1
|
Pierce AF, Shupe L, Fetz E, Yazdan-Shahmorad A. Flexible modeling of large-scale neural network stimulation: electrical and optical extensions to The Virtual Electrode Recording Tool for EXtracellular Potentials (VERTEX). BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.20.608687. [PMID: 39229104 PMCID: PMC11370401 DOI: 10.1101/2024.08.20.608687] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 09/05/2024]
Abstract
Computational models that predict effects of neural stimulation can be used as a preliminary tool to inform in-vivo research, reducing the costs, time, and ethical considerations involved. However, current models do not support the diverse neural stimulation techniques used in-vivo, including the expanding selection of electrodes, stimulation modalities, and stimulation paradigms. To develop a more comprehensive software, we created several extensions to The Virtual Electrode Recording Tool for EXtracellular Potentials (VERTEX), the MATLAB-based neural stimulation tool from Newcastle University. VERTEX simulates input currents in a large population of multi-compartment neurons within a small cortical slice to model electric field stimulation, while recording local field potentials (LFPs) and spiking activity. Our extensions to its existing electric field stimulation framework include multiple pairs of parametrically defined electrodes and biphasic, bipolar stimulation delivered at programmable delays. To support the growing use of optogenetic approaches for targeted neural stimulation, we introduced a feature that models optogenetic stimulation through an additional VERTEX input function that converts irradiance to currents at optogenetically responsive neurons. Finally, we added extensions to allow complex stimulation protocols including paired-pulse, spatiotemporal patterned, and closed-loop stimulation. We demonstrated our novel features using VERTEX's built-in functionalities, illustrating how these extensions can be used to efficiently and systematically test diverse, targeted, and individualized stimulation patterns.
Collapse
Affiliation(s)
- Anne F Pierce
- Department of Bioengineering, University of Washington, Seattle, WA 98195, USA
- Washington National Primate Research Center, Seattle, WA 98195, USA
| | - Larry Shupe
- Department of Physiology and Biophysics, University of Washington, Seattle WA 98195, USA
| | - Eberhard Fetz
- Washington National Primate Research Center, Seattle, WA 98195, USA
- Department of Physiology and Biophysics, University of Washington, Seattle WA 98195, USA
| | - Azadeh Yazdan-Shahmorad
- Department of Bioengineering, University of Washington, Seattle, WA 98195, USA
- Washington National Primate Research Center, Seattle, WA 98195, USA
- Department of Electrical and Computer Engineering, University of Washington, Seattle, WA 98195, USA
| |
Collapse
|
2
|
McAlinden N, Reiche CF, Clark AM, Scharf R, Cheng Y, Sharma R, Rieth L, Dawson MD, Angelucci A, Mathieson K, Blair S. In vivooptogenetics using a Utah Optrode Array with enhanced light output and spatial selectivity. J Neural Eng 2024; 21:046051. [PMID: 39084245 DOI: 10.1088/1741-2552/ad69c3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Accepted: 07/31/2024] [Indexed: 08/02/2024]
Abstract
Objective.Optogenetics allows the manipulation of neural circuitsin vivowith high spatial and temporal precision. However, combining this precision with control over a significant portion of the brain is technologically challenging (especially in larger animal models).Approach.Here, we have developed, optimised, and testedin vivo, the Utah Optrode Array (UOA), an electrically addressable array of optical needles and interstitial sites illuminated by 181μLEDs and used to optogenetically stimulate the brain. The device is specifically designed for non-human primate studies.Main results.Thinning the combinedμLED and needle backplane of the device from 300μm to 230μm improved the efficiency of light delivery to tissue by 80%, allowing lowerμLED drive currents, which improved power management and thermal performance. The spatial selectivity of each site was also improved by integrating an optical interposer to reduce stray light emission. These improvements were achieved using an innovative fabrication method to create an anodically bonded glass/silicon substrate with through-silicon vias etched, forming an optical interposer. Optical modelling was used to demonstrate that the tip structure of the device had a major influence on the illumination pattern. The thermal performance was evaluated through a combination of modelling and experiment, in order to ensure that cortical tissue temperatures did not rise by more than 1 °C. The device was testedin vivoin the visual cortex of macaque expressing ChR2-tdTomato in cortical neurons.Significance.It was shown that the UOA produced the strongest optogenetic response in the region surrounding the needle tips, and that the extent of the optogenetic response matched the predicted illumination profile based on optical modelling-demonstrating the improved spatial selectivity resulting from the optical interposer approach. Furthermore, different needle illumination sites generated different patterns of low-frequency potential activity.
Collapse
Affiliation(s)
- Niall McAlinden
- SUPA, Institute of Photonics, Department of Physics, University of Strathclyde, Glasgow, United Kingdom
| | - Christopher F Reiche
- Department of Electrical and Computer Engineering, University of Utah, Salt Lake City, UT, United States of America
| | - Andrew M Clark
- Department of Ophthalmology and Visual Science, Moran Eye Institute, University of Utah, Salt Lake City, UT, United States of America
| | - Robert Scharf
- Department of Electrical and Computer Engineering, University of Utah, Salt Lake City, UT, United States of America
| | - Yunzhou Cheng
- SUPA, Institute of Photonics, Department of Physics, University of Strathclyde, Glasgow, United Kingdom
| | - Rohit Sharma
- Department of Electrical and Computer Engineering, University of Utah, Salt Lake City, UT, United States of America
| | - Loren Rieth
- Department of Mechanical, Materials and Aerospace Engineering, West Virginia University, Morgantown, WV, United States of America
| | - Martin D Dawson
- SUPA, Institute of Photonics, Department of Physics, University of Strathclyde, Glasgow, United Kingdom
| | - Alessandra Angelucci
- Department of Ophthalmology and Visual Science, Moran Eye Institute, University of Utah, Salt Lake City, UT, United States of America
| | - Keith Mathieson
- SUPA, Institute of Photonics, Department of Physics, University of Strathclyde, Glasgow, United Kingdom
| | - Steve Blair
- Department of Electrical and Computer Engineering, University of Utah, Salt Lake City, UT, United States of America
| |
Collapse
|
3
|
Mondragón-González SL, Schreiweis C, Burguière E. Closed-loop recruitment of striatal interneurons prevents compulsive-like grooming behaviors. Nat Neurosci 2024; 27:1148-1156. [PMID: 38693349 PMCID: PMC11156588 DOI: 10.1038/s41593-024-01633-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Accepted: 03/27/2024] [Indexed: 05/03/2024]
Abstract
Compulsive behaviors have been associated with striatal hyperactivity. Parvalbumin-positive striatal interneurons (PVIs) in the striatum play a crucial role in regulating striatal activity and suppressing prepotent inappropriate actions. To investigate the potential role of striatal PVIs in regulating compulsive behaviors, we assessed excessive self-grooming-a behavioral metric of compulsive-like behavior-in male Sapap3 knockout mice (Sapap3-KO). Continuous optogenetic activation of PVIs in striatal areas receiving input from the lateral orbitofrontal cortex reduced self-grooming events in Sapap3-KO mice to wild-type levels. Aiming to shorten the critical time window for PVI recruitment, we then provided real-time closed-loop optogenetic stimulation of striatal PVIs, using a transient power increase in the 1-4 Hz frequency band in the orbitofrontal cortex as a predictive biomarker of grooming onsets. Targeted closed-loop stimulation at grooming onsets was as effective as continuous stimulation in reducing grooming events but required 87% less stimulation time, paving the way for adaptive stimulation therapeutic protocols.
Collapse
Affiliation(s)
- Sirenia Lizbeth Mondragón-González
- Institut du Cerveau - Paris Brain Institute - ICM, Sorbonne Université, Inserm, CNRS, AP-HP Hôpital de la Pitié Salpêtrière, Paris, France
| | - Christiane Schreiweis
- Institut du Cerveau - Paris Brain Institute - ICM, Sorbonne Université, Inserm, CNRS, AP-HP Hôpital de la Pitié Salpêtrière, Paris, France
| | - Eric Burguière
- Institut du Cerveau - Paris Brain Institute - ICM, Sorbonne Université, Inserm, CNRS, AP-HP Hôpital de la Pitié Salpêtrière, Paris, France.
| |
Collapse
|
4
|
McAlinden N, Reiche CF, Clark AM, Scharf R, Cheng Y, Sharma R, Rieth L, Dawson MD, Angelucci A, Mathieson K, Blair S. In vivo optogenetics using a Utah Optrode Array with enhanced light output and spatial selectivity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.18.585479. [PMID: 38562871 PMCID: PMC10983961 DOI: 10.1101/2024.03.18.585479] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
Optogenetics allows manipulation of neural circuits in vivo with high spatial and temporal precision. However, combining this precision with control over a significant portion of the brain is technologically challenging (especially in larger animal models). Here, we have developed, optimised, and tested in vivo, the Utah Optrode Array (UOA), an electrically addressable array of optical needles and interstitial sites illuminated by 181 µLEDs and used to optogenetically stimulate the brain. The device is specifically designed for non-human primate studies. Thinning the combined µLED and needle backplane of the device from 300 µm to 230 µm improved the efficiency of light delivery to tissue by 80%, allowing lower µLED drive currents, which improved power management and thermal performance. The spatial selectivity of each site was also improved by integrating an optical interposer to reduce stray light emission. These improvements were achieved using an innovative fabrication method to create an anodically bonded glass/silicon substrate with through-silicon vias etched, forming an optical interposer. Optical modelling was used to demonstrate that the tip structure of the device had a major influence on the illumination pattern. The thermal performance was evaluated through a combination of modelling and experiment, in order to ensure that cortical tissue temperatures did not rise by more than 1°C. The device was tested in vivo in the visual cortex of macaque expressing ChR2-tdTomato in cortical neurons. It was shown that the strongest optogenetic response occurred in the region surrounding the needle tips, and that the extent of the optogenetic response matched the predicted illumination profile based on optical modelling - demonstrating the improved spatial selectivity resulting from the optical interposer approach. Furthermore, different needle illumination sites generated different patterns of low-frequency potential (LFP) activity.
Collapse
|
5
|
Lee Y, Kim S, Cho YK, Kong C, Chang JW, Jun SB. Amygdala electrical stimulation for operant conditioning in rat navigation. Biomed Eng Lett 2024; 14:291-306. [PMID: 38374898 PMCID: PMC10874353 DOI: 10.1007/s13534-023-00336-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Revised: 11/12/2023] [Accepted: 11/17/2023] [Indexed: 02/21/2024] Open
Abstract
There have been several attempts to navigate the locomotion of animals by neuromodulation. The most common method is animal training with electrical brain stimulation for directional cues and rewards; the basic principle is to activate dopamine-mediated neural reward pathways such as the medial forebrain bundle (MFB) when the animal correctly follows the external commands. In this study, the amygdala, which is the brain region responsible for fear modulation, was targeted for punishment training. The brain regions of MFB, amygdala, and barrel cortex were electrically stimulated for reward, punishment, and directional cues, respectively. Electrical stimulation was applied to the amygdala of rats when they failed to follow directional commands. First, two different amygdala regions, i.e., basolateral amygdala (BLA) and central amygdala (CeA), were stimulated and compared in terms of behavior responses, success and correction rates for training, and gene expression for learning and memory. Then, the training was performed in three groups: group R (MFB stimulation for reward), group P (BLA stimulation for punishment), and group RP (both MFB and BLA stimulation for reward and punishment). In group P, after the training, RNA sequencing was conducted to detect gene expression and demonstrate the effect of punishment learning. Group P showed higher success rates than group R, and group RP exhibited the most effective locomotion control among the three groups. Gene expression results imply that BLA stimulation can be more effective as a punishment in the learning process than CeA stimulation. We developed a new method to navigate rat locomotion behaviors by applying amygdala stimulation.
Collapse
Affiliation(s)
- Youjin Lee
- Department of Electronic and Electrical Engineering, Ewha Womans University, Seoul, 03760 Republic of Korea
- Graduate Program in Smart Factory, Ewha Womans University, Seoul, 03760 Republic of Korea
| | - Soonyoung Kim
- Department of Electrical and Computer Engineering, Rice University, Houston, TX 77005 USA
| | - Yoon Kyung Cho
- Department of Electronic and Electrical Engineering, Ewha Womans University, Seoul, 03760 Republic of Korea
| | - Chanho Kong
- Department of Neurosurgery, Yonsei University College of Medicine, Seoul, 03722 Republic of Korea
| | - Jin Woo Chang
- Department of Neurosurgery, Yonsei University College of Medicine, Seoul, 03722 Republic of Korea
- Brain Korea 21 PLUS Project for Medical Science and Brain Research Institute, Yonsei University College of Medicine, Seoul, 03722 Republic of Korea
| | - Sang Beom Jun
- Department of Electronic and Electrical Engineering, Ewha Womans University, Seoul, 03760 Republic of Korea
- Graduate Program in Smart Factory, Ewha Womans University, Seoul, 03760 Republic of Korea
- Department of Brain and Cognitive Sciences, Ewha Womans University, Seoul, 03760 Republic of Korea
| |
Collapse
|
6
|
Cunha AB, Schuelke C, Mesri A, Ruud SK, Aizenshtadt A, Ferrari G, Heiskanen A, Asif A, Keller SS, Ramos-Moreno T, Kalvøy H, Martínez-Serrano A, Krauss S, Emnéus J, Sampietro M, Martinsen ØG. Development of a Smart Wireless Multisensor Platform for an Optogenetic Brain Implant. SENSORS (BASEL, SWITZERLAND) 2024; 24:575. [PMID: 38257668 PMCID: PMC11154348 DOI: 10.3390/s24020575] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Revised: 01/09/2024] [Accepted: 01/12/2024] [Indexed: 01/24/2024]
Abstract
Implantable cell replacement therapies promise to completely restore the function of neural structures, possibly changing how we currently perceive the onset of neurodegenerative diseases. One of the major clinical hurdles for the routine implementation of stem cell therapies is poor cell retention and survival, demanding the need to better understand these mechanisms while providing precise and scalable approaches to monitor these cell-based therapies in both pre-clinical and clinical scenarios. This poses significant multidisciplinary challenges regarding planning, defining the methodology and requirements, prototyping and different stages of testing. Aiming toward an optogenetic neural stem cell implant controlled by a smart wireless electronic frontend, we show how an iterative development methodology coupled with a modular design philosophy can mitigate some of these challenges. In this study, we present a miniaturized, wireless-controlled, modular multisensor platform with fully interfaced electronics featuring three different modules: an impedance analyzer, a potentiostat and an optical stimulator. We show the application of the platform for electrical impedance spectroscopy-based cell monitoring, optical stimulation to induce dopamine release from optogenetically modified neurons and a potentiostat for cyclic voltammetry and amperometric detection of dopamine release. The multisensor platform is designed to be used as an opto-electric headstage for future in vivo animal experiments.
Collapse
Affiliation(s)
- André B. Cunha
- Department of Physics, University of Oslo, Sem Sælands vei 24, 0371 Oslo, Norway; (A.B.C.); (C.S.); (S.K.R.)
| | - Christin Schuelke
- Department of Physics, University of Oslo, Sem Sælands vei 24, 0371 Oslo, Norway; (A.B.C.); (C.S.); (S.K.R.)
- Hybrid Technology Hub—Centre of Excellence, Institute of Basic Medical Sciences, P.O. Box 1110 Blindern, 0317 Oslo, Norway; (A.A.); (S.K.)
| | - Alireza Mesri
- Department of Electronics Information and Bioengineering, Politecnico di Milano, Piazza L. da Vinci 32, 20133 Milan, Italy; (A.M.); (G.F.); (M.S.)
| | - Simen K. Ruud
- Department of Physics, University of Oslo, Sem Sælands vei 24, 0371 Oslo, Norway; (A.B.C.); (C.S.); (S.K.R.)
| | - Aleksandra Aizenshtadt
- Hybrid Technology Hub—Centre of Excellence, Institute of Basic Medical Sciences, P.O. Box 1110 Blindern, 0317 Oslo, Norway; (A.A.); (S.K.)
| | - Giorgio Ferrari
- Department of Electronics Information and Bioengineering, Politecnico di Milano, Piazza L. da Vinci 32, 20133 Milan, Italy; (A.M.); (G.F.); (M.S.)
| | - Arto Heiskanen
- Department of Biotechnology and Biomedicine, Technical University of Denmark, 2800 Kongens Lyngby, Denmark; (A.H.); (A.A.); (J.E.)
| | - Afia Asif
- Department of Biotechnology and Biomedicine, Technical University of Denmark, 2800 Kongens Lyngby, Denmark; (A.H.); (A.A.); (J.E.)
| | - Stephan S. Keller
- National Centre for Nano Fabrication and Characterization, Technical University of Denmark, 2800 Kongens Lyngby, Denmark;
| | - Tania Ramos-Moreno
- Lund Stem Cell Center, Division of Neurosurgery, Department of Clinical Sciences, Faculty of Medicine, Lund University, 22184 Lund, Sweden;
| | - Håvard Kalvøy
- Department of Clinical and Biomedical Engineering, Oslo University Hospital, Sognsvannsveien 20, 0372 Oslo, Norway;
| | - Alberto Martínez-Serrano
- Department of Molecular Neurobiology, Center of Molecular Biology ‘Severo Ochoa’, Universidad Autónoma de Madrid, Calle Nicolás Cabrera 1, 28049 Madrid, Spain;
| | - Stefan Krauss
- Hybrid Technology Hub—Centre of Excellence, Institute of Basic Medical Sciences, P.O. Box 1110 Blindern, 0317 Oslo, Norway; (A.A.); (S.K.)
- Department of Immunology and Transfusion Medicine, Oslo University Hospital, P.O. Box 4950, 0424 Oslo, Norway
| | - Jenny Emnéus
- Department of Biotechnology and Biomedicine, Technical University of Denmark, 2800 Kongens Lyngby, Denmark; (A.H.); (A.A.); (J.E.)
| | - Marco Sampietro
- Department of Electronics Information and Bioengineering, Politecnico di Milano, Piazza L. da Vinci 32, 20133 Milan, Italy; (A.M.); (G.F.); (M.S.)
| | - Ørjan G. Martinsen
- Department of Physics, University of Oslo, Sem Sælands vei 24, 0371 Oslo, Norway; (A.B.C.); (C.S.); (S.K.R.)
- Department of Clinical and Biomedical Engineering, Oslo University Hospital, Sognsvannsveien 20, 0372 Oslo, Norway;
| |
Collapse
|
7
|
Kook YH, Lee H, Lee J, Jeong Y, Rho J, Heo WD, Lee S. AAV-compatible optogenetic tools for activating endogenous calcium channels in vivo. Mol Brain 2023; 16:73. [PMID: 37848907 PMCID: PMC10583393 DOI: 10.1186/s13041-023-01061-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Accepted: 09/28/2023] [Indexed: 10/19/2023] Open
Abstract
Calcium ions (Ca2+) play pivotal roles in regulating diverse brain functions, including cognition, emotion, locomotion, and learning and memory. These functions are intricately regulated by a variety of Ca2+-dependent cellular processes, encompassing synaptic plasticity, neuro/gliotransmitter release, and gene expression. In our previous work, we developed 'monster OptoSTIM1' (monSTIM1), an improved OptoSTIM1 that selectively activates Ca2+-release-activated Ca2+ (CRAC) channels in the plasma membrane through blue light, allowing precise control over intracellular Ca2+ signaling and specific brain functions. However, the large size of the coding sequence of monSTIM1 poses a limitation for its widespread use, as it exceeds the packaging capacity of adeno-associated virus (AAV). To address this constraint, we have introduced monSTIM1 variants with reduced coding sequence sizes and established AAV-based systems for expressing them in neurons and glial cells in the mouse brain. Upon expression by AAVs, these monSTIM1 variants significantly increased the expression levels of cFos in neurons and astrocytes in the hippocampal CA1 region following non-invasive light illumination. The use of monSTIM1 variants offers a promising avenue for investigating the spatiotemporal roles of Ca2+-mediated cellular activities in various brain functions. Furthermore, this toolkit holds potential as a therapeutic strategy for addressing brain disorders associated with aberrant Ca2+ signaling.
Collapse
Affiliation(s)
- Yeon Hee Kook
- Center for Cognition and Sociality, Institute for Basic Science (IBS), Daejeon, 34126, Republic of Korea
- Department of Bioscience and Biotechnology, Graduate School, Chungnam National University, Daejeon, 34134, Korea
| | - Hyoin Lee
- Center for Cognition and Sociality, Institute for Basic Science (IBS), Daejeon, 34126, Republic of Korea
| | - Jinsu Lee
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Republic of Korea
| | - Yeonji Jeong
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Republic of Korea
| | - Jaerang Rho
- Department of Bioscience and Biotechnology, Graduate School, Chungnam National University, Daejeon, 34134, Korea
| | - Won Do Heo
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Republic of Korea.
- KAIST Institute for the BioCentury, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Republic of Korea.
| | - Sangkyu Lee
- Center for Cognition and Sociality, Institute for Basic Science (IBS), Daejeon, 34126, Republic of Korea.
| |
Collapse
|
8
|
Lightning A, Bourzeix M, Beurrier C, Kuczewski N. Effects of discontinuous blue light stimulation on the electrophysiological properties of neurons lacking opsin expression in vitro: Implications for optogenetic experiments. Eur J Neurosci 2023; 57:885-899. [PMID: 36726326 DOI: 10.1111/ejn.15927] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Revised: 01/05/2023] [Accepted: 01/26/2023] [Indexed: 02/03/2023]
Abstract
Neuronal sensitivity to light stimulation can be a significant confounding factor for assays that use light to study neuronal processes, such as optogenetics and fluorescent imaging. While continuous one-photon (1P) blue light stimulation has been shown to be responsible for a decrease in firing activity in several neuronal subtypes, discontinuous 1P blue light stimulation commonly used in optogenetic experiments is supposed to have a negligible action. In the present report, we tested experimentally this theoretical prediction by assessing the effects produced by the most commonly used patterns of discontinuous 1P light stimulation on several electrophysiological parameters in brain slices. We found that, compared with continuous stimulation, the artefactual effect of light is reduced when discontinuous stimulation is used, especially when the duty cycle and light power are low.
Collapse
Affiliation(s)
- Anistasha Lightning
- Université Claude Bernard Lyon 1, CNRS, INSERM, Centre de Recherche en Neurosciences de Lyon CRNL U1028UMR5292, NEUROPOP, Bron, France
| | - Marie Bourzeix
- Aix Marseille Univ, CNRS UMR 7289, Institut de Neurosciences de la Timone (INT), Marseille, France
| | - Corinne Beurrier
- Aix Marseille Univ, CNRS UMR 7289, Institut de Neurosciences de la Timone (INT), Marseille, France
| | - Nicola Kuczewski
- Université Claude Bernard Lyon 1, CNRS, INSERM, Centre de Recherche en Neurosciences de Lyon CRNL U1028UMR5292, NEUROPOP, Bron, France
| |
Collapse
|
9
|
Wang AY, Sheng Y, Li W, Jung D, Junek GV, Liu H, Park J, Lee D, Wang M, Maharjan S, Kumashi S, Hao J, Zhang YS, Eggan K, Wang H. A Multimodal and Multifunctional CMOS Cellular Interfacing Array for Digital Physiology and Pathology Featuring an Ultra Dense Pixel Array and Reconfigurable Sampling Rate. IEEE TRANSACTIONS ON BIOMEDICAL CIRCUITS AND SYSTEMS 2022; 16:1057-1074. [PMID: 36417722 DOI: 10.1109/tbcas.2022.3224064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
The article presents a fully integrated multimodal and multifunctional CMOS biosensing/actuating array chip and system for multi-dimensional cellular/tissue characterization. The CMOS chip supports up to 1,568 simultaneous parallel readout channels across 21,952 individually addressable multimodal pixels with 13 μm × 13 μm 2-D pixel pitch along with 1,568 Pt reference electrodes. These features allow the CMOS array chip to perform multimodal physiological measurements on living cell/tissue samples with both high throughput and single-cell resolution. Each pixel supports three sensing and one actuating modalities, each reconfigurable for different functionalities, in the form of full array (FA) or fast scan (FS) voltage recording schemes, bright/dim optical detection, 2-/4-point impedance sensing (ZS), and biphasic current stimulation (BCS) with adjustable stimulation area for single-cell or tissue-level stimulation. Each multi-modal pixel contains an 8.84 μm × 11 μm Pt electrode, 4.16 μm × 7.2 μm photodiode (PD), and in-pixel circuits for PD measurements and pixel selection. The chip is fabricated in a standard 130nm BiCMOS process as a proof of concept. The on-chip electrodes are constructed by unique design and in-house post-CMOS fabrication processes, including a critical Al shorting of all pixels during fabrication and Al etching after fabrication that ensures a high-yield planar electrode array on CMOS with high biocompatibility and long-term measurement reliability. For demonstration, extensive biological testing is performed with human and mouse progenitor cells, in which multidimensional biophysiological data are acquired for comprehensive cellular characterization.
Collapse
|
10
|
Skoven CS, Tomasevic L, Kvitsiani D, Pakkenberg B, Dyrby TB, Siebner HR. Dose-response relationship between the variables of unilateral optogenetic stimulation and transcallosal evoked responses in rat motor cortex. Front Neurosci 2022; 16:968839. [PMID: 36213739 PMCID: PMC9539969 DOI: 10.3389/fnins.2022.968839] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Accepted: 08/19/2022] [Indexed: 11/17/2022] Open
Abstract
Efficient interhemispheric integration of neural activity between left and right primary motor cortex (M1) is critical for inter-limb motor control. We employed optogenetic stimulation to establish a framework for probing transcallosal M1–M1 interactions in rats. We performed optogenetic stimulation of excitatory neurons in right M1 of male Sprague-Dawley rats. We recorded the transcallosal evoked potential in contralateral left M1 via chronically implanted electrodes. Recordings were performed under anesthesia combination of dexmedetomidine and a low concentration of isoflurane. We systematically varied the stimulation intensity and duration to characterize the relationship between stimulation parameters in right M1 and the characteristics of the evoked intracortical potentials in left M1. Optogenetic stimulation of right M1 consistently evoked a transcallosal response in left M1 with a consistent negative peak (N1) that sometimes was preceded by a smaller positive peak (P1). Higher stimulation intensity or longer stimulation duration gradually increased N1 amplitude and reduced N1 variability across trials. A combination of stimulation intensities of 5–10 mW with stimulus durations of 1–10 ms were generally sufficient to elicit a robust transcallosal response in most animal, with our optic fiber setup. Optogenetically stimulated excitatory neurons in M1 can reliably evoke a transcallosal response in anesthetized rats. Characterizing the relationship between “stimulation dose” and “response magnitude” (i.e., the gain function) of transcallosal M1-to-M1 excitatory connections can be used to optimize the variables of optogenetic stimulation and ensure stimulation efficacy.
Collapse
Affiliation(s)
- Christian Stald Skoven
- Danish Research Centre for Magnetic Resonance, Centre for Functional and Diagnostic Imaging and Research, Copenhagen University Hospital Amager and Hvidovre, Copenhagen, Denmark
- Center for Functional Integrative Neuroscience, Aarhus University (AU), Aarhus, Denmark
- *Correspondence: Christian Stald Skoven,
| | - Leo Tomasevic
- Danish Research Centre for Magnetic Resonance, Centre for Functional and Diagnostic Imaging and Research, Copenhagen University Hospital Amager and Hvidovre, Copenhagen, Denmark
| | - Duda Kvitsiani
- Department of Molecular Biology and Genetics, Danish Research Institute of Translational Neuroscience, Aarhus University, Aarhus, Denmark
| | - Bente Pakkenberg
- Research Laboratory for Stereology and Neuroscience, Copenhagen University Hospital Bispebjerg and Frederiksberg, Copenhagen, Denmark
- Department of Clinical Medicine, Faculty of Medical and Health Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Tim Bjørn Dyrby
- Danish Research Centre for Magnetic Resonance, Centre for Functional and Diagnostic Imaging and Research, Copenhagen University Hospital Amager and Hvidovre, Copenhagen, Denmark
- Department of Applied Mathematics and Computer Science, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Hartwig Roman Siebner
- Danish Research Centre for Magnetic Resonance, Centre for Functional and Diagnostic Imaging and Research, Copenhagen University Hospital Amager and Hvidovre, Copenhagen, Denmark
- Department of Clinical Medicine, Faculty of Medical and Health Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Neurology, Copenhagen University Hospital Bispebjerg and Frederiksberg, Copenhagen, Denmark
- Hartwig Roman Siebner,
| |
Collapse
|
11
|
Bellusci L, Kim E, Garcia DuBar S, Gillis RA, Vicini S, Sahibzada N. Brainstem activation of GABAB receptors in the nucleus tractus solitarius increases gastric motility. Front Neurosci 2022; 16:961042. [PMID: 35983226 PMCID: PMC9379309 DOI: 10.3389/fnins.2022.961042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Accepted: 07/11/2022] [Indexed: 12/01/2022] Open
Abstract
Background and aim Local GABAergic signaling in the dorsal vagal complex (DVC) is essential to control gastric function. While the inhibitory GABAA receptor action on motility in the DVC is well-documented, the role of the GABAB receptor on gastric function is less well-established. Microinjection of baclofen, a selective GABAB receptor agonist, in the dorsal motor nucleus of the vagus (DMV) increases gastric tone and motility, while the effect on motility in the nucleus tractus solitarius (NTS) needs to be investigated. Previous in vitro studies showed that GABAB receptors exert a local inhibitory effect in unidentified NTS neurons. Since the NTS and DMV nuclei have differential control of gastric motility, we compared GABAB receptor activation in the NTS to that reported in the DMV. We microinjected baclofen unilaterally in the NTS while monitoring intragastric pressure and compared its action to optogenetic activation of somatostatin (SST) neurons in transgenic sst-Cre::channelrhodopsin-2 (ChR2) mice. We also performed patch-clamp recordings from SST and DMV neurons in brainstem slices from these mice. Methods In vivo drug injections and optogenetic stimulation were performed in fasted urethane/α-chloralose anesthetized male mice. Gastric tone and motility were monitored by an intragastric balloon inserted in the antrum and inflated with warm water to provide a baseline intragastric pressure (IGP). Coronal brainstem slices were obtained from the sst-Cre::ChR2 mice for interrogation with optogenetics and pharmacology using electrophysiology. Results The unilateral microinjection of baclofen into the NTS caused a robust increase in gastric tone and motility that was not affected by ipsilateral vagotomy. Optogenetic activation of SST neurons that followed baclofen effectively suppresses the gastric motility in vivo. In brain slices, baclofen suppressed spontaneous and light-activated inhibitory postsynaptic currents in SST and gastrointestinal-projection DMV neurons and produced outward currents. Conclusion Our results show that GABAB receptors in the NTS strongly increase gastric tone and motility. Optogenetic stimulation in vivo and in vitro suggests that these receptors activated by baclofen suppress the glutamatergic sensory vagal afferents in the NTS and also inhibit the interneurons and the inhibitory neurons that project to the DMV, which, in turn, increase motility via a cholinergic excitatory pathway to the stomach.
Collapse
|
12
|
Preliminary Study on Safety Assessment of 10 Hz Transcranial Alternating Current Stimulation in Rat Brain. APPLIED SCIENCES-BASEL 2022. [DOI: 10.3390/app12115299] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Assessment of the safety of transcranial electrical stimulation devices that contact the scalp and apply electrical stimulations to brain tissues is essential for the prevention of unexpected brain damage caused by electromagnetic fields. In particular, safety studies on transcranial alternating current stimulation (tACS) are needed for active applications to treat brain diseases and for the development of medical devices, because there is a lack of research on the safety of tACS, in contrast to transcranial direct current stimulation. In this study, the safety of tACS with selected parameters, i.e., a stimulation intensity of 1.0 to 2.0 mA, a frequency of 10 Hz, and a treatment time of 20 min, was examined at a preclinical stage using small animals (rats). The results of magnetic resonance imaging and histopathological imaging indicated that the conditions applied in this study provided safe tACS without damaging brain tissues or neuronal components in the acute phase. In addition, the temperature did not increase above 41 °C, which is a temperature limitation for contact-type medical devices, even after 20 min of tACS application.
Collapse
|
13
|
A Miniaturized Closed-Loop Optogenetic Brain Stimulation Device. ELECTRONICS 2022. [DOI: 10.3390/electronics11101591] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
This paper presents a tetherless and miniaturized closed-loop optogenetic brain stimulation device, designed as a back mountable device for laboratory mice. The device has the ability to sense the biomarkers corresponding to major depressive disorder (MDD) from local field potential (LFP), and produces a feedback signal to control the closed-loop operation after on-device processing of the sensed signals. MDD is a chronic neurological disorder and there are still many unanswered questions about the underlying neurological mechanisms behind its occurrence. Along with other brain stimulation paradigms, optogenetics has recently proved effective in the study of MDD. Most of these experiments have used tethered and connected devices. However, the use of tethered devices in optogenetic brain stimulation experiments has the drawback of hindering the free movement of the laboratory animal subjects undergoing stimulation. To address this issue, the proposed device is small, light-weight, untethered, and back-mountable. The device consists of: (i) an optrode which houses an electrode for collecting neural signals, an optical source for delivering light stimulations, and a temperature sensor for monitoring the temperature increase at the stimulation site, (ii) a neural sensor for acquisition and pre-processing of the neural signals to obtain LFP signals in the frequency range of 4 to 200 Hz, as electrophysiological biomarkers of MDD (iii) a classifier for classification of the signal into four classes: normal, abnormal alpha, abnormal theta, and abnormal gamma oscillations, (iv) a control algorithm to select stimulation parameters based on the input class, and (v) a stimulator for generating light stimulations. The design, implementation, and evaluation of the device are presented, and the results are discussed. The neural sensor and the stimulator are circular in shape with a radius of 8 mm. Pre-recorded neural signals from the mouse hippocampus are used for the evaluation of the device.
Collapse
|
14
|
Pallikaras V, Carter F, Velazquez-Martinez DN, Arvanitogiannis A, Shizgal P. The trade-off between pulse duration and power in optical excitation of midbrain dopamine neurons approximates Bloch's law. Behav Brain Res 2022; 419:113702. [PMID: 34864162 DOI: 10.1016/j.bbr.2021.113702] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2021] [Revised: 11/22/2021] [Accepted: 11/29/2021] [Indexed: 11/28/2022]
Abstract
Optogenetic experiments reveal functional roles of specific neurons. However, functional inferences have been limited by widespread adoption of a restricted set of stimulation parameters. Broader exploration of the parameter space can deepen insight into the mapping between selective neural activity and behavior. In this way, characteristics of the activated neural circuit, such as temporal integration, can be inferred. Our objective was to determine whether an equal-energy principle accounts for the interaction of pulse duration and optical power in optogenetic excitation. Six male TH::Cre rats worked for optogenetic (ChannelRhodopsin-2) stimulation of VTA dopamine neurons. We used a within-subject design to describe the trade-off between pulse duration and optical power in determining reward seeking. Parameters were customized for each subject based on behavioral effectiveness. Within a useful range of powers (~12.6-31.6 mW) the product of optical power and pulse duration required to produce a given level of reward seeking was roughly constant. Such reciprocity is consistent with Bloch's law, which posits an equal-energy principle of temporal summation over short durations in human vision. The trade-off between pulse duration and power broke down at higher powers. Thus, optical power and duration can be adjusted reciprocally for brief durations and lower powers, and power can be substituted for pulse duration to scale the region of excitation in behavioral optogenetic experiments. The findings demonstrate the utility of within-subject and trade-off designs in optogenetics and of parameter adjustment based on functional endpoints instead of physical properties of the stimulation.
Collapse
Affiliation(s)
- Vasilios Pallikaras
- Center for Studies in Behavioral Neurobiology, Department of Psychology, Concordia University, 7141 Sherbrooke St W., Montreal, QC H4B 1R6, Canada
| | - Francis Carter
- Center for Studies in Behavioral Neurobiology, Department of Psychology, Concordia University, 7141 Sherbrooke St W., Montreal, QC H4B 1R6, Canada
| | - David Natanael Velazquez-Martinez
- Center for Studies in Behavioral Neurobiology, Department of Psychology, Concordia University, 7141 Sherbrooke St W., Montreal, QC H4B 1R6, Canada
| | - Andreas Arvanitogiannis
- Center for Studies in Behavioral Neurobiology, Department of Psychology, Concordia University, 7141 Sherbrooke St W., Montreal, QC H4B 1R6, Canada
| | - Peter Shizgal
- Center for Studies in Behavioral Neurobiology, Department of Psychology, Concordia University, 7141 Sherbrooke St W., Montreal, QC H4B 1R6, Canada.
| |
Collapse
|
15
|
Acharya AR, Vandekerckhove B, Larsen LE, Delbeke J, Wadman WJ, Vonck K, Carette E, Meurs A, Vanfleteren J, Boon P, Missinne J, Raedt R. In vivoblue light illumination for optogenetic inhibition: effect on local temperature and excitability of the rat hippocampus. J Neural Eng 2021; 18. [PMID: 34951406 DOI: 10.1088/1741-2552/ac3ef4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Accepted: 12/01/2021] [Indexed: 11/12/2022]
Abstract
Objective.The blue light-activated inhibitory opsin, stGtACR2, is gaining prominence as a neuromodulatory tool due its ability to shunt-inhibit neurons and is being frequently used inin vivoexperimentation. However, experiments involving stGtACR2 use longer durations of blue light pulses, which inadvertently heat up the local brain tissue and confound experimental results. Therefore, the heating effects of illumination parameters used forin vivooptogenetic inhibition must be evaluated.Approach.To assess blue light (473 nm)-induced heating of the brain, we used a computational model as well as direct temperature measurements using a fiber Bragg grating (FBG). The effects of different light power densities (LPDs) and pulse durations on evoked potentials (EP) recorded from dentate gyrus were assessed. For opsin-negative rats, LPDs between 127 and 636 mW mm-2and pulse durations between 20 and 5120 ms were tested while for stGtACR2 expressing rats, LPD of 127 mW mm-2and pulse durations between 20 and 640 ms were tested.Main results.Increasing LPDs and pulse durations logarithmically increased the peak temperature and significantly decreased the population spike (PS) amplitude and latencies of EPs. For a pulse duration of 5120 ms, the tissue temperature increased by 0.6 °C-3.4 °C. All tested LPDs decreased the PS amplitude in opsin-negative rats, but 127 mW mm-2had comparatively minimal effects and a significant effect of increasing light pulse duration was seen from 320 ms and beyond. This corresponded with an average temperature increase of 0.2 °C-1.1 °C at the recorded site. Compared to opsin-negative rats, illumination in stGtACR2-expressing rats resulted in much greater inhibition of EPs.Significance.Our study demonstrates that light-induced heating of the brain can be accurately measuredin vivousing FBG sensors. Such light-induced heating alone can affect neuronal excitability. Useful neuromodulation by the activation of stGtACR2 is still possible while minimizing thermal effects.
Collapse
Affiliation(s)
- Anirudh R Acharya
- 4Brain Team, Department of Head and Skin, Ghent University, 9000 Ghent, Belgium
| | - Bram Vandekerckhove
- Center for Microsystems Technology, Imec and Ghent University, 9000 Ghent, Belgium
| | - Lars Emil Larsen
- 4Brain Team, Department of Head and Skin, Ghent University, 9000 Ghent, Belgium
| | - Jean Delbeke
- 4Brain Team, Department of Head and Skin, Ghent University, 9000 Ghent, Belgium
| | - Wytse J Wadman
- 4Brain Team, Department of Head and Skin, Ghent University, 9000 Ghent, Belgium
| | - Kristl Vonck
- 4Brain Team, Department of Head and Skin, Ghent University, 9000 Ghent, Belgium
| | - Evelien Carette
- 4Brain Team, Department of Head and Skin, Ghent University, 9000 Ghent, Belgium
| | - Alfred Meurs
- 4Brain Team, Department of Head and Skin, Ghent University, 9000 Ghent, Belgium
| | - Jan Vanfleteren
- Center for Microsystems Technology, Imec and Ghent University, 9000 Ghent, Belgium
| | - Paul Boon
- 4Brain Team, Department of Head and Skin, Ghent University, 9000 Ghent, Belgium
| | - Jeroen Missinne
- Center for Microsystems Technology, Imec and Ghent University, 9000 Ghent, Belgium
| | - Robrecht Raedt
- 4Brain Team, Department of Head and Skin, Ghent University, 9000 Ghent, Belgium
| |
Collapse
|
16
|
Wireless, battery-free, subdermally implantable platforms for transcranial and long-range optogenetics in freely moving animals. Proc Natl Acad Sci U S A 2021; 118:2025775118. [PMID: 34301889 DOI: 10.1073/pnas.2025775118] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Wireless, battery-free, and fully subdermally implantable optogenetic tools are poised to transform neurobiological research in freely moving animals. Current-generation wireless devices are sufficiently small, thin, and light for subdermal implantation, offering some advantages over tethered methods for naturalistic behavior. Yet current devices using wireless power delivery require invasive stimulus delivery, penetrating the skull and disrupting the blood-brain barrier. This can cause tissue displacement, neuronal damage, and scarring. Power delivery constraints also sharply curtail operational arena size. Here, we implement highly miniaturized, capacitive power storage on the platform of wireless subdermal implants. With approaches to digitally manage power delivery to optoelectronic components, we enable two classes of applications: transcranial optogenetic activation millimeters into the brain (validated using motor cortex stimulation to induce turning behaviors) and wireless optogenetics in arenas of more than 1 m2 in size. This methodology allows for previously impossible behavioral experiments leveraging the modern optogenetic toolkit.
Collapse
|
17
|
Klink PC, Aubry JF, Ferrera VP, Fox AS, Froudist-Walsh S, Jarraya B, Konofagou EE, Krauzlis RJ, Messinger A, Mitchell AS, Ortiz-Rios M, Oya H, Roberts AC, Roe AW, Rushworth MFS, Sallet J, Schmid MC, Schroeder CE, Tasserie J, Tsao DY, Uhrig L, Vanduffel W, Wilke M, Kagan I, Petkov CI. Combining brain perturbation and neuroimaging in non-human primates. Neuroimage 2021; 235:118017. [PMID: 33794355 PMCID: PMC11178240 DOI: 10.1016/j.neuroimage.2021.118017] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Revised: 03/07/2021] [Accepted: 03/22/2021] [Indexed: 12/11/2022] Open
Abstract
Brain perturbation studies allow detailed causal inferences of behavioral and neural processes. Because the combination of brain perturbation methods and neural measurement techniques is inherently challenging, research in humans has predominantly focused on non-invasive, indirect brain perturbations, or neurological lesion studies. Non-human primates have been indispensable as a neurobiological system that is highly similar to humans while simultaneously being more experimentally tractable, allowing visualization of the functional and structural impact of systematic brain perturbation. This review considers the state of the art in non-human primate brain perturbation with a focus on approaches that can be combined with neuroimaging. We consider both non-reversible (lesions) and reversible or temporary perturbations such as electrical, pharmacological, optical, optogenetic, chemogenetic, pathway-selective, and ultrasound based interference methods. Method-specific considerations from the research and development community are offered to facilitate research in this field and support further innovations. We conclude by identifying novel avenues for further research and innovation and by highlighting the clinical translational potential of the methods.
Collapse
Affiliation(s)
- P Christiaan Klink
- Department of Vision & Cognition, Netherlands Institute for Neuroscience, Meibergdreef 47, 1105 BA Amsterdam, the Netherlands.
| | - Jean-François Aubry
- Physics for Medicine Paris, Inserm U1273, CNRS UMR 8063, ESPCI Paris, PSL University, Paris, France
| | - Vincent P Ferrera
- Department of Neuroscience & Department of Psychiatry, Columbia University Medical Center, New York, NY, USA; Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY, USA
| | - Andrew S Fox
- Department of Psychology & California National Primate Research Center, University of California, Davis, CA, USA
| | | | - Béchir Jarraya
- NeuroSpin, Commissariat à l'Énergie Atomique et aux Énergies Alternatives (CEA), Institut National de la Santé et de la Recherche Médicale (INSERM), Cognitive Neuroimaging Unit, Université Paris-Saclay, France; Foch Hospital, UVSQ, Suresnes, France
| | - Elisa E Konofagou
- Ultrasound and Elasticity Imaging Laboratory, Department of Biomedical Engineering, Columbia University, New York, NY, USA; Department of Radiology, Columbia University, New York, NY, USA
| | - Richard J Krauzlis
- Laboratory of Sensorimotor Research, National Eye Institute, Bethesda, MD, USA
| | - Adam Messinger
- Laboratory of Brain and Cognition, National Institute of Mental Health, Bethesda, MD, USA
| | - Anna S Mitchell
- Department of Experimental Psychology, Oxford University, Oxford, United Kingdom
| | - Michael Ortiz-Rios
- Newcastle University Medical School, Newcastle upon Tyne NE1 7RU, United Kingdom; German Primate Center, Leibniz Institute for Primate Research, Kellnerweg 4, 37077 Göttingen, Germany
| | - Hiroyuki Oya
- Iowa Neuroscience Institute, Carver College of Medicine, University of Iowa, Iowa City, IA, USA; Department of Neurosurgery, University of Iowa, Iowa city, IA, USA
| | - Angela C Roberts
- Department of Physiology, Development and Neuroscience, Cambridge University, Cambridge, United Kingdom
| | - Anna Wang Roe
- Interdisciplinary Institute of Neuroscience and Technology, School of Medicine, Zhejiang University, Hangzhou 310029, China
| | | | - Jérôme Sallet
- Department of Experimental Psychology, Oxford University, Oxford, United Kingdom; Univ Lyon, Université Lyon 1, Inserm, Stem Cell and Brain Research Institute, U1208 Bron, France; Wellcome Centre for Integrative Neuroimaging, Department of Experimental Psychology, University of Oxford, Oxford, United Kingdom
| | - Michael Christoph Schmid
- Newcastle University Medical School, Newcastle upon Tyne NE1 7RU, United Kingdom; Faculty of Science and Medicine, University of Fribourg, Chemin du Musée 5, CH-1700 Fribourg, Switzerland
| | - Charles E Schroeder
- Nathan Kline Institute, Orangeburg, NY, USA; Columbia University, New York, NY, USA
| | - Jordy Tasserie
- NeuroSpin, Commissariat à l'Énergie Atomique et aux Énergies Alternatives (CEA), Institut National de la Santé et de la Recherche Médicale (INSERM), Cognitive Neuroimaging Unit, Université Paris-Saclay, France
| | - Doris Y Tsao
- Division of Biology and Biological Engineering, Tianqiao and Chrissy Chen Institute for Neuroscience; Howard Hughes Medical Institute; Computation and Neural Systems, Caltech, Pasadena, CA, USA
| | - Lynn Uhrig
- NeuroSpin, Commissariat à l'Énergie Atomique et aux Énergies Alternatives (CEA), Institut National de la Santé et de la Recherche Médicale (INSERM), Cognitive Neuroimaging Unit, Université Paris-Saclay, France
| | - Wim Vanduffel
- Laboratory for Neuro- and Psychophysiology, Neurosciences Department, KU Leuven Medical School, Leuven, Belgium; Leuven Brain Institute, KU Leuven, Leuven Belgium; Harvard Medical School, Boston, MA, USA; Massachusetts General Hospital, Martinos Center for Biomedical Imaging, Charlestown, MA, USA
| | - Melanie Wilke
- German Primate Center, Leibniz Institute for Primate Research, Kellnerweg 4, 37077 Göttingen, Germany; Department of Cognitive Neurology, University Medicine Göttingen, Göttingen, Germany
| | - Igor Kagan
- German Primate Center, Leibniz Institute for Primate Research, Kellnerweg 4, 37077 Göttingen, Germany.
| | - Christopher I Petkov
- Newcastle University Medical School, Newcastle upon Tyne NE1 7RU, United Kingdom.
| |
Collapse
|
18
|
Jeon S, Lee Y, Ryu D, Cho YK, Lee Y, Jun SB, Ji CH. Implantable Optrode Array for Optogenetic Modulation and Electrical Neural Recording. MICROMACHINES 2021; 12:mi12060725. [PMID: 34205473 PMCID: PMC8234104 DOI: 10.3390/mi12060725] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Revised: 06/14/2021] [Accepted: 06/16/2021] [Indexed: 02/06/2023]
Abstract
During the last decade, optogenetics has become an essential tool for neuroscience research due to its unrivaled feature of cell-type-specific neuromodulation. There have been several technological advances in light delivery devices. Among them, the combination of optogenetics and electrophysiology provides an opportunity for facilitating optogenetic approaches. In this study, a novel design of an optrode array was proposed for realizing optical modulation and electrophysiological recording. A 4 × 4 optrode array and five-channel recording electrodes were assembled as a disposable part, while a reusable part comprised an LED (light-emitting diode) source and a power line. After the characterization of the intensity of the light delivered at the fiber tips, in vivo animal experiment was performed with transgenic mice expressing channelrhodopsin, showing the effectiveness of optical activation and neural recording.
Collapse
Affiliation(s)
- Saeyeong Jeon
- Department of Electrical and Computer Engineering, Seoul National University, Seoul 08826, Korea; (S.J.); (D.R.)
| | - Youjin Lee
- Department of Electronic and Electrical Engineering, Ewha Womans University, Seoul 03760, Korea; (Y.L.); (Y.K.C.); (Y.L.); (S.B.J.)
- Graduate Program in Smart Factory, Ewha Womans University, Seoul 03760, Korea
| | - Daeho Ryu
- Department of Electrical and Computer Engineering, Seoul National University, Seoul 08826, Korea; (S.J.); (D.R.)
| | - Yoon Kyung Cho
- Department of Electronic and Electrical Engineering, Ewha Womans University, Seoul 03760, Korea; (Y.L.); (Y.K.C.); (Y.L.); (S.B.J.)
| | - Yena Lee
- Department of Electronic and Electrical Engineering, Ewha Womans University, Seoul 03760, Korea; (Y.L.); (Y.K.C.); (Y.L.); (S.B.J.)
| | - Sang Beom Jun
- Department of Electronic and Electrical Engineering, Ewha Womans University, Seoul 03760, Korea; (Y.L.); (Y.K.C.); (Y.L.); (S.B.J.)
- Graduate Program in Smart Factory, Ewha Womans University, Seoul 03760, Korea
- Department of Brain and Cognitive Sciences, Ewha Womans University, Seoul 03760, Korea
| | - Chang-Hyeon Ji
- Department of Electronic and Electrical Engineering, Ewha Womans University, Seoul 03760, Korea; (Y.L.); (Y.K.C.); (Y.L.); (S.B.J.)
- Graduate Program in Smart Factory, Ewha Womans University, Seoul 03760, Korea
- Correspondence: ; Tel.: +82-2-3277-3895
| |
Collapse
|
19
|
Essig J, Hunt JB, Felsen G. Inhibitory neurons in the superior colliculus mediate selection of spatially-directed movements. Commun Biol 2021; 4:719. [PMID: 34117346 PMCID: PMC8196039 DOI: 10.1038/s42003-021-02248-1] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [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: 05/18/2021] [Indexed: 02/05/2023] Open
Abstract
Decision making is a cognitive process that mediates behaviors critical for survival. Choosing spatial targets is an experimentally-tractable form of decision making that depends on the midbrain superior colliculus (SC). While physiological and computational studies have uncovered the functional topographic organization of the SC, the role of specific SC cell types in spatial choice is unknown. Here, we leveraged behavior, optogenetics, neural recordings and modeling to directly examine the contribution of GABAergic SC neurons to the selection of opposing spatial targets. Although GABAergic SC neurons comprise a heterogeneous population with local and long-range projections, our results demonstrate that GABAergic SC neurons do not locally suppress premotor output, suggesting that functional long-range inhibition instead plays a dominant role in spatial choice. An attractor model requiring only intrinsic SC circuitry was sufficient to account for our experimental observations. Overall, our study elucidates the role of GABAergic SC neurons in spatial choice.
Collapse
Affiliation(s)
- Jaclyn Essig
- Department of Physiology and Biophysics, and Neuroscience Program University of Colorado School of Medicine, Aurora, CO, USA
| | - Joshua B Hunt
- Department of Physiology and Biophysics, and Neuroscience Program University of Colorado School of Medicine, Aurora, CO, USA
| | - Gidon Felsen
- Department of Physiology and Biophysics, and Neuroscience Program University of Colorado School of Medicine, Aurora, CO, USA.
| |
Collapse
|
20
|
Karadas M, Olsson C, Winther Hansen N, Perrier JF, Webb JL, Huck A, Andersen UL, Thielscher A. In-vitro Recordings of Neural Magnetic Activity From the Auditory Brainstem Using Color Centers in Diamond: A Simulation Study. Front Neurosci 2021; 15:643614. [PMID: 34054404 PMCID: PMC8155532 DOI: 10.3389/fnins.2021.643614] [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: 12/18/2020] [Accepted: 04/12/2021] [Indexed: 11/13/2022] Open
Abstract
Magnetometry based on nitrogen-vacancy (NV) centers in diamond is a novel technique capable of measuring magnetic fields with high sensitivity and high spatial resolution. With the further advancements of these sensors, they may open up novel approaches for the 2D imaging of neural signals in vitro. In the present study, we investigate the feasibility of NV-based imaging by numerically simulating the magnetic signal from the auditory pathway of a rodent brainstem slice (ventral cochlear nucleus, VCN, to the medial trapezoid body, MNTB) as stimulated by both electric and optic stimulation. The resulting signal from these two stimulation methods are evaluated and compared. A realistic pathway model was created based on published data of the neural morphologies and channel dynamics of the globular bushy cells in the VCN and their axonal projections to the principal cells in the MNTB. The pathway dynamics in response to optic and electric stimulation and the emitted magnetic fields were estimated using the cable equation. For simulating the optic stimulation, the light distribution in brain tissue was numerically estimated and used to model the optogenetic neural excitation based on a four state channelrhodopsin-2 (ChR2) model. The corresponding heating was also estimated, using the bio-heat equation and was found to be low (<2°C) even at excessively strong optic signals. A peak magnetic field strength of ∼0.5 and ∼0.1 nT was calculated from the auditory brainstem pathway after electrical and optical stimulation, respectively. By increasing the stimulating light intensity four-fold (far exceeding commonly used intensities) the peak magnetic signal strength only increased to 0.2 nT. Thus, while optogenetic stimulation would be favorable to avoid artefacts in the recordings, electric stimulation achieves higher peak fields. The present simulation study predicts that high-resolution magnetic imaging of the action potentials traveling along the auditory brainstem pathway will only be possible for next generation NV sensors. However, the existing sensors already have sufficient sensitivity to support the magnetic sensing of cumulated neural signals sampled from larger parts of the pathway, which might be a promising intermediate step toward further maturing this novel technology.
Collapse
Affiliation(s)
- Mürsel Karadas
- Department of Health Technology, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Christoffer Olsson
- Department of Health Technology, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Nikolaj Winther Hansen
- Department of Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Jean-François Perrier
- Department of Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - James Luke Webb
- Department of Physics, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Alexander Huck
- Department of Physics, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Ulrik Lund Andersen
- Department of Physics, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Axel Thielscher
- Department of Health Technology, Technical University of Denmark, Kongens Lyngby, Denmark
- Danish Research Centre for Magnetic Resonance, Centre for Functional and Diagnostic Imaging and Research, Copenhagen University Hospital Hvidovre, Hvidovre, Denmark
| |
Collapse
|
21
|
Bartley AF, Fischer M, Bagley ME, Barnes JA, Burdette MK, Cannon KE, Bolding MS, Foulger SH, McMahon LL, Weick JP, Dobrunz LE. Feasibility of cerium-doped LSO particles as a scintillator for x-ray induced optogenetics. J Neural Eng 2021; 18:10.1088/1741-2552/abef89. [PMID: 33730704 PMCID: PMC8656171 DOI: 10.1088/1741-2552/abef89] [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: 10/12/2020] [Accepted: 03/17/2021] [Indexed: 11/11/2022]
Abstract
Objective.Non-invasive light delivery into the brain is needed forin vivooptogenetics to avoid physical damage. An innovative strategy could employ x-ray activation of radioluminescent particles (RLPs) to emit localized light. However, modulation of neuronal or synaptic function by x-ray induced radioluminescence from RLPs has not yet been demonstrated.Approach.Molecular and electrophysiological approaches were used to determine if x-ray dependent radioluminescence emitted from RLPs can activate light sensitive proteins. RLPs composed of cerium doped lutetium oxyorthosilicate (LSO:Ce), an inorganic scintillator that emits blue light, were used as they are biocompatible with neuronal function and synaptic transmission.Main results.We show that 30 min of x-ray exposure at a rate of 0.042 Gy s-1caused no change in the strength of basal glutamatergic transmission during extracellular field recordings in mouse hippocampal slices. Additionally, long-term potentiation, a robust measure of synaptic integrity, was induced after x-ray exposure and expressed at a magnitude not different from control conditions (absence of x-rays). We found that x-ray stimulation of RLPs elevated cAMP levels in HEK293T cells expressing OptoXR, a chimeric opsin receptor that combines the extracellular light-sensitive domain of rhodopsin with an intracellular second messenger signaling cascade. This demonstrates that x-ray radioluminescence from LSO:Ce particles can activate OptoXR. Next, we tested whether x-ray activation of the RLPs can enhance synaptic activity in whole-cell recordings from hippocampal neurons expressing channelrhodopsin-2, both in cell culture and acute hippocampal slices. Importantly, x-ray radioluminescence caused an increase in the frequency of spontaneous excitatory postsynaptic currents in both systems, indicating activation of channelrhodopsin-2 and excitation of neurons.Significance.Together, our results show that x-ray activation of LSO:Ce particles can heighten cellular and synaptic function. The combination of LSO:Ce inorganic scintillators and x-rays is therefore a viable method for optogenetics as an alternative to more invasive light delivery methods.
Collapse
Affiliation(s)
- Aundrea F Bartley
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, AL, United States of America
- Civitan International Research Center, University of Alabama at Birmingham, Birmingham, AL, United States of America
- Evelyn F. McKnight Brain Institute, University of Alabama at Birmingham, Birmingham, AL, United States of America
- Comprehensive Neuroscience Center, University of Alabama at Birmingham, Birmingham, AL, United States of America
| | - Máté Fischer
- Department of Neurosciences, University of New Mexico-Health Sciences Center, Albuquerque, NM, United States of America
| | - Micah E Bagley
- Department of Cell, Developmental, and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL, United States of America
| | - Justin A Barnes
- Department of Cell, Developmental, and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL, United States of America
| | - Mary K Burdette
- Department of Materials Science and Engineering, Clemson University, Anderson, SC, United States of America
| | - Kelli E Cannon
- Department of Vision Science, University of Alabama at Birmingham, Birmingham, AL, United States of America
| | - Mark S Bolding
- Department of Radiology, University of Alabama at Birmingham, Birmingham, AL, United States of America
| | - Stephen H Foulger
- Department of Materials Science and Engineering, Clemson University, Anderson, SC, United States of America
- Center for Optical Materials Science and Engineering Technologies, Clemson University, Anderson, SC, United States of America
- Department of Bioengineering, Clemson University, Clemson, SC, United States of America
| | - Lori L McMahon
- Department of Cell, Developmental, and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL, United States of America
- Civitan International Research Center, University of Alabama at Birmingham, Birmingham, AL, United States of America
- Evelyn F. McKnight Brain Institute, University of Alabama at Birmingham, Birmingham, AL, United States of America
- Comprehensive Neuroscience Center, University of Alabama at Birmingham, Birmingham, AL, United States of America
| | - Jason P Weick
- Department of Neurosciences, University of New Mexico-Health Sciences Center, Albuquerque, NM, United States of America
| | - Lynn E Dobrunz
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, AL, United States of America
- Civitan International Research Center, University of Alabama at Birmingham, Birmingham, AL, United States of America
- Evelyn F. McKnight Brain Institute, University of Alabama at Birmingham, Birmingham, AL, United States of America
- Comprehensive Neuroscience Center, University of Alabama at Birmingham, Birmingham, AL, United States of America
| |
Collapse
|
22
|
Chronic Optogenetic Stimulation in Freely Moving Rodents. Methods Mol Biol 2021. [PMID: 32865756 DOI: 10.1007/978-1-0716-0830-2_22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register]
Abstract
In vivo optogenetic strategies have been fundamental for the investigation of how neural circuits relate to behavior. While short-term experimental procedures are typically used in such studies, chronic stimulation during behavioral sessions has been largely unexplored. Here we describe a protocol for long-term optogenetic modulation of neuronal populations in freely moving animals.
Collapse
|
23
|
Thompson AC, Wise AK, Hart WL, Needham K, Fallon JB, Gunewardene N, Stoddart PR, Richardson RT. Hybrid optogenetic and electrical stimulation for greater spatial resolution and temporal fidelity of cochlear activation. J Neural Eng 2020; 17:056046. [PMID: 33036009 DOI: 10.1088/1741-2552/abbff0] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
OBJECTIVE Compared to electrical stimulation, optogenetic stimulation has the potential to improve the spatial precision of neural activation in neuroprostheses, but it requires intense light and has relatively poor temporal kinetics. We tested the effect of hybrid stimulation, which is the combination of subthreshold optical and electrical stimuli, on spectral and temporal fidelity in the cochlea by recording multiunit activity in the inferior colliculus of channelrhodopsin (H134R variant) transgenic mice. APPROACH Pulsed light or biphasic electrical pulses were delivered to cochlear spiral ganglion neurons of acutely deafened mice, either as individual stimuli or as hybrid stimuli for which the timing of the electrical pulse had a varied delay relative to the start of the optical pulse. Response thresholds, spread of activation and entrainment data were obtained from multi-unit recordings from the auditory midbrain. MAIN RESULTS Facilitation occurred when subthreshold electrical stimuli were applied at the end of, or up to 3.75 ms after subthreshold optical pulses. The spread of activation resulting from hybrid stimulation was significantly narrower than electrical-only and optical-only stimulation (p < 0.01), measured at equivalent suprathreshold levels of loudness that are relevant to cochlear implant users. Furthermore, temporal fidelity, measured as maximum following rates to 300 ms pulse trains bursts up to 240 Hz, was 2.4-fold greater than optical-only stimulation (p < 0.05). SIGNIFICANCE By significantly improving spectral resolution of electrical- and optical-only stimulation and the temporal fidelity of optical-only stimulation, hybrid stimulation has the potential to increase the number of perceptually independent stimulating channels in a cochlear implant.
Collapse
|
24
|
Bansal H, Gupta N, Roy S. Theoretical Analysis of Low-power Bidirectional Optogenetic Control of High-frequency Neural Codes with Single Spike Resolution. Neuroscience 2020; 449:165-188. [DOI: 10.1016/j.neuroscience.2020.09.022] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2020] [Revised: 09/04/2020] [Accepted: 09/07/2020] [Indexed: 02/06/2023]
|
25
|
Moreaux LC, Yatsenko D, Sacher WD, Choi J, Lee C, Kubat NJ, Cotton RJ, Boyden ES, Lin MZ, Tian L, Tolias AS, Poon JKS, Shepard KL, Roukes ML. Integrated Neurophotonics: Toward Dense Volumetric Interrogation of Brain Circuit Activity-at Depth and in Real Time. Neuron 2020; 108:66-92. [PMID: 33058767 PMCID: PMC8061790 DOI: 10.1016/j.neuron.2020.09.043] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Revised: 08/18/2020] [Accepted: 09/28/2020] [Indexed: 12/17/2022]
Abstract
We propose a new paradigm for dense functional imaging of brain activity to surmount the limitations of present methodologies. We term this approach "integrated neurophotonics"; it combines recent advances in microchip-based integrated photonic and electronic circuitry with those from optogenetics. This approach has the potential to enable lens-less functional imaging from within the brain itself to achieve dense, large-scale stimulation and recording of brain activity with cellular resolution at arbitrary depths. We perform a computational study of several prototype 3D architectures for implantable probe-array modules that are designed to provide fast and dense single-cell resolution (e.g., within a 1-mm3 volume of mouse cortex comprising ∼100,000 neurons). We describe progress toward realizing integrated neurophotonic imaging modules, which can be produced en masse with current semiconductor foundry protocols for chip manufacturing. Implantation of multiple modules can cover extended brain regions.
Collapse
Affiliation(s)
- Laurent C Moreaux
- Division of Physics, Mathematics and Astronomy, California Institute of Technology, Pasadena, CA 91125, USA.
| | - Dimitri Yatsenko
- Vathes LLC, Houston, TX 77030, USA; Center for Neuroscience and Artificial Intelligence and Department of Neuroscience, Baylor College of Medicine, Houston, TX 77030, USA
| | - Wesley D Sacher
- Kavli Nanoscience Institute, California Institute of Technology, Pasadena, CA 91125, USA; Division of Engineering and Applied Science, California Institute of Technology, Pasadena, CA 91125, USA; Max Planck Institute for Microstructure Physics, Halle, Germany
| | - Jaebin Choi
- Departments of Electrical Engineering and Biomedical Engineering, Columbia University, New York, NY 10027, USA
| | - Changhyuk Lee
- Departments of Electrical Engineering and Biomedical Engineering, Columbia University, New York, NY 10027, USA; Center for BioMicrosystems, Brain Science Institute, Korea Institute of Science and Technology, Korea
| | - Nicole J Kubat
- Division of Physics, Mathematics and Astronomy, California Institute of Technology, Pasadena, CA 91125, USA
| | - R James Cotton
- Shirley Ryan AbilityLab, Northwestern University, Chicago, IL 60611, USA; Center for Neuroscience and Artificial Intelligence and Department of Neuroscience, Baylor College of Medicine, Houston, TX 77030, USA
| | - Edward S Boyden
- Howard Hughes Medical Institute, Cambridge, MA, USA; McGovern Institute, MIT, Cambridge, USA; Koch Institute, MIT, Cambridge, USA; Departments of Brain and Cognitive Sciences, Media Arts and Sciences, and Biological Engineering, MIT, Cambridge, USA
| | - Michael Z Lin
- Departments of Neurobiology and Bioengineering, Stanford University, Stanford, CA 94305, USA
| | - Lin Tian
- Department of Biochemistry and Molecular Medicine, School of Medicine, University of California, Davis, CA 95616, USA
| | - Andreas S Tolias
- Vathes LLC, Houston, TX 77030, USA; Center for Neuroscience and Artificial Intelligence and Department of Neuroscience, Baylor College of Medicine, Houston, TX 77030, USA; Department of Electrical and Computer Engineering, Rice University, Houston, TX 77005, USA
| | - Joyce K S Poon
- Max Planck Institute for Microstructure Physics, Halle, Germany; Department of Electrical and Computer Engineering, University of Toronto, 10 King's College Rd., Toronto, ON M5S 3G4, Canada
| | - Kenneth L Shepard
- Departments of Electrical Engineering and Biomedical Engineering, Columbia University, New York, NY 10027, USA
| | - Michael L Roukes
- Division of Physics, Mathematics and Astronomy, California Institute of Technology, Pasadena, CA 91125, USA; Kavli Nanoscience Institute, California Institute of Technology, Pasadena, CA 91125, USA; Division of Engineering and Applied Science, California Institute of Technology, Pasadena, CA 91125, USA; Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA.
| |
Collapse
|
26
|
Bansal H, Gupta N, Roy S. Comparison of low-power, high-frequency and temporally precise optogenetic inhibition of spiking in NpHR, eNpHR3.0 and Jaws-expressing neurons. Biomed Phys Eng Express 2020; 6:045011. [PMID: 33444272 DOI: 10.1088/2057-1976/ab90a1] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
A detailed theoretical analysis of low-power, high-frequency and temporally precise optogenetic inhibition of neuronal spiking, with red-shifted opsins namely, NpHR, eNpHR3.0 and Jaws, has been presented. An accurate model for inhibition of spiking in these opsins expressed hippocampal neurons that includes the important rebound activity of chloride ions across the membrane has been formulated. The effect of various parameters including irradiance, pulse width, frequency, opsin-expression density and chloride concentration has been studied in detail. Theoretical simulations are in very good agreement with reported experimental results. The chloride concentration gradient directly affects the photocurrent and inhibition capacity in all three variants. eNpHR3.0 shows smallest inhibitory post-synaptic potential plateau at higher frequencies. The time delay between light stimulus and target spike is crucial to minimize irradiance and expression density thresholds for suppressing individual spike. Good practical values of photostimulation parameters have been obtained empirically for peak photocurrent, time delay and 100% spiking inhibition, at continuous and pulsed illumination. Under continuous illumination, complete inhibition of neural activity in Jaws-expressing neurons takes place at minimum irradiance of 0.2 mW mm-2 and expression density of 0.2 mS cm-2, whereas for pulsed stimulation, it is at minimum irradiance of 0.6 mW mm-2 and 5 ms pulse width, at 10 Hz. It is shown that Jaws and eNpHR3.0 are able to invoke single spike precise inhibition up to 160 and 200 Hz, respectively. The study is useful in designing new experiments, understanding temporal spike coding and bidirectional control, and curing neurological disorders.
Collapse
Affiliation(s)
- Himanshu Bansal
- Department of Physics and Computer Science, Dayalbagh Educational Institute, Agra-282005, India
| | | | | |
Collapse
|
27
|
Dieter A, Keppeler D, Moser T. Towards the optical cochlear implant: optogenetic approaches for hearing restoration. EMBO Mol Med 2020; 12:e11618. [PMID: 32227585 PMCID: PMC7136966 DOI: 10.15252/emmm.201911618] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Revised: 01/08/2020] [Accepted: 01/28/2020] [Indexed: 12/30/2022] Open
Abstract
Cochlear implants (CIs) are considered the most successful neuroprosthesis as they enable speech comprehension in the majority of half a million CI users suffering from sensorineural hearing loss. By electrically stimulating the auditory nerve, CIs constitute an interface re-connecting the brain and the auditory scene, providing the patient with information regarding the latter. However, since electric current is hard to focus in conductive environments such as the cochlea, the precision of electrical sound encoding-and thus quality of artificial hearing-is limited. Recently, optogenetic stimulation of the cochlea has been suggested as an alternative approach for hearing restoration. Cochlear optogenetics promises increased spectral selectivity of artificial sound encoding, hence improved hearing, as light can conveniently be confined in space to activate the auditory nerve within smaller tonotopic ranges. In this review, we discuss the latest experimental and technological developments of cochlear optogenetics and outline the remaining challenges on the way to clinical translation.
Collapse
Affiliation(s)
- Alexander Dieter
- Institute for Auditory Neuroscience and InnerEarLabUniversity Medical Center GöttingenGöttingenGermany
- Göttingen Graduate School for NeurosciencesBiophysics and Molecular BiosciencesUniversity of GöttingenGöttingenGermany
| | - Daniel Keppeler
- Institute for Auditory Neuroscience and InnerEarLabUniversity Medical Center GöttingenGöttingenGermany
| | - Tobias Moser
- Institute for Auditory Neuroscience and InnerEarLabUniversity Medical Center GöttingenGöttingenGermany
- Auditory Neuroscience and Optogenetics LaboratoryGerman Primate CenterGöttingenGermany
- Auditory Neuroscience GroupMax Planck Institute of Experimental MedicineGöttingenGermany
- Cluster of Excellence “Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells” (MBExC)University of GöttingenGöttingenGermany
| |
Collapse
|
28
|
Kim S, Kyung T, Chung JH, Kim N, Keum S, Lee J, Park H, Kim HM, Lee S, Shin HS, Do Heo W. Non-invasive optical control of endogenous Ca 2+ channels in awake mice. Nat Commun 2020; 11:210. [PMID: 31924789 PMCID: PMC6954201 DOI: 10.1038/s41467-019-14005-4] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2019] [Accepted: 12/05/2019] [Indexed: 02/08/2023] Open
Abstract
Optogenetic approaches for controlling Ca2+ channels provide powerful means for modulating diverse Ca2+-specific biological events in space and time. However, blue light-responsive photoreceptors are, in principle, considered inadequate for deep tissue stimulation unless accompanied by optic fiber insertion. Here, we present an ultra-light-sensitive optogenetic Ca2+ modulator, named monSTIM1 encompassing engineered cryptochrome2 for manipulating Ca2+ signaling in the brain of awake mice through non-invasive light delivery. Activation of monSTIM1 in either excitatory neurons or astrocytes of mice brain is able to induce Ca2+-dependent gene expression without any mechanical damage in the brain. Furthermore, we demonstrate that non-invasive Ca2+ modulation in neurons can be sufficiently and effectively translated into changes in behavioral phenotypes of awake mice. Optogenetic applications in the brain of live animals often require the use of optic fibers due to poor tissue-penetration of blue light. Here the authors present monSTIM1, an improved high sensitivity optogenetic tool able to modulate Ca2+ signaling in the brain of awake mice using non-invasive light stimulation.
Collapse
Affiliation(s)
- Sungsoo Kim
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
| | - Taeyoon Kyung
- Center for Cognition and Sociality, Institute for Basic Science (IBS), Daejeon, Republic of Korea.,David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Jae-Hee Chung
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
| | - Nury Kim
- Center for Cognition and Sociality, Institute for Basic Science (IBS), Daejeon, Republic of Korea
| | - Sehoon Keum
- Center for Cognition and Sociality, Institute for Basic Science (IBS), Daejeon, Republic of Korea
| | - Jinsu Lee
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
| | - Hyerim Park
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
| | - Ho Min Kim
- Center for Biomolecular and Cellular Structure, Institute for Basic Science (IBS), Daejeon, Republic of Korea.,Graduate School of Medical Science & Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
| | - Sangkyu Lee
- Center for Cognition and Sociality, Institute for Basic Science (IBS), Daejeon, Republic of Korea.
| | - Hee-Sup Shin
- Center for Cognition and Sociality, Institute for Basic Science (IBS), Daejeon, Republic of Korea.
| | - Won Do Heo
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea. .,Center for Cognition and Sociality, Institute for Basic Science (IBS), Daejeon, Republic of Korea. .,KAIST Institute for the BioCentury, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea.
| |
Collapse
|
29
|
Horváth ÁC, Borbély S, Boros ÖC, Komáromi L, Koppa P, Barthó P, Fekete Z. Infrared neural stimulation and inhibition using an implantable silicon photonic microdevice. MICROSYSTEMS & NANOENGINEERING 2020; 6:44. [PMID: 34567656 PMCID: PMC8433474 DOI: 10.1038/s41378-020-0153-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/04/2019] [Revised: 12/19/2019] [Accepted: 02/24/2020] [Indexed: 05/07/2023]
Abstract
Brain is one of the most temperature sensitive organs. Besides the fundamental role of temperature in cellular metabolism, thermal response of neuronal populations is also significant during the evolution of various neurodegenerative diseases. For such critical environmental factor, thorough mapping of cellular response to variations in temperature is desired in the living brain. So far, limited efforts have been made to create complex devices that are able to modulate temperature, and concurrently record multiple features of the stimulated region. In our work, the in vivo application of a multimodal photonic neural probe is demonstrated. Optical, thermal, and electrophysiological functions are monolithically integrated in a single device. The system facilitates spatial and temporal control of temperature distribution at high precision in the deep brain tissue through an embedded infrared waveguide, while it provides recording of the artefact-free electrical response of individual cells at multiple locations along the probe shaft. Spatial distribution of the optically induced temperature changes is evaluated through in vitro measurements and a validated multi-physical model. The operation of the multimodal microdevice is demonstrated in the rat neocortex and in the hippocampus to increase or suppress firing rate of stimulated neurons in a reversible manner using continuous wave infrared light (λ = 1550 nm). Our approach is envisioned to be a promising candidate as an advanced experimental toolset to reveal thermally evoked responses in the deep neural tissue.
Collapse
Affiliation(s)
- Ágoston Csaba Horváth
- Research Group for Implantable Microsystems, Faculty of Information Technology & Bionics, Pázmány Péter Catholic University, Budapest, Hungary
- Microsystems Laboratory, Institute for Technical Physics & Material Science, Centre for Energy Research, Budapest, Hungary
- Óbuda University Doctoral School on Materials Sciences and Technologies, Budapest, Hungary
| | - Sándor Borbély
- MTA TTK NAP Sleep Oscillations Research Group, Budapest, Hungary
- Department of Physiology and Neurobiology, Eötvös Loránd University, Budapest, Hungary
| | - Örs Csanád Boros
- Department of Atomic Physics, Budapest University of Technology & Economics, Budapest, Hungary
| | - Lili Komáromi
- Department of Atomic Physics, Budapest University of Technology & Economics, Budapest, Hungary
| | - Pál Koppa
- Department of Atomic Physics, Budapest University of Technology & Economics, Budapest, Hungary
| | - Péter Barthó
- MTA TTK NAP Sleep Oscillations Research Group, Budapest, Hungary
| | - Zoltán Fekete
- Research Group for Implantable Microsystems, Faculty of Information Technology & Bionics, Pázmány Péter Catholic University, Budapest, Hungary
- Microsystems Laboratory, Institute for Technical Physics & Material Science, Centre for Energy Research, Budapest, Hungary
| |
Collapse
|
30
|
Gagnon-Turcotte G, Keramidis I, Ethier C, De Koninck Y, Gosselin B. A Wireless Electro-Optic Headstage With a 0.13- μm CMOS Custom Integrated DWT Neural Signal Decoder for Closed-Loop Optogenetics. IEEE TRANSACTIONS ON BIOMEDICAL CIRCUITS AND SYSTEMS 2019; 13:1036-1051. [PMID: 31352352 DOI: 10.1109/tbcas.2019.2930498] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
We present a wireless electro-optic headstage that uses a 0.13- μm CMOS custom integrated circuit (IC) implementing a digital neural decoder (ND-IC) for enabling real-time closed-loop (CL) optogenetics. The ND-IC processes the neural activity data using three digital cores: 1) the detector core detects and extracts the action potential (AP) of individual neurons by using an adaptive threshold; 2) the data compression core compresses the detected AP by using an efficient Symmlet-2 discrete wavelet transform (DWT) processor for decreasing the amount of data to be transmitted by the low-power wireless link; and 3) the classification core sorts the compressed AP into separated clusters on the fly according to their wave shapes. The ND-IC encompasses several innovations: 1) the compression core decreases the complexity from O(n 2) to O(n · log(n)) compared to the previous solutions, while using two times less memory, thanks to the use of a new coefficient sorting tree; and 2) the AP classification core reuses both the compressed DWT coefficients to perform implicit dimensionality reduction, which allows for performing intensive signal processing on-chip, while increasing power and hardware efficiency. This core also reuses the signal standard deviation already computed by the AP detector core as threshold for performing automatic AP sorting. The headstage also introduces innovations by enabling a new wireless CL scheme between the neural data acquisition module and the optical stimulator. Our CL scheme uses the AP sorting and timing information produced by the ND-IC for detecting complex firing patterns within the brain. The headstage is also smaller (1.13 cm 3), lighter (3.0 g with a 40 mAh battery) and less invasive than the previous solutions, while providing a measured autonomy of 2h40, with the ND-IC. The whole system and the ND-IC are first validated in vivo in the LD thalamus of a Long-Evans rat, and then in freely-moving CL experiments involving a mouse virally expressing ChR2-mCherry in inhibitory neurons of the prelimbic cortex, and the results show that our system works well within an in vivo experimental setting with a freely moving mouse.
Collapse
|
31
|
Bartley AF, Abiraman K, Stewart LT, Hossain MI, Gahan DM, Kamath AV, Burdette MK, Andrabe S, Foulger SH, McMahon LL, Dobrunz LE. LSO:Ce Inorganic Scintillators Are Biocompatible With Neuronal and Circuit Function. Front Synaptic Neurosci 2019; 11:24. [PMID: 31551750 PMCID: PMC6733890 DOI: 10.3389/fnsyn.2019.00024] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Accepted: 08/06/2019] [Indexed: 12/19/2022] Open
Abstract
Optogenetics is widely used in neuroscience to control neural circuits. However, non-invasive methods for light delivery in brain are needed to avoid physical damage caused by current methods. One potential strategy could employ x-ray activation of radioluminescent particles (RPLs), enabling localized light generation within the brain. RPLs composed of inorganic scintillators can emit light at various wavelengths depending upon composition. Cerium doped lutetium oxyorthosilicate (LSO:Ce), an inorganic scintillator that emits blue light in response to x-ray or ultraviolet (UV) stimulation, could potentially be used to control neural circuits through activation of channelrhodopsin-2 (ChR2), a light-gated cation channel. Whether inorganic scintillators themselves negatively impact neuronal processes and synaptic function is unknown, and was investigated here using cellular, molecular, and electrophysiological approaches. As proof of principle, we applied UV stimulation to 4 μm LSO:Ce particles during whole-cell recording of CA1 pyramidal cells in acute hippocampal slices from mice that expressed ChR2 in glutamatergic neurons. We observed an increase in frequency and amplitude of spontaneous excitatory postsynaptic currents (sEPSCs), indicating activation of ChR2 and excitation of neurons. Importantly, LSO:Ce particles did not affect survival of primary mouse cortical neurons, even after 24 h of exposure. In extracellular dendritic field potential recordings, no change in the strength of basal glutamatergic transmission was observed during exposure to LSO:Ce microparticles. However, the amplitude of the fiber volley was slightly reduced with high stimulation. Additionally, there was a slight decrease in the frequency of sEPSCs in whole-cell voltage-clamp recordings from CA1 pyramidal cells, with no change in current amplitudes. The amplitude and frequency of spontaneous inhibitory postsynaptic currents were unchanged. Finally, long term potentiation (LTP), a synaptic modification believed to underlie learning and memory and a robust measure of synaptic integrity, was successfully induced, although the magnitude was slightly reduced. Together, these results show LSO:Ce particles are biocompatible even though there are modest effects on baseline synaptic function and long-term synaptic plasticity. Importantly, we show that light emitted from LSO:Ce particles is able to activate ChR2 and modify synaptic function. Therefore, LSO:Ce inorganic scintillators are potentially viable for use as a new light delivery system for optogenetics.
Collapse
Affiliation(s)
- Aundrea F. Bartley
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, AL, United States
- Civitan International Research Center, University of Alabama at Birmingham, Birmingham, AL, United States
- Evelyn F. McKnight Brain Institute, University of Alabama at Birmingham, Birmingham, AL, United States
- Comprehensive Neuroscience Center, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Kavitha Abiraman
- Comprehensive Neuroscience Center, University of Alabama at Birmingham, Birmingham, AL, United States
- Department of Cell, Developmental, and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Luke T. Stewart
- Comprehensive Neuroscience Center, University of Alabama at Birmingham, Birmingham, AL, United States
- Department of Cell, Developmental, and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Mohammed Iqbal Hossain
- Department of Pharmacology, University of Alabama at Birmingham, Birmingham, AL, United States
| | - David M. Gahan
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Abhishek V. Kamath
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Mary K. Burdette
- Department of Materials Science and Engineering, Clemson University, Anderson, SC, United States
| | - Shaida Andrabe
- Department of Pharmacology, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Stephen H. Foulger
- Department of Materials Science and Engineering, Clemson University, Anderson, SC, United States
- Center for Optical Materials Science and Engineering Technologies, Clemson University, Anderson, SC, United States
- Department of Bioengineering, Clemson University, Clemson, SC, United States
| | - Lori L. McMahon
- Civitan International Research Center, University of Alabama at Birmingham, Birmingham, AL, United States
- Evelyn F. McKnight Brain Institute, University of Alabama at Birmingham, Birmingham, AL, United States
- Comprehensive Neuroscience Center, University of Alabama at Birmingham, Birmingham, AL, United States
- Department of Cell, Developmental, and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Lynn E. Dobrunz
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, AL, United States
- Civitan International Research Center, University of Alabama at Birmingham, Birmingham, AL, United States
- Evelyn F. McKnight Brain Institute, University of Alabama at Birmingham, Birmingham, AL, United States
- Comprehensive Neuroscience Center, University of Alabama at Birmingham, Birmingham, AL, United States
| |
Collapse
|
32
|
Srinivasan S, Schelhaas B, Maimon B, Song H, Herr H. Retinal supplementation augments optogenetic stimulation efficacy in vivo. J Neural Eng 2019; 16:054002. [PMID: 31039554 DOI: 10.1088/1741-2552/ab1e22] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
OBJECTIVE Over the last two decades, optical control of neuronal activity in the central nervous system has seen rapid development, demonstrating the utility of optogenetics as both an experimental and therapeutic tool. Conversely, applications of optogenetics in the peripheral nervous system have been relatively constrained by the challenges of temporally variable opsin expression, light penetration and immune attack of non-native opsins. Whilst opsin expression can be increased significantly through high-concentration viral induction, subsequent attack by the immune system causes temporal decay and high variability in electrophysiological response. APPROACH In this study, we present a method to circumvent the aforementioned challenges by locally supplementing all-trans-retinal (ATR) (via a slow release pellet) to increase tissue photosensitivity in transgenic mice expressing channelrhodopsin 2 (ChR2) in nerves. MAIN RESULTS In mice supplemented with ATR, we demonstrate enhanced electrophysiological activation and fatigue tolerance in response to optical stimulation for six weeks. SIGNIFICANCE Local supplementation of ATR enables improved optogenetic stimulation efficacy in peripheral nerves. This method enables greater exploration of neurophysiology and development of clinically-viable optogenetic treatments in the peripheral nervous system.
Collapse
Affiliation(s)
- Shriya Srinivasan
- Center for Extreme Bionics, Massachusetts Institute of Technology, Cambridge, MA 02139, United States of America. Harvard-MIT Division of Health Sciences and Technology (HST), Massachusetts Institute of Technology, Cambridge, MA 02139, United States of America
| | | | | | | | | |
Collapse
|
33
|
Fabbrini F, Van den Haute C, De Vitis M, Baekelandt V, Vanduffel W, Vogels R. Probing the Mechanisms of Repetition Suppression in Inferior Temporal Cortex with Optogenetics. Curr Biol 2019; 29:1988-1998.e4. [PMID: 31178318 DOI: 10.1016/j.cub.2019.05.014] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Revised: 04/03/2019] [Accepted: 05/02/2019] [Indexed: 01/14/2023]
Abstract
Neurons in macaque inferior temporal (IT) cortex show a decrease in the response with stimulus repetition, known as repetition suppression (RS). Several mechanisms may contribute to RS in IT, such as firing rate-dependent fatigue and transsynaptic mechanisms, like synaptic depression or reduced input from neurons within the same area or from up- or downstream areas. We examined the role of firing rate fatigue and transsynaptic mechanisms by stimulating directly IT neurons using optogenetics and measured the effect of photo-stimulation on their responses using timing parameters that resulted in RS for visual stimuli. Photo-stimulation of IT neurons resulted in a marginally decreased probability of spiking activity to a subsequent photo-stimulation or to a subsequent low-contrast visual stimulus. This response reduction was small relative to that for repeated visual stimuli and was related to post-stimulation inhibition of the activity during the interval between adapter and test stimuli. Presentation of a visual adapter did not change the response to subsequent photo-stimulation. In neurons whose response to the visual adapter was inhibited by simultaneous photo-stimulation, RS to visual stimuli was unaffected. Overall, these data imply that RS in IT has a transsynaptic origin, with little or no contribution of intrinsic firing rate fatigue. In addition, they suggest a limited contribution of both local synaptic depression and reduced input from nearby IT neurons, whose responses were postulated to be decreased by firing rate fatigue, to RS in IT.
Collapse
Affiliation(s)
- Francesco Fabbrini
- Laboratory for Neuro- and Psychophysiology, KU Leuven, Leuven 3000, Belgium; Leuven Brain Institute, KU Leuven, Leuven 3000, Belgium
| | - Chris Van den Haute
- Leuven Brain Institute, KU Leuven, Leuven 3000, Belgium; Laboratory for Neurobiology and Gene Therapy, KU Leuven, Leuven 3000, Belgium
| | - Marina De Vitis
- Laboratory for Neuro- and Psychophysiology, KU Leuven, Leuven 3000, Belgium; Leuven Brain Institute, KU Leuven, Leuven 3000, Belgium; Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna 40126, Italy
| | - Veerle Baekelandt
- Leuven Brain Institute, KU Leuven, Leuven 3000, Belgium; Laboratory for Neurobiology and Gene Therapy, KU Leuven, Leuven 3000, Belgium
| | - Wim Vanduffel
- Laboratory for Neuro- and Psychophysiology, KU Leuven, Leuven 3000, Belgium; Leuven Brain Institute, KU Leuven, Leuven 3000, Belgium; Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown, MA 02129, USA; Department of Radiology, Harvard Medical School, Boston, MA 02115, USA
| | - Rufin Vogels
- Laboratory for Neuro- and Psychophysiology, KU Leuven, Leuven 3000, Belgium; Leuven Brain Institute, KU Leuven, Leuven 3000, Belgium.
| |
Collapse
|
34
|
Pezo F, Zambrano F, Uribe P, Ramírez‐Reveco A, Romero F, Sanchéz R. LED‐based red light photostimulation improves short‐term response of cooled boar semen exposed to thermal stress at 37°C. Andrologia 2019; 51:e13237. [DOI: 10.1111/and.13237] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2018] [Revised: 12/12/2018] [Accepted: 12/21/2018] [Indexed: 12/14/2022] Open
Affiliation(s)
- Felipe Pezo
- Laboratory of Reproductive Medicine and Molecular Endocrinology Center for Translational Medicine (CEMT‐BIOREN) Temuco Chile
| | - Fabiola Zambrano
- Laboratory of Reproductive Medicine and Molecular Endocrinology Center for Translational Medicine (CEMT‐BIOREN) Temuco Chile
- Department of Preclinical Sciences, Faculty of Medicine Universidad de La Frontera Temuco Chile
| | - Pamela Uribe
- Laboratory of Reproductive Medicine and Molecular Endocrinology Center for Translational Medicine (CEMT‐BIOREN) Temuco Chile
- Department of Internal Medicine, Faculty of Medicine Universidad de La Frontera Temuco Chile
| | - Alfredo Ramírez‐Reveco
- Laboratory of Cryobiology and Analysis of Spermatic Functionality, Institute of Animal Science, Faculty of Veterinary Sciences Universidad Austral Valdivia Chile
| | - Fernando Romero
- Department of Preclinical Sciences, Faculty of Medicine Universidad de La Frontera Temuco Chile
| | - Raúl Sanchéz
- Laboratory of Reproductive Medicine and Molecular Endocrinology Center for Translational Medicine (CEMT‐BIOREN) Temuco Chile
- Department of Preclinical Sciences, Faculty of Medicine Universidad de La Frontera Temuco Chile
| |
Collapse
|
35
|
Wirdatmadja S, Johari P, Desai A, Bae Y, Stachowiak EK, Stachowiak MK, Jornet JM, Balasubramaniam S. Analysis of Light Propagation on Physiological Properties of Neurons for Nanoscale Optogenetics. IEEE Trans Neural Syst Rehabil Eng 2019; 27:108-117. [PMID: 30624220 DOI: 10.1109/tnsre.2019.2891271] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Miniaturization of implantable devices is an important challenge for future brain-computer interface applications, and in particular for achieving precise neuron stimulation. For stimulation that utilizes light, i.e., optogenetics, the light propagation behavior and interaction at the nanoscale with elements within the neuron is an important factor that needs to be considered when designing the device. This paper analyzes the effect of light behavior for a single neuron stimulation and focuses on the impact from different cell shapes. Based on the Mie scattering theory, the paper analyzes how the shape of the soma and the nucleus contributes to the focusing effect resulting in an intensity increase, which ensures that neurons can assist in transferring light through the tissue toward the target cells. At the same time, this intensity increase can in turn also stimulate neighboring cells leading to interference within the neural circuits. This paper also analyzes the ideal placements of the device with respect to the angle and position within the cortex that can enable axonal biophoton communications, which can contain light within the cell to avoid the interference.
Collapse
|
36
|
Srinivasan SS, Maimon BE, Diaz M, Song H, Herr HM. Closed-loop functional optogenetic stimulation. Nat Commun 2018; 9:5303. [PMID: 30546051 PMCID: PMC6294002 DOI: 10.1038/s41467-018-07721-w] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2018] [Accepted: 11/16/2018] [Indexed: 12/30/2022] Open
Abstract
Optogenetics has been used to orchestrate temporal- and tissue-specific control of neural tissues and offers a wealth of unique advantages for neuromuscular control. Here, we establish a closed-loop functional optogenetic stimulation (CL-FOS) system to control ankle joint position in murine models. Using the measurement of either joint angle or fascicle length as a feedback signal, we compare the controllability of CL-FOS to closed-loop functional electrical stimulation (CL-FES) and demonstrate significantly greater accuracy, lower rise times and lower overshoot percentages. We demonstrate orderly recruitment of motor units and reduced fatigue when performing cyclical movements with CL-FOS compared with CL-FES. We develop and investigate a 3-phase, photo-kinetic model to elucidate the underlying mechanisms for temporal variations in optogenetically activated neuromusculature during closed-loop control experiments. Methods and insights from this study lay the groundwork for the development of closed-loop optogenetic neuromuscular stimulation therapies and devices for peripheral limb control.
Collapse
Affiliation(s)
- Shriya S Srinivasan
- MIT Media Lab, Center for Extreme Bionics, Massachusetts Institute of Technology, Cambridge, 02139, MA, USA.
- Harvard-MIT Division of Health Sciences and Technology (HST), Massachusetts Institute of Technology, Cambridge, 02139, MA, USA.
| | - Benjamin E Maimon
- MIT Media Lab, Center for Extreme Bionics, Massachusetts Institute of Technology, Cambridge, 02139, MA, USA
- Harvard-MIT Division of Health Sciences and Technology (HST), Massachusetts Institute of Technology, Cambridge, 02139, MA, USA
| | - Maurizio Diaz
- MIT Media Lab, Center for Extreme Bionics, Massachusetts Institute of Technology, Cambridge, 02139, MA, USA
- Department of Electrical Engineering and Computer Science, Massachusetts Institute of Technology, Cambridge, 02139, MA, USA
| | - Hyungeun Song
- MIT Media Lab, Center for Extreme Bionics, Massachusetts Institute of Technology, Cambridge, 02139, MA, USA
- Harvard-MIT Division of Health Sciences and Technology (HST), Massachusetts Institute of Technology, Cambridge, 02139, MA, USA
| | - Hugh M Herr
- MIT Media Lab, Center for Extreme Bionics, Massachusetts Institute of Technology, Cambridge, 02139, MA, USA.
| |
Collapse
|
37
|
Ait Ouares K, Beurrier C, Canepari M, Laverne G, Kuczewski N. Opto nongenetics inhibition of neuronal firing. Eur J Neurosci 2018; 49:6-26. [PMID: 30387216 DOI: 10.1111/ejn.14251] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Revised: 09/07/2018] [Accepted: 09/14/2018] [Indexed: 01/28/2023]
Abstract
Optogenetics is based on the selective expression of exogenous opsins by neurons allowing experimental control of their electrical activity using visible light. The interpretation of the results of optogenetic experiments is based on the assumption that light stimulation selectively acts on those neurons expressing the exogenous opsins without perturbing the activity of naive ones. Here, we report that light stimulation, of wavelengths and power in the range of those normally used in optogenetic experiments, consistently reduces the firing activity of naive Mitral Cells (MCs) and Tufted Neurons in the olfactory bulb as well as in Medium Spiny Neurons (MSNs) in the striatum. No such effect was observed for cerebellar Purkinje and hippocampal CA1 neurons. The effects on MC firing appear to be mainly due to a light-induced increase in tissue temperature, between 0.1 and 0.4°C, associated with the generation of a hyperpolarizing current and a modification of action potential (AP) shape. Therefore, light in the visible range can affect neuronal physiology in a cell-specific manner. Beside the implications for optogenetic studies, our results pave the way to investigating the use of visible light for therapeutic purposes in pathologies associated with neuronal hyperexcitability.
Collapse
Affiliation(s)
- Karima Ait Ouares
- Univ. Grenoble Alpes, CNRS, LIPhy, Grenoble, France.,Laboratories of Excellence, Ion Channel Science and Therapeutics, Grenoble, France
| | - Corinne Beurrier
- Univ. Grenoble Alpes, CNRS, LIPhy, Grenoble, France.,Laboratories of Excellence, Ion Channel Science and Therapeutics, Grenoble, France.,Aix Marseille University, CNRS, IBDM, Marseille, France
| | - Marco Canepari
- Univ. Grenoble Alpes, CNRS, LIPhy, Grenoble, France.,Laboratories of Excellence, Ion Channel Science and Therapeutics, Grenoble, France.,Institut National de la Santé et Recherche Médicale, Paris, France
| | | | - Nicola Kuczewski
- CNRS, UMR 5292, INSERM, U1028, Lyon, France.,Lyon Neuroscience Research Center, Neuroplasticity and neuropathology of olfactory perception Team, Lyon, France.,University Lyon, Lyon, Franc.,University Lyon1, Villeurbanne, France
| |
Collapse
|
38
|
Dubois A, Chiang CC, Smekens F, Jan S, Cuplov V, Palfi S, Chuang KS, Senova S, Pain F. Optical and thermal simulations for the design of optodes for minimally invasive optogenetics stimulation or photomodulation of deep and large cortical areas in non-human primate brain. J Neural Eng 2018; 15:065004. [PMID: 30190446 DOI: 10.1088/1741-2552/aadf97] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
The use of optogenetics or photobiomodulation in non-human primate (NHP) requires the ability to noninvasively stimulate large and deep cortical brain tissues volumes. In this context, the optical and geometrical parameters of optodes are critical. Methods and general guidelines to optimize these parameters have to be defined. OBJECTIVE We propose the design of an optode for safe and efficient optical stimulation of a large volume of NHP cortex, down to 3-5 mm depths without inserting fibers into the cortex. APPROACH Monte Carlo simulations of optical and thermal transport have been carried out using the Geant4 application for tomographic emission (GATE) platform. Parameters such as the fiber diameter, numerical aperture, number of fibers and their geometrical arrangement have been studied. Optimal hardware parameters are proposed to obtain homogeneous fluence above the fluence threshold for opsin activation without detrimental thermal effects. MAIN RESULTS The simulations show that a large fiber diameter and a large numerical aperture are preferable since they allow limiting power concentration and hence the resulting thermal increases at the brain surface. To obtain a volume of 200-500 mm3 of brain tissues receiving a fluence above the opsin activation threshold for optogenetics or below a phototocixity threshold for photobiomodulation, a 4 fibers configuration is proposed. The optimal distance between the fibers was found to be 4 mm. A practical implementation of the optode has been performed and the corresponding fluence and thermal maps have been simulated. SIGNIFICANCE The present study defines a method to optimize the design of optode and the choice of stimulation parameters for optogenetics and more generally light delivery to deep and large volumes of tissues in NHP brain with a controlled irradiance dosimetry. The general guidelines are the use of silica fibers with a large numerical aperture and a large diameter. The combination of several fibers is required if large volumes need to be stimulated while avoiding thermal effects.
Collapse
Affiliation(s)
- A Dubois
- IMNC, CNRS, Université Paris-Sud, Université Paris Saclay, Orsay F-91405, France
| | | | | | | | | | | | | | | | | |
Collapse
|
39
|
The Role of Cholinergic Midbrain Neurons in Startle and Prepulse Inhibition. J Neurosci 2018; 38:8798-8808. [PMID: 30171090 DOI: 10.1523/jneurosci.0984-18.2018] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Revised: 08/01/2018] [Accepted: 08/20/2018] [Indexed: 01/21/2023] Open
Abstract
One of the two major cholinergic centers of the mammalian brain is located in the midbrain, i.e., the pedunculopontine tegmentum (PPTg) and the adjacent laterodorsal tegmentum. These cholinergic neurons have been shown to be important for e.g., arousal, reward associations, and sleep. They also have been suggested to mediate sensorimotor gating, measured as prepulse inhibition of startle (PPI). PPI disruptions are a hallmark of schizophrenia and are observed in various other psychiatric disorders, where they are associated with, and often predictive of, other cognitive symptoms. PPI has been proposed to be mediated by a short midbrain circuitry including inhibitory cholinergic projections from PPTg to the startle pathway. Although the data indicating the involvement of the PPTg is very robust, some more recent evidence challenges that there is a cholinergic contribution to PPI. We here use transient optogenetic activation of specifically the cholinergic PPTg neurons in male and female rats to address their role in startle modulation in general, and in PPI specifically. Although we could confirm the crucial role of PPTg cholinergic neurons in associative reward learning, validating our experimental approach, we found that activation of cholinergic PPTg neurons did not inhibit startle responses. In contrast, activation of cholinergic PPTg neurons enhanced startle, which is in accordance with their general role in arousal and indicate a potential involvement in sensitization of startle. We conclude that noncholinergic PPTg neurons mediate PPI in contrast to the longstanding hypothetical view that PPI is mediated by cholinergic PPTg neurons.SIGNIFICANCE STATEMENT Activation of cholinergic neurons in the midbrain has been assumed to mediate prepulse inhibition of startle (PPI), a common measure of sensorimotor gating that is disrupted in schizophrenia and other psychiatric disorders. We here revisit this long-standing hypothesis using optogenetic activation of these specific neurons combined with startle testing in rats. In contrast to the hypothetical role of these neurons in startle modulation, we show that their activation leads to an increase of baseline startle and to prepulse facilitation. This supports recent data by others that have started to cast some doubt on the cholinergic hypothesis of PPI, and calls for a revision of the theoretical construct of PPI mechanisms.
Collapse
|
40
|
Optogenetic Tractography for anatomo-functional characterization of cortico-subcortical neural circuits in non-human primates. Sci Rep 2018; 8:3362. [PMID: 29463867 PMCID: PMC5820256 DOI: 10.1038/s41598-018-21486-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2017] [Accepted: 01/29/2018] [Indexed: 11/08/2022] Open
Abstract
Dissecting neural circuitry in non-human primates (NHP) is crucial to identify potential neuromodulation anatomical targets for the treatment of pharmacoresistant neuropsychiatric diseases by electrical neuromodulation. How targets of deep brain stimulation (DBS) and cortical targets of transcranial magnetic stimulation (TMS) compare and might complement one another is an important question. Combining optogenetics and tractography may enable anatomo-functional characterization of large brain cortico-subcortical neural pathways. For the proof-of-concept this approach was used in the NHP brain to characterize the motor cortico-subthalamic pathway (m_CSP) which might be involved in DBS action mechanism in Parkinson’s disease (PD). Rabies-G-pseudotyped and Rabies-G-VSVg-pseudotyped EIAV lentiviral vectors encoding the opsin ChR2 gene were stereotaxically injected into the subthalamic nucleus (STN) and were retrogradely transported to the layer of the motor cortex projecting to STN. A precise anatomical mapping of this pathway was then performed using histology-guided high angular resolution MRI tractography guiding accurately cortical photostimulation of m_CSP origins. Photoexcitation of m_CSP axon terminals or m_CSP cortical origins modified the spikes distribution for photosensitive STN neurons firing rate in non-equivalent ways. Optogenetic tractography might help design preclinical neuromodulation studies in NHP models of neuropsychiatric disease choosing the most appropriate target for the tested hypothesis.
Collapse
|
41
|
Wiegert JS, Mahn M, Prigge M, Printz Y, Yizhar O. Silencing Neurons: Tools, Applications, and Experimental Constraints. Neuron 2017; 95:504-529. [PMID: 28772120 DOI: 10.1016/j.neuron.2017.06.050] [Citation(s) in RCA: 173] [Impact Index Per Article: 24.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2017] [Revised: 06/29/2017] [Accepted: 06/30/2017] [Indexed: 12/16/2022]
Abstract
Reversible silencing of neuronal activity is a powerful approach for isolating the roles of specific neuronal populations in circuit dynamics and behavior. In contrast with neuronal excitation, for which the majority of studies have used a limited number of optogenetic and chemogenetic tools, the number of genetically encoded tools used for inhibition of neuronal activity has vastly expanded. Silencing strategies vary widely in their mechanism of action and in their spatial and temporal scales. Although such manipulations are commonly applied, the design and interpretation of neuronal silencing experiments present unique challenges, both technically and conceptually. Here, we review the most commonly used tools for silencing neuronal activity and provide an in-depth analysis of their mechanism of action and utility for particular experimental applications. We further discuss the considerations that need to be given to experimental design, analysis, and interpretation of collected data. Finally, we discuss future directions for the development of new silencing approaches in neuroscience.
Collapse
Affiliation(s)
- J Simon Wiegert
- Research Group Synaptic Wiring and Information Processing, Center for Molecular Neurobiology Hamburg, Falkenried 94, 20251 Hamburg, Germany
| | - Mathias Mahn
- Department of Neurobiology, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Matthias Prigge
- Department of Neurobiology, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Yoav Printz
- Department of Neurobiology, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Ofer Yizhar
- Department of Neurobiology, Weizmann Institute of Science, Rehovot 76100, Israel.
| |
Collapse
|