USA
- Program in Neurosciences & Mental Health, Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Physiology, University of Toronto, Toronto, Ontario, Canada
- Program in Neurosciences & Mental Health, Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Physiology, University of Toronto, Toronto, Ontario, Canada
| | - Blake Madruga
- Department of Neurology, UCLA David Geffen School of Medicine, Los Angeles, CA, USA
| | - Jiang Lan Fan
- UC Berkeley/UCSF Joint Program in Bioengineering, University of California Berkeley, Berkeley, CA, USA
| | - Fuu-Jiun Hwang
- Department of Neurosurgery, Stanford University Medical Center, Stanford, CA, USA
| | - Siri C van Keulen
- Department of Computer Science, Stanford University, Stanford, CA, USA
| | | | - Michelle M Pang
- Department of Neurobiology, Stanford University Medical Center, Stanford, CA, USA
| | - Sharon Su
- Department of Neurobiology, Stanford University Medical Center, Stanford, CA, USA
| | - Sungmoo Lee
- Department of Neurobiology, Stanford University Medical Center, Stanford, CA, USA
| | - Yukun A Hao
- Department of Neurobiology, Stanford University Medical Center, Stanford, CA, USA
- Department of Bioengineering, Stanford University, Stanford, CA, USA
| | - Guofeng Zhang
- Department of Neurobiology, Stanford University Medical Center, Stanford, CA, USA
| | - Dongyun Jiang
- Department of Neurobiology, Stanford University Medical Center, Stanford, CA, USA
| | - Lagnajeet Pradhan
- Department of Neurobiology, Stanford University Medical Center, Stanford, CA, USA
| | - Richard H Roth
- Department of Neurosurgery, Stanford University Medical Center, Stanford, CA, USA
| | - Yu Liu
- Department of Neurosurgery, Stanford University Medical Center, Stanford, CA, USA
- Department of Ophthalmology, Stanford University Medical Center, Stanford, CA, USA
| | - Conor C Dorian
- Department of Neurology, UCLA David Geffen School of Medicine, Los Angeles, CA, USA
| | - Austin L Reese
- Institute for Genomic Medicine, Columbia University, New York, NY, USA
| | - Adrian Negrean
- Department of Neuroscience, Columbia University, New York, NY, USA
| | - Attila Losonczy
- Department of Neuroscience, Columbia University, New York, NY, USA
- Mortimer B. Zuckerman Mind Brain Behavior Institute, New York, NY, USA
- Kavli Institute for Brain Science, New York, NY, USA
| | - Christopher D Makinson
- Institute for Genomic Medicine, Columbia University, New York, NY, USA
- Department of Neurology, Columbia University, New York, NY, USA
| | - Sui Wang
- Department of Ophthalmology, Stanford University Medical Center, Stanford, CA, USA
| | - Thomas R Clandinin
- Department of Neurobiology, Stanford University Medical Center, Stanford, CA, USA
| | - Ron O Dror
- Department of Computer Science, Stanford University, Stanford, CA, USA
- Department of Structural Biology, Stanford University School of Medicine, Stanford, CA, USA
- Institute for Computational and Mathematical Engineering, Stanford University, Stanford, CA, USA
- Department of Molecular and Cellular Physiology, Stanford University, Stanford, USA
| | - Jun B Ding
- Department of Neurosurgery, Stanford University Medical Center, Stanford, CA, USA
- Department of Neurology and Neurological Sciences, Stanford University Medical Center, Stanford, CA, USA
| | - Na Ji
- Program in Neurosciences & Mental Health, Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Molecular and Cell Biology, University of California Berkeley, Berkeley, CA, USA
- Department of Physics, University of California Berkeley, Berkeley, CA, USA
- Helen Wills Neuroscience Institute, University of California Berkeley, Berkeley, CA, USA
- Molecular Biophysics and Integrated Bioimaging Division, Lawrence Berkeley National Laboratory, Berkeley, CA, USA
| | - Peyman Golshani
- Department of Neurology, UCLA David Geffen School of Medicine, Los Angeles, CA, USA
- Semel Institute for Neuroscience and Human Behavior, UCLA David Geffen School of Medicine, Los Angeles, CA, USA
| | - Lisa M Giocomo
- Department of Neurobiology, Stanford University Medical Center, Stanford, CA, USA
| | - Guo-Qiang Bi
- Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Interdisciplinary Center for Brain Information, Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Michael Z Lin
- Department of Neurobiology, Stanford University Medical Center, Stanford, CA, USA.
- Department of Bioengineering, Stanford University, Stanford, CA, USA.
- Department of Chemical and Systems Biology, Stanford University, Stanford, USA.
| |
Collapse
|
14
|
Kalkunte NG, Delambre TE, Sohn S, Pickett M, Parekh S, Zoldan J. Engineering Alignment Has Mixed Effects on Human Induced Pluripotent Stem Cell Differentiated Cardiomyocyte Maturation. Tissue Eng Part A 2023; 29:322-332. [PMID: 36855326 DOI: 10.1089/ten.tea.2022.0172] [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] [Indexed: 03/02/2023] Open
Abstract
The potential of human induced pluripotent stem cell differentiated cardiomyocytes (hiPSC-CMs) is greatly limited by their functional immaturity. Strong relationships exist between cardiomyocyte (CM) structure and function, leading many in the field to seek ways to mature hiPSC-CMs by culturing on biomimetic substrates, specifically those that promote alignment. However, these in vitro models have so far failed to replicate the alignment that occurs during cardiac differentiation. We show that engineered alignment, incorporated before and during cardiac differentiation, affects hiPSC-CM electrochemical coupling and mitochondrial morphology. We successfully engineer alignment in differentiating human induced pluripotent stem cells (hiPSCs) as early as day 4. We uniquely apply optical redox imaging to monitor the metabolic changes occurring during cardiac differentiation. We couple this modality with cardiac-specific markers, which allows us to assess cardiac metabolism in heterogeneous cell populations. The engineered alignment drives hiPSC-CM differentiation toward the ventricular compact CM subtype and improves electrochemical coupling in the short term, at day 14 of differentiation. Moreover, we observe the glycolysis to oxidative phosphorylation switch throughout differentiation and CM development. On the subcellular scale, we note changes in mitochondrial morphology in the long term, at day 28 of differentiation. Our results demonstrate that cellular alignment accelerates hiPSC-CM maturity and emphasizes the interrelation of structure and function in cardiac development. We anticipate that combining engineered alignment with additional maturation strategies will result in improved development of mature CMs from hiPSCs and strongly improve cardiac tissue engineering.
Collapse
Affiliation(s)
- Nikhith G Kalkunte
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, Texas, USA
| | - Talia E Delambre
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, Texas, USA
| | - Sogu Sohn
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, Texas, USA
| | - Madison Pickett
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, Texas, USA
| | - Sapun Parekh
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, Texas, USA
| | - Janet Zoldan
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, Texas, USA
| |
Collapse
|
15
|
Tran O, Hughes HJ, Carter T, Török K. Development and characterization of novel jGCaMP8f calcium sensor variants with improved kinetics and fluorescence response range. Front Cell Neurosci 2023; 17:1155406. [PMID: 37275778 PMCID: PMC10234427 DOI: 10.3389/fncel.2023.1155406] [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: 01/31/2023] [Accepted: 04/28/2023] [Indexed: 06/07/2023] Open
Abstract
Introduction Genetically encoded biosensors for monitoring intracellular calcium changes have advanced our understanding of cell signaling and neuronal activity patterns in health and disease. Successful application of GCaMP biosensors to a wide range of biological questions requires that sensor properties such as brightness and dynamic range, ligand affinity and response kinetics be tuned to the specific conditions or phenomena to be investigated. Random as well as rational targeted mutations of such sensor molecules have led to a number of important breakthroughs in this field, including the calcium sensors GCaMP6f and GCaMP6fu. jGCaMP8f of the most recently developed generation is promising a step-change in in vivo imaging with further increased fluorescence dynamic range. Here, we critically examine the biophysical properties of jGCaMP8f and report development by rational design of two novel variants of jGCaMP8f. Methods We determined the in vitro biophysical properties of jGCaMP8f and selected variants by fluorescence spectroscopies and compared their performance monitoring intracellular Ca2+ transients with previously developed fast and bright GCaMP sensors by live cell imaging. Results We demonstrate that the physiologically highly relevant Mg2+ not only majorly affects the kinetic responses of GCaMPs but also their brightness and fluorescence dynamic range. We developed novel variants jGCaMP8f L27A which has threefold faster off-kinetics and jGCaMP8f F366H which shows a ∼3-fold greater dynamic range than jGCaMP8f, in vitro as well as in HEK293T cells and endothelial cell line HUVEC in response to ATP stimulation. Discussion We discuss the importance of optimization of biosensors for studying neurobiology in the context of the novel variants of jGCaMP8f. The jGCaMP8f F366H variant with a large dynamic range has the potential to improve in vivo imaging outcomes with increased signal-to-noise ratio. The L27A variant with faster kinetics than jGCaMP8f has larger cellular responses than previous fast GCaMP variants. The jGCaMP8f generation and novel improved variants presented here will further increase the application potential of GECIs in health and disease.
Collapse
|
16
|
Pollmann EH, Yin H, Uguz I, Dubey A, Wingel KE, Choi JS, Moazeni S, Gilhotra Y, Pavlovsky VA, Banees A, Boominathan V, Robinson J, Veeraraghavan A, Pieribone VA, Pesaran B, Shepard KL. Subdural CMOS optical probe (SCOPe) for bidirectional neural interfacing. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.02.07.527500. [PMID: 36798295 PMCID: PMC9934536 DOI: 10.1101/2023.02.07.527500] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/11/2023]
Abstract
Optical neurotechnologies use light to interface with neurons and can monitor and manipulate neural activity with high spatial-temporal precision over large cortical extents. While there has been significant progress in miniaturizing microscope for head-mounted configurations, these existing devices are still very bulky and could never be fully implanted. Any viable translation of these technologies to human use will require a much more noninvasive, fully implantable form factor. Here, we leverage advances in microelectronics and heterogeneous optoelectronic packaging to develop a transformative, ultrathin, miniaturized device for bidirectional optical stimulation and recording: the subdural CMOS Optical Probe (SCOPe). By being thin enough to lie entirely within the subdural space of the primate brain, SCOPe defines a path for the eventual human translation of a new generation of brain-machine interfaces based on light.
Collapse
|
17
|
Lukyanenko V, Muriel J, Garman D, Breydo L, Bloch RJ. Elevated Ca 2+ at the triad junction underlies dysregulation of Ca 2+ signaling in dysferlin-null skeletal muscle. Front Physiol 2022; 13:1032447. [PMID: 36406982 PMCID: PMC9669649 DOI: 10.3389/fphys.2022.1032447] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Accepted: 10/14/2022] [Indexed: 11/05/2022] Open
Abstract
Dysferlin-null A/J myofibers generate abnormal Ca2+ transients that are slightly reduced in amplitude compared to controls. These are further reduced in amplitude by hypoosmotic shock and often appear as Ca2+ waves (Lukyanenko et al., J. Physiol., 2017). Ca2+ waves are typically associated with Ca2+-induced Ca2+ release, or CICR, which can be myopathic. We tested the ability of a permeable Ca2+ chelator, BAPTA-AM, to inhibit CICR in injured dysferlin-null fibers and found that 10-50 nM BAPTA-AM suppressed all Ca2+ waves. The same concentrations of BAPTA-AM increased the amplitude of the Ca2+ transient in A/J fibers to wild type levels and protected transients against the loss of amplitude after hypoosmotic shock, as also seen in wild type fibers. Incubation with 10 nM BAPTA-AM led to intracellular BAPTA concentrations of ∼60 nM, as estimated with its fluorescent analog, Fluo-4AM. This should be sufficient to restore intracellular Ca2+ to levels seen in wild type muscle. Fluo-4AM was ∼10-fold less effective than BAPTA-AM, however, consistent with its lower affinity for Ca2+. EGTA, which has an affinity for Ca2+ similar to BAPTA, but with much slower kinetics of binding, was even less potent when introduced as the -AM derivative. By contrast, a dysferlin variant with GCaMP6fu in place of its C2A domain accumulated at triad junctions, like wild type dysferlin, and suppressed all abnormal Ca2+ signaling. GCaMP6fu introduced as a Venus chimera did not accumulate at junctions and failed to suppress abnormal Ca2+ signaling. Our results suggest that leak of Ca2+ into the triad junctional cleft underlies dysregulation of Ca2+ signaling in dysferlin-null myofibers, and that dysferlin's C2A domain suppresses abnormal Ca2+ signaling and protects muscle against injury by binding Ca2+ in the cleft.
Collapse
Affiliation(s)
- Valeriy Lukyanenko
- Department of Physiology, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Joaquin Muriel
- Department of Physiology, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Daniel Garman
- Department of Physiology, University of Maryland School of Medicine, Baltimore, MD, United States
- Program in Biochemistry and Molecular Biology, University of Maryland, Baltimore, MD, United States
| | - Leonid Breydo
- Formulation Development, Regeneron Pharmaceuticals, Tarrytown, NY, United States
| | - Robert J. Bloch
- Department of Physiology, University of Maryland School of Medicine, Baltimore, MD, United States
| |
Collapse
|
18
|
Ren L, Gopireddy RR, Perkins G, Zhang H, Timofeyev V, Lyu Y, Diloretto DA, Trinh P, Sirish P, Overton JL, Xu W, Grainger N, Xiang YK, Dedkova EN, Zhang XD, Yamoah EN, Navedo MF, Thai PN, Chiamvimonvat N. Disruption of mitochondria-sarcoplasmic reticulum microdomain connectomics contributes to sinus node dysfunction in heart failure. Proc Natl Acad Sci U S A 2022; 119:e2206708119. [PMID: 36044551 PMCID: PMC9456763 DOI: 10.1073/pnas.2206708119] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Accepted: 08/01/2022] [Indexed: 11/18/2022] Open
Abstract
The sinoatrial node (SAN), the leading pacemaker region, generates electrical impulses that propagate throughout the heart. SAN dysfunction with bradyarrhythmia is well documented in heart failure (HF). However, the underlying mechanisms are not completely understood. Mitochondria are critical to cellular processes that determine the life or death of the cell. The release of Ca2+ from the ryanodine receptors 2 (RyR2) on the sarcoplasmic reticulum (SR) at mitochondria-SR microdomains serves as the critical communication to match energy production to meet metabolic demands. Therefore, we tested the hypothesis that alterations in the mitochondria-SR connectomics contribute to SAN dysfunction in HF. We took advantage of a mouse model of chronic pressure overload-induced HF by transverse aortic constriction (TAC) and a SAN-specific CRISPR-Cas9-mediated knockdown of mitofusin-2 (Mfn2), the mitochondria-SR tethering GTPase protein. TAC mice exhibited impaired cardiac function with HF, cardiac fibrosis, and profound SAN dysfunction. Ultrastructural imaging using electron microscope (EM) tomography revealed abnormal mitochondrial structure with increased mitochondria-SR distance. The expression of Mfn2 was significantly down-regulated and showed reduced colocalization with RyR2 in HF SAN cells. Indeed, SAN-specific Mfn2 knockdown led to alterations in the mitochondria-SR microdomains and SAN dysfunction. Finally, disruptions in the mitochondria-SR microdomains resulted in abnormal mitochondrial Ca2+ handling, alterations in localized protein kinase A (PKA) activity, and impaired mitochondrial function in HF SAN cells. The current study provides insights into the role of mitochondria-SR microdomains in SAN automaticity and possible therapeutic targets for SAN dysfunction in HF patients.
Collapse
Affiliation(s)
- Lu Ren
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of California, Davis, CA 95616
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305
| | | | - Guy Perkins
- National Center for Microscopy and Imaging Research, University of California San Diego, La Jolla, CA 92093
| | - Hao Zhang
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305
| | - Valeriy Timofeyev
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of California, Davis, CA 95616
| | - Yankun Lyu
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of California, Davis, CA 95616
| | - Daphne A. Diloretto
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of California, Davis, CA 95616
| | - Pauline Trinh
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of California, Davis, CA 95616
| | - Padmini Sirish
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of California, Davis, CA 95616
| | - James L. Overton
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of California, Davis, CA 95616
| | - Wilson Xu
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of California, Davis, CA 95616
| | - Nathan Grainger
- Department of Physiology and Membrane Biology, University of California, Davis, CA 95616
| | - Yang K. Xiang
- Department of Pharmacology, University of California, Davis, CA 95616
| | - Elena N. Dedkova
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California, Davis, CA 95616
| | - Xiao-Dong Zhang
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of California, Davis, CA 95616
| | - Ebenezer N. Yamoah
- Department of Physiology and Cell Biology, University of Nevada, Reno, NV 89557
| | - Manuel F. Navedo
- Department of Pharmacology, University of California, Davis, CA 95616
| | - Phung N. Thai
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of California, Davis, CA 95616
- Department of Physiology and Cell Biology, University of Nevada, Reno, NV 89557
| | - Nipavan Chiamvimonvat
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of California, Davis, CA 95616
- Department of Pharmacology, University of California, Davis, CA 95616
- Department of Veterans Affairs, Northern California Health Care System, Mather, CA 95655
| |
Collapse
|
19
|
Agnew-Svoboda W, Ubina T, Figueroa Z, Wong YC, Vizcarra EA, Roebini B, Wilson EH, Fiacco TA, Riccomagno MM. A genetic tool for the longitudinal study of a subset of post-inflammatory reactive astrocytes. CELL REPORTS METHODS 2022; 2:100276. [PMID: 36046623 PMCID: PMC9421582 DOI: 10.1016/j.crmeth.2022.100276] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Revised: 06/01/2022] [Accepted: 07/22/2022] [Indexed: 11/29/2022]
Abstract
Astrocytes are vital support cells that ensure proper brain function. In brain disease, astrocytes reprogram into a reactive state that alters many of their cellular roles. A long-standing question in the field is whether downregulation of reactive astrocyte (RA) markers during resolution of inflammation is because these astrocytes revert back to a non-reactive state or die and are replaced. This has proven difficult to answer mainly because existing genetic tools cannot distinguish between healthy versus RAs. Here we describe the generation of an inducible genetic tool that can be used to specifically target and label a subset of RAs. Longitudinal analysis of an acute inflammation model using this tool revealed that the previously observed downregulation of RA markers after inflammation is likely due to changes in gene expression and not because of cell death. Our findings suggest that cellular changes associated with astrogliosis after acute inflammation are largely reversible.
Collapse
Affiliation(s)
- William Agnew-Svoboda
- Neuroscience Graduate Program, University of California, Riverside, Riverside, CA 92521, USA
- Department of Molecular, Cell, and Systems Biology, University of California, Riverside, Riverside, CA 92521, USA
| | - Teresa Ubina
- Neuroscience Graduate Program, University of California, Riverside, Riverside, CA 92521, USA
- Department of Molecular, Cell, and Systems Biology, University of California, Riverside, Riverside, CA 92521, USA
| | - Zoe Figueroa
- Neuroscience Graduate Program, University of California, Riverside, Riverside, CA 92521, USA
- Division of Biomedical Sciences, University of California, Riverside, Riverside, CA 92521, USA
| | - Yiu-Cheung Wong
- Department of Molecular, Cell, and Systems Biology, University of California, Riverside, Riverside, CA 92521, USA
| | - Edward A. Vizcarra
- Division of Biomedical Sciences, University of California, Riverside, Riverside, CA 92521, USA
- Biomedical Sciences Graduate Program, University of California, Riverside, Riverside, CA 92521, USA
| | - Bryan Roebini
- Department of Molecular, Cell, and Systems Biology, University of California, Riverside, Riverside, CA 92521, USA
| | - Emma H. Wilson
- Neuroscience Graduate Program, University of California, Riverside, Riverside, CA 92521, USA
- Division of Biomedical Sciences, University of California, Riverside, Riverside, CA 92521, USA
- Biomedical Sciences Graduate Program, University of California, Riverside, Riverside, CA 92521, USA
| | - Todd A. Fiacco
- Neuroscience Graduate Program, University of California, Riverside, Riverside, CA 92521, USA
- Department of Molecular, Cell, and Systems Biology, University of California, Riverside, Riverside, CA 92521, USA
- Biomedical Sciences Graduate Program, University of California, Riverside, Riverside, CA 92521, USA
| | - Martin M. Riccomagno
- Neuroscience Graduate Program, University of California, Riverside, Riverside, CA 92521, USA
- Department of Molecular, Cell, and Systems Biology, University of California, Riverside, Riverside, CA 92521, USA
- Biomedical Sciences Graduate Program, University of California, Riverside, Riverside, CA 92521, USA
| |
Collapse
|
20
|
Mahadevan A, Codadu NK, Parrish RR. Xenon LFP Analysis Platform Is a Novel Graphical User Interface for Analysis of Local Field Potential From Large-Scale MEA Recordings. Front Neurosci 2022; 16:904931. [PMID: 35844228 PMCID: PMC9285004 DOI: 10.3389/fnins.2022.904931] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2022] [Accepted: 06/08/2022] [Indexed: 11/24/2022] Open
Abstract
High-density multi-electrode array (HD-MEA) has enabled neuronal measurements at high spatial resolution to record local field potentials (LFP), extracellular action potentials, and network-wide extracellular recording on an extended spatial scale. While we have advanced recording systems with over 4,000 electrodes capable of recording data at over 20 kHz, it still presents computational challenges to handle, process, extract, and view information from these large recordings. We have created a computational method, and an open-source toolkit built in Python, rendered on a web browser using Plotly’s Dash for extracting and viewing the data and creating interactive visualization. In addition to extracting and viewing entire or small chunks of data sampled at lower or higher frequencies, respectively, it provides a framework to collect user inputs, analyze channel groups, generate raster plots, view quick summary measures for LFP activity, detect and isolate noise channels, and generate plots and visualization in both time and frequency domain. Incorporated into our Graphical User Interface (GUI), we also created a novel seizure detection method, which can be used to detect the onset of seizures in all or a selected group of channels and provide the following measures of seizures: distance, duration, and propagation across the region of interest. We demonstrate the utility of this toolkit, using datasets collected from an HD-MEA device comprising of 4,096 recording electrodes. For the current analysis, we demonstrate the toolkit and methods with a low sampling frequency dataset (300 Hz) and a group of approximately 400 channels. Using this toolkit, we present novel data demonstrating increased seizure propagation speed from brain slices of Scn1aHet mice compared to littermate controls. While there have been advances in HD-MEA recording systems with high spatial and temporal resolution, limited tools are available for researchers to view and process these big datasets. We now provide a user-friendly toolkit to analyze LFP activity obtained from large-scale MEA recordings with translatable applications to EEG recordings and demonstrate the utility of this new graphic user interface with novel biological findings.
Collapse
Affiliation(s)
- Arjun Mahadevan
- Department of Cellular and Molecular Biology, Xenon Pharmaceuticals Inc., Burnaby, BC, Canada
| | - Neela K. Codadu
- Department of Clinical and Experimental Epilepsy, Institute of Neurology, University College London, London, United Kingdom
| | - R. Ryley Parrish
- Department of Cellular and Molecular Biology, Xenon Pharmaceuticals Inc., Burnaby, BC, Canada
- *Correspondence: R. Ryley Parrish,
| |
Collapse
|
21
|
Faraj N, Duinkerken BHP, Carroll EC, Giepmans BNG. Microscopic modulation and analysis of islets of Langerhans in living zebrafish larvae. FEBS Lett 2022; 596:2497-2512. [DOI: 10.1002/1873-3468.14411] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Revised: 04/22/2022] [Accepted: 05/20/2022] [Indexed: 11/08/2022]
Affiliation(s)
- Noura Faraj
- Department of Biomedical Sciences of Cells and Systems, University of Groningen University Medical Center Groningen Groningen 9713AV The Netherlands
| | - B. H. Peter Duinkerken
- Department of Biomedical Sciences of Cells and Systems, University of Groningen University Medical Center Groningen Groningen 9713AV The Netherlands
| | - Elizabeth C. Carroll
- Department of Imaging Physics Delft University of Technology Delft, 2628 CJ The Netherlands
| | - Ben N. G. Giepmans
- Department of Biomedical Sciences of Cells and Systems, University of Groningen University Medical Center Groningen Groningen 9713AV The Netherlands
| |
Collapse
|
22
|
Application of piconewton forces to individual filopodia reveals mechanosensory role of L-type Ca 2+ channels. Biomaterials 2022; 284:121477. [PMID: 35395455 DOI: 10.1016/j.biomaterials.2022.121477] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Accepted: 03/18/2022] [Indexed: 11/02/2022]
Abstract
Filopodia are ubiquitous membrane projections that play crucial role in guiding cell migration on rigid substrates and through extracellular matrix by utilizing yet unknown mechanosensing molecular pathways. As recent studies show that Ca2+ channels localized to filopodia play an important role in regulation of their formation and since some Ca2+ channels are known to be mechanosensitive, force-dependent activity of filopodial Ca2+ channels might be linked to filopodia's mechanosensing function. We tested this hypothesis by monitoring changes in the intra-filopodial Ca2+ level in response to application of stretching force to individual filopodia of several cell types using optical tweezers. Results show that stretching forces of tens of pN strongly promote Ca2+ influx into filopodia, causing persistent Ca2+ oscillations that last for minutes even after the force is released. Several known mechanosensitive Ca2+ channels, such as Piezo 1, Piezo 2 and TRPV4, were found to be dispensable for the observed force-dependent Ca2+ influx, while L-type Ca2+ channels appear to be a key player in the discovered phenomenon. As previous studies have shown that intra-filopodial transient Ca2+ signals play an important role in guidance of cell migration, our results suggest that the force-dependent activation of L-type Ca2+ channels may contribute to this process. Overall, our study reveals an intricate interplay between mechanical forces and Ca2+ signaling in filopodia, providing novel mechanistic insights for the force-dependent filopodia functions in guidance of cell migration.
Collapse
|
23
|
Guan H, Li D, Park HC, Li A, Yue Y, Gau YTA, Li MJ, Bergles DE, Lu H, Li X. Deep-learning two-photon fiberscopy for video-rate brain imaging in freely-behaving mice. Nat Commun 2022; 13:1534. [PMID: 35318318 PMCID: PMC8940941 DOI: 10.1038/s41467-022-29236-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Accepted: 03/01/2022] [Indexed: 01/04/2023] Open
Abstract
Scanning two-photon (2P) fiberscopes (also termed endomicroscopes) have the potential to transform our understanding of how discrete neural activity patterns result in distinct behaviors, as they are capable of high resolution, sub cellular imaging yet small and light enough to allow free movement of mice. However, their acquisition speed is currently suboptimal, due to opto-mechanical size and weight constraints. Here we demonstrate significant advances in 2P fiberscopy that allow high resolution imaging at high speeds (26 fps) in freely-behaving mice. A high-speed scanner and a down-sampling scheme are developed to boost imaging speed, and a deep learning (DL) algorithm is introduced to recover image quality. For the DL algorithm, a two-stage learning transfer strategy is established to generate proper training datasets for enhancing the quality of in vivo images. Implementation enables video-rate imaging at ~26 fps, representing 10-fold improvement in imaging speed over the previous 2P fiberscopy technology while maintaining a high signal-to-noise ratio and imaging resolution. This DL-assisted 2P fiberscope is capable of imaging the arousal-induced activity changes in populations of layer2/3 pyramidal neurons in the primary motor cortex of freely-behaving mice, providing opportunities to define the neural basis of behavior.
Collapse
Affiliation(s)
- Honghua Guan
- Department of Electrical and Computer Engineering, Johns Hopkins University, Baltimore, MD, 21218, USA
| | - Dawei Li
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Hyeon-Cheol Park
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Ang Li
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Yuanlei Yue
- Department of Pharmacology and Physiology, School of Medicine and Health Sciences, George Washington University, Washington, DC, 20052, USA
| | - Yung-Tian A Gau
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Ming-Jun Li
- Science and Technology Division, Corning Incorporated, Corning, NY, 14831, USA
| | - Dwight E Bergles
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
- Johns Hopkins Kavli Neuroscience Discovery Institute, Baltimore, MD, 21218, USA
| | - Hui Lu
- Department of Pharmacology and Physiology, School of Medicine and Health Sciences, George Washington University, Washington, DC, 20052, USA
| | - Xingde Li
- Department of Electrical and Computer Engineering, Johns Hopkins University, Baltimore, MD, 21218, USA.
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA.
- Johns Hopkins Kavli Neuroscience Discovery Institute, Baltimore, MD, 21218, USA.
| |
Collapse
|
24
|
Approach to map nanotopography of cell surface receptors. Commun Biol 2022; 5:218. [PMID: 35264712 PMCID: PMC8907216 DOI: 10.1038/s42003-022-03152-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Accepted: 02/09/2022] [Indexed: 12/18/2022] Open
Abstract
Cells communicate with their environment via surface receptors, but nanoscopic receptor organization with respect to complex cell surface morphology remains unclear. This is mainly due to a lack of accessible, robust and high-resolution methods. Here, we present an approach for mapping the topography of receptors at the cell surface with nanometer precision. The method involves coating glass coverslips with glycine, which preserves the fine membrane morphology while allowing immobilized cells to be positioned close to the optical surface. We developed an advanced and simplified algorithm for the analysis of single-molecule localization data acquired in a biplane detection scheme. These advancements enable direct and quantitative mapping of protein distribution on ruffled plasma membranes with near isotropic 3D nanometer resolution. As demonstrated successfully for CD4 and CD45 receptors, the described workflow is a straightforward quantitative technique to study molecules and their interactions at the complex surface nanomorphology of differentiated metazoan cells. A super-resolution localisation-based method is shown to map receptor topography in immune cells, which allows quantitative study of molecules and their interactions at the complex surface nanomorphology of cells.
Collapse
|
25
|
Bais S, Norwillo A, Ruthel G, Herbert DR, Freedman BD, Greenberg RM. Schistosome TRPML channels play a role in neuromuscular activity and tegumental integrity. Biochimie 2022; 194:108-117. [PMID: 34990770 PMCID: PMC8950431 DOI: 10.1016/j.biochi.2021.12.018] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Revised: 12/16/2021] [Accepted: 12/31/2021] [Indexed: 11/02/2022]
Abstract
Schistosomiasis is a neglected tropical disease caused by parasitic flatworms of the genus Schistosoma. Mono-therapeutic treatment of this disease with the drug praziquantel, presents challenges such as inactivity against immature worms and inability to prevent reinfection. Importantly, ion channels are important targets for many current anthelmintics. Transient receptor potential (TRP) channels are important mediators of sensory signals with marked effects on cellular functions and signaling pathways. TRPML channels are a class of Ca2+-permeable TRP channels expressed on endolysosomal membranes. They regulate lysosomal function and trafficking, among other functions. Schistosoma mansoni is predicted to have a single TRPML gene (SmTRPML) with two splice variants differing by 12 amino acids. This study focuses on exploring the physiological properties of SmTRPML channels to better understand their role in schistosomes. In mammalian cells expressing SmTRPML, TRPML activators elicit a rise in intracellular Ca2+. In these cells, SmTRPML localizes both to lysosomes and the plasma membrane. These same TRPML activators elicit an increase in adult worm motility that is dependent on SmTRPML expression, indicating a role for these channels in parasite neuromuscular activity. Suppression of SmTRPML in adult worms, or exposure of adult worms to TRPML inhibitors, results in tegumental vacuolations, balloon-like surface exudates, and membrane blebbing, similar to that found following TRPML loss in other organisms. Together, these findings indicate that SmTRPML may regulate the function of the schistosome endolysosomal system. Further, the role of SmTRPML in neuromuscular activity and in parasite tegumental integrity establishes this channel as a candidate anti-schistosome drug target.
Collapse
Affiliation(s)
- Swarna Bais
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, USA.
| | - Abigail Norwillo
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, USA
| | - Gordon Ruthel
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, USA
| | - De'Broski R Herbert
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, USA
| | - Bruce D Freedman
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, USA
| | - Robert M Greenberg
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, USA
| |
Collapse
|
26
|
Peng Y, Schöneberg N, Esposito MS, Geiger JRP, Sharott A, Tovote P. Current approaches to characterize micro- and macroscale circuit mechanisms of Parkinson's disease in rodent models. Exp Neurol 2022; 351:114008. [PMID: 35149118 PMCID: PMC7612860 DOI: 10.1016/j.expneurol.2022.114008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Revised: 01/17/2022] [Accepted: 02/04/2022] [Indexed: 11/24/2022]
Abstract
Accelerating technological progress in experimental neuroscience is increasing the scale as well as specificity of both observational and perturbational approaches to study circuit physiology. While these techniques have also been used to study disease mechanisms, a wider adoption of these approaches in the field of experimental neurology would greatly facilitate our understanding of neurological dysfunctions and their potential treatments at cellular and circuit level. In this review, we will introduce classic and novel methods ranging from single-cell electrophysiological recordings to state-of-the-art calcium imaging and cell-type specific optogenetic or chemogenetic stimulation. We will focus on their application in rodent models of Parkinson’s disease while also presenting their use in the context of motor control and basal ganglia function. By highlighting the scope and limitations of each method, we will discuss how they can be used to study pathophysiological mechanisms at local and global circuit levels and how novel frameworks can help to bridge these scales.
Collapse
Affiliation(s)
- Yangfan Peng
- Institute of Neurophysiology, Charité - Universitätsmedizin Berlin, Freie Universität Berlin and Humboldt Universität zu Berlin, Charitéplatz 1, 10117 Berlin, Germany; Department of Neurology, Charité - Universitätsmedizin Berlin, Freie Universität Berlin and Humboldt Universität zu Berlin, Charitéplatz 1, 10117 Berlin, Germany; MRC Brain Network Dynamics Unit, University of Oxford, Mansfield Road, Oxford OX1 3TH, United Kingdom.
| | - Nina Schöneberg
- Institute of Clinical Neurobiology, University Hospital Wuerzburg, Versbacher Str. 5, 97078 Wuerzburg, Germany
| | - Maria Soledad Esposito
- Medical Physics Department, Centro Atomico Bariloche, Comision Nacional de Energia Atomica (CNEA), Consejo Nacional de Investigaciones Cientificas y Tecnicas (CONICET), Av. E. Bustillo 9500, R8402AGP San Carlos de Bariloche, Rio Negro, Argentina
| | - Jörg R P Geiger
- Institute of Neurophysiology, Charité - Universitätsmedizin Berlin, Freie Universität Berlin and Humboldt Universität zu Berlin, Charitéplatz 1, 10117 Berlin, Germany
| | - Andrew Sharott
- MRC Brain Network Dynamics Unit, University of Oxford, Mansfield Road, Oxford OX1 3TH, United Kingdom
| | - Philip Tovote
- Institute of Clinical Neurobiology, University Hospital Wuerzburg, Versbacher Str. 5, 97078 Wuerzburg, Germany; Center for Mental Health, University of Wuerzburg, Margarete-Höppel-Platz 1, 97080 Wuerzburg, Germany.
| |
Collapse
|
27
|
Genetically encoded photo-switchable molecular sensors for optoacoustic and super-resolution imaging. Nat Biotechnol 2022; 40:598-605. [PMID: 34845372 PMCID: PMC9005348 DOI: 10.1038/s41587-021-01100-5] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Accepted: 09/17/2021] [Indexed: 02/07/2023]
Abstract
Reversibly photo-switchable proteins are essential for many super-resolution fluorescence microscopic and optoacoustic imaging methods. However, they have yet to be used as sensors that measure the distribution of specific analytes at the nanoscale or in the tissues of live animals. Here we constructed the prototype of a photo-switchable Ca2+ sensor based on GCaMP5G that can be switched with 405/488-nm light and describe its molecular mechanisms at the structural level, including the importance of the interaction of the core barrel structure of the fluorescent protein with the Ca2+ receptor moiety. We demonstrate super-resolution imaging of Ca2+ concentration in cultured cells and optoacoustic Ca2+ imaging in implanted tumor cells in mice under controlled Ca2+ conditions. Finally, we show the generalizability of the concept by constructing examples of photo-switching maltose and dopamine sensors based on periplasmatic binding protein and G-protein-coupled receptor-based sensors.
Collapse
|
28
|
Hamilos AE, Spedicato G, Hong Y, Sun F, Li Y, Assad J. Slowly evolving dopaminergic activity modulates the moment-to-moment probability of reward-related self-timed movements. eLife 2021; 10:62583. [PMID: 34939925 PMCID: PMC8860451 DOI: 10.7554/elife.62583] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2020] [Accepted: 12/21/2021] [Indexed: 11/13/2022] Open
Abstract
Clues from human movement disorders have long suggested that the neurotransmitter dopamine plays a role in motor control, but how the endogenous dopaminergic system influences movement is unknown. Here we examined the relationship between dopaminergic signaling and the timing of reward-related movements in mice. Animals were trained to initiate licking after a self-timed interval following a start-timing cue; reward was delivered in response to movements initiated after a criterion time. The movement time was variable from trial-to-trial, as expected from previous studies. Surprisingly, dopaminergic signals ramped-up over seconds between the start-timing cue and the self-timed movement, with variable dynamics that predicted the movement/reward time on single trials. Steeply rising signals preceded early lick-initiation, whereas slowly rising signals preceded later initiation. Higher baseline signals also predicted earlier self-timed movements. Optogenetic activation of dopamine neurons during self-timing did not trigger immediate movements, but rather caused systematic early-shifting of movement initiation, whereas inhibition caused late-shifting, as if modulating the probability of movement. Consistent with this view, the dynamics of the endogenous dopaminergic signals quantitatively predicted the moment-by-moment probability of movement initiation on single trials. We propose that ramping dopaminergic signals, likely encoding dynamic reward expectation, can modulate the decision of when to move.
Collapse
Affiliation(s)
- Allison E Hamilos
- Department of Neurobiology, Harvard Medical School, Boston, United States
| | - Giulia Spedicato
- Department of Neurobiology, Harvard Medical School, Boston, United States
| | - Ye Hong
- Department of Neurobiology, Harvard Medical School, Boston, United States
| | - Fangmiao Sun
- State Key Laboratory of Membrane Biology, Peking University School of Life Science, Beijing, China
| | - Yulong Li
- State Key Laboratory of Membrane Biology, Peiking University School of Life Sciences, Beijing, China
| | - John Assad
- Department of Neurobiology, Harvard Medical School, Boston, United States
| |
Collapse
|
29
|
Clough M, Chen IA, Park SW, Ahrens AM, Stirman JN, Smith SL, Chen JL. Flexible simultaneous mesoscale two-photon imaging of neural activity at high speeds. Nat Commun 2021; 12:6638. [PMID: 34789730 PMCID: PMC8599611 DOI: 10.1038/s41467-021-26737-3] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Accepted: 10/05/2021] [Indexed: 12/02/2022] Open
Abstract
Understanding brain function requires monitoring local and global brain dynamics. Two-photon imaging of the brain across mesoscopic scales has presented trade-offs between imaging area and acquisition speed. We describe a flexible cellular resolution two-photon microscope capable of simultaneous video rate acquisition of four independently targetable brain regions spanning an approximate five-millimeter field of view. With this system, we demonstrate the ability to measure calcium activity across mouse sensorimotor cortex at behaviorally relevant timescales.
Collapse
Affiliation(s)
- Mitchell Clough
- Department of Biomedical Engineering, Boston University, Boston, MA, 02215, USA
| | - Ichun Anderson Chen
- Department of Biomedical Engineering, Boston University, Boston, MA, 02215, USA
- Center for Neurophotonics, Boston University, Boston, MA, 02215, USA
| | - Seong-Wook Park
- Department of Biomedical Engineering, Boston University, Boston, MA, 02215, USA
- Center for Neurophotonics, Boston University, Boston, MA, 02215, USA
| | - Allison M Ahrens
- Department of Biology, Boston University, Boston, MA, 02215, USA
| | - Jeffrey N Stirman
- Neuroscience Center, University of North Carolina School of Medicine, Chapel Hill, NC, 27599, USA
| | - Spencer L Smith
- Department of Electrical and Computer Engineering, University of California Santa Barbara, Santa Barbara, CA, 93106, USA
| | - Jerry L Chen
- Department of Biomedical Engineering, Boston University, Boston, MA, 02215, USA.
- Center for Neurophotonics, Boston University, Boston, MA, 02215, USA.
- Department of Biology, Boston University, Boston, MA, 02215, USA.
| |
Collapse
|
30
|
Deng X, Yao XQ, Berglund K, Dong B, Ouedraogo D, Ghane MA, Zhuo Y, McBean C, Wei ZZ, Gozem S, Yu SP, Wei L, Fang N, Mabb AM, Gadda G, Hamelberg D, Yang JJ. Tuning Protein Dynamics to Sense Rapid Endoplasmic-Reticulum Calcium Dynamics. Angew Chem Int Ed Engl 2021; 60:23289-23298. [PMID: 34436811 DOI: 10.1002/anie.202108443] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Indexed: 11/11/2022]
Abstract
Multi-scale calcium (Ca2+ ) dynamics, exhibiting wide-ranging temporal kinetics, constitutes a ubiquitous mode of signal transduction. We report a novel endoplasmic-reticulum (ER)-targeted Ca2+ indicator, R-CatchER, which showed superior kinetics in vitro (koff ≥2×103 s-1 , kon ≥7×106 M-1 s-1 ) and in multiple cell types. R-CatchER captured spatiotemporal ER Ca2+ dynamics in neurons and hotspots at dendritic branchpoints, enabled the first report of ER Ca2+ oscillations mediated by calcium sensing receptors (CaSRs), and revealed ER Ca2+ -based functional cooperativity of CaSR. We elucidate the mechanism of R-CatchER and propose a principle to rationally design genetically encoded Ca2+ indicators with a single Ca2+ -binding site and fast kinetics by tuning rapid fluorescent-protein dynamics and the electrostatic potential around the chromophore. The design principle is supported by the development of G-CatchER2, an upgrade of our previous (G-)CatchER with improved dynamic range. Our work may facilitate protein design, visualizing Ca2+ dynamics, and drug discovery.
Collapse
Affiliation(s)
- Xiaonan Deng
- Department of Chemistry, Center for Diagnostics and Therapeutics, Advanced Translational Imaging Facility, Georgia State University, 50 Decatur Street, 552 NSC, Atlanta, GA, 30303, USA
| | - Xin-Qiu Yao
- Department of Chemistry, Center for Diagnostics and Therapeutics, Advanced Translational Imaging Facility, Georgia State University, 50 Decatur Street, 552 NSC, Atlanta, GA, 30303, USA
| | - Ken Berglund
- Department of Neurosurgery, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Bin Dong
- Department of Chemistry, Center for Diagnostics and Therapeutics, Advanced Translational Imaging Facility, Georgia State University, 50 Decatur Street, 552 NSC, Atlanta, GA, 30303, USA
| | - Daniel Ouedraogo
- Department of Chemistry, Center for Diagnostics and Therapeutics, Advanced Translational Imaging Facility, Georgia State University, 50 Decatur Street, 552 NSC, Atlanta, GA, 30303, USA
| | - Mohammad A Ghane
- Neuroscience Institute, Georgia State University, Atlanta, GA, 30303, USA.,Center for Behavioral Neuroscience, Georgia State University, Atlanta, GA, 30303, USA
| | - You Zhuo
- Department of Chemistry, Center for Diagnostics and Therapeutics, Advanced Translational Imaging Facility, Georgia State University, 50 Decatur Street, 552 NSC, Atlanta, GA, 30303, USA
| | - Cheyenne McBean
- Department of Chemistry, Center for Diagnostics and Therapeutics, Advanced Translational Imaging Facility, Georgia State University, 50 Decatur Street, 552 NSC, Atlanta, GA, 30303, USA
| | - Zheng Zachory Wei
- Department of Anesthesiology, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Samer Gozem
- Department of Chemistry, Center for Diagnostics and Therapeutics, Advanced Translational Imaging Facility, Georgia State University, 50 Decatur Street, 552 NSC, Atlanta, GA, 30303, USA
| | - Shan P Yu
- Department of Anesthesiology, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Ling Wei
- Department of Anesthesiology, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Ning Fang
- Department of Chemistry, Center for Diagnostics and Therapeutics, Advanced Translational Imaging Facility, Georgia State University, 50 Decatur Street, 552 NSC, Atlanta, GA, 30303, USA
| | - Angela M Mabb
- Neuroscience Institute, Georgia State University, Atlanta, GA, 30303, USA.,Center for Behavioral Neuroscience, Georgia State University, Atlanta, GA, 30303, USA
| | - Giovanni Gadda
- Department of Chemistry, Center for Diagnostics and Therapeutics, Advanced Translational Imaging Facility, Georgia State University, 50 Decatur Street, 552 NSC, Atlanta, GA, 30303, USA
| | - Donald Hamelberg
- Department of Chemistry, Center for Diagnostics and Therapeutics, Advanced Translational Imaging Facility, Georgia State University, 50 Decatur Street, 552 NSC, Atlanta, GA, 30303, USA
| | - Jenny J Yang
- Department of Chemistry, Center for Diagnostics and Therapeutics, Advanced Translational Imaging Facility, Georgia State University, 50 Decatur Street, 552 NSC, Atlanta, GA, 30303, USA
| |
Collapse
|
31
|
Deng X, Yao X, Berglund K, Dong B, Ouedraogo D, Ghane MA, Zhuo Y, McBean C, Wei ZZ, Gozem S, Yu SP, Wei L, Fang N, Mabb AM, Gadda G, Hamelberg D, Yang JJ. Tuning Protein Dynamics to Sense Rapid Endoplasmic‐Reticulum Calcium Dynamics. Angew Chem Int Ed Engl 2021. [DOI: 10.1002/ange.202108443] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Affiliation(s)
- Xiaonan Deng
- Department of Chemistry Center for Diagnostics and Therapeutics Advanced Translational Imaging Facility Georgia State University 50 Decatur Street, 552 NSC Atlanta GA 30303 USA
| | - Xin‐Qiu Yao
- Department of Chemistry Center for Diagnostics and Therapeutics Advanced Translational Imaging Facility Georgia State University 50 Decatur Street, 552 NSC Atlanta GA 30303 USA
| | - Ken Berglund
- Department of Neurosurgery Emory University School of Medicine Atlanta GA 30322 USA
| | - Bin Dong
- Department of Chemistry Center for Diagnostics and Therapeutics Advanced Translational Imaging Facility Georgia State University 50 Decatur Street, 552 NSC Atlanta GA 30303 USA
| | - Daniel Ouedraogo
- Department of Chemistry Center for Diagnostics and Therapeutics Advanced Translational Imaging Facility Georgia State University 50 Decatur Street, 552 NSC Atlanta GA 30303 USA
| | - Mohammad A. Ghane
- Neuroscience Institute Georgia State University Atlanta GA 30303 USA
- Center for Behavioral Neuroscience Georgia State University Atlanta GA 30303 USA
| | - You Zhuo
- Department of Chemistry Center for Diagnostics and Therapeutics Advanced Translational Imaging Facility Georgia State University 50 Decatur Street, 552 NSC Atlanta GA 30303 USA
| | - Cheyenne McBean
- Department of Chemistry Center for Diagnostics and Therapeutics Advanced Translational Imaging Facility Georgia State University 50 Decatur Street, 552 NSC Atlanta GA 30303 USA
| | - Zheng Zachory Wei
- Department of Anesthesiology Emory University School of Medicine Atlanta GA 30322 USA
| | - Samer Gozem
- Department of Chemistry Center for Diagnostics and Therapeutics Advanced Translational Imaging Facility Georgia State University 50 Decatur Street, 552 NSC Atlanta GA 30303 USA
| | - Shan P. Yu
- Department of Anesthesiology Emory University School of Medicine Atlanta GA 30322 USA
| | - Ling Wei
- Department of Anesthesiology Emory University School of Medicine Atlanta GA 30322 USA
| | - Ning Fang
- Department of Chemistry Center for Diagnostics and Therapeutics Advanced Translational Imaging Facility Georgia State University 50 Decatur Street, 552 NSC Atlanta GA 30303 USA
| | - Angela M. Mabb
- Neuroscience Institute Georgia State University Atlanta GA 30303 USA
- Center for Behavioral Neuroscience Georgia State University Atlanta GA 30303 USA
| | - Giovanni Gadda
- Department of Chemistry Center for Diagnostics and Therapeutics Advanced Translational Imaging Facility Georgia State University 50 Decatur Street, 552 NSC Atlanta GA 30303 USA
| | - Donald Hamelberg
- Department of Chemistry Center for Diagnostics and Therapeutics Advanced Translational Imaging Facility Georgia State University 50 Decatur Street, 552 NSC Atlanta GA 30303 USA
| | - Jenny J. Yang
- Department of Chemistry Center for Diagnostics and Therapeutics Advanced Translational Imaging Facility Georgia State University 50 Decatur Street, 552 NSC Atlanta GA 30303 USA
| |
Collapse
|
32
|
Trautmann EM, O'Shea DJ, Sun X, Marshel JH, Crow A, Hsueh B, Vesuna S, Cofer L, Bohner G, Allen W, Kauvar I, Quirin S, MacDougall M, Chen Y, Whitmire MP, Ramakrishnan C, Sahani M, Seidemann E, Ryu SI, Deisseroth K, Shenoy KV. Dendritic calcium signals in rhesus macaque motor cortex drive an optical brain-computer interface. Nat Commun 2021; 12:3689. [PMID: 34140486 PMCID: PMC8211867 DOI: 10.1038/s41467-021-23884-5] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2019] [Accepted: 05/19/2021] [Indexed: 02/05/2023] Open
Abstract
Calcium imaging is a powerful tool for recording from large populations of neurons in vivo. Imaging in rhesus macaque motor cortex can enable the discovery of fundamental principles of motor cortical function and can inform the design of next generation brain-computer interfaces (BCIs). Surface two-photon imaging, however, cannot presently access somatic calcium signals of neurons from all layers of macaque motor cortex due to photon scattering. Here, we demonstrate an implant and imaging system capable of chronic, motion-stabilized two-photon imaging of neuronal calcium signals from macaques engaged in a motor task. By imaging apical dendrites, we achieved optical access to large populations of deep and superficial cortical neurons across dorsal premotor (PMd) and gyral primary motor (M1) cortices. Dendritic signals from individual neurons displayed tuning for different directions of arm movement. Combining several technical advances, we developed an optical BCI (oBCI) driven by these dendritic signalswhich successfully decoded movement direction online. By fusing two-photon functional imaging with CLARITY volumetric imaging, we verified that many imaged dendrites which contributed to oBCI decoding originated from layer 5 output neurons, including a putative Betz cell. This approach establishes new opportunities for studying motor control and designing BCIs via two photon imaging.
Collapse
Affiliation(s)
- Eric M Trautmann
- Neurosciences Graduate Program, Stanford University, Stanford, CA, USA.
- Department of Electrical Engineering, Stanford University, Stanford, CA, USA.
| | - Daniel J O'Shea
- Neurosciences Graduate Program, Stanford University, Stanford, CA, USA.
- Department of Electrical Engineering, Stanford University, Stanford, CA, USA.
| | - Xulu Sun
- Department of Biology, Stanford University, Stanford, CA, USA.
| | - James H Marshel
- Department of Bioengineering, Stanford University, Stanford, CA, USA
| | - Ailey Crow
- Department of Bioengineering, Stanford University, Stanford, CA, USA
| | - Brian Hsueh
- Neurosciences Graduate Program, Stanford University, Stanford, CA, USA
| | - Sam Vesuna
- Department of Bioengineering, Stanford University, Stanford, CA, USA
| | - Lucas Cofer
- Department of Electrical Engineering, Stanford University, Stanford, CA, USA
| | - Gergő Bohner
- Gatsby Computational Neuroscience Unit, University College London, London, UK
| | - Will Allen
- Neurosciences Graduate Program, Stanford University, Stanford, CA, USA
| | - Isaac Kauvar
- Neurosciences Graduate Program, Stanford University, Stanford, CA, USA
| | - Sean Quirin
- Department of Bioengineering, Stanford University, Stanford, CA, USA
| | | | - Yuzhi Chen
- Center for Perceptual Systems, University of Texas, Austin, TX, USA
- Department of Psychology, University of Texas, Austin, TX, USA
- Department of Neuroscience, University of Texas, Austin, TX, USA
| | - Matthew P Whitmire
- Center for Perceptual Systems, University of Texas, Austin, TX, USA
- Department of Psychology, University of Texas, Austin, TX, USA
- Department of Neuroscience, University of Texas, Austin, TX, USA
| | | | - Maneesh Sahani
- Gatsby Computational Neuroscience Unit, University College London, London, UK
| | - Eyal Seidemann
- Center for Perceptual Systems, University of Texas, Austin, TX, USA
- Department of Psychology, University of Texas, Austin, TX, USA
- Department of Neuroscience, University of Texas, Austin, TX, USA
| | - Stephen I Ryu
- Department of Electrical Engineering, Stanford University, Stanford, CA, USA
- Department of Neurosurgery, Palo Alto Medical Foundation, Palo Alto, CA, USA
| | - Karl Deisseroth
- Neurosciences Graduate Program, Stanford University, Stanford, CA, USA.
- Department of Bioengineering, Stanford University, Stanford, CA, USA.
- Department of Psychiatry and Behavioral Science, Stanford University, Stanford, CA, USA.
- Howard Hughes Medical Institute, Stanford University, Stanford, CA, USA.
- Wu Tsai Neuroscience Institute, Stanford University, Stanford, CA, USA.
| | - Krishna V Shenoy
- Neurosciences Graduate Program, Stanford University, Stanford, CA, USA.
- Department of Electrical Engineering, Stanford University, Stanford, CA, USA.
- Department of Bioengineering, Stanford University, Stanford, CA, USA.
- Howard Hughes Medical Institute, Stanford University, Stanford, CA, USA.
- Wu Tsai Neuroscience Institute, Stanford University, Stanford, CA, USA.
- Department of Neurobiology, Stanford University, Stanford, CA, USA.
| |
Collapse
|
33
|
Kaszas A, Szalay G, Slézia A, Bojdán A, Vanzetta I, Hangya B, Rózsa B, O'Connor R, Moreau D. Two-photon GCaMP6f imaging of infrared neural stimulation evoked calcium signals in mouse cortical neurons in vivo. Sci Rep 2021; 11:9775. [PMID: 33963220 PMCID: PMC8105372 DOI: 10.1038/s41598-021-89163-x] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Accepted: 04/22/2021] [Indexed: 02/07/2023] Open
Abstract
Infrared neural stimulation is a promising tool for stimulating the brain because it can be used to excite with high spatial precision without the need of delivering or inserting any exogenous agent into the tissue. Very few studies have explored its use in the brain, as most investigations have focused on sensory or motor nerve stimulation. Using intravital calcium imaging with the genetically encoded calcium indicator GCaMP6f, here we show that the application of infrared neural stimulation induces intracellular calcium signals in Layer 2/3 neurons in mouse cortex in vivo. The number of neurons exhibiting infrared-induced calcium response as well as the amplitude of those signals are shown to be both increasing with the energy density applied. By studying as well the spatial extent of the stimulation, we show that reproducibility of the stimulation is achieved mainly in the central part of the infrared beam path. Stimulating in vivo at such a degree of precision and without any exogenous chromophores enables multiple applications, from mapping the brain's connectome to applications in systems neuroscience and the development of new therapeutic tools for investigating the pathological brain.
Collapse
Affiliation(s)
- Attila Kaszas
- Mines Saint-Etienne, Centre CMP, Département BEL, F - 13541, Gardanne, France
- Institut de Neurosciences de la Timone, CNRS UMR 7289 & Aix-Marseille Université, 13005, Marseille, France
| | - Gergely Szalay
- Laboratory of 3D Functional Network and Dendritic Imaging, Institute of Experimental Medicine, Budapest, 1083, Hungary
| | - Andrea Slézia
- Mines Saint-Etienne, Centre CMP, Département BEL, F - 13541, Gardanne, France
- Lendület Laboratory of Systems Neuroscience, Institute of Experimental Medicine, Budapest, 1083, Hungary
| | - Alexandra Bojdán
- Laboratory of 3D Functional Network and Dendritic Imaging, Institute of Experimental Medicine, Budapest, 1083, Hungary
| | - Ivo Vanzetta
- Institut de Neurosciences de la Timone, CNRS UMR 7289 & Aix-Marseille Université, 13005, Marseille, France
| | - Balázs Hangya
- Lendület Laboratory of Systems Neuroscience, Institute of Experimental Medicine, Budapest, 1083, Hungary
| | - Balázs Rózsa
- Laboratory of 3D Functional Network and Dendritic Imaging, Institute of Experimental Medicine, Budapest, 1083, Hungary
- Two-Photon Laboratory, Faculty of Information Technology, Pázmány Péter Catholic University, Budapest, 1083, Hungary
| | - Rodney O'Connor
- Mines Saint-Etienne, Centre CMP, Département BEL, F - 13541, Gardanne, France
- Institut de Neurosciences de la Timone, CNRS UMR 7289 & Aix-Marseille Université, 13005, Marseille, France
| | - David Moreau
- Mines Saint-Etienne, Centre CMP, Département BEL, F - 13541, Gardanne, France.
| |
Collapse
|
34
|
Adelizzi B, Gielen V, Le Saux T, Dedecker P, Jullien L. Quantitative Model for Reversibly Photoswitchable Sensors. ACS Sens 2021; 6:1157-1165. [PMID: 33565309 PMCID: PMC8008439 DOI: 10.1021/acssensors.0c02414] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Accepted: 01/21/2021] [Indexed: 12/11/2022]
Abstract
Composed of a reversibly photoswitchable unit allosterically linked to a sensing module, reversibly photoswitchable sensors (rs-sensors) represent a new and attractive strategy to quantitatively read-out analyte concentrations. However, their kinetic response to illumination is complex, and much attention is required from the design to the application steps. Here, we exploit a generic kinetic model of rs-sensors which enables us to point to key thermokinetic parameters, such as dissociation constants and kinetic rates for exchange toward the analyte, and cross-sections for photoswitching. The application of the model allows to evaluate the robustness of the analyzed parameters and to introduce a methodology for their reliable use. Model and methodology have been experimentally tested on a newly reported calcium sensor based on a reversibly photoswitchable green fluorescent protein allosterically linked to a calcium-sensing module integrating calmodulin and an RS20 peptide.
Collapse
Affiliation(s)
- Beatrice Adelizzi
- PASTEUR,
Département de Chimie, École
Normale Supérieure, PSL University, Sorbonne Université,
CNRS, 24, rue Lhomond, Paris 75005, France
| | - Vincent Gielen
- Laboratory
for Nanobiology, Department of Chemistry, KU Leuven, Celestijnenlaan
200G, Heverlee 3001, Belgium
| | - Thomas Le Saux
- PASTEUR,
Département de Chimie, École
Normale Supérieure, PSL University, Sorbonne Université,
CNRS, 24, rue Lhomond, Paris 75005, France
| | - Peter Dedecker
- Laboratory
for Nanobiology, Department of Chemistry, KU Leuven, Celestijnenlaan
200G, Heverlee 3001, Belgium
| | - Ludovic Jullien
- PASTEUR,
Département de Chimie, École
Normale Supérieure, PSL University, Sorbonne Université,
CNRS, 24, rue Lhomond, Paris 75005, France
| |
Collapse
|
35
|
Li ES, Saha MS. Optimizing Calcium Detection Methods in Animal Systems: A Sandbox for Synthetic Biology. Biomolecules 2021; 11:343. [PMID: 33668387 PMCID: PMC7996158 DOI: 10.3390/biom11030343] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Revised: 02/19/2021] [Accepted: 02/21/2021] [Indexed: 12/16/2022] Open
Abstract
Since the 1970s, the emergence and expansion of novel methods for calcium ion (Ca2+) detection have found diverse applications in vitro and in vivo across a series of model animal systems. Matched with advances in fluorescence imaging techniques, the improvements in the functional range and stability of various calcium indicators have significantly enhanced more accurate study of intracellular Ca2+ dynamics and its effects on cell signaling, growth, differentiation, and regulation. Nonetheless, the current limitations broadly presented by organic calcium dyes, genetically encoded calcium indicators, and calcium-responsive nanoparticles suggest a potential path toward more rapid optimization by taking advantage of a synthetic biology approach. This engineering-oriented discipline applies principles of modularity and standardization to redesign and interrogate endogenous biological systems. This review will elucidate how novel synthetic biology technologies constructed for eukaryotic systems can offer a promising toolkit for interfacing with calcium signaling and overcoming barriers in order to accelerate the process of Ca2+ detection optimization.
Collapse
Affiliation(s)
| | - Margaret S. Saha
- Department of Biology, College of William and Mary, Williamsburg, VA 23185, USA;
| |
Collapse
|
36
|
P2X-GCaMPs as Versatile Tools for Imaging Extracellular ATP Signaling. eNeuro 2021; 8:ENEURO.0185-20.2020. [PMID: 33380526 PMCID: PMC7877454 DOI: 10.1523/eneuro.0185-20.2020] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2020] [Revised: 11/02/2020] [Accepted: 12/07/2020] [Indexed: 11/30/2022] Open
Abstract
ATP is an extracellular signaling molecule involved in numerous physiological and pathologic processes. However, in situ characterization of the spatiotemporal dynamic of extracellular ATP is still challenging because of the lack of sensor with appropriate specificity, sensitivity, and kinetics. Here, we report the development of biosensors based on the fusion of cation permeable ATP receptors (P2X) to genetically encoded calcium sensors [genetically encoded calcium indicator (GECI)]. By combining the features of P2X receptors with the high signal-to-noise ratio of GECIs, we generated ultrasensitive green and red fluorescent sniffers that detect nanomolar ATP concentrations in situ and also enable the tracking of P2X receptor activity. We provide the proof of concept that these sensors can dynamically track ATP release evoked by depolarization in mouse neurons or by extracellular hypotonicity. Targeting these P2X-based biosensors to diverse cell types should advance our knowledge of extracellular ATP dynamics in vivo.
Collapse
|
37
|
Weigand C, Kim SH, Brown E, Medina E, Mares M, Miller G, Harper JF, Choi WG. A Ratiometric Calcium Reporter CGf Reveals Calcium Dynamics Both in the Single Cell and Whole Plant Levels Under Heat Stress. FRONTIERS IN PLANT SCIENCE 2021; 12:777975. [PMID: 34975960 PMCID: PMC8718611 DOI: 10.3389/fpls.2021.777975] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Accepted: 11/17/2021] [Indexed: 05/02/2023]
Abstract
Land plants evolved to quickly sense and adapt to temperature changes, such as hot days and cold nights. Given that calcium (Ca2+) signaling networks are implicated in most abiotic stress responses, heat-triggered changes in cytosolic Ca2+ were investigated in Arabidopsis leaves and pollen. Plants were engineered with a reporter called CGf, a ratiometric, genetically encoded Ca2+ reporter with an mCherry reference domain fused to an intensiometric Ca2+ reporter GCaMP6f. Relative changes in [Ca2+]cyt were estimated based on CGf's apparent K D around 220 nM. The ratiometric output provided an opportunity to compare Ca2+ dynamics between different tissues, cell types, or subcellular locations. In leaves, CGf detected heat-triggered cytosolic Ca2+ signals, comprised of three different signatures showing similarly rapid rates of Ca2+ influx followed by differing rates of efflux (50% durations ranging from 5 to 19 min). These heat-triggered Ca2+ signals were approximately 1.5-fold greater in magnitude than blue light-triggered signals in the same leaves. In contrast, growing pollen tubes showed two different heat-triggered responses. Exposure to heat caused tip-focused steady growth [Ca2+]cyt oscillations to shift to a pattern characteristic of a growth arrest (22%), or an almost undetectable [Ca2+]cyt (78%). Together, these contrasting examples of heat-triggered Ca2+ responses in leaves and pollen highlight the diversity of Ca2+ signals in plants, inviting speculations about their differing kinetic features and biological functions.
Collapse
Affiliation(s)
- Chrystle Weigand
- Department of Biochemistry and Molecular Biology, University of Nevada, Reno, Reno, NV, United States
| | - Su-Hwa Kim
- Department of Biochemistry and Molecular Biology, University of Nevada, Reno, Reno, NV, United States
| | - Elizabeth Brown
- Department of Biochemistry and Molecular Biology, University of Nevada, Reno, Reno, NV, United States
| | - Emily Medina
- Department of Biochemistry and Molecular Biology, University of Nevada, Reno, Reno, NV, United States
| | - Moises Mares
- Department of Biochemistry and Molecular Biology, University of Nevada, Reno, Reno, NV, United States
| | - Gad Miller
- The Mina and Everard Goodman Faculty of Life Sciences, Bar Ilan University, Ramat-Gan, Israel
| | - Jeffrey F. Harper
- Department of Biochemistry and Molecular Biology, University of Nevada, Reno, Reno, NV, United States
- *Correspondence: Jeffrey F. Harper,
| | - Won-Gyu Choi
- Department of Biochemistry and Molecular Biology, University of Nevada, Reno, Reno, NV, United States
- Won-Gyu Choi,
| |
Collapse
|
38
|
Gräwe A, Stein V. Linker Engineering in the Context of Synthetic Protein Switches and Sensors. Trends Biotechnol 2020; 39:731-744. [PMID: 33293101 DOI: 10.1016/j.tibtech.2020.11.007] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Revised: 11/11/2020] [Accepted: 11/11/2020] [Indexed: 12/17/2022]
Abstract
Linkers play critical roles in the construction of synthetic protein switches and sensors as they functionally couple a receptor with an actuator. With an increasing number of molecular toolboxes and experimental strategies becoming available that can be applied to engineer protein switches and sensors with tailored response functions, optimising the connecting linkers remains an idiosyncratic and empiric process. This review aims to provide an in-depth analysis of linker motifs, the biophysical properties they confer, and how they impact the performance of synthetic protein switches and sensors while identifying trends, mechanisms, and strategies that underlie the most potent switches and sensors.
Collapse
Affiliation(s)
- Alexander Gräwe
- Department of Biology, TU Darmstadt, 64287 Darmstadt, Germany; Centre for Synthetic Biology, TU Darmstadt, 64283 Darmstadt, Germany
| | - Viktor Stein
- Department of Biology, TU Darmstadt, 64287 Darmstadt, Germany; Centre for Synthetic Biology, TU Darmstadt, 64283 Darmstadt, Germany.
| |
Collapse
|
39
|
Wakida NM, Gomez-Godinez V, Li H, Nguyen J, Kim EK, Dynes JL, Othy S, Lau AL, Ding P, Shi L, Carmona C, Thompson LM, Cahalan MD, Berns MW. Calcium Dynamics in Astrocytes During Cell Injury. Front Bioeng Biotechnol 2020; 8:912. [PMID: 32984268 PMCID: PMC7481337 DOI: 10.3389/fbioe.2020.00912] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Accepted: 07/15/2020] [Indexed: 12/31/2022] Open
Abstract
The changes in intracellular calcium concentration ([Ca2+]) following laser-induced cell injury in nearby cells were studied in primary mouse astrocytes selectively expressing the Ca2+ sensitive GFAP-Cre Salsa6f fluorescent tandem protein, in an Ast1 astrocyte cell line, and in primary mouse astrocytes loaded with Fluo4. Astrocytes in these three systems exhibit distinct changes in [Ca2+] following induced death of nearby cells. Changes in [Ca2+] appear to result from release of Ca2+ from intracellular organelles, as opposed to influx from the external medium. Salsa6f expressing astrocytes displayed dynamic Ca2+ changes throughout the phagocytic response, including lamellae protrusion, cytosolic signaling during vesicle formation, vesicle maturation, and vesicle tract formation. Our results demonstrate local changes in [Ca2+] are involved in the process of phagocytosis in astrocytes responding to cell corpses and/or debris.
Collapse
Affiliation(s)
- Nicole M Wakida
- Beckman Laser Institute & Medical Clinic, University of California, Irvine, Irvine, CA, United States
| | - Veronica Gomez-Godinez
- Institute of Engineering in Medicine, University of California, San Diego, San Diego, CA, United States
| | - Huayan Li
- Institute of Engineering in Medicine, University of California, San Diego, San Diego, CA, United States
| | - Jessica Nguyen
- Beckman Laser Institute & Medical Clinic, University of California, Irvine, Irvine, CA, United States
| | - Edward K Kim
- Beckman Laser Institute & Medical Clinic, University of California, Irvine, Irvine, CA, United States
| | - Joseph L Dynes
- Department of Physiology and Biophysics, School of Medicine, University of California, Irvine, Irvine, CA, United States
| | - Shivashankar Othy
- Department of Physiology and Biophysics, School of Medicine, University of California, Irvine, Irvine, CA, United States
| | - Alice L Lau
- Department of Psychiatry and Human Behavior, University of California, Irvine, Irvine, CA, United States
| | - Peng Ding
- Institute of Engineering in Medicine, University of California, San Diego, San Diego, CA, United States
| | - Linda Shi
- Institute of Engineering in Medicine, University of California, San Diego, San Diego, CA, United States
| | - Christopher Carmona
- Institute of Engineering in Medicine, University of California, San Diego, San Diego, CA, United States
| | - Leslie M Thompson
- Department of Psychiatry and Human Behavior, University of California, Irvine, Irvine, CA, United States.,Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, Irvine, CA, United States.,Department of Neurobiology and Behavior, University of California, Irvine, Irvine, CA, United States.,Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, Irvine, CA, United States
| | - Michael D Cahalan
- Department of Physiology and Biophysics, School of Medicine, University of California, Irvine, Irvine, CA, United States.,Institute for Immunology, University of California, Irvine, Irvine, CA, United States
| | - Michael W Berns
- Beckman Laser Institute & Medical Clinic, University of California, Irvine, Irvine, CA, United States.,Department of Biomedical Engineering, University of California, Irvine, Irvine, CA, United States.,Department of Developmental and Cell Biology, School of Biological Sciences, University of California, Irvine, Irvine, CA, United States
| |
Collapse
|
40
|
Stieger KC, Eles JR, Ludwig KA, Kozai TDY. In vivo microstimulation with cathodic and anodic asymmetric waveforms modulates spatiotemporal calcium dynamics in cortical neuropil and pyramidal neurons of male mice. J Neurosci Res 2020; 98:2072-2095. [PMID: 32592267 DOI: 10.1002/jnr.24676] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Revised: 05/26/2020] [Accepted: 05/27/2020] [Indexed: 12/12/2022]
Abstract
Electrical stimulation has been critical in the development of an understanding of brain function and disease. Despite its widespread use and obvious clinical potential, the mechanisms governing stimulation in the cortex remain largely unexplored in the context of pulse parameters. Modeling studies have suggested that modulation of stimulation pulse waveform may be able to control the probability of neuronal activation to selectively stimulate either cell bodies or passing fibers depending on the leading polarity. Thus, asymmetric waveforms with equal charge per phase (i.e., increasing the leading phase duration and proportionately decreasing the amplitude) may be able to activate a more spatially localized or distributed population of neurons if the leading phase is cathodic or anodic, respectively. Here, we use two-photon and mesoscale calcium imaging of GCaMP6s expressed in excitatory pyramidal neurons of male mice to investigate the role of pulse polarity and waveform asymmetry on the spatiotemporal properties of direct neuronal activation with 10-Hz electrical stimulation. We demonstrate that increasing cathodic asymmetry effectively reduces neuronal activation and results in a more spatially localized subpopulation of activated neurons without sacrificing the density of activated neurons around the electrode. Conversely, increasing anodic asymmetry increases the spatial spread of activation and highly resembles spatiotemporal calcium activity induced by conventional symmetric cathodic stimulation. These results suggest that stimulation polarity and asymmetry can be used to modulate the spatiotemporal dynamics of neuronal activity thus increasing the effective parameter space of electrical stimulation to restore sensation and study circuit dynamics.
Collapse
Affiliation(s)
- Kevin C Stieger
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, USA.,Center for the Neural Basis of Cognition, University of Pittsburgh, Carnegie Mellon University, Pittsburgh, PA, USA
| | - James R Eles
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, USA
| | - Kip A Ludwig
- Department of Biomedical Engineering, University of Wisconsin Madison, Madison, WI, USA.,Department of Neurological Surgery, University of Wisconsin Madison, Madison, WI, USA.,Wisconsin Institute for Translational Neuroengineering (WITNe), Madison, WI, USA
| | - Takashi D Y Kozai
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, USA.,Center for the Neural Basis of Cognition, University of Pittsburgh, Carnegie Mellon University, Pittsburgh, PA, USA.,Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA, USA.,McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, USA.,NeuroTech Center, University of Pittsburgh Brain Institute, Pittsburgh, PA, USA
| |
Collapse
|
41
|
Genetically encoded calcium indicators to probe complex brain circuit dynamics in vivo. Neurosci Res 2020; 169:2-8. [PMID: 32531233 DOI: 10.1016/j.neures.2020.05.013] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2020] [Revised: 05/13/2020] [Accepted: 05/27/2020] [Indexed: 12/28/2022]
Abstract
Over the past two decades, genetically encoded calcium indicators (GECIs) have been used extensively to report intracellular calcium (Ca2+) dynamics in order to readout neuronal and network activity in living tissue. Single wavelength GECIs, such as GCaMP, have been widely adapted due to advances in dynamic range, sensitivity, and kinetics. Additionally, recent efforts in protein engineering have expanded the GECI color palette to enable direct optical interrogation of more complex circuit dynamics. Here, I discuss the engineering, application, and future directions of the most recently developed GECIs for in vivo neuroscience research.
Collapse
|
42
|
Dexamethasone Treatment Increases the Intracellular Calcium Level Through TRPV6 in A549 Cells. Int J Mol Sci 2020; 21:ijms21031050. [PMID: 32033337 PMCID: PMC7037905 DOI: 10.3390/ijms21031050] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Revised: 02/03/2020] [Accepted: 02/04/2020] [Indexed: 12/04/2022] Open
Abstract
This study investigated the effect of dexamethasone (DEX) on intracellular calcium levels and the expressions of transient receptor potential cation channel subcomponent V member 6 (TRPV6), sodium-calcium exchanger 1 (NCX1), and plasma membrane calcium ATPase 1 (PMCA1) in A549 cells. The intracellular calcium level, by using the calcium indicator pGP-CMV-GCaMP6f, increased following DEX treatment for 6, 12, and 24 h in A549 cells. In addition, Rhod-4 assay after DEX treatment for 24 h showed that DEX increased the level of intracellular calcium. The expression of the calcium influx TRPV6 gene significantly increased, whereas the expressions of the calcium outflow NCX1 and PMCA1 genes significantly decreased with DEX treatment. The mRNA levels of surfactant protein genes SFTPA1, SFTPB, SFTPC, and SFTPD and the secreted airway mucin genes MUC1 and MUC5AC were investigated by treating cells with DEX. The DEX treatment decreased the mRNA levels of SFTPA1 and SFTPB but increased the mRNA levels of SFTPC and SFTPD. The MUC1 mRNA level was increased by DEX treatment, whereas MUC5AC mRNA was significantly decreased. These results indicate that DEX influences the intracellular calcium level through TRPV6, and affects pulmonary surfactant genes and secreted airway mucin genes in A549 cells.
Collapse
|
43
|
Eles JR, Kozai TDY. In vivo imaging of calcium and glutamate responses to intracortical microstimulation reveals distinct temporal responses of the neuropil and somatic compartments in layer II/III neurons. Biomaterials 2020; 234:119767. [PMID: 31954232 DOI: 10.1016/j.biomaterials.2020.119767] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Revised: 12/22/2019] [Accepted: 01/05/2020] [Indexed: 02/07/2023]
Abstract
OBJECTIVE Intracortical microelectrode implants can generate a tissue response hallmarked by glial scarring and neuron cell death within 100-150 μm of the biomaterial device. Many have proposed that any performance decline in intracortical microstimulation (ICMS) due to this foreign body tissue response could be offset by increasing the stimulation amplitude. The mechanisms of this approach are unclear, however, as there has not been consensus on how increasing amplitude affects the spatial and temporal recruitment patterns of ICMS. APPROACH We clarify these unknowns using in vivo two-photon imaging of mice transgenically expressing the calcium sensor GCaMP6s in Thy1 neurons or virally expressing the glutamate sensor iGluSnFr in neurons. Calcium and neurotransmitter activity are tracked in the neuronal somas and neuropil during long-train stimulation in Layer II/III of somatosensory cortex. MAIN RESULTS Neural calcium activity and glutamate release are dense and strongest within 20-40 μm around the electrode, falling off with distance from the electrode. Neuronal calcium increases with higher amplitude stimulations. During prolonged stimulation trains, a sub-population of somas fail to maintain calcium activity. Interestingly, neuropil calcium activity is 3-fold less correlated to somatic calcium activity for cells that drop-out during the long stimulation train compared to cells that sustain activity throughout the train. Glutamate release is apparent only within 20 μm of the electrode and is sustained for at least 10s after cessation of the 15 and 20 μA stimulation train, but not lower amplitudes. SIGNIFICANCE These results demonstrate that increasing amplitude can increase the radius and intensity of neural recruitment, but it also alters the temporal response of some neurons. Further, dense glutamate release is highest within the first 20 μm of the electrode site even at high amplitudes, suggesting that there may be spatial limitations to the amplitude parameter space. The glutamate elevation outlasts stimulation, suggesting that high-amplitude stimulation may affect neurotransmitter re-uptake. This ultimately suggests that increasing the amplitude of ICMS device stimulation may fundamentally alter the temporal neural response, which could have implications for using amplitude to improve the ICMS effect or "offset" the effects of glial scarring.
Collapse
Affiliation(s)
- James R Eles
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, USA
| | - Takashi D Y Kozai
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, USA; Center for the Neural Basis of Cognition, University of Pittsburgh, Carnegie Mellon University, Pittsburgh, PA, USA; Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA, USA; McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, USA; NeuroTech Center, University of Pittsburgh Brain Institute, Pittsburgh, PA, USA.
| |
Collapse
|
44
|
Helassa N, Nugues C, Rajamanoharan D, Burgoyne RD, Haynes LP. A centrosome-localized calcium signal is essential for mammalian cell mitosis. FASEB J 2019; 33:14602-14610. [PMID: 31682764 PMCID: PMC6910830 DOI: 10.1096/fj.201901662r] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2019] [Accepted: 09/23/2019] [Indexed: 02/02/2023]
Abstract
Mitosis defects can lead to premature ageing and cancer. Understanding mitosis regulation therefore has important implications for human disease. Early data suggested that calcium (Ca2+) signals could influence mitosis, but these have hitherto not been observed in mammalian cells. Here, we reveal a prolonged yet spatially restricted Ca2+ signal at the centrosomes of actively dividing cells. Local buffering of the centrosomal Ca2+ signals, by flash photolysis of the caged Ca2+ chelator diazo-2-acetoxymethyl ester, arrests mitosis. We also provide evidence that this Ca2+ signal emanates from the endoplasmic reticulum. In summary, we characterize a unique centrosomal Ca2+ signal as a functionally essential input into mitosis.-Helassa, N., Nugues, C., Rajamanoharan, D., Burgoyne, R. D., Haynes, L. P. A centrosome-localized calcium signal is essential for mammalian cell mitosis.
Collapse
Affiliation(s)
- Nordine Helassa
- Department of Cellular and Molecular Physiology, Institute of Translational Medicine, University of Liverpool, Liverpool, United Kingdom
| | - Charlotte Nugues
- Department of Cellular and Molecular Physiology, Institute of Translational Medicine, University of Liverpool, Liverpool, United Kingdom
| | - Dayani Rajamanoharan
- Department of Cellular and Molecular Physiology, Institute of Translational Medicine, University of Liverpool, Liverpool, United Kingdom
| | - Robert D. Burgoyne
- Department of Cellular and Molecular Physiology, Institute of Translational Medicine, University of Liverpool, Liverpool, United Kingdom
| | - Lee P. Haynes
- Department of Cellular and Molecular Physiology, Institute of Translational Medicine, University of Liverpool, Liverpool, United Kingdom
| |
Collapse
|
45
|
Andreoni A, Davis CM, Tian L. Measuring brain chemistry using genetically encoded fluorescent sensors. CURRENT OPINION IN BIOMEDICAL ENGINEERING 2019. [DOI: 10.1016/j.cobme.2019.09.008] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
46
|
Jung K, Kang J, Chung S, Park HJ. Dynamic causal modeling for calcium imaging: Exploration of differential effective connectivity for sensory processing in a barrel cortical column. Neuroimage 2019; 201:116008. [DOI: 10.1016/j.neuroimage.2019.116008] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2018] [Revised: 06/24/2019] [Accepted: 07/09/2019] [Indexed: 01/08/2023] Open
|
47
|
Oh J, Lee C, Kaang BK. Imaging and analysis of genetically encoded calcium indicators linking neural circuits and behaviors. THE KOREAN JOURNAL OF PHYSIOLOGY & PHARMACOLOGY : OFFICIAL JOURNAL OF THE KOREAN PHYSIOLOGICAL SOCIETY AND THE KOREAN SOCIETY OF PHARMACOLOGY 2019; 23:237-249. [PMID: 31297008 PMCID: PMC6609268 DOI: 10.4196/kjpp.2019.23.4.237] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Revised: 05/29/2019] [Accepted: 05/29/2019] [Indexed: 12/21/2022]
Abstract
Confirming the direct link between neural circuit activity and animal behavior has been a principal aim of neuroscience. The genetically encoded calcium indicator (GECI), which binds to calcium ions and emits fluorescence visualizing intracellular calcium concentration, enables detection of in vivo neuronal firing activity. Various GECIs have been developed and can be chosen for diverse purposes. These GECI-based signals can be acquired by several tools including two-photon microscopy and microendoscopy for precise or wide imaging at cellular to synaptic levels. In addition, the images from GECI signals can be analyzed with open source codes including constrained non-negative matrix factorization for endoscopy data (CNMF_E) and miniscope 1-photon-based calcium imaging signal extraction pipeline (MIN1PIPE), and considering parameters of the imaged brain regions (e.g., diameter or shape of soma or the resolution of recorded images), the real-time activity of each cell can be acquired and linked with animal behaviors. As a result, GECI signal analysis can be a powerful tool for revealing the functions of neuronal circuits related to specific behaviors.
Collapse
Affiliation(s)
- Jihae Oh
- School of Biological Sciences, Seoul National University, Seoul 08826, Korea
| | - Chiwoo Lee
- School of Biological Sciences, Seoul National University, Seoul 08826, Korea
| | - Bong-Kiun Kaang
- School of Biological Sciences, Seoul National University, Seoul 08826, Korea
| |
Collapse
|
48
|
Li P, Geng X, Jiang H, Caccavano A, Vicini S, Wu JY. Measuring Sharp Waves and Oscillatory Population Activity With the Genetically Encoded Calcium Indicator GCaMP6f. Front Cell Neurosci 2019; 13:274. [PMID: 31275115 PMCID: PMC6593119 DOI: 10.3389/fncel.2019.00274] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2019] [Accepted: 06/05/2019] [Indexed: 12/16/2022] Open
Abstract
GCaMP6f is among the most widely used genetically encoded calcium indicators for monitoring neuronal activity. Applications are at both the cellular and population levels. Here, we explore two important and under-explored issues. First, we have tested if GCaMP6f is sensitive enough for the detection of population activity with sparse firing, similar to the sensitivity of the local field potential (LFP). Second, we have tested if GCaMP6f is fast enough for the detection of fast network oscillations critical for the encoding and consolidation of memory. We have focused this study on the activity of the hippocampal network including sharp waves (SWs), carbachol-induced theta oscillations, and interictal-like spikes. We compare simultaneous LFP and optical GCaMP6f fluorescent recordings in Thy1-GCaMP6f mouse hippocampal slices. We observe that SWs produce a clear population GCaMP6f signal above noise with an average magnitude of 0.3% ΔF/F. This population signal is highly correlated with the LFP, albeit with a delay of 40.3 ms (SD 10.8 ms). The population GCaMP6f signal follows the LFP evoked by 20 Hz stimulation with high fidelity, while electrically evoked oscillations up to 40 Hz were detectable with reduced amplitude. GCaMP6f and LFP signals showed a large amplitude discrepancy. The amplitude of GCaMP6f fluorescence increased by a factor of 28.9 (SD 13.5) between spontaneous SWs and carbachol-induced theta bursts, while the LFP amplitude increased by a factor of 2.4 (SD 1.0). Our results suggest that GCaMP6f is a useful tool for applications commonly considered beyond the scope of genetically encoded calcium indicators. In particular, population GCaMP6f signals are sensitive enough for detecting synchronous network events with sparse firing and sub-threshold activity, as well as asynchronous events with only a nominal LFP. In addition, population GCaMP6f signals are fast enough for monitoring theta and beta oscillations (<25 Hz). Faster calcium indicators (e.g., GCaMP7) will further improve the frequency response for the detection of gamma band oscillations. The advantage of population optical over LFP recordings are that they are non-contact and free from stimulation artifacts. These features may be particularly useful for high-throughput recordings and applications sensitive to stimulus artifact, such as monitoring responses during continuous stimulation.
Collapse
Affiliation(s)
- Pinggan Li
- Department of Pediatric Neurology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
- Department of Neuroscience, Georgetown University, Washington, DC, United States
| | - Xinling Geng
- Department of Neuroscience, Georgetown University, Washington, DC, United States
- School of Biomedical Engineering, Capital Medical University, Beijing, China
| | - Huiyi Jiang
- Department of Neuroscience, Georgetown University, Washington, DC, United States
- Department of Pediatrics, The First Hospital of Jilin University, Changchun, China
| | - Adam Caccavano
- Department of Pharmacology and Physiology, Georgetown University, Washington, DC, United States
| | - Stefano Vicini
- Department of Pharmacology and Physiology, Georgetown University, Washington, DC, United States
| | - Jian-young Wu
- Department of Neuroscience, Georgetown University, Washington, DC, United States
| |
Collapse
|
49
|
Dürst CD, Wiegert JS, Helassa N, Kerruth S, Coates C, Schulze C, Geeves MA, Török K, Oertner TG. High-speed imaging of glutamate release with genetically encoded sensors. Nat Protoc 2019; 14:1401-1424. [PMID: 30988508 PMCID: PMC6751072 DOI: 10.1038/s41596-019-0143-9] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2018] [Accepted: 01/22/2019] [Indexed: 11/08/2022]
Abstract
The strength of an excitatory synapse depends on its ability to release glutamate and on the density of postsynaptic receptors. Genetically encoded glutamate indicators (GEGIs) allow eavesdropping on synaptic transmission at the level of cleft glutamate to investigate properties of the release machinery in detail. Based on the sensor iGluSnFR, we recently developed accelerated versions of GEGIs that allow investigation of synaptic release during 100-Hz trains. Here, we describe the detailed procedures for design and characterization of fast iGluSnFR variants in vitro, transfection of pyramidal cells in organotypic hippocampal cultures, and imaging of evoked glutamate transients with two-photon laser-scanning microscopy. As the released glutamate spreads from a point source-the fusing vesicle-it is possible to localize the vesicle fusion site with a precision exceeding the optical resolution of the microscope. By using a spiral scan path, the temporal resolution can be increased to 1 kHz to capture the peak amplitude of fast iGluSnFR transients. The typical time frame for these experiments is 30 min per synapse.
Collapse
Affiliation(s)
- Céline D Dürst
- Institute for Synaptic Physiology, Center for Molecular Neurobiology Hamburg, Hamburg, Germany
| | - J Simon Wiegert
- Institute for Synaptic Physiology, Center for Molecular Neurobiology Hamburg, Hamburg, Germany
- Research Group Synaptic Wiring and Information Processing, Center for Molecular Neurobiology Hamburg, Hamburg, Germany
| | - Nordine Helassa
- Molecular and Clinical Sciences Research Institute, St George's, University of London, London, UK
- Department of Cellular and Molecular Physiology, Institute of Translational Medicine, University of Liverpool, Liverpool, UK
| | - Silke Kerruth
- Molecular and Clinical Sciences Research Institute, St George's, University of London, London, UK
- Department of Biophysical Chemistry, J. Heyrovský Institute of Physical Chemistry, Prague, Czech Republic
| | - Catherine Coates
- Molecular and Clinical Sciences Research Institute, St George's, University of London, London, UK
| | - Christian Schulze
- Institute for Synaptic Physiology, Center for Molecular Neurobiology Hamburg, Hamburg, Germany
| | | | - Katalin Török
- Molecular and Clinical Sciences Research Institute, St George's, University of London, London, UK
| | - Thomas G Oertner
- Institute for Synaptic Physiology, Center for Molecular Neurobiology Hamburg, Hamburg, Germany.
| |
Collapse
|
50
|
Kerruth S, Coates C, Dürst CD, Oertner TG, Török K. The kinetic mechanisms of fast-decay red-fluorescent genetically encoded calcium indicators. J Biol Chem 2019; 294:3934-3946. [PMID: 30651353 PMCID: PMC6422079 DOI: 10.1074/jbc.ra118.004543] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2018] [Revised: 01/14/2019] [Indexed: 01/06/2023] Open
Abstract
Genetically encoded calcium indicators (GECIs) are useful reporters of cell-signaling, neuronal, and network activities. We have generated novel fast variants and investigated the kinetic mechanisms of two recently developed red-fluorescent GECIs (RGECIs), mApple-based jRGECO1a and mRuby-based jRCaMP1a. In the formation of fluorescent jRGECO1a and jRCaMP1a complexes, calcium binding is followed by rate-limiting isomerization. However, fluorescence decay of calcium-bound jRGECO1a follows a different pathway from its formation: dissociation of calcium occurs first, followed by the peptide, similarly to GCaMP-s. In contrast, fluorescence decay of calcium-bound jRCaMP1a occurs by the reversal of the on-pathway: peptide dissociation is followed by calcium. The mechanistic differences explain the generally slower off-kinetics of jRCaMP1a-type indicators compared with GCaMP-s and jRGECO1a-type GECI: the fluorescence decay rate of f-RCaMP1 was 21 s-1, compared with 109 s-1 for f-RGECO1 and f-RGECO2 (37 °C). Thus, the CaM-peptide interface is an important determinant of the kinetic responses of GECIs; however, the topology of the structural link to the fluorescent protein demonstrably affects the internal dynamics of the CaM-peptide complex. In the dendrites of hippocampal CA3 neurons, f-RGECO1 indicates calcium elevation in response to a 100 action potential train in a linear fashion, making the probe particularly useful for monitoring large-amplitude, fast signals, e.g. those in dendrites, muscle cells, and immune cells.
Collapse
Affiliation(s)
- Silke Kerruth
- From the Molecular and Clinical Sciences Research Institute, St. George's, University of London, London SW17 0RE, United Kingdom and
| | - Catherine Coates
- From the Molecular and Clinical Sciences Research Institute, St. George's, University of London, London SW17 0RE, United Kingdom and
| | - Céline D Dürst
- the Institute for Synaptic Physiology, Center for Molecular Neurobiology Hamburg, 20251 Hamburg, Germany
| | - Thomas G Oertner
- the Institute for Synaptic Physiology, Center for Molecular Neurobiology Hamburg, 20251 Hamburg, Germany
| | - Katalin Török
- From the Molecular and Clinical Sciences Research Institute, St. George's, University of London, London SW17 0RE, United Kingdom and
| |
Collapse
|