1
|
Franceschini A, Jin M, Chen CW, Silvestri L, Mastrodonato A, Denny CA. Brain-wide immunolabeling and tissue clearing applications for engram research. Neurobiol Learn Mem 2025; 218:108032. [PMID: 39922482 DOI: 10.1016/j.nlm.2025.108032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Revised: 01/28/2025] [Accepted: 02/05/2025] [Indexed: 02/10/2025]
Abstract
In recent years, there has been significant progress in memory research, driven by genetic and imaging technological advances that have given unprecedented access to individual memory traces or engrams. Although Karl Lashley argued since the 1930s that an engram is not confined to a particular area but rather distributed across the entire brain, most current studies have focused exclusively on a single or few brain regions. However, this compartmentalized approach overlooks the interactions between multiple brain regions, limiting our understanding of engram mechanisms. More recently, several studies have begun to investigate engrams across the brain, but research is still limited by a lack of standardized techniques capable of reconstructing multiple ensembles at single-cell resolution across the entire brain. In this review, we guide researchers through the latest technological advancements and discoveries in immediate early gene (IEG) techniques, tissue clearing methods, microscope modalities, and automated large-scale analysis. These innovations could propel the field forward in building brain-wide engram maps of normal and disease states, thus, providing unprecedented new insights. Ultimately, this review aims to bridge the gap between research focused on single brain regions and the need for a comprehensive understanding of whole-brain engrams, revealing new approaches for exploring the neuronal mechanisms underlying engrams.
Collapse
Affiliation(s)
- Alessandra Franceschini
- Department of Psychiatry, Columbia University Irving Medical Center (CUIMC), New York, NY 10032, USA; European Laboratory for Non-linear Spectroscopy (LENS), Sesto Fiorentino, 50019 Italy
| | - Michelle Jin
- Medical Scientist Training Program (MSTP), Columbia University Irving Medical Center (CUIMC), New York, NY 10032, USA; Neurobiology and Behavior (NB&B) Graduate Program, Columbia University, New York, NY 10027, USA
| | - Claire W Chen
- Cellular, Molecular, and Biomedical Sciences Graduate Program, Columbia University, New York, NY 10027, USA
| | - Ludovico Silvestri
- European Laboratory for Non-linear Spectroscopy (LENS), Sesto Fiorentino, 50019 Italy; Department of Physics and Astronomy, University of Florence, Sesto Fiorentino 50019, Italy
| | - Alessia Mastrodonato
- Department of Psychiatry, Columbia University Irving Medical Center (CUIMC), New York, NY 10032, USA; Division of Systems Neuroscience, Area Neuroscience, Research Foundation for Mental Hygiene, Inc. (RFMH) / New York State Psychiatric Institute (NYSPI), New York, NY 10032, USA.
| | - Christine Ann Denny
- Department of Psychiatry, Columbia University Irving Medical Center (CUIMC), New York, NY 10032, USA; Division of Systems Neuroscience, Area Neuroscience, Research Foundation for Mental Hygiene, Inc. (RFMH) / New York State Psychiatric Institute (NYSPI), New York, NY 10032, USA.
| |
Collapse
|
2
|
Kitzerow O, Christensen S, Hong J, Adam RJ, Zucker IH, Jensen‐Smith H, Wang H. Anatomical mapping of neural lineages expressing the transient receptor potential vanilloid type 1 receptor using a modified and combined PACT and CUBIC protocol for rapid tissue clearance. Acta Physiol (Oxf) 2025; 241:e14275. [PMID: 39821962 PMCID: PMC11737471 DOI: 10.1111/apha.14275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2024] [Revised: 11/13/2024] [Accepted: 01/01/2025] [Indexed: 01/19/2025]
Abstract
AIM Tissue clearance is a rapidly evolving technology that allows for the three-dimensional imaging of intact biological tissues. Preexisting tissue-clearing techniques, such as Passive Clarity Technique (PACT) and Clear Unobstructed Brain Imaging Cocktails and Computational Analysis (CUBIC), clear tissues adequately but have distinct disadvantages, such as taking extensive time to clear tissues and degradation of endogenous tissue fluorescence. We developed a new tissue-clearing technique combining PACT and CUBIC protocols to map the neural lineages expressing the transient receptor potential vanilloid type 1 (TRPV1) receptor. METHODS To test the effectiveness of this modified protocol, a TdTomato reporter mouse line was crossed with a separate mouse line containing Cre recombinase under the control of the TRPV1 promoter, which would result in TRPV1 cell lineages expressing green fluorescence protein (GFP). RESULTS Compared to the PACT protocol that requires several weeks to months for tissue clearance, our approach reached a satisfactory clearance within 3 days in all neural tissues as well as several non-neural tissues such as colon, duodenum, and pancreas. Compared to the CUBIC approach, all tissues reserved strong GFP fluorescence. Robust GFP fluorescence was visualized in sensory neuronal soma but not in sympathetic ganglia neuronal soma. On the other hand, GFP fluorescence in the TRPV1 cells appeared to be expressed throughout the epithelium of the duodenum and colon and the arteriole smooth muscle in all non-neuronal tissues. CONCLUSION This study shows that our combined PACT and CUBIC (CPC) protocol can clear tissues in significantly less time while preserving tissue integrity and fluorescence.
Collapse
Affiliation(s)
- Oliver Kitzerow
- Department of Genetics Cell Biology and AnatomyUniversity of Nebraska Medical CenterOmahaNebraskaUSA
- Deptrtment of AnesthesiologyUniversity of Nebraska Medical CenterOmahaNebraskaUSA
- Department of Medical EducationCreighton UniversityOmahaNebraskaUSA
| | - Samuel Christensen
- Deptrtment of AnesthesiologyUniversity of Nebraska Medical CenterOmahaNebraskaUSA
| | - Juan Hong
- Deptrtment of AnesthesiologyUniversity of Nebraska Medical CenterOmahaNebraskaUSA
| | - Ryan J. Adam
- Deptrtment of AnesthesiologyUniversity of Nebraska Medical CenterOmahaNebraskaUSA
| | - Irving H. Zucker
- Department of Cellular and Integrative PhysiologyUniversity of Nebraska Medical CenterOmahaNebraskaUSA
| | - Heather Jensen‐Smith
- Department of Genetics Cell Biology and AnatomyUniversity of Nebraska Medical CenterOmahaNebraskaUSA
- Fred and Pamela Buffett Cancer CenterUniversity of Nebraska Medical CenterOmahaNebraskaUSA
| | - Han‐Jun Wang
- Deptrtment of AnesthesiologyUniversity of Nebraska Medical CenterOmahaNebraskaUSA
| |
Collapse
|
3
|
Asahina Y, Aihara M, Miyai T, Tanaka A, Onodera H. Visualization of porcine and human aqueous humor outflow tract anatomies with transparency enhancement. Jpn J Ophthalmol 2025:10.1007/s10384-024-01151-6. [PMID: 39826072 DOI: 10.1007/s10384-024-01151-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Accepted: 11/11/2024] [Indexed: 01/20/2025]
Abstract
PURPOSE There is no established method for visualizing the three-dimensional (3D) structure of the aqueous humor outflow tract. This study attempted to visualize the 3D structures of porcine and human ocular tissues, particularly the aqueous humor outflow tract using a transparency reagent composed of 2, 2-thiodiethanol. STUDY DESIGN Clinical and experimental. METHODS The porcine eyes were collected in Japan, and the human eyes were imported from the United States. The human eyes were obtained from a 64-year-old Caucasian woman, arriving 7 days after her death. The specimens were formalin-fixed upon arrival, fluorescently labeled, optically cleared using a transparency-enhancing reagent, and visualized using a confocal microscope. RESULTS Both porcine and human eyes were visualized to the extent that the choroidal vessels were observed on gross examination. The aqueous humor outflow tract was clearly observed as a luminal structure in the porcine eye, mainly depicted by autofluorescence, and in the human eyes as a luminal structure continuing from the trabecular meshwork without fluorescence. CONCLUSION Observations using transparency-enhancing technology enabled us to obtain 3D images useful for visualizing ocular tissues, especially the aqueous humor outflow tract.
Collapse
Affiliation(s)
- Yuichi Asahina
- Department of Ophthalmology, Graduate School of Medicine, The University of Tokyo, 7-3-1, Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan.
| | - Makoto Aihara
- Department of Ophthalmology, Graduate School of Medicine, The University of Tokyo, 7-3-1, Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Takashi Miyai
- Department of Ophthalmology, Graduate School of Medicine, The University of Tokyo, 7-3-1, Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Asami Tanaka
- Photon Science Center, The University of Tokyo, 7-3-1, Hongo, Bunkyo-ku, Tokyo, 113-8656, Japan
| | - Hiroshi Onodera
- Institute for Photon Science and Technology, The University of Tokyo, 7-3-1, Hongo, Bunkyo-ku, Tokyo, 113-8656, Japan
| |
Collapse
|
4
|
König A, Cavanagh BL, Amado I, Kalra A, Ogon BA, Hinton PV, Kennedy OD. A novel workflow for multi-modal imaging of musculoskeletal tissues. J Anat 2025. [PMID: 39823263 DOI: 10.1111/joa.14202] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Revised: 12/02/2024] [Accepted: 12/04/2024] [Indexed: 01/19/2025] Open
Abstract
According to the World Health Organization (WHO) musculoskeletal conditions are a leading contributor to disability worldwide. This fact is often somewhat overlooked, since musculoskeletal conditions are less likely to be associated with mortality. Nonetheless, treatments, therapies and management of these conditions are extremely costly to national healthcare systems. As with all systemic conditions, biomedical imaging of relevant tissues plays a major role in understanding the fundamental biological processes involved in musculoskeletal health. However, the skeletal system with its relatively large proportion of dense, opaque (often mineralised) tissues can often be more challenging to image, and recently important advances have been made in imaging these complex musculoskeletal tissues. Thus, we here describe a novel workflow in which recent advanced imaging techniques have been modified and optimised for use in musculoskeletal tissues (specifically bone and cartilage). This will allow for investigations, of different phases of these tissues, at new and higher resolutions. Furthermore, the process has been designed to fit with the existing and standard processes which are typically used with these samples (i.e. μCT imaging and standard histology). The additional modalities which have been included here are second harmonic generation (SHG) imaging, tissue clearing, specifically the Passive Clear Lipid-exchanged Acrylamide-hybridised Rigid Imaging Tissue hYdrogel (CLARITY) method known as PACT, and then imaging of these tissues with confocal, multiphoton and light-sheet microscopy. This paper serves to introduce a combination of existing new methods and improvements in imaging of musculoskeletal tissues.
Collapse
Affiliation(s)
- Anya König
- Department of Anatomy and Regenerative Medicine, Tissue Engineering Research Group (TERG) Royal College of Surgeons Ireland (RCSI) University of Medicine and Health Sciences, Dublin, Ireland
| | - Brenton L Cavanagh
- Cellular and Molecular Imaging Core, Royal College of Surgeons Ireland (RCSI) University of Medicine and Health Sciences, Dublin, Ireland
| | - Isabel Amado
- Department of Anatomy and Regenerative Medicine, Tissue Engineering Research Group (TERG) Royal College of Surgeons Ireland (RCSI) University of Medicine and Health Sciences, Dublin, Ireland
| | - Amit Kalra
- Department of Anatomy and Regenerative Medicine, Tissue Engineering Research Group (TERG) Royal College of Surgeons Ireland (RCSI) University of Medicine and Health Sciences, Dublin, Ireland
| | - Bohnejie A Ogon
- Department of Anatomy and Regenerative Medicine, Tissue Engineering Research Group (TERG) Royal College of Surgeons Ireland (RCSI) University of Medicine and Health Sciences, Dublin, Ireland
| | - Paige V Hinton
- Department of Anatomy and Regenerative Medicine, Tissue Engineering Research Group (TERG) Royal College of Surgeons Ireland (RCSI) University of Medicine and Health Sciences, Dublin, Ireland
| | - Oran D Kennedy
- Department of Anatomy and Regenerative Medicine, Tissue Engineering Research Group (TERG) Royal College of Surgeons Ireland (RCSI) University of Medicine and Health Sciences, Dublin, Ireland
- Trinity Center for Biomedical Engineering, Trinity College Dublin, Dublin, Ireland
- Advanced Materials and BioEngineering Research (AMBER) Centre, Dublin, Ireland
| |
Collapse
|
5
|
Ni Y, Wu J, Liu F, Yi Y, Meng X, Gao X, Xiao L, Zhou W, Chen Z, Chu P, Xing D, Yuan Y, Ding D, Shen G, Yang M, Wu R, Wang L, Melo LMN, Lin S, Cheng X, Li G, Tasdogan A, Ubellacker JM, Zhao H, Fang S, Shen B. Deep imaging of LepR + stromal cells in optically cleared murine bone hemisections. Bone Res 2025; 13:6. [PMID: 39800733 PMCID: PMC11725602 DOI: 10.1038/s41413-024-00387-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 09/30/2024] [Accepted: 10/22/2024] [Indexed: 01/16/2025] Open
Abstract
Tissue clearing combined with high-resolution confocal imaging is a cutting-edge approach for dissecting the three-dimensional (3D) architecture of tissues and deciphering cellular spatial interactions under physiological and pathological conditions. Deciphering the spatial interaction of leptin receptor-expressing (LepR+) stromal cells with other compartments in the bone marrow is crucial for a deeper understanding of the stem cell niche and the skeletal tissue. In this study, we introduce an optimized protocol for the 3D analysis of skeletal tissues, enabling the visualization of hematopoietic and stromal cells, especially LepR+ stromal cells, within optically cleared bone hemisections. Our method preserves the 3D tissue architecture and is extendable to other hematopoietic sites such as calvaria and vertebrae. The protocol entails tissue fixation, decalcification, and cryosectioning to reveal the marrow cavity. Completed within approximately 12 days, this process yields highly transparent tissues that maintain genetically encoded or antibody-stained fluorescent signals. The bone hemisections are compatible with diverse antibody labeling strategies. Confocal microscopy of these transparent samples allows for qualitative and quantitative image analysis using Aivia or Bitplane Imaris software, assessing a spectrum of parameters. With proper storage, the fluorescent signal in the stained and cleared bone hemisections remains intact for at least 2-3 months. This protocol is robust, straightforward to implement, and highly reproducible, offering a valuable tool for tissue architecture and cellular interaction studies.
Collapse
Affiliation(s)
- Yuehan Ni
- College of Life Sciences, Beijing Normal University, 100875, Beijing, China
- National Institute of Biological Sciences, Beijing (NIBS), 102206, Beijing, China
| | - Jiamiao Wu
- National Institute of Biological Sciences, Beijing (NIBS), 102206, Beijing, China
- Peking University-Tsinghua University-National Institute of Biological Sciences Joint Graduate Program, School of Life Sciences, Tsinghua University, 100084, Beijing, China
| | - Fengqi Liu
- School of Biopharmacy, China Pharmaceutical University, 211198, Nanjing, China
| | - Yating Yi
- Chinese Institute for Brain Research, Beijing (CIBR), 102206, Beijing, China
| | - Xiangjiao Meng
- National Institute of Biological Sciences, Beijing (NIBS), 102206, Beijing, China
- Chinese Academy of Medical Sciences & Peking Union Medical College, 100730, Beijing, China
| | - Xiang Gao
- National Institute of Biological Sciences, Beijing (NIBS), 102206, Beijing, China
- Peking University-Tsinghua University-National Institute of Biological Sciences Joint Graduate Program, Academy for Advanced Interdisciplinary Studies, Peking University, 100871, Beijing, China
| | - Luyi Xiao
- National Institute of Biological Sciences, Beijing (NIBS), 102206, Beijing, China
| | - Weiwei Zhou
- National Institute of Biological Sciences, Beijing (NIBS), 102206, Beijing, China
- Peking University-Tsinghua University-National Institute of Biological Sciences Joint Graduate Program, School of Life Sciences, Tsinghua University, 100084, Beijing, China
| | - Zexi Chen
- Chinese Institute for Brain Research, Beijing (CIBR), 102206, Beijing, China
| | - Peng Chu
- National Institute of Biological Sciences, Beijing (NIBS), 102206, Beijing, China
- Peking University-Tsinghua University-National Institute of Biological Sciences Joint Graduate Program, School of Life Sciences, Tsinghua University, 100084, Beijing, China
| | - Dan Xing
- Arthritis Clinic and Research Center, Peking University People's Hospital, Peking University, 100044, Beijing, China
| | - Ye Yuan
- Arthritis Clinic and Research Center, Peking University People's Hospital, Peking University, 100044, Beijing, China
| | - Donghui Ding
- School of Biopharmacy, China Pharmaceutical University, 211198, Nanjing, China
| | - Ge Shen
- National Institute of Biological Sciences, Beijing (NIBS), 102206, Beijing, China
- Peking University-Tsinghua University-National Institute of Biological Sciences Joint Graduate Program, Academy for Advanced Interdisciplinary Studies, Peking University, 100871, Beijing, China
| | - Min Yang
- College of Life Sciences, Beijing Normal University, 100875, Beijing, China
- National Institute of Biological Sciences, Beijing (NIBS), 102206, Beijing, China
| | - Ronjie Wu
- Musculoskeletal Research Laboratory, Department of Orthopaedics & Traumatology & Li Ka Shing Institute of Health Sciences, Faculty of Medicine, The Chinese University of Hong Kong, 999077, Shatin, Hong Kong SAR, PR China
| | - Ling Wang
- Department of Radiology, Beijing Jishuitan Hospital, Capital Medical University, National Center for Orthopaedics, 100035, Beijing, China
| | - Luiza Martins Nascentes Melo
- Department of Dermatology, University Hospital Essen & German Cancer Consortium, Partner Site, Essen, 45147, Germany
| | - Sien Lin
- Musculoskeletal Research Laboratory, Department of Orthopaedics & Traumatology & Li Ka Shing Institute of Health Sciences, Faculty of Medicine, The Chinese University of Hong Kong, 999077, Shatin, Hong Kong SAR, PR China
| | - Xiaoguang Cheng
- Department of Radiology, Beijing Jishuitan Hospital, Capital Medical University, National Center for Orthopaedics, 100035, Beijing, China
| | - Gang Li
- Musculoskeletal Research Laboratory, Department of Orthopaedics & Traumatology & Li Ka Shing Institute of Health Sciences, Faculty of Medicine, The Chinese University of Hong Kong, 999077, Shatin, Hong Kong SAR, PR China
| | - Alpaslan Tasdogan
- Department of Dermatology, University Hospital Essen & German Cancer Consortium, Partner Site, Essen, 45147, Germany
| | - Jessalyn M Ubellacker
- Department of Molecular Metabolism, Harvard T.H. Chan School of Public Health, Boston, MA, 02115, USA
| | - Hu Zhao
- Chinese Institute for Brain Research, Beijing (CIBR), 102206, Beijing, China.
| | - Shentong Fang
- School of Biopharmacy, China Pharmaceutical University, 211198, Nanjing, China.
| | - Bo Shen
- National Institute of Biological Sciences, Beijing (NIBS), 102206, Beijing, China.
- Tsinghua Institute of Multidisciplinary Biomedical Research, Tsinghua University, 100084, Beijing, China.
| |
Collapse
|
6
|
Unal S, Mi R, Musicki B, Hoke A, Burnett AL. Mapping of functional erectogenic nerves on the rat prostate. J Sex Med 2025; 22:217-224. [PMID: 39657061 DOI: 10.1093/jsxmed/qdae174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2024] [Revised: 10/10/2024] [Accepted: 11/19/2024] [Indexed: 12/17/2024]
Abstract
BACKGROUND Preservation of erectogenic nerves during radical prostatectomy (RP) is hampered by limited understanding of their precise localization, due to their complex, intertwined paths, and the inherent individual variations across patients. Because erection utilizes a subset of cavernous nerves (CNs) that in response to sexual stimuli reveal phosphorylation of neuronal nitric oxide synthase (nNOS) on its stimulatory site Ser-1412, we hypothesized that delineation of nerves containing phosphorylated (P)-nNOS on Ser-1412 would establish the location of functional erectogenic nerves within the periprostatic region. AIM To identify the distribution and quantity of functional erection-relevant ([P-nNOS]-containing) nerves in the periprostatic area and discriminate them among the CNs distribution. We further evaluated whether functional communication exists between contralateral CNs. METHODS Young adult male Sprague-Dawley rats underwent electrical stimulation of the CNs to induce penile erection via phosphorylation of nNOS on Ser-1412 (6 V for 2 min, n = 6). No stimulation group served as control (n = 6). The prostate and adjacent structures were collected and processed for whole-mount double-staining with TuJ1 antibody (a pan-axonal marker) and P-nNOS (n = 3 for stimulation, n = 3 for no stimulation), or total nNOS and P-nNOS (n = 3 for stimulation, n = 3 for no stimulation), followed by modified optical clearing and microscopic examination. Nerve quantification was done by systematic counting. OUTCOMES Location and quantification of functional erectogenic nerves. RESULTS In the male rat, we obtained a map of P-nNOS-containing nerves in the periprostatic area, which are relevant for penile erection. Only 17.5% of all nerves, and only 28.4% of the total nNOS-containing nerves in the periprostatic region are functionally erectogenic nerves. Furthermore, there is no functional innervation between contralateral (stimulated and non-stimulated) CNs. CLINICAL IMPLICATIONS This basic science study is expected to provide a foundation for subsequent studies at the human level. Understanding the erection-relevant nerve distribution in the periprostatic area is expected to advance nerve-sparing RP at the human level to improve sexual function outcomes. STRENGTHS AND LIMITATIONS This is the first study to describe and quantitate a subset of functional erection-relevant (P-nNOS-containing) nerves in the periprostatic area. Our study differs from previous studies where nerves containing total nNOS were localized without specifying which nerves produce erection. However, because this study comprised a relatively small number of rats, further studies with a bigger sample size or other model animals are warranted. CONCLUSION Only a subset of nerve fibers in the periprostatic region represent functional erectogenic nerves, characterized by the expression of P-nNOS (Ser-1412).
Collapse
Affiliation(s)
- Selman Unal
- The James Buchanan Brady Urological Institute and Department of Urology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, United States
- Department of Urology, Ankara Yildirim Beyazit University School of Medicine, Ankara, 06800, Turkey
| | - Ruifa Mi
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, United States
| | - Biljana Musicki
- The James Buchanan Brady Urological Institute and Department of Urology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, United States
| | - Ahmet Hoke
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, United States
| | - Arthur L Burnett
- The James Buchanan Brady Urological Institute and Department of Urology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, United States
| |
Collapse
|
7
|
Zhang J, Qiao W, Jin R, Li H, Gong H, Chen SC, Luo Q, Yuan J. Optical sectioning methods in three-dimensional bioimaging. LIGHT, SCIENCE & APPLICATIONS 2025; 14:11. [PMID: 39741128 DOI: 10.1038/s41377-024-01677-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/11/2024] [Revised: 09/24/2024] [Accepted: 10/28/2024] [Indexed: 01/02/2025]
Abstract
In recent advancements in life sciences, optical microscopy has played a crucial role in acquiring high-quality three-dimensional structural and functional information. However, the quality of 3D images is often compromised due to the intense scattering effect in biological tissues, compounded by several issues such as limited spatiotemporal resolution, low signal-to-noise ratio, inadequate depth of penetration, and high phototoxicity. Although various optical sectioning techniques have been developed to address these challenges, each method adheres to distinct imaging principles for specific applications. As a result, the effective selection of suitable optical sectioning techniques across diverse imaging scenarios has become crucial yet challenging. This paper comprehensively overviews existing optical sectioning techniques and selection guidance under different imaging scenarios. Specifically, we categorize the microscope design based on the spatial relationship between the illumination and detection axis, i.e., on-axis and off-axis. This classification provides a unique perspective to compare the implementation and performances of various optical sectioning approaches. Lastly, we integrate selected optical sectioning methods on a custom-built off-axis imaging system and present a unique perspective for the future development of optical sectioning techniques.
Collapse
Affiliation(s)
- Jing Zhang
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology, Wuhan, China
- MoE Key Laboratory for Biomedical Photonics, Innovation Institute, Huazhong University of Science and Technology, Wuhan, China
| | - Wei Qiao
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology, Wuhan, China
- MoE Key Laboratory for Biomedical Photonics, Innovation Institute, Huazhong University of Science and Technology, Wuhan, China
| | - Rui Jin
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology, Wuhan, China
- MoE Key Laboratory for Biomedical Photonics, Innovation Institute, Huazhong University of Science and Technology, Wuhan, China
| | - Hongjin Li
- Hong Kong Center for Cerebro-Cardiovascular Health Engineering, N.T, Hong Kong, China
- Department of Biomedical Engineering, City University of Hong Kong, Hong Kong, China
| | - Hui Gong
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology, Wuhan, China
- MoE Key Laboratory for Biomedical Photonics, Innovation Institute, Huazhong University of Science and Technology, Wuhan, China
- HUST-Suzhou Institute for Brainsmatics, Suzhou, China
| | - Shih-Chi Chen
- Hong Kong Center for Cerebro-Cardiovascular Health Engineering, N.T, Hong Kong, China.
- Department of Mechanical and Automation Engineering, The Chinese University of Hong Kong, Shatin, N.T., Hong Kong, China.
| | - Qingming Luo
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology, Wuhan, China.
- MoE Key Laboratory for Biomedical Photonics, Innovation Institute, Huazhong University of Science and Technology, Wuhan, China.
- HUST-Suzhou Institute for Brainsmatics, Suzhou, China.
- School of Biomedical Engineering, Hainan University, Haikou, China.
| | - Jing Yuan
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology, Wuhan, China.
- MoE Key Laboratory for Biomedical Photonics, Innovation Institute, Huazhong University of Science and Technology, Wuhan, China.
- HUST-Suzhou Institute for Brainsmatics, Suzhou, China.
| |
Collapse
|
8
|
Delpech C, Schaeffer J, Vilallongue N, Delaunay A, Benadjal A, Blot B, Excoffier B, Plissonnier E, Gascon E, Albert F, Paccard A, Saintpierre A, Gasnier C, Zagar Y, Castellani V, Belin S, Chédotal A, Nawabi H. Axon guidance during mouse central nervous system regeneration is required for specific brain innervation. Dev Cell 2024; 59:3213-3228.e8. [PMID: 39353435 DOI: 10.1016/j.devcel.2024.09.005] [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: 10/10/2023] [Revised: 07/11/2024] [Accepted: 09/05/2024] [Indexed: 10/04/2024]
Abstract
Reconstructing functional neuronal circuits is one major challenge of central nervous system repair. Through activation of pro-growth signaling pathways, some neurons achieve long-distance axon regrowth. Yet, functional reconnection has hardly been obtained, as these regenerating axons fail to resume their initial trajectory and reinnervate their proper target. Axon guidance is considered to be active only during development. Here, using the mouse visual system, we show that axon guidance is still active in the adult brain in regenerative conditions. We highlight that regenerating retinal ganglion cell axons avoid one of their primary targets, the suprachiasmatic nucleus (SCN), due to Slit/Robo repulsive signaling. Together with promoting regeneration, silencing Slit/Robo in vivo enables regenerating axons to enter the SCN and form active synapses. The newly formed circuit is associated with neuronal activation and functional recovery. Our results provide evidence that axon guidance mechanisms are required to reconnect regenerating axons to specific brain nuclei.
Collapse
Affiliation(s)
- Céline Delpech
- Université Grenoble Alpes, Inserm U1216, Grenoble Institut Neurosciences, 38000 Grenoble, France
| | - Julia Schaeffer
- Université Grenoble Alpes, Inserm U1216, Grenoble Institut Neurosciences, 38000 Grenoble, France
| | - Noemie Vilallongue
- Université Grenoble Alpes, Inserm U1216, Grenoble Institut Neurosciences, 38000 Grenoble, France
| | - Apolline Delaunay
- Université Grenoble Alpes, Inserm U1216, Grenoble Institut Neurosciences, 38000 Grenoble, France
| | - Amin Benadjal
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, Paris, France
| | - Beatrice Blot
- Université Grenoble Alpes, Inserm U1216, Grenoble Institut Neurosciences, 38000 Grenoble, France
| | - Blandine Excoffier
- Université Grenoble Alpes, Inserm U1216, Grenoble Institut Neurosciences, 38000 Grenoble, France
| | - Elise Plissonnier
- Université Grenoble Alpes, Inserm U1216, Grenoble Institut Neurosciences, 38000 Grenoble, France
| | - Eduardo Gascon
- Aix Marseille University, CNRS, INT, Institute of Neurosci Timone, Marseille, France
| | - Floriane Albert
- Université Grenoble Alpes, Inserm U1216, Grenoble Institut Neurosciences, 38000 Grenoble, France
| | - Antoine Paccard
- Université Grenoble Alpes, Inserm U1216, Grenoble Institut Neurosciences, 38000 Grenoble, France
| | - Ana Saintpierre
- Université Grenoble Alpes, Inserm U1216, Grenoble Institut Neurosciences, 38000 Grenoble, France
| | - Celestin Gasnier
- Université Grenoble Alpes, Inserm U1216, Grenoble Institut Neurosciences, 38000 Grenoble, France
| | - Yvrick Zagar
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, Paris, France
| | - Valérie Castellani
- University Claude Bernard Lyon 1, MeLiS, CNRS UMR5284, INSERM U1314, Lyon, France
| | - Stephane Belin
- Université Grenoble Alpes, Inserm U1216, Grenoble Institut Neurosciences, 38000 Grenoble, France
| | - Alain Chédotal
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, Paris, France; University Claude Bernard Lyon 1, MeLiS, CNRS UMR5284, INSERM U1314, Lyon, France; Institut de pathologie, groupe hospitalier Est, Hospices Civils de Lyon, Lyon, France
| | - Homaira Nawabi
- Université Grenoble Alpes, Inserm U1216, Grenoble Institut Neurosciences, 38000 Grenoble, France.
| |
Collapse
|
9
|
Yu T, Zhong X, Li D, Zhu J, Tuchin VV, Zhu D. Delivery and kinetics of immersion optical clearing agents in tissues: Optical imaging from ex vivo to in vivo. Adv Drug Deliv Rev 2024; 215:115470. [PMID: 39481483 DOI: 10.1016/j.addr.2024.115470] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Revised: 09/30/2024] [Accepted: 10/27/2024] [Indexed: 11/02/2024]
Abstract
Advanced optical imaging provides a powerful tool for the structural and functional analysis of tissues with high resolution and contrast, but the imaging performance decreases as light propagates deeper into the tissue. Tissue optical clearing technique demonstrates an innovative way to realize deep-tissue imaging and have emerged substantially in the last two decades. Here, we briefly reviewed the basic principles of tissue optical clearing techniques in the view of delivery strategies via either free diffusion or external forces-driven advection, and the commonly-used optical techniques for monitoring kinetics of clearing agents in tissue, as well as their ex vivo to in vivo applications in multiple biomedical research fields. With future efforts on the even distribution of both clearing agents and probes, excavation of more effective clearing agents, and automation of tissue clearing processes, tissue optical clearing should provide more insights into the fundamental questions in biological events clinical diagnostics.
Collapse
Affiliation(s)
- Tingting Yu
- Britton Chance Center for Biomedical Photonics-MoE Key Laboratory for Biomedical Photonics, Advanced Biomedical Imaging Facility-Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology, Wuhan 430074, Hubei, China
| | - Xiang Zhong
- Britton Chance Center for Biomedical Photonics-MoE Key Laboratory for Biomedical Photonics, Advanced Biomedical Imaging Facility-Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology, Wuhan 430074, Hubei, China
| | - Dongyu Li
- Britton Chance Center for Biomedical Photonics-MoE Key Laboratory for Biomedical Photonics, Advanced Biomedical Imaging Facility-Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology, Wuhan 430074, Hubei, China; School of Optical and Electronic Information, Huazhong University of Science and Technology, Wuhan 430074, Hubei, China
| | - Jingtan Zhu
- Britton Chance Center for Biomedical Photonics-MoE Key Laboratory for Biomedical Photonics, Advanced Biomedical Imaging Facility-Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology, Wuhan 430074, Hubei, China
| | - Valery V Tuchin
- Institute of Physics and Science Medical Center, Saratov State University, Saratov 410012, Russia; Laboratory of Laser Molecular Imaging and Machine Learning, Tomsk State University, Tomsk 634050, Russia; Institute of Precision Mechanics and Control, FRS "Saratov Scientific Centre of the RAS", Saratov 410028, Russia
| | - Dan Zhu
- Britton Chance Center for Biomedical Photonics-MoE Key Laboratory for Biomedical Photonics, Advanced Biomedical Imaging Facility-Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology, Wuhan 430074, Hubei, China.
| |
Collapse
|
10
|
Han T, Wu J, Sheng P, Li Y, Tao Z, Qu L. Deep coupled registration and segmentation of multimodal whole-brain images. Bioinformatics 2024; 40:btae606. [PMID: 39400311 PMCID: PMC11543610 DOI: 10.1093/bioinformatics/btae606] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Revised: 08/07/2024] [Accepted: 10/11/2024] [Indexed: 10/15/2024] Open
Abstract
MOTIVATION Recent brain mapping efforts are producing large-scale whole-brain images using different imaging modalities. Accurate alignment and delineation of anatomical structures in these images are essential for numerous studies. These requirements are typically modeled as two distinct tasks: registration and segmentation. However, prevailing methods, fail to fully explore and utilize the inherent correlation and complementarity between the two tasks. Furthermore, variations in brain anatomy, brightness, and texture pose another formidable challenge in designing multi-modal similarity metrics. A high-throughput approach capable of overcoming the bottleneck of multi-modal similarity metric design, while effective leveraging the highly correlated and complementary nature of two tasks is highly desirable. RESULTS We introduce a deep learning framework for joint registration and segmentation of multi-modal brain images. Under this framework, registration and segmentation tasks are deeply coupled and collaborated at two hierarchical layers. In the inner layer, we establish a strong feature-level coupling between the two tasks by learning a unified common latent feature representation. In the outer layer, we introduce a mutually supervised dual-branch network to decouple latent features and facilitate task-level collaboration between registration and segmentation. Since the latent features we designed are also modality-independent, the bottleneck of designing multi-modal similarity metric is essentially addressed. Another merit offered by this framework is the interpretability of latent features, which allows intuitive manipulation of feature learning, thereby further enhancing network training efficiency and the performance of both tasks. Extensive experiments conducted on both multi-modal and mono-modal datasets of mouse and human brains demonstrate the superiority of our method. AVAILABILITY AND IMPLEMENTATION The code is available at https://github.com/tingtingup/DCRS.
Collapse
Affiliation(s)
- Tingting Han
- Ministry of Education Key Laboratory of Intelligent Computation and Signal Processing, Information Materials and Intelligent Sensing Laboratory of Anhui Province, School of Electronics and Information Engineering, Anhui University, Hefei, Anhui, 230601, China
| | - Jun Wu
- Ministry of Education Key Laboratory of Intelligent Computation and Signal Processing, Information Materials and Intelligent Sensing Laboratory of Anhui Province, School of Electronics and Information Engineering, Anhui University, Hefei, Anhui, 230601, China
| | - Pengpeng Sheng
- Ministry of Education Key Laboratory of Intelligent Computation and Signal Processing, Information Materials and Intelligent Sensing Laboratory of Anhui Province, School of Electronics and Information Engineering, Anhui University, Hefei, Anhui, 230601, China
| | - Yuanyuan Li
- Ministry of Education Key Laboratory of Intelligent Computation and Signal Processing, Information Materials and Intelligent Sensing Laboratory of Anhui Province, School of Electronics and Information Engineering, Anhui University, Hefei, Anhui, 230601, China
| | - ZaiYang Tao
- Ministry of Education Key Laboratory of Intelligent Computation and Signal Processing, Information Materials and Intelligent Sensing Laboratory of Anhui Province, School of Electronics and Information Engineering, Anhui University, Hefei, Anhui, 230601, China
| | - Lei Qu
- Ministry of Education Key Laboratory of Intelligent Computation and Signal Processing, Information Materials and Intelligent Sensing Laboratory of Anhui Province, School of Electronics and Information Engineering, Anhui University, Hefei, Anhui, 230601, China
- SEU-ALLEN Joint Center, Institute for Brain and Intelligence, Southeast University, Nanjing, Jiangsu, 210096, China
- Institute of Artiffcial Intelligence, Hefei Comprehensive National Science Center, Hefei, 231299, China
- Hefei National Laboratory, University of Science and Technology of China, Hefei, 230094, China
| |
Collapse
|
11
|
Zhou L, Shi W, Fu S, Li M, Chen J, Fang K, Li Y. High Refractive Index Imaging Buffer for Dual-Color 3D SMLM Imaging of Thick Samples. Anal Chem 2024; 96:15648-15656. [PMID: 39298273 DOI: 10.1021/acs.analchem.4c02893] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/21/2024]
Abstract
The current limitations of single-molecule localization microscopy (SMLM) in deep tissue imaging, primarily due to depth-dependent aberrations caused by refractive index (RI) mismatch, present a significant challenge in achieving high-resolution images at greater depths. To extend the imaging depth, we optimized the imaging buffer of SMLM with the RI matched to that of the objective immersion medium and systematically evaluated five different RI-matched buffers, focusing on their impact on the blinking behavior of red-absorbing dyes and the quality of reconstructed super-resolution images. Particularly, we found that clear unobstructed brain imaging cocktails-based imaging buffer could match the RI of oil and was able to clear the tissue samples. With the help of the RI-matched imaging buffers, we showed high-quality dual-color 3D SMLM images with imaging depths ranging from a few micrometers to tens of micrometers in both cultured cells and sectioned tissue samples. This advancement offers a practical and accessible method for high-resolution imaging at greater depths without the need for specialized optical equipment or expertise.
Collapse
Affiliation(s)
- Lulu Zhou
- Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen 518055, China
| | - Wei Shi
- Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen 518055, China
| | - Shuang Fu
- Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen 518055, China
| | - Mengfan Li
- Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen 518055, China
| | - Jianwei Chen
- Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen 518055, China
| | - Ke Fang
- Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen 518055, China
| | - Yiming Li
- Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen 518055, China
| |
Collapse
|
12
|
Pesce L, Ricci P, Sportelli G, Belcari N, Sancataldo G. Expansion and Light-Sheet Microscopy for Nanoscale 3D Imaging. SMALL METHODS 2024; 8:e2301715. [PMID: 38461540 DOI: 10.1002/smtd.202301715] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Revised: 02/10/2024] [Indexed: 03/12/2024]
Abstract
Expansion Microscopy (ExM) and Light-Sheet Fluorescence Microscopy (LSFM) are forefront imaging techniques that enable high-resolution visualization of biological specimens. ExM enhances nanoscale investigation using conventional fluorescence microscopes, while LSFM offers rapid, minimally invasive imaging over large volumes. This review explores the joint advancements of ExM and LSFM, focusing on the excellent performance of the integrated modality obtained from the combination of the two, which is refer to as ExLSFM. In doing so, the chemical processes required for ExM, the tailored optical setup of LSFM for examining expanded samples, and the adjustments in sample preparation for accurate data collection are emphasized. It is delve into various specimen types studied using this integrated method and assess its potential for future applications. The goal of this literature review is to enrich the comprehension of ExM and LSFM, encouraging their wider use and ongoing development, looking forward to the upcoming challenges, and anticipating innovations in these imaging techniques.
Collapse
Affiliation(s)
- Luca Pesce
- Department of Physics - Enrico Fermi, University of Pisa, Largo Pontecorvo, 3, Pisa, 56127, Italy
| | - Pietro Ricci
- Department of Applied Physics, University of Barcelona, C/Martí i Franquès, 1, Barcelona, 08028, Spain
| | - Giancarlo Sportelli
- Department of Physics - Enrico Fermi, University of Pisa, Largo Pontecorvo, 3, Pisa, 56127, Italy
| | - Nicola Belcari
- Department of Physics - Enrico Fermi, University of Pisa, Largo Pontecorvo, 3, Pisa, 56127, Italy
| | - Giuseppe Sancataldo
- Department of Physics - Emilio Segrè, University of Palermo, Viale delle Scienze, 18, Palermo, 90128, Italy
| |
Collapse
|
13
|
Li C, Li Y, Zhao H, Ding L. Enhancing brain image quality with 3D U-net for stripe removal in light sheet fluorescence microscopy. Brain Inform 2024; 11:24. [PMID: 39325110 PMCID: PMC11427638 DOI: 10.1186/s40708-024-00236-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Accepted: 08/19/2024] [Indexed: 09/27/2024] Open
Abstract
Light Sheet Fluorescence Microscopy (LSFM) is increasingly popular in neuroimaging for its ability to capture high-resolution 3D neural data. However, the presence of stripe noise significantly degrades image quality, particularly in complex 3D stripes with varying widths and brightness, posing challenges in neuroscience research. Existing stripe removal algorithms excel in suppressing noise and preserving details in 2D images with simple stripes but struggle with the complexity of 3D stripes. To address this, we propose a novel 3D U-net model for Stripe Removal in Light sheet fluorescence microscopy (USRL). This approach directly learns and removes stripes in 3D space across different scales, employing a dual-resolution strategy to effectively handle stripes of varying complexities. Additionally, we integrate a nonlinear mapping technique to normalize high dynamic range and unevenly distributed data before applying the stripe removal algorithm. We validate our method on diverse datasets, demonstrating substantial improvements in peak signal-to-noise ratio (PSNR) compared to existing algorithms. Moreover, our algorithm exhibits robust performance when applied to real LSFM data. Through extensive validation experiments, both on test sets and real-world data, our approach outperforms traditional methods, affirming its effectiveness in enhancing image quality. Furthermore, the adaptability of our algorithm extends beyond LSFM applications to encompass other imaging modalities. This versatility underscores its potential to enhance image usability across various research disciplines.
Collapse
Affiliation(s)
- Changshan Li
- Institute for Brain and Intelligence, Southeast University, Nanjing, China
| | - Youqi Li
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- Chinese Institute for Brain Research, Beijing, China
| | - Hu Zhao
- Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, China
- Chinese Institute for Brain Research, Beijing, China
| | - Liya Ding
- Institute for Brain and Intelligence, Southeast University, Nanjing, China.
| |
Collapse
|
14
|
Weiss KR, Huisken J, Khanjani N, Bakalov V, Engle ML, Krzyzanowski MC, Madden T, Maiese DR, Waterfield JR, Williams DN, Wood L, Wu X, Hamilton CM, Huggins W. T-CLEARE: a pilot community-driven tissue clearing protocol repository. Front Bioeng Biotechnol 2024; 12:1304622. [PMID: 39351064 PMCID: PMC11439823 DOI: 10.3389/fbioe.2024.1304622] [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: 02/23/2024] [Accepted: 06/18/2024] [Indexed: 10/04/2024] Open
Abstract
Selecting and implementing a tissue clearing protocol is challenging. Established more than 100 years ago, tissue clearing is still a rapidly evolving field of research. There are currently many published protocols to choose from, and each performs better or worse across a range of key evaluation factors (e.g., speed, cost, tissue stability, fluorescence quenching). Additionally, tissue clearing protocols are often optimized for specific experimental contexts, and applying an existing protocol to a new problem can require a lengthy period of adaptation by trial and error. Although the primary literature and review articles provide a useful starting point for optimization, there is growing recognition that results can vary dramatically with changes to tissue type or antibody used. To help address this issue, we have developed a novel, freely available repository of tissue clearing protocols named T-CLEARE (Tissue CLEAring protocol REpository; https://doryworkspace.org/doryviz). T-CLEARE incorporates community responses to an open survey designed to capture details not commonly found in the scientific literature, including modifications to published protocols required for specific use cases and instances when tissue clearing protocols did not perform well (negative results). The goal of T-CLEARE is to help the community share evaluations and modifications of tissue clearing protocols for various tissue types and potentially identify best-in-class methods for a given application.
Collapse
Affiliation(s)
- Kurt R. Weiss
- Morgridge Institute for Research, Madison, WI, United States
| | - Jan Huisken
- Morgridge Institute for Research, Madison, WI, United States
| | - Neda Khanjani
- Mark and Mary Stevens Neuroimaging and Informatics Institute, Laboratory of Neuro Imaging, Keck School of Medicine of University of Southern California, Los Angeles, CA, United States
| | - Vesselina Bakalov
- Bioinformatics and Computational Biology Program, RTI International, Durham, NC, United States
| | - Michelle L. Engle
- Bioinformatics and Computational Biology Program, RTI International, Durham, NC, United States
| | | | - Tom Madden
- Bioinformatics and Computational Biology Program, RTI International, Durham, NC, United States
| | - Deborah R. Maiese
- Bioinformatics and Computational Biology Program, RTI International, Durham, NC, United States
| | - Justin R. Waterfield
- Bioinformatics and Computational Biology Program, RTI International, Durham, NC, United States
| | - David N. Williams
- Bioinformatics and Computational Biology Program, RTI International, Durham, NC, United States
| | - Lauren Wood
- Bioinformatics and Computational Biology Program, RTI International, Durham, NC, United States
| | - Xin Wu
- Bioinformatics and Computational Biology Program, RTI International, Durham, NC, United States
| | - Carol M. Hamilton
- Bioinformatics and Computational Biology Program, RTI International, Durham, NC, United States
| | - Wayne Huggins
- Bioinformatics and Computational Biology Program, RTI International, Durham, NC, United States
| |
Collapse
|
15
|
Meo DD, Sorelli M, Ramazzotti J, Cheli F, Bradley S, Perego L, Lorenzon B, Mazzamuto G, Emmi A, Porzionato A, Caro RD, Garbelli R, Biancheri D, Pelorosso C, Conti V, Guerrini R, Pavone FS, Costantini I. Quantitative cytoarchitectural phenotyping of deparaffinized human brain tissues. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.10.612232. [PMID: 39314456 PMCID: PMC11419081 DOI: 10.1101/2024.09.10.612232] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 09/25/2024]
Abstract
Advanced 3D imaging techniques and image segmentation and classification methods can profoundly transform biomedical research by offering deep insights into the cytoarchitecture of the human brain in relation to pathological conditions. Here, we propose a comprehensive pipeline for performing 3D imaging and automated quantitative cellular phenotyping on Formalin-Fixed Paraffin-Embedded (FFPE) human brain specimens, a valuable yet underutilized resource. We exploited the versatility of our method by applying it to different human specimens from both adult and pediatric, normal and abnormal brain regions. Quantitative data on neuronal volume, ellipticity, local density, and spatial clustering level were obtained from a machine learning-based analysis of the 3D cytoarchitectural organization of cells identified by different molecular markers in two subjects with malformations of cortical development (MCD). This approach will grant access to a wide range of physiological and pathological paraffin-embedded clinical specimens, allowing for volumetric imaging and quantitative analysis of human brain samples at cellular resolution. Possible genotype-phenotype correlations can be unveiled, providing new insights into the pathogenesis of various brain diseases and enlarging treatment opportunities.
Collapse
Affiliation(s)
- Danila Di Meo
- European Laboratory for Non-linear Spectroscopy (LENS), University of Florence, Sesto Fiorentino, Italy
| | - Michele Sorelli
- European Laboratory for Non-linear Spectroscopy (LENS), University of Florence, Sesto Fiorentino, Italy
| | - Josephine Ramazzotti
- European Laboratory for Non-linear Spectroscopy (LENS), University of Florence, Sesto Fiorentino, Italy
| | - Franco Cheli
- European Laboratory for Non-linear Spectroscopy (LENS), University of Florence, Sesto Fiorentino, Italy
| | - Samuel Bradley
- European Laboratory for Non-linear Spectroscopy (LENS), University of Florence, Sesto Fiorentino, Italy
- Department of Physics and Astronomy, University of Florence, Sesto Fiorentino, Italy
| | - Laura Perego
- European Laboratory for Non-linear Spectroscopy (LENS), University of Florence, Sesto Fiorentino, Italy
| | - Beatrice Lorenzon
- European Laboratory for Non-linear Spectroscopy (LENS), University of Florence, Sesto Fiorentino, Italy
- Department of Physics and Astronomy, University of Florence, Sesto Fiorentino, Italy
| | - Giacomo Mazzamuto
- European Laboratory for Non-linear Spectroscopy (LENS), University of Florence, Sesto Fiorentino, Italy
- Department of Physics and Astronomy, University of Florence, Sesto Fiorentino, Italy
- National Research Council – National Institute of Optics (CNR-INO), Sesto Fiorentino, Italy
| | - Aron Emmi
- Institute of Human Anatomy, Department of Neuroscience, University of Padova, Italy
| | - Andrea Porzionato
- Institute of Human Anatomy, Department of Neuroscience, University of Padova, Italy
| | - Raffaele De Caro
- Institute of Human Anatomy, Department of Neuroscience, University of Padova, Italy
| | - Rita Garbelli
- Epilepsy Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta
| | - Dalila Biancheri
- Epilepsy Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta
| | - Cristiana Pelorosso
- Department of Neuroscience and Medical Genetics, Meyer Children’s Hospital IRCCS, Florence, Italy
| | - Valerio Conti
- Department of Neuroscience and Medical Genetics, Meyer Children’s Hospital IRCCS, Florence, Italy
| | - Renzo Guerrini
- Department of Neuroscience and Medical Genetics, Meyer Children’s Hospital IRCCS, Florence, Italy
- Department of Neuroscience, Psychology, Drug Research and Child Health, University of Florence, Florence, Italy
| | - Francesco S. Pavone
- European Laboratory for Non-linear Spectroscopy (LENS), University of Florence, Sesto Fiorentino, Italy
- Department of Physics and Astronomy, University of Florence, Sesto Fiorentino, Italy
- National Research Council – National Institute of Optics (CNR-INO), Sesto Fiorentino, Italy
| | - Irene Costantini
- European Laboratory for Non-linear Spectroscopy (LENS), University of Florence, Sesto Fiorentino, Italy
- National Research Council – National Institute of Optics (CNR-INO), Sesto Fiorentino, Italy
- Department of Biology, University of Florence, Italy
| |
Collapse
|
16
|
Talei Franzesi G, Gupta I, Hu M, Piatkveich K, Yildirim M, Zhao JP, Eom M, Han S, Park D, Andaraarachchi H, Li Z, Greenhagen J, Islam AM, Vashishtha P, Yaqoob Z, Pak N, Wissner-Gross AD, Martin-Alarcon D, Veinot J, So PT, Kortshagen U, Yoon YG, Sur M, Boyden ES. In Vivo Optical Clearing of Mammalian Brain. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.05.611421. [PMID: 39282466 PMCID: PMC11398509 DOI: 10.1101/2024.09.05.611421] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/16/2025]
Abstract
Established methods for imaging the living mammalian brain have, to date, taken optical properties of the tissue as fixed; we here demonstrate that it is possible to modify the optical properties of the brain itself to significantly enhance at-depth imaging while preserving native physiology. Using a small amount of any of several biocompatible materials to raise the refractive index of solutions superfusing the brain prior to imaging, we could increase several-fold the signals from the deepest cells normally visible and, under both one-photon and two-photon imaging, visualize cells previously too dim to see. The enhancement was observed for both anatomical and functional fluorescent reporters across a broad range of emission wavelengths. Importantly, visual tuning properties of cortical neurons in awake mice, and electrophysiological properties of neurons assessed ex vivo, were not altered by this procedure.
Collapse
|
17
|
Shih CP, Tang WC, Chen P, Chen BC. Applications of Lightsheet Fluorescence Microscopy by High Numerical Aperture Detection Lens. J Phys Chem B 2024; 128:8273-8289. [PMID: 39177503 PMCID: PMC11382282 DOI: 10.1021/acs.jpcb.4c01721] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/24/2024]
Abstract
This Review explores the evolution, improvements, and recent applications of Light Sheet Fluorescence Microscopy (LSFM) in biological research using a high numerical aperture detection objective (lens) for imaging subcellular structures. The Review begins with an overview of the development of LSFM, tracing its evolution from its inception to its current state and emphasizing key milestones and technological advancements over the years. Subsequently, we will discuss various improvements of LSFM techniques, covering advancements in hardware such as illumination strategies, optical designs, and sample preparation methods that have enhanced imaging capabilities and resolution. The advancements in data acquisition and processing are also included, which provides a brief overview of the recent development of artificial intelligence. Fluorescence probes that were commonly used in LSFM will be highlighted, together with some insights regarding the selection of potential probe candidates for future LSFM development. Furthermore, we also discuss recent advances in the application of LSFM with a focus on high numerical aperture detection objectives for various biological studies. For sample preparation techniques, there are discussions regarding fluorescence probe selection, tissue clearing protocols, and some insights into expansion microscopy. Integrated setups such as adaptive optics, single objective modification, and microfluidics will also be some of the key discussion points in this Review. We hope that this comprehensive Review will provide a holistic perspective on the historical development, technical enhancements, and cutting-edge applications of LSFM, showcasing its pivotal role and future potential in advancing biological research.
Collapse
Affiliation(s)
- Chun-Pei Shih
- Institute of Physics, Academia Sinica, Taipei 11529, Taiwan
- Department of Chemistry, National Taiwan University, Taipei 106319, Taiwan
- Nano Science and Technology Program, Taiwan International Graduate Program, Academia Sinica and National Taiwan University, Taipei 11529, Taiwan
| | - Wei-Chun Tang
- Research Center for Applied Sciences, Academia Sinica, Taipei 11529, Taiwan
| | - Peilin Chen
- Institute of Physics, Academia Sinica, Taipei 11529, Taiwan
- Research Center for Applied Sciences, Academia Sinica, Taipei 11529, Taiwan
| | - Bi-Chang Chen
- Research Center for Applied Sciences, Academia Sinica, Taipei 11529, Taiwan
| |
Collapse
|
18
|
Lin J, Mehra D, Marin Z, Wang X, Borges HM, Shen Q, Gałecki S, Haug J, Abbott DH, Dean KM. Mechanically sheared axially swept light-sheet microscopy. BIOMEDICAL OPTICS EXPRESS 2024; 15:5314-5327. [PMID: 39296406 PMCID: PMC11407235 DOI: 10.1364/boe.526145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Revised: 07/06/2024] [Accepted: 07/23/2024] [Indexed: 09/21/2024]
Abstract
We present a mechanically sheared image acquisition format for upright and open-top light-sheet microscopes that automatically places data in its proper spatial context. This approach, which reduces computational post-processing and eliminates unnecessary interpolation or duplication of the data, is demonstrated on an upright variant of axially swept light-sheet microscopy (ASLM) that achieves a field of view, measuring 774 × 435 microns, that is 3.2-fold larger than previous models and a raw and isotropic resolution of ∼460 nm. Combined, we demonstrate the power of this approach by imaging sub-diffraction beads, cleared biological tissues, and expanded specimens.
Collapse
Affiliation(s)
- Jinlong Lin
- Lyda Hill Department of Bioinformatics, UT Southwestern Medical Center, 6000 Harry Hines Blvd, Dallas, TX 75390, USA
- Cecil H. and Ida Green Center for Systems Biology, UT Southwestern Medical Center, 6000 Harry Hines Blvd, Dallas, TX 75390, USA
| | - Dushyant Mehra
- Lyda Hill Department of Bioinformatics, UT Southwestern Medical Center, 6000 Harry Hines Blvd, Dallas, TX 75390, USA
- Cecil H. and Ida Green Center for Systems Biology, UT Southwestern Medical Center, 6000 Harry Hines Blvd, Dallas, TX 75390, USA
| | - Zach Marin
- Lyda Hill Department of Bioinformatics, UT Southwestern Medical Center, 6000 Harry Hines Blvd, Dallas, TX 75390, USA
- Cecil H. and Ida Green Center for Systems Biology, UT Southwestern Medical Center, 6000 Harry Hines Blvd, Dallas, TX 75390, USA
- Max Perutz Labs, Department of Structural and Computational Biology, University of Vienna, Dr. Bohr-Gasse 9, 1030 Vienna, Austria
| | - Xiaoding Wang
- Lyda Hill Department of Bioinformatics, UT Southwestern Medical Center, 6000 Harry Hines Blvd, Dallas, TX 75390, USA
- Cecil H. and Ida Green Center for Systems Biology, UT Southwestern Medical Center, 6000 Harry Hines Blvd, Dallas, TX 75390, USA
| | - Hazel M. Borges
- Lyda Hill Department of Bioinformatics, UT Southwestern Medical Center, 6000 Harry Hines Blvd, Dallas, TX 75390, USA
- Cecil H. and Ida Green Center for Systems Biology, UT Southwestern Medical Center, 6000 Harry Hines Blvd, Dallas, TX 75390, USA
| | - Qionghua Shen
- Lyda Hill Department of Bioinformatics, UT Southwestern Medical Center, 6000 Harry Hines Blvd, Dallas, TX 75390, USA
- Cecil H. and Ida Green Center for Systems Biology, UT Southwestern Medical Center, 6000 Harry Hines Blvd, Dallas, TX 75390, USA
| | - Seweryn Gałecki
- Lyda Hill Department of Bioinformatics, UT Southwestern Medical Center, 6000 Harry Hines Blvd, Dallas, TX 75390, USA
- Cecil H. and Ida Green Center for Systems Biology, UT Southwestern Medical Center, 6000 Harry Hines Blvd, Dallas, TX 75390, USA
- Department of Systems Biology and Engineering, Silesian University of Technology, Akademicka 16, 44-100 Gliwice, Poland
| | - John Haug
- Lyda Hill Department of Bioinformatics, UT Southwestern Medical Center, 6000 Harry Hines Blvd, Dallas, TX 75390, USA
- Cecil H. and Ida Green Center for Systems Biology, UT Southwestern Medical Center, 6000 Harry Hines Blvd, Dallas, TX 75390, USA
| | - Derek H. Abbott
- Lyda Hill Department of Bioinformatics, UT Southwestern Medical Center, 6000 Harry Hines Blvd, Dallas, TX 75390, USA
- Cecil H. and Ida Green Center for Systems Biology, UT Southwestern Medical Center, 6000 Harry Hines Blvd, Dallas, TX 75390, USA
| | - Kevin M. Dean
- Lyda Hill Department of Bioinformatics, UT Southwestern Medical Center, 6000 Harry Hines Blvd, Dallas, TX 75390, USA
- Cecil H. and Ida Green Center for Systems Biology, UT Southwestern Medical Center, 6000 Harry Hines Blvd, Dallas, TX 75390, USA
| |
Collapse
|
19
|
Chen Y, Chauhan S, Gong C, Dayton H, Xu C, De La Cruz ED, Tsai YYW, Datta MS, Rosoklija GB, Dwork AJ, Mann JJ, Boldrini M, Leong KW, Dietrich LEP, Tomer R. Low-cost and scalable projected light-sheet microscopy for the high-resolution imaging of cleared tissue and living samples. Nat Biomed Eng 2024; 8:1109-1123. [PMID: 39209948 DOI: 10.1038/s41551-024-01249-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2023] [Accepted: 08/02/2024] [Indexed: 09/04/2024]
Abstract
Light-sheet fluorescence microscopy (LSFM) is a widely used technique for imaging cleared tissue and living samples. However, high-performance LSFM systems are typically expensive and not easily scalable. Here we introduce a low-cost, scalable and versatile LSFM framework, which we named 'projected light-sheet microscopy' (pLSM), with high imaging performance and small device and computational footprints. We characterized the capabilities of pLSM, which repurposes readily available consumer-grade components, optimized optics, over-network control architecture and software-driven light-sheet modulation, by performing high-resolution mapping of cleared mouse brains and of post-mortem pathological human brain samples, and via the molecular phenotyping of brain and blood-vessel organoids derived from human induced pluripotent stem cells. We also report a method that leverages pLSM for the live imaging of the dynamics of sparsely labelled multi-layered bacterial pellicle biofilms at an air-liquid interface. pLSM can make high-resolution LSFM for biomedical applications more accessible, affordable and scalable.
Collapse
Affiliation(s)
- Yannan Chen
- Department of Biological Sciences, Columbia University, New York, NY, USA
- Department of Biomedical Engineering, Columbia University, New York, NY, USA
| | - Shradha Chauhan
- Department of Biological Sciences, Columbia University, New York, NY, USA
| | - Cheng Gong
- Department of Biological Sciences, Columbia University, New York, NY, USA
- Department of Biomedical Engineering, Columbia University, New York, NY, USA
| | - Hannah Dayton
- Department of Biological Sciences, Columbia University, New York, NY, USA
| | - Cong Xu
- Department of Biomedical Engineering, Columbia University, New York, NY, USA
| | | | - Yu-Young Wesley Tsai
- Department of Biological Sciences, Columbia University, New York, NY, USA
- Mortimer B. Zuckerman Mind Brain and Behavior Institute, Columbia University, New York, NY, USA
| | - Malika S Datta
- Department of Biological Sciences, Columbia University, New York, NY, USA
- Mortimer B. Zuckerman Mind Brain and Behavior Institute, Columbia University, New York, NY, USA
| | - Gorazd B Rosoklija
- Department of Psychiatry, Columbia University Irving Medical Center, New York State Psychiatric Institute, New York, NY, USA
| | - Andrew J Dwork
- Department of Psychiatry, Columbia University Irving Medical Center, New York State Psychiatric Institute, New York, NY, USA
- Department of Pathology and Cell Biology, Columbia University, New York, NY, USA
| | - J John Mann
- Department of Psychiatry, Columbia University Irving Medical Center, New York State Psychiatric Institute, New York, NY, USA
| | - Maura Boldrini
- Department of Psychiatry, Columbia University Irving Medical Center, New York State Psychiatric Institute, New York, NY, USA
| | - Kam W Leong
- Department of Biomedical Engineering, Columbia University, New York, NY, USA
| | - Lars E P Dietrich
- Department of Biological Sciences, Columbia University, New York, NY, USA
| | - Raju Tomer
- Department of Biological Sciences, Columbia University, New York, NY, USA.
- Department of Biomedical Engineering, Columbia University, New York, NY, USA.
- Mortimer B. Zuckerman Mind Brain and Behavior Institute, Columbia University, New York, NY, USA.
| |
Collapse
|
20
|
Farrar Z, Afshar A, Zare A, Mussin NM, Kaliyev AA, Zhilisbayeva KR, Mahdipour M, Tamadon A. Tissue clearing and three-dimensional imaging of intact tissues: a review on FACT protocol. J Histotechnol 2024; 47:126-142. [PMID: 38752929 DOI: 10.1080/01478885.2024.2352695] [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] [Accepted: 05/03/2024] [Indexed: 08/31/2024]
Abstract
FACT is a developed technique for clearing tissues that does not use acrylamide. Since the removal of lipids is crucial for transparency and efficient antibody staining throughout the tissue, especially for microscopy and imaging, it is a harmful process that can cause the loss of important biological molecules such as proteins. The FACT technique overcomes this by chemically bonding the membrane and intracellular proteins with the extracellular matrix, creating a massive 3D hydrogel matrix and providing structural support to fortify the tissue during processing. Compared to other acrylamide-based techniques, the FACT technique requires less labor and harmful chemicals and is therefore considered a more suitable option. In this study, we describe the complete FACT protocol for antibody staining and imaging of whole-cleared tissues while preserving structure and improving image quality. The protocol includes tissue perfusion, fixation, clearing, antibody staining, refractive index matching (RIM) (), microscopy, and imaging. The timing for each step varies depending on the size, weight, and type of tissue, as well as the type of immunostaining. We provide an example of the FACT protocol using mouse brain tissue, which demonstrates its suitability for molecular interrogation analysis of large tissues. The FACT technique has been successfully performed on different types of tissues, making it a favorable choice for a variety of applications.
Collapse
Affiliation(s)
- Zohreh Farrar
- Student Research Committee, Bushehr University of Medical Sciences, Bushehr, Iran
| | - Alireza Afshar
- Student Research Committee, Bushehr University of Medical Sciences, Bushehr, Iran
| | - Afshin Zare
- Department of Research and Development, PerciaVista R&D Co., Shiraz, Iran
| | - Nadiar M Mussin
- Department of General Surgery, West Kazakhstan Marat Ospanov Medical University, Aktobe, Kazakhstan
| | - Asset A Kaliyev
- Department of General Surgery, West Kazakhstan Marat Ospanov Medical University, Aktobe, Kazakhstan
| | - Kulyash R Zhilisbayeva
- Department of Scientific Work, West Kazakhstan Marat Ospanov Medical University, Aktobe, Kazakhstan
| | - Mahdi Mahdipour
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Applied Cell Sciences, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Amin Tamadon
- Department of Research and Development, PerciaVista R&D Co., Shiraz, Iran
- Department of Natural Sciences, West Kazakhstan Marat Ospanov Medical University, Aktobe, Kazakhstan
| |
Collapse
|
21
|
Mino T, Nonaka H, Hamachi I. Molecular anchoring and fluorescent labeling in animals compatible with tissue clearing for 3D imaging. Curr Opin Chem Biol 2024; 81:102474. [PMID: 38838505 DOI: 10.1016/j.cbpa.2024.102474] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 05/14/2024] [Accepted: 05/16/2024] [Indexed: 06/07/2024]
Abstract
Analyzing the quantity and distribution of molecules throughout intact biological tissue is crucial for understanding various biological phenomena. Traditional methods involving destructive extraction result in the loss of spatial information. Conversely, tissue-clearing techniques combined with fluorescence imaging have recently emerged as a powerful tool for deep tissue imaging without sacrificing spatial coverage. Key to this approach is the anchoring and labeling of targets in intact tissue. In this review, methods for anchoring and labeling proteins, lipids, carbohydrates, and small molecules are presented. Future directions include the development of activity-based probes that work in vivo and mark transient events with spatial information to enable a deeper understanding of biological phenomena.
Collapse
Affiliation(s)
- Takeharu Mino
- Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering, Kyoto University, Kyoto 615-8510, Japan
| | - Hiroshi Nonaka
- Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering, Kyoto University, Kyoto 615-8510, Japan; ERATO (Exploratory Research for Advanced Technology, JST), Tokyo 102-0075, Japan
| | - Itaru Hamachi
- Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering, Kyoto University, Kyoto 615-8510, Japan; ERATO (Exploratory Research for Advanced Technology, JST), Tokyo 102-0075, Japan.
| |
Collapse
|
22
|
Tamura I, Sakamoto DM, Yi B, Saito Y, Yamada N, Morimoto J, Takakusagi Y, Kuroda M, Kubota SI, Yatabe H, Kobayashi M, Harada H, Tainaka K, Sando S. Click3D: Click reaction across deep tissues for whole-organ 3D fluorescence imaging. SCIENCE ADVANCES 2024; 10:eado8471. [PMID: 39018410 PMCID: PMC466963 DOI: 10.1126/sciadv.ado8471] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/25/2024] [Accepted: 06/11/2024] [Indexed: 07/19/2024]
Abstract
Click chemistry offers various applications through efficient bioorthogonal reactions. In bioimaging, pretargeting strategies have often been used, using click reactions between molecular probes with a click handle and reporter molecules that make them observable. Recent efforts have integrated tissue-clearing techniques with fluorescent labeling through click chemistry, allowing high-resolution three-dimensional fluorescence imaging. Nevertheless, these techniques have faced a challenge in limited staining depth, confining their use to imaging tissue sections or partial organs. In this study, we introduce Click3D, a method for thoroughly staining whole organs using click chemistry. We identified click reaction conditions that improve staining depth with our custom-developed assay. The Click3D protocol exhibits a greater staining depth compared to conventional methods. Using Click3D, we have successfully achieved whole-kidney imaging of nascent RNA and whole-tumor imaging of hypoxia. We have also accomplished whole-brain imaging of hypoxia by using the clickable hypoxia probe, which has a small size and, therefore, has high permeability to cross the blood-brain barrier.
Collapse
Affiliation(s)
- Iori Tamura
- Department of Chemistry and Biotechnology, Graduate School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan
| | - Daichi M. Sakamoto
- Department of Chemistry and Biotechnology, Graduate School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan
| | - Bo Yi
- Department of Chemistry and Biotechnology, Graduate School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan
| | - Yutaro Saito
- Department of Chemistry and Biotechnology, Graduate School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan
| | - Naoki Yamada
- Department of Chemistry and Biotechnology, Graduate School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan
| | - Jumpei Morimoto
- Department of Chemistry and Biotechnology, Graduate School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan
| | - Yoichi Takakusagi
- Quantum Hyperpolarized MRI Research Team, Institute for Quantum Life Science, National Institutes for Quantum Science and Technology, 4-9-1 Anagawa, Inage-ku, Chiba 263-8555, Japan
- Institute for Quantum Medical Science, National Institutes for Quantum Science and Technology, 4-9-1 Anagawa, Inage-ku, Chiba 263-8555, Japan
| | - Masafumi Kuroda
- International Research Center for Neurointelligence (WPI-IRCN), The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Shimpei I. Kubota
- Division of Molecular Psychoimmunology, Institute for Genetic Medicine, Graduate School of Medicine, Hokkaido University, Kita-15, Nishi-7, Kita-ku, Sapporo, Hokkaido 060-0815, Japan
| | - Hiroyuki Yatabe
- Department of Chemistry and Biotechnology, Graduate School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan
| | - Minoru Kobayashi
- Laboratory of Cancer Cell Biology, Graduate School of Biostudies, Kyoto University, Yoshida Konoe-cho, Sakyo-ku, Kyoto 606-8501, Japan
- Department of Genome Dynamics, Radiation Biology Center, Graduate School of Biostudies, Kyoto University, Yoshida Konoe-cho, Sakyo-ku, Kyoto 606-8501, Japan
| | - Hiroshi Harada
- Laboratory of Cancer Cell Biology, Graduate School of Biostudies, Kyoto University, Yoshida Konoe-cho, Sakyo-ku, Kyoto 606-8501, Japan
- Department of Genome Dynamics, Radiation Biology Center, Graduate School of Biostudies, Kyoto University, Yoshida Konoe-cho, Sakyo-ku, Kyoto 606-8501, Japan
| | - Kazuki Tainaka
- Department of System Pathology for Neurological Disorders, Brain Research Institute, Niigata University, 1-757 Asahimachidori, Chuo-ku, Niigata 951-8585, Japan
| | - Shinsuke Sando
- Department of Chemistry and Biotechnology, Graduate School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan
- Department of Bioengineering, Graduate School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan
| |
Collapse
|
23
|
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
|
24
|
Ding Y, Huang Y, Gao P, Thai A, Chilaparasetti AN, Gopi M, Xu X, Li C. Brain image data processing using collaborative data workflows on Texera. Front Neural Circuits 2024; 18:1398884. [PMID: 39050044 PMCID: PMC11266044 DOI: 10.3389/fncir.2024.1398884] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2024] [Accepted: 06/20/2024] [Indexed: 07/27/2024] Open
Abstract
In the realm of neuroscience, mapping the three-dimensional (3D) neural circuitry and architecture of the brain is important for advancing our understanding of neural circuit organization and function. This study presents a novel pipeline that transforms mouse brain samples into detailed 3D brain models using a collaborative data analytics platform called "Texera." The user-friendly Texera platform allows for effective interdisciplinary collaboration between team members in neuroscience, computer vision, and data processing. Our pipeline utilizes the tile images from a serial two-photon tomography/TissueCyte system, then stitches tile images into brain section images, and constructs 3D whole-brain image datasets. The resulting 3D data supports downstream analyses, including 3D whole-brain registration, atlas-based segmentation, cell counting, and high-resolution volumetric visualization. Using this platform, we implemented specialized optimization methods and obtained significant performance enhancement in workflow operations. We expect the neuroscience community can adopt our approach for large-scale image-based data processing and analysis.
Collapse
Affiliation(s)
- Yunyan Ding
- Department of Computer Science, University of California, Irvine, Irvine, CA, United States
| | - Yicong Huang
- Department of Computer Science, University of California, Irvine, Irvine, CA, United States
| | - Pan Gao
- Department of Anatomy and Neurobiology, School of Medicine, University of California, Irvine, Irvine, CA, United States
| | - Andy Thai
- Department of Computer Science, University of California, Irvine, Irvine, CA, United States
| | | | - M. Gopi
- Department of Computer Science, University of California, Irvine, Irvine, CA, United States
| | - Xiangmin Xu
- Department of Computer Science, University of California, Irvine, Irvine, CA, United States
- Department of Anatomy and Neurobiology, School of Medicine, University of California, Irvine, Irvine, CA, United States
- Department of Biomedical Engineering, University of California, Irvine, Irvine, CA, United States
- The Center for Neural Circuit Mapping, University of California, Irvine, Irvine, CA, United States
| | - Chen Li
- Department of Computer Science, University of California, Irvine, Irvine, CA, United States
- The Center for Neural Circuit Mapping, University of California, Irvine, Irvine, CA, United States
| |
Collapse
|
25
|
Feng R, Xie J, Gao L. EDTP enhances and protects the fluorescent signal of GFP in cleared and expanded tissues. Sci Rep 2024; 14:15279. [PMID: 38961181 PMCID: PMC11222453 DOI: 10.1038/s41598-024-66398-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Accepted: 07/01/2024] [Indexed: 07/05/2024] Open
Abstract
Advanced 3D high-resolution imaging techniques are essential for investigating biological challenges, such as neural circuit analysis and tumor microenvironment in intact tissues. However, the fluorescence signal emitted by endogenous fluorescent proteins in cleared or expanded biological samples gradually diminishes with repeated irradiation and prolonged imaging, compromising its ability to accurately depict the underlying scientific problem. We have developed a strategy to preserve fluorescence in cleared and expanded tissue samples during prolonged high-resolution three-dimensional imaging. We evaluated various compounds at different concentrations to determine their ability to enhance fluorescence intensity and resistance to photobleaching while maintaining the structural integrity of the tissue. Specifically, we investigated the impact of EDTP utilization on GFP, as it has been observed to significantly improve fluorescence intensity, resistance to photobleaching, and maintain fluorescence during extended room temperature storage. This breakthrough will facilitate extended hydrophilic and hydrogel-based clearing and expansion methods for achieving long-term high-resolution 3D imaging of cleared biological tissues by effectively safeguarding fluorescent proteins within the tissue.
Collapse
Affiliation(s)
- Ruili Feng
- Fudan University, Shanghai, 200433, China.
- Key Laboratory of Structural Biology of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, 310024, Zhejiang, China.
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, 310024, Zhejiang, China.
| | - Jiongfang Xie
- Fudan University, Shanghai, 200433, China
- Key Laboratory of Structural Biology of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, 310024, Zhejiang, China
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, 310024, Zhejiang, China
| | - Liang Gao
- Fudan University, Shanghai, 200433, China
- Key Laboratory of Structural Biology of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, 310024, Zhejiang, China
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, 310024, Zhejiang, China
| |
Collapse
|
26
|
Soumier A, Lio G, Demily C. Current and future applications of light-sheet imaging for identifying molecular and developmental processes in autism spectrum disorders. Mol Psychiatry 2024; 29:2274-2284. [PMID: 38443634 DOI: 10.1038/s41380-024-02487-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Revised: 02/09/2024] [Accepted: 02/13/2024] [Indexed: 03/07/2024]
Abstract
Autism spectrum disorder (ASD) is identified by a set of neurodevelopmental divergences that typically affect the social communication domain. ASD is also characterized by heterogeneous cognitive impairments and is associated with cooccurring physical and medical conditions. As behaviors emerge as the brain matures, it is particularly essential to identify any gaps in neurodevelopmental trajectories during early perinatal life. Here, we introduce the potential of light-sheet imaging for studying developmental biology and cross-scale interactions among genetic, cellular, molecular and macroscale levels of circuitry and connectivity. We first report the core principles of light-sheet imaging and the recent progress in studying brain development in preclinical animal models and human organoids. We also present studies using light-sheet imaging to understand the development and function of other organs, such as the skin and gastrointestinal tract. We also provide information on the potential of light-sheet imaging in preclinical drug development. Finally, we speculate on the translational benefits of light-sheet imaging for studying individual brain-body interactions in advancing ASD research and creating personalized interventions.
Collapse
Affiliation(s)
- Amelie Soumier
- Le Vinatier Hospital Center, 95 boulevard Pinel, 69675, Bron cedex, France.
- iMIND, Center of Excellence for Autism, 95 boulevard Pinel, 69675, Bron cedex, France.
- Institute of Cognitive Science Marc Jeannerod, CNRS, UMR 5229, 67 boulevard Pinel, 69675, Bron cedex, France.
- University Claude Bernard Lyon 1, 43 boulevard du 11 Novembre 1918, 69622, Villeurbanne cedex, France.
| | - Guillaume Lio
- Le Vinatier Hospital Center, 95 boulevard Pinel, 69675, Bron cedex, France
- iMIND, Center of Excellence for Autism, 95 boulevard Pinel, 69675, Bron cedex, France
- Institute of Cognitive Science Marc Jeannerod, CNRS, UMR 5229, 67 boulevard Pinel, 69675, Bron cedex, France
| | - Caroline Demily
- Le Vinatier Hospital Center, 95 boulevard Pinel, 69675, Bron cedex, France
- iMIND, Center of Excellence for Autism, 95 boulevard Pinel, 69675, Bron cedex, France
- Institute of Cognitive Science Marc Jeannerod, CNRS, UMR 5229, 67 boulevard Pinel, 69675, Bron cedex, France
- University Claude Bernard Lyon 1, 43 boulevard du 11 Novembre 1918, 69622, Villeurbanne cedex, France
| |
Collapse
|
27
|
Otomo K, Omura T, Nozawa Y, Edwards SJ, Sato Y, Saito Y, Yagishita S, Uchida H, Watakabe Y, Naitou K, Yanai R, Sahara N, Takagi S, Katayama R, Iwata Y, Shiokawa T, Hayakawa Y, Otsuka K, Watanabe-Takano H, Haneda Y, Fukuhara S, Fujiwara M, Nii T, Meno C, Takeshita N, Yashiro K, Rosales Rocabado JM, Kaku M, Yamada T, Oishi Y, Koike H, Cheng Y, Sekine K, Koga JI, Sugiyama K, Kimura K, Karube F, Kim H, Manabe I, Nemoto T, Tainaka K, Hamada A, Brismar H, Susaki EA. descSPIM: an affordable and easy-to-build light-sheet microscope optimized for tissue clearing techniques. Nat Commun 2024; 15:4941. [PMID: 38866781 PMCID: PMC11169475 DOI: 10.1038/s41467-024-49131-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Accepted: 05/24/2024] [Indexed: 06/14/2024] Open
Abstract
Despite widespread adoption of tissue clearing techniques in recent years, poor access to suitable light-sheet fluorescence microscopes remains a major obstacle for biomedical end-users. Here, we present descSPIM (desktop-equipped SPIM for cleared specimens), a low-cost ($20,000-50,000), low-expertise (one-day installation by a non-expert), yet practical do-it-yourself light-sheet microscope as a solution for this bottleneck. Even the most fundamental configuration of descSPIM enables multi-color imaging of whole mouse brains and a cancer cell line-derived xenograft tumor mass for the visualization of neurocircuitry, assessment of drug distribution, and pathological examination by false-colored hematoxylin and eosin staining in a three-dimensional manner. Academically open-sourced ( https://github.com/dbsb-juntendo/descSPIM ), descSPIM allows routine three-dimensional imaging of cleared samples in minutes. Thus, the dissemination of descSPIM will accelerate biomedical discoveries driven by tissue clearing technologies.
Collapse
Affiliation(s)
- Kohei Otomo
- Department of Biochemistry and Systems Biomedicine, Juntendo University Graduate School of Medicine, Tokyo, Japan
- Biochemistry II, Juntendo University School of Medicine, Tokyo, Japan
- Nakatani Biomedical Spatialomics Hub, Juntendo University Graduate School of Medicine, Tokyo, Japan
- Division of Biophotonics, National Institute for Physiological Sciences, National Institutes of Natural Sciences, Okazaki, Japan
- Biophotonics Research Group, Exploratory Research Center on Life and Living Systems, National Institutes of Natural Sciences, Okazaki, Japan
| | - Takaki Omura
- Department of Biochemistry and Systems Biomedicine, Juntendo University Graduate School of Medicine, Tokyo, Japan
- Nakatani Biomedical Spatialomics Hub, Juntendo University Graduate School of Medicine, Tokyo, Japan
- Department of Neurosurgery, University of Tokyo, Tokyo, Japan
- Department of Neurosurgery and Neuro-Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Yuki Nozawa
- Biochemistry II, Juntendo University School of Medicine, Tokyo, Japan
| | - Steven J Edwards
- Science for Life Laboratory, Department of Applied Physics, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Yukihiko Sato
- Department of Biochemistry and Systems Biomedicine, Juntendo University Graduate School of Medicine, Tokyo, Japan
- Nakatani Biomedical Spatialomics Hub, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Yuri Saito
- Department of Biochemistry and Systems Biomedicine, Juntendo University Graduate School of Medicine, Tokyo, Japan
- Nakatani Biomedical Spatialomics Hub, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Shigehiro Yagishita
- Department of Pharmacology and Therapeutics, Fundamental Innovative Oncology Core, National Cancer Center Research Institute, Tokyo, Japan
- Division of Molecular Pharmacology, National Cancer Center Research Institute, Tokyo, Japan
| | - Hitoshi Uchida
- Department of System Pathology for Neurological Disorders, Brain Research Institute, Niigata University, Niigata, Japan
| | - Yuki Watakabe
- Division of Biophotonics, National Institute for Physiological Sciences, National Institutes of Natural Sciences, Okazaki, Japan
- Biophotonics Research Group, Exploratory Research Center on Life and Living Systems, National Institutes of Natural Sciences, Okazaki, Japan
| | - Kiyotada Naitou
- Department of Basic Veterinary Science, Joint Faculty of Veterinary Medicine, Kagoshima University, Kagoshima, Japan
| | - Rin Yanai
- Advanced Neuroimaging Center, Institute for Quantum Medical Sciences, National Institutes for Quantum Science and Technology, Chiba, Japan
| | - Naruhiko Sahara
- Advanced Neuroimaging Center, Institute for Quantum Medical Sciences, National Institutes for Quantum Science and Technology, Chiba, Japan
| | - Satoshi Takagi
- Division of Experimental Chemotherapy, Cancer Chemotherapy Center, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Ryohei Katayama
- Division of Experimental Chemotherapy, Cancer Chemotherapy Center, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Yusuke Iwata
- Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Toshiro Shiokawa
- Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Yoku Hayakawa
- Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Kensuke Otsuka
- Biology and Environmental Chemistry Division, Sustainable System Research Laboratory, Central Research Institute of Electric Power Industry, Chiba, Japan
| | - Haruko Watanabe-Takano
- Department of Molecular Pathophysiology, Institute for Advanced Medical Sciences, Nippon Medical School, Tokyo, Japan
| | - Yuka Haneda
- Department of Molecular Pathophysiology, Institute for Advanced Medical Sciences, Nippon Medical School, Tokyo, Japan
| | - Shigetomo Fukuhara
- Department of Molecular Pathophysiology, Institute for Advanced Medical Sciences, Nippon Medical School, Tokyo, Japan
| | - Miku Fujiwara
- Department of Developmental Biology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Takenobu Nii
- Department of Developmental Biology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Chikara Meno
- Department of Developmental Biology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Naoki Takeshita
- Anatomy and Developmental Biology, Kyoto Prefectural University of Medicine, Kyoto, Japan
- Department of Pediatrics, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Kenta Yashiro
- Anatomy and Developmental Biology, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Juan Marcelo Rosales Rocabado
- Division of Bio-prosthodontics, Faculty of Dentistry & Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
| | - Masaru Kaku
- Division of Bio-prosthodontics, Faculty of Dentistry & Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
| | - Tatsuya Yamada
- Department of Biochemistry, University of Nebraska-Lincoln, Lincoln, USA
| | - Yumiko Oishi
- Department of Meidical Biochemistry, Tokyo Medical and Dental University, Tokyo, Japan
| | - Hiroyuki Koike
- Department of Meidical Biochemistry, Tokyo Medical and Dental University, Tokyo, Japan
| | - Yinglan Cheng
- Department of Meidical Biochemistry, Tokyo Medical and Dental University, Tokyo, Japan
| | - Keisuke Sekine
- Laboratory of Cancer Cell Systems, National Cancer Center Research Institute, Tokyo, Japan
| | - Jun-Ichiro Koga
- The Second Department of Internal Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Kaori Sugiyama
- Institute for Advanced Research of Biosystem Dynamics, Research Institute for Science and Engineering, Waseda University, Tokyo, Japan
| | - Kenichi Kimura
- Life Science Center for Survival Dynamics, Tsukuba Advanced Research Alliance (TARA), University of Tsukuba, Tsukuba, Japan
| | - Fuyuki Karube
- Lab of Histology and Cytology, Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Hyeree Kim
- Department of Systems Medicine, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Ichiro Manabe
- Department of Systems Medicine, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Tomomi Nemoto
- Division of Biophotonics, National Institute for Physiological Sciences, National Institutes of Natural Sciences, Okazaki, Japan
- Biophotonics Research Group, Exploratory Research Center on Life and Living Systems, National Institutes of Natural Sciences, Okazaki, Japan
| | - Kazuki Tainaka
- Department of System Pathology for Neurological Disorders, Brain Research Institute, Niigata University, Niigata, Japan
| | - Akinobu Hamada
- Department of Pharmacology and Therapeutics, Fundamental Innovative Oncology Core, National Cancer Center Research Institute, Tokyo, Japan
- Division of Molecular Pharmacology, National Cancer Center Research Institute, Tokyo, Japan
| | - Hjalmar Brismar
- Science for Life Laboratory, Department of Applied Physics, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Etsuo A Susaki
- Department of Biochemistry and Systems Biomedicine, Juntendo University Graduate School of Medicine, Tokyo, Japan.
- Biochemistry II, Juntendo University School of Medicine, Tokyo, Japan.
- Nakatani Biomedical Spatialomics Hub, Juntendo University Graduate School of Medicine, Tokyo, Japan.
| |
Collapse
|
28
|
Lin YH, Wang LW, Chen YH, Chan YC, Hu SH, Wu SY, Chiang CS, Huang GJ, Yang SD, Chu SW, Wang KC, Lin CH, Huang PH, Cheng HJ, Chen BC, Chu LA. Revealing intact neuronal circuitry in centimeter-sized formalin-fixed paraffin-embedded brain. eLife 2024; 13:RP93212. [PMID: 38775133 PMCID: PMC11111220 DOI: 10.7554/elife.93212] [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] [Indexed: 05/24/2024] Open
Abstract
Tissue-clearing and labeling techniques have revolutionized brain-wide imaging and analysis, yet their application to clinical formalin-fixed paraffin-embedded (FFPE) blocks remains challenging. We introduce HIF-Clear, a novel method for efficiently clearing and labeling centimeter-thick FFPE specimens using elevated temperature and concentrated detergents. HIF-Clear with multi-round immunolabeling reveals neuron circuitry regulating multiple neurotransmitter systems in a whole FFPE mouse brain and is able to be used as the evaluation of disease treatment efficiency. HIF-Clear also supports expansion microscopy and can be performed on a non-sectioned 15-year-old FFPE specimen, as well as a 3-month formalin-fixed mouse brain. Thus, HIF-Clear represents a feasible approach for researching archived FFPE specimens for future neuroscientific and 3D neuropathological analyses.
Collapse
Affiliation(s)
- Ya-Hui Lin
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua UniversityHsinchuTaiwan
- Brain Research Center, National Tsing Hua UniversityHsinchuTaiwan
| | - Li-Wen Wang
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua UniversityHsinchuTaiwan
- Brain Research Center, National Tsing Hua UniversityHsinchuTaiwan
| | - Yen-Hui Chen
- Institute of Biomedical Sciences, Academia SinicaTaipeiTaiwan
| | - Yi-Chieh Chan
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua UniversityHsinchuTaiwan
| | - Shang-Hsiu Hu
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua UniversityHsinchuTaiwan
| | - Sheng-Yan Wu
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua UniversityHsinchuTaiwan
| | - Chi-Shiun Chiang
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua UniversityHsinchuTaiwan
| | - Guan-Jie Huang
- Department of Physics, National Taiwan UniversityTaipeiTaiwan
| | - Shang-Da Yang
- Institute of Photonics Technologies, National Tsing Hua UniversityHsinchuTaiwan
| | - Shi-Wei Chu
- Department of Physics, National Taiwan UniversityTaipeiTaiwan
| | - Kuo-Chuan Wang
- Department of Neurosurgery, National Taiwan University HospitalTaipeiTaiwan
| | - Chin-Hsien Lin
- Department of Neurosurgery, National Taiwan University HospitalTaipeiTaiwan
| | - Pei-Hsin Huang
- Department of Pathology, National Taiwan University HospitalTaipeiTaiwan
| | | | - Bi-Chang Chen
- Research Center for Applied Sciences, Academia SinicaTaipeiTaiwan
| | - Li-An Chu
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua UniversityHsinchuTaiwan
- Brain Research Center, National Tsing Hua UniversityHsinchuTaiwan
| |
Collapse
|
29
|
Zheng J, Wu YC, Cai X, Phan P, Er EE, Zhao Z, Lee SSY. Correlative multiscale 3D imaging of mouse primary and metastatic tumors by sequential light sheet and confocal fluorescence microscopy. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.14.594162. [PMID: 38798657 PMCID: PMC11118317 DOI: 10.1101/2024.05.14.594162] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
Three-dimensional (3D) optical microscopy, combined with advanced tissue clearing, permits in situ interrogation of the tumor microenvironment (TME) in large volumetric tumors for preclinical cancer research. Light sheet (also known as ultramicroscopy) and confocal fluorescence microscopy are often used to achieve macroscopic and microscopic 3D images of optically cleared tumor tissues, respectively. Although each technique offers distinct fields of view (FOVs) and spatial resolution, the combination of these two optical microscopy techniques to obtain correlative multiscale 3D images from the same tumor tissues has not yet been explored. To establish correlative multiscale 3D optical microscopy, we developed a method for optically marking defined regions of interest (ROIs) within a cleared mouse tumor by employing a UV light-activated visible dye and Z-axis position-selective UV irradiation in a light sheet microscope system. By integrating this method with subsequent tissue processing, including physical ROI marking, reversal of tissue clearing, tissue macrosectioning, and multiplex immunofluorescence, we established a workflow that enables the tracking and 3D imaging of ROIs within tumor tissues through sequential light sheet and confocal fluorescence microscopy. This approach allowed for quantitative 3D spatial analysis of the immune response in the TME of a mouse mammary tumor following cancer immunotherapy at multiple spatial scales. The workflow also facilitated the direct localization of a metastatic lesion within a whole mouse brain. These results demonstrate that our ROI tracking method and its associated workflow offer a novel approach for correlative multiscale 3D optical microscopy, with the potential to provide new insights into tumor heterogeneity, metastasis, and response to therapy at various spatial levels.
Collapse
|
30
|
Deng Y, Zhu J, Liu X, Dai J, Yu T, Zhu D. A robust vessel-labeling pipeline with high tissue clearing compatibility for 3D mapping of vascular networks. iScience 2024; 27:109730. [PMID: 38706842 PMCID: PMC11068851 DOI: 10.1016/j.isci.2024.109730] [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: 11/15/2023] [Revised: 02/23/2024] [Accepted: 04/09/2024] [Indexed: 05/07/2024] Open
Abstract
The combination of vessel-labeling, tissue-clearing, and light-sheet imaging techniques provides a potent tool for accurately mapping vascular networks, enabling the assessment of vascular remodeling in vascular-related disorders. However, most vascular labeling methods face challenges such as inadequate labeling efficiency or poor compatibility with current tissue clearing technology, which significantly undermines the image quality. To address this limitation, we introduce a vessel-labeling pipeline, termed Ultralabel, which relies on a specially designed dye hydrogel containing lysine-fixable dextran and gelatins for double enhancement. Ultralabel demonstrates not only excellent vessel-labeling capability but also strong compatibility with all tissue clearing methods tested, which outperforms other vessel-labeling methods. Consequently, Ultralabel enables fine mapping of vascular networks in diverse organs, as well as multi-color labeling alongside other labeling techniques. Ultralabel should provide a robust and user-friendly method for obtaining 3D vascular networks in different biomedical applications.
Collapse
Affiliation(s)
- Yating Deng
- Britton Chance Center for Biomedical Photonics- MoE Key Laboratory for Biomedical Photonics, Advanced Biomedical Imaging Facility, Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology, Wuhan 430074, Hubei, China
| | - Jingtan Zhu
- Britton Chance Center for Biomedical Photonics- MoE Key Laboratory for Biomedical Photonics, Advanced Biomedical Imaging Facility, Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology, Wuhan 430074, Hubei, China
| | - Xiaomei Liu
- Britton Chance Center for Biomedical Photonics- MoE Key Laboratory for Biomedical Photonics, Advanced Biomedical Imaging Facility, Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology, Wuhan 430074, Hubei, China
| | - Junyao Dai
- Britton Chance Center for Biomedical Photonics- MoE Key Laboratory for Biomedical Photonics, Advanced Biomedical Imaging Facility, Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology, Wuhan 430074, Hubei, China
| | - Tingting Yu
- Britton Chance Center for Biomedical Photonics- MoE Key Laboratory for Biomedical Photonics, Advanced Biomedical Imaging Facility, Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology, Wuhan 430074, Hubei, China
| | - Dan Zhu
- Britton Chance Center for Biomedical Photonics- MoE Key Laboratory for Biomedical Photonics, Advanced Biomedical Imaging Facility, Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology, Wuhan 430074, Hubei, China
| |
Collapse
|
31
|
He C, Yuan Y, Gong C, Wang X, Lyu G. Applications of Tissue Clearing in Central and Peripheral Nerves. Neuroscience 2024; 546:104-117. [PMID: 38570062 DOI: 10.1016/j.neuroscience.2024.03.030] [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: 09/17/2023] [Revised: 03/25/2024] [Accepted: 03/27/2024] [Indexed: 04/05/2024]
Abstract
The techniques of tissue clearing have been proposed and applied in anatomical and biomedical research since the 19th century. As we all know, the original study of the nervous system relied on serial ultrathin sections and stereoscopic techniques. The 3D visualization of the nervous system was established by software splicing and reconstruction. With the development of science and technology, microscope equipment had constantly been upgraded. Despite the great progress that has been made in this field, the workload is too complex, and it needs high technical requirements. Abundant mistakes due to manual sections were inescapable and structural integrity remained questionable. According to the classification of tissue transparency methods, we introduced the latest application of transparency methods in central and peripheral nerve research from optical imaging, molecular markers and data analysis. This review summarizes the application of transparent technology in neural pathways. We hope to provide some inspiration for the continuous optimization of tissue clearing methods.
Collapse
Affiliation(s)
- Cheng He
- Department of Anatomy, Medical School of Nantong University, Nantong, China
| | - Ye Yuan
- Department of Anatomy, Medical School of Nantong University, Nantong, China
| | - Chuanhui Gong
- Department of Anatomy, Medical School of Nantong University, Nantong, China
| | - Xueying Wang
- Medical School of Nantong University, Nantong, China
| | - Guangming Lyu
- Department of Anatomy, Medical School of Nantong University, Nantong, China; Department of Anatomy, Institute of Neurobiology, Jiangsu Key Laboratory of Neuroregeneration, Medical School of Nantong University, Nantong, China.
| |
Collapse
|
32
|
Zheng W, Mu H, Chen Z, Liu J, Xia D, Cheng Y, Jing Q, Lau PM, Tang J, Bi GQ, Wu F, Wang H. NEATmap: a high-efficiency deep learning approach for whole mouse brain neuronal activity trace mapping. Natl Sci Rev 2024; 11:nwae109. [PMID: 38831937 PMCID: PMC11145917 DOI: 10.1093/nsr/nwae109] [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: 09/13/2023] [Revised: 01/26/2024] [Accepted: 02/25/2024] [Indexed: 06/05/2024] Open
Abstract
Quantitative analysis of activated neurons in mouse brains by a specific stimulation is usually a primary step to locate the responsive neurons throughout the brain. However, it is challenging to comprehensively and consistently analyze the neuronal activity trace in whole brains of a large cohort of mice from many terabytes of volumetric imaging data. Here, we introduce NEATmap, a deep learning-based high-efficiency, high-precision and user-friendly software for whole-brain neuronal activity trace mapping by automated segmentation and quantitative analysis of immunofluorescence labeled c-Fos+ neurons. We applied NEATmap to study the brain-wide differentiated neuronal activation in response to physical and psychological stressors in cohorts of mice.
Collapse
Affiliation(s)
- Weijie Zheng
- AHU-IAI AI Joint Laboratory, Anhui University, Hefei 230039, China
- Anhui Province Key Laboratory of Biomedical Imaging and Intelligent Processing, Institute of Artificial Intelligence, Hefei Comprehensive National Science Center, Hefei 230088, China
- National Engineering Laboratory for Brain-inspired Intelligence Technology and Application, School of Information Science and Technology, University of Science and Technology of China, Hefei 230026, China
| | - Huawei Mu
- Anhui Province Key Laboratory of Biomedical Imaging and Intelligent Processing, Institute of Artificial Intelligence, Hefei Comprehensive National Science Center, Hefei 230088, China
- Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China
| | - Zhiyi Chen
- Anhui Province Key Laboratory of Biomedical Imaging and Intelligent Processing, Institute of Artificial Intelligence, Hefei Comprehensive National Science Center, Hefei 230088, China
- National Engineering Laboratory for Brain-inspired Intelligence Technology and Application, School of Information Science and Technology, University of Science and Technology of China, Hefei 230026, China
| | - Jiajun Liu
- Anhui Province Key Laboratory of Biomedical Imaging and Intelligent Processing, Institute of Artificial Intelligence, Hefei Comprehensive National Science Center, Hefei 230088, China
- National Engineering Laboratory for Brain-inspired Intelligence Technology and Application, School of Information Science and Technology, University of Science and Technology of China, Hefei 230026, China
| | - Debin Xia
- Anhui Province Key Laboratory of Biomedical Imaging and Intelligent Processing, Institute of Artificial Intelligence, Hefei Comprehensive National Science Center, Hefei 230088, China
- National Engineering Laboratory for Brain-inspired Intelligence Technology and Application, School of Information Science and Technology, University of Science and Technology of China, Hefei 230026, China
| | - Yuxiao Cheng
- Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China
| | - Qi Jing
- Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China
| | - Pak-Ming Lau
- Anhui Province Key Laboratory of Biomedical Imaging and Intelligent Processing, Institute of Artificial Intelligence, Hefei Comprehensive National Science Center, Hefei 230088, China
- Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China
- Interdisciplinary Center for Brain Information, Brain Cognition and Brain Disease Institute, Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Jin Tang
- AHU-IAI AI Joint Laboratory, Anhui University, Hefei 230039, China
- Anhui Province Key Laboratory of Biomedical Imaging and Intelligent Processing, Institute of Artificial Intelligence, Hefei Comprehensive National Science Center, Hefei 230088, China
| | - Guo-Qiang Bi
- Anhui Province Key Laboratory of Biomedical Imaging and Intelligent Processing, Institute of Artificial Intelligence, Hefei Comprehensive National Science Center, Hefei 230088, China
- Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China
- Interdisciplinary Center for Brain Information, Brain Cognition and Brain Disease Institute, Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Feng Wu
- Anhui Province Key Laboratory of Biomedical Imaging and Intelligent Processing, Institute of Artificial Intelligence, Hefei Comprehensive National Science Center, Hefei 230088, China
- National Engineering Laboratory for Brain-inspired Intelligence Technology and Application, School of Information Science and Technology, University of Science and Technology of China, Hefei 230026, China
| | - Hao Wang
- Anhui Province Key Laboratory of Biomedical Imaging and Intelligent Processing, Institute of Artificial Intelligence, Hefei Comprehensive National Science Center, Hefei 230088, China
- National Engineering Laboratory for Brain-inspired Intelligence Technology and Application, School of Information Science and Technology, University of Science and Technology of China, Hefei 230026, China
| |
Collapse
|
33
|
Chen FD, Sharma A, Roszko DA, Xue T, Mu X, Luo X, Chua H, Lo PGQ, Sacher WD, Poon JKS. Development of wafer-scale multifunctional nanophotonic neural probes for brain activity mapping. LAB ON A CHIP 2024; 24:2397-2417. [PMID: 38623840 DOI: 10.1039/d3lc00931a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/17/2024]
Abstract
Optical techniques, such as optogenetic stimulation and functional fluorescence imaging, have been revolutionary for neuroscience by enabling neural circuit analysis with cell-type specificity. To probe deep brain regions, implantable light sources are crucial. Silicon photonics, commonly used for data communications, shows great promise in creating implantable devices with complex optical systems in a compact form factor compatible with high volume manufacturing practices. This article reviews recent developments of wafer-scale multifunctional nanophotonic neural probes. The probes can be realized on 200 or 300 mm wafers in commercial foundries and integrate light emitters for photostimulation, microelectrodes for electrophysiological recording, and microfluidic channels for chemical delivery and sampling. By integrating active optical devices to the probes, denser emitter arrays, enhanced on-chip biosensing, and increased ease of use may be realized. Silicon photonics technology makes possible highly versatile implantable neural probes that can transform neuroscience experiments.
Collapse
Affiliation(s)
- Fu Der Chen
- Max Planck Institute of Microstructure Physics, Weinberg 2, 06120 Halle, Germany.
- Department of Electrical and Computer Engineering, University of Toronto, 10 King's College Road, Toronto, Ontario M5S 3G4, Canada
| | - Ankita Sharma
- Max Planck Institute of Microstructure Physics, Weinberg 2, 06120 Halle, Germany.
- Department of Electrical and Computer Engineering, University of Toronto, 10 King's College Road, Toronto, Ontario M5S 3G4, Canada
| | - David A Roszko
- Max Planck Institute of Microstructure Physics, Weinberg 2, 06120 Halle, Germany.
- Department of Electrical and Computer Engineering, University of Toronto, 10 King's College Road, Toronto, Ontario M5S 3G4, Canada
| | - Tianyuan Xue
- Max Planck Institute of Microstructure Physics, Weinberg 2, 06120 Halle, Germany.
- Department of Electrical and Computer Engineering, University of Toronto, 10 King's College Road, Toronto, Ontario M5S 3G4, Canada
| | - Xin Mu
- Max Planck Institute of Microstructure Physics, Weinberg 2, 06120 Halle, Germany.
- Department of Electrical and Computer Engineering, University of Toronto, 10 King's College Road, Toronto, Ontario M5S 3G4, Canada
| | - Xianshu Luo
- Advanced Micro Foundry Pte Ltd, 11 Science Park Road, Singapore Science Park II, 117685, Singapore
| | - Hongyao Chua
- Advanced Micro Foundry Pte Ltd, 11 Science Park Road, Singapore Science Park II, 117685, Singapore
| | - Patrick Guo-Qiang Lo
- Advanced Micro Foundry Pte Ltd, 11 Science Park Road, Singapore Science Park II, 117685, Singapore
| | - Wesley D Sacher
- Max Planck Institute of Microstructure Physics, Weinberg 2, 06120 Halle, Germany.
| | - Joyce K S Poon
- Max Planck Institute of Microstructure Physics, Weinberg 2, 06120 Halle, Germany.
- Department of Electrical and Computer Engineering, University of Toronto, 10 King's College Road, Toronto, Ontario M5S 3G4, Canada
| |
Collapse
|
34
|
Zeng Y, Wang Y. Complete Neuron Reconstruction Based on Branch Confidence. Brain Sci 2024; 14:396. [PMID: 38672045 PMCID: PMC11047972 DOI: 10.3390/brainsci14040396] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Revised: 04/04/2024] [Accepted: 04/09/2024] [Indexed: 04/28/2024] Open
Abstract
In the past few years, significant advancements in microscopic imaging technology have led to the production of numerous high-resolution images capturing brain neurons at the micrometer scale. The reconstructed structure of neurons from neuronal images can serve as a valuable reference for research in brain diseases and neuroscience. Currently, there lacks an accurate and efficient method for neuron reconstruction. Manual reconstruction remains the primary approach, offering high accuracy but requiring significant time investment. While some automatic reconstruction methods are faster, they often sacrifice accuracy and cannot be directly relied upon. Therefore, the primary goal of this paper is to develop a neuron reconstruction tool that is both efficient and accurate. The tool aids users in reconstructing complete neurons by calculating the confidence of branches during the reconstruction process. The method models the neuron reconstruction as multiple Markov chains, and calculates the confidence of the connections between branches by simulating the reconstruction artifacts in the results. Users iteratively modify low-confidence branches to ensure precise and efficient neuron reconstruction. Experiments on both the publicly accessible BigNeuron dataset and a self-created Whole-Brain dataset demonstrate that the tool achieves high accuracy similar to manual reconstruction, while significantly reducing reconstruction time.
Collapse
Affiliation(s)
- Ying Zeng
- School of Computer Science and Technology, Shanghai University, Shanghai 200444, China;
- Guangdong Institute of Intelligence Science and Technology, Zhuhai 519031, China
| | - Yimin Wang
- Guangdong Institute of Intelligence Science and Technology, Zhuhai 519031, China
| |
Collapse
|
35
|
Gao P, Rivera M, Lin X, Holmes TC, Zhao H, Xu X. Immunolabeling-compatible PEGASOS tissue clearing for high-resolution whole mouse brain imaging. Front Neural Circuits 2024; 18:1345692. [PMID: 38694272 PMCID: PMC11061518 DOI: 10.3389/fncir.2024.1345692] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Accepted: 03/13/2024] [Indexed: 05/04/2024] Open
Abstract
Novel brain clearing methods revolutionize imaging by increasing visualization throughout the brain at high resolution. However, combining the standard tool of immunostaining targets of interest with clearing methods has lagged behind. We integrate whole-mount immunostaining with PEGASOS tissue clearing, referred to as iPEGASOS (immunostaining-compatible PEGASOS), to address the challenge of signal quenching during clearing processes. iPEGASOS effectively enhances molecular-genetically targeted fluorescent signals that are otherwise compromised during conventional clearing procedures. Additionally, we demonstrate the utility of iPEGASOS for visualizing neurochemical markers or viral labels to augment visualization that transgenic mouse lines cannot provide. Our study encompasses three distinct applications, each showcasing the versatility and efficacy of this approach. We employ whole-mount immunostaining to enhance molecular signals in transgenic reporter mouse lines to visualize the whole-brain spatial distribution of specific cellular populations. We also significantly improve the visualization of neural circuit connections by enhancing signals from viral tracers injected into the brain. Last, we show immunostaining without genetic markers to selectively label beta-amyloid deposits in a mouse model of Alzheimer's disease, facilitating the comprehensive whole-brain study of pathological features.
Collapse
Affiliation(s)
- Pan Gao
- Department of Anatomy and Neurobiology, School of Medicine, University of California, Irvine, Irvine, CA, United States
| | - Matthew Rivera
- Department of Anatomy and Neurobiology, School of Medicine, University of California, Irvine, Irvine, CA, United States
| | - Xiaoxiao Lin
- Department of Anatomy and Neurobiology, School of Medicine, University of California, Irvine, Irvine, CA, United States
| | - Todd C. Holmes
- Department of Physiology and Biophysics, School of Medicine, University of California, Irvine, Irvine, CA, United States
- Center for Neural Circuit Mapping, University of California, Irvine, Irvine, CA, United States
| | - Hu Zhao
- Chinese Institute for Brain Research, Beijing, China
| | - Xiangmin Xu
- Department of Anatomy and Neurobiology, School of Medicine, University of California, Irvine, Irvine, CA, United States
- Center for Neural Circuit Mapping, University of California, Irvine, Irvine, CA, United States
| |
Collapse
|
36
|
Lin J, Mehra D, Marin Z, Wang X, Borges HM, Shen Q, Gałecki S, Haug J, Dean KM. Mechanically Sheared Axially Swept Light-Sheet Microscopy. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.10.588892. [PMID: 38645073 PMCID: PMC11030395 DOI: 10.1101/2024.04.10.588892] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/23/2024]
Abstract
We present a mechanically sheared image acquisition format for upright and open-top light-sheet microscopes that automatically places data in its proper spatial context. This approach, which reduces computational post-processing and eliminates unnecessary interpolation or duplication of the data, is demonstrated on an upright variant of Axially Swept Light-Sheet Microscopy (ASLM) that achieves a field of view, measuring 774 x 435 microns, that is 3.2-fold larger than previous models and a raw and isotropic resolution of ∼420 nm. Combined, we demonstrate the power of this approach by imaging sub-diffraction beads, cleared biological tissues, and expanded specimens.
Collapse
|
37
|
Mai H, Luo J, Hoeher L, Al-Maskari R, Horvath I, Chen Y, Kofler F, Piraud M, Paetzold JC, Modamio J, Todorov M, Elsner M, Hellal F, Ertürk A. Whole-body cellular mapping in mouse using standard IgG antibodies. Nat Biotechnol 2024; 42:617-627. [PMID: 37430076 PMCID: PMC11021200 DOI: 10.1038/s41587-023-01846-0] [Citation(s) in RCA: 40] [Impact Index Per Article: 40.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Accepted: 05/26/2023] [Indexed: 07/12/2023]
Abstract
Whole-body imaging techniques play a vital role in exploring the interplay of physiological systems in maintaining health and driving disease. We introduce wildDISCO, a new approach for whole-body immunolabeling, optical clearing and imaging in mice, circumventing the need for transgenic reporter animals or nanobody labeling and so overcoming existing technical limitations. We identified heptakis(2,6-di-O-methyl)-β-cyclodextrin as a potent enhancer of cholesterol extraction and membrane permeabilization, enabling deep, homogeneous penetration of standard antibodies without aggregation. WildDISCO facilitates imaging of peripheral nervous systems, lymphatic vessels and immune cells in whole mice at cellular resolution by labeling diverse endogenous proteins. Additionally, we examined rare proliferating cells and the effects of biological perturbations, as demonstrated in germ-free mice. We applied wildDISCO to map tertiary lymphoid structures in the context of breast cancer, considering both primary tumor and metastases throughout the mouse body. An atlas of high-resolution images showcasing mouse nervous, lymphatic and vascular systems is accessible at http://discotechnologies.org/wildDISCO/atlas/index.php .
Collapse
Affiliation(s)
- Hongcheng Mai
- Institute for Tissue Engineering and Regenerative Medicine, Helmholtz Center Munich, Neuherberg, Germany
- Institute for Stroke and Dementia Research, Medical Centre of the University of Munich, Ludwig-Maximilians University of Munich, Munich, Germany
- Munich Medical Research School, Munich, Germany
- Deep Piction GmbH, Munich, Germany
| | - Jie Luo
- Institute for Tissue Engineering and Regenerative Medicine, Helmholtz Center Munich, Neuherberg, Germany
- Institute for Stroke and Dementia Research, Medical Centre of the University of Munich, Ludwig-Maximilians University of Munich, Munich, Germany
- Deep Piction GmbH, Munich, Germany
| | - Luciano Hoeher
- Institute for Tissue Engineering and Regenerative Medicine, Helmholtz Center Munich, Neuherberg, Germany
| | - Rami Al-Maskari
- Institute for Tissue Engineering and Regenerative Medicine, Helmholtz Center Munich, Neuherberg, Germany
- TUM School of Computation, Information and Technology, Technical University of Munich, Munich, Germany
| | - Izabela Horvath
- Institute for Tissue Engineering and Regenerative Medicine, Helmholtz Center Munich, Neuherberg, Germany
- TUM School of Computation, Information and Technology, Technical University of Munich, Munich, Germany
| | - Ying Chen
- Institute for Tissue Engineering and Regenerative Medicine, Helmholtz Center Munich, Neuherberg, Germany
- Institute for Stroke and Dementia Research, Medical Centre of the University of Munich, Ludwig-Maximilians University of Munich, Munich, Germany
- Faculty of Medicine, Ludwig-Maximilians University of Munich, Munich, Germany
| | - Florian Kofler
- Helmholtz Al, Helmholtz Center Munich, Neuherberg, Germany
- Department of Informatics, Technical University of Munich, Munich, Germany
- TranslaTUM - Central Institute for Translational Cancer Research, Technical University of Munich, Munich, Germany
- Department of Diagnostic and Interventional Neuroradiology, School of Medicine, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Marie Piraud
- Helmholtz Al, Helmholtz Center Munich, Neuherberg, Germany
| | - Johannes C Paetzold
- Institute for Tissue Engineering and Regenerative Medicine, Helmholtz Center Munich, Neuherberg, Germany
- Department of Computing, Imperial College London, London, UK
| | - Jennifer Modamio
- Institute for Tissue Engineering and Regenerative Medicine, Helmholtz Center Munich, Neuherberg, Germany
| | - Mihail Todorov
- Institute for Tissue Engineering and Regenerative Medicine, Helmholtz Center Munich, Neuherberg, Germany
- Institute for Stroke and Dementia Research, Medical Centre of the University of Munich, Ludwig-Maximilians University of Munich, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Markus Elsner
- Institute for Tissue Engineering and Regenerative Medicine, Helmholtz Center Munich, Neuherberg, Germany
| | - Farida Hellal
- Institute for Tissue Engineering and Regenerative Medicine, Helmholtz Center Munich, Neuherberg, Germany
- Institute for Stroke and Dementia Research, Medical Centre of the University of Munich, Ludwig-Maximilians University of Munich, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Ali Ertürk
- Institute for Tissue Engineering and Regenerative Medicine, Helmholtz Center Munich, Neuherberg, Germany.
- Institute for Stroke and Dementia Research, Medical Centre of the University of Munich, Ludwig-Maximilians University of Munich, Munich, Germany.
- Deep Piction GmbH, Munich, Germany.
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany.
- Graduate School of Neuroscience (GSN), Munich, Germany.
| |
Collapse
|
38
|
Zhang D, Cleveland AH, Krimitza E, Han K, Yi C, Stout AL, Zou W, Dorsey JF, Gong Y, Fan Y. Spatial analysis of tissue immunity and vascularity by light sheet fluorescence microscopy. Nat Protoc 2024; 19:1053-1082. [PMID: 38212641 DOI: 10.1038/s41596-023-00941-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Accepted: 10/25/2023] [Indexed: 01/13/2024]
Abstract
The pathogenesis of cancer and cardiovascular diseases is subjected to spatiotemporal regulation by the tissue microenvironment. Multiplex visualization of the microenvironmental components, including immune cells, vasculature and tissue hypoxia, provides critical information underlying the disease progression and therapy resistance, which is often limited by imaging depth and resolution in large-volume tissues. To this end, light sheet fluorescence microscopy, following tissue clarification and immunostaining, may generate three-dimensional high-resolution images at a whole-organ level. Here we provide a detailed description of light sheet fluorescence microscopy imaging analysis of immune cell composition, vascularization, tissue perfusion and hypoxia in mouse normal brains and hearts, as well as brain tumors. We describe a procedure for visualizing tissue vascularization, perfusion and hypoxia with a transgenic vascular labeling system. We provide the procedures for tissue collection, tissue semi-clearing and immunostaining. We further describe standard methods for analyzing tissue immunity and vascularity. We anticipate that this method will facilitate the spatial illustration of structure and function of the tissue microenvironmental components in cancer and cardiovascular diseases. The procedure requires 1-2 weeks and can be performed by users with expertise in general molecular biology.
Collapse
Affiliation(s)
- Duo Zhang
- Department of Radiation Oncology, University of Pennsylvania, Philadelphia, PA, USA
| | - Abigail H Cleveland
- Department of Radiation Oncology, University of Pennsylvania, Philadelphia, PA, USA
| | - Elisavet Krimitza
- Department of Radiation Oncology, University of Pennsylvania, Philadelphia, PA, USA
| | - Katherine Han
- Department of Radiation Oncology, University of Pennsylvania, Philadelphia, PA, USA
| | - Chenlong Yi
- Department of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Andrea L Stout
- Department of Cell and Developmental Biology, University of Pennsylvania, Philadelphia, PA, USA
| | - Wei Zou
- Department of Radiation Oncology, University of Pennsylvania, Philadelphia, PA, USA
| | - Jay F Dorsey
- Department of Radiation Oncology, University of Pennsylvania, Philadelphia, PA, USA
| | - Yanqing Gong
- Department of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| | - Yi Fan
- Department of Radiation Oncology, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
39
|
Skovbjerg G, Fritzen AM, Svendsen CSA, Perens J, Skytte JL, Lund C, Lund J, Madsen MR, Roostalu U, Hecksher-Sørensen J, Clemmensen C. Atlas of exercise-induced brain activation in mice. Mol Metab 2024; 82:101907. [PMID: 38428817 PMCID: PMC10943479 DOI: 10.1016/j.molmet.2024.101907] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Accepted: 02/25/2024] [Indexed: 03/03/2024] Open
Abstract
OBJECTIVES There is significant interest in uncovering the mechanisms through which exercise enhances cognition, memory, and mood, and lowers the risk of neurodegenerative diseases. In this study, we utilize forced treadmill running and distance-matched voluntary wheel running, coupled with light sheet 3D brain imaging and c-Fos immunohistochemistry, to generate a comprehensive atlas of exercise-induced brain activation in mice. METHODS To investigate the effects of exercise on brain activity, we compared whole-brain activation profiles of mice subjected to treadmill running with mice subjected to distance-matched wheel running. Male mice were assigned to one of four groups: a) an acute bout of voluntary wheel running, b) confinement to a cage with a locked running wheel, c) forced treadmill running, or d) placement on an inactive treadmill. Immediately following each exercise or control intervention, blood samples were collected for plasma analysis, and brains were collected for whole-brain c-Fos quantification. RESULTS Our dataset reveals 255 brain regions activated by acute exercise in mice, the majority of which have not previously been linked to exercise. We find a broad response of 140 regulated brain regions that are shared between voluntary wheel running and treadmill running, while 32 brain regions are uniquely regulated by wheel running and 83 brain regions uniquely regulated by treadmill running. In contrast to voluntary wheel running, forced treadmill running triggers activity in brain regions associated with stress, fear, and pain. CONCLUSIONS Our findings demonstrate a significant overlap in neuronal activation signatures between voluntary wheel running and distance-matched forced treadmill running. However, our analysis also reveals notable differences and subtle nuances between these two widely used paradigms. The comprehensive dataset is accessible online at www.neuropedia.dk, with the aim of enabling future research directed towards unraveling the neurobiological response to exercise.
Collapse
Affiliation(s)
- Grethe Skovbjerg
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; Gubra, Hørsholm, Denmark
| | - Andreas Mæchel Fritzen
- August Krogh Section for Molecular Physiology, Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Copenhagen, Denmark; Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Charlotte Sashi Aier Svendsen
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | | | | | - Camilla Lund
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Jens Lund
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | | | | | | | - Christoffer Clemmensen
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
40
|
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
|
41
|
Willekens SMA, Morini F, Mediavilla T, Nilsson E, Orädd G, Hahn M, Chotiwan N, Visa M, Berggren PO, Ilegems E, Överby AK, Ahlgren U, Marcellino D. An MR-based brain template and atlas for optical projection tomography and light sheet fluorescence microscopy in neuroscience. Front Neurosci 2024; 18:1328815. [PMID: 38601090 PMCID: PMC11004350 DOI: 10.3389/fnins.2024.1328815] [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: 10/27/2023] [Accepted: 03/11/2024] [Indexed: 04/12/2024] Open
Abstract
Introduction Optical Projection Tomography (OPT) and light sheet fluorescence microscopy (LSFM) are high resolution optical imaging techniques, ideally suited for ex vivo 3D whole mouse brain imaging. Although they exhibit high specificity for their targets, the anatomical detail provided by tissue autofluorescence remains limited. Methods T1-weighted images were acquired from 19 BABB or DBE cleared brains to create an MR template using serial longitudinal registration. Afterwards, fluorescent OPT and LSFM images were coregistered/normalized to the MR template to create fusion images. Results Volumetric calculations revealed a significant difference between BABB and DBE cleared brains, leading to develop two optimized templates, with associated tissue priors and brain atlas, for BABB (OCUM) and DBE (iOCUM). By creating fusion images, we identified virus infected brain regions, mapped dopamine transporter and translocator protein expression, and traced innervation from the eye along the optic tract to the thalamus and superior colliculus using cholera toxin B. Fusion images allowed for precise anatomical identification of fluorescent signal in the detailed anatomical context provided by MR. Discussion The possibility to anatomically map fluorescent signals on magnetic resonance (MR) images, widely used in clinical and preclinical neuroscience, would greatly benefit applications of optical imaging of mouse brain. These specific MR templates for cleared brains enable a broad range of neuroscientific applications integrating 3D optical brain imaging.
Collapse
Affiliation(s)
- Stefanie M. A. Willekens
- Department of Clinical Microbiology, Umeå University, Umeå, Sweden
- The Laboratory for Molecular Infection Medicine Sweden (MIMS), Umeå University, Umeå, Sweden
- Department of Medical and Translational Biology, Umeå University, Umeå, Sweden
| | - Federico Morini
- Department of Medical and Translational Biology, Umeå University, Umeå, Sweden
| | - Tomas Mediavilla
- Department of Medical and Translational Biology, Umeå University, Umeå, Sweden
| | - Emma Nilsson
- Department of Clinical Microbiology, Umeå University, Umeå, Sweden
- The Laboratory for Molecular Infection Medicine Sweden (MIMS), Umeå University, Umeå, Sweden
| | - Greger Orädd
- Department of Medical and Translational Biology, Umeå University, Umeå, Sweden
| | - Max Hahn
- Department of Medical and Translational Biology, Umeå University, Umeå, Sweden
| | - Nunya Chotiwan
- Department of Clinical Microbiology, Umeå University, Umeå, Sweden
- The Laboratory for Molecular Infection Medicine Sweden (MIMS), Umeå University, Umeå, Sweden
| | - Montse Visa
- The Rolf Luft Research Centre for Diabetes and Endocrinology, Karolinska Institutet, Stockholm, Sweden
| | - Per-Olof Berggren
- The Rolf Luft Research Centre for Diabetes and Endocrinology, Karolinska Institutet, Stockholm, Sweden
| | - Erwin Ilegems
- The Rolf Luft Research Centre for Diabetes and Endocrinology, Karolinska Institutet, Stockholm, Sweden
| | - Anna K. Överby
- Department of Clinical Microbiology, Umeå University, Umeå, Sweden
- The Laboratory for Molecular Infection Medicine Sweden (MIMS), Umeå University, Umeå, Sweden
| | - Ulf Ahlgren
- Department of Medical and Translational Biology, Umeå University, Umeå, Sweden
| | - Daniel Marcellino
- Department of Medical and Translational Biology, Umeå University, Umeå, Sweden
| |
Collapse
|
42
|
Yin J, Liang R, Hou H, Miao Y, Yu L. Light sheet fluorescence microscopy with active optical manipulation. OPTICS LETTERS 2024; 49:1193-1196. [PMID: 38426971 DOI: 10.1364/ol.515280] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Accepted: 01/31/2024] [Indexed: 03/02/2024]
Abstract
We present a light sheet fluorescence microscopy (LSFM) with active optical manipulation by using linear optical tweezers (LOTs). In this method, two coaxially transmitting laser beams of different wavelengths are shaped using cylindrical lenses to form a linear optical trapping perpendicular to the optical axis and an excitation light sheet (LS) parallel to the optical axis, respectively. Multiple large-sized polystyrene fluorescent microspheres are stably captured by LOTs, and their rotation angles around specific rotation axes are precisely controlled. During a sample rotation, the stationary excitation LS scans the sample to obtain fluorescence sectioning images of the sample at different angles.
Collapse
|
43
|
Solé-Guardia G, Luijten M, Geenen B, Claassen JAHR, Litjens G, de Leeuw FE, Wiesmann M, Kiliaan AJ. Three-dimensional identification of microvascular pathology and neurovascular inflammation in severe white matter hyperintensity: a case report. Sci Rep 2024; 14:5004. [PMID: 38424226 PMCID: PMC10904845 DOI: 10.1038/s41598-024-55733-y] [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: 09/18/2023] [Accepted: 02/27/2024] [Indexed: 03/02/2024] Open
Abstract
White matter hyperintensities (WMH) are the most prevalent markers of cerebral small vessel disease (SVD), which is the major vascular risk factor for dementia. Microvascular pathology and neuroinflammation are suggested to drive the transition from normal-appearing white matter (NAWM) to WMH, particularly in individuals with hypertension. However, current imaging techniques cannot capture ongoing NAWM changes. The transition from NAWM into WMH is a continuous process, yet white matter lesions are often examined dichotomously, which may explain their underlying heterogeneity. Therefore, we examined microvascular and neurovascular inflammation pathology in NAWM and severe WMH three-dimensionally, along with gradual magnetic resonance imaging (MRI) fluid-attenuated inversion recovery (FLAIR) signal (sub-)segmentation. In WMH, the vascular network exhibited reduced length and complexity compared to NAWM. Neuroinflammation was more severe in WMH. Vascular inflammation was more pronounced in NAWM, suggesting its potential significance in converting NAWM into WMH. Moreover, the (sub-)segmentation of FLAIR signal displayed varying degrees of vascular pathology, particularly within WMH regions. These findings highlight the intricate interplay between microvascular pathology and neuroinflammation in the transition from NAWM to WMH. Further examination of neurovascular inflammation across MRI-visible alterations could aid deepening our understanding on WMH conversion, and therewith how to improve the prognosis of SVD.
Collapse
Affiliation(s)
- Gemma Solé-Guardia
- Department of Medical Imaging, Anatomy, Donders Institute for Brain, Cognition & Behavior, Preclinical Imaging Center PRIME, Radboud Alzheimer Center, Radboud university medical center, 6525 EZ, Nijmegen, PO Box 9101, The Netherlands
| | - Matthijs Luijten
- Department of Medical Imaging, Anatomy, Donders Institute for Brain, Cognition & Behavior, Preclinical Imaging Center PRIME, Radboud Alzheimer Center, Radboud university medical center, 6525 EZ, Nijmegen, PO Box 9101, The Netherlands
| | - Bram Geenen
- Department of Medical Imaging, Anatomy, Donders Institute for Brain, Cognition & Behavior, Preclinical Imaging Center PRIME, Radboud Alzheimer Center, Radboud university medical center, 6525 EZ, Nijmegen, PO Box 9101, The Netherlands
| | - Jurgen A H R Claassen
- Department of Geriatrics, Donders Institute for Brain, Cognition & Behavior, Radboud Alzheimer Center, Radboud university medical center, Nijmegen, The Netherlands
- Department of Cardiovascular Sciences, University of Leicester, Leicester, UK
| | - Geert Litjens
- Department of Pathology, Radboud university medical center, Nijmegen, The Netherlands
- Computational Pathology Group, Research Institute for Medical Innovation, Radboud university medical center, Nijmegen, The Netherlands
| | - Frank-Erik de Leeuw
- Department of Neurology, Donders Institute for Brain, Cognition & Behavior, Radboud university medical center, Nijmegen, The Netherlands
| | - Maximilian Wiesmann
- Department of Medical Imaging, Anatomy, Donders Institute for Brain, Cognition & Behavior, Preclinical Imaging Center PRIME, Radboud Alzheimer Center, Radboud university medical center, 6525 EZ, Nijmegen, PO Box 9101, The Netherlands
| | - Amanda J Kiliaan
- Department of Medical Imaging, Anatomy, Donders Institute for Brain, Cognition & Behavior, Preclinical Imaging Center PRIME, Radboud Alzheimer Center, Radboud university medical center, 6525 EZ, Nijmegen, PO Box 9101, The Netherlands.
| |
Collapse
|
44
|
Sakamoto DM, Tamura I, Yi B, Hasegawa S, Saito Y, Yamada N, Takakusagi Y, Kubota SI, Kobayashi M, Harada H, Hanaoka K, Taki M, Nangaku M, Tainaka K, Sando S. Whole-Body and Whole-Organ 3D Imaging of Hypoxia Using an Activatable Covalent Fluorescent Probe Compatible with Tissue Clearing. ACS NANO 2024; 18:5167-5179. [PMID: 38301048 DOI: 10.1021/acsnano.3c12716] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/03/2024]
Abstract
Elucidation of biological phenomena requires imaging of microenvironments in vivo. Although the seamless visualization of in vivo hypoxia from the level of whole-body to single-cell has great potential to discover unknown phenomena in biological and medical fields, no methodology for achieving it has been established thus far. Here, we report the whole-body and whole-organ imaging of hypoxia, an important microenvironment, at single-cell resolution using activatable covalent fluorescent probes compatible with tissue clearing. We initially focused on overcoming the incompatibility of fluorescent dyes and refractive index matching solutions (RIMSs), which has greatly hindered the development of fluorescent molecular probes in the field of tissue clearing. The fluorescent dyes compatible with RIMS were then incorporated into the development of activatable covalent fluorescent probes for hypoxia. We combined the probes with tissue clearing, achieving comprehensive single-cell-resolution imaging of hypoxia in a whole mouse body and whole organs.
Collapse
Affiliation(s)
- Daichi M Sakamoto
- Department of Chemistry and Biotechnology, Graduate School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan
| | - Iori Tamura
- Department of Chemistry and Biotechnology, Graduate School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan
| | - Bo Yi
- Department of Chemistry and Biotechnology, Graduate School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan
| | - Sho Hasegawa
- Division of Nephrology and Endocrinology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8654, Japan
| | - Yutaro Saito
- Department of Chemistry and Biotechnology, Graduate School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan
| | - Naoki Yamada
- Department of Chemistry and Biotechnology, Graduate School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan
| | - Yoichi Takakusagi
- Quantum Hyperpolarized MRI Team, Institute for Quantum Life Science, National Institutes for Quantum Science and Technology, 4-9-1 Anagawa, Inage, Chiba-city 263-8555, Japan
- Institute for Quantum Medical Science, National Institutes for Quantum Science and Technology, 4-9-1 Anagawa, Inage, Chiba-city 263-8555, Japan
| | - Shimpei I Kubota
- Division of Molecular Psychoimmunology, Institute for Genetic Medicine, Graduate School of Medicine, Hokkaido University, Kita-15, Nishi-7, Kita-ku, Sapporo, Hokkaido 060-0815, Japan
| | - Minoru Kobayashi
- Laboratory of Cancer Cell Biology, Graduate School of Biostudies, Kyoto University, Yoshida Konoe-cho, Sakyo-ku, Kyoto 606-8501, Japan
- Department of Genome Dynamics, Radiation Biology Center, Graduate School of Biostudies, Kyoto University, Yoshida Konoe-cho, Sakyo-ku, Kyoto 606-8501, Japan
| | - Hiroshi Harada
- Laboratory of Cancer Cell Biology, Graduate School of Biostudies, Kyoto University, Yoshida Konoe-cho, Sakyo-ku, Kyoto 606-8501, Japan
- Department of Genome Dynamics, Radiation Biology Center, Graduate School of Biostudies, Kyoto University, Yoshida Konoe-cho, Sakyo-ku, Kyoto 606-8501, Japan
| | - Kenjiro Hanaoka
- Division of Analytical Chemistry for Drug Discovery, Graduate School of Pharmaceutical Sciences, Keio University, 1-5-30 Shibakoen, Minato-ku, Tokyo 105-8512, Japan
| | - Masayasu Taki
- Institute of Transformative Bio-Molecules, Nagoya University, Furo, Chikusa, Nagoya 464-8601, Japan
| | - Masaomi Nangaku
- Division of Nephrology and Endocrinology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8654, Japan
| | - Kazuki Tainaka
- Department of System Pathology for Neurological Disorders, Brain Research Institute, Niigata University, 1-757 Asahimachidori, Chuo-ku, Niigata 951-8585, Japan
- Gftd DeSci, Gftd DAO, Nishikawa Building, 20 Kikuicho, Shinjuku-ku, Tokyo 162-0044, Japan
| | - Shinsuke Sando
- Department of Chemistry and Biotechnology, Graduate School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan
- Department of Bioengineering, Graduate School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan
| |
Collapse
|
45
|
Gómez-de-Mariscal E, Del Rosario M, Pylvänäinen JW, Jacquemet G, Henriques R. Harnessing artificial intelligence to reduce phototoxicity in live imaging. J Cell Sci 2024; 137:jcs261545. [PMID: 38324353 PMCID: PMC10912813 DOI: 10.1242/jcs.261545] [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] [Indexed: 02/08/2024] Open
Abstract
Fluorescence microscopy is essential for studying living cells, tissues and organisms. However, the fluorescent light that switches on fluorescent molecules also harms the samples, jeopardizing the validity of results - particularly in techniques such as super-resolution microscopy, which demands extended illumination. Artificial intelligence (AI)-enabled software capable of denoising, image restoration, temporal interpolation or cross-modal style transfer has great potential to rescue live imaging data and limit photodamage. Yet we believe the focus should be on maintaining light-induced damage at levels that preserve natural cell behaviour. In this Opinion piece, we argue that a shift in role for AIs is needed - AI should be used to extract rich insights from gentle imaging rather than recover compromised data from harsh illumination. Although AI can enhance imaging, our ultimate goal should be to uncover biological truths, not just retrieve data. It is essential to prioritize minimizing photodamage over merely pushing technical limits. Our approach is aimed towards gentle acquisition and observation of undisturbed living systems, aligning with the essence of live-cell fluorescence microscopy.
Collapse
Affiliation(s)
| | | | - Joanna W. Pylvänäinen
- Faculty of Science and Engineering, Cell Biology, Åbo Akademi University, Turku 20500, Finland
| | - Guillaume Jacquemet
- Faculty of Science and Engineering, Cell Biology, Åbo Akademi University, Turku 20500, Finland
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku 20520, Finland
- Turku Bioimaging, University of Turku and Åbo Akademi University, Turku 20520, Finland
- InFLAMES Research Flagship Center, Åbo Akademi University, Turku 20100, Finland
| | - Ricardo Henriques
- Instituto Gulbenkian de Ciência, Oeiras 2780-156, Portugal
- UCL Laboratory for Molecular Cell Biology, University College London, London WC1E 6BT, UK
| |
Collapse
|
46
|
Yamada H, Makino SI, Okunaga I, Miyake T, Yamamoto-Nonaka K, Oliva Trejo JA, Tominaga T, Empitu MA, Kadariswantiningsih IN, Kerever A, Komiya A, Ichikawa T, Arikawa-Hirasawa E, Yanagita M, Asanuma K. Beyond 2D: A scalable and highly sensitive method for a comprehensive 3D analysis of kidney biopsy tissue. PNAS NEXUS 2024; 3:pgad433. [PMID: 38193136 PMCID: PMC10772983 DOI: 10.1093/pnasnexus/pgad433] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Accepted: 11/06/2023] [Indexed: 01/10/2024]
Abstract
The spatial organization of various cell populations is critical for the major physiological and pathological processes in the kidneys. Most evaluation of these processes typically comes from a conventional 2D tissue cross-section, visualizing a limited amount of cell organization. Therefore, the 2D analysis of kidney biopsy introduces selection bias. The 2D analysis potentially omits key pathological findings outside a 1- to 10-μm thin-sectioned area and lacks information on tissue organization, especially in a particular irregular structure such as crescentic glomeruli. In this study, we introduce an easy-to-use and scalable method for obtaining high-quality images of molecules of interest in a large tissue volume, enabling a comprehensive evaluation of the 3D organization and cellular composition of kidney tissue, especially the glomerular structure. We show that CUBIC and ScaleS clearing protocols could allow a 3D analysis of the kidney tissues in human and animal models of kidney disease. We also demonstrate that the paraffin-embedded human biopsy specimens previously examined via 2D evaluation could be applicable to 3D analysis, showing a potential utilization of this method in kidney biopsy tissue collected in the past. In summary, the 3D analysis of kidney biopsy provides a more comprehensive analysis and a minimized selection bias than 2D tissue analysis. Additionally, this method enables a quantitative evaluation of particular kidney structures and their surrounding tissues, with the potential utilization from basic science investigation to applied diagnostics in nephrology.
Collapse
Affiliation(s)
- Hiroyuki Yamada
- Department of Nephrology, Graduate School of Medicine, Chiba University, Chiba 260-8677, Japan
- The Laboratory for Kidney Research (TMK Project), Medical Innovation Center, Graduate School of Medicine, Kyoto University, Kyoto 606-8397, Japan
- Department of Nephrology, Graduate School of Medicine, Kyoto University, Kyoto 606-8507, Japan
- Department of Primary Care and Emergency, Graduate School of Medicine, Kyoto University, Kyoto 606-8507, Japan
| | - Shin-ichi Makino
- Department of Nephrology, Graduate School of Medicine, Chiba University, Chiba 260-8677, Japan
- The Laboratory for Kidney Research (TMK Project), Medical Innovation Center, Graduate School of Medicine, Kyoto University, Kyoto 606-8397, Japan
- Department of Nephrology, Graduate School of Medicine, Kyoto University, Kyoto 606-8507, Japan
| | - Issei Okunaga
- Department of Nephrology, Graduate School of Medicine, Chiba University, Chiba 260-8677, Japan
| | - Takafumi Miyake
- The Laboratory for Kidney Research (TMK Project), Medical Innovation Center, Graduate School of Medicine, Kyoto University, Kyoto 606-8397, Japan
- Department of Nephrology, Graduate School of Medicine, Kyoto University, Kyoto 606-8507, Japan
| | - Kanae Yamamoto-Nonaka
- The Laboratory for Kidney Research (TMK Project), Medical Innovation Center, Graduate School of Medicine, Kyoto University, Kyoto 606-8397, Japan
- Department of Nephrology, Graduate School of Medicine, Kyoto University, Kyoto 606-8507, Japan
| | - Juan Alejandro Oliva Trejo
- The Laboratory for Kidney Research (TMK Project), Medical Innovation Center, Graduate School of Medicine, Kyoto University, Kyoto 606-8397, Japan
| | - Takahiro Tominaga
- Department of Nephrology, Graduate School of Medicine, Chiba University, Chiba 260-8677, Japan
| | - Maulana A Empitu
- Department of Nephrology, Graduate School of Medicine, Chiba University, Chiba 260-8677, Japan
| | | | - Aurelien Kerever
- Research Institute for Diseases of Old Age, Graduate School of Medicine, Juntendo University, Tokyo 113-8421, Japan
| | - Akira Komiya
- Department of Urology, Graduate School of Medicine, Chiba University, Chiba 260-8677, Japan
| | - Tomohiko Ichikawa
- Department of Urology, Graduate School of Medicine, Chiba University, Chiba 260-8677, Japan
| | - Eri Arikawa-Hirasawa
- Research Institute for Diseases of Old Age, Graduate School of Medicine, Juntendo University, Tokyo 113-8421, Japan
| | - Motoko Yanagita
- The Laboratory for Kidney Research (TMK Project), Medical Innovation Center, Graduate School of Medicine, Kyoto University, Kyoto 606-8397, Japan
- Department of Nephrology, Graduate School of Medicine, Kyoto University, Kyoto 606-8507, Japan
- Institute for the Advanced Study of Human Biology (ASHBi), Kyoto University, Kyoto 606-8303, Japan
| | - Katsuhiko Asanuma
- Department of Nephrology, Graduate School of Medicine, Chiba University, Chiba 260-8677, Japan
- The Laboratory for Kidney Research (TMK Project), Medical Innovation Center, Graduate School of Medicine, Kyoto University, Kyoto 606-8397, Japan
- Department of Nephrology, Graduate School of Medicine, Kyoto University, Kyoto 606-8507, Japan
| |
Collapse
|
47
|
Murakami E, Nakamori M, Nakatani K, Shibata T, Tainaka K. Intracerebral Distribution of CAG Repeat-Binding Small Molecule Visualized by Whole-Brain Imaging. Bioconjug Chem 2023; 34:2187-2193. [PMID: 37948852 DOI: 10.1021/acs.bioconjchem.3c00470] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2023]
Abstract
Understanding the pharmacokinetics of drug candidates of interest in the brain and evaluating drug delivery to the brain are important for developing drugs targeting the brain. Previously, we demonstrated that a CAG repeat-binding small molecule, naphthyridine-azaquinolone (NA), resulted in repeat contraction in mouse models of dentatorubral-pallidoluysian atrophy and Huntington's disease caused by aberrant expansion of CAG repeats. However, the intracerebral distribution and drug deliverability of NA remain unclear. Here, we report three-dimensional whole-brain imaging of an externally administered small molecule using tissue clearing and light sheet fluorescence microscopy (LSFM). We designed and synthesized an Alexa594-labeled NA derivative with a primary amine for whole-brain imaging (NA-Alexa594-NH2), revealing the intracerebral distribution of NA-Alexa594-NH2 after intraparenchymal and intracerebroventricular administrations by whole-brain imaging combined with tissue clearing and LSFM. We also clarified that intranasally administered NA-Alexa594-NH2 was delivered into the brain via multiple nose-to-brain pathways by tracking the time-dependent change in the intracerebral distribution. Whole-brain imaging of small molecules by tissue clearing and LSFM is useful for elucidating not only the intracerebral distribution but also the drug delivery pathways into the brain.
Collapse
Affiliation(s)
- Eitaro Murakami
- Department of Regulatory Bioorganic Chemistry, SANKEN (The Institute of Scientific and Industrial Research), Osaka University, Osaka 567-0047, Japan
| | - Masayuki Nakamori
- Department of Neurology, Osaka University Graduate School of Medicine, Osaka 565-0871, Japan
| | - Kazuhiko Nakatani
- Department of Regulatory Bioorganic Chemistry, SANKEN (The Institute of Scientific and Industrial Research), Osaka University, Osaka 567-0047, Japan
| | - Tomonori Shibata
- Department of Regulatory Bioorganic Chemistry, SANKEN (The Institute of Scientific and Industrial Research), Osaka University, Osaka 567-0047, Japan
| | - Kazuki Tainaka
- Department of System Pathology for Neurological Disorders, Center for Bioresources, Brain Research Institute, Niigata University, Niigata 951-8585, Japan
| |
Collapse
|
48
|
Kang YG, Park K, Hyeon MG, Yang TD, Choi Y. Three-dimensional imaging in reflection phase microscopy with minimal axial scanning. OPTICS EXPRESS 2023; 31:44741-44753. [PMID: 38178536 DOI: 10.1364/oe.510519] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Accepted: 11/29/2023] [Indexed: 01/06/2024]
Abstract
Reflection phase microscopy is a valuable tool for acquiring three-dimensional (3D) images of objects due to its capability of optical sectioning. The conventional method of constructing a 3D map is capturing 2D images at each depth with a mechanical scanning finer than the optical sectioning. This not only compromises sample stability but also slows down the acquisition process, imposing limitations on its practical applications. In this study, we utilized a reflection phase microscope to acquire 2D images at depth locations significantly spaced apart, far beyond the range of optical sectioning. By employing a numerical propagation, we successfully filled the information gap between the acquisition layers, and then constructed complete 3D maps of objects with substantially reduced number of axial scans. Our experimental results also demonstrated the effectiveness of this approach in enhancing imaging speed while maintaining the accuracy of the reconstructed 3D structures. This technique has the potential to improve the applicability of reflection phase microscopy in diverse fields such as bioimaging and material science.
Collapse
|
49
|
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: 1.0] [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
|
50
|
Lee SH, Son HJ. Second Wave, Late-Stage Neuroinflammation in Cleared Brains of Aged 5xFAD Alzheimer's Mice Detected by Macrolaser Light Sheet Microscopy Imaging. Int J Mol Sci 2023; 24:17058. [PMID: 38069392 PMCID: PMC10707588 DOI: 10.3390/ijms242317058] [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: 11/12/2023] [Revised: 11/27/2023] [Accepted: 11/29/2023] [Indexed: 12/18/2023] Open
Abstract
This study leverages the innovative imaging capabilities of macrolaser light-sheet microscopy to elucidate the 3D spatial visualization of AD-associated neuropathologic networks in the transparent brains of 44-week-old 5xFAD mice. Brain samples from ten AD and seven control mice were prepared through a hydrophilic tissue-clearing pipeline and immunostained with thioflavin S (β-amyloid), anti-CD11b antibody (microglia), and anti-ACSA-2 antibody (astrocytes). The 5xFAD group exhibited significantly higher average total surface volumes of β-amyloid accumulation than the control group (AD, 898,634,368 µm3 [383,355,488-1,324,986,752]; control, 33,320,178 µm3 [11,156,785-65,390,988], p = 0.0006). Within the AD group, there was significant interindividual and interindividual variability concerning the number and surface volume of individual amyloid particles throughout the entire brain. In the context of neuroinflammation, the 5xFAD group showed significantly higher average total surface volumes of anti-ACSA-2-labeled astrocytes (AD, 59,064,360 µm3 [27,815,500-222,619,280]; control, 20,272,722 µm3 [9,317,288-27,223,352], p = 0.0047) and anti-CD11b labeled microglia (AD, 51,210,100 µm3 [15,309,118-135,532,144]; control, 23,461,593 µm3 [14,499,170-27,924,110], p = 0.0162) than the control group. Contrary to the long-standing finding that early-stage neuroinflammation precedes the subsequent later-stage of neurodegeneration, our data reveal that the second wave, late-stage active neuroinflammation persists in the aged AD brains, even as they continue to show signs of ongoing neurodegeneration and significant amyloid accumulation.
Collapse
Affiliation(s)
- Suk Hyun Lee
- Department of Radiology, Hallym University Kangnam Sacred Heart Hospital, Hallym University College of Medicine, Seoul 07441, Republic of Korea
| | - Hye Joo Son
- Department of Nuclear Medicine, Dankook University Medical Center, Dankook University College of Medicine, Cheonan 31116, Republic of Korea
| |
Collapse
|