1
|
Zheng R, Xue Z, You M. Optogenetic Tools for Regulating RNA Metabolism and Functions. Chembiochem 2024; 25:e202400615. [PMID: 39316432 DOI: 10.1002/cbic.202400615] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Revised: 09/22/2024] [Accepted: 09/24/2024] [Indexed: 09/25/2024]
Abstract
RNA molecules play a vital role in linking genetic information with various cellular processes. In recent years, a variety of optogenetic tools have been engineered for regulating cellular RNA metabolism and functions. These highly desirable tools can offer non-intrusive control with spatial precision, remote operation, and biocompatibility. Here, we would like to review these currently available approaches that can regulate RNAs with light: from non-genetically encodable chemically modified oligonucleotides to genetically encoded RNA aptamers that recognize photosensitive small-molecule or protein ligands. Some key applications of these optogenetic tools will also be highlighted to illustrate how they have been used for regulating all aspects of the RNA life cycle: from RNA synthesis, maturation, modification, and translation to their degradation, localization, and phase separation control. Some current challenges and potential practical utilizations of these RNA optogenetic tools will also be discussed.
Collapse
Affiliation(s)
- Ru Zheng
- Department of Chemistry, University of Massachusetts, Amherst, MA, 01003, USA
| | - Zhaolin Xue
- Department of Chemistry, University of Massachusetts, Amherst, MA, 01003, USA
| | - Mingxu You
- Department of Chemistry, University of Massachusetts, Amherst, MA, 01003, USA
| |
Collapse
|
2
|
Zhu M, Fang Y, Sun Y, Li S, Yu J, Xiong B, Wen C, Zhou B, Huang B, Yin H, Xu H. Sonogenetics in the Treatment of Chronic Diseases: A New Method for Cell Regulation. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2407373. [PMID: 39488795 DOI: 10.1002/advs.202407373] [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: 08/28/2024] [Revised: 10/21/2024] [Indexed: 11/04/2024]
Abstract
Sonogenetics is an innovative technology that integrates ultrasound with genetic editing to precisely modulate cellular activities in a non-invasive manner. This method entails introducing and activating mechanosensitive channels on the cell membrane of specific cells using gene delivery vectors. When exposed to ultrasound, these channels can be manipulated to open or close, thereby impacting cellular functions. Sonogenetics is currently being used extensively in the treatment of various chronic diseases, including Parkinson's disease, vision restoration, and cancer therapy. This paper provides a comprehensive review of key components of sonogenetics and focuses on evaluating its prospects and potential challenges in the treatment of chronic disease.
Collapse
Affiliation(s)
- Mingrui Zhu
- Department of Ultrasound, Institute of Ultrasound in Medicine and Engineering, Zhongshan Hospital, Fudan University, Shanghai, 200032, P. R. China
| | - Yan Fang
- Department of Ultrasound, Huashan Hospital, Fudan University, Shanghai, 200040, P. R. China
| | - Yikang Sun
- Department of Ultrasound, Institute of Ultrasound in Medicine and Engineering, Zhongshan Hospital, Fudan University, Shanghai, 200032, P. R. China
| | - Shaoyue Li
- Department of Medical Ultrasound, Center of Minimally Invasive Treatment for Tumor, Shanghai Tenth People's Hospital, Ultrasound Research and Education Institute, Clinical Research Center for Interventional Medicine, School of Medicine, Tongji University, Shanghai, 200072, P. R. China
| | - Jifeng Yu
- Department of Ultrasound, Institute of Ultrasound in Medicine and Engineering, Zhongshan Hospital, Fudan University, Shanghai, 200032, P. R. China
| | - Bing Xiong
- Department of Ultrasound, Institute of Ultrasound in Medicine and Engineering, Zhongshan Hospital, Fudan University, Shanghai, 200032, P. R. China
| | - Congjian Wen
- Department of Ultrasound, Institute of Ultrasound in Medicine and Engineering, Zhongshan Hospital, Fudan University, Shanghai, 200032, P. R. China
| | - Boyang Zhou
- Department of Ultrasound, Institute of Ultrasound in Medicine and Engineering, Zhongshan Hospital, Fudan University, Shanghai, 200032, P. R. China
| | - Bin Huang
- Zhejiang Hospital, Hangzhou, 310013, P. R. China
| | - Haohao Yin
- Department of Ultrasound, Institute of Ultrasound in Medicine and Engineering, Zhongshan Hospital, Fudan University, Shanghai, 200032, P. R. China
| | - Huixiong Xu
- Department of Ultrasound, Institute of Ultrasound in Medicine and Engineering, Zhongshan Hospital, Fudan University, Shanghai, 200032, P. R. China
| |
Collapse
|
3
|
Weiler S, Velez-Fort M, Margrie TW. Overcoming off-target optical stimulation-evoked cortical activity in the mouse brain in vivo. iScience 2024; 27:111152. [PMID: 39524362 PMCID: PMC11543908 DOI: 10.1016/j.isci.2024.111152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Revised: 09/21/2024] [Accepted: 10/08/2024] [Indexed: 11/16/2024] Open
Abstract
Exogenous opsins allow for in vivo interrogation of brain circuits at unprecedented temporal and spatial precision. Here, we found that optical fiber laser stimulation at wavelengths of 637, 594, or 473 nm within the cortex of mice lacking expression of exogenous opsins resulted in a strong neuronal response in the contralateral visual cortex. Evoked responses were observed even at low laser intensities (fiber tip power 1 mW) and most pronounced at 637 nm. We took advantage of retinal light adaptation by using a dim external light source (20 lux) that abolished the 594 and 473 nm-evoked neuronal responses even at high laser intensities (15 mW). The prevention of 637 nm-evoked responses, however, could only be achieved for stimulation intensities ≤ 2.5 mW. This highlights the need for careful selection of light wavelengths and intensities for optogenetic experiments. Additionally, retinal light adaptation offers an effective solution to minimize unintended activation.
Collapse
Affiliation(s)
- Simon Weiler
- Sainsbury Wellcome Centre for Neuronal Circuits and Behavior, University College London, 25 Howland Street, London W1T 4JG, UK
| | - Mateo Velez-Fort
- Sainsbury Wellcome Centre for Neuronal Circuits and Behavior, University College London, 25 Howland Street, London W1T 4JG, UK
| | - Troy W. Margrie
- Sainsbury Wellcome Centre for Neuronal Circuits and Behavior, University College London, 25 Howland Street, London W1T 4JG, UK
| |
Collapse
|
4
|
Yamakou ME, Zhu J, Martens EA. Inverse stochastic resonance in adaptive small-world neural networks. CHAOS (WOODBURY, N.Y.) 2024; 34:113119. [PMID: 39504100 DOI: 10.1063/5.0225760] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Accepted: 10/16/2024] [Indexed: 11/08/2024]
Abstract
Inverse stochastic resonance (ISR) is a counterintuitive phenomenon where noise reduces the oscillation frequency of an oscillator to a minimum occurring at an intermediate noise intensity, and sometimes even to the complete absence of oscillations. In neuroscience, ISR was first experimentally verified with cerebellar Purkinje neurons [Buchin et al., PLOS Comput. Biol. 12, e1005000 (2016)]. These experiments showed that ISR enables a locally optimal information transfer between the input and output spike train of neurons. Subsequent studies have further demonstrated the efficiency of information processing and transfer in neural networks with small-world network topology. We have conducted a numerical investigation into the impact of adaptivity on ISR in a small-world network of noisy FitzHugh-Nagumo (FHN) neurons, operating in a bi-metastable regime consisting of a metastable fixed point and a metastable limit cycle. Our results show that the degree of ISR is highly dependent on the value of the FHN model's timescale separation parameter ε. The network structure undergoes dynamic adaptation via mechanisms of either spike-time-dependent plasticity (STDP) with potentiation-/depression-domination parameter P or homeostatic structural plasticity (HSP) with rewiring frequency F. We demonstrate that both STDP and HSP amplify the effect of ISR when ε lies within the bi-stability region of FHN neurons. Specifically, at larger values of ε within the bi-stability regime, higher rewiring frequencies F are observed to enhance ISR at intermediate (weak) synaptic noise intensities, while values of P consistent with depression-domination (potentiation-domination) consistently enhance (deteriorate) ISR. Moreover, although STDP and HSP control parameters may jointly enhance ISR, P has a greater impact on improving ISR compared to F. Our findings inform future ISR enhancement strategies in noisy artificial neural circuits, aiming to optimize local information transfer between input and output spike trains in neuromorphic systems and prompt venues for experiments in neural networks.
Collapse
Affiliation(s)
- Marius E Yamakou
- Department of Data Science, Friedrich-Alexander-Universität Erlangen-Nürnberg, Cauerstr. 11, 91058 Erlangen, Germany
| | - Jinjie Zhu
- State Key Laboratory of Mechanics and Control for Aerospace Structures, College of Aerospace Engineering, Nanjing University of Aeronautics and Astronautics, Nanjing 210016, China
| | - Erik A Martens
- Centre for Mathematical Sciences, Lund University, Sölvegatan 18B, 221 00 Lund, Sweden
| |
Collapse
|
5
|
Phalip A, Netser S, Wagner S. Understanding the neurobiology of social behavior through exploring brain-wide dynamics of neural activity. Neurosci Biobehav Rev 2024; 165:105856. [PMID: 39159735 DOI: 10.1016/j.neubiorev.2024.105856] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Revised: 08/11/2024] [Accepted: 08/14/2024] [Indexed: 08/21/2024]
Abstract
Social behavior is highly complex and adaptable. It can be divided into multiple temporal stages: detection, approach, and consummatory behavior. Each stage can be further divided into several cognitive and behavioral processes, such as perceiving social cues, evaluating the social and non-social contexts, and recognizing the internal/emotional state of others. Recent studies have identified numerous brain-wide circuits implicated in social behavior and suggested the existence of partially overlapping functional brain networks underlying various types of social and non-social behavior. However, understanding the brain-wide dynamics underlying social behavior remains challenging, and several brain-scale dynamics (macro-, meso-, and micro-scale levels) need to be integrated. Here, we suggest leveraging new tools and concepts to explore social brain networks and integrate those different levels. These include studying the expression of immediate-early genes throughout the entire brain to impartially define the structure of the neuronal networks involved in a given social behavior. Then, network dynamics could be investigated using electrode arrays or multi-channel fiber photometry. Finally, tools like high-density silicon probes and miniscopes can probe neural activity in specific areas and across neuronal populations at the single-cell level.
Collapse
Affiliation(s)
- Adèle Phalip
- Sagol Department of Neurobiology, Faculty of Natural Sciences, University of Haifa, Haifa, Israel.
| | - Shai Netser
- Sagol Department of Neurobiology, Faculty of Natural Sciences, University of Haifa, Haifa, Israel
| | - Shlomo Wagner
- Sagol Department of Neurobiology, Faculty of Natural Sciences, University of Haifa, Haifa, Israel
| |
Collapse
|
6
|
Jons CK, Cheng D, Dong C, Meany EL, Nassi JJ, Appel EA. Viral Vector Eluting Lenses for Single-Step Targeted Expression of Genetically-Encoded Activity Sensors for in Vivo Microendoscopic Calcium Imaging. Macromol Biosci 2024:e2400359. [PMID: 39283817 DOI: 10.1002/mabi.202400359] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Indexed: 09/25/2024]
Abstract
Optical methods for studying the brain offer powerful approaches for understanding how neural activity underlies complex behavior. These methods typically rely on genetically encoded sensors and actuators to monitor and control neural activity. For microendoscopic calcium imaging, injection of a virus followed by implantation of a lens probe is required to express a calcium sensor and enable optical access to the target brain region. This two-step process poses several challenges, chief among them being the risks associated with mistargeting and/or misalignment between virus expression zone, lens probe and target brain region. Here, an adeno-associated virus (AAV)-eluting polymer coating is engineered for gradient refractive index (GRIN) lenses enabling the expression of a genetically encoded calcium indicator (GCaMP) directly within the brain region of interest upon implantation of the lens. This approach requires only one surgical step and guarantees alignment between GCaMP expression and lens in the brain. Additionally, the slow virus release from these coatings increases the working time for surgical implantation, expanding the brain regions and species amenable to this approach. These enhanced capabilities should accelerate neuroscience research utilizing optical methods and advance the understanding of the neural circuit mechanisms underlying brain function and behavior in health and disease.
Collapse
Affiliation(s)
- Carolyn K Jons
- Department of Materials Science & Engineering, Stanford University, Stanford, CA, 94305, USA
| | - David Cheng
- Inscopix - A Bruker Company, 1212 Terra Bella Ave. Suite 200, Mountain View, CA, 94043, USA
| | - Changxin Dong
- Department of Materials Science & Engineering, Stanford University, Stanford, CA, 94305, USA
| | - Emily L Meany
- Department of Bioengineering, Stanford University, Stanford, CA, 94305, USA
| | - Jonathan J Nassi
- Inscopix - A Bruker Company, 1212 Terra Bella Ave. Suite 200, Mountain View, CA, 94043, USA
| | - Eric A Appel
- Department of Materials Science & Engineering, Stanford University, Stanford, CA, 94305, USA
- Department of Bioengineering, Stanford University, Stanford, CA, 94305, USA
- Department of Pediatrics (Endocrinology), Stanford University, Stanford, CA, 94305, USA
- ChEM-H Institute, Stanford University, Stanford, CA, 94305, USA
- Woods Institute for the Environment, Stanford University, Stanford, CA, 94305, USA
| |
Collapse
|
7
|
Jons CK, Cheng D, Dong C, Meany EL, Nassi JJ, Appel EA. Viral vector eluting lenses for single-step targeted expression of genetically-encoded activity sensors for in vivo microendoscopic calcium imaging. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.11.09.566491. [PMID: 38014217 PMCID: PMC10680654 DOI: 10.1101/2023.11.09.566491] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2023]
Abstract
Optical methods for studying the brain offer powerful approaches for understanding how neural activity underlies complex behavior. These methods typically rely on genetically encoded sensors and actuators to monitor and control neural activity. For microendoscopic calcium imaging, injection of a virus followed by implantation of a lens probe is required to express a calcium sensor and enable optical access to the target brain region. This two-step process poses several challenges, chief among them being the risks associated with mistargeting and/or misalignment between virus expression zone, lens probe and target brain region. Here, we engineer an adeno-associated virus (AAV)-eluting polymer coating for gradient refractive index (GRIN) lenses enabling expression of a genetically encoded calcium indicator (GCaMP) directly within the brain region of interest upon implantation of the lens. This approach requires only one surgical step and guarantees alignment between GCaMP expression and lens in the brain. Additionally, the slow virus release from these coatings increases the working time for surgical implantation, expanding the brain regions and species amenable to this approach. These enhanced capabilities should accelerate neuroscience research utilizing optical methods and advance our understanding of the neural circuit mechanisms underlying brain function and behavior in health and disease.
Collapse
|
8
|
Lewis CM, Hoffmann A, Helmchen F. Linking brain activity across scales with simultaneous opto- and electrophysiology. NEUROPHOTONICS 2024; 11:033403. [PMID: 37662552 PMCID: PMC10472193 DOI: 10.1117/1.nph.11.3.033403] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Revised: 07/19/2023] [Accepted: 07/24/2023] [Indexed: 09/05/2023]
Abstract
The brain enables adaptive behavior via the dynamic coordination of diverse neuronal signals across spatial and temporal scales: from fast action potential patterns in microcircuits to slower patterns of distributed activity in brain-wide networks. Understanding principles of multiscale dynamics requires simultaneous monitoring of signals in multiple, distributed network nodes. Combining optical and electrical recordings of brain activity is promising for collecting data across multiple scales and can reveal aspects of coordinated dynamics invisible to standard, single-modality approaches. We review recent progress in combining opto- and electrophysiology, focusing on mouse studies that shed new light on the function of single neurons by embedding their activity in the context of brain-wide activity patterns. Optical and electrical readouts can be tailored to desired scales to tackle specific questions. For example, fast dynamics in single cells or local populations recorded with multi-electrode arrays can be related to simultaneously acquired optical signals that report activity in specified subpopulations of neurons, in non-neuronal cells, or in neuromodulatory pathways. Conversely, two-photon imaging can be used to densely monitor activity in local circuits while sampling electrical activity in distant brain areas at the same time. The refinement of combined approaches will continue to reveal previously inaccessible and under-appreciated aspects of coordinated brain activity.
Collapse
Affiliation(s)
| | - Adrian Hoffmann
- University of Zurich, Brain Research Institute, Zurich, Switzerland
- University of Zurich, Neuroscience Center Zurich, Zurich, Switzerland
| | - Fritjof Helmchen
- University of Zurich, Brain Research Institute, Zurich, Switzerland
- University of Zurich, Neuroscience Center Zurich, Zurich, Switzerland
- University of Zurich, University Research Priority Program, Adaptive Brain Circuits in Development and Learning, Zurich, Switzerland
| |
Collapse
|
9
|
Xia F, Rimoli CV, Akemann W, Ventalon C, Bourdieu L, Gigan S, de Aguiar HB. Neurophotonics beyond the surface: unmasking the brain's complexity exploiting optical scattering. NEUROPHOTONICS 2024; 11:S11510. [PMID: 38617592 PMCID: PMC11014413 DOI: 10.1117/1.nph.11.s1.s11510] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Revised: 03/07/2024] [Accepted: 03/14/2024] [Indexed: 04/16/2024]
Abstract
The intricate nature of the brain necessitates the application of advanced probing techniques to comprehensively study and understand its working mechanisms. Neurophotonics offers minimally invasive methods to probe the brain using optics at cellular and even molecular levels. However, multiple challenges persist, especially concerning imaging depth, field of view, speed, and biocompatibility. A major hindrance to solving these challenges in optics is the scattering nature of the brain. This perspective highlights the potential of complex media optics, a specialized area of study focused on light propagation in materials with intricate heterogeneous optical properties, in advancing and improving neuronal readouts for structural imaging and optical recordings of neuronal activity. Key strategies include wavefront shaping techniques and computational imaging and sensing techniques that exploit scattering properties for enhanced performance. We discuss the potential merger of the two fields as well as potential challenges and perspectives toward longer term in vivo applications.
Collapse
Affiliation(s)
- Fei Xia
- Sorbonne Université, Collège de France, Laboratoire Kastler Brossel, ENS-Université PSL, CNRS, Paris, France
| | - Caio Vaz Rimoli
- Sorbonne Université, Collège de France, Laboratoire Kastler Brossel, ENS-Université PSL, CNRS, Paris, France
- Université PSL, Institut de Biologie de l’ENS, École Normale Supérieure, CNRS, INSERM, Paris, France
| | - Walther Akemann
- Université PSL, Institut de Biologie de l’ENS, École Normale Supérieure, CNRS, INSERM, Paris, France
| | - Cathie Ventalon
- Université PSL, Institut de Biologie de l’ENS, École Normale Supérieure, CNRS, INSERM, Paris, France
| | - Laurent Bourdieu
- Université PSL, Institut de Biologie de l’ENS, École Normale Supérieure, CNRS, INSERM, Paris, France
| | - Sylvain Gigan
- Sorbonne Université, Collège de France, Laboratoire Kastler Brossel, ENS-Université PSL, CNRS, Paris, France
| | - Hilton B. de Aguiar
- Sorbonne Université, Collège de France, Laboratoire Kastler Brossel, ENS-Université PSL, CNRS, Paris, France
| |
Collapse
|
10
|
Tang F, Yan F, Zhong Y, Li J, Gong H, Li X. Optogenetic Brain-Computer Interfaces. Bioengineering (Basel) 2024; 11:821. [PMID: 39199779 PMCID: PMC11351350 DOI: 10.3390/bioengineering11080821] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Revised: 07/23/2024] [Accepted: 07/24/2024] [Indexed: 09/01/2024] Open
Abstract
The brain-computer interface (BCI) is one of the most powerful tools in neuroscience and generally includes a recording system, a processor system, and a stimulation system. Optogenetics has the advantages of bidirectional regulation, high spatiotemporal resolution, and cell-specific regulation, which expands the application scenarios of BCIs. In recent years, optogenetic BCIs have become widely used in the lab with the development of materials and software. The systems were designed to be more integrated, lightweight, biocompatible, and power efficient, as were the wireless transmission and chip-level embedded BCIs. The software is also constantly improving, with better real-time performance and accuracy and lower power consumption. On the other hand, as a cutting-edge technology spanning multidisciplinary fields including molecular biology, neuroscience, material engineering, and information processing, optogenetic BCIs have great application potential in neural decoding, enhancing brain function, and treating neural diseases. Here, we review the development and application of optogenetic BCIs. In the future, combined with other functional imaging techniques such as near-infrared spectroscopy (fNIRS) and functional magnetic resonance imaging (fMRI), optogenetic BCIs can modulate the function of specific circuits, facilitate neurological rehabilitation, assist perception, establish a brain-to-brain interface, and be applied in wider application scenarios.
Collapse
Affiliation(s)
- Feifang Tang
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics, MoE Key Laboratory for Biomedical Photonics, Huazhong University of Science and Technology, Wuhan 430074, China; (F.T.); (F.Y.); (Y.Z.); (J.L.); (H.G.)
| | - Feiyang Yan
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics, MoE Key Laboratory for Biomedical Photonics, Huazhong University of Science and Technology, Wuhan 430074, China; (F.T.); (F.Y.); (Y.Z.); (J.L.); (H.G.)
| | - Yushan Zhong
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics, MoE Key Laboratory for Biomedical Photonics, Huazhong University of Science and Technology, Wuhan 430074, China; (F.T.); (F.Y.); (Y.Z.); (J.L.); (H.G.)
| | - Jinqian Li
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics, MoE Key Laboratory for Biomedical Photonics, Huazhong University of Science and Technology, Wuhan 430074, China; (F.T.); (F.Y.); (Y.Z.); (J.L.); (H.G.)
| | - Hui Gong
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics, MoE Key Laboratory for Biomedical Photonics, Huazhong University of Science and Technology, Wuhan 430074, China; (F.T.); (F.Y.); (Y.Z.); (J.L.); (H.G.)
| | - Xiangning Li
- Key Laboratory of Biomedical Engineering of Hainan Province, School of Biomedical Engineering, Hainan University, Haikou 570228, China
| |
Collapse
|
11
|
Tian W, Jia Q, Lin J, Luo J, He D, Yang J, Guo T, Guo H, Guo Y, Zhang W, Chen F, Ye Y, Liu J, Xu M, Deng C, Cui B, Su D, Wang H, Lu Y, Xiao J, Liu H, Yang J, Hou Z, Wang S. Remote neurostimulation through an endogenous ion channel using a near-infrared light-activatable nanoagonist. SCIENCE ADVANCES 2024; 10:eadn0367. [PMID: 39121219 PMCID: PMC11313869 DOI: 10.1126/sciadv.adn0367] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Accepted: 07/02/2024] [Indexed: 08/11/2024]
Abstract
The development of noninvasive approaches to precisely control neural activity in mammals is highly desirable. Here, we used the ion channel transient receptor potential ankyrin-repeat 1 (TRPA1) as a proof of principle, demonstrating remote near-infrared (NIR) activation of endogenous neuronal channels in mice through an engineered nanoagonist. This achievement enables specific neurostimulation in nongenetically modified mice. Initially, target-based screening identified flavins as photopharmacological agonists, allowing for the photoactivation of TRPA1 in sensory neurons upon ultraviolet A/blue light illumination. Subsequently, upconversion nanoparticles (UCNPs) were customized with an emission spectrum aligned to flavin absorption and conjugated with flavin adenine dinucleotide, creating a nanoagonist capable of NIR activation of TRPA1. Following the intrathecal injection of the nanoagonist, noninvasive NIR stimulation allows precise bidirectional control of nociception in mice through remote activation of spinal TRPA1. This study demonstrates a noninvasive NIR neurostimulation method with the potential for adaptation to various endogenous ion channels and neural processes by combining photochemical toolboxes with customized UCNPs.
Collapse
Affiliation(s)
- Weifeng Tian
- The Affiliated TCM Hospital of Guangzhou Medical University, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
- Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
- Institute of Organoid Technology, Kunming Medical University, Kunming, China
- Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
| | - Qi Jia
- Department of Orthopedic Oncology, Shanghai Changzheng Hospital, Second Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Jiewen Lin
- The Affiliated TCM Hospital of Guangzhou Medical University, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Jiamin Luo
- The Affiliated TCM Hospital of Guangzhou Medical University, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Dongmei He
- The Affiliated TCM Hospital of Guangzhou Medical University, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Jie Yang
- The Affiliated TCM Hospital of Guangzhou Medical University, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Tao Guo
- The Affiliated TCM Hospital of Guangzhou Medical University, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Huiling Guo
- The Affiliated TCM Hospital of Guangzhou Medical University, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Yusheng Guo
- GMU-GIBH Joint School of Life Sciences, The Guangdong-Hong Kong-Macao Joint Laboratory for Cell Fate Regulation and Diseases, The Affiliated TCM Hospital of Guangzhou Medical University, State Key Laboratory of Respiratory Disease, GMU-GIBH Joint School of Life Sciences, Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, Guangzhou Medical University, Guangzhou, China
| | - Wenjie Zhang
- The Affiliated TCM Hospital of Guangzhou Medical University, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Feiyu Chen
- The Affiliated TCM Hospital of Guangzhou Medical University, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Ying Ye
- The Affiliated TCM Hospital of Guangzhou Medical University, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Jingjing Liu
- The Affiliated TCM Hospital of Guangzhou Medical University, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Mindong Xu
- The Affiliated TCM Hospital of Guangzhou Medical University, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Chengjie Deng
- Cell Biology and Molecular Biology Laboratory of Experimental Teaching Center, Faculty of Basic Medical Science, Kunming Medical University, Kunming, China
| | - Boxiang Cui
- The Affiliated TCM Hospital of Guangzhou Medical University, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Deyuan Su
- Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
| | - Hao Wang
- Department of Neurobiology and Department of Neurosurgery of Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yi Lu
- Department of Anesthesiology, The Affiliated Traditional Chinese Medicine Hospital, Guangzhou Medical University, Guangzhou, China
| | - Jianru Xiao
- Department of Orthopedic Oncology, Shanghai Changzheng Hospital, Second Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Heng Liu
- GMU-GIBH Joint School of Life Sciences, The Guangdong-Hong Kong-Macao Joint Laboratory for Cell Fate Regulation and Diseases, The Affiliated TCM Hospital of Guangzhou Medical University, State Key Laboratory of Respiratory Disease, GMU-GIBH Joint School of Life Sciences, Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, Guangzhou Medical University, Guangzhou, China
| | - Jian Yang
- Department of Biological Sciences, Columbia University, New York, NY 10027, USA
| | - Zhiyao Hou
- The Affiliated TCM Hospital of Guangzhou Medical University, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Shu Wang
- The Affiliated TCM Hospital of Guangzhou Medical University, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
- Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
12
|
Zhang Q, Li T, Xu M, Islam B, Wang J. Application of Optogenetics in Neurodegenerative Diseases. Cell Mol Neurobiol 2024; 44:57. [PMID: 39060759 PMCID: PMC11281982 DOI: 10.1007/s10571-024-01486-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Accepted: 06/15/2024] [Indexed: 07/28/2024]
Abstract
Optogenetics, a revolutionary technique integrating optical and genetic methodologies, offers unparalleled precision in spatial targeting and temporal resolution for cellular control. This approach enables the selective manipulation of specific neuronal populations, inducing subtle electrical changes that significantly impact complex neural circuitry. As optogenetics precisely targets and modulates neuronal activity, it holds the potential for significant breakthroughs in understanding and potentially altering the course of neurodegenerative diseases, characterized by selective neuronal loss leading to functional deficits within the nervous system. The integration of optogenetics into neurodegenerative disease research has significantly advanced in the field, offering new insights and paving the way for innovative treatment strategies. Its application in clinical settings, although still in the nascent stages, suggests a promising future for addressing some of the most challenging aspects of neurodegenerative disorders. In this review, we provide a comprehensive overview of these research undertakings.
Collapse
Affiliation(s)
- Qian Zhang
- Xiangya School of Public Health, Central South University, Changsha, 410078, Hunan, People's Republic of China
| | - Tianjiao Li
- Xiangya School of Public Health, Central South University, Changsha, 410078, Hunan, People's Republic of China
| | - Mengying Xu
- Xiangya School of Public Health, Central South University, Changsha, 410078, Hunan, People's Republic of China
| | - Binish Islam
- Xiangya School of Public Health, Central South University, Changsha, 410078, Hunan, People's Republic of China
| | - Jianwu Wang
- Xiangya School of Public Health, Central South University, Changsha, 410078, Hunan, People's Republic of China.
| |
Collapse
|
13
|
Hernández-Morales M, Morales-Weil K, Han SM, Han V, Tran T, Benner EJ, Pegram K, Meanor J, Miller EW, Kramer RH, Liu C. Electrophysiological Mechanisms and Validation of Ferritin-Based Magnetogenetics for Remote Control of Neurons. J Neurosci 2024; 44:e1717232024. [PMID: 38777598 PMCID: PMC11270515 DOI: 10.1523/jneurosci.1717-23.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Revised: 05/07/2024] [Accepted: 05/10/2024] [Indexed: 05/25/2024] Open
Abstract
Magnetogenetics was developed to remotely control genetically targeted neurons. A variant of magnetogenetics uses magnetic fields to activate transient receptor potential vanilloid (TRPV) channels when coupled with ferritin. Stimulation with static or RF magnetic fields of neurons expressing these channels induces Ca2+ transients and modulates behavior. However, the validity of ferritin-based magnetogenetics has been questioned due to controversies surrounding the underlying mechanisms and deficits in reproducibility. Here, we validated the magnetogenetic approach Ferritin-iron Redistribution to Ion Channels (FeRIC) using electrophysiological (Ephys) and imaging techniques. Previously, interference from RF stimulation rendered patch-clamp recordings inaccessible for magnetogenetics. We solved this limitation for FeRIC, and we studied the bioelectrical properties of neurons expressing TRPV4 (nonselective cation channel) and transmembrane member 16A (TMEM16A; chloride-permeable channel) coupled to ferritin (FeRIC channels) under RF stimulation. We used cultured neurons obtained from the rat hippocampus of either sex. We show that RF decreases the membrane resistance (Rm) and depolarizes the membrane potential in neurons expressing TRPV4FeRIC RF does not directly trigger action potential firing but increases the neuronal basal spiking frequency. In neurons expressing TMEM16AFeRIC, RF decreases the Rm, hyperpolarizes the membrane potential, and decreases the spiking frequency. Additionally, we corroborated the previously described biochemical mechanism responsible for RF-induced activation of ferritin-coupled ion channels. We solved an enduring problem for ferritin-based magnetogenetics, obtaining direct Ephys evidence of RF-induced activation of ferritin-coupled ion channels. We found that RF does not yield instantaneous changes in neuronal membrane potentials. Instead, RF produces responses that are long-lasting and moderate, but effective in controlling the bioelectrical properties of neurons.
Collapse
Affiliation(s)
- Miriam Hernández-Morales
- Department of Electrical Engineering and Computer Sciences, University of California, Berkeley, California 94720
- Helen Wills Neuroscience Institute, University of California, Berkeley, California 94720
| | - Koyam Morales-Weil
- Department of Electrical Engineering and Computer Sciences, University of California, Berkeley, California 94720
- Helen Wills Neuroscience Institute, University of California, Berkeley, California 94720
| | - Sang Min Han
- Department of Electrical Engineering and Computer Sciences, University of California, Berkeley, California 94720
- Helen Wills Neuroscience Institute, University of California, Berkeley, California 94720
| | - Victor Han
- Department of Electrical Engineering and Computer Sciences, University of California, Berkeley, California 94720
- Helen Wills Neuroscience Institute, University of California, Berkeley, California 94720
| | - Tiffany Tran
- Helen Wills Neuroscience Institute, University of California, Berkeley, California 94720
| | - Eric J Benner
- Division of Neonatology, Department of Pediatrics, Duke University Medical Center, Jean and George Brumley, Jr. Neonatal-Perinatal Institute, Durham, North Carolina 27710
| | - Kelly Pegram
- Division of Neonatology, Department of Pediatrics, Duke University Medical Center, Jean and George Brumley, Jr. Neonatal-Perinatal Institute, Durham, North Carolina 27710
| | - Jenna Meanor
- Division of Neonatology, Department of Pediatrics, Duke University Medical Center, Jean and George Brumley, Jr. Neonatal-Perinatal Institute, Durham, North Carolina 27710
| | - Evan W Miller
- Helen Wills Neuroscience Institute, University of California, Berkeley, California 94720
- Department of Chemistry, University of California, Berkeley, California 94720
- Department of Molecular and Cell Biology, University of California, Berkeley, California 94720
| | - Richard H Kramer
- Department of Molecular and Cell Biology, University of California, Berkeley, California 94720
| | - Chunlei Liu
- Department of Electrical Engineering and Computer Sciences, University of California, Berkeley, California 94720
- Helen Wills Neuroscience Institute, University of California, Berkeley, California 94720
| |
Collapse
|
14
|
Kim H, Jung SO, Lee S, Lee Y. Bioluminescent Systems for Theranostic Applications. Int J Mol Sci 2024; 25:7563. [PMID: 39062805 PMCID: PMC11277111 DOI: 10.3390/ijms25147563] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 07/03/2024] [Accepted: 07/08/2024] [Indexed: 07/28/2024] Open
Abstract
Bioluminescence, the light produced by biochemical reactions involving luciferases in living organisms, has been extensively investigated for various applications. It has attracted particular interest as an internal light source for theranostic applications due to its safe and efficient characteristics that overcome the limited penetration of conventional external light sources. Recent advancements in protein engineering technologies and protein delivery platforms have expanded the application of bioluminescence to a wide range of theranostic areas, including bioimaging, biosensing, photodynamic therapy, and optogenetics. This comprehensive review presents the fundamental concepts of bioluminescence and explores its recent applications across diverse fields. Moreover, it discusses future research directions based on the current status of bioluminescent systems for further expansion of their potential.
Collapse
Affiliation(s)
- Hyemin Kim
- Department of Cosmetics Engineering, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea; (S.O.J.); (S.L.); (Y.L.)
| | | | | | | |
Collapse
|
15
|
Lamm GHU, Zabelskii D, Balandin T, Gordeliy V, Wachtveitl J. Calcium-Sensitive Microbial Rhodopsin VirChR1: A Femtosecond to Second Photocycle Study. J Phys Chem Lett 2024; 15:5510-5516. [PMID: 38749015 DOI: 10.1021/acs.jpclett.4c00693] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/24/2024]
Abstract
Viral rhodopsins are light-gated cation channels representing a novel class of microbial rhodopsins. For viral rhodopsin 1 subfamily members VirChR1 and OLPVR1, channel activity is abolished above a certain calcium concentration. Here we present a calcium-dependent spectroscopic analysis of VirChR1 on the femtosecond to second time scale. Unlike channelrhodopsin-2, VirChR1 possesses two intermediate states P1 and P2 on the ultrafast time scale, similar to J and K in ion-pumping rhodopsins. Subsequently, we observe multifaceted photocycle kinetics with up to seven intermediate states. Calcium predominantly affects the last photocycle steps, including the appearance of additional intermediates P6Ca and P7 representing the blocked channel. Furthermore, the photocycle of the counterion variant D80N is drastically altered, yielding intermediates with different spectra and kinetics compared to those of the wt. These findings demonstrate the central role of the counterion within the defined reaction sequence of microbial rhodopsins that ultimately defines the protein function.
Collapse
Affiliation(s)
- Gerrit H U Lamm
- Institute of Physical and Theoretical Chemistry, Goethe University Frankfurt, 60438 Frankfurt am Main, Germany
| | | | - Taras Balandin
- Institute of Biological Information Processing (IBI-7: Structural Biochemistry), Forschungszentrum Jülich, 52428 Jülich, Germany
- JuStruct: Jülich Center for Structural Biology, Forschungszentrum Jülich, 52428 Jülich, Germany
| | - Valentin Gordeliy
- Institute of Biological Information Processing (IBI-7: Structural Biochemistry), Forschungszentrum Jülich, 52428 Jülich, Germany
- JuStruct: Jülich Center for Structural Biology, Forschungszentrum Jülich, 52428 Jülich, Germany
- University Grenoble Alpes, CEA, CNRS, Institute de Biologie Structurale (IBS), 38000 Grenoble, France
| | - Josef Wachtveitl
- Institute of Physical and Theoretical Chemistry, Goethe University Frankfurt, 60438 Frankfurt am Main, Germany
| |
Collapse
|
16
|
Terada Y, Toyoizumi T. Chaotic neural dynamics facilitate probabilistic computations through sampling. Proc Natl Acad Sci U S A 2024; 121:e2312992121. [PMID: 38648479 PMCID: PMC11067032 DOI: 10.1073/pnas.2312992121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Accepted: 02/13/2024] [Indexed: 04/25/2024] Open
Abstract
Cortical neurons exhibit highly variable responses over trials and time. Theoretical works posit that this variability arises potentially from chaotic network dynamics of recurrently connected neurons. Here, we demonstrate that chaotic neural dynamics, formed through synaptic learning, allow networks to perform sensory cue integration in a sampling-based implementation. We show that the emergent chaotic dynamics provide neural substrates for generating samples not only of a static variable but also of a dynamical trajectory, where generic recurrent networks acquire these abilities with a biologically plausible learning rule through trial and error. Furthermore, the networks generalize their experience in the stimulus-evoked samples to the inference without partial or all sensory information, which suggests a computational role of spontaneous activity as a representation of the priors as well as a tractable biological computation for marginal distributions. These findings suggest that chaotic neural dynamics may serve for the brain function as a Bayesian generative model.
Collapse
Affiliation(s)
- Yu Terada
- Laboratory for Neural Computation and Adaptation, RIKEN Center for Brain Science, Saitama351-0198, Japan
- Department of Neurobiology, University of California, San Diego, La Jolla, CA92093
- The Institute for Physics of Intelligence, The University of Tokyo, Tokyo113-0033, Japan
| | - Taro Toyoizumi
- Laboratory for Neural Computation and Adaptation, RIKEN Center for Brain Science, Saitama351-0198, Japan
- Department of Mathematical Informatics, Graduate School of Information Science and Technology, The University of Tokyo, Tokyo113-8656, Japan
| |
Collapse
|
17
|
Ghanem M, Espinosa C, Chung P, Reincke M, Harrison N, Phongpreecha T, Shome S, Saarunya G, Berson E, James T, Xie F, Shu CH, Hazra D, Mataraso S, Kim Y, Seong D, Chakraborty D, Studer M, Xue L, Marić I, Chang AL, Tjoa E, Gaudillière B, Tawfik VL, Mackey S, Aghaeepour N. Comprehensive overview of the anesthesiology research landscape: A machine Learning Analysis of 737 NIH-funded anesthesiology primary Investigator's publication trends. Heliyon 2024; 10:e29050. [PMID: 38623206 PMCID: PMC11016610 DOI: 10.1016/j.heliyon.2024.e29050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 02/24/2024] [Accepted: 03/28/2024] [Indexed: 04/17/2024] Open
Abstract
Background Anesthesiology plays a crucial role in perioperative care, critical care, and pain management, impacting patient experiences and clinical outcomes. However, our understanding of the anesthesiology research landscape is limited. Accordingly, we initiated a data-driven analysis through topic modeling to uncover research trends, enabling informed decision-making and fostering progress within the field. Methods The easyPubMed R package was used to collect 32,300 PubMed abstracts spanning from 2000 to 2022. These abstracts were authored by 737 Anesthesiology Principal Investigators (PIs) who were recipients of National Institute of Health (NIH) funding from 2010 to 2022. Abstracts were preprocessed, vectorized, and analyzed with the state-of-the-art BERTopic algorithm to identify pillar topics and trending subtopics within anesthesiology research. Temporal trends were assessed using the Mann-Kendall test. Results The publishing journals with most abstracts in this dataset were Anesthesia & Analgesia 1133, Anesthesiology 992, and Pain 671. Eight pillar topics were identified and categorized as basic or clinical sciences based on a hierarchical clustering analysis. Amongst the pillar topics, "Cells & Proteomics" had both the highest annual and total number of abstracts. Interestingly, there was an overall upward trend for all topics spanning the years 2000-2022. However, when focusing on the period from 2015 to 2022, topics "Cells & Proteomics" and "Pulmonology" exhibit a downward trajectory. Additionally, various subtopics were identified, with notable increasing trends in "Aneurysms", "Covid 19 Pandemic", and "Artificial intelligence & Machine Learning". Conclusion Our work offers a comprehensive analysis of the anesthesiology research landscape by providing insights into pillar topics, and trending subtopics. These findings contribute to a better understanding of anesthesiology research and can guide future directions.
Collapse
Affiliation(s)
- Marc Ghanem
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Camilo Espinosa
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Immunology Program, Stanford University School of Medicine, Stanford, CA, USA
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
- Department of Biomedical Data Science, Stanford University, Stanford, CA, USA
| | - Philip Chung
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Momsen Reincke
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Natasha Harrison
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
- Department of Biomedical Data Science, Stanford University, Stanford, CA, USA
| | - Thanaphong Phongpreecha
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Department of Biomedical Data Science, Stanford University, Stanford, CA, USA
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Sayane Shome
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
- Department of Biomedical Data Science, Stanford University, Stanford, CA, USA
| | - Geetha Saarunya
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
- Department of Biomedical Data Science, Stanford University, Stanford, CA, USA
| | - Eloise Berson
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Department of Biomedical Data Science, Stanford University, Stanford, CA, USA
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Tomin James
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Feng Xie
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
- Department of Biomedical Data Science, Stanford University, Stanford, CA, USA
| | - Chi-Hung Shu
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Debapriya Hazra
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
- Department of Biomedical Data Science, Stanford University, Stanford, CA, USA
| | - Samson Mataraso
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
- Department of Biomedical Data Science, Stanford University, Stanford, CA, USA
| | - Yeasul Kim
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
- Department of Biomedical Data Science, Stanford University, Stanford, CA, USA
| | - David Seong
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Immunology Program, Stanford University School of Medicine, Stanford, CA, USA
- Medical Scientist Training Program, Stanford University School of Medicine, Stanford, CA, USA
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA, USA
| | - Dipro Chakraborty
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
- Department of Biomedical Data Science, Stanford University, Stanford, CA, USA
| | - Manuel Studer
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Lei Xue
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
- Department of Biomedical Data Science, Stanford University, Stanford, CA, USA
| | - Ivana Marić
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
| | - Alan L. Chang
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
- Department of Biomedical Data Science, Stanford University, Stanford, CA, USA
| | - Erico Tjoa
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Brice Gaudillière
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Vivianne L. Tawfik
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Sean Mackey
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Nima Aghaeepour
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
- Department of Biomedical Data Science, Stanford University, Stanford, CA, USA
| |
Collapse
|
18
|
Shibukawa A, Higuchi R, Song G, Mikami H, Sudo Y, Jang M. Large-volume focus control at 10 MHz refresh rate via fast line-scanning amplitude-encoded scattering-assisted holography. Nat Commun 2024; 15:2926. [PMID: 38589389 PMCID: PMC11001868 DOI: 10.1038/s41467-024-47009-w] [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: 05/30/2023] [Accepted: 03/11/2024] [Indexed: 04/10/2024] Open
Abstract
The capability of focus control has been central to optical technologies that require both high temporal and spatial resolutions. However, existing varifocal lens schemes are commonly limited to the response time on the microsecond timescale and share the fundamental trade-off between the response time and the tuning power. Here, we propose an ultrafast holographic focusing method enabled by translating the speed of a fast 1D beam scanner into the speed of the complex wavefront modulation of a relatively slow 2D spatial light modulator. Using a pair of a digital micromirror device and a resonant scanner, we demonstrate an unprecedented refresh rate of focus control of 31 MHz, which is more than 1,000 times faster than the switching rate of a digital micromirror device. We also show that multiple micrometer-sized focal spots can be independently addressed in a range of over 1 MHz within a large volume of 5 mm × 5 mm × 5.5 mm, validating the superior spatiotemporal characteristics of the proposed technique - high temporal and spatial precision, high tuning power, and random accessibility in a three-dimensional space. The demonstrated scheme offers a new route towards three-dimensional light manipulation in the 100 MHz regime.
Collapse
Affiliation(s)
- Atsushi Shibukawa
- Research Institute for Electronic Science, Hokkaido University, Sapporo, 001-0020, Japan
| | - Ryota Higuchi
- Research Institute for Electronic Science, Hokkaido University, Sapporo, 001-0020, Japan
| | - Gookho Song
- Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology, Daejeon, 34141, Republic of Korea
| | - Hideharu Mikami
- Research Institute for Electronic Science, Hokkaido University, Sapporo, 001-0020, Japan.
| | - Yuki Sudo
- Faculty of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, 700-8530, Japan.
| | - Mooseok Jang
- Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology, Daejeon, 34141, Republic of Korea.
| |
Collapse
|
19
|
Yu H, Song L, Duan X, Zhu D, Li N, Pan R, Xu R, Yu X, Ye F, Jiang X, Ye H, Pan Z, Wei S, Jiang Z. Optogenetics in taste research: A decade of enlightenment. Oral Dis 2024; 30:903-913. [PMID: 36620868 DOI: 10.1111/odi.14498] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Revised: 12/03/2022] [Accepted: 01/05/2023] [Indexed: 01/10/2023]
Abstract
The electrophysiological function of the tongue involves complicated activities in taste sense, producing the perceptions of salty, sweet, bitter, and sour. However, therapies and prevention of taste loss arising from dysfunction in electrophysiological activity require further fundamental research. Optogenetics has revolutionized neuroscience and brought the study of sensory system to a higher level in taste. The year 2022 marks a decade of developments of optogenetics in taste since this technology was adopted from neuroscience and applied to the taste research. This review summarizes a decade of advances that define near-term translation with optogenetic tools, and newly-discovered mechanisms with the applications of these tools. The main limitations and opportunities for optogenetics in taste research are also discussed.
Collapse
Affiliation(s)
- Hanshu Yu
- Zhejiang University School of Medicine, Hangzhou, China
| | - Luyao Song
- Zhejiang University School of Medicine, Hangzhou, China
| | - Xiangyao Duan
- Zhejiang University School of Medicine, Hangzhou, China
| | - Danji Zhu
- Key Laboratory of Oral Biomedical Research of Zhejiang Province, School of Stomatology, Zhejiang Provincial Clinical Research Centre for Oral Diseases, Cancer Centre of Zhejiang University, Stomatology Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Na Li
- Key Laboratory of Oral Biomedical Research of Zhejiang Province, School of Stomatology, Zhejiang Provincial Clinical Research Centre for Oral Diseases, Cancer Centre of Zhejiang University, Stomatology Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Runxin Pan
- Zhejiang University School of Medicine, Hangzhou, China
| | - Rui Xu
- Zhejiang University School of Medicine, Hangzhou, China
| | - Xinying Yu
- Zhejiang University School of Medicine, Hangzhou, China
| | - Fengkai Ye
- Zhejiang University School of Medicine, Hangzhou, China
| | - Xinrui Jiang
- Zhejiang University School of Medicine, Hangzhou, China
| | - Han Ye
- Zhejiang University School of Medicine, Hangzhou, China
| | - Zikang Pan
- Zhejiang University School of Medicine, Hangzhou, China
| | - Sixing Wei
- Zhejiang University School of Medicine, Hangzhou, China
| | - Zhiwei Jiang
- Key Laboratory of Oral Biomedical Research of Zhejiang Province, School of Stomatology, Zhejiang Provincial Clinical Research Centre for Oral Diseases, Cancer Centre of Zhejiang University, Stomatology Hospital, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
20
|
Xia F, Rimoli CV, Akemann W, Ventalon C, Bourdieu L, Gigan S, de Aguiar HB. Neurophotonics beyond the Surface: Unmasking the Brain's Complexity Exploiting Optical Scattering. ARXIV 2024:arXiv:2403.14809v1. [PMID: 38562443 PMCID: PMC10984001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
The intricate nature of the brain necessitates the application of advanced probing techniques to comprehensively study and understand its working mechanisms. Neurophotonics offers minimally invasive methods to probe the brain using optics at cellular and even molecular levels. However, multiple challenges persist, especially concerning imaging depth, field of view, speed, and biocompatibility. A major hindrance to solving these challenges in optics is the scattering nature of the brain. This perspective highlights the potential of complex media optics, a specialized area of study focused on light propagation in materials with intricate heterogeneous optical properties, in advancing and improving neuronal readouts for structural imaging and optical recordings of neuronal activity. Key strategies include wavefront shaping techniques and computational imaging and sensing techniques that exploit scattering properties for enhanced performance. We discuss the potential merger of the two fields as well as potential challenges and perspectives toward longer term in vivo applications.
Collapse
Affiliation(s)
- Fei Xia
- Laboratoire Kastler Brossel, ENS-Université PSL, CNRS, Sorbonne Université, Collège de France, 24 rue Lhomond, 75005 Paris, France
| | - Caio Vaz Rimoli
- Laboratoire Kastler Brossel, ENS-Université PSL, CNRS, Sorbonne Université, Collège de France, 24 rue Lhomond, 75005 Paris, France
- Institut de Biologie de l'ENS (IBENS), École Normale Supérieure, CNRS, INSERM, Université PSL, Paris, France
| | - Walther Akemann
- Institut de Biologie de l'ENS (IBENS), École Normale Supérieure, CNRS, INSERM, Université PSL, Paris, France
| | - Cathie Ventalon
- Institut de Biologie de l'ENS (IBENS), École Normale Supérieure, CNRS, INSERM, Université PSL, Paris, France
| | - Laurent Bourdieu
- Institut de Biologie de l'ENS (IBENS), École Normale Supérieure, CNRS, INSERM, Université PSL, Paris, France
| | - Sylvain Gigan
- Laboratoire Kastler Brossel, ENS-Université PSL, CNRS, Sorbonne Université, Collège de France, 24 rue Lhomond, 75005 Paris, France
| | - Hilton B de Aguiar
- Laboratoire Kastler Brossel, ENS-Université PSL, CNRS, Sorbonne Université, Collège de France, 24 rue Lhomond, 75005 Paris, France
| |
Collapse
|
21
|
Mulholland HN, Jayakumar H, Farinella DM, Smith GB. All-optical interrogation of millimeter-scale networks and application to developing ferret cortex. J Neurosci Methods 2024; 403:110051. [PMID: 38145718 PMCID: PMC10872452 DOI: 10.1016/j.jneumeth.2023.110051] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Revised: 12/13/2023] [Accepted: 12/21/2023] [Indexed: 12/27/2023]
Abstract
BACKGROUND Perception and behavior require coordinated activity of thousands of neurons operating in networks that span millimeters of brain area. In vivo calcium imaging approaches have proven exceptionally powerful for examining the structure of these networks at large scales, and optogenetics can allow for causal manipulations of large populations of neurons. However, realizing the full potential of these techniques requires the ability to simultaneously measure and manipulate distinct circuit elements on the scale of millimeters. NEW METHOD We describe an opto-macroscope, an artifact-free, all-optical system capable of delivering patterned optogenetic stimulation with high spatial and temporal resolution across millimeters of brain while simultaneously imaging functional neural activity. RESULTS We find that this approach provides direct manipulation of cortical regions ranging from hundreds of microns to several millimeters in area, allowing for the perturbation of individual brain areas or networks of functional domains. Using this system we find that spatially complex endogenous networks in the developing ferret visual cortex can be readily reactivated by precisely designed patterned optogenetic stimuli. COMPARISON WITH EXISTING METHODS Our opto-macroscope extends current all-optical optogenetic approaches which operate on a cellular scale with multiphoton stimulation, and are poorly suited to investigate the millimeter-scale of many functional networks. It also builds upon other mesoscopic optogenetic techniques that lack simultaneous optical readouts of neural activity. CONCLUSIONS The large-scale all-optical capabilities of our system make it a powerful new tool for investigating the contribution of cortical domains and brain areas to the functional neural networks that underlie perception and behavior.
Collapse
Affiliation(s)
- Haleigh N Mulholland
- Optical Imaging and Brain Sciences Medical Discovery Team, Department of Neuroscience, University of Minnesota, 2021 6th Street SE, Minneapolis, MN 55455, USA
| | - Harishankar Jayakumar
- Optical Imaging and Brain Sciences Medical Discovery Team, Department of Neuroscience, University of Minnesota, 2021 6th Street SE, Minneapolis, MN 55455, USA
| | - Deano M Farinella
- Optical Imaging and Brain Sciences Medical Discovery Team, Department of Neuroscience, University of Minnesota, 2021 6th Street SE, Minneapolis, MN 55455, USA
| | - Gordon B Smith
- Optical Imaging and Brain Sciences Medical Discovery Team, Department of Neuroscience, University of Minnesota, 2021 6th Street SE, Minneapolis, MN 55455, USA.
| |
Collapse
|
22
|
Lin Q, Li Z, Wang B, Zhou M, Xie Y, Wang D, Hou C, Wang R, Liu X, Sun X, Shan H, Chen Z, Wu H, Yang Y, Fei C, Chen Z. Acoustic hologram-induced virtual in vivo enhanced waveguide (AH-VIEW). SCIENCE ADVANCES 2024; 10:eadl2232. [PMID: 38354252 DOI: 10.1126/sciadv.adl2232] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Accepted: 01/12/2024] [Indexed: 02/16/2024]
Abstract
Optical imaging and phototherapy in deep tissues face notable challenges due to light scattering. We use encoded acoustic holograms to generate three-dimensional acoustic fields within the target medium, enabling instantaneous and robust modulation of the volumetric refractive index, thereby noninvasively controlling the trajectory of light. Through this approach, we achieved a remarkable 24.3% increase in tissue heating rate in vitro photothermal effect tests on porcine skin. In vivo photoacoustic imaging of mouse brain vasculature exhibits an improved signal-to-noise ratio through the intact scalp and skull. These findings demonstrate that our strategy can effectively suppress light scattering in complex biological tissues by inducing low-angle scattering, achieving an effective depth reaching the millimeter scale. The versatility of this strategy extends its potential applications to neuroscience, lithography, and additive manufacturing.
Collapse
Affiliation(s)
- Qibo Lin
- State Key Laboratory of Precision Manufacturing for Extreme Service Performance, College of Mechanical and Electrical Engineering, Central South University, Changsha 410083, China
| | - Zhaoxi Li
- School of Microelectronics, Xidian University, Xi'an 710071, China
| | - Bo Wang
- Department of Biomedical Engineering, School of Basic Medical Science, Central South University, Changsha 410083, China
| | - Mengqing Zhou
- School of Microelectronics, Xidian University, Xi'an 710071, China
| | - Yang Xie
- State Key Laboratory of Precision Manufacturing for Extreme Service Performance, College of Mechanical and Electrical Engineering, Central South University, Changsha 410083, China
| | - Danfeng Wang
- State Key Laboratory of Precision Manufacturing for Extreme Service Performance, College of Mechanical and Electrical Engineering, Central South University, Changsha 410083, China
| | - Chenxue Hou
- School of Microelectronics, Xidian University, Xi'an 710071, China
| | - Runyu Wang
- State Key Laboratory of Precision Manufacturing for Extreme Service Performance, College of Mechanical and Electrical Engineering, Central South University, Changsha 410083, China
| | - Xiangdong Liu
- State Key Laboratory of Precision Manufacturing for Extreme Service Performance, College of Mechanical and Electrical Engineering, Central South University, Changsha 410083, China
| | - Xin Sun
- State Key Laboratory of Precision Manufacturing for Extreme Service Performance, College of Mechanical and Electrical Engineering, Central South University, Changsha 410083, China
| | - Han Shan
- State Key Laboratory of Precision Manufacturing for Extreme Service Performance, College of Mechanical and Electrical Engineering, Central South University, Changsha 410083, China
| | - Ziyan Chen
- State Key Laboratory of Precision Manufacturing for Extreme Service Performance, College of Mechanical and Electrical Engineering, Central South University, Changsha 410083, China
| | - Huayi Wu
- State Key Laboratory of Precision Manufacturing for Extreme Service Performance, College of Mechanical and Electrical Engineering, Central South University, Changsha 410083, China
| | - Yintang Yang
- School of Microelectronics, Xidian University, Xi'an 710071, China
| | - Chunlong Fei
- School of Microelectronics, Xidian University, Xi'an 710071, China
| | - Zeyu Chen
- State Key Laboratory of Precision Manufacturing for Extreme Service Performance, College of Mechanical and Electrical Engineering, Central South University, Changsha 410083, China
- Furong Laboratory (Precision Medicine), Changsha 410008, China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Xiangya Hospital, Central South University, Changsha 410008, China
- Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Central South University, Changsha 410008, China
| |
Collapse
|
23
|
Dong B, Everly RM, Mahapatra S, Carlsen MS, Ma S, Zhang C. Unleashing Precision and Freedom in Optical Manipulation: Software-Assisted Real-Time Precision Opto-Control of Intracellular Molecular Activities and Cell Functions. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.09.579709. [PMID: 38405826 PMCID: PMC10888777 DOI: 10.1101/2024.02.09.579709] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/27/2024]
Abstract
The traditional method in biological science to regulate cell functions often employs chemical interventions, which commonly lack precision in space and time. While optical manipulation offers superior spatial precision, existing technologies are constrained by limitations in flexibility, accuracy, and response time. Here, we present an adaptable and interactive optical manipulation platform that integrates laser scanning, chemical sensing, synchronized multi-laser control, adaptable target selection, flexible decision-making, and real-time monitoring of sample responses. This software-assisted real-time precision opto-control (S-RPOC) platform facilitates automatic target selection driven by optical signals while permitting user-defined manual delineation. It allows the treatment of mobile or stationary targets with varying laser dosages and wavelengths simultaneously at diffraction-limited spatial precision and optimal accuracy. Significantly, S-RPOC showcases versatile capabilities including adaptive photobleaching, comprehensive quantification of protein dynamics, selective organelle perturbation, control of cell division, and manipulation of individual cell behaviors within a population. With its unprecedented spatiotemporal precision and adaptable decision-making, S-RPOC holds the potential for extensive applications in biological science.
Collapse
|
24
|
Altahini S, Arnoux I, Stroh A. Optogenetics 2.0: challenges and solutions towards a quantitative probing of neural circuits. Biol Chem 2024; 405:43-54. [PMID: 37650383 DOI: 10.1515/hsz-2023-0194] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Accepted: 08/02/2023] [Indexed: 09/01/2023]
Abstract
To exploit the full potential of optogenetics, we need to titrate and tailor optogenetic methods to emulate naturalistic circuit function. For that, the following prerequisites need to be met: first, we need to target opsin expression not only to genetically defined neurons per se, but to specifically target a functional node. Second, we need to assess the scope of optogenetic modulation, i.e. the fraction of optogenetically modulated neurons. Third, we need to integrate optogenetic control in a closed loop setting. Fourth, we need to further safe and stable gene expression and light delivery to bring optogenetics to the clinics. Here, we review these concepts for the human and rodent brain.
Collapse
Affiliation(s)
- Saleh Altahini
- Leibniz Institute for Resilience Research, D-55122 Mainz, Germany
| | - Isabelle Arnoux
- Cerebral Physiopathology Laboratory, Center for Interdisciplinary Research in Biology, College de France, Centre national de la recherche scientifique, Institut national de la santé et de la recherche médicale, Université PSL, F-75005 Paris, France
| | - Albrecht Stroh
- Leibniz Institute for Resilience Research, D-55122 Mainz, Germany
- Institute of Pathophysiology, University Medical Center Mainz, D-55128 Mainz, Germany
| |
Collapse
|
25
|
Kumar A, Jain H, Paul A, Thakur S, Biswas SK. Regularized cost function in wavefront shaping for advancing the contrast of structured light. APPLIED OPTICS 2024; 63:595-603. [PMID: 38294369 DOI: 10.1364/ao.506920] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Accepted: 12/15/2023] [Indexed: 02/01/2024]
Abstract
The cost function in the iterative optimization algorithms is one of the sensitive optimization controllers that plays a crucial role in feedback based wavefront shaping for constructing well-resolved complex structured light through scattering media. There has been a trade-off between resolution and the contrast enhancement of the structured light in wavefront shaping. We have developed an ℓ 2-norm based quadratic cost function (L2QN) and proposed a regularized cost function (RCF) for advancing the contrast and maintaining the high resolution of structured light. Both the simulations and experiments have been performed, and it has been found that the proposed RCF significantly advances the contrast and structural uniformity for focusing light through scattering media as well as for diffused reflection mode. The potential applications of the method demonstrated in this study can be extended into holographic displays, structured light illumination microscopy, photo-lithography, photothermal treatments, dosimetry, laser materials processing, and energy control inside and outside an incubation system.
Collapse
|
26
|
Song L, Wang H, Peng R. Advances in the Regulation of Neural Function by Infrared Light. Int J Mol Sci 2024; 25:928. [PMID: 38256001 PMCID: PMC10815576 DOI: 10.3390/ijms25020928] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 01/02/2024] [Accepted: 01/10/2024] [Indexed: 01/24/2024] Open
Abstract
In recent years, with the rapid development of optical technology, infrared light has been increasingly used in biomedical fields. Research has shown that infrared light could play roles in light stimulation and biological regulation. Infrared light has been used to regulate neural function due to its high spatial resolution, safety and neural sensitivity and has been considered a useful method to replace traditional neural regulation approaches. Infrared neuromodulation methods have been used for neural activation, central nervous system disorder treatment and cognitive enhancement. Research on the regulation of neural function by infrared light stimulation began only recently, and the underlying mechanism remains unclear. This article reviews the characteristics of infrared light, the advantages and disadvantages of infrared neuromodulation, its effects on improving individual health, and its mechanism. This article aims to provide a reference for future research on the use of infrared neural regulation to treat neuropsychological disorders.
Collapse
Affiliation(s)
| | - Hui Wang
- Beijing Institute of Radiation Medicine, Beijing 100850, China
| | - Ruiyun Peng
- Beijing Institute of Radiation Medicine, Beijing 100850, China
| |
Collapse
|
27
|
Qi L, Wang F, Sun X, Li H, Zhang K, Li J. Recent advances in tissue repair of the blood-brain barrier after stroke. J Tissue Eng 2024; 15:20417314241226551. [PMID: 38304736 PMCID: PMC10832427 DOI: 10.1177/20417314241226551] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Accepted: 12/31/2023] [Indexed: 02/03/2024] Open
Abstract
The selective permeability of the blood-brain barrier (BBB) enables the necessary exchange of substances between the brain parenchyma and circulating blood and is important for the normal functioning of the central nervous system. Ischemic stroke inflicts damage upon the BBB, triggering adverse stroke outcomes such as cerebral edema, hemorrhagic transformation, and aggravated neuroinflammation. Therefore, effective repair of the damaged BBB after stroke and neovascularization that allows for the unique selective transfer of substances from the BBB after stroke is necessary and important for the recovery of brain function. This review focuses on four important therapies that have effects of BBB tissue repair after stroke in the last seven years. Most of these new therapies show increased expression of BBB tight-junction proteins, and some show beneficial results in terms of enhanced pericyte coverage at the injured vessels. This review also briefly outlines three effective classes of approaches and their mechanisms for promoting neoangiogenesis following a stroke.
Collapse
Affiliation(s)
- Liujie Qi
- School of Material Science and Engineering & Henan Key Laboratory of Advanced Magnesium Alloy & Key Laboratory of Materials Processing and Mold (Ministry of Education), Zhengzhou University, Zhengzhou, PR China
| | - Fei Wang
- School of Material Science and Engineering & Henan Key Laboratory of Advanced Magnesium Alloy & Key Laboratory of Materials Processing and Mold (Ministry of Education), Zhengzhou University, Zhengzhou, PR China
| | - Xiaojing Sun
- School of Material Science and Engineering & Henan Key Laboratory of Advanced Magnesium Alloy & Key Laboratory of Materials Processing and Mold (Ministry of Education), Zhengzhou University, Zhengzhou, PR China
| | - Hang Li
- School of Material Science and Engineering & Henan Key Laboratory of Advanced Magnesium Alloy & Key Laboratory of Materials Processing and Mold (Ministry of Education), Zhengzhou University, Zhengzhou, PR China
| | - Kun Zhang
- School of Life Science, Zhengzhou University, Zhengzhou, PR China
| | - Jingan Li
- School of Material Science and Engineering & Henan Key Laboratory of Advanced Magnesium Alloy & Key Laboratory of Materials Processing and Mold (Ministry of Education), Zhengzhou University, Zhengzhou, PR China
| |
Collapse
|
28
|
Yamakou ME, Desroches M, Rodrigues S. Synchronization in STDP-driven memristive neural networks with time-varying topology. J Biol Phys 2023; 49:483-507. [PMID: 37656327 PMCID: PMC10651826 DOI: 10.1007/s10867-023-09642-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2023] [Accepted: 08/07/2023] [Indexed: 09/02/2023] Open
Abstract
Synchronization is a widespread phenomenon in the brain. Despite numerous studies, the specific parameter configurations of the synaptic network structure and learning rules needed to achieve robust and enduring synchronization in neurons driven by spike-timing-dependent plasticity (STDP) and temporal networks subject to homeostatic structural plasticity (HSP) rules remain unclear. Here, we bridge this gap by determining the configurations required to achieve high and stable degrees of complete synchronization (CS) and phase synchronization (PS) in time-varying small-world and random neural networks driven by STDP and HSP. In particular, we found that decreasing P (which enhances the strengthening effect of STDP on the average synaptic weight) and increasing F (which speeds up the swapping rate of synapses between neurons) always lead to higher and more stable degrees of CS and PS in small-world and random networks, provided that the network parameters such as the synaptic time delay [Formula: see text], the average degree [Formula: see text], and the rewiring probability [Formula: see text] have some appropriate values. When [Formula: see text], [Formula: see text], and [Formula: see text] are not fixed at these appropriate values, the degree and stability of CS and PS may increase or decrease when F increases, depending on the network topology. It is also found that the time delay [Formula: see text] can induce intermittent CS and PS whose occurrence is independent F. Our results could have applications in designing neuromorphic circuits for optimal information processing and transmission via synchronization phenomena.
Collapse
Affiliation(s)
- Marius E Yamakou
- Department of Data Science, Friedrich-Alexander-Universität Erlangen-Nürnberg, Cauerstr. 11, 91058, Erlangen, Germany.
- Max-Planck-Institut für Mathematik in den Naturwissenschaften, Inselstr. 22, 04103, Leipzig, Germany.
| | - Mathieu Desroches
- MathNeuro Project-Team, Inria Center at Université Côte d'Azur, 2004 route des Lucioles - BP 93, 06902, Cedex, Sophia Antipolis, France
| | - Serafim Rodrigues
- Mathematical, Computational and Experimental Neuroscience, Basque Center for Applied Mathematics, Alameda de Mazzaredo 14, 48009, Bilbao, Spain
- Ikerbasque, Basque Foundation for Science, Plaza Euskadi 5, 48009, Bilbao, Spain
| |
Collapse
|
29
|
Telliez C, De Sars V, Emiliani V, Ronzitti E. Descanned fast light targeting (deFLiT) two-photon optogenetics. BIOMEDICAL OPTICS EXPRESS 2023; 14:6222-6232. [PMID: 38420304 PMCID: PMC10898566 DOI: 10.1364/boe.499445] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 09/11/2023] [Accepted: 09/25/2023] [Indexed: 03/02/2024]
Abstract
Two-photon light-targeting optogenetics allows controlling selected subsets of neurons with near single-cell resolution and high temporal precision. To push forward this approach, we recently proposed a fast light-targeting strategy (FLiT) to rapidly scan multiple holograms tiled on a spatial light modulator (SLM). This allowed generating sub-ms timely-controlled switch of light patterns enabling to reduce the power budget for multi-target excitation and increase the temporal precision for relative spike tuning in a circuit. Here, we modified the optical design of FLiT by including a de-scan unit (deFLiT) to keep the holographic illumination centered at the middle of the objective pupil independently of the position of the tiled hologram on the SLM. This enables enlarging the number of usable holograms and reaching extended on-axis excitation volumes, and therefore increasing even further the power gain and temporal precision of conventional FLiT.
Collapse
Affiliation(s)
- Cecile Telliez
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, 75012 Paris, France
| | - Vincent De Sars
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, 75012 Paris, France
| | - Valentina Emiliani
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, 75012 Paris, France
| | - Emiliano Ronzitti
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, 75012 Paris, France
| |
Collapse
|
30
|
Junge S, Ricci Signorini ME, Al Masri M, Gülink J, Brüning H, Kasperek L, Szepes M, Bakar M, Gruh I, Heisterkamp A, Torres-Mapa ML. A micro-LED array based platform for spatio-temporal optogenetic control of various cardiac models. Sci Rep 2023; 13:19490. [PMID: 37945622 PMCID: PMC10636122 DOI: 10.1038/s41598-023-46149-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Accepted: 10/27/2023] [Indexed: 11/12/2023] Open
Abstract
Optogenetics relies on dynamic spatial and temporal control of light to address emerging fundamental and therapeutic questions in cardiac research. In this work, a compact micro-LED array, consisting of 16 × 16 pixels, is incorporated in a widefield fluorescence microscope for controlled light stimulation. We describe the optical design of the system that allows the micro-LED array to fully cover the field of view regardless of the imaging objective used. Various multicellular cardiac models are used in the experiments such as channelrhodopsin-2 expressing aggregates of cardiomyocytes, termed cardiac bodies, and bioartificial cardiac tissues derived from human induced pluripotent stem cells. The pacing efficiencies of the cardiac bodies and bioartificial cardiac tissues were characterized as a function of illumination time, number of switched-on pixels and frequency of stimulation. To demonstrate dynamic stimulation, steering of calcium waves in HL-1 cell monolayer expressing channelrhodopsin-2 was performed by applying different configurations of patterned light. This work shows that micro-LED arrays are powerful light sources for optogenetic control of contraction and calcium waves in cardiac monolayers, multicellular bodies as well as three-dimensional artificial cardiac tissues.
Collapse
Affiliation(s)
- Sebastian Junge
- Institute of Quantum Optics, Gottfried Wilhelm Leibniz University, 30167, Hannover, Germany
- Lower Saxony Centre for Biomedical Engineering, Implant Research and Development (NIFE), 30625, Hannover, Germany
| | - Maria Elena Ricci Signorini
- Department of Cardiac, Thoracic-, Transplantation and Vascular Surgery, Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Hannover Medical School, 30625, Hannover, Germany
| | - Masa Al Masri
- Institute of Quantum Optics, Gottfried Wilhelm Leibniz University, 30167, Hannover, Germany
- Lower Saxony Centre for Biomedical Engineering, Implant Research and Development (NIFE), 30625, Hannover, Germany
| | - Jan Gülink
- QubeDot GmbH, Wilhelmsgarten 3, 38100, Brunswick, Germany
| | - Heiko Brüning
- QubeDot GmbH, Wilhelmsgarten 3, 38100, Brunswick, Germany
| | - Leon Kasperek
- Institute of Quantum Optics, Gottfried Wilhelm Leibniz University, 30167, Hannover, Germany
- Lower Saxony Centre for Biomedical Engineering, Implant Research and Development (NIFE), 30625, Hannover, Germany
| | - Monika Szepes
- Department of Cardiac, Thoracic-, Transplantation and Vascular Surgery, Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Hannover Medical School, 30625, Hannover, Germany
| | - Mine Bakar
- Department of Cardiac, Thoracic-, Transplantation and Vascular Surgery, Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Hannover Medical School, 30625, Hannover, Germany
| | - Ina Gruh
- Department of Cardiac, Thoracic-, Transplantation and Vascular Surgery, Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Hannover Medical School, 30625, Hannover, Germany
| | - Alexander Heisterkamp
- Institute of Quantum Optics, Gottfried Wilhelm Leibniz University, 30167, Hannover, Germany
- Lower Saxony Centre for Biomedical Engineering, Implant Research and Development (NIFE), 30625, Hannover, Germany
| | - Maria Leilani Torres-Mapa
- Institute of Quantum Optics, Gottfried Wilhelm Leibniz University, 30167, Hannover, Germany.
- Lower Saxony Centre for Biomedical Engineering, Implant Research and Development (NIFE), 30625, Hannover, Germany.
| |
Collapse
|
31
|
Gupta P, Rathi P, Gupta R, Baldi H, Coquerel Q, Debnath A, Derami HG, Raman B, Singamaneni S. Neuronal maturation-dependent nano-neuro interaction and modulation. NANOSCALE HORIZONS 2023; 8:1537-1555. [PMID: 37672212 PMCID: PMC10615777 DOI: 10.1039/d3nh00258f] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/07/2023]
Abstract
Nanotechnology-enabled neuromodulation is a promising minimally-invasive tool in neuroscience and engineering for both fundamental studies and clinical applications. However, the nano-neuro interaction at different stages of maturation of a neural network and its implications for the nano-neuromodulation remain unclear. Here, we report heterogeneous to homogeneous transformation of neuromodulation in a progressively maturing neural network. Utilizing plasmonic-fluors as ultrabright fluorescent nanolabels, we reveal that negative surface charge of nanoparticles renders selective nano-neuro interaction with a strong correlation between the maturation stage of the individual neurons in the neural network and the density of the nanoparticles bound on the neurons. In stark contrast to homogeneous neuromodulation in a mature neural network reported so far, the maturation-dependent density of the nanoparticles bound to neurons in a developing neural network resulted in a heterogeneous optical neuromodulation (i.e., simultaneous excitation and inhibition of neural network activity). This study advances our understanding of nano-neuro interactions and nano-neuromodulation with potential applications in minimally-invasive technologies for treating neuronal disorders in parts of the mammalian brain where neurogenesis persists throughout aging.
Collapse
Affiliation(s)
- Prashant Gupta
- Department of Mechanical Engineering and Materials Science, and Institute of Materials Science and Engineering, Washington University in St. Louis, St. Louis, MO, 63130, USA.
| | - Priya Rathi
- Department of Mechanical Engineering and Materials Science, and Institute of Materials Science and Engineering, Washington University in St. Louis, St. Louis, MO, 63130, USA.
| | - Rohit Gupta
- Department of Mechanical Engineering and Materials Science, and Institute of Materials Science and Engineering, Washington University in St. Louis, St. Louis, MO, 63130, USA.
| | - Harsh Baldi
- Department of Mechanical Engineering and Materials Science, and Institute of Materials Science and Engineering, Washington University in St. Louis, St. Louis, MO, 63130, USA.
| | - Quentin Coquerel
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO, 63130, USA.
| | - Avishek Debnath
- Department of Mechanical Engineering and Materials Science, and Institute of Materials Science and Engineering, Washington University in St. Louis, St. Louis, MO, 63130, USA.
| | - Hamed Gholami Derami
- Department of Mechanical Engineering and Materials Science, and Institute of Materials Science and Engineering, Washington University in St. Louis, St. Louis, MO, 63130, USA.
| | - Baranidharan Raman
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO, 63130, USA.
| | - Srikanth Singamaneni
- Department of Mechanical Engineering and Materials Science, and Institute of Materials Science and Engineering, Washington University in St. Louis, St. Louis, MO, 63130, USA.
| |
Collapse
|
32
|
Mitroshina E, Kalinina E, Vedunova M. Optogenetics in Alzheimer's Disease: Focus on Astrocytes. Antioxidants (Basel) 2023; 12:1856. [PMID: 37891935 PMCID: PMC10604138 DOI: 10.3390/antiox12101856] [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: 09/04/2023] [Revised: 09/27/2023] [Accepted: 10/10/2023] [Indexed: 10/29/2023] Open
Abstract
Alzheimer's disease (AD) is the most common form of dementia, resulting in disability and mortality. The global incidence of AD is consistently surging. Although numerous therapeutic agents with promising potential have been developed, none have successfully treated AD to date. Consequently, the pursuit of novel methodologies to address neurodegenerative processes in AD remains a paramount endeavor. A particularly promising avenue in this search is optogenetics, enabling the manipulation of neuronal activity. In recent years, research attention has pivoted from neurons to glial cells. This review aims to consider the potential of the optogenetic correction of astrocyte metabolism as a promising strategy for correcting AD-related disorders. The initial segment of the review centers on the role of astrocytes in the genesis of neurodegeneration. Astrocytes have been implicated in several pathological processes associated with AD, encompassing the clearance of β-amyloid, neuroinflammation, excitotoxicity, oxidative stress, and lipid metabolism (along with a critical role in apolipoprotein E function). The effect of astrocyte-neuronal interactions will also be scrutinized. Furthermore, the review delves into a number of studies indicating that changes in cellular calcium (Ca2+) signaling are one of the causes of neurodegeneration. The review's latter section presents insights into the application of various optogenetic tools to manipulate astrocytic function as a means to counteract neurodegenerative changes.
Collapse
Affiliation(s)
- Elena Mitroshina
- Institute of Biology and Biomedicine, Lobachevsky State University of Nizhny Novgorod, 23 Gagarin Avenue, 603022 Nizhny Novgorod, Russia (M.V.)
| | | | | |
Collapse
|
33
|
Kim S, Moon HS, Vo TT, Kim CH, Im GH, Lee S, Choi M, Kim SG. Whole-brain mapping of effective connectivity by fMRI with cortex-wide patterned optogenetics. Neuron 2023; 111:1732-1747.e6. [PMID: 37001524 DOI: 10.1016/j.neuron.2023.03.002] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Revised: 11/23/2022] [Accepted: 03/02/2023] [Indexed: 04/03/2023]
Abstract
Functional magnetic resonance imaging (fMRI) with optogenetic neural manipulation is a powerful tool that enables brain-wide mapping of effective functional networks. To achieve flexible manipulation of neural excitation throughout the mouse cortex, we incorporated spatiotemporal programmable optogenetic stimuli generated by a digital micromirror device into an MRI scanner via an optical fiber bundle. This approach offered versatility in space and time in planning the photostimulation pattern, combined with in situ optical imaging and cell-type-specific or circuit-specific genetic targeting in individual mice. Brain-wide effective connectivity obtained by fMRI with optogenetic stimulation of atlas-based cortical regions is generally congruent with anatomically defined axonal tracing data but is affected by the types of anesthetics that act selectively on specific connections. fMRI combined with flexible optogenetics opens a new path to investigate dynamic changes in functional brain states in the same animal through high-throughput brain-wide effective connectivity mapping.
Collapse
Affiliation(s)
- Seonghoon Kim
- Center for Neuroscience Imaging Research, Institute for Basic Science, Suwon, Republic of Korea; School of Biological Sciences, Seoul National University, Seoul, Republic of Korea
| | - Hyun Seok Moon
- Center for Neuroscience Imaging Research, Institute for Basic Science, Suwon, Republic of Korea; Department of Biomedical Engineering, Sungkyunkwan University, Suwon, Republic of Korea; Department of Intelligent Precision Healthcare Convergence, Sungkyunkwan University, Suwon, Republic of Korea
| | - Thanh Tan Vo
- Center for Neuroscience Imaging Research, Institute for Basic Science, Suwon, Republic of Korea; Department of Biomedical Engineering, Sungkyunkwan University, Suwon, Republic of Korea; Department of Intelligent Precision Healthcare Convergence, Sungkyunkwan University, Suwon, Republic of Korea
| | - Chang-Ho Kim
- School of Biological Sciences, Seoul National University, Seoul, Republic of Korea; Institute of Molecular Biology and Genetics, Seoul National University, Seoul, Republic of Korea
| | - Geun Ho Im
- Center for Neuroscience Imaging Research, Institute for Basic Science, Suwon, Republic of Korea
| | - Sungho Lee
- School of Biological Sciences, Seoul National University, Seoul, Republic of Korea
| | - Myunghwan Choi
- Center for Neuroscience Imaging Research, Institute for Basic Science, Suwon, Republic of Korea; School of Biological Sciences, Seoul National University, Seoul, Republic of Korea; Institute of Molecular Biology and Genetics, Seoul National University, Seoul, Republic of Korea.
| | - Seong-Gi Kim
- Center for Neuroscience Imaging Research, Institute for Basic Science, Suwon, Republic of Korea; Department of Biomedical Engineering, Sungkyunkwan University, Suwon, Republic of Korea; Department of Intelligent Precision Healthcare Convergence, Sungkyunkwan University, Suwon, Republic of Korea.
| |
Collapse
|
34
|
Losero E, Jagannath S, Pezzoli M, Goblot V, Babashah H, Lashuel HA, Galland C, Quack N. Neuronal growth on high-aspect-ratio diamond nanopillar arrays for biosensing applications. Sci Rep 2023; 13:5909. [PMID: 37041255 PMCID: PMC10090193 DOI: 10.1038/s41598-023-32235-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Accepted: 03/24/2023] [Indexed: 04/13/2023] Open
Abstract
Monitoring neuronal activity with simultaneously high spatial and temporal resolution in living cell cultures is crucial to advance understanding of the development and functioning of our brain, and to gain further insights in the origin of brain disorders. While it has been demonstrated that the quantum sensing capabilities of nitrogen-vacancy (NV) centers in diamond allow real time detection of action potentials from large neurons in marine invertebrates, quantum monitoring of mammalian neurons (presenting much smaller dimensions and thus producing much lower signal and requiring higher spatial resolution) has hitherto remained elusive. In this context, diamond nanostructuring can offer the opportunity to boost the diamond platform sensitivity to the required level. However, a comprehensive analysis of the impact of a nanostructured diamond surface on the neuronal viability and growth was lacking. Here, we pattern a single crystal diamond surface with large-scale nanopillar arrays and we successfully demonstrate growth of a network of living and functional primary mouse hippocampal neurons on it. Our study on geometrical parameters reveals preferential growth along the nanopillar grid axes with excellent physical contact between cell membrane and nanopillar apex. Our results suggest that neuron growth can be tailored on diamond nanopillars to realize a nanophotonic quantum sensing platform for wide-field and label-free neuronal activity recording with sub-cellular resolution.
Collapse
Affiliation(s)
- Elena Losero
- School of Basic Sciences, Institute of Physics, EPFL, Rte Cantonale, 1015, Lausanne, Switzerland.
- Division of Quantum Metrology and Nanotechnologies, Istituto Nazionale di Ricerca Metrologica (INRiM), Strada delle Cacce 91, 10135, Torino, Italy.
- School of Engineering, Institute of Electrical and Micro Engineering, EPFL, Rte Cantonale, 1015, Lausanne, Switzerland.
| | - Somanath Jagannath
- School of Life Sciences, EPFL, Rte Cantonale, 1015, Lausanne, Switzerland
| | - Maurizio Pezzoli
- School of Life Sciences, EPFL, Rte Cantonale, 1015, Lausanne, Switzerland
| | - Valentin Goblot
- School of Basic Sciences, Institute of Physics, EPFL, Rte Cantonale, 1015, Lausanne, Switzerland
| | - Hossein Babashah
- School of Basic Sciences, Institute of Physics, EPFL, Rte Cantonale, 1015, Lausanne, Switzerland
| | - Hilal A Lashuel
- School of Life Sciences, EPFL, Rte Cantonale, 1015, Lausanne, Switzerland
| | - Christophe Galland
- School of Basic Sciences, Institute of Physics, EPFL, Rte Cantonale, 1015, Lausanne, Switzerland
| | - Niels Quack
- School of Aerospace, Mechanical and Mechatronic Engineering, The University of Sydney, Sydney, NSW, Australia
- School of Engineering, Institute of Electrical and Micro Engineering, EPFL, Rte Cantonale, 1015, Lausanne, Switzerland
| |
Collapse
|
35
|
Daw TB, El-Nahal HG, Basso MA, Jun EJ, Bautista AR, Samulski RJ, Sommer MA, Bohlen MO. Direct Comparison of Epifluorescence and Immunostaining for Assessing Viral Mediated Gene Expression in the Primate Brain. Hum Gene Ther 2023; 34:228-246. [PMID: 36719771 PMCID: PMC10031143 DOI: 10.1089/hum.2022.194] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2022] [Accepted: 01/03/2023] [Indexed: 02/01/2023] Open
Abstract
Viral vector technologies are commonly used in neuroscience research to understand and manipulate neural circuits, but successful applications of these technologies in non-human primate models have been inconsistent. An essential component to improve these technologies is an impartial and accurate assessment of the effectiveness of different viral constructs in the primate brain. We tested a diverse array of viral vectors delivered to the brain and extraocular muscles of macaques and compared three methods for histological assessment of viral-mediated fluorescent transgene expression: epifluorescence (Epi), immunofluorescence (IF), and immunohistochemistry (IHC). Importantly, IF and IHC identified a greater number of transduced neurons compared to Epi. Furthermore, IF and IHC reliably provided enhanced visualization of transgene in most cellular compartments (i.e., dendritic, axonal, and terminal fields), whereas the degree of labeling provided by Epi was inconsistent and predominantly restricted to somas and apical dendrites. Because Epi signals are unamplified (in contrast to IF and IHC), Epi may provide a more veridical assessment for the amount of accumulated transgene and, thus, the potential to chemogenetically or optogenetically manipulate neuronal activity. The comparatively weak Epi signals suggest that the current generations of viral constructs, regardless of delivered transgene, are not optimized for primates. This reinforces an emerging viewpoint that viral vectors tailored for the primate brain are necessary for basic research and human gene therapy.
Collapse
Affiliation(s)
- Tierney B. Daw
- Department of Neurobiology, Duke University School of Medicine, Durham, North Carolina, USA
| | - Hala G. El-Nahal
- Department of Biomedical Engineering, Duke University, Durham, North Carolina, USA
| | - Michele A. Basso
- Fuster Laboratory of Cognitive Neuroscience, Department of Psychiatry and Biobehavioral Sciences, Jane and Terry Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles, Los Angeles, California, USA
- Department of Biological Structure, Washington National Primate Research Center, University of Washington, Seattle, Seattle, Washington, USA
- Department of Physiology and Biophysics, Washington National Primate Research Center, University of Washington, Seattle, Seattle, Washington, USA
| | - Elizabeth J. Jun
- Fuster Laboratory of Cognitive Neuroscience, Department of Psychiatry and Biobehavioral Sciences, Jane and Terry Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles, Los Angeles, California, USA
| | - Alex R. Bautista
- Fuster Laboratory of Cognitive Neuroscience, Department of Psychiatry and Biobehavioral Sciences, Jane and Terry Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles, Los Angeles, California, USA
| | - R. Jude Samulski
- Gene Therapy Center and Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
- R&D Department, Asklepios BioPharmaceutical, Inc. (AskBio), Research Triangle, North Carolina, USA
| | - Marc A. Sommer
- Department of Neurobiology, Duke University School of Medicine, Durham, North Carolina, USA
- Department of Biomedical Engineering, Duke University, Durham, North Carolina, USA
- Department of Psychology and Neuroscience, Duke University, Durham, North Carolina, USA
- Center for Cognitive Neuroscience, Duke Institute for Brain Sciences, Duke University, Durham, North Carolina, USA
| | - Martin O. Bohlen
- Department of Biomedical Engineering, Duke University, Durham, North Carolina, USA
| |
Collapse
|
36
|
Wang Y, Garg R, Cohen-Karni D, Cohen-Karni T. Neural modulation with photothermally active nanomaterials. NATURE REVIEWS BIOENGINEERING 2023; 1:193-207. [PMID: 39221032 PMCID: PMC11364367 DOI: 10.1038/s44222-023-00022-y] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 01/02/2023] [Indexed: 09/04/2024]
Abstract
Modulating neural electrophysiology with high precision is essential for understanding neural communication and for the diagnosis and treatment of neural disorders. Photothermal modulation offers a remote and non-genetic method for neural modulation with high spatiotemporal resolution and specificity. This technique induces highly localized and transient temperature changes at the cell membrane interfaced with photothermally active nanomaterials. This rapid temperature change affects the electrical properties of the cell membrane or temperature-sensitive ion channels. In this Review, we discuss the fundamental material properties and illumination conditions that are necessary for nanomaterial-assisted photothermal neural excitation and inhibition. We examine how this versatile technique allows direct investigation of neural electrophysiology and signalling pathways in two-dimensional and three-dimensional cell cultures and tissues, and highlight the scientific and technological challenges in terms of cellular specificity, light delivery and biointerface stability on the road to clinical translation.
Collapse
Affiliation(s)
- Yingqiao Wang
- Department of Materials Science and Engineering, Carnegie Mellon University, Pittsburgh, PA, USA
- These authors contributed equally: Yingqiao Wang, Raghav Garg
| | - Raghav Garg
- Department of Materials Science and Engineering, Carnegie Mellon University, Pittsburgh, PA, USA
- These authors contributed equally: Yingqiao Wang, Raghav Garg
| | - Devora Cohen-Karni
- Preclinical education biochemistry, Lake Erie College of Osteopathic Medicine at Seton Hill, Greensburg, PA, USA
| | - Tzahi Cohen-Karni
- Department of Materials Science and Engineering, Carnegie Mellon University, Pittsburgh, PA, USA
- Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, PA, USA
| |
Collapse
|
37
|
Detection and modulation of neurodegenerative processes using graphene-based nanomaterials: Nanoarchitectonics and applications. Adv Colloid Interface Sci 2023; 311:102824. [PMID: 36549182 DOI: 10.1016/j.cis.2022.102824] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Revised: 12/02/2022] [Accepted: 12/08/2022] [Indexed: 12/14/2022]
Abstract
Neurodegenerative disorders (NDDs) are caused by progressive loss of functional neurons following the aggregation and fibrillation of proteins in the central nervous system. The incidence rate continues to rise alarmingly worldwide, particularly in aged population, and the success of treatment remains limited to symptomatic relief. Graphene nanomaterials (GNs) have attracted immense interest on the account of their unique physicochemical and optoelectronic properties. The research over the past two decades has recognized their ability to interact with aggregation-prone neuronal proteins, regulate autophagy and modulate the electrophysiology of neuronal cells. Graphene can prevent the formation of higher order protein aggregates and facilitate the clearance of such deposits. In this review, after highlighting the role of protein fibrillation in neurodegeneration, we have discussed how GN-protein interactions can be exploited for preventing neurodegeneration. A comprehensive understanding of such interactions would contribute to the exploration of novel modalities for controlling neurodegenerative processes.
Collapse
|
38
|
Luo J, Xue N, Chen J. A Review: Research Progress of Neural Probes for Brain Research and Brain-Computer Interface. BIOSENSORS 2022; 12:bios12121167. [PMID: 36551135 PMCID: PMC9775442 DOI: 10.3390/bios12121167] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Revised: 12/07/2022] [Accepted: 12/13/2022] [Indexed: 06/01/2023]
Abstract
Neural probes, as an invasive physiological tool at the mesoscopic scale, can decipher the code of brain connections and communications from the cellular or even molecular level, and realize information fusion between the human body and external machines. In addition to traditional electrodes, two new types of neural probes have been developed in recent years: optoprobes based on optogenetics and magnetrodes that record neural magnetic signals. In this review, we give a comprehensive overview of these three kinds of neural probes. We firstly discuss the development of microelectrodes and strategies for their flexibility, which is mainly represented by the selection of flexible substrates and new electrode materials. Subsequently, the concept of optogenetics is introduced, followed by the review of several novel structures of optoprobes, which are divided into multifunctional optoprobes integrated with microfluidic channels, artifact-free optoprobes, three-dimensional drivable optoprobes, and flexible optoprobes. At last, we introduce the fundamental perspectives of magnetoresistive (MR) sensors and then review the research progress of magnetrodes based on it.
Collapse
Affiliation(s)
- Jiahui Luo
- State Key Laboratory of Transducer Technology, Aerospace Information Research Institute, Chinese Academy of Sciences, Beijing 100190, China
- School of Electronic, Electrical and Communication Engineering, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Ning Xue
- State Key Laboratory of Transducer Technology, Aerospace Information Research Institute, Chinese Academy of Sciences, Beijing 100190, China
- School of Electronic, Electrical and Communication Engineering, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jiamin Chen
- State Key Laboratory of Transducer Technology, Aerospace Information Research Institute, Chinese Academy of Sciences, Beijing 100190, China
- School of Electronic, Electrical and Communication Engineering, University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
39
|
Feng Z, Ducos B, Scerbo P, Aujard I, Jullien L, Bensimon D. The Development and Application of Opto-Chemical Tools in the Zebrafish. Molecules 2022; 27:6231. [PMID: 36234767 PMCID: PMC9572478 DOI: 10.3390/molecules27196231] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Revised: 09/19/2022] [Accepted: 09/20/2022] [Indexed: 11/18/2022] Open
Abstract
The zebrafish is one of the most widely adopted animal models in both basic and translational research. This popularity of the zebrafish results from several advantages such as a high degree of similarity to the human genome, the ease of genetic and chemical perturbations, external fertilization with high fecundity, transparent and fast-developing embryos, and relatively low cost-effective maintenance. In particular, body translucency is a unique feature of zebrafish that is not adequately obtained with other vertebrate organisms. The animal's distinctive optical clarity and small size therefore make it a successful model for optical modulation and observation. Furthermore, the convenience of microinjection and high embryonic permeability readily allow for efficient delivery of large and small molecules into live animals. Finally, the numerous number of siblings obtained from a single pair of animals offers large replicates and improved statistical analysis of the results. In this review, we describe the development of opto-chemical tools based on various strategies that control biological activities with unprecedented spatiotemporal resolution. We also discuss the reported applications of these tools in zebrafish and highlight the current challenges and future possibilities of opto-chemical approaches, particularly at the single cell level.
Collapse
Affiliation(s)
- Zhiping Feng
- Department of Chemical and Systems Biology, Stanford University, Stanford, CA 94305, USA
| | - Bertrand Ducos
- Laboratoire de Physique de l’Ecole Normale Supérieure, Paris Sciences Letters University, Sorbonne Université, Université de Paris, Centre National de la Recherche Scientifique, 24 Rue Lhomond, 75005 Paris, France
- High Throughput qPCR Core Facility, Ecole Normale Supérieure, Paris Sciences Letters University, 46 Rue d’Ulm, 75005 Paris, France
| | - Pierluigi Scerbo
- Laboratoire de Physique de l’Ecole Normale Supérieure, Paris Sciences Letters University, Sorbonne Université, Université de Paris, Centre National de la Recherche Scientifique, 24 Rue Lhomond, 75005 Paris, France
- Inovarion, 75005 Paris, France
| | - Isabelle Aujard
- Laboratoire PASTEUR, Département de Chimie, Ecole Normale Supérieure, Paris Sciences Letters University, Sorbonne Université, Centre National de la Recherche Scientifique, 24 Rue Lhomond, 75005 Paris, France
| | - Ludovic Jullien
- Laboratoire PASTEUR, Département de Chimie, Ecole Normale Supérieure, Paris Sciences Letters University, Sorbonne Université, Centre National de la Recherche Scientifique, 24 Rue Lhomond, 75005 Paris, France
| | - David Bensimon
- Laboratoire de Physique de l’Ecole Normale Supérieure, Paris Sciences Letters University, Sorbonne Université, Université de Paris, Centre National de la Recherche Scientifique, 24 Rue Lhomond, 75005 Paris, France
- Department of Chemistry and Biochemistry, University of California, Los Angeles, CA 90095, USA
| |
Collapse
|
40
|
Zhang D, Liu S, Guan J, Mou F. "Motile-targeting" drug delivery platforms based on micro/nanorobots for tumor therapy. Front Bioeng Biotechnol 2022; 10:1002171. [PMID: 36185435 PMCID: PMC9523273 DOI: 10.3389/fbioe.2022.1002171] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2022] [Accepted: 08/25/2022] [Indexed: 11/13/2022] Open
Abstract
Traditional drug delivery systems opened the gate for tumor-targeted therapy, but they generally took advantage of enhanced permeability and retention or ligand-receptor mediated interaction, and thus suffered from limited recognition range (<0.5 nm) and low targeting efficiency (0.7%, median). Alternatively, micro/nanorobots (MNRs) may act as emerging "motile-targeting" drug delivery platforms to deliver therapeutic payloads, thereby making a giant step toward effective and safe cancer treatment due to their autonomous movement and navigation in biological media. This review focuses on the most recent developments of MNRs in "motile-targeting" drug delivery. After a brief introduction to traditional tumor-targeted drug delivery strategies and various MNRs, the representative applications of MNRs in "motile-targeting" drug delivery are systematically streamlined in terms of the propelling mechanisms. Following a discussion of the current challenges of each type of MNR in biomedical applications, as well as future prospects, several promising designs for MNRs that could benefit in "motile-targeting" drug delivery are proposed. This work is expected to attract and motivate researchers from different communities to advance the creation and practical application of the "motile-targeting" drug delivery platforms.
Collapse
Affiliation(s)
| | | | | | - Fangzhi Mou
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Wuhan University of Technology, Wuhan, China
| |
Collapse
|
41
|
Rojvirat CP, Berlin JR, Nguyen TD. Evaluating spatial and network properties of NMDA-dependent neuronal connectivity in mixed cortical cultures. Brain Res 2022; 1787:147919. [PMID: 35436447 PMCID: PMC11470759 DOI: 10.1016/j.brainres.2022.147919] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Revised: 04/08/2022] [Accepted: 04/11/2022] [Indexed: 11/24/2022]
Abstract
A technique combining fluorescence imaging with Ca2+ indicators and single-cell laser scanning photostimulation of caged glutamate (LSPS) allowed identification of functional connections between individual neurons in mixed cultures of rat neocortical cells as well as observation of synchronous spontaneous activity among neurons. LSPS performed on large numbers of neurons yielded maps of functional connections between neurons and allowed calculation of neuronal network parameters. LSPS also provided an indirect measure of excitability of neurons targeted for photostimulation. By repeating LSPS sessions with the same neurons, stability of connections and change in the number and strength of connections were also determined. Experiments were conducted in the presence of bicuculline to study in detail the properties of excitatory neurotransmission. The AMPA receptor inhibitor, 6-Cyano-7-nitroquinoxaline-2,3-dione (CNQX), abolished synchronous neuronal activity but had no effect on connections mapped by LSPS. In contrast, the NMDA receptor inhibitor, 2-Amino-5-phosphono-pentanoic acid (APV), dramatically decreased the number of functional connections between neurons while also affecting synchronous spontaneous activity. Functional connections were also decreased by increasing extracellular Mg2+ concentration. These data demonstrated that LSPS mapping interrogates NMDA receptor-dependent connectivity between neurons in the network. In addition, a GluN2A-specific inhibitor, NVP-AAM077, decreased the number and strength of connections between neurons as well as neuron excitability. Conversely, the GluN2A-specific positive modulator, GNE-0723, increased these same properties. These data showed that LSPS can be used to directly study perturbations in the properties of NMDA receptor-dependent connectivity in neuronal networks. This approach should be applicable in a wide variety of in vitro and in vivo experimental preparations.
Collapse
Affiliation(s)
- Catherine P Rojvirat
- Department of Pharmacology, Physiology and Neuroscience, New Jersey Medical School, Rutgers University, 185 South Orange Avenue, Newark, NJ 07101-1709, United States; School of Graduate Studies, Rutgers Biomedical and Health Sciences Campus-Newark, Rutgers University, 185 South Orange Avenue, Newark, NJ 07101-1709, United States.
| | - Joshua R Berlin
- Department of Pharmacology, Physiology and Neuroscience, New Jersey Medical School, Rutgers University, 185 South Orange Avenue, Newark, NJ 07101-1709, United States.
| | - Tuan D Nguyen
- Department of Physics, The College of New Jersey, 2000 Pennington Rd., Ewing, NJ 08628, United States.
| |
Collapse
|
42
|
Russell LE, Dalgleish HWP, Nutbrown R, Gauld OM, Herrmann D, Fişek M, Packer AM, Häusser M. All-optical interrogation of neural circuits in behaving mice. Nat Protoc 2022; 17:1579-1620. [PMID: 35478249 PMCID: PMC7616378 DOI: 10.1038/s41596-022-00691-w] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Accepted: 02/09/2022] [Indexed: 12/22/2022]
Abstract
Recent advances combining two-photon calcium imaging and two-photon optogenetics with computer-generated holography now allow us to read and write the activity of large populations of neurons in vivo at cellular resolution and with high temporal resolution. Such 'all-optical' techniques enable experimenters to probe the effects of functionally defined neurons on neural circuit function and behavioral output with new levels of precision. This greatly increases flexibility, resolution, targeting specificity and throughput compared with alternative approaches based on electrophysiology and/or one-photon optogenetics and can interrogate larger and more densely labeled populations of neurons than current voltage imaging-based implementations. This protocol describes the experimental workflow for all-optical interrogation experiments in awake, behaving head-fixed mice. We describe modular procedures for the setup and calibration of an all-optical system (~3 h), the preparation of an indicator and opsin-expressing and task-performing animal (~3-6 weeks), the characterization of functional and photostimulation responses (~2 h per field of view) and the design and implementation of an all-optical experiment (achievable within the timescale of a normal behavioral experiment; ~3-5 h per field of view). We discuss optimizations for efficiently selecting and targeting neuronal ensembles for photostimulation sequences, as well as generating photostimulation response maps from the imaging data that can be used to examine the impact of photostimulation on the local circuit. We demonstrate the utility of this strategy in three brain areas by using different experimental setups. This approach can in principle be adapted to any brain area to probe functional connectivity in neural circuits and investigate the relationship between neural circuit activity and behavior.
Collapse
Affiliation(s)
- Lloyd E Russell
- Wolfson Institute for Biomedical Research, University College London, London, UK
| | - Henry W P Dalgleish
- Wolfson Institute for Biomedical Research, University College London, London, UK
- Sainsbury Wellcome Centre, University College London, London, UK
| | - Rebecca Nutbrown
- Wolfson Institute for Biomedical Research, University College London, London, UK
| | - Oliver M Gauld
- Wolfson Institute for Biomedical Research, University College London, London, UK
| | - Dustin Herrmann
- Wolfson Institute for Biomedical Research, University College London, London, UK
| | - Mehmet Fişek
- Wolfson Institute for Biomedical Research, University College London, London, UK
| | - Adam M Packer
- Wolfson Institute for Biomedical Research, University College London, London, UK.
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK.
| | - Michael Häusser
- Wolfson Institute for Biomedical Research, University College London, London, UK.
| |
Collapse
|
43
|
Mineur YS, Mose TN, Vanopdenbosch L, Etherington IM, Ogbejesi C, Islam A, Pineda CM, Crouse RB, Zhou W, Thompson DC, Bentham MP, Picciotto MR. Hippocampal acetylcholine modulates stress-related behaviors independent of specific cholinergic inputs. Mol Psychiatry 2022; 27:1829-1838. [PMID: 34997190 PMCID: PMC9106825 DOI: 10.1038/s41380-021-01404-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Revised: 11/15/2021] [Accepted: 11/24/2021] [Indexed: 11/08/2022]
Abstract
Acetylcholine (ACh) levels are elevated in actively depressed subjects. Conversely, antagonism of either nicotinic or muscarinic ACh receptors can have antidepressant effects in humans and decrease stress-relevant behaviors in rodents. Consistent with a role for ACh in mediating maladaptive responses to stress, brain ACh levels increase in response to stressful challenges, whereas systemically blocking acetylcholinesterase (AChE, the primary ACh degradative enzyme) elicits depression-like symptoms in human subjects, and selectively blocking AChE in the hippocampus increases relevant behaviors in rodents. We used an ACh sensor to characterize stress-evoked ACh release, then used chemogenetic, optogenetic and pharmacological approaches to determine whether cholinergic inputs from the medial septum/diagonal bands of Broca (MSDBB) or ChAT-positive neurons intrinsic to the hippocampus mediate stress-relevant behaviors in mice. Chemogenetic inhibition or activation of MSDBB cholinergic neurons did not result in significant behavioral effects, while inhibition attenuated the behavioral effects of physostigmine. In contrast, optogenetic stimulation of septohippocampal terminals or selective chemogenetic activation of ChAT-positive inputs to hippocampus increased stress-related behaviors. Finally, stimulation of sparse ChAT-positive hippocampal neurons increased stress-related behaviors in one ChAT-Cre line, which were attenuated by local infusion of cholinergic antagonists. These studies suggest that ACh signaling results in maladaptive behavioral responses to stress if the balance of signaling is shifted toward increased hippocampal engagement.
Collapse
Affiliation(s)
- Yann S Mineur
- Department of Psychiatry, Yale University School of Medicine, 34 Park Street, 3rd Floor Research, New Haven, CT, 06508, USA
| | - Tenna N Mose
- Department of Psychiatry, Yale University School of Medicine, 34 Park Street, 3rd Floor Research, New Haven, CT, 06508, USA
| | - Laura Vanopdenbosch
- Department of Psychiatry, Yale University School of Medicine, 34 Park Street, 3rd Floor Research, New Haven, CT, 06508, USA
| | - Ian M Etherington
- Department of Psychiatry, Yale University School of Medicine, 34 Park Street, 3rd Floor Research, New Haven, CT, 06508, USA
| | - Chika Ogbejesi
- Department of Psychiatry, Yale University School of Medicine, 34 Park Street, 3rd Floor Research, New Haven, CT, 06508, USA
| | - Ashraful Islam
- Department of Psychiatry, Yale University School of Medicine, 34 Park Street, 3rd Floor Research, New Haven, CT, 06508, USA
| | - Cristiana M Pineda
- Department of Psychiatry, Yale University School of Medicine, 34 Park Street, 3rd Floor Research, New Haven, CT, 06508, USA
| | - Richard B Crouse
- Department of Psychiatry, Yale University School of Medicine, 34 Park Street, 3rd Floor Research, New Haven, CT, 06508, USA
| | - Wenliang Zhou
- Department of Psychiatry, Yale University School of Medicine, 34 Park Street, 3rd Floor Research, New Haven, CT, 06508, USA
| | - David C Thompson
- Department of Psychiatry, Yale University School of Medicine, 34 Park Street, 3rd Floor Research, New Haven, CT, 06508, USA
| | - Matthew P Bentham
- Department of Psychiatry, Yale University School of Medicine, 34 Park Street, 3rd Floor Research, New Haven, CT, 06508, USA
| | - Marina R Picciotto
- Department of Psychiatry, Yale University School of Medicine, 34 Park Street, 3rd Floor Research, New Haven, CT, 06508, USA.
| |
Collapse
|
44
|
Mirzayi P, Shobeiri P, Kalantari A, Perry G, Rezaei N. Optogenetics: implications for Alzheimer's disease research and therapy. Mol Brain 2022; 15:20. [PMID: 35197102 PMCID: PMC8867657 DOI: 10.1186/s13041-022-00905-y] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Accepted: 02/10/2022] [Indexed: 12/16/2022] Open
Abstract
Alzheimer’s disease (AD), a critical neurodegenerative condition, has a wide range of effects on brain activity. Synaptic plasticity and neuronal circuits are the most vulnerable in Alzheimer’s disease, but the exact mechanism is unknown. Incorporating optogenetics into the study of AD has resulted in a significant leap in this field during the last decades, kicking off a revolution in our knowledge of the networks that underpin cognitive functions. In Alzheimer's disease, optogenetics can help to reduce and reverse neural circuit and memory impairments. Here we review how optogenetically driven methods have helped expand our knowledge of Alzheimer's disease, and how optogenetic interventions hint at a future translation into therapeutic possibilities for further utilization in clinical settings. In conclusion, neuroscience has witnessed one of its largest revolutions following the introduction of optogenetics into the field.
Collapse
Affiliation(s)
- Parsa Mirzayi
- School of Medicine, Tehran University of Medical Sciences (TUMS), Children's Medical Center Hospital, Dr. Qarib St., Keshavarz Blvd, 14194, Tehran, Iran.,Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Parnian Shobeiri
- School of Medicine, Tehran University of Medical Sciences (TUMS), Children's Medical Center Hospital, Dr. Qarib St., Keshavarz Blvd, 14194, Tehran, Iran.,Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, Iran.,Non-Communicable Diseases Research Center, Endocrinology and Metabolism Population Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran.,Research Center for Immunodeficiencies, Pediatrics Center of Excellence, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Amirali Kalantari
- School of Medicine, Tehran University of Medical Sciences (TUMS), Children's Medical Center Hospital, Dr. Qarib St., Keshavarz Blvd, 14194, Tehran, Iran.,Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - George Perry
- Department of Biology and Neurosciences Institute, University of Texas at San Antonio (UTSA), San Antonio, TX, USA
| | - Nima Rezaei
- Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, Iran. .,Research Center for Immunodeficiencies, Pediatrics Center of Excellence, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran. .,Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran. .,Research Center for Immunodeficiencies, Children's Medical Center, Dr. Gharib St, Keshavarz Blvd, Tehran, Iran.
| |
Collapse
|
45
|
Gupta P, Shinde A, Illath K, Kar S, Nagai M, Tseng FG, Santra TS. Microfluidic platforms for single neuron analysis. Mater Today Bio 2022; 13:100222. [PMID: 35243297 PMCID: PMC8866890 DOI: 10.1016/j.mtbio.2022.100222] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Revised: 02/05/2022] [Accepted: 02/14/2022] [Indexed: 11/29/2022]
Abstract
Single-neuron actions are the basis of brain function, as clinical sequelae, neuronal dysfunction or failure for most of the central nervous system (CNS) diseases and injuries can be identified via tracing single-neurons. The bulk analysis methods tend to miscue critical information by assessing the population-averaged outcomes. However, its primary requisite in neuroscience to analyze single-neurons and to understand dynamic interplay of neurons and their environment. Microfluidic systems enable precise control over nano-to femto-liter volumes via adjusting device geometry, surface characteristics, and flow-dynamics, thus facilitating a well-defined micro-environment with spatio-temporal control for single-neuron analysis. The microfluidic platform not only offers a comprehensive landscape to study brain cell diversity at the level of transcriptome, genome, and/or epigenome of individual cells but also has a substantial role in deciphering complex dynamics of brain development and brain-related disorders. In this review, we highlight recent advances of microfluidic devices for single-neuron analysis, i.e., single-neuron trapping, single-neuron dynamics, single-neuron proteomics, single-neuron transcriptomics, drug delivery at the single-neuron level, single axon guidance, and single-neuron differentiation. Moreover, we also emphasize limitations and future challenges of single-neuron analysis by focusing on key performances of throughput and multiparametric activity analysis on microfluidic platforms.
Collapse
|
46
|
Wang Y, Li Y, Yang X, Wu X, Wang Y, Pei W. Analysis of Electromagnetic Interference and Shielding in the μLED Optrode Based on Finite Element Method. FRONTIERS IN NANOTECHNOLOGY 2021. [DOI: 10.3389/fnano.2021.758484] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Monolithic integrated μLED optrode has promising applications in optogenetics due to their ability to achieve more optical channels in a smaller footprint. The current used to drive the μLED will cause electromagnetic interference (EMI) noise to the recording electrodes at a very close distance. Utilizing a grounded metal shielding layer between the active device and the electrode can potentially reduce the interference. In this paper, multi-dimensional μLED optrode models are set up according to the real device. By numerically analyzing the electromagnetic interference between the μLED and recording electrodes, several optimized shielding schemes are evaluated by simulations and experiments. Some important process and layout parameters that may influence the shielding effect are studied through the finite element method (FEM). Different circuit models based on the corresponding test environment are built to analyze the simulation and experiment results. A new PCB with a shielding layer has been designed and initially verified. The proposed novel computational model can analyze EMI quantitatively, which could facilitate the design of low-noise μLED optrode with reasonable shielding and packaging.
Collapse
|
47
|
Genetics, pathogenesis and therapeutic developments for Usher syndrome type 2. Hum Genet 2021; 141:737-758. [PMID: 34331125 DOI: 10.1007/s00439-021-02324-w] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Accepted: 07/24/2021] [Indexed: 12/28/2022]
Abstract
Usher syndrome (USH) is a rare, autosomal recessively inherited disorder resulting in a combination of sensorineural hearing loss and a progressive loss of vision resulting from retinitis pigmentosa (RP), occasionally accompanied by an altered vestibular function. More and more evidence is building up indicating that also sleep deprivation, olfactory dysfunction, deficits in tactile perception and reduced sperm motility are part of the disease etiology. USH can be clinically classified into three different types, of which Usher syndrome type 2 (USH2) is the most prevalent. In this review, we, therefore, assess the genetic and clinical aspects, available models and therapeutic developments for USH2. Mutations in USH2A, ADGRV1 and WHRN have been described to be responsible for USH2, with USH2A being the most frequently mutated USH-associated gene, explaining 50% of all cases. The proteins encoded by the USH2 genes together function in a dynamic protein complex that, among others, is found at the photoreceptor periciliary membrane and at the base of the hair bundles of inner ear hair cells. To unravel the pathogenic mechanisms underlying USH2, patient-derived cellular models and animal models including mouse, zebrafish and drosophila, have been generated that all in part mimic the USH phenotype. Multiple cellular and genetic therapeutic approaches are currently under development for USH2, mainly focused on preserving or partially restoring the visual function of which one is already in the clinical phase. These developments are opening a new gate towards a possible treatment for USH2 patients.
Collapse
|
48
|
Optogenetically-inspired neuromodulation: Translating basic discoveries into therapeutic strategies. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2021; 159:187-219. [PMID: 34446246 DOI: 10.1016/bs.irn.2021.06.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Optogenetic tools allow for the selective activation, inhibition or modulation of genetically-defined neural circuits with incredible temporal precision. Over the past decade, application of these tools in preclinical models of psychiatric disease has advanced our understanding the neural circuit basis of maladaptive behaviors in these disorders. Despite their power as an investigational tool, optogenetics cannot yet be applied in the clinical for the treatment of neurological and psychiatric disorders. To date, deep brain stimulation (DBS) is the only clinical treatment that can be used to achieve circuit-specific neuromodulation in the context of psychiatric. Despite its increasing clinical indications, the mechanisms underlying the therapeutic effects of DBS for psychiatric disorders are poorly understood, which makes optimization difficult. We discuss the variety of optogenetic tools available for preclinical research, and how these tools have been leveraged to reverse-engineer the mechanisms underlying DBS for movement and compulsive disorders. We review studies that have used optogenetics to induce plasticity within defined basal ganglia circuits, to alter neural circuit function and evaluate the corresponding effects on motor and compulsive behaviors. While not immediately applicable to patient populations, the translational power of optogenetics is in inspiring novel DBS protocols by providing a rationale for targeting defined neural circuits to ameliorate specific behavioral symptoms, and by establishing optimal stimulation paradigms that could selectively compensate for pathological synaptic plasticity within these defined neural circuits.
Collapse
|
49
|
Repina NA, McClave T, Johnson HJ, Bao X, Kane RS, Schaffer DV. Engineered Illumination Devices for Optogenetic Control of Cellular Signaling Dynamics. Cell Rep 2021; 31:107737. [PMID: 32521262 PMCID: PMC9357365 DOI: 10.1016/j.celrep.2020.107737] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Revised: 03/09/2020] [Accepted: 05/14/2020] [Indexed: 10/31/2022] Open
Abstract
Spatially and temporally varying patterns of morphogen signals during development drive cell fate specification at the proper location and time. However, current in vitro methods typically do not allow for precise, dynamic spatiotemporal control of morphogen signaling and are thus insufficient to readily study how morphogen dynamics affect cell behavior. Here, we show that optogenetic Wnt/β-catenin pathway activation can be controlled at user-defined intensities, temporal sequences, and spatial patterns using engineered illumination devices for optogenetic photostimulation and light activation at variable amplitudes (LAVA). By patterning human embryonic stem cell (hESC) cultures with varying light intensities, LAVA devices enabled dose-responsive control of optoWnt activation and Brachyury expression. Furthermore, time-varying and spatially localized patterns of light revealed tissue patterning that models the embryonic presentation of Wnt signals in vitro. LAVA devices thus provide a low-cost, user-friendly method for high-throughput and spatiotemporal optogenetic control of cell signaling for applications in developmental and cell biology.
Collapse
Affiliation(s)
- Nicole A Repina
- Department of Bioengineering, University of California, Berkeley, Berkeley, CA 94720, USA; Graduate Program in Bioengineering, University of California, San Francisco, San Francisco, CA, USA; Graduate Program in Bioengineering, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Thomas McClave
- Department of Physics, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Hunter J Johnson
- Department of Bioengineering, University of California, Berkeley, Berkeley, CA 94720, USA; Graduate Program in Bioengineering, University of California, San Francisco, San Francisco, CA, USA; Graduate Program in Bioengineering, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Xiaoping Bao
- Department of Chemical and Biomolecular Engineering, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Ravi S Kane
- School of Chemical and Biomolecular Engineering, Georgia Institute of Technology, Atlanta, GA 30332, USA.
| | - David V Schaffer
- Department of Bioengineering, University of California, Berkeley, Berkeley, CA 94720, USA; Department of Chemical and Biomolecular Engineering, University of California, Berkeley, Berkeley, CA 94720, USA; Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, CA 94720, USA; Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA.
| |
Collapse
|
50
|
Li X, Liu C, Wang R. Light Modulation of Brain and Development of Relevant Equipment. J Alzheimers Dis 2021; 74:29-41. [PMID: 32039856 DOI: 10.3233/jad-191240] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Light modulation plays an important role in understanding the pathology of brain disorders and improving brain function. Optogenetic techniques can activate or silence targeted neurons with high temporal and spatial accuracy and provide precise control, and have recently become a method for quick manipulation of genetically identified types of neurons. Photobiomodulation (PBM) is light therapy that utilizes non-ionizing light sources, including lasers, light emitting diodes, or broadband light. It provides a safe means of modulating brain activity without any irreversible damage and has established optimal treatment parameters in clinical practice. This manuscript reviews 1) how optogenetic approaches have been used to dissect neural circuits in animal models of Alzheimer's disease, Parkinson's disease, and depression, and 2) how low level transcranial lasers and LED stimulation in humans improves brain activity patterns in these diseases. State-of-the-art brain machine interfaces that can record neural activity and stimulate neurons with light have good prospects in the future.
Collapse
Affiliation(s)
- Xiaoran Li
- School of Information and Electronics, Beijing Institute of Technology, Beijing, China
| | - Chunyan Liu
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China.,Beijing Key Laboratory of Neuromodulation, Beijing, China
| | - Rong Wang
- Central Laboratory, Xuanwu Hospital, Capital Medical University, Beijing Geriatric Medical Research Center, Beijing, China.,Beijing Institute for Brain Disorders, Beijing, China
| |
Collapse
|