1
|
Dai B, You S, Wang K, Long Y, Chen J, Upreti N, Peng J, Zheng L, Chang C, Huang TJ, Guan Y, Zhuang S, Zhang D. Deep learning-enabled filter-free fluorescence microscope. SCIENCE ADVANCES 2025; 11:eadq2494. [PMID: 39742468 DOI: 10.1126/sciadv.adq2494] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/04/2024] [Accepted: 11/25/2024] [Indexed: 01/03/2025]
Abstract
Optical filtering is an indispensable part of fluorescence microscopy for selectively highlighting molecules labeled with a specific fluorophore and suppressing background noise. However, the utilization of optical filtering sets increases the complexity, size, and cost of microscopic systems, making them less suitable for multifluorescence channel, high-speed imaging. Here, we present filter-free fluorescence microscopic imaging enabled with deep learning-based digital spectral filtering. This approach allows for automatic fluorescence channel selection after image acquisition and accurate prediction of fluorescence by computing color changes due to spectral shifts with the presence of excitation scattering. Fluorescence prediction for cells and tissues labeled with various fluorophores was demonstrated under different magnification powers. The technique offers accurate identification of labeling with robust sensitivity and specificity, achieving consistent results with the reference standard. Beyond fluorescence microscopy, the deep learning-enabled spectral filtering strategy has the potential to drive the development of other biomedical applications, including cytometry and endoscopy.
Collapse
Affiliation(s)
- Bo Dai
- Engineering Research Center of Optical Instrument and System, the Ministry of Education, Shanghai Key Laboratory of Modern Optical System, University of Shanghai for Science and Technology, Shanghai 200093, China
| | - Shaojie You
- Engineering Research Center of Optical Instrument and System, the Ministry of Education, Shanghai Key Laboratory of Modern Optical System, University of Shanghai for Science and Technology, Shanghai 200093, China
| | - Kan Wang
- Department of Neurology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai 200127, China
| | - Yan Long
- Engineering Research Center of Optical Instrument and System, the Ministry of Education, Shanghai Key Laboratory of Modern Optical System, University of Shanghai for Science and Technology, Shanghai 200093, China
| | - Junyi Chen
- Engineering Research Center of Optical Instrument and System, the Ministry of Education, Shanghai Key Laboratory of Modern Optical System, University of Shanghai for Science and Technology, Shanghai 200093, China
| | - Neil Upreti
- Department of Mechanical Engineering and Materials Science, Duke University, Durham, NC 27709, USA
| | - Jing Peng
- Department of Neurology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai 200127, China
| | - Lulu Zheng
- Engineering Research Center of Optical Instrument and System, the Ministry of Education, Shanghai Key Laboratory of Modern Optical System, University of Shanghai for Science and Technology, Shanghai 200093, China
| | - Chenliang Chang
- Engineering Research Center of Optical Instrument and System, the Ministry of Education, Shanghai Key Laboratory of Modern Optical System, University of Shanghai for Science and Technology, Shanghai 200093, China
| | - Tony Jun Huang
- Department of Mechanical Engineering and Materials Science, Duke University, Durham, NC 27709, USA
| | - Yangtai Guan
- Department of Neurology, Punan Branch of Renji Hospital, School of Medicine, Shanghai Jiaotong University (Punan Hospital in Pudong New District, Shanghai), Shanghai 200125, China
| | - Songlin Zhuang
- Engineering Research Center of Optical Instrument and System, the Ministry of Education, Shanghai Key Laboratory of Modern Optical System, University of Shanghai for Science and Technology, Shanghai 200093, China
| | - Dawei Zhang
- Engineering Research Center of Optical Instrument and System, the Ministry of Education, Shanghai Key Laboratory of Modern Optical System, University of Shanghai for Science and Technology, Shanghai 200093, China
| |
Collapse
|
2
|
McNaughton BL. Neuronal 'Ensemble' Recording and the Search for the Cell Assembly: A Personal History. Hippocampus 2025; 35:e23669. [PMID: 39676610 DOI: 10.1002/hipo.23669] [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: 08/20/2024] [Revised: 11/20/2024] [Accepted: 11/26/2024] [Indexed: 12/17/2024]
Abstract
This contribution is part of the special issue on the Hippocampus focused on personal histories of advances in knowledge on the hippocampus and related structures. An account is offered of the author's role in the development of neural ensemble recording: stereo recording (stereotrodes, tetrodes) and the use of this approach to search for evidence of Hebb's "cell assemblies" and "phase sequences", the holy grail of the neuroscience of learning and memory.
Collapse
Affiliation(s)
- Bruce L McNaughton
- Department of Neurobiology and Behavior, University of California Irvine, Irvine, USA
- Department of Neuroscience, The University of Lethbridge, Irvine, USA
| |
Collapse
|
3
|
Tian C, Tang F, Wang M, Zhou J, Yue X, Luan F, Zhuang X. Sensitive detection of H2S in the environment with electrochemiluminescence and fluorescence double-mode sensor constructed by Eu2O3@CDs NPs. SENSORS AND ACTUATORS B: CHEMICAL 2025; 422:136582. [DOI: 10.1016/j.snb.2024.136582] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
4
|
Pang W, Yuan C, Zhong T, Huang X, Pan Y, Qu J, Nie L, Zhou Y, Lai P. Diagnostic and therapeutic optical imaging in cardiovascular diseases. iScience 2024; 27:111216. [PMID: 39569375 PMCID: PMC11576408 DOI: 10.1016/j.isci.2024.111216] [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] [Indexed: 11/22/2024] Open
Abstract
Cardiovascular disease (CVD) is one of the most prevalent health threats globally. Traditional diagnostic methods for CVDs, including electrocardiography, ultrasound, and cardiac magnetic resonance imaging, have inherent limitations in real-time monitoring and high-resolution visualization of cardiovascular pathophysiology. In recent years, optical imaging technology has gained considerable attention as a non-invasive, high-resolution, real-time monitoring solution in the study and diagnosis of CVD. This review discusses the latest advancements, and applications of optical techniques in cardiac imaging. We compare the advantages of optical imaging over traditional modalities and especially scrutinize techniques such as optical coherence tomography, photoacoustic imaging, and fluorescence imaging. We summarize their investigations in atherosclerosis, myocardial infarction, and heart valve disease, etc. Additionally, we discuss challenges like deep-tissue imaging and high spatiotemporal resolution adjustment, and review existing solutions such as multimodal integration, artificial intelligence, and enhanced optical probes. This article aims to drive further development in optical imaging technologies to provide more precise and efficient tools for early diagnosis, pathological mechanism exploration, and treatment of CVD.
Collapse
Affiliation(s)
- Weiran Pang
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Hong Kong SAR, China
- Medical Research Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510080, China
| | - Chuqi Yuan
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Hong Kong SAR, China
| | - Tianting Zhong
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Hong Kong SAR, China
| | - Xiazi Huang
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Hong Kong SAR, China
| | - Yue Pan
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China
- Nanchang Research Institute, Sun Yat-Sen University, Nanchang 330096, China
| | - Junle Qu
- Key Laboratory of Optoelectronic Devices and Systems of Ministry of Education and Guangdong Province, College of Physics and Optoelectronic Engineering, Shenzhen 518060, China
| | - Liming Nie
- Medical Research Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510080, China
| | - Yingying Zhou
- College of Professional and Continuing Education, The Hong Kong Polytechnic University, Hong Kong SAR, China
| | - Puxiang Lai
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Hong Kong SAR, China
- The Joint Research Centre for Biosensing and Precision Theranostics, The Hong Kong Polytechnic University, Hong Kong SAR, China
| |
Collapse
|
5
|
Xiao G, Cai Y, Zhang Y, Xie J, Wu L, Xie H, Wu J, Dai Q. Mesoscale neuronal granular trial variability in vivo illustrated by nonlinear recurrent network in silico. Nat Commun 2024; 15:9894. [PMID: 39548098 PMCID: PMC11567969 DOI: 10.1038/s41467-024-54346-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Accepted: 11/06/2024] [Indexed: 11/17/2024] Open
Abstract
Large-scale neural recording with single-neuron resolution has revealed the functional complexity of the neural systems. However, even under well-designed task conditions, the cortex-wide network exhibits highly dynamic trial variability, posing challenges to the conventional trial-averaged analysis. To study mesoscale trial variability, we conducted a comparative study between fluorescence imaging of layer-2/3 neurons in vivo and network simulation in silico. We imaged up to 40,000 cortical neurons' triggered responses by deep brain stimulus (DBS). And we build an in silico network to reproduce the biological phenomena we observed in vivo. We proved the existence of ineluctable trial variability and found it influenced by input amplitude and range. Moreover, we demonstrated that a spatially heterogeneous coding community accounts for more reliable inter-trial coding despite single-unit trial variability. A deeper understanding of trial variability from the perspective of a dynamical system may lead to uncovering intellectual abilities such as parallel coding and creativity.
Collapse
Affiliation(s)
- Guihua Xiao
- Beijing National Research Center for Information Science and Technology, Tsinghua University, Beijing, China
- Department of Automation, Tsinghua University, Beijing, China
- Institute for Brain and Cognitive Sciences, Tsinghua University, Beijing, China
| | - Yeyi Cai
- Department of Automation, Tsinghua University, Beijing, China
- Institute for Brain and Cognitive Sciences, Tsinghua University, Beijing, China
| | - Yuanlong Zhang
- Department of Automation, Tsinghua University, Beijing, China
- Institute for Brain and Cognitive Sciences, Tsinghua University, Beijing, China
| | - Jingyu Xie
- Department of Automation, Tsinghua University, Beijing, China
- Institute for Brain and Cognitive Sciences, Tsinghua University, Beijing, China
| | - Lifan Wu
- Department of Automation, Tsinghua University, Beijing, China
- Institute for Brain and Cognitive Sciences, Tsinghua University, Beijing, China
| | - Hao Xie
- Department of Automation, Tsinghua University, Beijing, China
- Institute for Brain and Cognitive Sciences, Tsinghua University, Beijing, China
| | - Jiamin Wu
- Beijing National Research Center for Information Science and Technology, Tsinghua University, Beijing, China.
- Department of Automation, Tsinghua University, Beijing, China.
- Institute for Brain and Cognitive Sciences, Tsinghua University, Beijing, China.
- IDG/McGovern Institute for Brain Research, Tsinghua University, Beijing, China.
| | - Qionghai Dai
- Beijing National Research Center for Information Science and Technology, Tsinghua University, Beijing, China.
- Department of Automation, Tsinghua University, Beijing, China.
- Institute for Brain and Cognitive Sciences, Tsinghua University, Beijing, China.
- IDG/McGovern Institute for Brain Research, Tsinghua University, Beijing, China.
| |
Collapse
|
6
|
Bai L, Cong L, Shi Z, Zhao Y, Zhang Y, Lu B, Zhang J, Xiong ZQ, Xu N, Mu Y, Wang K. Volumetric voltage imaging of neuronal populations in the mouse brain by confocal light-field microscopy. Nat Methods 2024; 21:2160-2170. [PMID: 39379535 DOI: 10.1038/s41592-024-02458-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Accepted: 09/06/2024] [Indexed: 10/10/2024]
Abstract
Voltage imaging measures neuronal activity directly and holds promise for understanding information processing within individual neurons and across populations. However, imaging voltage over large neuronal populations has been challenging owing to the simultaneous requirements of high imaging speed and signal-to-noise ratio, large volume coverage and low photobleaching rate. Here, to overcome this challenge, we developed a confocal light-field microscope that surpassed the traditional limits in speed and noise performance by incorporating a speed-enhanced camera, a fast and robust scanning mechanism, laser-speckle-noise elimination and optimized light efficiency. With this method, we achieved simultaneous recording from more than 300 spiking neurons within an 800-µm-diameter and 180-µm-thick volume in the mouse cortex, for more than 20 min. By integrating the spatial and voltage activity profiles, we have mapped three-dimensional neural coordination patterns in awake mouse brains. Our method is robust for routine application in volumetric voltage imaging.
Collapse
Affiliation(s)
- Lu Bai
- Institute of Neuroscience, Key Laboratory of Brain Cognition and Brain-Inspired Intelligence Technology, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Lin Cong
- Institute of Neuroscience, Key Laboratory of Brain Cognition and Brain-Inspired Intelligence Technology, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| | - Ziqi Shi
- Institute of Neuroscience, Key Laboratory of Brain Cognition and Brain-Inspired Intelligence Technology, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Yuchen Zhao
- Institute of Neuroscience, Key Laboratory of Brain Cognition and Brain-Inspired Intelligence Technology, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Yujie Zhang
- Institute of Neuroscience, Key Laboratory of Brain Cognition and Brain-Inspired Intelligence Technology, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Bin Lu
- Institute of Neuroscience, Key Laboratory of Brain Cognition and Brain-Inspired Intelligence Technology, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| | - Jing Zhang
- Institute of Neuroscience, Key Laboratory of Brain Cognition and Brain-Inspired Intelligence Technology, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| | - Zhi-Qi Xiong
- Institute of Neuroscience, Key Laboratory of Brain Cognition and Brain-Inspired Intelligence Technology, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
- Shanghai Center for Brain Science and Brain-Inspired Intelligence Technology, Shanghai, China
| | - Ninglong Xu
- Institute of Neuroscience, Key Laboratory of Brain Cognition and Brain-Inspired Intelligence Technology, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
- Shanghai Center for Brain Science and Brain-Inspired Intelligence Technology, Shanghai, China
| | - Yu Mu
- Institute of Neuroscience, Key Laboratory of Brain Cognition and Brain-Inspired Intelligence Technology, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Kai Wang
- Institute of Neuroscience, Key Laboratory of Brain Cognition and Brain-Inspired Intelligence Technology, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China.
- University of Chinese Academy of Sciences, Beijing, China.
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China.
| |
Collapse
|
7
|
Zhang Y, Wang M, Zhu Q, Guo Y, Liu B, Li J, Yao X, Kong C, Zhang Y, Huang Y, Qi H, Wu J, Guo ZV, Dai Q. Long-term mesoscale imaging of 3D intercellular dynamics across a mammalian organ. Cell 2024; 187:6104-6122.e25. [PMID: 39276776 DOI: 10.1016/j.cell.2024.08.026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Revised: 06/06/2024] [Accepted: 08/13/2024] [Indexed: 09/17/2024]
Abstract
A comprehensive understanding of physio-pathological processes necessitates non-invasive intravital three-dimensional (3D) imaging over varying spatial and temporal scales. However, huge data throughput, optical heterogeneity, surface irregularity, and phototoxicity pose great challenges, leading to an inevitable trade-off between volume size, resolution, speed, sample health, and system complexity. Here, we introduce a compact real-time, ultra-large-scale, high-resolution 3D mesoscope (RUSH3D), achieving uniform resolutions of 2.6 × 2.6 × 6 μm3 across a volume of 8,000 × 6,000 × 400 μm3 at 20 Hz with low phototoxicity. Through the integration of multiple computational imaging techniques, RUSH3D facilitates a 13-fold improvement in data throughput and an orders-of-magnitude reduction in system size and cost. With these advantages, we observed premovement neural activity and cross-day visual representational drift across the mouse cortex, the formation and progression of multiple germinal centers in mouse inguinal lymph nodes, and heterogeneous immune responses following traumatic brain injury-all at single-cell resolution, opening up a horizon for intravital mesoscale study of large-scale intercellular interactions at the organ level.
Collapse
Affiliation(s)
- Yuanlong Zhang
- Department of Automation, Tsinghua University, Beijing 100084, China; Institute for Brain and Cognitive Sciences, Tsinghua University, Beijing 100084, China; Beijing Key Laboratory of Multi-dimension & Multi-scale Computational Photography (MMCP), Tsinghua University, Beijing 100084, China; IDG/McGovern Institute for Brain Research, Tsinghua University, Beijing 100084, China
| | - Mingrui Wang
- Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen 518071, China
| | - Qiyu Zhu
- IDG/McGovern Institute for Brain Research, Tsinghua University, Beijing 100084, China; School of Basic Medical Sciences, Tsinghua University, Beijing 100084, China; Tsinghua-Peking Center for Life Sciences, Beijing 100084, China
| | - Yuduo Guo
- Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen 518071, China
| | - Bo Liu
- School of Basic Medical Sciences, Tsinghua University, Beijing 100084, China; Tsinghua-Peking Center for Life Sciences, Beijing 100084, China; Laboratory of Dynamic Immunobiology, Institute for Immunology, Tsinghua University, Beijing 100084, China
| | - Jiamin Li
- IDG/McGovern Institute for Brain Research, Tsinghua University, Beijing 100084, China; School of Basic Medical Sciences, Tsinghua University, Beijing 100084, China; Tsinghua-Peking Center for Life Sciences, Beijing 100084, China
| | - Xiao Yao
- IDG/McGovern Institute for Brain Research, Tsinghua University, Beijing 100084, China; School of Basic Medical Sciences, Tsinghua University, Beijing 100084, China; Tsinghua-Peking Center for Life Sciences, Beijing 100084, China
| | - Chui Kong
- School of Information Science and Technology, Fudan University, Shanghai 200433, China
| | - Yi Zhang
- Department of Automation, Tsinghua University, Beijing 100084, China; Institute for Brain and Cognitive Sciences, Tsinghua University, Beijing 100084, China; Beijing Key Laboratory of Multi-dimension & Multi-scale Computational Photography (MMCP), Tsinghua University, Beijing 100084, China; IDG/McGovern Institute for Brain Research, Tsinghua University, Beijing 100084, China
| | - Yuchao Huang
- IDG/McGovern Institute for Brain Research, Tsinghua University, Beijing 100084, China; School of Basic Medical Sciences, Tsinghua University, Beijing 100084, China; Tsinghua-Peking Center for Life Sciences, Beijing 100084, China
| | - Hai Qi
- School of Basic Medical Sciences, Tsinghua University, Beijing 100084, China; Tsinghua-Peking Center for Life Sciences, Beijing 100084, China; Laboratory of Dynamic Immunobiology, Institute for Immunology, Tsinghua University, Beijing 100084, China
| | - Jiamin Wu
- Department of Automation, Tsinghua University, Beijing 100084, China; Institute for Brain and Cognitive Sciences, Tsinghua University, Beijing 100084, China; Beijing Key Laboratory of Multi-dimension & Multi-scale Computational Photography (MMCP), Tsinghua University, Beijing 100084, China; IDG/McGovern Institute for Brain Research, Tsinghua University, Beijing 100084, China.
| | - Zengcai V Guo
- IDG/McGovern Institute for Brain Research, Tsinghua University, Beijing 100084, China; School of Basic Medical Sciences, Tsinghua University, Beijing 100084, China; Tsinghua-Peking Center for Life Sciences, Beijing 100084, China.
| | - Qionghai Dai
- Department of Automation, Tsinghua University, Beijing 100084, China; Institute for Brain and Cognitive Sciences, Tsinghua University, Beijing 100084, China; Beijing Key Laboratory of Multi-dimension & Multi-scale Computational Photography (MMCP), Tsinghua University, Beijing 100084, China; IDG/McGovern Institute for Brain Research, Tsinghua University, Beijing 100084, China.
| |
Collapse
|
8
|
Farrants H, Shuai Y, Lemon WC, Monroy Hernandez C, Zhang D, Yang S, Patel R, Qiao G, Frei MS, Plutkis SE, Grimm JB, Hanson TL, Tomaska F, Turner GC, Stringer C, Keller PJ, Beyene AG, Chen Y, Liang Y, Lavis LD, Schreiter ER. A modular chemigenetic calcium indicator for multiplexed in vivo functional imaging. Nat Methods 2024; 21:1916-1925. [PMID: 39304767 PMCID: PMC11466818 DOI: 10.1038/s41592-024-02411-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Accepted: 08/12/2024] [Indexed: 09/22/2024]
Abstract
Genetically encoded fluorescent calcium indicators allow cellular-resolution recording of physiology. However, bright, genetically targetable indicators that can be multiplexed with existing tools in vivo are needed for simultaneous imaging of multiple signals. Here we describe WHaloCaMP, a modular chemigenetic calcium indicator built from bright dye-ligands and protein sensor domains. Fluorescence change in WHaloCaMP results from reversible quenching of the bound dye via a strategically placed tryptophan. WHaloCaMP is compatible with rhodamine dye-ligands that fluoresce from green to near-infrared, including several that efficiently label the brain in animals. When bound to a near-infrared dye-ligand, WHaloCaMP shows a 7× increase in fluorescence intensity and a 2.1-ns increase in fluorescence lifetime upon calcium binding. We use WHaloCaMP1a to image Ca2+ responses in vivo in flies and mice, to perform three-color multiplexed functional imaging of hundreds of neurons and astrocytes in zebrafish larvae and to quantify Ca2+ concentration using fluorescence lifetime imaging microscopy (FLIM).
Collapse
Affiliation(s)
- Helen Farrants
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA, USA.
| | - Yichun Shuai
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA, USA
| | - William C Lemon
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA, USA
| | | | - Deng Zhang
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA, USA
| | - Shang Yang
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA, USA
| | - Ronak Patel
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA, USA
| | - Guanda Qiao
- Department of Diagnostic Radiology and Nuclear Medicine, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Michelle S Frei
- Department of Chemical Biology, Max Planck Institute for Medical Research, Heidelberg, Germany
- Department of Pharmacology, University of California San Diego, La Jolla, CA, USA
| | - Sarah E Plutkis
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA, USA
| | - Jonathan B Grimm
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA, USA
| | - Timothy L Hanson
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA, USA
| | - Filip Tomaska
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA, USA
- Department of Electrical and Computer Engineering, Center for BioEngineering, Neuroscience Research Institute, University of California, Santa Barbara, Santa Barbara, CA, USA
| | - Glenn C Turner
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA, USA
| | - Carsen Stringer
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA, USA
| | - Philipp J Keller
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA, USA
| | - Abraham G Beyene
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA, USA
| | - Yao Chen
- Department of Neuroscience, Washington University in St. Louis, St. Louis, MO, USA
| | - Yajie Liang
- Department of Diagnostic Radiology and Nuclear Medicine, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Luke D Lavis
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA, USA
| | - Eric R Schreiter
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA, USA.
| |
Collapse
|
9
|
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
|
10
|
Li L, Zhang B, Zhao W, Sheng D, Yin L, Sheng X, Yao D. Multimodal Technologies for Closed-Loop Neural Modulation and Sensing. Adv Healthc Mater 2024; 13:e2303289. [PMID: 38640468 DOI: 10.1002/adhm.202303289] [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: 09/27/2023] [Revised: 03/11/2024] [Indexed: 04/21/2024]
Abstract
Existing methods for studying neural circuits and treating neurological disorders are typically based on physical and chemical cues to manipulate and record neural activities. These approaches often involve predefined, rigid, and unchangeable signal patterns, which cannot be adjusted in real time according to the patient's condition or neural activities. With the continuous development of neural interfaces, conducting in vivo research on adaptive and modifiable treatments for neurological diseases and neural circuits is now possible. In this review, current and potential integration of various modalities to achieve precise, closed-loop modulation, and sensing in neural systems are summarized. Advanced materials, devices, or systems that generate or detect electrical, magnetic, optical, acoustic, or chemical signals are highlighted and utilized to interact with neural cells, tissues, and networks for closed-loop interrogation. Further, the significance of developing closed-loop techniques for diagnostics and treatment of neurological disorders such as epilepsy, depression, rehabilitation of spinal cord injury patients, and exploration of brain neural circuit functionality is elaborated.
Collapse
Affiliation(s)
- Lizhu Li
- Sichuan Provincial Key Laboratory for Human Disease Gene Study and the Center for Medical Genetics, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, 611731, China
| | - Bozhen Zhang
- School of Materials Science and Engineering, The Key Laboratory of Advanced Materials of Ministry of Education, State Key Laboratory of New Ceramics and Fine Processing, Laboratory of Flexible Electronics Technology, Tsinghua University, Beijing, 100084, China
| | - Wenxin Zhao
- Department of Electronic Engineering, Beijing National Research Center for Information Science and Technology, Institute for Precision Medicine, Laboratory of Flexible Electronics Technology, IDG/McGovern Institute for Brain Research, Tsinghua University, Beijing, 100084, China
| | - David Sheng
- Department of Electronic Engineering, Beijing National Research Center for Information Science and Technology, Institute for Precision Medicine, Laboratory of Flexible Electronics Technology, IDG/McGovern Institute for Brain Research, Tsinghua University, Beijing, 100084, China
| | - Lan Yin
- School of Materials Science and Engineering, The Key Laboratory of Advanced Materials of Ministry of Education, State Key Laboratory of New Ceramics and Fine Processing, Laboratory of Flexible Electronics Technology, Tsinghua University, Beijing, 100084, China
| | - Xing Sheng
- Department of Electronic Engineering, Beijing National Research Center for Information Science and Technology, Institute for Precision Medicine, Laboratory of Flexible Electronics Technology, IDG/McGovern Institute for Brain Research, Tsinghua University, Beijing, 100084, China
| | - Dezhong Yao
- Sichuan Provincial Key Laboratory for Human Disease Gene Study and the Center for Medical Genetics, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, 611731, China
| |
Collapse
|
11
|
Kobayashi R, Shinomoto S. Inference of monosynaptic connections from parallel spike trains: A review. Neurosci Res 2024:S0168-0102(24)00097-X. [PMID: 39098768 DOI: 10.1016/j.neures.2024.07.006] [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: 03/19/2024] [Revised: 07/12/2024] [Accepted: 07/19/2024] [Indexed: 08/06/2024]
Abstract
This article presents a mini-review about the progress in inferring monosynaptic connections from spike trains of multiple neurons over the past twenty years. First, we explain a variety of meanings of "neuronal connectivity" in different research areas of neuroscience, such as structural connectivity, monosynaptic connectivity, and functional connectivity. Among these, we focus on the methods used to infer the monosynaptic connectivity from spike data. We then summarize the inference methods based on two main approaches, i.e., correlation-based and model-based approaches. Finally, we describe available source codes for connectivity inference and future challenges. Although inference will never be perfect, the accuracy of identifying the monosynaptic connections has improved dramatically in recent years due to continuous efforts.
Collapse
Affiliation(s)
- Ryota Kobayashi
- Graduate School of Frontier Sciences, The University of Tokyo, Chiba 277-8561, Japan; Mathematics and Informatics Center, The University of Tokyo, Tokyo 113-8656, Japan.
| | - Shigeru Shinomoto
- Graduate School of Biostudies, Kyoto University, Kyoto 606-8501, Japan; Research Organization of Open Innovation and Collaboration, Ritsumeikan University, Osaka 567-8570, Japan
| |
Collapse
|
12
|
Liao Z, Gonzalez KC, Li DM, Yang CM, Holder D, McClain NE, Zhang G, Evans SW, Chavarha M, Simko J, Makinson CD, Lin MZ, Losonczy A, Negrean A. Functional architecture of intracellular oscillations in hippocampal dendrites. Nat Commun 2024; 15:6295. [PMID: 39060234 PMCID: PMC11282248 DOI: 10.1038/s41467-024-50546-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Accepted: 07/10/2024] [Indexed: 07/28/2024] Open
Abstract
Fast electrical signaling in dendrites is central to neural computations that support adaptive behaviors. Conventional techniques lack temporal and spatial resolution and the ability to track underlying membrane potential dynamics present across the complex three-dimensional dendritic arbor in vivo. Here, we perform fast two-photon imaging of dendritic and somatic membrane potential dynamics in single pyramidal cells in the CA1 region of the mouse hippocampus during awake behavior. We study the dynamics of subthreshold membrane potential and suprathreshold dendritic events throughout the dendritic arbor in vivo by combining voltage imaging with simultaneous local field potential recording, post hoc morphological reconstruction, and a spatial navigation task. We systematically quantify the modulation of local event rates by locomotion in distinct dendritic regions, report an advancing gradient of dendritic theta phase along the basal-tuft axis, and describe a predominant hyperpolarization of the dendritic arbor during sharp-wave ripples. Finally, we find that spatial tuning of dendritic representations dynamically reorganizes following place field formation. Our data reveal how the organization of electrical signaling in dendrites maps onto the anatomy of the dendritic tree across behavior, oscillatory network, and functional cell states.
Collapse
Affiliation(s)
- Zhenrui Liao
- Department of Neuroscience, Columbia University, New York, USA
- Mortimer B. Zuckerman Mind Brain Behavior Institute, Columbia University, New York, USA
| | - Kevin C Gonzalez
- Department of Neuroscience, Columbia University, New York, USA
- Mortimer B. Zuckerman Mind Brain Behavior Institute, Columbia University, New York, USA
| | - Deborah M Li
- Department of Neuroscience, Columbia University, New York, USA
- Mortimer B. Zuckerman Mind Brain Behavior Institute, Columbia University, New York, USA
| | - Catalina M Yang
- Department of Neuroscience, Columbia University, New York, USA
- Mortimer B. Zuckerman Mind Brain Behavior Institute, Columbia University, New York, USA
| | - Donald Holder
- Department of Neuroscience, Columbia University, New York, USA
- Mortimer B. Zuckerman Mind Brain Behavior Institute, Columbia University, New York, USA
| | - Natalie E McClain
- Department of Neuroscience, Columbia University, New York, USA
- Mortimer B. Zuckerman Mind Brain Behavior Institute, Columbia University, New York, USA
| | - Guofeng Zhang
- Department of Neurobiology, Stanford University, Stanford, USA
| | - Stephen W Evans
- Department of Neurobiology, Stanford University, Stanford, USA
- The Boulder Creek Research Institute, Los Altos, USA
| | - Mariya Chavarha
- Department of Bioengineering, Stanford University, Stanford, USA
| | - Jane Simko
- Department of Neuroscience, Columbia University, New York, USA
- Department of Neurology, Columbia University, New York, USA
| | - Christopher D Makinson
- Department of Neuroscience, Columbia University, New York, USA
- Department of Neurology, Columbia University, New York, USA
| | - Michael Z Lin
- Department of Neurobiology, Stanford University, Stanford, USA
- Department of Bioengineering, Stanford University, Stanford, USA
| | - Attila Losonczy
- Department of Neuroscience, Columbia University, New York, USA.
- Mortimer B. Zuckerman Mind Brain Behavior Institute, Columbia University, New York, USA.
- Kavli Institute for Brain Science, Columbia University, New York, USA.
| | - Adrian Negrean
- Department of Neuroscience, Columbia University, New York, USA.
- Mortimer B. Zuckerman Mind Brain Behavior Institute, Columbia University, New York, USA.
- Allen Institute for Neural Dynamics, Seattle, USA.
| |
Collapse
|
13
|
Elleman AV, Milicic N, Williams DJ, Simko J, Liu CJ, Haynes AL, Ehrlich DE, Makinson CD, Du Bois J. Behavioral control through the direct, focal silencing of neuronal activity. Cell Chem Biol 2024; 31:1324-1335.e20. [PMID: 38729162 PMCID: PMC11260259 DOI: 10.1016/j.chembiol.2024.04.003] [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: 07/24/2023] [Revised: 01/02/2024] [Accepted: 04/09/2024] [Indexed: 05/12/2024]
Abstract
The ability to optically stimulate and inhibit neurons has revolutionized neuroscience research. Here, we present a direct, potent, user-friendly chemical approach for optically silencing neurons. We have rendered saxitoxin (STX), a naturally occurring paralytic agent, transiently inert through chemical protection with a previously undisclosed nitrobenzyl-derived photocleavable group. Exposing the caged toxin, STX-bpc, to a brief (5 ms) pulse of light effects rapid release of a potent STX derivative and transient, spatially precise blockade of voltage-gated sodium channels (NaVs). We demonstrate the efficacy of STX-bpc for parametrically manipulating action potentials in mammalian neurons and brain slice. Additionally, we show the effectiveness of this reagent for silencing neural activity by dissecting sensory-evoked swimming in larval zebrafish. Photo-uncaging of STX-bpc is a straightforward method for non-invasive, reversible, spatiotemporally precise neural silencing without the need for genetic access, thus removing barriers for comparative research.
Collapse
Affiliation(s)
- Anna V Elleman
- Department of Chemistry, Stanford University, 333 Campus Drive, Stanford, CA 94305, USA
| | - Nikola Milicic
- Department of Integrative Biology, University of Wisconsin-Madison, 121 Integrative Biology Research Building, 1117 W Johnson St, Madison, WI 53706, USA
| | - Damian J Williams
- Department of Neurology, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, 710 W 168th St, New York, NY 10032, USA
| | - Jane Simko
- Department of Neurology, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, 710 W 168th St, New York, NY 10032, USA
| | - Christine J Liu
- Department of Neurology, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, 710 W 168th St, New York, NY 10032, USA; Department of Neuroscience, Columbia University, Jerome L. Greene Science Center, 3227 Broadway, MC 9872, New York, NY 10027, USA
| | - Allison L Haynes
- Department of Chemistry, Stanford University, 333 Campus Drive, Stanford, CA 94305, USA
| | - David E Ehrlich
- Department of Integrative Biology, University of Wisconsin-Madison, 121 Integrative Biology Research Building, 1117 W Johnson St, Madison, WI 53706, USA.
| | - Christopher D Makinson
- Department of Neurology, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, 710 W 168th St, New York, NY 10032, USA; Department of Neuroscience, Columbia University, Jerome L. Greene Science Center, 3227 Broadway, MC 9872, New York, NY 10027, USA.
| | - J Du Bois
- Department of Chemistry, Stanford University, 333 Campus Drive, Stanford, CA 94305, USA.
| |
Collapse
|
14
|
Nevelchuk S, Brawek B, Schwarz N, Valiente-Gabioud A, Wuttke TV, Kovalchuk Y, Koch H, Höllig A, Steiner F, Figarella K, Griesbeck O, Garaschuk O. Morphotype-specific calcium signaling in human microglia. J Neuroinflammation 2024; 21:175. [PMID: 39020359 PMCID: PMC11256502 DOI: 10.1186/s12974-024-03169-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2024] [Accepted: 07/09/2024] [Indexed: 07/19/2024] Open
Abstract
BACKGROUND Key functions of Ca2+ signaling in rodent microglia include monitoring the brain state as well as the surrounding neuronal activity and sensing the danger or damage in their vicinity. Microglial Ca2+ dyshomeostasis is a disease hallmark in many mouse models of neurological disorders but the Ca2+ signal properties of human microglia remain unknown. METHODS We developed a novel genetically-encoded ratiometric Ca2+ indicator, targeting microglial cells in the freshly resected human tissue, organotypically cultured tissue slices and analyzed in situ ongoing Ca2+ signaling of decades-old microglia dwelling in their native microenvironment. RESULTS The data revealed marked compartmentalization of Ca2+ signals, with signal properties differing across the compartments and resident morphotypes. The basal Ca2+ levels were low in ramified and high in ameboid microglia. The fraction of cells with ongoing Ca2+ signaling, the fraction and the amplitude of process Ca2+ signals and the duration of somatic Ca2+ signals decreased when moving from ramified via hypertrophic to ameboid microglia. In contrast, the size of active compartments, the fraction and amplitude of somatic Ca2+ signals and the duration of process Ca2+ signals increased along this pathway.
Collapse
Affiliation(s)
- Sofia Nevelchuk
- Department of Neurophysiology, Institute of Physiology, Eberhard Karls University of Tübingen, Keplerstr. 15, 72074, Tübingen, Germany
| | - Bianca Brawek
- Department of Neurophysiology, Institute of Physiology, Eberhard Karls University of Tübingen, Keplerstr. 15, 72074, Tübingen, Germany
| | - Niklas Schwarz
- Department of Neurology and Epileptology, Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany
| | - Ariel Valiente-Gabioud
- Tools for Bio-Imaging, Max-Planck-Institute for Biological Intelligence, Martinsried, Germany
| | - Thomas V Wuttke
- Department of Neurology and Epileptology, Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany
- Department of Neurosurgery, University of Tübingen, Tübingen, Germany
| | - Yury Kovalchuk
- Department of Neurophysiology, Institute of Physiology, Eberhard Karls University of Tübingen, Keplerstr. 15, 72074, Tübingen, Germany
| | - Henner Koch
- Department of Epileptology, Neurology, RWTH Aachen University Hospital, Aachen, Germany
| | - Anke Höllig
- Department of Neurosurgery, RWTH Aachen University, Aachen, Germany
| | - Frederik Steiner
- Department of Neurophysiology, Institute of Physiology, Eberhard Karls University of Tübingen, Keplerstr. 15, 72074, Tübingen, Germany
| | - Katherine Figarella
- Department of Neurophysiology, Institute of Physiology, Eberhard Karls University of Tübingen, Keplerstr. 15, 72074, Tübingen, Germany
- Department of Anesthesiology, Critical Care and Pain Medicine, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Oliver Griesbeck
- Tools for Bio-Imaging, Max-Planck-Institute for Biological Intelligence, Martinsried, Germany
| | - Olga Garaschuk
- Department of Neurophysiology, Institute of Physiology, Eberhard Karls University of Tübingen, Keplerstr. 15, 72074, Tübingen, Germany.
| |
Collapse
|
15
|
Olorocisimo JP, Ohta Y, Regonia PR, Castillo VCG, Yoshimoto J, Takehara H, Sasagawa K, Ohta J. Brain-implantable needle-type CMOS imaging device enables multi-layer dissection of seizure calcium dynamics in the hippocampus. J Neural Eng 2024; 21:046022. [PMID: 38925109 DOI: 10.1088/1741-2552/ad5c03] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Accepted: 06/26/2024] [Indexed: 06/28/2024]
Abstract
Objective: Current neuronal imaging methods mostly use bulky lenses that either impede animal behavior or prohibit multi-depth imaging. To overcome these limitations, we developed a lightweight lensless biophotonic system for neuronal imaging, enabling compact and simultaneous visualization of multiple brain layers.Approach: Our developed 'CIS-NAIST' device integrates a micro-CMOS image sensor, thin-film fluorescence filter, micro-LEDs, and a needle-shaped flexible printed circuit. With this device, we monitored neuronal calcium dynamics during seizures across the different layers of the hippocampus and employed machine learning techniques for seizure classification and prediction.Main results: The CIS-NAIST device revealed distinct calcium activity patterns across the CA1, molecular interlayer, and dentate gyrus. Our findings indicated an elevated calcium amplitude activity specifically in the dentate gyrus compared to other layers. Then, leveraging the multi-layer data obtained from the device, we successfully classified seizure calcium activity and predicted seizure behavior using Long Short-Term Memory and Hidden Markov models.Significance: Taken together, our 'CIS-NAIST' device offers an effective and minimally invasive method of seizure monitoring that can help elucidate the mechanisms of temporal lobe epilepsy.
Collapse
Affiliation(s)
| | - Yasumi Ohta
- Division of Materials Science, Nara Institute of Science and Technology, Ikoma, Japan
| | - Paul R Regonia
- Department of Computer Science, University of the Philippines Diliman, Manila, The Philippines
| | - Virgil C G Castillo
- Division of Materials Science, Nara Institute of Science and Technology, Ikoma, Japan
| | - Junichiro Yoshimoto
- Department of Biomedical Data Science, Fujita Health University School of Medicine, Toyoake, Japan
| | - Hironari Takehara
- Division of Materials Science, Nara Institute of Science and Technology, Ikoma, Japan
| | - Kiyotaka Sasagawa
- Division of Materials Science, Nara Institute of Science and Technology, Ikoma, Japan
| | - Jun Ohta
- Division of Materials Science, Nara Institute of Science and Technology, Ikoma, Japan
| |
Collapse
|
16
|
Phil Brooks F, Davis HC, Wong-Campos JD, Cohen AE. Optical constraints on two-photon voltage imaging. NEUROPHOTONICS 2024; 11:035007. [PMID: 39139631 PMCID: PMC11321468 DOI: 10.1117/1.nph.11.3.035007] [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: 05/28/2024] [Revised: 07/23/2024] [Accepted: 07/25/2024] [Indexed: 08/15/2024]
Abstract
Significance Genetically encoded voltage indicators (GEVIs) are a valuable tool for studying neural circuits in vivo, but the relative merits and limitations of one-photon (1P) versus two-photon (2P) voltage imaging are not well characterized. Aim We consider the optical and biophysical constraints particular to 1P and 2P voltage imaging and compare the imaging properties of commonly used GEVIs under 1P and 2P excitation. Approach We measure the brightness and voltage sensitivity of voltage indicators from commonly used classes under 1P and 2P illumination. We also measure the decrease in fluorescence as a function of depth in the mouse brain. We develop a simple model of the number of measurable cells as a function of reporter properties, imaging parameters, and desired signal-to-noise ratio (SNR). We then discuss how the performance of voltage imaging would be affected by sensor improvements and by recently introduced advanced imaging modalities. Results Compared with 1P excitation, 2P excitation requires ∼ 10 4 -fold more illumination power per cell to produce similar photon count rates. For voltage imaging with JEDI-2P in the mouse cortex with a target SNR of 10 (spike height to baseline shot noise), a measurement bandwidth of 1 kHz, a thermally limited laser power of 200 mW, and an imaging depth of > 300 μ m , 2P voltage imaging using an 80-MHz source can record from no more than ∼ 12 neurons simultaneously. Conclusions Due to the stringent photon-count requirements of voltage imaging and the modest voltage sensitivity of existing reporters, 2P voltage imaging in vivo faces a stringent tradeoff between shot noise and tissue photodamage. 2P imaging of hundreds of neurons with high SNR at a depth of > 300 μ m will require either major improvements in 2P GEVIs or qualitatively new approaches to imaging.
Collapse
Affiliation(s)
- F. Phil Brooks
- Harvard University, Department of Chemistry and Chemical Biology, Cambridge, Massachusetts, United States
| | - Hunter C. Davis
- Harvard University, Department of Chemistry and Chemical Biology, Cambridge, Massachusetts, United States
| | - J. David Wong-Campos
- Harvard University, Department of Chemistry and Chemical Biology, Cambridge, Massachusetts, United States
| | - Adam E. Cohen
- Harvard University, Department of Chemistry and Chemical Biology, Cambridge, Massachusetts, United States
| |
Collapse
|
17
|
Chen X, Li J, Roy S, Ullah Z, Gu J, Huang H, Yu C, Wang X, Wang H, Zhang Y, Guo B. Development of Polymethine Dyes for NIR-II Fluorescence Imaging and Therapy. Adv Healthc Mater 2024; 13:e2304506. [PMID: 38441392 DOI: 10.1002/adhm.202304506] [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: 12/18/2023] [Revised: 02/29/2024] [Indexed: 03/16/2024]
Abstract
Fluorescence imaging in the second near-infrared window (NIR-II) is burgeoning because of its higher imaging fidelity in monitoring physiological and pathological processes than clinical visible/the second near-infrared window fluorescence imaging. Notably, the imaging fidelity is heavily dependent on fluorescence agents. So far, indocyanine green, one of the polymethine dyes, with good biocompatibility and renal clearance is the only dye approved by the Food and Drug Administration, but it shows relatively low NIR-II brightness. Importantly, tremendous efforts are devoted to synthesizing polymethine dyes for imaging preclinically and clinically. They have shown feasibility in the customization of structure and properties to fulfill various needs in imaging and therapy. Herein, a timely update on NIR-II polymethine dyes, with a special focus on molecular design strategies for fluorescent, photoacoustic, and multimodal imaging, is offered. Furthermore, the progress of polymethine dyes in sensing pathological biomarkers and even reporting drug release is illustrated. Moreover, the NIR-II fluorescence imaging-guided therapies with polymethine dyes are summarized regarding chemo-, photothermal, photodynamic, and multimodal approaches. In addition, artificial intelligence is pointed out for its potential to expedite dye development. This comprehensive review will inspire interest among a wide audience and offer a handbook for people with an interest in NIR-II polymethine dyes.
Collapse
Affiliation(s)
- Xin Chen
- School of Science, Shenzhen Key Laboratory of Flexible Printed Electronics Technology, Shenzhen Key Laboratory of Advanced Functional Carbon Materials Research and Comprehensive Application, Harbin Institute of Technology, Shenzhen, 518055, China
| | - Jieyan Li
- School of Science, Shenzhen Key Laboratory of Flexible Printed Electronics Technology, Shenzhen Key Laboratory of Advanced Functional Carbon Materials Research and Comprehensive Application, Harbin Institute of Technology, Shenzhen, 518055, China
| | - Shubham Roy
- School of Science, Shenzhen Key Laboratory of Flexible Printed Electronics Technology, Shenzhen Key Laboratory of Advanced Functional Carbon Materials Research and Comprehensive Application, Harbin Institute of Technology, Shenzhen, 518055, China
| | - Zia Ullah
- School of Science, Shenzhen Key Laboratory of Flexible Printed Electronics Technology, Shenzhen Key Laboratory of Advanced Functional Carbon Materials Research and Comprehensive Application, Harbin Institute of Technology, Shenzhen, 518055, China
| | - Jingsi Gu
- Education Center and Experiments and Innovations, Harbin Institute of Technology, Shenzhen, 518055, China
| | - Haiyan Huang
- School of Science, Shenzhen Key Laboratory of Flexible Printed Electronics Technology, Shenzhen Key Laboratory of Advanced Functional Carbon Materials Research and Comprehensive Application, Harbin Institute of Technology, Shenzhen, 518055, China
| | - Chen Yu
- School of Science, Shenzhen Key Laboratory of Flexible Printed Electronics Technology, Shenzhen Key Laboratory of Advanced Functional Carbon Materials Research and Comprehensive Application, Harbin Institute of Technology, Shenzhen, 518055, China
- College of Chemistry and Environmental Engineering, Shenzhen University, Shenzhen, 518060, China
| | - Xuejin Wang
- School of Science, Shenzhen Key Laboratory of Flexible Printed Electronics Technology, Shenzhen Key Laboratory of Advanced Functional Carbon Materials Research and Comprehensive Application, Harbin Institute of Technology, Shenzhen, 518055, China
| | - Han Wang
- School of Science, Shenzhen Key Laboratory of Flexible Printed Electronics Technology, Shenzhen Key Laboratory of Advanced Functional Carbon Materials Research and Comprehensive Application, Harbin Institute of Technology, Shenzhen, 518055, China
| | - Yinghe Zhang
- School of Science, Shenzhen Key Laboratory of Flexible Printed Electronics Technology, Shenzhen Key Laboratory of Advanced Functional Carbon Materials Research and Comprehensive Application, Harbin Institute of Technology, Shenzhen, 518055, China
| | - Bing Guo
- School of Science, Shenzhen Key Laboratory of Flexible Printed Electronics Technology, Shenzhen Key Laboratory of Advanced Functional Carbon Materials Research and Comprehensive Application, Harbin Institute of Technology, Shenzhen, 518055, China
| |
Collapse
|
18
|
Xiao S, Cunningham WJ, Kondabolu K, Lowet E, Moya MV, Mount RA, Ravasio C, Bortz E, Shaw D, Economo MN, Han X, Mertz J. Large-scale deep tissue voltage imaging with targeted-illumination confocal microscopy. Nat Methods 2024; 21:1094-1102. [PMID: 38840033 PMCID: PMC11500676 DOI: 10.1038/s41592-024-02275-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Accepted: 04/09/2024] [Indexed: 06/07/2024]
Abstract
Voltage imaging with cellular specificity has been made possible by advances in genetically encoded voltage indicators. However, the kilohertz rates required for voltage imaging lead to weak signals. Moreover, out-of-focus fluorescence and tissue scattering produce background that both undermines the signal-to-noise ratio and induces crosstalk between cells, making reliable in vivo imaging in densely labeled tissue highly challenging. We describe a microscope that combines the distinct advantages of targeted illumination and confocal gating while also maximizing signal detection efficiency. The resulting benefits in signal-to-noise ratio and crosstalk reduction are quantified experimentally and theoretically. Our microscope provides a versatile solution for enabling high-fidelity in vivo voltage imaging at large scales and penetration depths, which we demonstrate across a wide range of imaging conditions and different genetically encoded voltage indicator classes.
Collapse
Affiliation(s)
- Sheng Xiao
- Department of Biomedical Engineering, Boston University, Boston, MA, USA.
| | | | | | - Eric Lowet
- Department of Biomedical Engineering, Boston University, Boston, MA, USA
- Department of Neuroscience, Erasmus MC, Rotterdam, the Netherlands
| | - Maria V Moya
- Department of Biomedical Engineering, Boston University, Boston, MA, USA
| | - Rebecca A Mount
- Department of Biomedical Engineering, Boston University, Boston, MA, USA
| | - Cara Ravasio
- Department of Biomedical Engineering, Boston University, Boston, MA, USA
| | - Emma Bortz
- Department of Biomedical Engineering, Boston University, Boston, MA, USA
| | - Dana Shaw
- Graduate Program for Neuroscience, Boston University, Boston, MA, USA
- Neurophotonics Center, Boston University, Boston, MA, USA
| | - Michael N Economo
- Department of Biomedical Engineering, Boston University, Boston, MA, USA
- Neurophotonics Center, Boston University, Boston, MA, USA
| | - Xue Han
- Department of Biomedical Engineering, Boston University, Boston, MA, USA
- Neurophotonics Center, Boston University, Boston, MA, USA
| | - Jerome Mertz
- Department of Biomedical Engineering, Boston University, Boston, MA, USA
- Neurophotonics Center, Boston University, Boston, MA, USA
| |
Collapse
|
19
|
Targeted illumination confocal microscopy enables in vivo voltage imaging in thick tissue. Nat Methods 2024; 21:948-949. [PMID: 38840034 DOI: 10.1038/s41592-024-02276-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/07/2024]
|
20
|
Li Y. Differential behaviors of calcium-induced calcium release in one dimensional dendrite by Nernst-Planck equation, cable model and pure diffusion model. Cogn Neurodyn 2024; 18:1285-1305. [PMID: 38826668 PMCID: PMC11143177 DOI: 10.1007/s11571-023-09952-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Revised: 02/16/2023] [Accepted: 03/08/2023] [Indexed: 06/04/2024] Open
Abstract
The source and dynamics of calcium is the key factor that regulates dendritic integration. Apart from the voltage-gated and ligand-gated calcium influx, an important source of calcium is from inner store of endoplasmic reticulum with a regenerative process of calcium-induced calcium release (CICR). To trigger this process, inositol 1,4,5-trisphosphate (IP3) and calcium are needed to satisfy certain requirements. The aim of our paper is to investigate how the CICR depends on the dynamics of membrane potential. We utilize one dimensional dendritic model to calculate membrane potential by Nernst-Planck Equation (NPE) and cable model and Pure Diffusion (PD) model, computational simulations are carried out to inject the calcium influx by synaptic stimulation and to predict subsequent CICR and calcium wave propagation. Our results demonstrate that CICR initiation and calcium wave propagation have much difference between electro-diffusion process of NPE and cable model. We find that cable model has lower threshold of IP3 stimulation to trigger CICR but is more difficult for calcium propagation than NPE, PD model requires even higher threshold of IP3 to initiate CICR process and calcium duration is shorter than NPE; the regenerative calcium wave propagates with faster speed in NPE than that in cable model and in PD model. Our work addresses the important role of electro-diffusion dynamics of charged ions in regulating CICR process in dendritic structure; and provides theoretical predictions for neurological process which requires sustaining calcium for downstream signaling processes.
Collapse
Affiliation(s)
- Yinyun Li
- School of Systems Science, Beijing Normal University, Beijing, 100875 China
- Department of Mathematics and Statistics, Washington State University Vancouver, Vancouver, USA
| |
Collapse
|
21
|
Mandracchia B, Zheng C, Rajendran S, Liu W, Forghani P, Xu C, Jia S. High-speed optical imaging with sCMOS pixel reassignment. Nat Commun 2024; 15:4598. [PMID: 38816394 PMCID: PMC11139943 DOI: 10.1038/s41467-024-48987-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Accepted: 05/13/2024] [Indexed: 06/01/2024] Open
Abstract
Fluorescence microscopy has undergone rapid advancements, offering unprecedented visualization of biological events and shedding light on the intricate mechanisms governing living organisms. However, the exploration of rapid biological dynamics still poses a significant challenge due to the limitations of current digital camera architectures and the inherent compromise between imaging speed and other capabilities. Here, we introduce sHAPR, a high-speed acquisition technique that leverages the operating principles of sCMOS cameras to capture fast cellular and subcellular processes. sHAPR harnesses custom fiber optics to convert microscopy images into one-dimensional recordings, enabling acquisition at the maximum camera readout rate, typically between 25 and 250 kHz. We have demonstrated the utility of sHAPR with a variety of phantom and dynamic systems, including high-throughput flow cytometry, cardiomyocyte contraction, and neuronal calcium waves, using a standard epi-fluorescence microscope. sHAPR is highly adaptable and can be integrated into existing microscopy systems without requiring extensive platform modifications. This method pushes the boundaries of current fluorescence imaging capabilities, opening up new avenues for investigating high-speed biological phenomena.
Collapse
Affiliation(s)
- Biagio Mandracchia
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
- E.T.S.I. Telecomunicación, Universidad de Valladolid, Valladolid, Spain
| | - Corey Zheng
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Suraj Rajendran
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Wenhao Liu
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Parvin Forghani
- Department of Pediatrics, School of Medicine, Emory University, Atlanta, GA, USA
| | - Chunhui Xu
- Department of Pediatrics, School of Medicine, Emory University, Atlanta, GA, USA
- Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, USA
| | - Shu Jia
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA.
- Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, USA.
| |
Collapse
|
22
|
Zhang J, Newman J, Wang Z, Qian Y, Feliciano-Ramos P, Guo W, Honda T, Chen ZS, Linghu C, Etienne-Cummings R, Fossum E, Boyden E, Wilson M. Pixel-wise programmability enables dynamic high-SNR cameras for high-speed microscopy. Nat Commun 2024; 15:4480. [PMID: 38802338 PMCID: PMC11530699 DOI: 10.1038/s41467-024-48765-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2023] [Accepted: 04/30/2024] [Indexed: 05/29/2024] Open
Abstract
High-speed wide-field fluorescence microscopy has the potential to capture biological processes with exceptional spatiotemporal resolution. However, conventional cameras suffer from low signal-to-noise ratio at high frame rates, limiting their ability to detect faint fluorescent events. Here, we introduce an image sensor where each pixel has individually programmable sampling speed and phase, so that pixels can be arranged to simultaneously sample at high speed with a high signal-to-noise ratio. In high-speed voltage imaging experiments, our image sensor significantly increases the output signal-to-noise ratio compared to a low-noise scientific CMOS camera (~2-3 folds). This signal-to-noise ratio gain enables the detection of weak neuronal action potentials and subthreshold activities missed by the standard scientific CMOS cameras. Our camera with flexible pixel exposure configurations offers versatile sampling strategies to improve signal quality in various experimental conditions.
Collapse
Affiliation(s)
- Jie Zhang
- Picower Institute for Learning and Memory, MIT, Cambridge, MA, USA.
- Department of Brain and Cognitive Sciences, MIT, Cambridge, MA, USA.
| | - Jonathan Newman
- Picower Institute for Learning and Memory, MIT, Cambridge, MA, USA
- Department of Brain and Cognitive Sciences, MIT, Cambridge, MA, USA
| | - Zeguan Wang
- Department of Brain and Cognitive Sciences, MIT, Cambridge, MA, USA
- McGovern Institute for Brain Research, MIT, Cambridge, MA, USA
| | - Yong Qian
- Department of Brain and Cognitive Sciences, MIT, Cambridge, MA, USA
- McGovern Institute for Brain Research, MIT, Cambridge, MA, USA
| | - Pedro Feliciano-Ramos
- Picower Institute for Learning and Memory, MIT, Cambridge, MA, USA
- Department of Brain and Cognitive Sciences, MIT, Cambridge, MA, USA
| | - Wei Guo
- Picower Institute for Learning and Memory, MIT, Cambridge, MA, USA
- Department of Brain and Cognitive Sciences, MIT, Cambridge, MA, USA
| | - Takato Honda
- Picower Institute for Learning and Memory, MIT, Cambridge, MA, USA
- Department of Brain and Cognitive Sciences, MIT, Cambridge, MA, USA
| | - Zhe Sage Chen
- Department of Psychiatry, NYU Grossman School of Medicine, New York, NY, USA
| | - Changyang Linghu
- Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, MI, USA
| | - Ralph Etienne-Cummings
- Department of Electrical and Computer Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Eric Fossum
- Thayer School of Engineering, Dartmouth College, Hanover, NH, USA
| | - Edward Boyden
- Department of Brain and Cognitive Sciences, MIT, Cambridge, MA, USA
- McGovern Institute for Brain Research, MIT, Cambridge, MA, USA
| | - Matthew Wilson
- Picower Institute for Learning and Memory, MIT, Cambridge, MA, USA
- Department of Brain and Cognitive Sciences, MIT, Cambridge, MA, USA
| |
Collapse
|
23
|
Brooks FP, Davis HC, Wong-Campos JD, Cohen AE. Optical constraints on two-photon voltage imaging. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.11.18.567441. [PMID: 38014011 PMCID: PMC10680948 DOI: 10.1101/2023.11.18.567441] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2023]
Abstract
Significance Genetically encoded voltage indicators (GEVIs) are a valuable tool for studying neural circuits in vivo, but the relative merits and limitations of one-photon (1P) vs. two-photon (2P) voltage imaging are not well characterized. Aim We consider the optical and biophysical constraints particular to 1P and 2P voltage imaging and compare the imaging properties of commonly used GEVIs under 1P and 2P excitation. Approach We measure brightness and voltage sensitivity of voltage indicators from commonly used classes under 1P and 2P illumination. We also measure the decrease in fluorescence as a function of depth in mouse brain. We develop a simple model of the number of measurable cells as a function of reporter properties, imaging parameters, and desired signal-to-noise ratio (SNR). We then discuss how the performance of voltage imaging would be affected by sensor improvements and by recently introduced advanced imaging modalities. Results Compared to 1P excitation, 2P excitation requires ~104-fold more illumination power per cell to produce similar photon count rates. For voltage imaging with JEDI-2P in mouse cortex with a target SNR of 10 (spike height:baseline shot noise), a measurement bandwidth of 1 kHz, a thermally limited laser power of 200 mW, and an imaging depth of > 300 μm, 2P voltage imaging using an 80 MHz source can record from no more 12 cells simultaneously. Conclusions Due to the stringent photon-count requirements of voltage imaging and the modest voltage sensitivity of existing reporters, 2P voltage imaging in vivo faces a stringent tradeoff between shot noise and tissue photodamage. 2P imaging of hundreds of neurons with high SNR at depth > 300 μm will require either major improvements in 2P GEVIs or qualitatively new approaches to imaging.
Collapse
Affiliation(s)
- F Phil Brooks
- Department of Chemistry and Chemical Biology, Harvard University
| | - Hunter C Davis
- Department of Chemistry and Chemical Biology, Harvard University
| | | | - Adam E Cohen
- Department of Chemistry and Chemical Biology, Harvard University
| |
Collapse
|
24
|
Manley J, Lu S, Barber K, Demas J, Kim H, Meyer D, Traub FM, Vaziri A. Simultaneous, cortex-wide dynamics of up to 1 million neurons reveal unbounded scaling of dimensionality with neuron number. Neuron 2024; 112:1694-1709.e5. [PMID: 38452763 PMCID: PMC11098699 DOI: 10.1016/j.neuron.2024.02.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Revised: 05/18/2023] [Accepted: 02/14/2024] [Indexed: 03/09/2024]
Abstract
The brain's remarkable properties arise from the collective activity of millions of neurons. Widespread application of dimensionality reduction to multi-neuron recordings implies that neural dynamics can be approximated by low-dimensional "latent" signals reflecting neural computations. However, can such low-dimensional representations truly explain the vast range of brain activity, and if not, what is the appropriate resolution and scale of recording to capture them? Imaging neural activity at cellular resolution and near-simultaneously across the mouse cortex, we demonstrate an unbounded scaling of dimensionality with neuron number in populations up to 1 million neurons. Although half of the neural variance is contained within sixteen dimensions correlated with behavior, our discovered scaling of dimensionality corresponds to an ever-increasing number of neuronal ensembles without immediate behavioral or sensory correlates. The activity patterns underlying these higher dimensions are fine grained and cortex wide, highlighting that large-scale, cellular-resolution recording is required to uncover the full substrates of neuronal computations.
Collapse
Affiliation(s)
- Jason Manley
- Laboratory of Neurotechnology and Biophysics, The Rockefeller University, New York, NY 10065, USA; The Kavli Neural Systems Institute, The Rockefeller University, New York, NY 10065, USA
| | - Sihao Lu
- Laboratory of Neurotechnology and Biophysics, The Rockefeller University, New York, NY 10065, USA
| | - Kevin Barber
- Laboratory of Neurotechnology and Biophysics, The Rockefeller University, New York, NY 10065, USA
| | - Jeffrey Demas
- Laboratory of Neurotechnology and Biophysics, The Rockefeller University, New York, NY 10065, USA; The Kavli Neural Systems Institute, The Rockefeller University, New York, NY 10065, USA
| | - Hyewon Kim
- Laboratory of Neurotechnology and Biophysics, The Rockefeller University, New York, NY 10065, USA
| | - David Meyer
- Laboratory of Neurotechnology and Biophysics, The Rockefeller University, New York, NY 10065, USA
| | - Francisca Martínez Traub
- Laboratory of Neurotechnology and Biophysics, The Rockefeller University, New York, NY 10065, USA
| | - Alipasha Vaziri
- Laboratory of Neurotechnology and Biophysics, The Rockefeller University, New York, NY 10065, USA; The Kavli Neural Systems Institute, The Rockefeller University, New York, NY 10065, USA.
| |
Collapse
|
25
|
Zhang J, Newman J, Wang Z, Qian Y, Feliciano-Ramos P, Guo W, Honda T, Chen ZS, Linghu C, Etienne-Cummings R, Fossum E, Boyden E, Wilson M. Pixel-wise programmability enables dynamic high-SNR cameras for high-speed microscopy. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.06.27.546748. [PMID: 37425952 PMCID: PMC10327006 DOI: 10.1101/2023.06.27.546748] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/11/2023]
Abstract
High-speed wide-field fluorescence microscopy has the potential to capture biological processes with exceptional spatiotemporal resolution. However, conventional cameras suffer from low signal-to-noise ratio at high frame rates, limiting their ability to detect faint fluorescent events. Here, we introduce an image sensor where each pixel has individually programmable sampling speed and phase, so that pixels can be arranged to simultaneously sample at high speed with a high signal-to-noise ratio. In high-speed voltage imaging experiments, our image sensor significantly increases the output signal-to-noise ratio compared to a low-noise scientific CMOS camera (~2-3 folds). This signal-to-noise ratio gain enables the detection of weak neuronal action potentials and subthreshold activities missed by the standard scientific CMOS cameras. Our camera with flexible pixel exposure configurations offers versatile sampling strategies to improve signal quality in various experimental conditions.
Collapse
Affiliation(s)
- Jie Zhang
- Picower Institute for Learning and Memory, MIT, Cambridge, MA, USA
- Department of Brain and Cognitive Sciences, MIT, Cambridge, MA, USA
| | - Jonathan Newman
- Picower Institute for Learning and Memory, MIT, Cambridge, MA, USA
- Department of Brain and Cognitive Sciences, MIT, Cambridge, MA, USA
| | - Zeguan Wang
- Department of Brain and Cognitive Sciences, MIT, Cambridge, MA, USA
- McGovern Institute for Brain Research, MIT, Cambridge, MA, USA
| | - Yong Qian
- Department of Brain and Cognitive Sciences, MIT, Cambridge, MA, USA
- McGovern Institute for Brain Research, MIT, Cambridge, MA, USA
| | - Pedro Feliciano-Ramos
- Picower Institute for Learning and Memory, MIT, Cambridge, MA, USA
- Department of Brain and Cognitive Sciences, MIT, Cambridge, MA, USA
| | - Wei Guo
- Picower Institute for Learning and Memory, MIT, Cambridge, MA, USA
- Department of Brain and Cognitive Sciences, MIT, Cambridge, MA, USA
| | - Takato Honda
- Picower Institute for Learning and Memory, MIT, Cambridge, MA, USA
- Department of Brain and Cognitive Sciences, MIT, Cambridge, MA, USA
| | - Zhe Sage Chen
- Department of Psychiatry, NYU Grossman School of Medicine, New York, NY, USA
| | - Changyang Linghu
- Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, MI, USA
| | - Ralph Etienne-Cummings
- Department of Electrical and Computer Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Eric Fossum
- Thayer School of Engineering, Dartmouth College, Hanover, NH, USA
| | - Edward Boyden
- Department of Brain and Cognitive Sciences, MIT, Cambridge, MA, USA
- McGovern Institute for Brain Research, MIT, Cambridge, MA, USA
| | - Matthew Wilson
- Picower Institute for Learning and Memory, MIT, Cambridge, MA, USA
- Department of Brain and Cognitive Sciences, MIT, Cambridge, MA, USA
| |
Collapse
|
26
|
Shang CF, Wang YF, Zhao MT, Fan QX, Zhao S, Qian Y, Xu SJ, Mu Y, Hao J, Du JL. Real-time analysis of large-scale neuronal imaging enables closed-loop investigation of neural dynamics. Nat Neurosci 2024; 27:1014-1018. [PMID: 38467902 DOI: 10.1038/s41593-024-01595-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Accepted: 02/07/2024] [Indexed: 03/13/2024]
Abstract
Large-scale imaging of neuronal activities is crucial for understanding brain functions. However, it is challenging to analyze large-scale imaging data in real time, preventing closed-loop investigation of neural circuitry. Here we develop a real-time analysis system with a field programmable gate array-graphics processing unit design for an up to 500-megabyte-per-second image stream. Adapted to whole-brain imaging of awake larval zebrafish, the system timely extracts activity from up to 100,000 neurons and enables closed-loop perturbations of neural dynamics.
Collapse
Affiliation(s)
- Chun-Feng Shang
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
- Guangdong-Hongkong-Macau Institute of CNS Regeneration, Ministry of Education CNS Regeneration Collaborative Joint Laboratory, Jinan University, Guangzhou, China
- Shenzhen Institute of Neuroscience, Shenzhen, China
| | - Yu-Fan Wang
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Mei-Ting Zhao
- Institute of Automation, Chinese Academy of Sciences, Beijing, China
- Guangdong Institute of Artificial Intelligence and Advanced Computing, Guangzhou, China
| | - Qiu-Xiang Fan
- Institute of Automation, Chinese Academy of Sciences, Beijing, China
- Guangdong Institute of Artificial Intelligence and Advanced Computing, Guangzhou, China
| | - Shan Zhao
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Yu Qian
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| | - Sheng-Jin Xu
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Yu Mu
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China.
- University of Chinese Academy of Sciences, Beijing, China.
| | - Jie Hao
- University of Chinese Academy of Sciences, Beijing, China.
- Institute of Automation, Chinese Academy of Sciences, Beijing, China.
- Guangdong Institute of Artificial Intelligence and Advanced Computing, Guangzhou, China.
| | - Jiu-Lin Du
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China.
- University of Chinese Academy of Sciences, Beijing, China.
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China.
| |
Collapse
|
27
|
Wu Y, Xu Z, Liang S, Wang L, Wang M, Jia H, Chen X, Zhao Z, Liao X. NeuroSeg-III: efficient neuron segmentation in two-photon Ca 2+ imaging data using self-supervised learning. BIOMEDICAL OPTICS EXPRESS 2024; 15:2910-2925. [PMID: 38855703 PMCID: PMC11161377 DOI: 10.1364/boe.521478] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Revised: 03/28/2024] [Accepted: 03/30/2024] [Indexed: 06/11/2024]
Abstract
Two-photon Ca2+ imaging technology increasingly plays an essential role in neuroscience research. However, the requirement for extensive professional annotation poses a significant challenge to improving the performance of neuron segmentation models. Here, we present NeuroSeg-III, an innovative self-supervised learning approach specifically designed to achieve fast and precise segmentation of neurons in imaging data. This approach consists of two modules: a self-supervised pre-training network and a segmentation network. After pre-training the encoder of the segmentation network via a self-supervised learning method without any annotated data, we only need to fine-tune the segmentation network with a small amount of annotated data. The segmentation network is designed with YOLOv8s, FasterNet, efficient multi-scale attention mechanism (EMA), and bi-directional feature pyramid network (BiFPN), which enhanced the model's segmentation accuracy while reducing the computational cost and parameters. The generalization of our approach was validated across different Ca2+ indicators and scales of imaging data. Significantly, the proposed neuron segmentation approach exhibits exceptional speed and accuracy, surpassing the current state-of-the-art benchmarks when evaluated using a publicly available dataset. The results underscore the effectiveness of NeuroSeg-III, with employing an efficient training strategy tailored for two-photon Ca2+ imaging data and delivering remarkable precision in neuron segmentation.
Collapse
Affiliation(s)
- Yukun Wu
- Guangxi Key Laboratory of Special Biomedicine and Advanced Institute for Brain and Intelligence, School of Medicine, Guangxi University, Nanning 530004, China
| | - Zhehao Xu
- Center for Neurointelligence, School of Medicine, Chongqing University, Chongqing 400030, China
| | - Shanshan Liang
- Brain Research Center, State Key Laboratory of Trauma and Chemical Poisoning, Third Military Medical University, Chongqing 400038, China
| | - Lukang Wang
- Brain Research Center, State Key Laboratory of Trauma and Chemical Poisoning, Third Military Medical University, Chongqing 400038, China
| | - Meng Wang
- Center for Neurointelligence, School of Medicine, Chongqing University, Chongqing 400030, China
| | - Hongbo Jia
- Guangxi Key Laboratory of Special Biomedicine and Advanced Institute for Brain and Intelligence, School of Medicine, Guangxi University, Nanning 530004, China
- Brain Research Instrument Innovation Center, Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou 215163, Jiangsu, China
| | - Xiaowei Chen
- Guangxi Key Laboratory of Special Biomedicine and Advanced Institute for Brain and Intelligence, School of Medicine, Guangxi University, Nanning 530004, China
- Chongqing Institute for Brain and Intelligence, Guangyang Bay Laboratory, Chongqing 400064, China
| | - Zhikai Zhao
- Center for Neurointelligence, School of Medicine, Chongqing University, Chongqing 400030, China
| | - Xiang Liao
- Center for Neurointelligence, School of Medicine, Chongqing University, Chongqing 400030, China
| |
Collapse
|
28
|
Akbari N, Tatarsky RL, Kolkman KE, Fetcho JR, Xu C, Bass AH. Label-free, whole-brain in vivo mapping in an adult vertebrate with third harmonic generation microscopy. J Comp Neurol 2024; 532:e25614. [PMID: 38616537 PMCID: PMC11069316 DOI: 10.1002/cne.25614] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Revised: 02/16/2024] [Accepted: 03/24/2024] [Indexed: 04/16/2024]
Abstract
Comprehensive understanding of interconnected networks within the brain requires access to high resolution information within large field of views and over time. Currently, methods that enable mapping structural changes of the entire brain in vivo are extremely limited. Third harmonic generation (THG) can resolve myelinated structures, blood vessels, and cell bodies throughout the brain without the need for any exogenous labeling. Together with deep penetration of long wavelengths, this enables in vivo brain-mapping of large fractions of the brain in small animals and over time. Here, we demonstrate that THG microscopy allows non-invasive label-free mapping of the entire brain of an adult vertebrate, Danionella dracula, which is a miniature species of cyprinid fish. We show this capability in multiple brain regions and in particular the identification of major commissural fiber bundles in the midbrain and the hindbrain. These features provide readily discernable landmarks for navigation and identification of regional-specific neuronal groups and even single neurons during in vivo experiments. We further show how this label-free technique can easily be coupled with fluorescence microscopy and used as a comparative tool for studies of other species with similar body features to Danionella, such as zebrafish (Danio rerio) and tetras (Trochilocharax ornatus). This new evidence, building on previous studies, demonstrates how small size and relative transparency, combined with the unique capabilities of THG microscopy, can enable label-free access to the entire adult vertebrate brain.
Collapse
Affiliation(s)
- Najva Akbari
- School of Applied and Engineering Physics, Cornell University, Ithaca, NY USA 14850
- Present address: Department of Biology, Stanford University, Stanford, CA USA 94305
| | - Rose L. Tatarsky
- Department of Neurobiology and Behavior, Cornell University, Ithaca, NY USA 14850
| | - Kristine E. Kolkman
- Department of Neurobiology and Behavior, Cornell University, Ithaca, NY USA 14850
| | - Joseph R. Fetcho
- Department of Neurobiology and Behavior, Cornell University, Ithaca, NY USA 14850
| | - Chris Xu
- School of Applied and Engineering Physics, Cornell University, Ithaca, NY USA 14850
| | - Andrew H. Bass
- Department of Neurobiology and Behavior, Cornell University, Ithaca, NY USA 14850
| |
Collapse
|
29
|
Branning JM, Faughnan KA, Tomson AA, Bell GJ, Isbell SM, DeGroot A, Jameson L, Kilroy K, Smith M, Smith R, Mottel L, Branning EG, Worrall Z, Anderson F, Panditaradyula A, Yang W, Abdelmalek J, Brake J, Cash KJ. Multifunction fluorescence open source in vivo/in vitro imaging system (openIVIS). PLoS One 2024; 19:e0299875. [PMID: 38498588 PMCID: PMC10947658 DOI: 10.1371/journal.pone.0299875] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Accepted: 02/18/2024] [Indexed: 03/20/2024] Open
Abstract
The widespread availability and diversity of open-source microcontrollers paired with off-the-shelf electronics and 3D printed technology has led to the creation of a wide range of low-cost scientific instruments, including microscopes, spectrometers, sensors, data loggers, and other tools that can be used for research, education, and experimentation. These devices can be used to explore a wide range of scientific topics, from biology and chemistry to physics and engineering. In this study, we designed and built a multifunction fluorescent open source in vivo/in vitro imaging system (openIVIS) system that integrates a Raspberry Pi with commercial cameras and LEDs with 3D printed structures combined with an acrylic housing. Our openIVIS provides three excitation wavelengths of 460 nm, 520 nm, and 630 nm integrated with Python control software to enable fluorescent measurements across the full visible light spectrum. To demonstrate the potential applications of our system, we tested its performance against a diverse set of experiments including laboratory assays (measuring fluorescent dyes, using optical nanosensors, and DNA gel electrophoresis) to potentially fieldable applications (plant and mineral imaging). We also tested the potential use for a high school biology environment by imaging small animals and tracking their development over the course of ten days. Our system demonstrated its ability to measure a wide dynamic range fluorescent response from millimolar to picomolar concentrations in the same sample while measuring responses across visible wavelengths. These results demonstrate the power and flexibility of open-source hardware and software and how it can be integrated with customizable manufacturing to create low-cost scientific instruments with a wide range of applications. Our study provides a promising model for the development of low-cost instruments that can be used in both research and education.
Collapse
Affiliation(s)
- John M. Branning
- Quantitative Biosciences and Engineering, Colorado School of Mines, Golden, Colorado, United States of America
- The MITRE Corporation, Bedford, Massachusetts, United States of America
| | - Kealy A. Faughnan
- Chemical and Biological Engineering Department, Colorado School of Mines, Golden, Colorado, United States of America
| | - Austin A. Tomson
- Mechanical Engineering, Colorado School of Mines, Golden, Colorado, United States of America
| | - Grant J. Bell
- Quantitative Biosciences and Engineering, Colorado School of Mines, Golden, Colorado, United States of America
| | - Sydney M. Isbell
- Chemical and Biological Engineering Department, Colorado School of Mines, Golden, Colorado, United States of America
| | - Allen DeGroot
- Electrical Engineering, Colorado School of Mines, Golden, Colorado, United States of America
| | - Lydia Jameson
- Electrical Engineering, Colorado School of Mines, Golden, Colorado, United States of America
| | - Kramer Kilroy
- Mechanical Engineering, Colorado School of Mines, Golden, Colorado, United States of America
| | - Michael Smith
- Mechanical Engineering, Colorado School of Mines, Golden, Colorado, United States of America
| | - Robert Smith
- Electrical Engineering, Colorado School of Mines, Golden, Colorado, United States of America
| | - Landon Mottel
- Arvada West High School, Arvada, Colorado, United States of America
| | - Elizabeth G. Branning
- Colorado Early Colleges Castle Rock, Castle Rock, Colorado, United States of America
| | - Zoe Worrall
- Department of Engineering, Harvey Mudd College, Claremont, California, United States of America
| | - Frances Anderson
- Department of Engineering, Harvey Mudd College, Claremont, California, United States of America
| | - Ashrit Panditaradyula
- Department of Engineering, Harvey Mudd College, Claremont, California, United States of America
| | - William Yang
- Department of Engineering, Harvey Mudd College, Claremont, California, United States of America
| | - Joseph Abdelmalek
- Department of Engineering, Harvey Mudd College, Claremont, California, United States of America
| | - Joshua Brake
- Department of Engineering, Harvey Mudd College, Claremont, California, United States of America
| | - Kevin J. Cash
- Quantitative Biosciences and Engineering, Colorado School of Mines, Golden, Colorado, United States of America
- Chemical and Biological Engineering Department, Colorado School of Mines, Golden, Colorado, United States of America
| |
Collapse
|
30
|
Manley J, Vaziri A. Whole-brain neural substrates of behavioral variability in the larval zebrafish. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.03.583208. [PMID: 38496592 PMCID: PMC10942351 DOI: 10.1101/2024.03.03.583208] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/19/2024]
Abstract
Animals engaged in naturalistic behavior can exhibit a large degree of behavioral variability even under sensory invariant conditions. Such behavioral variability can include not only variations of the same behavior, but also variability across qualitatively different behaviors driven by divergent cognitive states, such as fight-or-flight decisions. However, the neural circuit mechanisms that generate such divergent behaviors across trials are not well understood. To investigate this question, here we studied the visual-evoked responses of larval zebrafish to moving objects of various sizes, which we found exhibited highly variable and divergent responses across repetitions of the same stimulus. Given that the neuronal circuits underlying such behaviors span sensory, motor, and other brain areas, we built a novel Fourier light field microscope which enables high-resolution, whole-brain imaging of larval zebrafish during behavior. This enabled us to screen for neural loci which exhibited activity patterns correlated with behavioral variability. We found that despite the highly variable activity of single neurons, visual stimuli were robustly encoded at the population level, and the visual-encoding dimensions of neural activity did not explain behavioral variability. This robustness despite apparent single neuron variability was due to the multi-dimensional geometry of the neuronal population dynamics: almost all neural dimensions that were variable across individual trials, i.e. the "noise" modes, were orthogonal to those encoding for sensory information. Investigating this neuronal variability further, we identified two sparsely-distributed, brain-wide neuronal populations whose pre-motor activity predicted whether the larva would respond to a stimulus and, if so, which direction it would turn on a single-trial level. These populations predicted single-trial behavior seconds before stimulus onset, indicating they encoded time-varying internal modulating behavior, perhaps organizing behavior over longer timescales or enabling flexible behavior routines dependent on the animal's internal state. Our results provide the first whole-brain confirmation that sensory, motor, and internal variables are encoded in a highly mixed fashion throughout the brain and demonstrate that de-mixing each of these components at the neuronal population level is critical to understanding the mechanisms underlying the brain's remarkable flexibility and robustness.
Collapse
Affiliation(s)
- Jason Manley
- Laboratory of Neurotechnology and Biophysics, The Rockefeller University, New York, NY 10065, USA
- The Kavli Neural Systems Institute, The Rockefeller University, New York, NY 10065, USA
| | - Alipasha Vaziri
- Laboratory of Neurotechnology and Biophysics, The Rockefeller University, New York, NY 10065, USA
- The Kavli Neural Systems Institute, The Rockefeller University, New York, NY 10065, USA
| |
Collapse
|
31
|
Pimenta S, Freitas JR, Correia JH. Flexible neural probes: a review of the current advantages, drawbacks, and future demands. J Zhejiang Univ Sci B 2024; 25:153-167. [PMID: 38303498 PMCID: PMC10835206 DOI: 10.1631/jzus.b2300337] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Accepted: 08/04/2023] [Indexed: 02/03/2024]
Abstract
Brain diseases affect millions of people and have a huge social and economic impact. The use of neural probes for studies in animals has been the main approach to increasing knowledge about neural network functioning. Ultimately, neuroscientists are trying to develop new and more effective therapeutic approaches to treating neurological disorders. The implementation of neural probes with multifunctionalities (electrical, optical, and fluidic interactions) has been increasing in the last few years, leading to the creation of devices with high temporal and spatial resolution. Increasing the applicability of, and elements integrated into, neural probes has also led to the necessity to create flexible interfaces, reducing neural tissue damage during probe implantation and increasing the quality of neural acquisition data. In this paper, we review the fabrication, characterization, and validation of several types of flexible neural probes, exploring the main advantages and drawbacks of these devices. Finally, future developments and applications are covered. Overall, this review aims to present the currently available flexible devices and future appropriate avenues for development as possible guidance for future engineered devices.
Collapse
Affiliation(s)
- Sara Pimenta
- CMEMS-UMinho, University of Minho, Guimares 4800-058, Portugal.
- LABBELS-Associate Laboratory, Braga/Guimares, Portugal.
| | - Joo R Freitas
- CMEMS-UMinho, University of Minho, Guimares 4800-058, Portugal
| | - Jos H Correia
- CMEMS-UMinho, University of Minho, Guimares 4800-058, Portugal
- LABBELS-Associate Laboratory, Braga/Guimares, Portugal
| |
Collapse
|
32
|
Liao Z, Gonzalez KC, Li DM, Yang CM, Holder D, McClain NE, Zhang G, Evans SW, Chavarha M, Yi J, Makinson CD, Lin MZ, Losonczy A, Negrean A. Functional architecture of intracellular oscillations in hippocampal dendrites. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.12.579750. [PMID: 38405778 PMCID: PMC10888786 DOI: 10.1101/2024.02.12.579750] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/27/2024]
Abstract
Fast electrical signaling in dendrites is central to neural computations that support adaptive behaviors. Conventional techniques lack temporal and spatial resolution and the ability to track underlying membrane potential dynamics present across the complex three-dimensional dendritic arbor in vivo. Here, we perform fast two-photon imaging of dendritic and somatic membrane potential dynamics in single pyramidal cells in the CA1 region of the mouse hippocampus during awake behavior. We study the dynamics of subthreshold membrane potential and suprathreshold dendritic events throughout the dendritic arbor in vivo by combining voltage imaging with simultaneous local field potential recording, post hoc morphological reconstruction, and a spatial navigation task. We systematically quantify the modulation of local event rates by locomotion in distinct dendritic regions and report an advancing gradient of dendritic theta phase along the basal-tuft axis, then describe a predominant hyperpolarization of the dendritic arbor during sharp-wave ripples. Finally, we find spatial tuning of dendritic representations dynamically reorganizes following place field formation. Our data reveal how the organization of electrical signaling in dendrites maps onto the anatomy of the dendritic tree across behavior, oscillatory network, and functional cell states.
Collapse
Affiliation(s)
- Zhenrui Liao
- Department of Neuroscience, Columbia University, New York, United States
- Mortimer B. Zuckerman Mind Brain Behavior Institute, Columbia University, New York, United States
| | - Kevin C. Gonzalez
- Department of Neuroscience, Columbia University, New York, United States
- Mortimer B. Zuckerman Mind Brain Behavior Institute, Columbia University, New York, United States
| | - Deborah M. Li
- Department of Neuroscience, Columbia University, New York, United States
- Mortimer B. Zuckerman Mind Brain Behavior Institute, Columbia University, New York, United States
| | - Catalina M. Yang
- Department of Neuroscience, Columbia University, New York, United States
- Mortimer B. Zuckerman Mind Brain Behavior Institute, Columbia University, New York, United States
| | - Donald Holder
- Department of Neuroscience, Columbia University, New York, United States
- Mortimer B. Zuckerman Mind Brain Behavior Institute, Columbia University, New York, United States
| | - Natalie E. McClain
- Department of Neuroscience, Columbia University, New York, United States
- Mortimer B. Zuckerman Mind Brain Behavior Institute, Columbia University, New York, United States
| | - Guofeng Zhang
- Department of Neurobiology, Stanford University, Stanford, United States
| | - Stephen W. Evans
- Department of Neurobiology, Stanford University, Stanford, United States
| | - Mariya Chavarha
- Department of Bioengineering, Stanford University, Stanford, United States
| | - Jane Yi
- Department of Neuroscience, Columbia University, New York, United States
- Department of Neurology, Columbia University, New York, United States
| | - Christopher D. Makinson
- Department of Neuroscience, Columbia University, New York, United States
- Department of Neurology, Columbia University, New York, United States
| | - Michael Z. Lin
- Department of Neurobiology, Stanford University, Stanford, United States
- Department of Bioengineering, Stanford University, Stanford, United States
| | - Attila Losonczy
- Department of Neuroscience, Columbia University, New York, United States
- Mortimer B. Zuckerman Mind Brain Behavior Institute, Columbia University, New York, United States
- Kavli Institute for Brain Science, Columbia University, New York, United States
| | - Adrian Negrean
- Department of Neuroscience, Columbia University, New York, United States
- Mortimer B. Zuckerman Mind Brain Behavior Institute, Columbia University, New York, United States
| |
Collapse
|
33
|
Wu J, Chen Y, Veeraraghavan A, Seidemann E, Robinson JT. Mesoscopic calcium imaging in a head-unrestrained male non-human primate using a lensless microscope. Nat Commun 2024; 15:1271. [PMID: 38341403 PMCID: PMC10858944 DOI: 10.1038/s41467-024-45417-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Accepted: 01/22/2024] [Indexed: 02/12/2024] Open
Abstract
Mesoscopic calcium imaging enables studies of cell-type specific neural activity over large areas. A growing body of literature suggests that neural activity can be different when animals are free to move compared to when they are restrained. Unfortunately, existing systems for imaging calcium dynamics over large areas in non-human primates (NHPs) are table-top devices that require restraint of the animal's head. Here, we demonstrate an imaging device capable of imaging mesoscale calcium activity in a head-unrestrained male non-human primate. We successfully miniaturize our system by replacing lenses with an optical mask and computational algorithms. The resulting lensless microscope can fit comfortably on an NHP, allowing its head to move freely while imaging. We are able to measure orientation columns maps over a 20 mm2 field-of-view in a head-unrestrained macaque. Our work establishes mesoscopic imaging using a lensless microscope as a powerful approach for studying neural activity under more naturalistic conditions.
Collapse
Affiliation(s)
- Jimin Wu
- Department of Bioengineering, Rice University, 6100 Main Street, Houston, TX, 77005, USA
| | - Yuzhi Chen
- Department of Neuroscience, University of Texas at Austin, 100 E 24th St., Austin, TX, 78712, USA
- Department of Psychology, University of Texas at Austin, 108 E Dean Keeton St., Austin, TX, 78712, USA
| | - Ashok Veeraraghavan
- Department of Electrical and Computer Engineering, Rice University, 6100 Main Street, Houston, TX, 77005, USA
- Department of Computer Science, Rice University, 6100 Main Street, Houston, TX, 77005, USA
| | - Eyal Seidemann
- Department of Neuroscience, University of Texas at Austin, 100 E 24th St., Austin, TX, 78712, USA.
- Department of Psychology, University of Texas at Austin, 108 E Dean Keeton St., Austin, TX, 78712, USA.
| | - Jacob T Robinson
- Department of Bioengineering, Rice University, 6100 Main Street, Houston, TX, 77005, USA.
- Department of Electrical and Computer Engineering, Rice University, 6100 Main Street, Houston, TX, 77005, USA.
- Department of Neuroscience, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, USA.
| |
Collapse
|
34
|
Yang Y, Jiang Q, Zhang F. Nanocrystals for Deep-Tissue In Vivo Luminescence Imaging in the Near-Infrared Region. Chem Rev 2024; 124:554-628. [PMID: 37991799 DOI: 10.1021/acs.chemrev.3c00506] [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: 11/23/2023]
Abstract
In vivo imaging technologies have emerged as a powerful tool for both fundamental research and clinical practice. In particular, luminescence imaging in the tissue-transparent near-infrared (NIR, 700-1700 nm) region offers tremendous potential for visualizing biological architectures and pathophysiological events in living subjects with deep tissue penetration and high imaging contrast owing to the reduced light-tissue interactions of absorption, scattering, and autofluorescence. The distinctive quantum effects of nanocrystals have been harnessed to achieve exceptional photophysical properties, establishing them as a promising category of luminescent probes. In this comprehensive review, the interactions between light and biological tissues, as well as the advantages of NIR light for in vivo luminescence imaging, are initially elaborated. Subsequently, we focus on achieving deep tissue penetration and improved imaging contrast by optimizing the performance of nanocrystal fluorophores. The ingenious design strategies of NIR nanocrystal probes are discussed, along with their respective biomedical applications in versatile in vivo luminescence imaging modalities. Finally, thought-provoking reflections on the challenges and prospects for future clinical translation of nanocrystal-based in vivo luminescence imaging in the NIR region are wisely provided.
Collapse
Affiliation(s)
- Yang Yang
- College of Energy Materials and Chemistry, State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, Inner Mongolia University, Hohhot 010021, China
| | - Qunying Jiang
- College of Energy Materials and Chemistry, State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, Inner Mongolia University, Hohhot 010021, China
| | - Fan Zhang
- College of Energy Materials and Chemistry, State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, Inner Mongolia University, Hohhot 010021, China
- Department of Chemistry, State Key Laboratory of Molecular Engineering of Polymers, Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials, Fudan University, Shanghai 200433, China
| |
Collapse
|
35
|
Manley J, Demas J, Kim H, Traub FM, Vaziri A. Simultaneous, cortex-wide and cellular-resolution neuronal population dynamics reveal an unbounded scaling of dimensionality with neuron number. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.15.575721. [PMID: 38293036 PMCID: PMC10827059 DOI: 10.1101/2024.01.15.575721] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/01/2024]
Abstract
The brain's remarkable properties arise from collective activity of millions of neurons. Widespread application of dimensionality reduction to multi-neuron recordings implies that neural dynamics can be approximated by low-dimensional "latent" signals reflecting neural computations. However, what would be the biological utility of such a redundant and metabolically costly encoding scheme and what is the appropriate resolution and scale of neural recording to understand brain function? Imaging the activity of one million neurons at cellular resolution and near-simultaneously across mouse cortex, we demonstrate an unbounded scaling of dimensionality with neuron number. While half of the neural variance lies within sixteen behavior-related dimensions, we find this unbounded scaling of dimensionality to correspond to an ever-increasing number of internal variables without immediate behavioral correlates. The activity patterns underlying these higher dimensions are fine-grained and cortex-wide, highlighting that large-scale recording is required to uncover the full neural substrates of internal and potentially cognitive processes.
Collapse
Affiliation(s)
- Jason Manley
- Laboratory of Neurotechnology and Biophysics, The Rockefeller University, New York, NY 10065, USA
- The Kavli Neural Systems Institute, The Rockefeller University, New York, NY 10065, USA
| | - Jeffrey Demas
- Laboratory of Neurotechnology and Biophysics, The Rockefeller University, New York, NY 10065, USA
- The Kavli Neural Systems Institute, The Rockefeller University, New York, NY 10065, USA
| | - Hyewon Kim
- Laboratory of Neurotechnology and Biophysics, The Rockefeller University, New York, NY 10065, USA
| | - Francisca Martínez Traub
- Laboratory of Neurotechnology and Biophysics, The Rockefeller University, New York, NY 10065, USA
| | - Alipasha Vaziri
- Laboratory of Neurotechnology and Biophysics, The Rockefeller University, New York, NY 10065, USA
- The Kavli Neural Systems Institute, The Rockefeller University, New York, NY 10065, USA
- Lead Contact
| |
Collapse
|
36
|
Singh RK, Nayak NP, Behl T, Arora R, Anwer MK, Gulati M, Bungau SG, Brisc MC. Exploring the Intersection of Geophysics and Diagnostic Imaging in the Health Sciences. Diagnostics (Basel) 2024; 14:139. [PMID: 38248016 PMCID: PMC11154438 DOI: 10.3390/diagnostics14020139] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Revised: 01/03/2024] [Accepted: 01/05/2024] [Indexed: 01/23/2024] Open
Abstract
To develop diagnostic imaging approaches, this paper emphasizes the transformational potential of merging geophysics with health sciences. Diagnostic imaging technology improvements have transformed the health sciences by enabling earlier and more precise disease identification, individualized therapy, and improved patient care. This review article examines the connection between geophysics and diagnostic imaging in the field of health sciences. Geophysics, which is typically used to explore Earth's subsurface, has provided new uses of its methodology in the medical field, providing innovative solutions to pressing medical problems. The article examines the different geophysical techniques like electrical imaging, seismic imaging, and geophysics and their corresponding imaging techniques used in health sciences like tomography, magnetic resonance imaging, ultrasound imaging, etc. The examination includes the description, similarities, differences, and challenges associated with these techniques and how modified geophysical techniques can be used in imaging methods in health sciences. Examining the progression of each method from geophysics to medical imaging and its contributions to illness diagnosis, treatment planning, and monitoring are highlighted. Also, the utilization of geophysical data analysis techniques like signal processing and inversion techniques in image processing in health sciences has been briefly explained, along with different mathematical and computational tools in geophysics and how they can be implemented for image processing in health sciences. The key findings include the development of machine learning and artificial intelligence in geophysics-driven medical imaging, demonstrating the revolutionary effects of data-driven methods on precision, speed, and predictive modeling.
Collapse
Affiliation(s)
- Rahul Kumar Singh
- Energy Cluster, University of Petroleum and Energy Studies, Dehradun 248007, Uttarakhand, India; (R.K.S.); (N.P.N.)
| | - Nirlipta Priyadarshini Nayak
- Energy Cluster, University of Petroleum and Energy Studies, Dehradun 248007, Uttarakhand, India; (R.K.S.); (N.P.N.)
| | - Tapan Behl
- Amity School of Pharmaceutical Sciences, Amity University, Mohali 140306, Punjab, India
| | - Rashmi Arora
- Chitkara College of Pharmacy, Chitkara University, Rajpura 140401, Punjab, India;
| | - Md. Khalid Anwer
- Department of Pharmaceutics, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Alkharj 11942, Saudi Arabia;
| | - Monica Gulati
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara 1444411, Punjab, India;
- Australian Research Centre in Complementary and Integrative Medicine, Faculty of Health, University of Technology Sydney, Ultimo, NSW 20227, Australia
| | - Simona Gabriela Bungau
- Department of Pharmacy, Faculty of Medicine and Pharmacy, University of Oradea, 410028 Oradea, Romania
- Doctoral School of Biological and Biomedical Sciences, University of Oradea, 410087 Oradea, Romania
| | - Mihaela Cristina Brisc
- Department of Medical Disciplines, Faculty of Medicine and Pharmacy, University of Oradea, 410073 Oradea, Romania;
| |
Collapse
|
37
|
Milicevic KD, Barbeau BL, Lovic DD, Patel AA, Ivanova VO, Antic SD. Physiological features of parvalbumin-expressing GABAergic interneurons contributing to high-frequency oscillations in the cerebral cortex. CURRENT RESEARCH IN NEUROBIOLOGY 2023; 6:100121. [PMID: 38616956 PMCID: PMC11015061 DOI: 10.1016/j.crneur.2023.100121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 11/13/2023] [Accepted: 12/01/2023] [Indexed: 04/16/2024] Open
Abstract
Parvalbumin-expressing (PV+) inhibitory interneurons drive gamma oscillations (30-80 Hz), which underlie higher cognitive functions. In this review, we discuss two groups/aspects of fundamental properties of PV+ interneurons. In the first group (dubbed Before Axon), we list properties representing optimal synaptic integration in PV+ interneurons designed to support fast oscillations. For example: [i] Information can neither enter nor leave the neocortex without the engagement of fast PV+ -mediated inhibition; [ii] Voltage responses in PV+ interneuron dendrites integrate linearly to reduce impact of the fluctuations in the afferent drive; and [iii] Reversed somatodendritic Rm gradient accelerates the time courses of synaptic potentials arriving at the soma. In the second group (dubbed After Axon), we list morphological and biophysical properties responsible for (a) short synaptic delays, and (b) efficient postsynaptic outcomes. For example: [i] Fast-spiking ability that allows PV+ interneurons to outpace other cortical neurons (pyramidal neurons). [ii] Myelinated axon (which is only found in the PV+ subclass of interneurons) to secure fast-spiking at the initial axon segment; and [iii] Inhibitory autapses - autoinhibition, which assures brief biphasic voltage transients and supports postinhibitory rebounds. Recent advent of scientific tools, such as viral strategies to target PV cells and the ability to monitor PV cells via in vivo imaging during behavior, will aid in defining the role of PV cells in the CNS. Given the link between PV+ interneurons and cognition, in the future, it would be useful to carry out physiological recordings in the PV+ cell type selectively and characterize if and how psychiatric and neurological diseases affect initiation and propagation of electrical signals in this cortical sub-circuit. Voltage imaging may allow fast recordings of electrical signals from many PV+ interneurons simultaneously.
Collapse
Affiliation(s)
- Katarina D. Milicevic
- University of Connecticut Health, School of Medicine, Institute for Systems Genomics, Farmington, CT, 06030, USA
- University of Belgrade, Faculty of Biology, Center for Laser Microscopy, Belgrade, 11000, Serbia
| | - Brianna L. Barbeau
- University of Connecticut Health, School of Medicine, Institute for Systems Genomics, Farmington, CT, 06030, USA
| | - Darko D. Lovic
- University of Connecticut Health, School of Medicine, Institute for Systems Genomics, Farmington, CT, 06030, USA
- University of Belgrade, Faculty of Biology, Center for Laser Microscopy, Belgrade, 11000, Serbia
| | - Aayushi A. Patel
- University of Connecticut Health, School of Medicine, Institute for Systems Genomics, Farmington, CT, 06030, USA
| | - Violetta O. Ivanova
- University of Connecticut Health, School of Medicine, Institute for Systems Genomics, Farmington, CT, 06030, USA
| | - Srdjan D. Antic
- University of Connecticut Health, School of Medicine, Institute for Systems Genomics, Farmington, CT, 06030, USA
| |
Collapse
|
38
|
Davidson CJ, Mascarin AT, Yahya MA, Rubio FJ, Gheidi A. Approaches and considerations of studying neuronal ensembles: a brief review. Front Cell Neurosci 2023; 17:1310724. [PMID: 38155864 PMCID: PMC10752959 DOI: 10.3389/fncel.2023.1310724] [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: 10/09/2023] [Accepted: 11/27/2023] [Indexed: 12/30/2023] Open
Abstract
First theorized by Hebb, neuronal ensembles have provided a framework for understanding how the mammalian brain operates, especially regarding learning and memory. Neuronal ensembles are discrete, sparsely distributed groups of neurons that become activated in response to a specific stimulus and are thought to provide an internal representation of the world. Beyond the study of region-wide or projection-wide activation, the study of ensembles offers increased specificity and resolution to identify and target specific memories or associations. Neuroscientists interested in the neurobiology of learning, memory, and motivated behavior have used electrophysiological-, calcium-, and protein-based proxies of neuronal activity in preclinical models to better understand the neurobiology of learned and motivated behaviors. Although these three approaches may be used to pursue the same general goal of studying neuronal ensembles, technical differences lead to inconsistencies in the output and interpretation of data. This mini-review highlights some of the methodologies used in electrophysiological-, calcium-, and protein-based studies of neuronal ensembles and discusses their strengths and weaknesses.
Collapse
Affiliation(s)
- Cameron J. Davidson
- William Beaumont School of Medicine, Oakland University, Rochester, MI, United States
| | - Alixandria T. Mascarin
- Department of Psychiatry and Behavioral Neurosciences, Wayne State University School of Medicine, Detroit, MI, United States
| | - Majd A. Yahya
- Department of Psychiatry and Behavioral Neurosciences, Wayne State University School of Medicine, Detroit, MI, United States
| | - F. Javier Rubio
- Neuronal Ensembles in Addiction Section, Behavioral Neuroscience Research Branch, Intramural Research Program/National Institute on Drug Abuse/National Institutes of Health, Bethesda, MD, United States
| | - Ali Gheidi
- Department of Biomedical Sciences, Mercer University, Macon, GA, United States
| |
Collapse
|
39
|
Zhou ZC, Gordon-Fennell A, Piantadosi SC, Ji N, Smith SL, Bruchas MR, Stuber GD. Deep-brain optical recording of neural dynamics during behavior. Neuron 2023; 111:3716-3738. [PMID: 37804833 PMCID: PMC10843303 DOI: 10.1016/j.neuron.2023.09.006] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Revised: 08/24/2023] [Accepted: 09/06/2023] [Indexed: 10/09/2023]
Abstract
In vivo fluorescence recording techniques have produced landmark discoveries in neuroscience, providing insight into how single cell and circuit-level computations mediate sensory processing and generate complex behaviors. While much attention has been given to recording from cortical brain regions, deep-brain fluorescence recording is more complex because it requires additional measures to gain optical access to harder to reach brain nuclei. Here we discuss detailed considerations and tradeoffs regarding deep-brain fluorescence recording techniques and provide a comprehensive guide for all major steps involved, from project planning to data analysis. The goal is to impart guidance for new and experienced investigators seeking to use in vivo deep fluorescence optical recordings in awake, behaving rodent models.
Collapse
Affiliation(s)
- Zhe Charles Zhou
- Department of Anesthesiology and Pain Medicine, University of Washington, Seattle, WA 98195, USA; Center for Neurobiology of Addiction, Pain, and Emotion, University of Washington, Seattle, WA 98195, USA
| | - Adam Gordon-Fennell
- Department of Anesthesiology and Pain Medicine, University of Washington, Seattle, WA 98195, USA; Center for Neurobiology of Addiction, Pain, and Emotion, University of Washington, Seattle, WA 98195, USA
| | - Sean C Piantadosi
- Department of Anesthesiology and Pain Medicine, University of Washington, Seattle, WA 98195, USA; Center for Neurobiology of Addiction, Pain, and Emotion, University of Washington, Seattle, WA 98195, USA
| | - Na Ji
- Department of Physics, University of California, Berkeley, Berkeley, CA 94720, USA; Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA; Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, CA 94720, USA; Molecular Biophysics and Integrated Bioimaging Division, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA
| | - Spencer LaVere Smith
- Department of Electrical and Computer Engineering, University of California Santa Barbara, Santa Barbara, CA 93106, USA
| | - Michael R Bruchas
- Department of Anesthesiology and Pain Medicine, University of Washington, Seattle, WA 98195, USA; Center for Neurobiology of Addiction, Pain, and Emotion, University of Washington, Seattle, WA 98195, USA; Department of Pharmacology, University of Washington, Seattle, WA 98195, USA; Department of Bioengineering, University of Washington, Seattle, WA 98195, USA.
| | - Garret D Stuber
- Department of Anesthesiology and Pain Medicine, University of Washington, Seattle, WA 98195, USA; Center for Neurobiology of Addiction, Pain, and Emotion, University of Washington, Seattle, WA 98195, USA; Department of Pharmacology, University of Washington, Seattle, WA 98195, USA.
| |
Collapse
|
40
|
Zhang Z, Wu Y, Xuan Z, Xu H, Yin S, Meng Z. Self-assembly of three-dimensional liver organoids: virtual reconstruction via endocytosed polymer dots for refactoring the fine structure. Biomater Sci 2023; 11:7867-7883. [PMID: 37902572 DOI: 10.1039/d3bm01174g] [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: 10/31/2023]
Abstract
In vitro culture of organoids holds considerable promise for the treatment of diseases or the provision of artificial organs. Traditional 2D differentiation from mesenchymal stem cells (MSCs) faces challenges in replicating the development of embryonic organs at the cellular level; conversely, the cultivation of 3D organoids exhibits potential for application. It is crucial for clinicians and technology researchers to acquire insights into organoid tissue differentiation, autonomous morphogenesis, as well as 3D assembly processes in vitro. In this investigation, novel 3D organoids capable of engendering complex liver-like tissues in vitro were cultured, and a class of high-luminance semiconductor polymer dots (Pdots) was employed to monitor the self-assembly process of 3D liver organoid tissues and cellular interaction and migration dynamics. Three-dimensional liver-bud (3D-LB) organoid tissues were derived through the interplay of induced MSCs, Wharton's Jelly, and human umbilical vein endothelial cells (HUVECs), and their structural characteristics were determined during the liver-bud organoid development; ultimately, the co-cultured organoid spatial cellular clusters resembling a truffle were successfully replicated. Utilizing R8-Pdots with remarkable resolution and biocompatibility, the structural elements of functional and vascularized organs derived from liver organoid tissues were adeptly reconstituted, and this investigation shall contribute to a further understanding of human hepato-developmental physiology and liver-disease modeling.
Collapse
Affiliation(s)
- Ze Zhang
- Department of Hepatobiliary-Pancreatic Surgery, China-Japan Union Hospital of Jilin University, Jilin University, No. 126 Xiantai Street, Changchun, Jilin 130000, P. R. China.
| | - Yuyang Wu
- State Key Laboratory of Integrated Optoelectronic, College of Electronic Science and Engineering, Jilin University, No. 2699 Qianjin Street, Changchun, Jilin 130012, P. R. China.
| | - Zhilu Xuan
- Department of Obstetrics & Gynecology, The First Hospital of Jilin University, Changchun, Jilin 130012, P. R. China
| | - Haotian Xu
- Department of Hepatobiliary-Pancreatic Surgery, China-Japan Union Hospital of Jilin University, Jilin University, No. 126 Xiantai Street, Changchun, Jilin 130000, P. R. China.
| | - Shengyan Yin
- State Key Laboratory of Integrated Optoelectronic, College of Electronic Science and Engineering, Jilin University, No. 2699 Qianjin Street, Changchun, Jilin 130012, P. R. China.
| | - Zihui Meng
- Department of Hepatobiliary-Pancreatic Surgery, China-Japan Union Hospital of Jilin University, Jilin University, No. 126 Xiantai Street, Changchun, Jilin 130000, P. R. China.
| |
Collapse
|
41
|
Xu S, Momin M, Ahmed S, Hossain A, Veeramuthu L, Pandiyan A, Kuo CC, Zhou T. Illuminating the Brain: Advances and Perspectives in Optoelectronics for Neural Activity Monitoring and Modulation. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2023; 35:e2303267. [PMID: 37726261 DOI: 10.1002/adma.202303267] [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: 04/07/2023] [Revised: 05/30/2023] [Indexed: 09/21/2023]
Abstract
Optogenetic modulation of brain neural activity that combines optical and electrical modes in a unitary neural system has recently gained robust momentum. Controlling illumination spatial coverage, designing light-activated modulators, and developing wireless light delivery and data transmission are crucial for maximizing the use of optical neuromodulation. To this end, biocompatible electrodes with enhanced optoelectrical performance, device integration for multiplexed addressing, wireless transmission, and multimodal operation in soft systems have been developed. This review provides an outlook for uniformly illuminating large brain areas while spatiotemporally imaging the neural responses upon optoelectrical stimulation with little artifacts. Representative concepts and important breakthroughs, such as head-mounted illumination, multiple implanted optical fibers, and micro-light-delivery devices, are discussed. Examples of techniques that incorporate electrophysiological monitoring and optoelectrical stimulation are presented. Challenges and perspectives are posed for further research efforts toward high-density optoelectrical neural interface modulation, with the potential for nonpharmacological neurological disease treatments and wireless optoelectrical stimulation.
Collapse
Affiliation(s)
- Shumao Xu
- Department of Engineering Science and Mechanics, Center for Neural Engineering, The Pennsylvania State University, Pennsylvania, 16802, USA
| | - Marzia Momin
- Department of Engineering Science and Mechanics, Center for Neural Engineering, The Pennsylvania State University, Pennsylvania, 16802, USA
| | - Salahuddin Ahmed
- Department of Engineering Science and Mechanics, Center for Neural Engineering, The Pennsylvania State University, Pennsylvania, 16802, USA
| | - Arafat Hossain
- Department of Electrical Engineering, The Pennsylvania State University, Pennsylvania, 16802, USA
| | - Loganathan Veeramuthu
- Department of Molecular Science and Engineering, National Taipei University of Technology, Taipei, 10608, Republic of China
| | - Archana Pandiyan
- Department of Molecular Science and Engineering, National Taipei University of Technology, Taipei, 10608, Republic of China
| | - Chi-Ching Kuo
- Department of Molecular Science and Engineering, National Taipei University of Technology, Taipei, 10608, Republic of China
| | - Tao Zhou
- Department of Engineering Science and Mechanics, Center for Neural Engineering, The Pennsylvania State University, Pennsylvania, 16802, USA
| |
Collapse
|
42
|
Matchynski JI, Cilley TS, Sadik N, Makki KM, Wu M, Manwar R, Woznicki AR, Kallakuri S, Arfken CL, Hope BT, Avanaki K, Conti AC, Perrine SA. Quantification of prefrontal cortical neuronal ensembles following conditioned fear learning in a Fos-LacZ transgenic rat with photoacoustic imaging in Vivo. PHOTOACOUSTICS 2023; 33:100551. [PMID: 38021296 PMCID: PMC10658601 DOI: 10.1016/j.pacs.2023.100551] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 05/19/2023] [Accepted: 08/26/2023] [Indexed: 12/01/2023]
Abstract
Understanding the neurobiology of complex behaviors requires measurement of activity in the discrete population of active neurons, neuronal ensembles, which control the behavior. Conventional neuroimaging techniques ineffectively measure neuronal ensemble activity in the brain in vivo because they assess the average regional neuronal activity instead of the specific activity of the neuronal ensemble that mediates the behavior. Our functional molecular photoacoustic tomography (FM-PAT) system allows direct imaging of Fos-dependent neuronal ensemble activation in Fos-LacZ transgenic rats in vivo. We tested four experimental conditions and found increased FM-PAT signal in prefrontal cortical areas in rats undergoing conditioned fear or novel context exposure. A parallel immunofluorescence ex vivo study of Fos expression found similar findings. These findings demonstrate the ability of FM-PAT to measure Fos-expressing neuronal ensembles directly in vivo and support a mechanistic role for the prefrontal cortex in higher-order processing of response to specific stimuli or environmental cues.
Collapse
Affiliation(s)
- James I Matchynski
- Department of Psychiatry and Behavioral Neurosciences, Wayne State University School of Medicine, Detroit, MI, USA
- Translational Neuroscience Program, Wayne State University School of Medicine, Detroit, MI, USA
- John D. Dingell Veterans Affairs Medical Center, Detroit, MI, USA
- Wayne State MD/PhD Program, Wayne State University School of Medicine, Detroit, MI, USA
| | - Timothy S Cilley
- Department of Psychiatry and Behavioral Neurosciences, Wayne State University School of Medicine, Detroit, MI, USA
| | - Nareen Sadik
- Department of Psychiatry and Behavioral Neurosciences, Wayne State University School of Medicine, Detroit, MI, USA
| | - Kassem M Makki
- John D. Dingell Veterans Affairs Medical Center, Detroit, MI, USA
| | - Min Wu
- John D. Dingell Veterans Affairs Medical Center, Detroit, MI, USA
| | - Rayyan Manwar
- University of Illinois at Chicago, Department of Bioengineering, Chicago, IL, USA
| | | | - Srinivasu Kallakuri
- Department of Psychiatry and Behavioral Neurosciences, Wayne State University School of Medicine, Detroit, MI, USA
| | - Cynthia L Arfken
- Department of Psychiatry and Behavioral Neurosciences, Wayne State University School of Medicine, Detroit, MI, USA
- Translational Neuroscience Program, Wayne State University School of Medicine, Detroit, MI, USA
| | - Bruce T Hope
- The National Institute on Drug Abuse (NIDA), Intramural Research Program, Baltimore, MD, USA
| | - Kamran Avanaki
- University of Illinois at Chicago, Department of Bioengineering, Chicago, IL, USA
| | - Alana C Conti
- Department of Psychiatry and Behavioral Neurosciences, Wayne State University School of Medicine, Detroit, MI, USA
- Translational Neuroscience Program, Wayne State University School of Medicine, Detroit, MI, USA
- John D. Dingell Veterans Affairs Medical Center, Detroit, MI, USA
| | - Shane A Perrine
- Department of Psychiatry and Behavioral Neurosciences, Wayne State University School of Medicine, Detroit, MI, USA
- Translational Neuroscience Program, Wayne State University School of Medicine, Detroit, MI, USA
| |
Collapse
|
43
|
Patel N, Ouellet V, Paquet-Mercier F, Chetoui N, Bélanger E, Paquet ME, Godin AG, Marquet P. A robust and reliable methodology to perform GECI-based multi-time point neuronal calcium imaging within mixed cultures of human iPSC-derived cortical neurons. Front Neurosci 2023; 17:1247397. [PMID: 37817802 PMCID: PMC10560759 DOI: 10.3389/fnins.2023.1247397] [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: 06/26/2023] [Accepted: 08/16/2023] [Indexed: 10/12/2023] Open
Abstract
Introduction Human induced pluripotent stem cells (iPSCs), with their ability to generate human neural cells (astrocytes and neurons) from patients, hold great promise for understanding the pathophysiology of major neuropsychiatric diseases such as schizophrenia and bipolar disorders, which includes alterations in cerebral development. Indeed, the in vitro neurodifferentiation of iPSCs, while recapitulating certain major stages of neurodevelopment in vivo, makes it possible to obtain networks of living human neurons. The culture model presented is particularly attractive within this framework since it involves iPSC-derived neural cells, which more specifically differentiate into cortical neurons of diverse types (in particular glutamatergic and GABAergic) and astrocytes. However, these in vitro neuronal networks, which may be heterogeneous in their degree of differentiation, remain challenging to bring to an appropriate level of maturation. It is therefore necessary to develop tools capable of analyzing a large number of cells to assess this maturation process. Calcium (Ca2+) imaging, which has been extensively developed, undoubtedly offers an incredibly good approach, particularly in its versions using genetically encoded calcium indicators. However, in the context of these iPSC-derived neural cell cultures, there is a lack of studies that propose Ca2+ imaging methods that can finely characterize the evolution of neuronal maturation during the neurodifferentiation process. Methods In this study, we propose a robust and reliable method for specifically measuring neuronal activity at two different time points of the neurodifferentiation process in such human neural cultures. To this end, we have developed a specific Ca2+ signal analysis procedure and tested a series of different AAV serotypes to obtain expression levels of GCaMP6f under the control of the neuron-specific human synapsin1 (hSyn) promoter. Results The retro serotype has been found to be the most efficient in driving the expression of the GCaMP6f and is compatible with multi-time point neuronal Ca2+ imaging in our human iPSC-derived neural cultures. An AAV2/retro carrying GCaMP6f under the hSyn promoter (AAV2/retro-hSyn-GCaMP6f) is an efficient vector that we have identified. To establish the method, calcium measurements were carried out at two time points in the neurodifferentiation process with both hSyn and CAG promoters, the latter being known to provide high transient gene expression across various cell types. Discussion Our results stress that this methodology involving AAV2/retro-hSyn-GCaMP6f is suitable for specifically measuring neuronal calcium activities over multiple time points and is compatible with the neurodifferentiation process in our mixed human neural cultures.
Collapse
Affiliation(s)
- Niraj Patel
- Department of Psychiatry and Neuroscience, Laval University, Quebec, QC, Canada
- CERVO Brain Research Centre, Laval University, Quebec, QC, Canada
| | - Vincent Ouellet
- Department of Psychiatry and Neuroscience, Laval University, Quebec, QC, Canada
- CERVO Brain Research Centre, Laval University, Quebec, QC, Canada
| | | | - Nizar Chetoui
- CERVO Brain Research Centre, Laval University, Quebec, QC, Canada
| | - Erik Bélanger
- CERVO Brain Research Centre, Laval University, Quebec, QC, Canada
| | - Marie-Eve Paquet
- CERVO Brain Research Centre, Laval University, Quebec, QC, Canada
- Department of Biochemistry, Microbiology and Bioinformatics, Laval University, Quebec, QC, Canada
| | - Antoine G. Godin
- Department of Psychiatry and Neuroscience, Laval University, Quebec, QC, Canada
- CERVO Brain Research Centre, Laval University, Quebec, QC, Canada
- Centre for Optics, Photonics and Lasers (COPL), Laval University, Quebec, QC, Canada
| | - Pierre Marquet
- Department of Psychiatry and Neuroscience, Laval University, Quebec, QC, Canada
- CERVO Brain Research Centre, Laval University, Quebec, QC, Canada
- Centre for Optics, Photonics and Lasers (COPL), Laval University, Quebec, QC, Canada
| |
Collapse
|
44
|
Luo X, Liu Z, Xu R. Adult tissue-specific stem cell interaction: novel technologies and research advances. Front Cell Dev Biol 2023; 11:1220694. [PMID: 37808078 PMCID: PMC10551553 DOI: 10.3389/fcell.2023.1220694] [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: 05/11/2023] [Accepted: 09/11/2023] [Indexed: 10/10/2023] Open
Abstract
Adult tissue-specific stem cells play a dominant role in tissue homeostasis and regeneration. Various in vivo markers of adult tissue-specific stem cells have been increasingly reported by lineage tracing in genetic mouse models, indicating that marked cells differentiation is crucial during homeostasis and regeneration. How adult tissue-specific stem cells with indicated markers contact the adjacent lineage with indicated markers is of significance to be studied. Novel methods bring future findings. Recent advances in lineage tracing, synthetic receptor systems, proximity labeling, and transcriptomics have enabled easier and more accurate cell behavior visualization and qualitative and quantitative analysis of cell-cell interactions than ever before. These technological innovations have prompted researchers to re-evaluate previous experimental results, providing increasingly compelling experimental results for understanding the mechanisms of cell-cell interactions. This review aimed to describe the recent methodological advances of dual enzyme lineage tracing system, the synthetic receptor system, proximity labeling, single-cell RNA sequencing and spatial transcriptomics in the study of adult tissue-specific stem cells interactions. An enhanced understanding of the mechanisms of adult tissue-specific stem cells interaction is important for tissue regeneration and maintenance of homeostasis in organisms.
Collapse
Affiliation(s)
| | | | - Ruoshi Xu
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| |
Collapse
|
45
|
Nadtochiy A, Luu P, Fraser SE, Truong TV. VoDEx: a Python library for time annotation and management of volumetric functional imaging data. Bioinformatics 2023; 39:btad568. [PMID: 37699009 PMCID: PMC10562951 DOI: 10.1093/bioinformatics/btad568] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Revised: 08/30/2023] [Accepted: 09/11/2023] [Indexed: 09/14/2023] Open
Abstract
SUMMARY In functional imaging studies, accurately synchronizing the time course of experimental manipulations and stimulus presentations with resulting imaging data is crucial for analysis. Current software tools lack such functionality, requiring manual processing of the experimental and imaging data, which is error-prone and potentially non-reproducible. We present VoDEx, an open-source Python library that streamlines the data management and analysis of functional imaging data. VoDEx synchronizes the experimental timeline and events (e.g. presented stimuli, recorded behavior) with imaging data. VoDEx provides tools for logging and storing the timeline annotation, and enables retrieval of imaging data based on specific time-based and manipulation-based experimental conditions. AVAILABILITY AND IMPLEMENTATION VoDEx is an open-source Python library and can be installed via the "pip install" command. It is released under a BSD license, and its source code is publicly accessible on GitHub (https://github.com/LemonJust/vodex). A graphical interface is available as a napari-vodex plugin, which can be installed through the napari plugins menu or using "pip install." The source code for the napari plugin is available on GitHub (https://github.com/LemonJust/napari-vodex). The software version at the time of submission is archived at Zenodo (version v1.0.18, https://zenodo.org/record/8061531).
Collapse
Affiliation(s)
- Anna Nadtochiy
- Department of Quantitative and Computational Biology, University of Southern California, Los Angeles, CA 90089, United States
- Translational Imaging Center, University of Southern California, Los Angeles, CA 90089, United States
| | - Peter Luu
- Translational Imaging Center, University of Southern California, Los Angeles, CA 90089, United States
- Division of Molecular and Computational Biology, Department of Biological Sciences, University of Southern California, Los Angeles, CA 90089, United States
| | - Scott E Fraser
- Department of Quantitative and Computational Biology, University of Southern California, Los Angeles, CA 90089, United States
- Translational Imaging Center, University of Southern California, Los Angeles, CA 90089, United States
- Division of Molecular and Computational Biology, Department of Biological Sciences, University of Southern California, Los Angeles, CA 90089, United States
| | - Thai V Truong
- Translational Imaging Center, University of Southern California, Los Angeles, CA 90089, United States
- Division of Molecular and Computational Biology, Department of Biological Sciences, University of Southern California, Los Angeles, CA 90089, United States
| |
Collapse
|
46
|
Xiao S, Cunningham WJ, Kondabolu K, Lowet E, Moya MV, Mount R, Ravasio C, Economo MN, Han X, Mertz J. Large-scale deep tissue voltage imaging with targeted illumination confocal microscopy. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.07.21.548930. [PMID: 37502929 PMCID: PMC10370169 DOI: 10.1101/2023.07.21.548930] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/29/2023]
Abstract
Voltage imaging with cellular specificity has been made possible by the tremendous advances in genetically encoded voltage indicators (GEVIs). However, the kilohertz rates required for voltage imaging lead to weak signals. Moreover, out-of-focus fluorescence and tissue scattering produce background that both undermines signal-to-noise ratio (SNR) and induces crosstalk between cells, making reliable in vivo imaging in densely labeled tissue highly challenging. We describe a microscope that combines the distinct advantages of targeted illumination and confocal gating, while also maximizing signal detection efficiency. The resulting benefits in SNR and crosstalk reduction are quantified experimentally and theoretically. Our microscope provides a versatile solution for enabling high-fidelity in vivo voltage imaging at large scales and penetration depths, which we demonstrate across a wide range of imaging conditions and different GEVI classes.
Collapse
Affiliation(s)
- Sheng Xiao
- Department of Biomedical Engineering, Boston University, Boston MA 02215
| | | | | | - Eric Lowet
- Department of Biomedical Engineering, Boston University, Boston MA 02215
| | - Maria V. Moya
- Department of Biomedical Engineering, Boston University, Boston MA 02215
| | - Rebecca Mount
- Department of Biomedical Engineering, Boston University, Boston MA 02215
| | - Cara Ravasio
- Department of Biomedical Engineering, Boston University, Boston MA 02215
| | - Michael N. Economo
- Department of Biomedical Engineering, Boston University, Boston MA 02215
- Neurophotonics Center, Boston University, Boston MA, 02215
| | - Xue Han
- Department of Biomedical Engineering, Boston University, Boston MA 02215
- Neurophotonics Center, Boston University, Boston MA, 02215
| | - Jerome Mertz
- Department of Biomedical Engineering, Boston University, Boston MA 02215
- Neurophotonics Center, Boston University, Boston MA, 02215
| |
Collapse
|
47
|
Farrants H, Shuai Y, Lemon WC, Hernandez CM, Yang S, Patel R, Qiao G, Frei MS, Grimm JB, Hanson TL, Tomaska F, Turner GC, Stringer C, Keller PJ, Beyene AG, Chen Y, Liang Y, Lavis LD, Schreiter ER. A modular chemigenetic calcium indicator enables in vivo functional imaging with near-infrared light. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.07.18.549527. [PMID: 37503182 PMCID: PMC10370049 DOI: 10.1101/2023.07.18.549527] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/29/2023]
Abstract
Genetically encoded fluorescent calcium indicators have revolutionized neuroscience and other biological fields by allowing cellular-resolution recording of physiology during behavior. However, we currently lack bright, genetically targetable indicators in the near infrared that can be used in animals. Here, we describe WHaloCaMP, a modular chemigenetic calcium indicator built from bright dye-ligands and protein sensor domains that can be genetically targeted to specific cell populations. Fluorescence change in WHaloCaMP results from reversible quenching of the bound dye via a strategically placed tryptophan. WHaloCaMP is compatible with rhodamine dye-ligands that fluoresce from green to near-infrared, including several dye-ligands that efficiently label the central nervous system in animals. When bound to a near-infrared dye-ligand, WHaloCaMP1a is more than twice as bright as jGCaMP8s, and shows a 7× increase in fluorescence intensity and a 2.1 ns increase in fluorescence lifetime upon calcium binding. We use WHaloCaMP1a with near-infrared fluorescence emission to image Ca2+ responses in flies and mice, to perform three-color multiplexed functional imaging of hundreds of neurons and astrocytes in zebrafish larvae, and to quantitate calcium concentration using fluorescence lifetime imaging microscopy (FLIM).
Collapse
Affiliation(s)
- Helen Farrants
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA, USA
| | - Yichun Shuai
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA, USA
| | - William C Lemon
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA, USA
| | | | - Shang Yang
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA, USA
| | - Ronak Patel
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA, USA
| | - Guanda Qiao
- Department of Diagnostic Radiology and Nuclear Medicine, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Michelle S Frei
- Department of Chemical Biology, Max Planck Institute for Medical Research, Jahnstrasse 29, 69120 Heidelberg, Germany
| | - Jonathan B Grimm
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA, USA
| | - Timothy L Hanson
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA, USA
| | - Filip Tomaska
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA, USA
| | - Glenn C Turner
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA, USA
| | - Carsen Stringer
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA, USA
| | - Philipp J Keller
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA, USA
| | - Abraham G Beyene
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA, USA
| | - Yao Chen
- Department of Neuroscience, Washington University in St. Louis, St. Louis, MO 63110, USA
| | - Yajie Liang
- Department of Diagnostic Radiology and Nuclear Medicine, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Luke D Lavis
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA, USA
| | - Eric R Schreiter
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA, USA
| |
Collapse
|
48
|
Affiliation(s)
- Xinyang Li
- Department of Automation, Tsinghua University, Beijing, China
| | - Yuanlong Zhang
- Department of Automation, Tsinghua University, Beijing, China
| | - Jiamin Wu
- Department of Automation, Tsinghua University, Beijing, China.
| | - Qionghai Dai
- Department of Automation, Tsinghua University, Beijing, China.
| |
Collapse
|
49
|
Tian H, Davis HC, Wong-Campos JD, Park P, Fan LZ, Gmeiner B, Begum S, Werley CA, Borja GB, Upadhyay H, Shah H, Jacques J, Qi Y, Parot V, Deisseroth K, Cohen AE. Video-based pooled screening yields improved far-red genetically encoded voltage indicators. Nat Methods 2023; 20:1082-1094. [PMID: 36624211 PMCID: PMC10329731 DOI: 10.1038/s41592-022-01743-5] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Accepted: 11/28/2022] [Indexed: 01/11/2023]
Abstract
Video-based screening of pooled libraries is a powerful approach for directed evolution of biosensors because it enables selection along multiple dimensions simultaneously from large libraries. Here we develop a screening platform, Photopick, which achieves precise phenotype-activated photoselection over a large field of view (2.3 × 2.3 mm, containing >103 cells, per shot). We used the Photopick platform to evolve archaerhodopsin-derived genetically encoded voltage indicators (GEVIs) with improved signal-to-noise ratio (QuasAr6a) and kinetics (QuasAr6b). These GEVIs gave improved signals in cultured neurons and in live mouse brains. By combining targeted in vivo optogenetic stimulation with high-precision voltage imaging, we characterized inhibitory synaptic coupling between individual cortical NDNF (neuron-derived neurotrophic factor) interneurons, and excitatory electrical synapses between individual hippocampal parvalbumin neurons. The QuasAr6 GEVIs are powerful tools for all-optical electrophysiology and the Photopick approach could be adapted to evolve a broad range of biosensors.
Collapse
Affiliation(s)
- He Tian
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA, USA
| | - Hunter C Davis
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA, USA
| | - J David Wong-Campos
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA, USA
| | - Pojeong Park
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA, USA
| | - Linlin Z Fan
- Department of Bioengineering, Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA
| | - Benjamin Gmeiner
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA, USA
| | - Shahinoor Begum
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA, USA
| | | | | | | | | | | | - Yitong Qi
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA, USA
| | - Vicente Parot
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA, USA
- Institute for Biological and Medical Engineering, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Karl Deisseroth
- Department of Bioengineering, Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA
- Howard Hughes Medical Institute, Chevy Chase, MA, USA
| | - Adam E Cohen
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA, USA.
- Department of Physics, Harvard University, Cambridge, MA, USA.
| |
Collapse
|
50
|
Bowman AJ, Huang C, Schnitzer MJ, Kasevich MA. Wide-field fluorescence lifetime imaging of neuron spiking and subthreshold activity in vivo. Science 2023; 380:1270-1275. [PMID: 37347862 PMCID: PMC10361454 DOI: 10.1126/science.adf9725] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Accepted: 05/16/2023] [Indexed: 06/24/2023]
Abstract
The development of voltage-sensitive fluorescent probes suggests fluorescence lifetime as a promising readout for electrical activity in biological systems. Existing approaches fail to achieve the speed and sensitivity required for voltage imaging in neuroscience applications. We demonstrated that wide-field electro-optic fluorescence lifetime imaging microscopy (EO-FLIM) allows lifetime imaging at kilohertz frame-acquisition rates, spatially resolving action potential propagation and subthreshold neural activity in live adult Drosophila. Lifetime resolutions of <5 picoseconds at 1 kilohertz were achieved for single-cell voltage recordings. Lifetime readout is limited by photon shot noise, and the method provides strong rejection of motion artifacts and technical noise sources. Recordings revealed local transmembrane depolarizations, two types of spikes with distinct fluorescence lifetimes, and phase locking of spikes to an external mechanical stimulus.
Collapse
Affiliation(s)
- Adam J Bowman
- Physics Department, Stanford University, Stanford, CA 94305, USA
| | - Cheng Huang
- James H. Clark Center, Stanford University, Stanford, CA 94305, USA
| | - Mark J Schnitzer
- James H. Clark Center, Stanford University, Stanford, CA 94305, USA
- CNC Program, Stanford University, Stanford, CA 94305, USA
- Howard Hughes Medical Institute, Stanford University, Stanford, CA 94305, USA
| | - Mark A Kasevich
- Physics Department, Stanford University, Stanford, CA 94305, USA
| |
Collapse
|