1
|
Pereira AR, Rooney LM, Gomes IB, Simões M, McConnell G. The impact of methylparaben and chlorine on the architecture of Stenotrophomonas maltophilia biofilms. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 951:175646. [PMID: 39168334 DOI: 10.1016/j.scitotenv.2024.175646] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 07/30/2024] [Accepted: 08/17/2024] [Indexed: 08/23/2024]
Abstract
The biofilm architecture is significantly influenced by external environmental conditions. Biofilms grown on drinking water distribution systems (DWDS) are exposed to environmental contaminants, including parabens, and disinfection strategies, such as chlorine. Although changes in biofilm density and culturability from chemical exposure are widely reported, little is known about the effects of parabens and chlorine on biofilm morphology and architecture. This is the first study evaluating architectural changes in Stenotrophomonas maltophilia colony biofilms (representatives of bacterial communities presented in DWDS) induced by the exposure to methylparaben (MP) at environmental (15 μg/L) and in-use (15 mg/L) concentrations, and chlorine at 5 mg/L, using widefield epi-fluorescence mesoscopy with Mesolens. The GFP fluorescence of colony biofilms allowed the visualization of internal structures and Nile Red fluorescence permitted the inspection of the distribution of lipids. Our data show that exposure to MP triggers physiological and morphological adaptation in mature colony biofilms by increasing the complexity of internal structures, which may confer protection to embedded cells from external chemical molecules. These architectural modifications include changes in lipid distribution as an adaptive response to MP exposure. Although chlorine exposure affected colony biofilm diameter and architecture, the colony roundness was completely affected by the simultaneous presence of MP and chlorine. This work is pioneer in using Mesolens to highlight the risks of exposure to emerging environmental contaminants (MP), by affecting the architecture of biofilms formed by drinking water (DW) bacteria, even when combined with routine disinfection strategies.
Collapse
Affiliation(s)
- Ana Rita Pereira
- LEPABE - Laboratory for Process Engineering, Environment, Biotechnology and Energy, Faculty of Engineering, University of Porto, Rua Dr. Roberto Frias, 4200-465 Porto, Portugal; ALiCE - Associate Laboratory in Chemical Engineering, Faculty of Engineering, University of Porto, Rua Dr. Roberto Frias, 4200-465 Porto, Portugal.
| | - Liam M Rooney
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, 161 Cathedral Street, Glasgow G4 0RE, UK
| | - Inês B Gomes
- LEPABE - Laboratory for Process Engineering, Environment, Biotechnology and Energy, Faculty of Engineering, University of Porto, Rua Dr. Roberto Frias, 4200-465 Porto, Portugal; ALiCE - Associate Laboratory in Chemical Engineering, Faculty of Engineering, University of Porto, Rua Dr. Roberto Frias, 4200-465 Porto, Portugal
| | - Manuel Simões
- LEPABE - Laboratory for Process Engineering, Environment, Biotechnology and Energy, Faculty of Engineering, University of Porto, Rua Dr. Roberto Frias, 4200-465 Porto, Portugal; ALiCE - Associate Laboratory in Chemical Engineering, Faculty of Engineering, University of Porto, Rua Dr. Roberto Frias, 4200-465 Porto, Portugal
| | - Gail McConnell
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, 161 Cathedral Street, Glasgow G4 0RE, UK
| |
Collapse
|
2
|
Rooney LM, Bottura B, Baxter K, Amos WB, Hoskisson PA, McConnell G. Addressing multiscale microbial challenges using the Mesolens. J Microsc 2024; 296:139-144. [PMID: 36692253 DOI: 10.1111/jmi.13172] [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: 12/06/2022] [Revised: 01/18/2023] [Accepted: 01/18/2023] [Indexed: 01/25/2023]
Abstract
We provide a brief review of the development and application of the Mesolens and its impact on microbiology. Microbial specimens such as infected tissue samples, colonies surfaces, and biofilms are routinely collected at the mesoscale. This means that they are relatively large multimillimetre-sized samples which contain microscopic detail that must be observed to answer important questions across various sectors. The Mesolens presents the ideal imaging method to study these specimens as no other optical microscope can thanks to its unique combination of low magnification and high numerical aperture providing large field-of-view, high-resolution imaging. We demonstrate the current applications of the Mesolens to microbial imaging and go on to outline the huge potential of the Mesolens to impact other key areas of microbiology.
Collapse
Affiliation(s)
- Liam M Rooney
- Strathclyde Institute for Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, UK
| | - Beatrice Bottura
- Strathclyde Institute for Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, UK
| | - Katherine Baxter
- Strathclyde Institute for Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, UK
| | - William B Amos
- Strathclyde Institute for Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, UK
| | - Paul A Hoskisson
- Strathclyde Institute for Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, UK
| | - Gail McConnell
- Strathclyde Institute for Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, UK
| |
Collapse
|
3
|
Ma H, Chen M, Xu J, Yang Y, Zhao Y, Liu Y. An Omni-Mesoscope for multiscale high-throughput quantitative phase imaging of cellular dynamics and high-content molecular characterization. SCIENCE ADVANCES 2024; 10:eadq5009. [PMID: 39413179 PMCID: PMC11482309 DOI: 10.1126/sciadv.adq5009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Accepted: 09/12/2024] [Indexed: 10/18/2024]
Abstract
The mesoscope has emerged as a powerful imaging tool in biomedical research, yet its high cost and low resolution have limited its broader application. Here, we introduce the Omni-Mesoscope, a high-spatial-temporal and multimodal mesoscopic imaging platform built from cost-efficient off-the-shelf components. This system uniquely merges the capabilities of label-free quantitative phase microscopy to capture live-cell morphodynamics across thousands of cells with highly multiplexed fluorescence imaging for comprehensive molecular characterization. This Omni-Mesoscope offers a mesoscale field of view of ~5 square millimeters with a high spatial resolution down to 700 nanometers, enabling the capture of detailed subcellular features. We demonstrate its capability in delineating molecular characteristics underlying rare morphodynamic cellular phenomena, including cancer cell responses to chemotherapy and the emergence of polyploidy in drug-resistant cells. We also integrate expansion technique to enhance three-dimensional volumetric super-resolution imaging of thicker tissues, opening the avenues for biological exploration at unprecedented scales and resolutions.
Collapse
Affiliation(s)
- Hongqiang Ma
- Departments of Medicine and Bioengineering, University of Pittsburgh, Pittsburgh, PA 15213, USA
- Department of Bioengineering, Beckman Institute for Advanced Science and Technology, University of Illinois Urbana-Champaign, Urbana, IL 61801, USA
| | - Maomao Chen
- Departments of Medicine and Bioengineering, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Jianquan Xu
- Departments of Medicine and Bioengineering, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Yaxin Yang
- Department of Bioengineering, Beckman Institute for Advanced Science and Technology, University of Illinois Urbana-Champaign, Urbana, IL 61801, USA
| | - Yongxin Zhao
- Departments of Biological Sciences and Biomedical Engineering, Carnegie Mellon University, Pittsburgh, PA 15213, USA
- Viron Molecular Medicine Institute, Boston, MA 02201, USA
| | - Yang Liu
- Departments of Medicine and Bioengineering, University of Pittsburgh, Pittsburgh, PA 15213, USA
- Department of Bioengineering, Beckman Institute for Advanced Science and Technology, University of Illinois Urbana-Champaign, Urbana, IL 61801, USA
- Departments of Bioengineering and Electrical and Computer Engineering, Beckman Institute for Advanced Science and Technology, Cancer Center at Illinois, University of Illinois Urbana-Champaign, Urbana, IL 61801, USA
| |
Collapse
|
4
|
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
|
5
|
Geiser A, Currie S, Al-Hasani H, Chadt A, McConnell G, Gould GW. A novel 3D imaging approach for quantification of GLUT4 levels across the intact myocardium. J Cell Sci 2024; 137:jcs262146. [PMID: 38958032 DOI: 10.1242/jcs.262146] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Accepted: 06/24/2024] [Indexed: 07/04/2024] Open
Abstract
Cellular heterogeneity is a well-accepted feature of tissues, and both transcriptional and metabolic diversity have been revealed by numerous approaches, including optical imaging. However, the high magnification objective lenses needed for high-resolution imaging provides information from only small layers of tissue, which can result in poor cell statistics. There is therefore an unmet need for an imaging modality that can provide detailed molecular and cellular insight within intact tissue samples in 3D. Using GFP-tagged GLUT4 as proof of concept, we present here a novel optical mesoscopy approach that allows precise measurement of the spatial location of GLUT4 within specific anatomical structures across the myocardium in ultrathick sections (5 mm×5 mm×3 mm) of intact mouse heart. We reveal distinct GLUT4 distribution patterns across cardiac walls and highlight specific changes in GLUT4 expression levels in response to high fat diet-feeding, and we identify sex-dependent differences in expression patterns. This method is applicable to any target that can be labelled for light microscopy, and to other complex tissues when organ structure needs to be considered simultaneously with cellular detail.
Collapse
Affiliation(s)
- Angéline Geiser
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, UK
| | - Susan Currie
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, UK
| | - Hadi Al-Hasani
- Institute for Clinical Biochemistry and Pathobiochemistry, German Diabetes Center (DDZ), Leibniz-Center for Diabetes Research at the Heinrich Heine University, Medical faculty, Düsseldorf, Germany
- German Center for Diabetes Research, Partner Düsseldorf, München-Neuherberg, Germany
| | - Alexandra Chadt
- Institute for Clinical Biochemistry and Pathobiochemistry, German Diabetes Center (DDZ), Leibniz-Center for Diabetes Research at the Heinrich Heine University, Medical faculty, Düsseldorf, Germany
- German Center for Diabetes Research, Partner Düsseldorf, München-Neuherberg, Germany
| | - Gail McConnell
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, UK
| | - Gwyn W Gould
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, UK
| |
Collapse
|
6
|
Kramer SN, Antarasen J, Reinholt CR, Kisley L. A practical guide to light-sheet microscopy for nanoscale imaging: Looking beyond the cell. JOURNAL OF APPLIED PHYSICS 2024; 136:091101. [PMID: 39247785 PMCID: PMC11380115 DOI: 10.1063/5.0218262] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Accepted: 08/12/2024] [Indexed: 09/10/2024]
Abstract
We present a comprehensive guide to light-sheet microscopy (LSM) to assist scientists in navigating the practical implementation of this microscopy technique. Emphasizing the applicability of LSM to image both static microscale and nanoscale features, as well as diffusion dynamics, we present the fundamental concepts of microscopy, progressing through beam profile considerations, to image reconstruction. We outline key practical decisions in constructing a home-built system and provide insight into the alignment and calibration processes. We briefly discuss the conditions necessary for constructing a continuous 3D image and introduce our home-built code for data analysis. By providing this guide, we aim to alleviate the challenges associated with designing and constructing LSM systems and offer scientists new to LSM a valuable resource in navigating this complex field.
Collapse
Affiliation(s)
- Stephanie N Kramer
- Department of Physics, Case Western Reserve University, Rockefeller Building, 2076 Adelbert Road, Cleveland, Ohio 44106, USA
| | - Jeanpun Antarasen
- Department of Physics, Case Western Reserve University, Rockefeller Building, 2076 Adelbert Road, Cleveland, Ohio 44106, USA
| | - Cole R Reinholt
- Department of Physics, Case Western Reserve University, Rockefeller Building, 2076 Adelbert Road, Cleveland, Ohio 44106, USA
| | | |
Collapse
|
7
|
Clapperton M, Kunanandam T, Florea CD, Douglas CM, McConnell G. Multimodal optical mesoscopy reveals the quantity and spatial distribution of Gram-positive biofilms in ex vivo tonsils. J Microsc 2024; 295:121-130. [PMID: 38296824 DOI: 10.1111/jmi.13266] [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: 11/28/2023] [Revised: 01/12/2024] [Accepted: 01/15/2024] [Indexed: 02/02/2024]
Abstract
Biofilms are known to be present in tonsils, but little is known about their spatial location and size distribution throughout the tonsil. Studies of the location and distribution of biofilms in tonsil specimens have thus far been limited to either high-magnification methods such as electron microscopy, which enables high-resolution imaging but only from a tiny tissue volume, or lower magnification techniques such as light microscopy, which allow imaging of larger specimens but with poor spatial resolution. To overcome these limitations, we report the use of multimodal optical mesoscopy to visualise and quantify the number and spatial distribution of Gram-positive biofilms in fresh, excised paediatric tonsils. This methodology supports simultaneous imaging of both the tonsil host and biofilms in whole mounts of tissue up to 5 mm × 5 mm × 3 mm with subcellular resolution throughout. A quantitative assessment of 36 tonsil specimens revealed no statistically significant difference between biofilm presence on the tonsil surface and the interior of the tonsil. This new quantitative mesoscale imaging approach may prove useful in understanding the role of biofilms in tonsillar diseases and other infections.
Collapse
Affiliation(s)
- Megan Clapperton
- Department of Physics, SUPA, University of Strathclyde, Glasgow, UK
| | - Tash Kunanandam
- Department of Otolaryngology - Head and Neck Surgery, Royal Hospital for Children, Glasgow, UK
| | - Catalina D Florea
- Department of Otolaryngology - Head and Neck Surgery, Royal Hospital for Children, Glasgow, UK
| | - Catriona M Douglas
- Department of Otolaryngology - Head and Neck Surgery, Queen Elizabeth University Hospital, Glasgow, UK
| | - Gail McConnell
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, UK
| |
Collapse
|
8
|
Ma H, Chen M, Xu J, Zhao Y, Liu Y. An Omni-Mesoscope for multiscale high-throughput quantitative phase imaging of cellular dynamics and high-content molecular characterization. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.18.604137. [PMID: 39071371 PMCID: PMC11275778 DOI: 10.1101/2024.07.18.604137] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/30/2024]
Abstract
The mesoscope has emerged as a powerful imaging tool in biomedical research, yet its high cost and low resolution have limited its broader application. Here, we introduce the Omni-Mesoscope, a cost-effective high-spatial-temporal, multimodal, and multiplex mesoscopic imaging platform built from cost-efficient off-the-shelf components. This system uniquely merges the capabilities of quantitative phase microscopy to capture live-cell dynamics over a large cell population with highly multiplexed fluorescence imaging for comprehensive molecular characterization. This integration facilitates simultaneous tracking of live-cell morphodynamics across thousands of cells, alongside high-content molecular analysis at the single-cell level. Furthermore, the Omni-Mesoscope offers a mesoscale field of view of approximately 5 mm 2 with a high spatial resolution down to 700 nm, enabling the capture of information-rich images with detailed sub-cellular features. We demonstrate such capability in delineating molecular characteristics underlying rare dynamic cellular phenomena, such as cancer cell responses to chemotherapy and the emergence of polyploidy in drug-resistant cells. Moreover, the cost-effectiveness and the simplicity of our Omni-Mesoscope democratizes mesoscopic imaging, making it accessible across diverse biomedical research fields. To further demonstrate its versatility, we integrate expansion microscopy to enhance 3D volumetric super-resolution imaging of thicker tissues, opening new avenues for biological exploration at unprecedented scales and resolutions.
Collapse
|
9
|
Glaser A, Chandrashekar J, Vasquez S, Arshadi C, Ouellette N, Jiang X, Baka J, Kovacs G, Woodard M, Seshamani S, Cao K, Clack N, Recknagel A, Grim A, Balaram P, Turschak E, Hooper M, Liddell A, Rohde J, Hellevik A, Takasaki K, Erion Barner L, Logsdon M, Chronopoulos C, de Vries S, Ting J, Perlmutter S, Kalmbach B, Dembrow N, Tasic B, Reid RC, Feng D, Svoboda K. Expansion-assisted selective plane illumination microscopy for nanoscale imaging of centimeter-scale tissues. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.06.08.544277. [PMID: 37425699 PMCID: PMC10327101 DOI: 10.1101/2023.06.08.544277] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/11/2023]
Abstract
Recent advances in tissue processing, labeling, and fluorescence microscopy are providing unprecedented views of the structure of cells and tissues at sub-diffraction resolutions and near single molecule sensitivity, driving discoveries in diverse fields of biology, including neuroscience. Biological tissue is organized over scales of nanometers to centimeters. Harnessing molecular imaging across intact, three-dimensional samples on this scale requires new types of microscopes with larger fields of view and working distance, as well as higher throughput. We present a new expansion-assisted selective plane illumination microscope (ExA-SPIM) with aberration-free 1×1×3 μm optical resolution over a large field of view (10.6×8.0 mm 2 ) and working distance (35 mm) at speeds up to 946 megavoxels/sec. Combined with new tissue clearing and expansion methods, the microscope allows imaging centimeter-scale samples with 250×250×750 nm optical resolution (4× expansion), including entire mouse brains, with high contrast and without sectioning. We illustrate ExA-SPIM by reconstructing individual neurons across the mouse brain, imaging cortico-spinal neurons in the macaque motor cortex, and visualizing axons in human white matter.
Collapse
|
10
|
Yang R, Xiao T, Cheng Y, Li A, Qu J, Liang R, Bao S, Wang X, Wang J, Suo J, Luo Q, Dai Q. Sharing massive biomedical data at magnitudes lower bandwidth using implicit neural function. Proc Natl Acad Sci U S A 2024; 121:e2320870121. [PMID: 38959033 PMCID: PMC11252806 DOI: 10.1073/pnas.2320870121] [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: 11/28/2023] [Accepted: 05/21/2024] [Indexed: 07/04/2024] Open
Abstract
Efficient storage and sharing of massive biomedical data would open up their wide accessibility to different institutions and disciplines. However, compressors tailored for natural photos/videos are rapidly limited for biomedical data, while emerging deep learning-based methods demand huge training data and are difficult to generalize. Here, we propose to conduct Biomedical data compRession with Implicit nEural Function (BRIEF) by representing the target data with compact neural networks, which are data specific and thus have no generalization issues. Benefiting from the strong representation capability of implicit neural function, BRIEF achieves 2[Formula: see text]3 orders of magnitude compression on diverse biomedical data at significantly higher fidelity than existing techniques. Besides, BRIEF is of consistent performance across the whole data volume, and supports customized spatially varying fidelity. BRIEF's multifold advantageous features also serve reliable downstream tasks at low bandwidth. Our approach will facilitate low-bandwidth data sharing and promote collaboration and progress in the biomedical field.
Collapse
Affiliation(s)
- Runzhao Yang
- Department of Automation, Tsinghua University, Beijing100084, China
- Institute of Brain and Cognitive Sciences, Tsinghua University, Beijing100084, China
- Shanghai Artificial Intelligence Laboratory, Shanghai200232, China
| | - Tingxiong Xiao
- Department of Automation, Tsinghua University, Beijing100084, China
| | - Yuxiao Cheng
- Department of Automation, Tsinghua University, Beijing100084, China
| | - Anan Li
- Britton Chance Center and MoE Key Laboratory for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology, Wuhan430074, China
- Huazhong University of Science and Technology-Suzhou Institute for Brainsmatics, Jiangsu Industrial Technology Research Institute, Suzhou215123, China
| | - Jinyuan Qu
- Department of Automation, Tsinghua University, Beijing100084, China
| | - Rui Liang
- School of Biomedical Engineering, Hainan University, Haikou570228, China
| | - Shengda Bao
- Britton Chance Center and MoE Key Laboratory for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology, Wuhan430074, China
| | - Xiaofeng Wang
- Britton Chance Center and MoE Key Laboratory for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology, Wuhan430074, China
| | - Jue Wang
- Department of Automation, Tsinghua University, Beijing100084, China
| | - Jinli Suo
- Department of Automation, Tsinghua University, Beijing100084, China
- Institute of Brain and Cognitive Sciences, Tsinghua University, Beijing100084, China
- Shanghai Artificial Intelligence Laboratory, Shanghai200232, China
| | - Qingming Luo
- School of Biomedical Engineering, Hainan University, Haikou570228, China
| | - Qionghai Dai
- Department of Automation, Tsinghua University, Beijing100084, China
- Institute of Brain and Cognitive Sciences, Tsinghua University, Beijing100084, China
| |
Collapse
|
11
|
Foylan S, Schniete JK, Kölln LS, Dempster J, Hansen CG, Shaw M, Bushell TJ, McConnell G. Mesoscale standing wave imaging. J Microsc 2024; 295:33-41. [PMID: 37156549 DOI: 10.1111/jmi.13189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Revised: 05/03/2023] [Accepted: 05/04/2023] [Indexed: 05/10/2023]
Abstract
Standing wave (SW) microscopy is a method that uses an interference pattern to excite fluorescence from labelled cellular structures and produces high-resolution images of three-dimensional objects in a two-dimensional dataset. SW microscopy is performed with high-magnification, high-numerical aperture objective lenses, and while this results in high-resolution images, the field of view is very small. Here we report upscaling of this interference imaging method from the microscale to the mesoscale using the Mesolens, which has the unusual combination of a low-magnification and high-numerical aperture. With this method, we produce SW images within a field of view of 4.4 mm × 3.0 mm that can readily accommodate over 16,000 cells in a single dataset. We demonstrate the method using both single-wavelength excitation and the multi-wavelength SW method TartanSW. We show application of the method for imaging of fixed and living cells specimens, with the first application of SW imaging to study cells under flow conditions.
Collapse
Affiliation(s)
- Shannan Foylan
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, UK
| | - Jana Katharina Schniete
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, UK
| | - Lisa Sophie Kölln
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, UK
| | - John Dempster
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, UK
| | - Carsten Gram Hansen
- Institute for Regeneration and Repair, Queen's Medical Research Institute, University of Edinburgh Centre for Inflammation Research, Edinburgh, UK
| | - Michael Shaw
- Faculty of Engineering Sciences, Department of Computer Science, University College London, London, UK
- Biometrology Group, National Physical Laboratory, Teddington, UK
| | - Trevor John Bushell
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, UK
| | - Gail McConnell
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, UK
| |
Collapse
|
12
|
Peckham M, Neumann U, Culley S. Introduction to women in microscopy: Volume 2. J Microsc 2024; 295:3-5. [PMID: 38860535 DOI: 10.1111/jmi.13337] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/12/2024]
Affiliation(s)
- Michelle Peckham
- School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, UK
| | - Ulla Neumann
- Max Planck Institute for Plant Breeding Research, Köln, Germany
| | - Siân Culley
- Randall Centre for Cell and Molecular Biophysics, Faculty of Life Sciences and Medicine, King's College London, London, UK
| |
Collapse
|
13
|
Mohammadi FS, Shabani H, Zarei M. Fast and robust feature-based stitching algorithm for microscopic images. Sci Rep 2024; 14:13304. [PMID: 38858367 PMCID: PMC11164998 DOI: 10.1038/s41598-024-61970-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2023] [Accepted: 05/13/2024] [Indexed: 06/12/2024] Open
Abstract
The limited field of view of high-resolution microscopic images hinders the study of biological samples in a single shot. Stitching of microscope images (tiles) captured by the whole-slide imaging (WSI) technique solves this problem. However, stitching is challenging due to the repetitive textures of tissues, the non-informative background part of the slide, and the large number of tiles that impact performance and computational time. To address these challenges, we proposed the Fast and Robust Microscopic Image Stitching (FRMIS) algorithm, which relies on pairwise and global alignment. The speeded up robust features (SURF) were extracted and matched within a small part of the overlapping region to compute the transformation and align two neighboring tiles. In cases where the transformation could not be computed due to an insufficient number of matched features, features were extracted from the entire overlapping region. This enhances the efficiency of the algorithm since most of the computational load is related to pairwise registration and reduces misalignment that may occur by matching duplicated features in tiles with repetitive textures. Then, global alignment was achieved by constructing a weighted graph where the weight of each edge is determined by the normalized inverse of the number of matched features between two tiles. FRMIS has been evaluated on experimental and synthetic datasets from different modalities with different numbers of tiles and overlaps, demonstrating faster stitching time compared to existing algorithms such as the Microscopy Image Stitching Tool (MIST) toolbox. FRMIS outperforms MIST by 481% for bright-field, 259% for phase-contrast, and 282% for fluorescence modalities, while also being robust to uneven illumination.
Collapse
Affiliation(s)
| | - Hasti Shabani
- Institute of Medical Science and Technology, Shahid Beheshti University, Tehran, Iran.
| | - Mojtaba Zarei
- Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| |
Collapse
|
14
|
Vladimirov N, Voigt FF, Naert T, Araujo GR, Cai R, Reuss AM, Zhao S, Schmid P, Hildebrand S, Schaettin M, Groos D, Mateos JM, Bethge P, Yamamoto T, Aerne V, Roebroeck A, Ertürk A, Aguzzi A, Ziegler U, Stoeckli E, Baudis L, Lienkamp SS, Helmchen F. Benchtop mesoSPIM: a next-generation open-source light-sheet microscope for cleared samples. Nat Commun 2024; 15:2679. [PMID: 38538644 PMCID: PMC10973490 DOI: 10.1038/s41467-024-46770-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Accepted: 03/08/2024] [Indexed: 04/04/2024] Open
Abstract
In 2015, we launched the mesoSPIM initiative, an open-source project for making light-sheet microscopy of large cleared tissues more accessible. Meanwhile, the demand for imaging larger samples at higher speed and resolution has increased, requiring major improvements in the capabilities of such microscopes. Here, we introduce the next-generation mesoSPIM ("Benchtop") with a significantly increased field of view, improved resolution, higher throughput, more affordable cost, and simpler assembly compared to the original version. We develop an optical method for testing detection objectives that enables us to select objectives optimal for light-sheet imaging with large-sensor cameras. The improved mesoSPIM achieves high spatial resolution (1.5 µm laterally, 3.3 µm axially) across the entire field of view, magnification up to 20×, and supports sample sizes ranging from sub-mm up to several centimeters while being compatible with multiple clearing techniques. The microscope serves a broad range of applications in neuroscience, developmental biology, pathology, and even physics.
Collapse
Grants
- U01 NS090475 NINDS NIH HHS
- This work was supported by the University Research Priority Program (URPP) “Adaptive Brain Circuits in Development and Learning (AdaBD)” of the University of Zurich (N.V., E.S. and F.H.). Additionally, F.F.V. is supported by an HFSP fellowship (LT00687), T.N. received funding from H2020 Marie Skłodowska-Curie Actions (xenCAKUT - 891127), A.R. and S.H. were supported by a Dutch Science Foundation VIDI Grant (14637), and A.R. was supported by an ERC Starting Grant (MULTICONNECT, 639938). Further funding support came from the Swiss National Science Foundation (SNF grant nos. 31003B-170269, 310030_192617 and CRSII5-18O316 to F.H., 310030_189102 to S.S.L., 200020_204950 to L.B., G.R.A, and V.A.); from an ERC Starting Grant by the European Union’s Horizon 2020 Research and Innovation Programme (grant agreement no. 804474, DiRECT, S.S.L); and the US Brain Initiative (1U01NS090475-01, F.H.).
Collapse
Affiliation(s)
- Nikita Vladimirov
- Brain Research Institute, University of Zurich, Zurich, Switzerland.
- University Research Priority Program (URPP), Adaptive Brain Circuits in Development and Learning, University of Zurich, Zurich, Switzerland.
- Center for Microscopy and Image Analysis (ZMB), University of Zurich, Zurich, Switzerland.
| | - Fabian F Voigt
- Brain Research Institute, University of Zurich, Zurich, Switzerland
- Neuroscience Center Zurich (ZNZ), University of Zurich, Zurich, Switzerland
- Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA, USA
| | - Thomas Naert
- Institute of Anatomy and Zurich Kidney Center (ZKC), University of Zurich, Zurich, Switzerland
| | | | - Ruiyao Cai
- Institute for Tissue Engineering and Regenerative Medicine (iTERM), Helmholtz Center Munich, Neuherberg, Germany
- Institute for Stroke and Dementia Research, Klinikum der Universität München, Ludwig-Maximilians University Munich, Munich, Germany
- Department of Biology, Stanford University, Stanford, CA, USA
| | - Anna Maria Reuss
- Neuroscience Center Zurich (ZNZ), University of Zurich, Zurich, Switzerland
- Institute of Neuropathology, University Hospital Zurich, Zurich, Switzerland
| | - Shan Zhao
- Department of Quantitative Biomedicine, University of Zurich, Zurich, Switzerland
| | - Patricia Schmid
- Institute of Anatomy and Zurich Kidney Center (ZKC), University of Zurich, Zurich, Switzerland
| | - Sven Hildebrand
- Department of Cognitive Neuroscience, Faculty of Psychology & Neuroscience, Maastricht University, Maastricht, The Netherlands
| | - Martina Schaettin
- Neuroscience Center Zurich (ZNZ), University of Zurich, Zurich, Switzerland
- Department of Molecular Life Sciences, University of Zurich, Zurich, Switzerland
| | - Dominik Groos
- Brain Research Institute, University of Zurich, Zurich, Switzerland
| | - José María Mateos
- Center for Microscopy and Image Analysis (ZMB), University of Zurich, Zurich, Switzerland
| | - Philipp Bethge
- Brain Research Institute, University of Zurich, Zurich, Switzerland
- Neuroscience Center Zurich (ZNZ), University of Zurich, Zurich, Switzerland
| | - Taiyo Yamamoto
- Institute of Anatomy and Zurich Kidney Center (ZKC), University of Zurich, Zurich, Switzerland
| | - Valentino Aerne
- Department of Physics, University of Zurich, Zurich, Switzerland
| | - Alard Roebroeck
- Department of Cognitive Neuroscience, Faculty of Psychology & Neuroscience, Maastricht University, Maastricht, The Netherlands
| | - Ali Ertürk
- Institute for Tissue Engineering and Regenerative Medicine (iTERM), Helmholtz Center Munich, Neuherberg, Germany
- Institute for Stroke and Dementia Research, Klinikum der Universität München, Ludwig-Maximilians University Munich, Munich, Germany
| | - Adriano Aguzzi
- Neuroscience Center Zurich (ZNZ), University of Zurich, Zurich, Switzerland
- Institute of Neuropathology, University Hospital Zurich, Zurich, Switzerland
| | - Urs Ziegler
- Center for Microscopy and Image Analysis (ZMB), University of Zurich, Zurich, Switzerland
| | - Esther Stoeckli
- University Research Priority Program (URPP), Adaptive Brain Circuits in Development and Learning, University of Zurich, Zurich, Switzerland
- Neuroscience Center Zurich (ZNZ), University of Zurich, Zurich, Switzerland
- Department of Molecular Life Sciences, University of Zurich, Zurich, Switzerland
| | - Laura Baudis
- Department of Physics, University of Zurich, Zurich, Switzerland
| | - Soeren S Lienkamp
- Institute of Anatomy and Zurich Kidney Center (ZKC), University of Zurich, Zurich, Switzerland
| | - Fritjof Helmchen
- Brain Research Institute, University of Zurich, Zurich, Switzerland.
- University Research Priority Program (URPP), Adaptive Brain Circuits in Development and Learning, University of Zurich, Zurich, Switzerland.
- Neuroscience Center Zurich (ZNZ), University of Zurich, Zurich, Switzerland.
| |
Collapse
|
15
|
Ichimura T, Kakizuka T, Sato Y, Fujioka Y, Ohba Y, Horikawa K, Nagai T. Strength in numbers: Unleashing the potential of trans-scale scope AMATERAS for massive cell quantification. Biophys Physicobiol 2024; 21:e211017. [PMID: 39175860 PMCID: PMC11338690 DOI: 10.2142/biophysico.bppb-v21.s017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Accepted: 03/22/2024] [Indexed: 08/24/2024] Open
Abstract
Singularity biology is a scientific field that targets drastic state changes in multicellular systems, aiming to discover the key cells that induce the state change and investigate the mechanisms behind them. To achieve this goal, we developed a trans-scale optical imaging system (trans-scale scope), that is capable of capturing both macroscale changes across the entire system and the micro-scale behavior of individual cells, surpassing the cell observation capabilities of traditional microscopes. We developed two units of the trans-scale scope, named AMATERAS-1 and -2, which demonstrated the ability to observe multicellular systems consisting of over one million cells in a single field of view with sub-cellular resolution. This flagship instrument has been used to observe the dynamics of various cell species, with the advantage of being able to observe a large number of cells, allowing the detection and analysis of rare events and cells such as leader cells in multicellular pattern formation and cells that spontaneously initiate calcium waves. In this paper, we present the design concept of AMATERAS, the optical configuration, and several examples of observations, and demonstrate how the strength-in-numbers works in life sciences.
Collapse
Affiliation(s)
- Taro Ichimura
- Transdimensional Life Imaging Division, Institute for Open and Transdisciplinary Research Initiatives, Osaka University, Suita, Osaka 565-0871, Japan
| | - Taishi Kakizuka
- Department of Biomolecular Science and Engineering, SANKEN, Osaka University, Ibaraki, Osaka 567-0047, Japan
| | - Yuki Sato
- Department of Anatomy and Cell Biology, Graduate School of Medical Sciences, Kyushu University, Higashi-ku, Fukuoka 812-8582, Japan
| | - Yoichiro Fujioka
- Department of Cell Physiology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Hokkaido 060-8638, Japan
| | - Yusuke Ohba
- Department of Cell Physiology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Hokkaido 060-8638, Japan
| | - Kazuki Horikawa
- Department of Optical Imaging, Advanced Research Promotion Center, Tokushima University, Tokushima 770-8503, Japan
| | - Takeharu Nagai
- Transdimensional Life Imaging Division, Institute for Open and Transdisciplinary Research Initiatives, Osaka University, Suita, Osaka 565-0871, Japan
- Department of Biomolecular Science and Engineering, SANKEN, Osaka University, Ibaraki, Osaka 567-0047, Japan
- Research Institute for Electronic Science, Hokkaido University, Sapporo, Hokkaido 001-0020, Japan
| |
Collapse
|
16
|
Watanabe TM, Shiina T. Visualizing Singularity Phenomenon. Biophys Physicobiol 2024; 21:e211011. [PMID: 39175854 PMCID: PMC11338682 DOI: 10.2142/biophysico.bppb-v21.s011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Accepted: 02/20/2024] [Indexed: 08/24/2024] Open
Affiliation(s)
- Tomonobu M Watanabe
- Department of Stem Cell Biology, Research Institute for Radiation Biology and Medicine, Hiroshima University, Hiroshima 734-8553, Japan
- Laboratory for Comprehensive Bioimaging, RIKEN Center for Biosystems Dynamics Research (BDR), Kobe, Hyogo 650-0047, Japan
| | - Tsuyoshi Shiina
- Advanced Biomedical Engineering Systems Laboratory, SIT Research Laboratories, Graduate School of Engineering and Science, Shibaura Institute of Technology, Koto-ku, Tokyo 135-8548, Japan
| |
Collapse
|
17
|
Amos WB. Principles of microscopy for ophthalmologists. Eye (Lond) 2024:10.1038/s41433-024-02970-0. [PMID: 38374367 DOI: 10.1038/s41433-024-02970-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 01/02/2024] [Accepted: 01/26/2024] [Indexed: 02/21/2024] Open
Abstract
This short review begins with the theories of Airy, Rayleigh and Abbe on microscope resolution. Next, the principal developments in microscopy in the last half-century are examined for relevance to ophthalmology: confocal microscopy, photoactivation light microscopy (PALM), stochastic optical reconstruction microscopy (STORM), stimulated emission depletion (STED), structured illumination (SI), 2-photon and multiphoton excitation microscopy with a focused beam. Except for confocal, these are difficult to apply to the eye in vivo, as are the interference methods available in microscopes. However, interferometry in the form of coherence tomography is now a major ophthalmic method which has diverged from microscopy. Multiphoton excitation microscopy with an unfocussed beam is a new, low-damage microscope method so-far not exploited in ophthalmoscopy. The Mesolens, which throws off the historic limitations in microscopy set by the human eye, is described as a possible future aid to ophthalmology of the anterior eye.
Collapse
Affiliation(s)
- William Bradshaw Amos
- Structural Studies Division, UKRI MRC Laboratory of Molecular Biology, Francis Crick Avenue, Cambridge, CB2 0QH, UK.
| |
Collapse
|
18
|
Voigt FF, Reuss AM, Naert T, Hildebrand S, Schaettin M, Hotz AL, Whitehead L, Bahl A, Neuhauss SCF, Roebroeck A, Stoeckli ET, Lienkamp SS, Aguzzi A, Helmchen F. Reflective multi-immersion microscope objectives inspired by the Schmidt telescope. Nat Biotechnol 2024; 42:65-71. [PMID: 36997681 PMCID: PMC10791577 DOI: 10.1038/s41587-023-01717-8] [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: 09/20/2022] [Accepted: 02/20/2023] [Indexed: 04/03/2023]
Abstract
Imaging large, cleared samples requires microscope objectives that combine a large field of view (FOV) with a long working distance (WD) and a high numerical aperture (NA). Ideally, such objectives should be compatible with a wide range of immersion media, which is challenging to achieve with conventional lens-based objective designs. Here we introduce the multi-immersion 'Schmidt objective' consisting of a spherical mirror and an aspherical correction plate as a solution to this problem. We demonstrate that a multi-photon variant of the Schmidt objective is compatible with all homogeneous immersion media and achieves an NA of 1.08 at a refractive index of 1.56, 1.1-mm FOV and 11-mm WD. We highlight its versatility by imaging cleared samples in various media ranging from air and water to benzyl alcohol/benzyl benzoate, dibenzyl ether and ethyl cinnamate and by imaging of neuronal activity in larval zebrafish in vivo. In principle, the concept can be extended to any imaging modality, including wide-field, confocal and light-sheet microscopy.
Collapse
Affiliation(s)
- Fabian F Voigt
- Brain Research Institute, University of Zurich, Zurich, Switzerland.
- Neuroscience Center Zurich, University of Zurich, Zurich, Switzerland.
- Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA, USA.
| | - Anna Maria Reuss
- Neuroscience Center Zurich, University of Zurich, Zurich, Switzerland
- Institute of Neuropathology, University Hospital Zurich, Zurich, Switzerland
| | - Thomas Naert
- Institute of Anatomy, University of Zurich, Zurich, Switzerland
| | - Sven Hildebrand
- Department of Cognitive Neuroscience, Faculty of Psychology & Neuroscience, Maastricht University, Maastricht, the Netherlands
| | - Martina Schaettin
- Neuroscience Center Zurich, University of Zurich, Zurich, Switzerland
- Department of Molecular Life Sciences, University of Zurich, Zurich, Switzerland
| | - Adriana L Hotz
- Neuroscience Center Zurich, University of Zurich, Zurich, Switzerland
- Department of Molecular Life Sciences, University of Zurich, Zurich, Switzerland
| | - Lachlan Whitehead
- Walter and Eliza Hall Institute of Medical Research, Melbourne, Victoria, Australia
- Department of Medical Biology, University of Melbourne, Melbourne, Victoria, Australia
| | - Armin Bahl
- Centre for the Advanced Study of Collective Behaviour, University of Konstanz, Konstanz, Germany
| | - Stephan C F Neuhauss
- Neuroscience Center Zurich, University of Zurich, Zurich, Switzerland
- Department of Molecular Life Sciences, University of Zurich, Zurich, Switzerland
| | - Alard Roebroeck
- Department of Cognitive Neuroscience, Faculty of Psychology & Neuroscience, Maastricht University, Maastricht, the Netherlands
| | - Esther T Stoeckli
- Neuroscience Center Zurich, University of Zurich, Zurich, Switzerland
- Department of Molecular Life Sciences, University of Zurich, Zurich, Switzerland
- University Research Priority Program (URPP), Adaptive Brain Circuits in Development and Learning (AdaBD), University of Zürich, Zurich, Switzerland
| | | | - Adriano Aguzzi
- Neuroscience Center Zurich, University of Zurich, Zurich, Switzerland
- Institute of Neuropathology, University Hospital Zurich, Zurich, Switzerland
| | - Fritjof Helmchen
- Brain Research Institute, University of Zurich, Zurich, Switzerland
- Neuroscience Center Zurich, University of Zurich, Zurich, Switzerland
- University Research Priority Program (URPP), Adaptive Brain Circuits in Development and Learning (AdaBD), University of Zürich, Zurich, Switzerland
| |
Collapse
|
19
|
Rooney LM, Dupuy LX, Hoskisson PA, McConnell G. Construction and characterisation of a structured, tuneable, and transparent 3D culture platform for soil bacteria. MICROBIOLOGY (READING, ENGLAND) 2024; 170:001429. [PMID: 38289644 PMCID: PMC10866023 DOI: 10.1099/mic.0.001429] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Accepted: 01/19/2024] [Indexed: 02/01/2024]
Abstract
We have developed a tuneable workflow for the study of soil microbes in an imitative 3D soil environment that is compatible with routine and advanced optical imaging, is chemically customisable, and is reliably refractive index matched based on the carbon catabolism of the study organism. We demonstrate our transparent soil pipeline with two representative soil organisms, Bacillus subtilis and Streptomyces coelicolor, and visualise their colonisation behaviours using fluorescence microscopy and mesoscopy. This spatially structured, 3D approach to microbial culture has the potential to further study the behaviour of bacteria in conditions matching their native environment and could be expanded to study microbial interactions, such as competition and warfare.
Collapse
Affiliation(s)
- Liam M. Rooney
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, UK
| | - Lionel X. Dupuy
- The James Hutton Institute, Invergowrie, Dundee, DD2 5DA, UK
- Present address: Department of Conservation of Natural Resources, Neiker, Basque Institute for Agricultural Research and Development, Derio, Spain
- Present address: Ikerbasque, Basque Foundation for Science, Bilbao, Spain
| | - Paul A. Hoskisson
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, UK
| | - Gail McConnell
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, UK
| |
Collapse
|
20
|
Baxter KJ, Sargison FA, Fitzgerald JR, McConnell G, Hoskisson PA. Time-lapse mesoscopy of Candida albicans and Staphylococcus aureus dual-species biofilms reveals a structural role for the hyphae of C. albicans in biofilm formation. MICROBIOLOGY (READING, ENGLAND) 2024; 170:001426. [PMID: 38261525 PMCID: PMC10866020 DOI: 10.1099/mic.0.001426] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Accepted: 01/02/2024] [Indexed: 01/25/2024]
Abstract
Polymicrobial infection with Candida albicans and Staphylococcus aureus may result in a concomitant increase in virulence and resistance to antimicrobial drugs. This enhanced pathogenicity phenotype is mediated by numerous factors, including metabolic processes and direct interaction of S. aureus with C. albicans hyphae. The overall structure of biofilms is known to contribute to their recalcitrance to treatment, although the dynamics of direct interaction between species and how it contributes to pathogenicity is poorly understood. To address this, a novel time-lapse mesoscopic optical imaging method was developed to enable the formation of C. albicans/S. aureus whole dual-species biofilms to be followed. It was found that yeast-form or hyphal-form C. albicans in the biofilm founder population profoundly affects the structure of the biofilm as it matures. Different sub-populations of C. albicans and S. aureus arise within each biofilm as a result of the different C. albicans morphotypes, resulting in distinct sub-regions. These data reveal that C. albicans cell morphology is pivotal in the development of global biofilm architecture and the emergence of colony macrostructures and may temporally influence synergy in infection.
Collapse
Affiliation(s)
- Katherine J. Baxter
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, 161 Cathedral Street, Glasgow, G4 0RE, UK
| | - Fiona A. Sargison
- The Roslin Institute, University of Edinburgh, Easter Bush Campus, Edinburgh, EH25 9RG, UK
| | - J. Ross Fitzgerald
- The Roslin Institute, University of Edinburgh, Easter Bush Campus, Edinburgh, EH25 9RG, UK
| | - Gail McConnell
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, 161 Cathedral Street, Glasgow, G4 0RE, UK
| | - Paul A. Hoskisson
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, 161 Cathedral Street, Glasgow, G4 0RE, UK
| |
Collapse
|
21
|
Imaging the biological microcosmos with a tiny telescope. Nat Biotechnol 2024; 42:36-37. [PMID: 37002344 DOI: 10.1038/s41587-023-01740-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/03/2023]
|
22
|
Vladimirov N, Voigt FF, Naert T, Araujo GR, Cai R, Reuss AM, Zhao S, Schmid P, Hildebrand S, Schaettin M, Groos D, Mateos JM, Bethge P, Yamamoto T, Aerne V, Roebroeck A, Ertürk A, Aguzzi A, Ziegler U, Stoeckli E, Baudis L, Lienkamp SS, Helmchen F. The Benchtop mesoSPIM: a next-generation open-source light-sheet microscope for large cleared samples. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.06.16.545256. [PMID: 38168219 PMCID: PMC10760166 DOI: 10.1101/2023.06.16.545256] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/05/2024]
Abstract
In 2015, we launched the mesoSPIM initiative (www.mesospim.org), an open-source project for making light-sheet microscopy of large cleared tissues more accessible. Meanwhile, the demand for imaging larger samples at higher speed and resolution has increased, requiring major improvements in the capabilities of light-sheet microscopy. Here, we introduce the next-generation mesoSPIM ("Benchtop") with significantly increased field of view, improved resolution, higher throughput, more affordable cost and simpler assembly compared to the original version. We developed a new method for testing objectives, enabling us to select detection objectives optimal for light-sheet imaging with large-sensor sCMOS cameras. The new mesoSPIM achieves high spatial resolution (1.5 μm laterally, 3.3 μm axially) across the entire field of view, a magnification up to 20x, and supports sample sizes ranging from sub-mm up to several centimetres, while being compatible with multiple clearing techniques. The new microscope serves a broad range of applications in neuroscience, developmental biology, and even physics.
Collapse
Affiliation(s)
- Nikita Vladimirov
- Brain Research Institute, University of Zurich, Zurich, Switzerland
- University Research Priority Program (URPP) Adaptive Brain Circuits in Development and Learning (AdaBD), University of Zurich, Zurich, Switzerland
- Center for Microscopy and Image Analysis (ZMB), University of Zurich, Zurich, Switzerland
| | - Fabian F. Voigt
- Brain Research Institute, University of Zurich, Zurich, Switzerland
- Neuroscience Center Zurich, University of Zurich, Zurich, Switzerland
- Present address: Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA, USA
| | - Thomas Naert
- Institute of Anatomy and Zurich Kidney Center (ZKC), University of Zurich, Zurich, Switzerland
| | | | - Ruiyao Cai
- Present address: Department of Biology, Stanford University, Stanford, CA, USA
- Institute for Tissue Engineering and Regenerative Medicine (iTERM), Helmholtz Center Munich, Neuherberg, Germany
- Institute for Stroke and Dementia Research, Klinikum der Universität München, Ludwig-Maximilians University Munich, Munich, German
| | - Anna Maria Reuss
- Neuroscience Center Zurich, University of Zurich, Zurich, Switzerland
- Institute of Neuropathology, University Hospital Zurich, Zurich, Switzerland
| | - Shan Zhao
- Department of Quantitative Biomedicine, University of Zurich, Zurich, Switzerland
| | - Patricia Schmid
- Institute of Anatomy and Zurich Kidney Center (ZKC), University of Zurich, Zurich, Switzerland
| | - Sven Hildebrand
- Department of Cognitive Neuroscience, Faculty of Psychology & Neuroscience, Maastricht University, Maastricht, the Netherlands
| | - Martina Schaettin
- Neuroscience Center Zurich, University of Zurich, Zurich, Switzerland
- Department of Molecular Life Sciences, University of Zurich, Zurich, Switzerland
| | - Dominik Groos
- Brain Research Institute, University of Zurich, Zurich, Switzerland
| | - José María Mateos
- Center for Microscopy and Image Analysis (ZMB), University of Zurich, Zurich, Switzerland
| | - Philipp Bethge
- Brain Research Institute, University of Zurich, Zurich, Switzerland
- Neuroscience Center Zurich, University of Zurich, Zurich, Switzerland
| | - Taiyo Yamamoto
- Institute of Anatomy and Zurich Kidney Center (ZKC), University of Zurich, Zurich, Switzerland
| | - Valentino Aerne
- Department of Physics, University of Zurich, Zurich, Switzerland
| | - Alard Roebroeck
- Department of Cognitive Neuroscience, Faculty of Psychology & Neuroscience, Maastricht University, Maastricht, the Netherlands
| | - Ali Ertürk
- Institute for Tissue Engineering and Regenerative Medicine (iTERM), Helmholtz Center Munich, Neuherberg, Germany
- Institute for Stroke and Dementia Research, Klinikum der Universität München, Ludwig-Maximilians University Munich, Munich, German
| | - Adriano Aguzzi
- Neuroscience Center Zurich, University of Zurich, Zurich, Switzerland
- Institute of Neuropathology, University Hospital Zurich, Zurich, Switzerland
| | - Urs Ziegler
- Center for Microscopy and Image Analysis (ZMB), University of Zurich, Zurich, Switzerland
| | - Esther Stoeckli
- University Research Priority Program (URPP) Adaptive Brain Circuits in Development and Learning (AdaBD), University of Zurich, Zurich, Switzerland
- Neuroscience Center Zurich, University of Zurich, Zurich, Switzerland
- Department of Molecular Life Sciences, University of Zurich, Zurich, Switzerland
| | - Laura Baudis
- Department of Physics, University of Zurich, Zurich, Switzerland
| | - Soeren S. Lienkamp
- Institute of Anatomy and Zurich Kidney Center (ZKC), University of Zurich, Zurich, Switzerland
| | - Fritjof Helmchen
- Brain Research Institute, University of Zurich, Zurich, Switzerland
- University Research Priority Program (URPP) Adaptive Brain Circuits in Development and Learning (AdaBD), University of Zurich, Zurich, Switzerland
- Neuroscience Center Zurich, University of Zurich, Zurich, Switzerland
| |
Collapse
|
23
|
Smith TD. Vespers and vampires: A lifelong microscopic search for the smallest of things. Anat Rec (Hoboken) 2023; 306:2670-2680. [PMID: 35202504 DOI: 10.1002/ar.24907] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Revised: 02/15/2022] [Accepted: 02/15/2022] [Indexed: 11/11/2022]
Affiliation(s)
- Timothy D Smith
- School of Physical Therapy, Slippery Rock University, Slippery Rock, Pennsylvania, USA
| |
Collapse
|
24
|
Balasubramanian H, Hobson CM, Chew TL, Aaron JS. Imagining the future of optical microscopy: everything, everywhere, all at once. Commun Biol 2023; 6:1096. [PMID: 37898673 PMCID: PMC10613274 DOI: 10.1038/s42003-023-05468-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Accepted: 10/16/2023] [Indexed: 10/30/2023] Open
Abstract
The optical microscope has revolutionized biology since at least the 17th Century. Since then, it has progressed from a largely observational tool to a powerful bioanalytical platform. However, realizing its full potential to study live specimens is hindered by a daunting array of technical challenges. Here, we delve into the current state of live imaging to explore the barriers that must be overcome and the possibilities that lie ahead. We venture to envision a future where we can visualize and study everything, everywhere, all at once - from the intricate inner workings of a single cell to the dynamic interplay across entire organisms, and a world where scientists could access the necessary microscopy technologies anywhere.
Collapse
Affiliation(s)
| | - Chad M Hobson
- Advanced Imaging Center; Howard Hughes Medical Institute Janelia Research Campus, Ashburn, VA, 20147, USA
| | - Teng-Leong Chew
- Advanced Imaging Center; Howard Hughes Medical Institute Janelia Research Campus, Ashburn, VA, 20147, USA
| | - Jesse S Aaron
- Advanced Imaging Center; Howard Hughes Medical Institute Janelia Research Campus, Ashburn, VA, 20147, USA.
| |
Collapse
|
25
|
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
|
26
|
Malik R, Khare K. Single-shot extended field of view imaging using point spread function engineering. JOURNAL OF THE OPTICAL SOCIETY OF AMERICA. A, OPTICS, IMAGE SCIENCE, AND VISION 2023; 40:1066-1075. [PMID: 37706760 DOI: 10.1364/josaa.484734] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/02/2023] [Accepted: 04/17/2023] [Indexed: 09/15/2023]
Abstract
We present a single-shot computational imaging system employing pupil phase engineering to extend the field of view (FOV) beyond the physical sensor limit. Our approach uses a point spread function in the form of a multiple-point impulse response (MPIR). Unlike the traditional point-to-point imaging model used by most traditional optical imaging systems, the proposed MPIR model can collect information from within and outside the sensor boundary. The detected raw image despite being scrambled can be decoded via a sparse optimization algorithm to get extended FOV imaging performance. We provide a thorough analysis of MPIR design regarding the number of impulses and their spatial extent. Increasing the number of impulses in MPIR of a given spatial extent leads to better information gathering within the detector region; however, it also reduces contrast in the raw data. Therefore, a trade-off between increasing the information and keeping adequate contrast in the detected data is necessary to achieve high-quality reconstruction. We first illustrate this trade-off with a simulation study and present experimental results on a suitably designed extended FOV imaging system. We demonstrate reconstructed images with a 4× gain in pixels over the native detection area without loss of spatial resolution. The proposed system design considerations are generic and can be applied to various imaging systems for extended FOV performance.
Collapse
|
27
|
Mohanan S, Corbett AD. Understanding the limits of remote focusing. OPTICS EXPRESS 2023; 31:16281-16294. [PMID: 37157710 DOI: 10.1364/oe.485635] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/10/2023]
Abstract
It has previously been demonstrated in both simulation and experiment that well aligned remote focusing microscopes exhibit residual spherical aberration outside the focal plane. In this work, compensation of the residual spherical aberration is provided by the correction collar on the primary objective, controlled by a high precision stepper motor. A Shack-Hartmann wave front sensor is used to demonstrate the magnitude of the spherical aberration generated by the correction collar matches that predicted by an optical model of the objective lens. The limited impact of spherical aberration compensation on the diffraction limited range of the remote focusing system is described through a consideration of both on-axis and off-axis comatic and astigmatic aberrations, which are an inherent feature of remote focusing microscopes.
Collapse
|
28
|
Cremin K, Duxbury SJN, Rosko J, Soyer OS. Formation and emergent dynamics of spatially organized microbial systems. Interface Focus 2023; 13:20220062. [PMID: 36789239 PMCID: PMC9912014 DOI: 10.1098/rsfs.2022.0062] [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: 10/25/2022] [Accepted: 12/19/2022] [Indexed: 02/12/2023] Open
Abstract
Spatial organization is the norm rather than the exception in the microbial world. While the study of microbial physiology has been dominated by studies in well-mixed cultures, there is now increasing interest in understanding the role of spatial organization in microbial physiology, coexistence and evolution. Where studied, spatial organization has been shown to influence all three of these aspects. In this mini review and perspective article, we emphasize that the dynamics within spatially organized microbial systems (SOMS) are governed by feedbacks between local physico-chemical conditions, cell physiology and movement, and evolution. These feedbacks can give rise to emergent dynamics, which need to be studied through a combination of spatio-temporal measurements and mathematical models. We highlight the initial formation of SOMS and their emergent dynamics as two open areas of investigation for future studies. These studies will benefit from the development of model systems that can mimic natural ones in terms of species composition and spatial structure.
Collapse
Affiliation(s)
- Kelsey Cremin
- School of Life Sciences, University of Warwick, Coventry CV4 7AL, UK
| | | | - Jerko Rosko
- School of Life Sciences, University of Warwick, Coventry CV4 7AL, UK
| | - Orkun S. Soyer
- School of Life Sciences, University of Warwick, Coventry CV4 7AL, UK
| |
Collapse
|
29
|
Yang X, Harfouche M, Zhou KC, Kreiss L, Xu S, Chandra Konda P, Kim K, Horstmeyer R. Multi-modal imaging using a cascaded microscope design. OPTICS LETTERS 2023; 48:1658-1661. [PMID: 37221734 DOI: 10.1364/ol.471380] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Accepted: 01/13/2023] [Indexed: 05/25/2023]
Abstract
We present a multi-modal fiber array snapshot technique (M-FAST) based on an array of 96 compact cameras placed behind a primary objective lens and a fiber bundle array. Our technique is capable of large-area, high-resolution, multi-channel video acquisition. The proposed design provides two key improvements to prior cascaded imaging system approaches: a novel optical arrangement that accommodates the use of planar camera arrays, and a new ability to acquire multi-modal image data acquisition. M-FAST is a multi-modal, scalable imaging system that can acquire snapshot dual-channel fluorescence images as well as differential phase contrast measurements over a large 6.59 mm × 9.74 mm field-of-view at 2.2-μm center full-pitch resolution.
Collapse
|
30
|
Nöbauer T, Zhang Y, Kim H, Vaziri A. Mesoscale volumetric light field (MesoLF) imaging of neuroactivity across cortical areas at 18 Hz. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.20.533476. [PMID: 36993596 PMCID: PMC10055306 DOI: 10.1101/2023.03.20.533476] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/31/2023]
Abstract
Various implementations of mesoscopes provide optical access for calcium imaging across multi-millimeter fields-of-view (FOV) in the mammalian brain. However, capturing the activity of the neuronal population within such FOVs near-simultaneously and in a volumetric fashion has remained challenging since approaches for imaging scattering brain tissues typically are based on sequential acquisition. Here, we present a modular, mesoscale light field (MesoLF) imaging hardware and software solution that allows recording from thousands of neurons within volumes of 4000 × 200 μm, located at up to 400 μm depth in the mouse cortex, at 18 volumes per second. Our optical design and computational approach enable up to hour-long recording of ~10,000 neurons across multiple cortical areas in mice using workstation-grade computing resources.
Collapse
Affiliation(s)
- Tobias Nöbauer
- Laboratory of Neurotechnology and Biophysics, The Rockefeller University, New York, NY, USA
| | - Yuanlong Zhang
- Laboratory of Neurotechnology and Biophysics, The Rockefeller University, New York, NY, USA
- Department of Automation, Tsinghua University, Beijing, China
| | - Hyewon Kim
- Laboratory of Neurotechnology and Biophysics, The Rockefeller University, New York, NY, USA
| | - Alipasha Vaziri
- Laboratory of Neurotechnology and Biophysics, The Rockefeller University, New York, NY, USA
- The Kavli Neural Systems Institute, The Rockefeller University, New York, NY, USA
| |
Collapse
|
31
|
Nelson MS, Liu Y, Wilson HM, Li B, Rosado-Mendez IM, Rogers JD, Block WF, Eliceiri KW. Multiscale Label-Free Imaging of Fibrillar Collagen in the Tumor Microenvironment. Methods Mol Biol 2023; 2614:187-235. [PMID: 36587127 DOI: 10.1007/978-1-0716-2914-7_13] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
With recent advances in cancer therapeutics, there is a great need for improved imaging methods for characterizing cancer onset and progression in a quantitative and actionable way. Collagen, the most abundant extracellular matrix protein in the tumor microenvironment (and the body in general), plays a multifaceted role, both hindering and promoting cancer invasion and progression. Collagen deposition can defend the tumor with immunosuppressive effects, while aligned collagen fiber structures can enable tumor cell migration, aiding invasion and metastasis. Given the complex role of collagen fiber organization and topology, imaging has been a tool of choice to characterize these changes on multiple spatial scales, from the organ and tumor scale to cellular and subcellular level. Macroscale density already aids in the detection and diagnosis of solid cancers, but progress is being made to integrate finer microscale features into the process. Here we review imaging modalities ranging from optical methods of second harmonic generation (SHG), polarized light microscopy (PLM), and optical coherence tomography (OCT) to the medical imaging approaches of ultrasound and magnetic resonance imaging (MRI). These methods have enabled scientists and clinicians to better understand the impact collagen structure has on the tumor environment, at both the bulk scale (density) and microscale (fibrillar structure) levels. We focus on imaging methods with the potential to both examine the collagen structure in as natural a state as possible and still be clinically amenable, with an emphasis on label-free strategies, exploiting intrinsic optical properties of collagen fibers.
Collapse
Affiliation(s)
- Michael S Nelson
- Center for Quantitative Cell Imaging, University of Wisconsin-Madison, Madison, WI, USA.,Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, USA
| | - Yuming Liu
- Center for Quantitative Cell Imaging, University of Wisconsin-Madison, Madison, WI, USA
| | - Helen M Wilson
- Center for Quantitative Cell Imaging, University of Wisconsin-Madison, Madison, WI, USA.,Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, USA
| | - Bin Li
- Center for Quantitative Cell Imaging, University of Wisconsin-Madison, Madison, WI, USA.,Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, USA.,Morgridge Institute for Research, Madison, WI, USA
| | - Ivan M Rosado-Mendez
- Department of Medical Physics, University of Wisconsin-Madison, Madison, WI, USA
| | - Jeremy D Rogers
- Morgridge Institute for Research, Madison, WI, USA.,McPherson Eye Research Institute, University of Wisconsin-Madison, Madison, WI, USA
| | - Walter F Block
- Department of Medical Physics, University of Wisconsin-Madison, Madison, WI, USA
| | - Kevin W Eliceiri
- Center for Quantitative Cell Imaging, University of Wisconsin-Madison, Madison, WI, USA. .,Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, USA. .,Morgridge Institute for Research, Madison, WI, USA. .,Department of Medical Physics, University of Wisconsin-Madison, Madison, WI, USA. .,McPherson Eye Research Institute, University of Wisconsin-Madison, Madison, WI, USA.
| |
Collapse
|
32
|
Shao W, Chang M, Emmerich K, Kanold PO, Mumm JS, Yi J. Mesoscopic oblique plane microscopy with a diffractive light-sheet for large-scale 4D cellular resolution imaging. OPTICA 2022; 9:1374-1385. [PMID: 38384442 PMCID: PMC10881189 DOI: 10.1364/optica.471101] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Accepted: 10/31/2022] [Indexed: 02/23/2024]
Abstract
Fundamental understanding of large-scale dynamic connectivity within a living organism requires volumetric imaging over a large field of view (FOV) at biologically relevant speed and resolution. However, most microscopy methods make trade-offs between FOV and axial resolution, making it challenging to observe highly dynamic processes at cellular resolution in 3D across mesoscopic scales (e.g., whole zebrafish larva). To overcome this limitation, we have developed mesoscopic oblique plane microscopy (Meso-OPM) with a diffractive light sheet. By augmenting the illumination angle of the light sheet with a transmission grating, we improved the axial resolution approximately sixfold over existing methods and approximately twofold beyond the diffraction limitation of the primary objective lens. We demonstrated a FOV up to 5.4 mm × 3.3 mm with resolution of 2.5 μm × 3 μm × 6 μm, allowing volumetric imaging of 3D cellular structures with a single scan. Applying Meso-OPM for in vivo imaging of zebrafish larvae, we report here in toto whole-body volumetric recordings of neuronal activity at 2 Hz volume rate and whole-body volumetric recordings of blood flow dynamics at 5 Hz with 3D cellular resolution.
Collapse
Affiliation(s)
- Wenjun Shao
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, Maryland 21231, USA
- Department of Ophthalmology, Johns Hopkins University, Baltimore, Maryland 21231, USA
| | - Minzi Chang
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, Maryland 21231, USA
| | - Kevin Emmerich
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, Maryland 21231, USA
| | - Patrick O. Kanold
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, Maryland 21231, USA
| | - Jeff S. Mumm
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, Maryland 21231, USA
| | - Ji Yi
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, Maryland 21231, USA
- Department of Ophthalmology, Johns Hopkins University, Baltimore, Maryland 21231, USA
| |
Collapse
|
33
|
Thomson EE, Harfouche M, Kim K, Konda PC, Seitz CW, Cooke C, Xu S, Jacobs WS, Blazing R, Chen Y, Sharma S, Dunn TW, Park J, Horstmeyer RW, Naumann EA. Gigapixel imaging with a novel multi-camera array microscope. eLife 2022; 11:e74988. [PMID: 36515989 PMCID: PMC9917455 DOI: 10.7554/elife.74988] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Accepted: 11/23/2022] [Indexed: 12/15/2022] Open
Abstract
The dynamics of living organisms are organized across many spatial scales. However, current cost-effective imaging systems can measure only a subset of these scales at once. We have created a scalable multi-camera array microscope (MCAM) that enables comprehensive high-resolution recording from multiple spatial scales simultaneously, ranging from structures that approach the cellular scale to large-group behavioral dynamics. By collecting data from up to 96 cameras, we computationally generate gigapixel-scale images and movies with a field of view over hundreds of square centimeters at an optical resolution of 18 µm. This allows us to observe the behavior and fine anatomical features of numerous freely moving model organisms on multiple spatial scales, including larval zebrafish, fruit flies, nematodes, carpenter ants, and slime mold. Further, the MCAM architecture allows stereoscopic tracking of the z-position of organisms using the overlapping field of view from adjacent cameras. Overall, by removing the bottlenecks imposed by single-camera image acquisition systems, the MCAM provides a powerful platform for investigating detailed biological features and behavioral processes of small model organisms across a wide range of spatial scales.
Collapse
Affiliation(s)
- Eric E Thomson
- Department of Neurobiology, Duke School of MedicineDurhamUnited States
| | | | - Kanghyun Kim
- Biomedical Engineering, Duke UniversityDurhamUnited States
| | - Pavan C Konda
- Biomedical Engineering, Duke UniversityDurhamUnited States
| | - Catherine W Seitz
- Department of Neurobiology, Duke School of MedicineDurhamUnited States
| | - Colin Cooke
- Biomedical Engineering, Duke UniversityDurhamUnited States
| | - Shiqi Xu
- Biomedical Engineering, Duke UniversityDurhamUnited States
| | - Whitney S Jacobs
- Department of Neurobiology, Duke School of MedicineDurhamUnited States
| | - Robin Blazing
- Department of Neurobiology, Duke School of MedicineDurhamUnited States
| | - Yang Chen
- Department of Neurobiology, Duke School of MedicineDurhamUnited States
| | | | - Timothy W Dunn
- Biomedical Engineering, Duke UniversityDurhamUnited States
| | | | - Roarke W Horstmeyer
- Ramona Optics IncDurhamUnited States
- Biomedical Engineering, Duke UniversityDurhamUnited States
| | - Eva A Naumann
- Department of Neurobiology, Duke School of MedicineDurhamUnited States
| |
Collapse
|
34
|
An integrated imaging sensor for aberration-corrected 3D photography. Nature 2022; 612:62-71. [PMID: 36261533 DOI: 10.1038/s41586-022-05306-8] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Accepted: 09/01/2022] [Indexed: 11/08/2022]
Abstract
Planar digital image sensors facilitate broad applications in a wide range of areas1-5, and the number of pixels has scaled up rapidly in recent years2,6. However, the practical performance of imaging systems is fundamentally limited by spatially nonuniform optical aberrations originating from imperfect lenses or environmental disturbances7,8. Here we propose an integrated scanning light-field imaging sensor, termed a meta-imaging sensor, to achieve high-speed aberration-corrected three-dimensional photography for universal applications without additional hardware modifications. Instead of directly detecting a two-dimensional intensity projection, the meta-imaging sensor captures extra-fine four-dimensional light-field distributions through a vibrating coded microlens array, enabling flexible and precise synthesis of complex-field-modulated images in post-processing. Using the sensor, we achieve high-performance photography up to a gigapixel with a single spherical lens without a data prior, leading to orders-of-magnitude reductions in system capacity and costs for optical imaging. Even in the presence of dynamic atmosphere turbulence, the meta-imaging sensor enables multisite aberration correction across 1,000 arcseconds on an 80-centimetre ground-based telescope without reducing the acquisition speed, paving the way for high-resolution synoptic sky surveys. Moreover, high-density accurate depth maps can be retrieved simultaneously, facilitating diverse applications from autonomous driving to industrial inspections.
Collapse
|
35
|
Bottura B, Rooney LM, Hoskisson PA, McConnell G. Intra-colony channel morphology in Escherichia coli biofilms is governed by nutrient availability and substrate stiffness. Biofilm 2022; 4:100084. [PMID: 36254115 PMCID: PMC9568850 DOI: 10.1016/j.bioflm.2022.100084] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Revised: 09/16/2022] [Accepted: 09/18/2022] [Indexed: 02/02/2023] Open
Abstract
Nutrient-transporting channels have been recently discovered in mature Escherichia coli biofilms, however the relationship between intra-colony channel structure and the surrounding environmental conditions is poorly understood. Using a combination of fluorescence mesoscopy and a purpose-designed open-source quantitative image analysis pipeline, we show that growth substrate composition and nutrient availability have a profound effect on the morphology of intra-colony channels in mature E. coli biofilms. Under all nutrient conditions, intra-colony channel width was observed to increase non-linearly with radial distance from the centre of the biofilm. Notably, the channels were around 25% wider at the centre of carbon-limited biofilms compared to nitrogen-limited biofilms. Channel density also differed in colonies grown on rich and minimal media, with the former creating a network of tightly packed channels and the latter leading to well-separated, wider channels with defined edges. Our approach paves the way for measurement of internal patterns in a wide range of biofilms, offering the potential for new insights into infection and pathogenicity.
Collapse
Affiliation(s)
- Beatrice Bottura
- Department of Physics, SUPA, University of Strathclyde, G4 0NG, Glasgow, UK,Corresponding author.
| | - Liam M. Rooney
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, G4 0RE, Glasgow, UK
| | - Paul A. Hoskisson
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, G4 0RE, Glasgow, UK
| | - Gail McConnell
- Department of Physics, SUPA, University of Strathclyde, G4 0NG, Glasgow, UK
| |
Collapse
|
36
|
Battistella E, Schniete J, Wesencraft K, Quintana JF, McConnell G. Light-sheet mesoscopy with the Mesolens provides fast sub-cellular resolution imaging throughout large tissue volumes. iScience 2022; 25:104797. [PMID: 36034214 PMCID: PMC9404659 DOI: 10.1016/j.isci.2022.104797] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Revised: 05/27/2022] [Accepted: 07/14/2022] [Indexed: 11/19/2022] Open
Abstract
Rapid imaging of large biological tissue specimens such as ultrathick sections of mouse brain cannot easily be performed with a standard microscope. Optical mesoscopy offers a solution, but thus far imaging has been too slow to be useful for routine use. We have developed two different illuminators for light-sheet mesoscopy with the Mesolens and we demonstrate their use in high-speed optical mesoscale imaging of large tissue specimens. The first light-sheet approach uses Gaussian optics and is straightforward to implement. It provides excellent lateral resolution and high-speed imaging, but the axial resolution is poor. The second light-sheet is a more complex Airy light-sheet that provides sub-cellular resolution in three dimensions that is comparable in quality to point-scanning confocal mesoscopy, but the light-sheet method of illuminating the specimen reduces the imaging time by a factor of 14. This creates new possibilities for high-content, higher-throughput optical bioimaging at the mesoscale.
Collapse
Affiliation(s)
- Eliana Battistella
- Department of Physics, SUPA, University of Strathclyde, 107 Rottenrow East, Glasgow G4 0NG, UK
| | - Jan Schniete
- Department of Physics, SUPA, University of Strathclyde, 107 Rottenrow East, Glasgow G4 0NG, UK
| | - Katrina Wesencraft
- Department of Physics, SUPA, University of Strathclyde, 107 Rottenrow East, Glasgow G4 0NG, UK
| | - Juan F. Quintana
- Wellcome Centre for Integrative Parasitology, Institute of Biodiversity Animal Health and Comparative Medicine, University of Glasgow, Glasgow, UK
| | - Gail McConnell
- Department of Physics, SUPA, University of Strathclyde, 107 Rottenrow East, Glasgow G4 0NG, UK
| |
Collapse
|
37
|
Jiang S, Guo C, Song P, Wang T, Wang R, Zhang T, Wu Q, Pandey R, Zheng G. High-throughput digital pathology via a handheld, multiplexed, and AI-powered ptychographic whole slide scanner. LAB ON A CHIP 2022; 22:2657-2670. [PMID: 35583207 DOI: 10.1039/d2lc00084a] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
The recent advent of whole slide imaging (WSI) systems has moved digital pathology closer to diagnostic applications and clinical practices. Integrating WSI with machine learning promises the growth of this field in upcoming years. Here we report the design and implementation of a handheld, colour-multiplexed, and AI-powered ptychographic whole slide scanner for digital pathology applications. This handheld scanner is built using low-cost and off-the-shelf components, including red, green, and blue laser diodes for sample illumination, a modified stage for programmable sample positioning, and a synchronized image sensor pair for data acquisition. We smear a monolayer of goat blood cells on the main sensor for high-resolution lensless coded ptychographic imaging. The synchronized secondary sensor acts as a non-contact encoder for precisely tracking the absolute object position for ptychographic reconstruction. For WSI, we introduce a new phase-contrast-based focus metric for post-acquisition autofocusing of both stained and unstained specimens. We show that the scanner can resolve the 388-nm linewidth on the resolution target and acquire gigapixel images with a 14 mm × 11 mm area in ∼70 seconds. The imaging performance is validated with regular stained pathology slides, unstained thyroid smears, and malaria-infected blood smears. The deep neural network developed in this study further enables high-throughput cytometric analysis using the recovered complex amplitude. The reported do-it-yourself scanner offers a portable solution to transform the high-end WSI system into one that can be made widely available at a low cost. The capability of high-throughput quantitative phase imaging may also find applications in rapid on-site evaluations.
Collapse
Affiliation(s)
- Shaowei Jiang
- Department of Biomedical Engineering, University of Connecticut, Storrs, CT, 06269, USA.
| | - Chengfei Guo
- Department of Biomedical Engineering, University of Connecticut, Storrs, CT, 06269, USA.
| | - Pengming Song
- Department of Biomedical Engineering, University of Connecticut, Storrs, CT, 06269, USA.
| | - Tianbo Wang
- Department of Biomedical Engineering, University of Connecticut, Storrs, CT, 06269, USA.
| | - Ruihai Wang
- Department of Biomedical Engineering, University of Connecticut, Storrs, CT, 06269, USA.
| | - Terrance Zhang
- Department of Biomedical Engineering, University of Connecticut, Storrs, CT, 06269, USA.
| | - Qian Wu
- Pathology and Laboratory Medicine, University of Connecticut Health Centre, Farmington, CT, 06030, USA
| | - Rishikesh Pandey
- Department of Biomedical Engineering, University of Connecticut, Storrs, CT, 06269, USA.
| | - Guoan Zheng
- Department of Biomedical Engineering, University of Connecticut, Storrs, CT, 06269, USA.
| |
Collapse
|
38
|
Dyer L, Parker A, Paphiti K, Sanderson J. Lightsheet Microscopy. Curr Protoc 2022; 2:e448. [PMID: 35838628 DOI: 10.1002/cpz1.448] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
In this paper, we review lightsheet (selective plane illumination) microscopy for mouse developmental biologists. There are different means of forming the illumination sheet, and we discuss these. We explain how we introduced the lightsheet microscope economically into our core facility and present our results on fixed and living samples. We also describe methods of clearing fixed samples for three-dimensional imaging and discuss the various means of preparing samples with particular reference to mouse cilia, adipose spheroids, and cochleae. © 2022 The Authors. Current Protocols published by Wiley Periodicals LLC.
Collapse
Affiliation(s)
- Laura Dyer
- MRC Harwell Institute, Mammalian Genetics Unit, Harwell Campus, Oxfordshire, UK
| | - Andrew Parker
- MRC Harwell Institute, Mammalian Genetics Unit, Harwell Campus, Oxfordshire, UK
| | - Keanu Paphiti
- MRC Harwell Institute, Mammalian Genetics Unit, Harwell Campus, Oxfordshire, UK
| | - Jeremy Sanderson
- MRC Harwell Institute, Mammalian Genetics Unit, Harwell Campus, Oxfordshire, UK
| |
Collapse
|
39
|
Battistella E, Quintana JF, McConnell G. Application of Light-Sheet Mesoscopy to Image Host-Pathogen Interactions in Intact Organs. Front Cell Infect Microbiol 2022; 12:903957. [PMID: 35774409 PMCID: PMC9237429 DOI: 10.3389/fcimb.2022.903957] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Accepted: 05/17/2022] [Indexed: 11/17/2022] Open
Abstract
Human African Trypanosomiasis (HAT) is a disease caused by the extracellular parasite Trypanosoma brucei that affects the central nervous system (CNS) during the chronic stage of the infection, inducing neuroinflammation, coma, and death if left untreated. However, little is known about the structural change happening in the brain as result of the infection. So far, infection-induced neuroinflammation has been observed with conventional methods, such as immunohistochemistry, electron microscopy, and 2-photon microscopy only in small portions of the brain, which may not be representative of the disease. In this paper, we have used a newly-developed light-sheet illuminator to image the level of neuroinflammation in chronically infected mice and compared it to naïve controls. This system was developed for imaging in combination with the Mesolens objective lens, providing fast sub-cellular resolution for tens of mm3-large imaging volumes. The mouse brain specimens were cleared using CUBIC+, followed by antibody staining to locate Glial Fibrillary Acid Protein (GFAP) expressing cells, primarily astrocytes and ependymocytes, used here as a proxy for cell reactivity and gliosis. The large capture volume allowed us to detect GFAP+ cells and spatially resolve the response to T. brucei infection. Based on morphometric analyses and spatial distribution of GFAP+ cells, our data demonstrates a significant increase in cell dendrite branching around the lateral ventricle, as well as dorsal and ventral third ventricles, that are negatively correlated with the branch extension in distal sites from the circumventricular spaces. To our knowledge, this is the first report highlighting the potential of light-sheet mesoscopy to characterise the inflammatory responses of the mouse brain to parasitic infection at the cellular level in intact cleared organs, opening new avenues for the development of new mesoscale imaging techniques for the study of host-pathogen interactions.
Collapse
Affiliation(s)
- Eliana Battistella
- Department of Physics, SUPA, University of Strathclyde, Glasgow, United Kingdom
| | - Juan F. Quintana
- Wellcome Centre for Integrative Parasitology (WCIP) University of Glasgow, Glasgow, United Kingdom
- Institute of Biodiversity, Animal Health, and Comparative Medicine (IBAHCM). University of Glasgow, Glasgow, United Kingdom
| | - Gail McConnell
- Department of Physics, SUPA, University of Strathclyde, Glasgow, United Kingdom
| |
Collapse
|
40
|
Munck S, Cawthorne C, Escamilla‐Ayala A, Kerstens A, Gabarre S, Wesencraft K, Battistella E, Craig R, Reynaud EG, Swoger J, McConnell G. Challenges and advances in optical 3D mesoscale imaging. J Microsc 2022; 286:201-219. [PMID: 35460574 PMCID: PMC9325079 DOI: 10.1111/jmi.13109] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Revised: 03/02/2022] [Accepted: 04/14/2022] [Indexed: 12/14/2022]
Abstract
Optical mesoscale imaging is a rapidly developing field that allows the visualisation of larger samples than is possible with standard light microscopy, and fills a gap between cell and organism resolution. It spans from advanced fluorescence imaging of micrometric cell clusters to centimetre-size complete organisms. However, with larger volume specimens, new problems arise. Imaging deeper into tissues at high resolution poses challenges ranging from optical distortions to shadowing from opaque structures. This manuscript discusses the latest developments in mesoscale imaging and highlights limitations, namely labelling, clearing, absorption, scattering, and also sample handling. We then focus on approaches that seek to turn mesoscale imaging into a more quantitative technique, analogous to quantitative tomography in medical imaging, highlighting a future role for digital and physical phantoms as well as artificial intelligence.
Collapse
Affiliation(s)
- Sebastian Munck
- VIB‐KU Leuven Center for Brain & Disease ResearchLight Microscopy Expertise Unit & VIB BioImaging CoreLeuvenBelgium
- KU Leuven Department of NeurosciencesLeuven Brain InstituteLeuvenBelgium
| | | | - Abril Escamilla‐Ayala
- VIB‐KU Leuven Center for Brain & Disease ResearchLight Microscopy Expertise Unit & VIB BioImaging CoreLeuvenBelgium
- KU Leuven Department of NeurosciencesLeuven Brain InstituteLeuvenBelgium
| | - Axelle Kerstens
- VIB‐KU Leuven Center for Brain & Disease ResearchLight Microscopy Expertise Unit & VIB BioImaging CoreLeuvenBelgium
- KU Leuven Department of NeurosciencesLeuven Brain InstituteLeuvenBelgium
| | - Sergio Gabarre
- VIB‐KU Leuven Center for Brain & Disease ResearchLight Microscopy Expertise Unit & VIB BioImaging CoreLeuvenBelgium
- KU Leuven Department of NeurosciencesLeuven Brain InstituteLeuvenBelgium
| | | | | | - Rebecca Craig
- Department of Physics, SUPAUniversity of StrathclydeGlasgowUK
| | - Emmanuel G. Reynaud
- School of Biomolecular and Biomedical ScienceUniversity College DublinDublinBelfieldIreland
| | - Jim Swoger
- European Molecular Biology Laboratory (EMBL) BarcelonaBarcelonaSpain
| | - Gail McConnell
- Department of Physics, SUPAUniversity of StrathclydeGlasgowUK
| |
Collapse
|
41
|
Jiang S, Guo C, Wang T, Liu J, Song P, Zhang T, Wang R, Feng B, Zheng G. Blood-Coated Sensor for High-Throughput Ptychographic Cytometry on a Blu-ray Disc. ACS Sens 2022; 7:1058-1067. [PMID: 35393855 DOI: 10.1021/acssensors.1c02704] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
The Blu-ray drive is an engineering masterpiece that integrates disc rotation, pickup head translation, and three lasers in a compact and portable format. Here, we integrate a blood-coated image sensor with a modified Blu-ray drive for high-throughput cytometric analysis of various biospecimens. In this device, samples are mounted on the rotating Blu-ray disc and illuminated by the built-in lasers from the pickup head. The resulting coherent diffraction patterns are then recorded by the blood-coated image sensor. The rich spatial features of the blood-cell monolayer help down-modulate the object information for sensor detection, thus forming a high-resolution computational biolens with a theoretically unlimited field of view. With the acquired data, we develop a lensless coherent diffraction imaging modality termed rotational ptychography for image reconstruction. We show that our device can resolve the 435 nm line width on the resolution target and has a field of view only limited by the size of the Blu-ray disc. To demonstrate its applications, we perform high-throughput urinalysis by locating disease-related calcium oxalate crystals over the entire microscope slide. We also quantify different types of cells on a blood smear with an acquisition speed of ∼10,000 cells per second. For in vitro experiments, we monitor live bacterial cultures over the entire Petri dish with single-cell resolution. Using biological cells as a computational lens could enable new intriguing imaging devices for point-of-care diagnostics. Modifying a Blu-ray drive with the blood-coated sensor further allows the spread of high-throughput optical microscopy from well-equipped laboratories to citizen scientists worldwide.
Collapse
Affiliation(s)
- Shaowei Jiang
- Department of Biomedical Engineering, University of Connecticut, Storrs, Connecticut 06269, United States
| | - Chengfei Guo
- Department of Biomedical Engineering, University of Connecticut, Storrs, Connecticut 06269, United States
| | - Tianbo Wang
- Department of Biomedical Engineering, University of Connecticut, Storrs, Connecticut 06269, United States
| | - Jia Liu
- Department of Biomedical Engineering, University of Connecticut, Storrs, Connecticut 06269, United States
| | - Pengming Song
- Department of Biomedical Engineering, University of Connecticut, Storrs, Connecticut 06269, United States
| | - Terrance Zhang
- Department of Biomedical Engineering, University of Connecticut, Storrs, Connecticut 06269, United States
| | - Ruihai Wang
- Department of Biomedical Engineering, University of Connecticut, Storrs, Connecticut 06269, United States
| | - Bin Feng
- Department of Biomedical Engineering, University of Connecticut, Storrs, Connecticut 06269, United States
| | - Guoan Zheng
- Department of Biomedical Engineering, University of Connecticut, Storrs, Connecticut 06269, United States
| |
Collapse
|
42
|
Neary-Zajiczek L, Essmann C, Rau A, Bano S, Clancy N, Jansen M, Heptinstall L, Miranda E, Gander A, Pawar V, Fernandez-Reyes D, Shaw M, Davidson B, Stoyanov D. Stain-free identification of tissue pathology using a generative adversarial network to infer nanomechanical signatures. NANOSCALE ADVANCES 2021; 3:6403-6414. [PMID: 34913024 PMCID: PMC8577366 DOI: 10.1039/d1na00527h] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Accepted: 09/01/2021] [Indexed: 06/14/2023]
Abstract
Intraoperative frozen section analysis can be used to improve the accuracy of tumour margin estimation during cancer resection surgery through rapid processing and pathological assessment of excised tissue. Its applicability is limited in some cases due to the additional risks associated with prolonged surgery, largely from the time-consuming staining procedure. Our work uses a measurable property of bulk tissue to bypass the staining process: as tumour cells proliferate, they influence the surrounding extra-cellular matrix, and the resulting change in elastic modulus provides a signature of the underlying pathology. In this work we accurately localise atomic force microscopy measurements of human liver tissue samples and train a generative adversarial network to infer elastic modulus from low-resolution images of unstained tissue sections. Pathology is predicted through unsupervised clustering of parameters characterizing the distributions of inferred values, achieving 89% accuracy for all samples based on the nominal assessment (n = 28), and 95% for samples that have been validated by two independent pathologists through post hoc staining (n = 20). Our results demonstrate that this technique could increase the feasibility of intraoperative frozen section analysis for use during resection surgery and improve patient outcomes.
Collapse
Affiliation(s)
- Lydia Neary-Zajiczek
- Wellcome/EPSRC Centre for Surgical and Interventional Sciences London W1W 7TS UK
- Department of Computer Science, University College London London WC1E 6BT UK
| | - Clara Essmann
- Department of Computer Science, University College London London WC1E 6BT UK
| | - Anita Rau
- Wellcome/EPSRC Centre for Surgical and Interventional Sciences London W1W 7TS UK
| | - Sophia Bano
- Wellcome/EPSRC Centre for Surgical and Interventional Sciences London W1W 7TS UK
| | - Neil Clancy
- Wellcome/EPSRC Centre for Surgical and Interventional Sciences London W1W 7TS UK
- Department of Medical Physics and Biomedical Engineering, University College London London WC1E 6BT UK
| | - Marnix Jansen
- Department of Pathology, UCL Cancer Institute, University College London London WC1E 6BT UK
| | | | - Elena Miranda
- Biobank and Pathology Translational Technology Platform, UCL Cancer Institute, University College London London WC1E 6BT UK
| | - Amir Gander
- Department of Surgical Biotechnology, University College London London WC1E 6BT UK
| | - Vijay Pawar
- Department of Computer Science, University College London London WC1E 6BT UK
| | | | - Michael Shaw
- Wellcome/EPSRC Centre for Surgical and Interventional Sciences London W1W 7TS UK
- Department of Computer Science, University College London London WC1E 6BT UK
- National Physical Laboratory Teddington TW11 0LW UK
| | - Brian Davidson
- Wellcome/EPSRC Centre for Surgical and Interventional Sciences London W1W 7TS UK
| | - Danail Stoyanov
- Wellcome/EPSRC Centre for Surgical and Interventional Sciences London W1W 7TS UK
| |
Collapse
|
43
|
Ichimura T, Kakizuka T, Horikawa K, Seiriki K, Kasai A, Hashimoto H, Fujita K, Watanabe TM, Nagai T. Exploring rare cellular activity in more than one million cells by a transscale scope. Sci Rep 2021; 11:16539. [PMID: 34400683 PMCID: PMC8368064 DOI: 10.1038/s41598-021-95930-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Accepted: 08/03/2021] [Indexed: 02/07/2023] Open
Abstract
In many phenomena of biological systems, not a majority, but a minority of cells act on the entire multicellular system causing drastic changes in the system properties. To understand the mechanisms underlying such phenomena, it is essential to observe the spatiotemporal dynamics of a huge population of cells at sub-cellular resolution, which is difficult with conventional tools such as microscopy and flow cytometry. Here, we describe an imaging system named AMATERAS that enables optical imaging with an over-one-centimeter field-of-view and a-few-micrometer spatial resolution. This trans-scale-scope has a simple configuration, composed of a low-power lens for machine vision and a hundred-megapixel image sensor. We demonstrated its high cell-throughput, capable of simultaneously observing more than one million cells. We applied it to dynamic imaging of calcium ions in HeLa cells and cyclic-adenosine-monophosphate in Dictyostelium discoideum, and successfully detected less than 0.01% of rare cells and observed multicellular events induced by these cells.
Collapse
Affiliation(s)
- T Ichimura
- Transdimensional Life Imaging Division, Institute for Open and Transdisciplinary Research Initiatives, Osaka University, Yamadaoka 2-1, Suita, Osaka, 565-0871, Japan.
- PRESTO, Japan Science and Technology Agency, Tokyo, 113-0033, Japan.
| | - T Kakizuka
- Transdimensional Life Imaging Division, Institute for Open and Transdisciplinary Research Initiatives, Osaka University, Yamadaoka 2-1, Suita, Osaka, 565-0871, Japan
| | - K Horikawa
- Department of Optical Imaging, Advanced Research Promotion Center, Tokushima University, Kuramoto-cho 3-18-15, Tokushima, Tokushima, 770-8503, Japan
| | - K Seiriki
- Laboratory of Molecular Neuropharmacology, Graduate School of Pharmaceutical Sciences, Osaka University, Yamadaoka 1-6, Suita, Osaka, 565-0871, Japan
| | - A Kasai
- Laboratory of Molecular Neuropharmacology, Graduate School of Pharmaceutical Sciences, Osaka University, Yamadaoka 1-6, Suita, Osaka, 565-0871, Japan
| | - H Hashimoto
- Transdimensional Life Imaging Division, Institute for Open and Transdisciplinary Research Initiatives, Osaka University, Yamadaoka 2-1, Suita, Osaka, 565-0871, Japan
- Laboratory of Molecular Neuropharmacology, Graduate School of Pharmaceutical Sciences, Osaka University, Yamadaoka 1-6, Suita, Osaka, 565-0871, Japan
- Institute for Transdisciplinary Graduate Degree Programs, Osaka University, Yamadaoka 1-1, Suita, Osaka, 565-0871, Japan
| | - K Fujita
- Transdimensional Life Imaging Division, Institute for Open and Transdisciplinary Research Initiatives, Osaka University, Yamadaoka 2-1, Suita, Osaka, 565-0871, Japan
- Department of Applied Physics, Graduate School of Engineering, Osaka University, Yamadaoka 2-1, Suita, Osaka, 565-0871, Japan
| | - T M Watanabe
- Laboratory for Comprehensive Bioimaging, RIKEN Center for Biosystems Dynamics Research (BDR), Minatomachi-minami 2-2-3, Chuo-ku, Kobe, Hyogo, 650-0047, Japan
- Department of Stem Cell Biology, Research Institute for Radiation Biology and Medicine, Hiroshima University, Kasumi 1-2-3, Minami-ku, Hiroshima, 734-8553, Japan
| | - T Nagai
- Transdimensional Life Imaging Division, Institute for Open and Transdisciplinary Research Initiatives, Osaka University, Yamadaoka 2-1, Suita, Osaka, 565-0871, Japan.
- SANKEN (The Institute of Scientific and Industrial Research), Osaka University, Mihogaoka 8-1, Ibaraki, Osaka, 567-0047, Japan.
| |
Collapse
|
44
|
Park J, Brady DJ, Zheng G, Tian L, Gao L. Review of bio-optical imaging systems with a high space-bandwidth product. ADVANCED PHOTONICS 2021; 3:044001. [PMID: 35178513 PMCID: PMC8849623 DOI: 10.1117/1.ap.3.4.044001] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/14/2023]
Abstract
Optical imaging has served as a primary method to collect information about biosystems across scales-from functionalities of tissues to morphological structures of cells and even at biomolecular levels. However, to adequately characterize a complex biosystem, an imaging system with a number of resolvable points, referred to as a space-bandwidth product (SBP), in excess of one billion is typically needed. Since a gigapixel-scale far exceeds the capacity of current optical imagers, compromises must be made to obtain either a low spatial resolution or a narrow field-of-view (FOV). The problem originates from constituent refractive optics-the larger the aperture, the more challenging the correction of lens aberrations. Therefore, it is impractical for a conventional optical imaging system to achieve an SBP over hundreds of millions. To address this unmet need, a variety of high-SBP imagers have emerged over the past decade, enabling an unprecedented resolution and FOV beyond the limit of conventional optics. We provide a comprehensive survey of high-SBP imaging techniques, exploring their underlying principles and applications in bioimaging.
Collapse
Affiliation(s)
- Jongchan Park
- University of California, Department of Bioengineering, Los Angeles, California, United States
| | - David J. Brady
- University of Arizona, James C. Wyant College of Optical Sciences, Tucson, Arizona, United States
| | - Guoan Zheng
- University of Connecticut, Department of Biomedical Engineering, Storrs, Connecticut, United States
- University of Connecticut, Department of Electrical and Computer Engineering, Storrs, Connecticut, United States
| | - Lei Tian
- Boston University, Department of Electrical and Computer Engineering, Boston, Massachusetts, United States
| | - Liang Gao
- University of California, Department of Bioengineering, Los Angeles, California, United States
| |
Collapse
|
45
|
Shaw M, Claveau R, Manescu P, Elmi M, Brown BJ, Scrimgeour R, Kölln LS, McConnell G, Fernandez-Reyes D. Optical mesoscopy, machine learning, and computational microscopy enable high information content diagnostic imaging of blood films. J Pathol 2021; 255:62-71. [PMID: 34096621 DOI: 10.1002/path.5738] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2021] [Revised: 05/19/2021] [Accepted: 06/03/2021] [Indexed: 12/11/2022]
Abstract
Automated image-based assessment of blood films has tremendous potential to support clinical haematology within overstretched healthcare systems. To achieve this, efficient and reliable digital capture of the rich diagnostic information contained within a blood film is a critical first step. However, this is often challenging, and in many cases entirely unfeasible, with the microscopes typically used in haematology due to the fundamental trade-off between magnification and spatial resolution. To address this, we investigated three state-of-the-art approaches to microscopic imaging of blood films which leverage recent advances in optical and computational imaging and analysis to increase the information capture capacity of the optical microscope: optical mesoscopy, which uses a giant microscope objective (Mesolens) to enable high-resolution imaging at low magnification; Fourier ptychographic microscopy, a computational imaging method which relies on oblique illumination with a series of LEDs to capture high-resolution information; and deep neural networks which can be trained to increase the quality of low magnification, low resolution images. We compare and contrast the performance of these techniques for blood film imaging for the exemplar case of Giemsa-stained peripheral blood smears. Using computational image analysis and shape-based object classification, we demonstrate their use for automated analysis of red blood cell morphology and visualization and detection of small blood-borne parasites such as the malarial parasite Plasmodium falciparum. Our results demonstrate that these new methods greatly increase the information capturing capacity of the light microscope, with transformative potential for haematology and more generally across digital pathology. © 2021 The Authors. The Journal of Pathology published by John Wiley & Sons, Ltd. on behalf of The Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Michael Shaw
- Department of Computer Science, Faculty of Engineering Sciences, University College London, London, UK.,Biometrology Group, National Physical Laboratory, Teddington, UK
| | - Rémy Claveau
- Department of Computer Science, Faculty of Engineering Sciences, University College London, London, UK
| | - Petru Manescu
- Department of Computer Science, Faculty of Engineering Sciences, University College London, London, UK
| | - Muna Elmi
- Department of Computer Science, Faculty of Engineering Sciences, University College London, London, UK
| | - Biobele J Brown
- Department of Paediatrics, College of Medicine of University of Ibadan, University College Hospital, Ibadan, Nigeria
| | - Ross Scrimgeour
- Department of Physics, SUPA, University of Strathclyde, Glasgow, UK
| | - Lisa S Kölln
- Department of Physics, SUPA, University of Strathclyde, Glasgow, UK
| | - Gail McConnell
- Department of Physics, SUPA, University of Strathclyde, Glasgow, UK
| | - Delmiro Fernandez-Reyes
- Department of Computer Science, Faculty of Engineering Sciences, University College London, London, UK.,Department of Paediatrics, College of Medicine of University of Ibadan, University College Hospital, Ibadan, Nigeria
| |
Collapse
|
46
|
Munck S, Swoger J, Coll-Lladó M, Gritti N, Vande Velde G. Maximizing content across scales: Moving multimodal microscopy and mesoscopy toward molecular imaging. Curr Opin Chem Biol 2021; 63:188-199. [PMID: 34198170 DOI: 10.1016/j.cbpa.2021.05.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 05/06/2021] [Accepted: 05/16/2021] [Indexed: 10/21/2022]
Abstract
Molecular imaging aims to depict the molecules in living patients. However, because this aim is still far beyond reach, patchworks of different solutions need to be used to tackle this overarching goal. From the vast toolbox of imaging techniques, we focus on those recent advances in optical microscopy that image molecules and cells at the submicron to centimeter scale. Mesoscopic imaging covers the "imaging gap" between techniques such as confocal microscopy and magnetic resonance imagingthat image entire live samples but with limited resolution. Microscopy focuses on the cellular level; mesoscopy visualizes the organization of molecules and cells into tissues and organs. The correlation between these techniques allows us to combine disciplines ranging from whole body imaging to basic research of model systems. We review current developments focused on improving microscopic and mesoscopic imaging technologies and on hardware and software that push the current sensitivity and resolution boundaries.
Collapse
Affiliation(s)
- Sebastian Munck
- VIB-KU Leuven Center for Brain & Disease Research, Light Microscopy Expertise Unit & VIB BioImaging Core, O&N4 Herestraat 49 box 602, Leuven, 3000, Belgium; KU Leuven Department of Neurosciences, Leuven Brain Institute, O&N4 Herestraat 49 box 602, Leuven, 3000, Belgium
| | - Jim Swoger
- European Molecular Biology Laboratory (EMBL) Barcelona, Barcelona, 08003, Spain
| | | | - Nicola Gritti
- European Molecular Biology Laboratory (EMBL) Barcelona, Barcelona, 08003, Spain
| | - Greetje Vande Velde
- Department of Imaging and Pathology, Faculty of Medicine, KU Leuven, Leuven, Belgium.
| |
Collapse
|
47
|
Dmitriev RI, Intes X, Barroso MM. Luminescence lifetime imaging of three-dimensional biological objects. J Cell Sci 2021; 134:1-17. [PMID: 33961054 PMCID: PMC8126452 DOI: 10.1242/jcs.254763] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
A major focus of current biological studies is to fill the knowledge gaps between cell, tissue and organism scales. To this end, a wide array of contemporary optical analytical tools enable multiparameter quantitative imaging of live and fixed cells, three-dimensional (3D) systems, tissues, organs and organisms in the context of their complex spatiotemporal biological and molecular features. In particular, the modalities of luminescence lifetime imaging, comprising fluorescence lifetime imaging (FLI) and phosphorescence lifetime imaging microscopy (PLIM), in synergy with Förster resonance energy transfer (FRET) assays, provide a wealth of information. On the application side, the luminescence lifetime of endogenous molecules inside cells and tissues, overexpressed fluorescent protein fusion biosensor constructs or probes delivered externally provide molecular insights at multiple scales into protein-protein interaction networks, cellular metabolism, dynamics of molecular oxygen and hypoxia, physiologically important ions, and other physical and physiological parameters. Luminescence lifetime imaging offers a unique window into the physiological and structural environment of cells and tissues, enabling a new level of functional and molecular analysis in addition to providing 3D spatially resolved and longitudinal measurements that can range from microscopic to macroscopic scale. We provide an overview of luminescence lifetime imaging and summarize key biological applications from cells and tissues to organisms.
Collapse
Affiliation(s)
- Ruslan I. Dmitriev
- Tissue Engineering and Biomaterials Group, Department of
Human Structure and Repair, Faculty of Medicine and Health Sciences,
Ghent University, Ghent 9000,
Belgium
| | - Xavier Intes
- Department of Biomedical Engineering, Center for
Modeling, Simulation and Imaging for Medicine (CeMSIM),
Rensselaer Polytechnic Institute, Troy, NY
12180-3590, USA
| | - Margarida M. Barroso
- Department of Molecular and Cellular
Physiology, Albany Medical College,
Albany, NY 12208, USA
| |
Collapse
|
48
|
Fast, cell-resolution, contiguous-wide two-photon imaging to reveal functional network architectures across multi-modal cortical areas. Neuron 2021; 109:1810-1824.e9. [PMID: 33878295 DOI: 10.1016/j.neuron.2021.03.032] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Revised: 02/11/2021] [Accepted: 03/25/2021] [Indexed: 02/07/2023]
Abstract
Fast and wide field-of-view imaging with single-cell resolution, high signal-to-noise ratio, and no optical aberrations have the potential to inspire new avenues of investigations in biology. However, such imaging is challenging because of the inevitable tradeoffs among these parameters. Here, we overcome these tradeoffs by combining a resonant scanning system, a large objective with low magnification and high numerical aperture, and highly sensitive large-aperture photodetectors. The result is a practically aberration-free, fast-scanning high optical invariant two-photon microscopy (FASHIO-2PM) that enables calcium imaging from a large network composed of ∼16,000 neurons at 7.5 Hz from a 9 mm2 contiguous image plane, including more than 10 sensory-motor and higher-order areas of the cerebral cortex in awake mice. Network analysis based on single-cell activities revealed that the brain exhibits small-world rather than scale-free behavior. The FASHIO-2PM is expected to enable studies on biological dynamics by simultaneously monitoring macroscopic activities and their compositional elements.
Collapse
|
49
|
Iyer S, Mukherjee S, Kumar M. Watching the embryo: Evolution of the microscope for the study of embryogenesis. Bioessays 2021; 43:e2000238. [PMID: 33837551 DOI: 10.1002/bies.202000238] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Revised: 03/09/2021] [Accepted: 03/10/2021] [Indexed: 11/08/2022]
Abstract
Embryos and microscopes share a long, remarkable history and biologists have always been intrigued to watch how embryos develop under the microscope. Here we discuss the advances in microscopy which have greatly influenced our current understanding of embryogenesis. We highlight the evolution of microscopes and the optical technologies that have been instrumental in studying various developmental processes. These imaging modalities provide mechanistic insights into the dynamic cellular and molecular events which drive lineage commitment and morphogenetic changes in the developing embryo. We begin the journey with a brief history of microscopy to study embryos. First, we review the principles and optics of light, fluorescence, confocal, and electron microscopy which have been key techniques for imaging cellular and molecular events during embryonic development. Next, we discuss recent key imaging modalities such as light-sheet microscopy, which are suitable for whole embryo imaging. Further, we highlight imaging techniques like multiphoton and super resolution microscopy for beyond light diffraction limit, high resolution imaging. Lastly, we review some of the scattering-based imaging methods and techniques used for imaging human embryos.
Collapse
Affiliation(s)
- Sharada Iyer
- Academy of Scientific and Innovative Research (AcCSIR), CSIR-CCMB campus, Uppal road, Hyderabad, 500007, India.,CSIR - Centre for Cellular and Molecular Biology, Hyderabad, India
| | | | - Megha Kumar
- CSIR - Centre for Cellular and Molecular Biology, Hyderabad, India
| |
Collapse
|
50
|
Zhuang C, Cao J, Zhang R, Xiao G, Hu J, Xie H, Dai Q. Real-time brain-wide multi-planar microscopy for simultaneous cortex and hippocampus imaging at the cellular resolution in mice. BIOMEDICAL OPTICS EXPRESS 2021; 12:1858-1868. [PMID: 33996203 PMCID: PMC8086472 DOI: 10.1364/boe.418229] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 02/19/2021] [Accepted: 02/25/2021] [Indexed: 06/12/2023]
Abstract
Interactions between the cerebral cortex and the deep cerebellar nuclei play important roles in cognitive processes. However, conventional microscopes fail to dynamically record cellular structures in distinct brain regions and at different depths, which requires high resolution, large field of view (FOV), and depth of field (DOF). Here we propose a single-photon excited fluorescence microscopy technique that performs simultaneous cortex and hippocampus imaging, enabled by a customized microscope and a chronic optical window. After we implant a glass microwindow above the hippocampus, the surface of the hippocampus is shifted to the superficial plane. We demonstrate that the proposed technique is able to image cellular structures and blood vessel dynamics in the cortex and the hippocampus in in vivo experiments, and is compatible with various mesoscopic systems.
Collapse
Affiliation(s)
- Chaowei Zhuang
- Department of Automation, Tsinghua University, Beijing 100084, China
- These authors contributed equally to this work
| | - Jiangbei Cao
- Department of Anesthesiology, the First Medical Center, Chinese PLA General Hospital, Beijing 100853, China
- These authors contributed equally to this work
| | - Rujin Zhang
- Department of Anesthesiology, the First Medical Center, Chinese PLA General Hospital, Beijing 100853, China
- Medical Company, CPLA Unit No. 32139, Beijing 101200, China
- These authors contributed equally to this work
| | - Guihua Xiao
- Department of Automation, Tsinghua University, Beijing 100084, China
- Institute of Brain and Cognitive Science, Tsinghua University, Beijing 100084, China
| | - Jing Hu
- State Key Laboratory of Modern Optical Instrumentation, Zhejiang University, Hangzhou 310027, China
| | - Hao Xie
- Department of Automation, Tsinghua University, Beijing 100084, China
- Institute of Brain and Cognitive Science, Tsinghua University, Beijing 100084, China
| | - Qionghai Dai
- Department of Automation, Tsinghua University, Beijing 100084, China
- Institute of Brain and Cognitive Science, Tsinghua University, Beijing 100084, China
- IDG/McGovern Institute for Brain Research, Tsinghua University, Beijing 100084, China
| |
Collapse
|