1
|
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 2024:115470. [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] [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-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
| | - 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
|
2
|
Kitamura E, Imai N. Molecular and Cellular Neurobiology of Spreading Depolarization/Depression and Migraine: A Narrative Review. Int J Mol Sci 2024; 25:11163. [PMID: 39456943 PMCID: PMC11508361 DOI: 10.3390/ijms252011163] [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: 08/26/2024] [Revised: 10/11/2024] [Accepted: 10/16/2024] [Indexed: 10/28/2024] Open
Abstract
Migraine is a prevalent neurological disorder, particularly among individuals aged 20-50 years, with significant social and economic impacts. Despite its high prevalence, the pathogenesis of migraine remains unclear. In this review, we provide a comprehensive overview of cortical spreading depolarization/depression (CSD) and its close association with migraine aura, focusing on its role in understanding migraine pathogenesis and therapeutic interventions. We discuss historical studies that have demonstrated the role of CSD in the visual phenomenon of migraine aura, along with modern imaging techniques confirming its propagation across the occipital cortex. Animal studies are examined to indicate that CSD is not exclusive to migraines; it also occurs in other neurological conditions. At the cellular level, we review how CSD is characterized by ionic changes and excitotoxicity, leading to neuronal and glial responses. We explore how CSD activates the trigeminal nervous system and upregulates the expression of calcitonin gene-related peptides (CGRP), thereby contributing to migraine pain. Factors such as genetics, obesity, and environmental conditions that influence the CSD threshold are discussed, suggesting potential therapeutic targets. Current treatments for migraine, including prophylactic agents and CGRP-targeting drugs, are evaluated in the context of their expected effects on suppressing CSD activity. Additionally, we highlight emerging therapies such as intranasal insulin-like growth factor 1 and vagus nerve stimulation, which have shown promise in reducing CSD susceptibility and frequency. By elucidating the molecular and cellular mechanisms of CSD, this review aims to enhance the understanding of migraine pathogenesis and support the development of targeted therapeutic strategies.
Collapse
Affiliation(s)
- Eiji Kitamura
- Department of Neurology, Kitasato University School of Medicine, Sagamihara 252-0329, Japan;
| | - Noboru Imai
- Department of Neurology and Headache Center, Japanese Red Cross Shizuoka Hospital, Shizuoka 420-0853, Japan
| |
Collapse
|
3
|
Kang JH, Yang JK, Cho KH, Lee OH, Kwon H, Kim SY, Kim S, Ko YT. Intracalvariosseous administration of donepezil microspheres protects against cognitive impairment by virtue of long-lasting brain exposure in mice. Theranostics 2024; 14:6708-6725. [PMID: 39479440 PMCID: PMC11519799 DOI: 10.7150/thno.100986] [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: 07/15/2024] [Accepted: 09/30/2024] [Indexed: 11/02/2024] Open
Abstract
Rationale: Recent studies have demonstrated the direct connections between the skull bone marrow, meninges, and brain. In an effort to explore these connections for the purpose of brain drug delivery, we previously proposed the direct application of CNS drugs into the diploic space between the outer and inner cortex of the skull, namely, intracalvariosseous administration (ICO). It was successfully demonstrated that small molecular to large colloidal drugs can readily reach the brain after ICO in mice and rabbits. Here, we report that a single ICO of donepezil microspheres protects cognitive impairment in Alzheimer mouse models over a month-long period. Methods: Donepezil-loaded long-acting microspheres (DPZ@LAM) were prepared with biodegradable poly(DL-lactide-co-glycolide) (PLGA). Pharmacokinetic study and behavioral test were performed to determine the brain exposure and therapeutic effects after ICO of DPZ@LAM in scopolamine-induced memory-deficient mice. Results: DPZ@LAM were capable of a month-long and precisely controlled drug release. After a single ICO of DPZ@LAM, DPZ concentration in brain sustained above the effective therapeutic levels for four weeks. The long-lasting brain exposure also led to significantly recovered cognitive function in scopolamine-induced memory-deficient mice, along with decreased acetylcholinesterase activity and increased brain-derived neurotrophic factor. Conclusions: ICO allows for BBB-bypassing brain drug delivery through the direct connection between the skull bone marrow and brain, providing an alternative approach for the treatment of neurodegenerative diseases with otherwise BBB impermeable CNS drugs.
Collapse
Affiliation(s)
- Ji Hee Kang
- College of Pharmacy and Gachon Institute of Pharmaceutical Sciences, Gachon University, Incheon, 21936, Republic of Korea
| | - Jin-Kyoung Yang
- Department of Chemical Engineering, Dong-Eui University, Busan, 47340, Republic of Korea
| | - Kyo Hee Cho
- College of Pharmacy and Gachon Institute of Pharmaceutical Sciences, Gachon University, Incheon, 21936, Republic of Korea
| | - O Hyun Lee
- College of Pharmacy and Gachon Institute of Pharmaceutical Sciences, Gachon University, Incheon, 21936, Republic of Korea
| | - Hayoon Kwon
- Chemical & Biological Integrative Research Center, Korea Institute of Science and Technology, Seoul, 02792, Republic of Korea
| | - Sun Yeou Kim
- College of Pharmacy and Gachon Institute of Pharmaceutical Sciences, Gachon University, Incheon, 21936, Republic of Korea
| | - Sehoon Kim
- Chemical & Biological Integrative Research Center, Korea Institute of Science and Technology, Seoul, 02792, Republic of Korea
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, 02841, Republic of Korea
| | - Young Tag Ko
- College of Pharmacy and Gachon Institute of Pharmaceutical Sciences, Gachon University, Incheon, 21936, Republic of Korea
| |
Collapse
|
4
|
Roostalu U, Hansen HH, Hecksher-Sørensen J. 3D light-sheet fluorescence microscopy in preclinical and clinical drug discovery. Drug Discov Today 2024; 29:104196. [PMID: 39368696 DOI: 10.1016/j.drudis.2024.104196] [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: 06/19/2024] [Revised: 09/10/2024] [Accepted: 09/30/2024] [Indexed: 10/07/2024]
Abstract
Light-sheet fluorescence microscopy (LSFM) combined with tissue clearing has emerged as a powerful technology in drug discovery. LSFM is applicable to a variety of samples, from rodent organs to clinical tissue biopsies, and has been used for characterizing drug targets in tissues, demonstrating the biodistribution of pharmaceuticals and determining their efficacy and mode of action. LSFM is scalable to high-throughput analysis and provides resolution down to the single cell level. In this review, we describe the advantages of implementing LSFM into the drug discovery pipeline and highlight recent advances in this field.
Collapse
|
5
|
Kuhn AM, Bosis KE, Wohleb ES. Neuroimmunomodulation: The History of Science in Psychoneuroimmunology. Neuroimmunomodulation 2024; 31:211-229. [PMID: 39369707 DOI: 10.1159/000541592] [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: 05/15/2024] [Accepted: 09/23/2024] [Indexed: 10/08/2024] Open
Abstract
BACKGROUND From the original studies investigating the effects of adrenal gland secretion to modern high-throughput multidimensional analyses, stress research has been a topic of scientific interest spanning just over a century. SUMMARY The objective of this review was to provide historical context for influential discoveries, surprising findings, and preclinical models in stress-related neuroimmune research. Furthermore, we summarize this work and present a current understanding of the stress pathways and their effects on the immune system and behavior. We focus on recent work demonstrating stress-induced immune changes within the brain and highlight studies investigating stress effects on microglia. Lastly, we conclude with potential areas for future investigation concerning microglia heterogeneity, bone marrow niches, and sex differences. KEY MESSAGES Stress is a phenomenon that ties together not only the central and peripheral nervous system, but the immune system as well. The cumulative effects of stress can enhance or suppress immune function, based on the intensity and duration of the stressor. These stress-induced immune alterations are associated with neurobiological changes, including structural remodeling of neurons and decreased neurogenesis, and these contribute to the development of behavioral and cognitive deficits. As such, research in this field has revealed important insights into neuroimmune communication as well as molecular and cellular mediators of complex behaviors relevant to psychiatric disorders.
Collapse
Affiliation(s)
- Alexander M Kuhn
- Department of Pharmacology, Physiology, and Neurobiology, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Kelly E Bosis
- Neuroscience Graduate Program, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Eric S Wohleb
- Department of Pharmacology, Physiology, and Neurobiology, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| |
Collapse
|
6
|
Zanluqui NG, McGavern DB. Why do central nervous system barriers host a diverse immune landscape? Trends Immunol 2024; 45:738-749. [PMID: 39299888 PMCID: PMC11471389 DOI: 10.1016/j.it.2024.08.009] [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: 08/14/2024] [Revised: 08/27/2024] [Accepted: 08/27/2024] [Indexed: 09/22/2024]
Abstract
The meninges in vertebrates comprise three layers (dura, arachnoid, pia mater), representing an important barrier surrounding and protecting the central nervous system (CNS). The most exterior CNS barrier, the dura mater, is unique because it resembles a peripheral tissue. It hosts a rich immune landscape, lymphatic vessels, and fenestrated vasculature, allowing microbes and other threats from the blood to extravasate into the meninges, potentially reaching the underlying CNS. The highly specialized large venous drainage system in the dura is especially susceptible to infection. Here, we explore specializations in the CNS barrier system from an anatomical and immunological perspective and posit that the dura mater evolved an elaborate innate and adaptive immune system in specific locations within it to protect underlying CNS tissue against invading pathogens.
Collapse
Affiliation(s)
- Nagela G Zanluqui
- Viral Immunology and Intravital Imaging Section, National Institute of Neurological Disorders and Stroke (NINDS), National Institute of Health (NIH), Bethesda, MD, USA
| | - Dorian B McGavern
- Viral Immunology and Intravital Imaging Section, National Institute of Neurological Disorders and Stroke (NINDS), National Institute of Health (NIH), Bethesda, MD, USA.
| |
Collapse
|
7
|
Wang Z, Xiao X, Zhou Z, Chen Y, Xia T, Sheng X, Han Y, Gong W, Si K. FLUID: a fluorescence-friendly lipid-compatible ultrafast clearing method. BIOMEDICAL OPTICS EXPRESS 2024; 15:5609-5624. [PMID: 39421767 PMCID: PMC11482171 DOI: 10.1364/boe.533072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Revised: 08/14/2024] [Accepted: 08/14/2024] [Indexed: 10/19/2024]
Abstract
Many clearing methods achieve high transparency by removing lipid components from tissues, which damages microstructure and limits their application in lipid research. As for methods which preserve lipid, it is difficult to balance transparency, fluorescence preservation and clearing speed. In this study, we propose a rapid water-based clearing method that is fluorescence-friendly and preserves lipid components. FLUID allows for preservation of endogenous fluorescence over 60 days. It shows negligible tissue distortion and is compatible with various types of fluorescent labeling and tissue staining methods. High quality imaging of human brain tissue and compatibility with pathological staining demonstrated the potential of our method for three-dimensional (3D) biopsy and clinical pathological diagnosis.
Collapse
Affiliation(s)
- Zizheng Wang
- Department of Psychiatry of the First Affiliated Hospital, Zhejiang University School of Medicine, College of Optical Science and Engineering, Zhejiang University, Hangzhou 310058, China
| | - Xiao Xiao
- Department of Psychiatry of the First Affiliated Hospital, Zhejiang University School of Medicine, College of Optical Science and Engineering, Zhejiang University, Hangzhou 310058, China
- Nanhu Brain-computer Interface Institute, Hangzhou 311100, China
| | - Ziwen Zhou
- Department of Psychiatry of the First Affiliated Hospital, Zhejiang University School of Medicine, College of Optical Science and Engineering, Zhejiang University, Hangzhou 310058, China
| | - Yunyin Chen
- Liangzhu Laboratory, MOE Frontier Science Center for Brain Science and Brain-machine Integration, State Key Laboratory of Brain-machine Intelligence, Zhejiang University, 1369 West Wenyi Road, Hangzhou 311121, China
| | - Tianqi Xia
- Liangzhu Laboratory, MOE Frontier Science Center for Brain Science and Brain-machine Integration, State Key Laboratory of Brain-machine Intelligence, Zhejiang University, 1369 West Wenyi Road, Hangzhou 311121, China
| | - Xiangyi Sheng
- NHC and CAMS Key Laboratory of Medical Neurobiology, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou 310058, China
| | - Yiping Han
- NHC and CAMS Key Laboratory of Medical Neurobiology, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou 310058, China
| | - Wei Gong
- Nanhu Brain-computer Interface Institute, Hangzhou 311100, China
- NHC and CAMS Key Laboratory of Medical Neurobiology, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou 310058, China
- Lingang Laboratory, Shanghai 200031, China
| | - Ke Si
- Department of Psychiatry of the First Affiliated Hospital, Zhejiang University School of Medicine, College of Optical Science and Engineering, Zhejiang University, Hangzhou 310058, China
- Liangzhu Laboratory, MOE Frontier Science Center for Brain Science and Brain-machine Integration, State Key Laboratory of Brain-machine Intelligence, Zhejiang University, 1369 West Wenyi Road, Hangzhou 311121, China
- NHC and CAMS Key Laboratory of Medical Neurobiology, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou 310058, China
| |
Collapse
|
8
|
Betsholtz C, Engelhardt B, Koh GY, McDonald DM, Proulx ST, Siegenthaler J. Advances and controversies in meningeal biology. Nat Neurosci 2024:10.1038/s41593-024-01701-8. [PMID: 39333784 DOI: 10.1038/s41593-024-01701-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Accepted: 06/12/2024] [Indexed: 09/30/2024]
Abstract
The dura, arachnoid and pia mater, as the constituent layers of the meninges, along with cerebrospinal fluid in the subarachnoid space and ventricles, are essential protectors of the brain and spinal cord. Complemented by immune cells, blood vessels, lymphatic vessels and nerves, these connective tissue layers have held many secrets that have only recently begun to be revealed. Each meningeal layer is now known to have molecularly distinct types of fibroblasts. Cerebrospinal fluid clearance through peripheral lymphatics and lymph nodes is well documented, but its routes and flow dynamics are debated. Advances made in meningeal immune functions are also debated. This Review considers the cellular and molecular structure and function of the dura, arachnoid and pia mater in the context of conventional views, recent progress, and what is uncertain or unknown. The hallmarks of meningeal pathophysiology are identified toward developing a more complete understanding of the meninges in health and disease.
Collapse
Affiliation(s)
- Christer Betsholtz
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Uppsala, Sweden and Department of Medicine-Huddinge, Karolinska Institutet, Huddinge, Sweden
| | | | - Gou Young Koh
- Center for Vascular Research, Institute for Basic Science and Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon, Republic of Korea
| | - Donald M McDonald
- Department of Anatomy, Cardiovascular Research Institute, and UCSF Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA, USA
| | - Steven T Proulx
- Theodor Kocher Institute, University of Bern, Bern, Switzerland
| | - Julie Siegenthaler
- Department of Pediatrics, Section of Developmental Biology, University of Colorado, Anschutz Medical Campus Aurora, Colorado, CO, USA.
| |
Collapse
|
9
|
Zhu J, Liu X, Liu Z, Deng Y, Xu J, Liu K, Zhang R, Meng X, Fei P, Yu T, Zhu D. SOLID: minimizing tissue distortion for brain-wide profiling of diverse architectures. Nat Commun 2024; 15:8303. [PMID: 39333107 PMCID: PMC11436996 DOI: 10.1038/s41467-024-52560-7] [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: 01/15/2024] [Accepted: 09/12/2024] [Indexed: 09/29/2024] Open
Abstract
Brain-wide profiling of diverse biological components is fundamental for understanding complex brain pathology. Despite the availability in whole-brain imaging, it is still challenging to conduct multiplexed, brain-wide analysis with current tissue clearing techniques. Here, we propose SOLID, a hydrophobic tissue clearing method that can minimize tissue distortion while offering impressive clearing performance. SOLID achieves high-quality imaging of multi-color labeled mouse brain, and the acquired datasets can be effectively registered to the Allen Brain Atlas via commonly-used algorithms. SOLID enables generation of neural and vascular maps within one mouse brain, as well as tracing of specific neural projections labeled with viruses. SOLID also allows cross-channel investigations of β-amyloid plaques and neurovascular lesions in the reconstructed all-in-one panorama, providing quantitative insights into structural interactions at different stages of Alzheimer's disease. Altogether, SOLID provides a robust pipeline for whole-brain mapping, which may widen the utility of tissue clearing techniques in diverse neuroscience research.
Collapse
Affiliation(s)
- 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, Hubei, 430074, 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, Hubei, 430074, China
| | - Zhang 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, Hubei, 430074, China
| | - 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, Hubei, 430074, China
| | - Jianyi Xu
- 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, Hubei, 430074, China
| | - Kunxing 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, Hubei, 430074, China
| | - Ruiying Zhang
- 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, Hubei, 430074, China
| | - Xizhi Meng
- 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, Hubei, 430074, China
| | - Peng Fei
- School of Optical Electronic Information, Huazhong University of Science and Technology, Wuhan, Hubei, 430074, 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, Hubei, 430074, 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, Hubei, 430074, China.
| |
Collapse
|
10
|
Ouyang Q, Qin R, Li Q, Huang P, Lin C, Xu Q, Quan W, Fang F, Zhu Y, Liao J, Wu K. A novel m-xylylene-diamine/glucose based-supramolecular eutectogels with tissue clearing for three dimensional histological imaging. Colloids Surf B Biointerfaces 2024; 245:114262. [PMID: 39303383 DOI: 10.1016/j.colsurfb.2024.114262] [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: 02/13/2024] [Revised: 05/27/2024] [Accepted: 09/17/2024] [Indexed: 09/22/2024]
Abstract
Hydrogel-based tissue clearing technologies have shown significant promise for deep-tissue imaging and subcellular-level optical 3D reconstruction of whole organs. This study proposes a novel approach utilizing a deep eutectic solvent (DES) formulated with glucose and m-xylylene-diamine (MXDA) to create a highly efficient tissue-clearing hydrogel system named the passive hydrogel clearing system (PHCS). PHCS achieved efficient tissue clearing through a single-step tissue gelation process. The resulting hydrogel-tissue complex exhibited thermoreversible properties, transitioning into a sol state upon heating and vice versa upon cooling. Notably, PHCS enabled media embedding, facilitating immunofluorescence histopathology. Additionally, the system demonstrated compatibility with various fluorescent probes, particularly lipophilic dyes. Our study successfully employed PHCS for the reconstruction of vascular structures within the intestine, enabling the generation of a 3D pathology model. These findings suggest that PHCS is a promising novel method for fabricating hydrogels for tissue clearing and holds great potential for application as a mounting medium for morphological imaging.
Collapse
Affiliation(s)
- Qianqian Ouyang
- The second Affiliated Hospital of Guangdong Medical University, Zhanjiang 524023, China; Marine Biomedical Research Institution, Guangdong Medical University, Zhanjiang 524023, PR China; The Marine Biomedical Research Institute of Guangdong Zhanjiang, Zhanjiang 524023, PR China
| | - Ruixiu Qin
- Marine Biomedical Research Institution, Guangdong Medical University, Zhanjiang 524023, PR China; The Marine Biomedical Research Institute of Guangdong Zhanjiang, Zhanjiang 524023, PR China
| | - Qian Li
- Marine Biomedical Research Institution, Guangdong Medical University, Zhanjiang 524023, PR China
| | - Peixin Huang
- Marine Biomedical Research Institution, Guangdong Medical University, Zhanjiang 524023, PR China
| | - Changmei Lin
- Marine Biomedical Research Institution, Guangdong Medical University, Zhanjiang 524023, PR China
| | - Qingbao Xu
- Marine Biomedical Research Institution, Guangdong Medical University, Zhanjiang 524023, PR China; The Marine Biomedical Research Institute of Guangdong Zhanjiang, Zhanjiang 524023, PR China
| | - Weiyan Quan
- Marine Biomedical Research Institution, Guangdong Medical University, Zhanjiang 524023, PR China; The Marine Biomedical Research Institute of Guangdong Zhanjiang, Zhanjiang 524023, PR China
| | - Fang Fang
- The second Affiliated Hospital of Guangdong Medical University, Zhanjiang 524023, China
| | - Yuzhen Zhu
- Marine Biomedical Research Institution, Guangdong Medical University, Zhanjiang 524023, PR China; The Marine Biomedical Research Institute of Guangdong Zhanjiang, Zhanjiang 524023, PR China
| | - Jing Liao
- The Affiliated Hospital of Guangdong Medical University, Zhanjiang 524002, PR China.
| | - Kefeng Wu
- The second Affiliated Hospital of Guangdong Medical University, Zhanjiang 524023, China; Marine Biomedical Research Institution, Guangdong Medical University, Zhanjiang 524023, PR China; The Marine Biomedical Research Institute of Guangdong Zhanjiang, Zhanjiang 524023, PR China.
| |
Collapse
|
11
|
Cao X, Li X, Li M, Sun J, Gao Z, Li X, Li Q, Shao Z, Fan C, Sun J. Light-Sheet Microscopic Imaging of Whole-Mouse Vascular Network with Fluorescent Microsphere Perfusion. ACS Biomater Sci Eng 2024; 10:5609-5616. [PMID: 38775700 DOI: 10.1021/acsbiomaterials.4c00546] [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] [Indexed: 09/10/2024]
Abstract
Visualizing the whole vascular network system is crucial for understanding the pathogenesis of specific diseases and devising targeted therapeutic interventions. Although the combination of light sheet microscopy and tissue-clearing methods has emerged as a promising approach for investigating the blood vascular network, leveraging the spatial resolution down to the capillary level and the ability to image centimeter-scale samples remains difficult. Especially, as the resolution improves, the issue of photobleaching outside the field of view poses a challenge to image the whole vascular network of adult mice at capillary resolution. Here, we devise a fluorescent microsphere vascular perfusion method to enable labeling of the whole vascular network in adult mice, which overcomes the photobleaching limit during the imaging of large samples. Moreover, by combining the utilization of a large-scale light-sheet microscope and tissue clearing protocols for whole-mouse samples, we achieve the capillary-level imaging resolution (3.2 × 3.2 × 6.5 μm) of the whole vascular network with dimensions of 45 × 15 × 82 mm in adult mice. This method thus holds great potential to deliver mesoscopic resolution images of various tissue organs for whole-animal imaging.
Collapse
Affiliation(s)
- Xiaojie Cao
- School of Biomedical Engineering, School of Chemistry and Chemical Engineering, National Center for Translational Medicine, Shanghai Jiao Tong University, 200240 Shanghai, China
| | - Xiaoyan Li
- School of Biomedical Engineering, School of Chemistry and Chemical Engineering, National Center for Translational Medicine, Shanghai Jiao Tong University, 200240 Shanghai, China
| | - Min Li
- School of Biomedical Engineering, School of Chemistry and Chemical Engineering, National Center for Translational Medicine, Shanghai Jiao Tong University, 200240 Shanghai, China
| | - Jiawei Sun
- School of Biomedical Engineering, School of Chemistry and Chemical Engineering, National Center for Translational Medicine, Shanghai Jiao Tong University, 200240 Shanghai, China
| | - Zhaoshuai Gao
- School of Biomedical Engineering, School of Chemistry and Chemical Engineering, National Center for Translational Medicine, Shanghai Jiao Tong University, 200240 Shanghai, China
| | - Xiaowei Li
- School of Biomedical Engineering, School of Chemistry and Chemical Engineering, National Center for Translational Medicine, Shanghai Jiao Tong University, 200240 Shanghai, China
| | - Qian Li
- School of Biomedical Engineering, School of Chemistry and Chemical Engineering, National Center for Translational Medicine, Shanghai Jiao Tong University, 200240 Shanghai, China
| | - Zhifeng Shao
- School of Biomedical Engineering, School of Chemistry and Chemical Engineering, National Center for Translational Medicine, Shanghai Jiao Tong University, 200240 Shanghai, China
| | - Chunhai Fan
- School of Biomedical Engineering, School of Chemistry and Chemical Engineering, National Center for Translational Medicine, Shanghai Jiao Tong University, 200240 Shanghai, China
| | - Jielin Sun
- School of Biomedical Engineering, School of Chemistry and Chemical Engineering, National Center for Translational Medicine, Shanghai Jiao Tong University, 200240 Shanghai, China
| |
Collapse
|
12
|
Fridy PC, Farrell RJ, Molloy KR, Keegan S, Wang J, Jacobs EY, Li Y, Trivedi J, Sehgal V, Fenyö D, Wu Z, Chait BT, Rout MP. A new generation of nanobody research tools using improved mass spectrometry-based discovery methods. J Biol Chem 2024; 300:107623. [PMID: 39098531 PMCID: PMC11401214 DOI: 10.1016/j.jbc.2024.107623] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Revised: 07/01/2024] [Accepted: 07/22/2024] [Indexed: 08/06/2024] Open
Abstract
Single-domain antibodies ("nanobodies") derived from the variable region of camelid heavy-chain only antibody variants have proven to be widely useful tools for research, therapeutic, and diagnostic applications. In addition to traditional display techniques, methods to generate nanobodies using direct detection by mass spectrometry and DNA sequencing have been highly effective. However, certain technical challenges have limited widespread application. We have optimized a new pipeline for this approach that greatly improves screening sensitivity, depth of antibody coverage, antigen compatibility, and overall hit rate and affinity. We have applied this improved methodology to generate significantly higher affinity nanobody repertoires against widely used targets in biological research-i.e., GFP, tdTomato, GST, and mouse, rabbit, and goat immunoglobulin G. We have characterized these reagents in affinity isolations and tissue immunofluorescence microscopy, identifying those that are optimal for these particularly demanding applications, and engineering dimeric constructs for ultra-high affinity. This study thus provides new nanobody tools directly applicable to a wide variety of research problems, and improved techniques enabling future nanobody development against diverse targets.
Collapse
Affiliation(s)
- Peter C Fridy
- Laboratory of Cellular and Structural Biology, The Rockefeller University, New York, New York, USA
| | - Ryan J Farrell
- Laboratory of Brain Development and Repair, The Rockefeller University, New York, New York, USA; Department of Biochemistry, Weill Cornell Medicine, New York, New York, USA
| | - Kelly R Molloy
- Laboratory of Mass Spectrometry and Gaseous Ion Chemistry, The Rockefeller University, New York, New York, USA
| | - Sarah Keegan
- Department of Biochemistry and Molecular Pharmacology, Institute for Systems Genetics, NYU Grossman School of Medicine, New York, New York, USA
| | - Junjie Wang
- Laboratory of Mass Spectrometry and Gaseous Ion Chemistry, The Rockefeller University, New York, New York, USA
| | - Erica Y Jacobs
- Laboratory of Mass Spectrometry and Gaseous Ion Chemistry, The Rockefeller University, New York, New York, USA; Chemistry Department, St John's University, Queens, New York, USA
| | - Yinyin Li
- Laboratory of Mass Spectrometry and Gaseous Ion Chemistry, The Rockefeller University, New York, New York, USA
| | - Jill Trivedi
- Laboratory of Cellular and Structural Biology, The Rockefeller University, New York, New York, USA
| | - Viren Sehgal
- Laboratory of Cellular and Structural Biology, The Rockefeller University, New York, New York, USA
| | - David Fenyö
- Department of Biochemistry and Molecular Pharmacology, Institute for Systems Genetics, NYU Grossman School of Medicine, New York, New York, USA
| | - Zhuhao Wu
- Laboratory of Brain Development and Repair, The Rockefeller University, New York, New York, USA
| | - Brian T Chait
- Laboratory of Mass Spectrometry and Gaseous Ion Chemistry, The Rockefeller University, New York, New York, USA.
| | - Michael P Rout
- Laboratory of Cellular and Structural Biology, The Rockefeller University, New York, New York, USA.
| |
Collapse
|
13
|
Weng D, Yang L, Xie Y. Engineering and characterization of GFP-targeting nanobody: Expression, purification, and post-translational modification analysis. Protein Expr Purif 2024; 221:106501. [PMID: 38782081 DOI: 10.1016/j.pep.2024.106501] [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: 02/21/2024] [Revised: 05/07/2024] [Accepted: 05/09/2024] [Indexed: 05/25/2024]
Abstract
Nanobodies are single-variable domain antibodies with excellent properties, which are evolving as versatile tools to guide cognate antigens in vitro and in vivo for biological research, diagnosis, and treatment. Given their simple structure, nanobodies are readily produced in multiple systems. However, selecting an appropriate expression system is crucial because different conditions might cause proteins to produce different folds or post-translational modifications (PTMs), and these differences often result in different functions. At present, the strategies of PTMs are rarely reported. The GFP nanobody can specifically target the GFP protein. Here, we engineered a GFP nanobody fused with 6 × His tag and Fc tag, respectively, and expressed in bacteria and mammalian cells. The 6 × His-GFP-nanobody was produced from Escherichia coli at high yields and the pull-down assay indicated that it can precipitate the GFP protein. Meanwhile, the Fc-GFP-nanobody can be expressed in HEK293T cells, and the co-immunoprecipitation experiment can trace and target the GFP-tagged protein in vivo. Furthermore, some different PTMs in antigen-binding regions have been identified after using mass spectrometry (MS) to analyze the GFP nanobodies, which are expressed in prokaryotes and eukaryotes. In this study, a GFP nanobody was designed, and its binding ability was verified by using the eukaryotic and prokaryotic protein expression systems. In addition, this GFP nanobody was transformed into a useful instrument for more in-depth functional investigations of GFP fusion proteins. MS was further used to explore the reason for the difference in binding ability, providing a novel perspective for the study of GFP nanobodies and protein expression purification.
Collapse
Affiliation(s)
- Dunchu Weng
- The Ministry of Education Key Laboratory of Laboratory Medical Diagnostics, The College of Laboratory Medicine, Chongqing Medical University, Chongqing, 400016, China
| | - Lin Yang
- The Ministry of Education Key Laboratory of Laboratory Medical Diagnostics, The College of Laboratory Medicine, Chongqing Medical University, Chongqing, 400016, China
| | - Yajun Xie
- The Ministry of Education Key Laboratory of Laboratory Medical Diagnostics, The College of Laboratory Medicine, Chongqing Medical University, Chongqing, 400016, China.
| |
Collapse
|
14
|
Xu C, Nedergaard M, Fowell DJ, Friedl P, Ji N. Multiphoton fluorescence microscopy for in vivo imaging. Cell 2024; 187:4458-4487. [PMID: 39178829 PMCID: PMC11373887 DOI: 10.1016/j.cell.2024.07.036] [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: 04/23/2024] [Revised: 07/18/2024] [Accepted: 07/22/2024] [Indexed: 08/26/2024]
Abstract
Multiphoton fluorescence microscopy (MPFM) has been a game-changer for optical imaging, particularly for studying biological tissues deep within living organisms. MPFM overcomes the strong scattering of light in heterogeneous tissue by utilizing nonlinear excitation that confines fluorescence emission mostly to the microscope focal volume. This enables high-resolution imaging deep within intact tissue and has opened new avenues for structural and functional studies. MPFM has found widespread applications and has led to numerous scientific discoveries and insights into complex biological processes. Today, MPFM is an indispensable tool in many research communities. Its versatility and effectiveness make it a go-to technique for researchers investigating biological phenomena at the cellular and subcellular levels in their native environments. In this Review, the principles, implementations, capabilities, and limitations of MPFM are presented. Three application areas of MPFM, neuroscience, cancer biology, and immunology, are reviewed in detail and serve as examples for applying MPFM to biological research.
Collapse
Affiliation(s)
- Chris Xu
- School of Applied and Engineering Physics, Cornell University, Ithaca, NY 14850, USA
| | - Maiken Nedergaard
- Center for Translational Neuromedicine, Faculty of Health and Medical Sciences, University of Copenhagen, Nørre Alle 3B, 2200 Copenhagen, Denmark; University of Rochester Medical School, 601 Elmwood Avenue, Rochester, NY 14642, USA
| | - Deborah J Fowell
- Department of Microbiology & Immunology, Cornell University, Ithaca, NY 14853, USA
| | - Peter Friedl
- Department of Medical BioSciences, Radboud University Medical Centre, Geert Grooteplein 26-28, Nijmegen HB 6500, the Netherlands
| | - Na Ji
- Department of Neuroscience, Department of Physics, University of California Berkeley, Berkeley, CA 94720, USA.
| |
Collapse
|
15
|
Li Y, Lu S, Zhang Z, Li X, Li Y, Li X, Xiong L. Fluorescent Pdots Facilitate High-Resolution Mapping of the Intact Meningeal Vascular Network and Eye-Brain Connections. ACS NANO 2024; 18:22080-22094. [PMID: 39102350 DOI: 10.1021/acsnano.4c05333] [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: 08/07/2024]
Abstract
Meningeal vascular network is significant in neurology and neurosurgery. However, high-resolution imaging of intact meningeal vascular network is lacking. In this work, we develop a practical experimental method to ensure that the intact meninges are morphologically unfolded and fixed in an agarose gel. With the help of high-brightness polymer dots (Pdots) as probe, macroscopic and detailed imaging of the vascular network on the intact dorsal meninges can be performed. Meningeal vessels are symmetrically distributed along the superior sagittal sinus, and the distribution of meningeal vessels had a certain degree of hierarchy. The meninges are thicker blood vessels and capillary networks from the outside to the inside. Moreover, the diameter of the capillaries is 3.96 ± 0.89 μm. Interestingly, meningeal primo vessels in the central nervous system of mice is imaged with the diameter of 4.18 ± 1.18 μm, which has not been reported previously. It is worth mentioning that we found that orthotopic xenografts of brain tumors caused the appearance of corneal neovascularization and morphological changes in optic nerve microvessels. In conclusion, our work provides an effective Pdots-based imaging method for follow-up research on meningeal vascular-related diseases, and illustrates that the eye can serve as a window for the prevention and diagnosis of brain diseases.
Collapse
Affiliation(s)
- Yuqiao Li
- Shanghai Med-X Engineering Center for Medical Equipment and Technology School of Biomedical Engineering Shanghai Jiao Tong University Shanghai 200030, P. R. China
| | - Shuting Lu
- Shanghai Med-X Engineering Center for Medical Equipment and Technology School of Biomedical Engineering Shanghai Jiao Tong University Shanghai 200030, P. R. China
| | - Zhuang Zhang
- Shanghai Med-X Engineering Center for Medical Equipment and Technology School of Biomedical Engineering Shanghai Jiao Tong University Shanghai 200030, P. R. China
| | - Xiaoyan Li
- Shanghai Med-X Engineering Center for Medical Equipment and Technology School of Biomedical Engineering Shanghai Jiao Tong University Shanghai 200030, P. R. China
| | - Yankun Li
- Shanghai Med-X Engineering Center for Medical Equipment and Technology School of Biomedical Engineering Shanghai Jiao Tong University Shanghai 200030, P. R. China
| | - Xiaowei Li
- Shanghai Med-X Engineering Center for Medical Equipment and Technology School of Biomedical Engineering Shanghai Jiao Tong University Shanghai 200030, P. R. China
| | - Liqin Xiong
- Shanghai Med-X Engineering Center for Medical Equipment and Technology School of Biomedical Engineering Shanghai Jiao Tong University Shanghai 200030, P. R. China
| |
Collapse
|
16
|
Huber D, Rabl L, Orsini C, Labek K, Viviani R. The fMRI global signal and its association with the signal from cranial bone. Neuroimage 2024; 297:120754. [PMID: 39059682 DOI: 10.1016/j.neuroimage.2024.120754] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Revised: 07/21/2024] [Accepted: 07/24/2024] [Indexed: 07/28/2024] Open
Abstract
The nature of the global signal, i.e. the average signal from sequential functional imaging scans of the brain or the cortex, is not well understood, but is thought to include vascular and neural components. Using resting state data, we report on the strong association between the global signal and the average signal from the part of the volume that includes the cranial bone and subdural vessels and venous collectors, separated from each other and the subdural space by multispectral segmentation procedures. While subdural vessels carried a signal with a phase delay relative to the cortex, the association with the cortical signal was strongest in the parts of the scan corresponding to the laminae of the cranial bone, reaching 80% shared variance in some individuals. These findings suggest that in resting state data vascular components may play a prominent role in the genesis of fluctuations of the global signal. Evidence from other studies on the existence of neural sources of the global signal suggests that it may reflect the action of multiple mechanisms (including cerebrovascular reactivity and autonomic control) concurrently acting to regulate global cerebral perfusion.
Collapse
Affiliation(s)
- Daniel Huber
- Institute of Psychology, University of Innsbruck, Innsbruck, Austria
| | - Luna Rabl
- Institute of Psychology, University of Innsbruck, Innsbruck, Austria
| | - Chiara Orsini
- Institute of Psychology, University of Innsbruck, Innsbruck, Austria
| | - Karin Labek
- Institute of Psychology, University of Innsbruck, Innsbruck, Austria
| | - Roberto Viviani
- Institute of Psychology, University of Innsbruck, Innsbruck, Austria; Psychiatry and Psychotherapy Clinic, University of Ulm, Ulm, Germany.
| |
Collapse
|
17
|
Du S, Drieu A, Cheng Y, Storck SE, Rustenhoven J, Mamuladze T, Bhattarai B, Brioschi S, Nguyen K, Ou F, Cao J, Rodrigues PF, Smirnov I, DeNardo D, Ginhoux F, Cella M, Colonna M, Kipnis J. Brain-Engrafted Monocyte-derived Macrophages from Blood and Skull-Bone Marrow Exhibit Distinct Identities from Microglia. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.08.606900. [PMID: 39211090 PMCID: PMC11361186 DOI: 10.1101/2024.08.08.606900] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
Microglia are thought to originate exclusively from primitive macrophage progenitors in the yolk sac (YS) and to persist throughout life without much contribution from definitive hematopoiesis. Here, using lineage tracing, pharmacological manipulation, and RNA-sequencing, we elucidated the presence and characteristics of monocyte-derived macrophages (MDMs) in the brain parenchyma at baseline and during microglia repopulation, and defined the core transcriptional signatures of brain-engrafted MDMs. Lineage tracing mouse models revealed that MDMs transiently express CD206 during brain engraftment as CD206 + microglia precursors in the YS. We found that brain-engrafted MDMs exhibit transcriptional and epigenetic characteristics akin to meningeal macrophages, likely due to environmental imprinting within the meningeal space. Utilizing parabiosis and skull transplantation, we demonstrated that monocytes from both peripheral blood and skull bone marrow can repopulate microglia-depleted brains. Our results reveal the heterogeneous origins and cellular dynamics of brain parenchymal macrophages at baseline and in models of microglia depletion.
Collapse
|
18
|
Jacob AM, Lindemann AF, Wagenpfeil J, Geiger S, Layer YC, Salam B, Panahabadi S, Kurt D, Wintergerst MWM, Schildberg FA, Kuetting D, Attenberger UI, Abdullah Z, Böhner AMC. Autofluorescence-based tissue characterization enhances clinical prospects of light-sheet-microscopy. Sci Rep 2024; 14:18033. [PMID: 39098935 PMCID: PMC11298517 DOI: 10.1038/s41598-024-67366-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Accepted: 07/10/2024] [Indexed: 08/06/2024] Open
Abstract
Light sheet fluorescence microscopy (LSFM) is a transformative imaging method that enables the visualization of non-dissected specimen in real-time 3D. Optical clearing of tissues is essential for LSFM, typically employing toxic solvents. Here, we test the applicability of a non-hazardous alternative, ethyl cinnamate (ECi). We comprehensively characterized autofluorescence (AF) spectra in diverse murine tissues-ocular globe, knee, and liver-employing LSFM under various excitation wavelengths (405-785 nm) to test the feasibility of unstained samples for diagnostic purposes, in particular regarding percutaneous biopsies, as they constitute to most harvested type of tissue sample in clinical routine. Ocular globe structures were best discerned with 640 nm excitation. Knee tissue showed complex variation in AF spectra variation influenced by tissue depth and structure. Liver exhibited a unique AF pattern, likely linked to vasculature. Hepatic tissue samples were used to demonstrate the compatibility of our protocol for antibody staining. Furthermore, we employed machine learning to augment raw images and segment liver structures based on AF spectra. Radiologists rated representative samples transferred to the clinical assessment software. Learning-generated images scored highest in quality. Additionally, we investigated an actual murine biopsy. Our study pioneers the application of AF spectra for tissue characterization and diagnostic potential of optically cleared unstained percutaneous biopsies, contributing to the clinical translation of LSFM.
Collapse
Affiliation(s)
- Alice M Jacob
- Institute of Molecular Medicine and Experimental Immunology, Medical Faculty, University Hospital Bonn, University of Bonn, Venusberg-Campus 1, 53127, Bonn, Germany
| | - Anna F Lindemann
- Institute of Molecular Medicine and Experimental Immunology, Medical Faculty, University Hospital Bonn, University of Bonn, Venusberg-Campus 1, 53127, Bonn, Germany
| | - Julia Wagenpfeil
- Clinics for Diagnostic and Interventional Radiology, University Hospital Bonn, Venusberg-Campus 1, 53127, Bonn, Germany
| | - Sergej Geiger
- Clinics for Diagnostic and Interventional Radiology, University Hospital Bonn, Venusberg-Campus 1, 53127, Bonn, Germany
| | - Yannik C Layer
- Clinics for Diagnostic and Interventional Radiology, University Hospital Bonn, Venusberg-Campus 1, 53127, Bonn, Germany
| | - Babak Salam
- Clinics for Diagnostic and Interventional Radiology, University Hospital Bonn, Venusberg-Campus 1, 53127, Bonn, Germany
| | - Sarah Panahabadi
- Clinic for Diagnostic and Interventional Neuroradiology, University Hospital Bonn, Venusberg-Campus 1, 53127, Bonn, Germany
| | - Darius Kurt
- Clinics for Diagnostic and Interventional Radiology, University Hospital Bonn, Venusberg-Campus 1, 53127, Bonn, Germany
| | | | - Frank A Schildberg
- Clinic for Orthopedics and Trauma Surgery, University Hospital Bonn, Venusberg-Campus 1, 53127, Bonn, Germany
| | - Daniel Kuetting
- Clinics for Diagnostic and Interventional Radiology, University Hospital Bonn, Venusberg-Campus 1, 53127, Bonn, Germany
| | - Ulrike I Attenberger
- Clinics for Diagnostic and Interventional Radiology, University Hospital Bonn, Venusberg-Campus 1, 53127, Bonn, Germany
- Department of Biomedical Imaging and Image-Guided Therapy, Medical University of Vienna and General Hospital, Waehringer Guertel 18-20, 1090, Vienna, Austria
| | - Zeinab Abdullah
- Institute of Molecular Medicine and Experimental Immunology, Medical Faculty, University Hospital Bonn, University of Bonn, Venusberg-Campus 1, 53127, Bonn, Germany
| | - Alexander M C Böhner
- Clinics for Diagnostic and Interventional Radiology, University Hospital Bonn, Venusberg-Campus 1, 53127, Bonn, Germany.
| |
Collapse
|
19
|
Salvador AFM, Abduljawad N, Kipnis J. Meningeal Lymphatics in Central Nervous System Diseases. Annu Rev Neurosci 2024; 47:323-344. [PMID: 38648267 DOI: 10.1146/annurev-neuro-113023-103045] [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: 04/25/2024]
Abstract
Since its recent discovery, the meningeal lymphatic system has reshaped our understanding of central nervous system (CNS) fluid exchange, waste clearance, immune cell trafficking, and immune privilege. Meningeal lymphatics have also been demonstrated to functionally modify the outcome of neurological disorders and their responses to treatment, including brain tumors, inflammatory diseases such as multiple sclerosis, CNS injuries, and neurodegenerative disorders such as Alzheimer's and Parkinson's diseases. In this review, we discuss recent evidence of the contribution of meningeal lymphatics to neurological diseases, as well as the available experimental methods for manipulating meningeal lymphatics in these conditions. Finally, we also provide a discussion of the pressing questions and challenges in utilizing meningeal lymphatics as a prime target for CNS therapeutic intervention and possibly drug delivery for brain disorders.
Collapse
Affiliation(s)
- Andrea Francesca M Salvador
- Brain Immunology and Glia (BIG) Center and Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri, USA;
| | - Nora Abduljawad
- Neuroscience Graduate Program, Washington University School of Medicine, St. Louis, Missouri, USA
- Brain Immunology and Glia (BIG) Center and Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri, USA;
| | - Jonathan Kipnis
- Neuroscience Graduate Program, Washington University School of Medicine, St. Louis, Missouri, USA
- Brain Immunology and Glia (BIG) Center and Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri, USA;
| |
Collapse
|
20
|
Kaag Rasmussen M, Møllgård K, Bork PAR, Weikop P, Esmail T, Drici L, Wewer Albrechtsen NJ, Carlsen JF, Huynh NPT, Ghitani N, Mann M, Goldman SA, Mori Y, Chesler AT, Nedergaard M. Trigeminal ganglion neurons are directly activated by influx of CSF solutes in a migraine model. Science 2024; 385:80-86. [PMID: 38963846 DOI: 10.1126/science.adl0544] [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: 09/27/2023] [Accepted: 05/01/2024] [Indexed: 07/06/2024]
Abstract
Classical migraine patients experience aura, which is transient neurological deficits associated with cortical spreading depression (CSD), preceding headache attacks. It is not currently understood how a pathological event in cortex can affect peripheral sensory neurons. In this study, we show that cerebrospinal fluid (CSF) flows into the trigeminal ganglion, establishing nonsynaptic signaling between brain and trigeminal cells. After CSD, ~11% of the CSF proteome is altered, with up-regulation of proteins that directly activate receptors in the trigeminal ganglion. CSF collected from animals exposed to CSD activates trigeminal neurons in naïve mice in part by CSF-borne calcitonin gene-related peptide (CGRP). We identify a communication pathway between the central and peripheral nervous system that might explain the relationship between migrainous aura and headache.
Collapse
Affiliation(s)
- Martin Kaag Rasmussen
- Center for Translational Neuromedicine, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Kjeld Møllgård
- Department of Cellular and Molecular Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Peter A R Bork
- Center for Translational Neuromedicine, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Pia Weikop
- Center for Translational Neuromedicine, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Tina Esmail
- Center for Translational Neuromedicine, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Lylia Drici
- NNF Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Nicolai J Wewer Albrechtsen
- NNF Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
- Department for Clinical Biochemistry, University Hospital Copenhagen - Bispebjerg, Copenhagen, 2400 Copenhagen, Denmark
| | - Jonathan Frederik Carlsen
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
- Department of Radiology, Copenhagen University Hospital-Rigshospitalet, 2100 Copenhagen, Denmark
| | - Nguyen P T Huynh
- Center for Translational Neuromedicine, Division of Glial Disease and Therapeutics, University of Rochester Medical Center, Rochester, NY 14642, USA
- Sana Biotechnology, Cambridge, MA 02139, USA
| | - Nima Ghitani
- National Center for Complementary and Integrative Health (NCCIH), Bethesda, MD 20892, USA
| | - Matthias Mann
- NNF Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
- Department of Proteomics and Signal Transduction, Max-Planck Institute of Biochemistry, 82152 Martinsried, Germany
| | - Steven A Goldman
- Center for Translational Neuromedicine, University of Copenhagen, 2200 Copenhagen, Denmark
- Center for Translational Neuromedicine, Division of Glial Disease and Therapeutics, University of Rochester Medical Center, Rochester, NY 14642, USA
- Sana Biotechnology, Cambridge, MA 02139, USA
| | - Yuki Mori
- Center for Translational Neuromedicine, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Alexander T Chesler
- National Center for Complementary and Integrative Health (NCCIH), Bethesda, MD 20892, USA
- National Institute of Neurological Disorders and Stroke, Bethesda, MD 20892, USA
| | - Maiken Nedergaard
- Center for Translational Neuromedicine, University of Copenhagen, 2200 Copenhagen, Denmark
- Center for Translational Neuromedicine, Division of Glial Disease and Therapeutics, University of Rochester Medical Center, Rochester, NY 14642, USA
| |
Collapse
|
21
|
Tissue histology in 3D. Nat Methods 2024; 21:1133. [PMID: 38997594 DOI: 10.1038/s41592-024-02361-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/14/2024]
|
22
|
Le Thuc O, García-Cáceres C. Obesity-induced inflammation: connecting the periphery to the brain. Nat Metab 2024; 6:1237-1252. [PMID: 38997442 DOI: 10.1038/s42255-024-01079-8] [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: 11/03/2022] [Accepted: 06/11/2024] [Indexed: 07/14/2024]
Abstract
Obesity is often associated with a chronic, low-grade inflammatory state affecting the entire body. This sustained inflammatory state disrupts the coordinated communication between the periphery and the brain, which has a crucial role in maintaining homeostasis through humoural, nutrient-mediated, immune and nervous signalling pathways. The inflammatory changes induced by obesity specifically affect communication interfaces, including the blood-brain barrier, glymphatic system and meninges. Consequently, brain areas near the third ventricle, including the hypothalamus and other cognition-relevant regions, become susceptible to impairments, resulting in energy homeostasis dysregulation and an elevated risk of cognitive impairments such as Alzheimer's disease and dementia. This Review explores the intricate communication between the brain and the periphery, highlighting the effect of obesity-induced inflammation on brain function.
Collapse
Affiliation(s)
- Ophélia Le Thuc
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center at Helmholtz Zentrum München, Neuherberg, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Cristina García-Cáceres
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center at Helmholtz Zentrum München, Neuherberg, Germany.
- German Center for Diabetes Research (DZD), Neuherberg, Germany.
- Medizinische Klinik und Poliklinik IV, Klinikum der Universität, Ludwig-Maximilians-Universität München, Munich, Germany.
| |
Collapse
|
23
|
Kaltenecker D, Al-Maskari R, Negwer M, Hoeher L, Kofler F, Zhao S, Todorov M, Rong Z, Paetzold JC, Wiestler B, Piraud M, Rueckert D, Geppert J, Morigny P, Rohm M, Menze BH, Herzig S, Berriel Diaz M, Ertürk A. Virtual reality-empowered deep-learning analysis of brain cells. Nat Methods 2024; 21:1306-1315. [PMID: 38649742 PMCID: PMC11239522 DOI: 10.1038/s41592-024-02245-2] [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: 06/03/2022] [Accepted: 03/12/2024] [Indexed: 04/25/2024]
Abstract
Automated detection of specific cells in three-dimensional datasets such as whole-brain light-sheet image stacks is challenging. Here, we present DELiVR, a virtual reality-trained deep-learning pipeline for detecting c-Fos+ cells as markers for neuronal activity in cleared mouse brains. Virtual reality annotation substantially accelerated training data generation, enabling DELiVR to outperform state-of-the-art cell-segmenting approaches. Our pipeline is available in a user-friendly Docker container that runs with a standalone Fiji plugin. DELiVR features a comprehensive toolkit for data visualization and can be customized to other cell types of interest, as we did here for microglia somata, using Fiji for dataset-specific training. We applied DELiVR to investigate cancer-related brain activity, unveiling an activation pattern that distinguishes weight-stable cancer from cancers associated with weight loss. Overall, DELiVR is a robust deep-learning tool that does not require advanced coding skills to analyze whole-brain imaging data in health and disease.
Collapse
Affiliation(s)
- Doris Kaltenecker
- Institute for Diabetes and Cancer (IDC), Helmholtz Munich, Neuherberg, Germany
- Joint Heidelberg-IDC Translational Diabetes Program, Heidelberg University Hospital, Heidelberg, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
- Institute for Stroke and Dementia Research, Klinikum der Universität München, Ludwig-Maximilians-Universität LMU, Munich, Germany
| | - Rami Al-Maskari
- Institute for Stroke and Dementia Research, Klinikum der Universität München, Ludwig-Maximilians-Universität LMU, Munich, Germany
- Institute for Tissue Engineering and Regenerative Medicine, Helmholtz Munich, Neuherberg, Germany
- Department of Computer Science, TUM Computation, Information and Technology, Technical University of Munich (TUM), Munich, Germany
- Center for Translational Cancer Research of the TUM (TranslaTUM), Munich, Germany
| | - Moritz Negwer
- Institute for Tissue Engineering and Regenerative Medicine, Helmholtz Munich, Neuherberg, Germany
| | - Luciano Hoeher
- Institute for Tissue Engineering and Regenerative Medicine, Helmholtz Munich, Neuherberg, Germany
| | - Florian Kofler
- Department of Computer Science, TUM Computation, Information and Technology, Technical University of Munich (TUM), Munich, Germany
- Center for Translational Cancer Research of the TUM (TranslaTUM), Munich, Germany
- Department of Diagnostic and Interventional Neuroradiology, School of Medicine, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
- Helmholtz AI, Helmholtz Munich, Neuherberg, Germany
| | - Shan Zhao
- Institute for Stroke and Dementia Research, Klinikum der Universität München, Ludwig-Maximilians-Universität LMU, Munich, Germany
- Institute for Tissue Engineering and Regenerative Medicine, Helmholtz Munich, Neuherberg, Germany
| | - Mihail Todorov
- Institute for Stroke and Dementia Research, Klinikum der Universität München, Ludwig-Maximilians-Universität LMU, Munich, Germany
- Institute for Tissue Engineering and Regenerative Medicine, Helmholtz Munich, Neuherberg, Germany
| | - Zhouyi Rong
- Institute for Stroke and Dementia Research, Klinikum der Universität München, Ludwig-Maximilians-Universität LMU, Munich, Germany
- Institute for Tissue Engineering and Regenerative Medicine, Helmholtz Munich, Neuherberg, Germany
| | - Johannes Christian Paetzold
- Institute for Tissue Engineering and Regenerative Medicine, Helmholtz Munich, Neuherberg, Germany
- Center for Translational Cancer Research of the TUM (TranslaTUM), Munich, Germany
- Department of Computing, Imperial College London, London, United Kingdom
| | - Benedikt Wiestler
- Department of Diagnostic and Interventional Neuroradiology, School of Medicine, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Marie Piraud
- Helmholtz AI, Helmholtz Munich, Neuherberg, Germany
| | - Daniel Rueckert
- Department of Computing, Imperial College London, London, United Kingdom
| | - Julia Geppert
- Institute for Diabetes and Cancer (IDC), Helmholtz Munich, Neuherberg, Germany
- Joint Heidelberg-IDC Translational Diabetes Program, Heidelberg University Hospital, Heidelberg, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Pauline Morigny
- Institute for Diabetes and Cancer (IDC), Helmholtz Munich, Neuherberg, Germany
- Joint Heidelberg-IDC Translational Diabetes Program, Heidelberg University Hospital, Heidelberg, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Maria Rohm
- Institute for Diabetes and Cancer (IDC), Helmholtz Munich, Neuherberg, Germany
- Joint Heidelberg-IDC Translational Diabetes Program, Heidelberg University Hospital, Heidelberg, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Bjoern H Menze
- Department of Computer Science, TUM Computation, Information and Technology, Technical University of Munich (TUM), Munich, Germany
- Department for Quantitative Biomedicine, University of Zurich, Zurich, Switzerland
| | - Stephan Herzig
- Institute for Diabetes and Cancer (IDC), Helmholtz Munich, Neuherberg, Germany
- Joint Heidelberg-IDC Translational Diabetes Program, Heidelberg University Hospital, Heidelberg, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
- Chair Molecular Metabolic Control, TU Munich, Munich, Germany
| | - Mauricio Berriel Diaz
- Institute for Diabetes and Cancer (IDC), Helmholtz Munich, Neuherberg, Germany.
- Joint Heidelberg-IDC Translational Diabetes Program, Heidelberg University Hospital, Heidelberg, Germany.
- German Center for Diabetes Research (DZD), Neuherberg, Germany.
| | - Ali Ertürk
- Institute for Stroke and Dementia Research, Klinikum der Universität München, Ludwig-Maximilians-Universität LMU, Munich, Germany.
- Institute for Tissue Engineering and Regenerative Medicine, Helmholtz Munich, Neuherberg, Germany.
- School of Medicine, Koç University, İstanbul, Turkey.
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany.
- Deep Piction, Munich, Germany.
| |
Collapse
|
24
|
Whiteley AE, Ma D, Wang L, Yu SY, Yin C, Price TT, Simon BG, Xu KR, Marsh KA, Brockman ML, Prioleau TM, Zhou KI, Cui X, Fecci PE, Jeck WR, McCall CM, Neff JL, Sipkins DA. Breast cancer exploits neural signaling pathways for bone-to-meninges metastasis. Science 2024; 384:eadh5548. [PMID: 38900896 DOI: 10.1126/science.adh5548] [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: 03/11/2023] [Accepted: 04/23/2024] [Indexed: 06/22/2024]
Abstract
The molecular mechanisms that regulate breast cancer cell (BCC) metastasis and proliferation within the leptomeninges (LM) are poorly understood, which limits the development of effective therapies. In this work, we show that BCCs in mice can invade the LM by abluminal migration along blood vessels that connect vertebral or calvarial bone marrow and meninges, bypassing the blood-brain barrier. This process is dependent on BCC engagement with vascular basement membrane laminin through expression of the neuronal pathfinding molecule integrin α6. Once in the LM, BCCs colocalize with perivascular meningeal macrophages and induce their expression of the prosurvival neurotrophin glial-derived neurotrophic factor (GDNF). Intrathecal GDNF blockade, macrophage-specific GDNF ablation, or deletion of the GDNF receptor neural cell adhesion molecule (NCAM) from BCCs inhibits breast cancer growth within the LM. These data suggest integrin α6 and the GDNF signaling axis as new therapeutic targets against breast cancer LM metastasis.
Collapse
Affiliation(s)
- Andrew E Whiteley
- Department of Medicine, Division of Hematologic Malignancies and Cellular Therapy, Duke University, Durham, NC 27710, USA
| | - Danhui Ma
- Department of Medicine, Division of Hematologic Malignancies and Cellular Therapy, Duke University, Durham, NC 27710, USA
| | - Lihua Wang
- Department of Medicine, Division of Hematologic Malignancies and Cellular Therapy, Duke University, Durham, NC 27710, USA
| | - Seok-Yeong Yu
- Department of Medicine, Division of Hematologic Malignancies and Cellular Therapy, Duke University, Durham, NC 27710, USA
| | - Claire Yin
- Department of Medicine, Division of Hematologic Malignancies and Cellular Therapy, Duke University, Durham, NC 27710, USA
| | - Trevor T Price
- Department of Medicine, Division of Hematologic Malignancies and Cellular Therapy, Duke University, Durham, NC 27710, USA
| | - Brennan G Simon
- Department of Medicine, Division of Hematologic Malignancies and Cellular Therapy, Duke University, Durham, NC 27710, USA
| | - Katie R Xu
- Department of Medicine, Division of Hematologic Malignancies and Cellular Therapy, Duke University, Durham, NC 27710, USA
| | - Kathleen A Marsh
- Department of Medicine, Division of Hematologic Malignancies and Cellular Therapy, Duke University, Durham, NC 27710, USA
| | - Maegan L Brockman
- Department of Medicine, Division of Hematologic Malignancies and Cellular Therapy, Duke University, Durham, NC 27710, USA
| | - Tatiana M Prioleau
- Department of Medicine, Division of Hematologic Malignancies and Cellular Therapy, Duke University, Durham, NC 27710, USA
| | - Katherine I Zhou
- Department of Medicine, Division of Hematologic Malignancies and Cellular Therapy, Duke University, Durham, NC 27710, USA
| | - Xiuyu Cui
- Department of Neurosurgery, Duke University, Durham, NC 27710, USA
| | - Peter E Fecci
- Department of Neurosurgery, Duke University, Durham, NC 27710, USA
| | - William R Jeck
- Department of Pathology, Duke University, Durham, NC 27710, USA
| | - Chad M McCall
- Department of Pathology, Duke University, Durham, NC 27710, USA
| | - Jadee L Neff
- Department of Pathology, Duke University, Durham, NC 27710, USA
| | - Dorothy A Sipkins
- Department of Medicine, Division of Hematologic Malignancies and Cellular Therapy, Duke University, Durham, NC 27710, USA
| |
Collapse
|
25
|
Eden T, Schaffrath AZ, Wesolowski J, Stähler T, Tode N, Richter N, Schäfer W, Hambach J, Hermans-Borgmeyer I, Woens J, Le Gall CM, Wendler S, Linke-Winnebeck C, Stobbe M, Budnicki I, Wanney A, Heitz Y, Schimmelpfennig L, Schweitzer L, Zimmer D, Stahl E, Seyfried F, Gebhardt AJ, Dieckow L, Riecken K, Fehse B, Bannas P, Magnus T, Verdoes M, Figdor CG, Hartlepp KF, Schleer H, Füner J, Tomas NM, Haag F, Rissiek B, Mann AM, Menzel S, Koch-Nolte F. Generation of nanobodies from transgenic 'LamaMice' lacking an endogenous immunoglobulin repertoire. Nat Commun 2024; 15:4728. [PMID: 38830864 PMCID: PMC11148044 DOI: 10.1038/s41467-024-48735-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Accepted: 05/02/2024] [Indexed: 06/05/2024] Open
Abstract
Due to their exceptional solubility and stability, nanobodies have emerged as powerful building blocks for research tools and therapeutics. However, their generation in llamas is cumbersome and costly. Here, by inserting an engineered llama immunoglobulin heavy chain (IgH) locus into IgH-deficient mice, we generate a transgenic mouse line, which we refer to as 'LamaMouse'. We demonstrate that LamaMice solely express llama IgH molecules without association to Igκ or λ light chains. Immunization of LamaMice with AAV8, the receptor-binding domain of the SARS-CoV-2 spike protein, IgE, IgG2c, and CLEC9A enabled us to readily select respective target-specific nanobodies using classical hybridoma and phage display technologies, single B cell screening, and direct cloning of the nanobody-repertoire into a mammalian expression vector. Our work shows that the LamaMouse represents a flexible and broadly applicable platform for a facilitated selection of target-specific nanobodies.
Collapse
Affiliation(s)
- Thomas Eden
- Institute of Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Alessa Z Schaffrath
- Institute of Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Janusz Wesolowski
- Institute of Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Tobias Stähler
- Institute of Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Natalie Tode
- Institute of Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Nathalie Richter
- Institute of Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Waldemar Schäfer
- Institute of Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Julia Hambach
- Institute of Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Irm Hermans-Borgmeyer
- Center for Molecular Neurobiology Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Jannis Woens
- Research Department Cell and Gene Therapy, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Camille M Le Gall
- Department of Medical BioSciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Sabrina Wendler
- ChromoTek GmbH, Martinsried, Germany - A part of Proteintech Group, Martinsried, Germany
| | | | - Martina Stobbe
- ChromoTek GmbH, Martinsried, Germany - A part of Proteintech Group, Martinsried, Germany
| | | | | | | | | | | | | | | | - Fabienne Seyfried
- Institute of Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Anna J Gebhardt
- Institute of Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Lynn Dieckow
- Institute of Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Kristoffer Riecken
- Research Department Cell and Gene Therapy, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Boris Fehse
- Research Department Cell and Gene Therapy, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Peter Bannas
- Department of Radiology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Tim Magnus
- Department of Neurology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Martijn Verdoes
- Department of Medical BioSciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Carl G Figdor
- Department of Medical BioSciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Klaus F Hartlepp
- ChromoTek GmbH, Martinsried, Germany - A part of Proteintech Group, Martinsried, Germany
| | | | | | - Nicola M Tomas
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Friedrich Haag
- Institute of Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Björn Rissiek
- Department of Neurology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Anna M Mann
- Institute of Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Stephan Menzel
- Institute of Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Core Facility Nanobodies, University of Bonn, Bonn, Germany
| | - Friedrich Koch-Nolte
- Institute of Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.
| |
Collapse
|
26
|
Dourson AJ, Fadaka AO, Warshak AM, Paranjpe A, Weinhaus B, Queme LF, Hofmann MC, Evans HM, Donmez OA, Forney C, Weirauch MT, Kottyan LC, Lucas D, Deepe GS, Jankowski MP. Macrophage memories of early-life injury drive neonatal nociceptive priming. Cell Rep 2024; 43:114129. [PMID: 38640063 PMCID: PMC11197107 DOI: 10.1016/j.celrep.2024.114129] [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: 02/14/2023] [Revised: 01/05/2024] [Accepted: 04/04/2024] [Indexed: 04/21/2024] Open
Abstract
The developing peripheral nervous and immune systems are functionally distinct from those of adults. These systems are vulnerable to early-life injury, which influences outcomes related to nociception following subsequent injury later in life (i.e., "neonatal nociceptive priming"). The underpinnings of this phenomenon are unclear, although previous work indicates that macrophages are trained by inflammation and injury. Our findings show that macrophages are both necessary and partially sufficient to drive neonatal nociceptive priming, possibly due to a long-lasting remodeling in chromatin structure. The p75 neurotrophic factor receptor is an important effector in regulating neonatal nociceptive priming through modulation of the inflammatory profile of rodent and human macrophages. This "pain memory" is long lasting in females and can be transferred to a naive host to alter sex-specific pain-related behaviors. This study reveals a mechanism by which acute, neonatal post-surgical pain drives a peripheral immune-related predisposition to persistent pain following a subsequent injury.
Collapse
Affiliation(s)
- Adam J Dourson
- Department of Anesthesia, Division of Pain Management, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Adewale O Fadaka
- Department of Anesthesia, Division of Pain Management, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Anna M Warshak
- Department of Anesthesia, Division of Pain Management, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Aditi Paranjpe
- Biomedical Informatics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Benjamin Weinhaus
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Medical Center, Cincinnati, OH, USA
| | - Luis F Queme
- Department of Anesthesia, Division of Pain Management, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Megan C Hofmann
- Department of Anesthesia, Division of Pain Management, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Heather M Evans
- Division of Infectious Diseases, University of Cincinnati, Cincinnati, OH, USA
| | - Omer A Donmez
- Center for Autoimmune Genomics and Etiology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Carmy Forney
- Center for Autoimmune Genomics and Etiology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Matthew T Weirauch
- Center for Autoimmune Genomics and Etiology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA; Division of Allergy and Immunology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA; Divisions of Biomedical Informatics and Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Leah C Kottyan
- Center for Autoimmune Genomics and Etiology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA; Division of Allergy and Immunology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA; Department of Pediatrics, College of Medicine, University of Cincinnati, Cincinnati, OH 45229, USA
| | - Daniel Lucas
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Medical Center, Cincinnati, OH, USA; Department of Pediatrics, College of Medicine, University of Cincinnati, Cincinnati, OH 45229, USA
| | - George S Deepe
- Division of Infectious Diseases, Department of Medicine, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Michael P Jankowski
- Department of Anesthesia, Division of Pain Management, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA; Department of Pediatrics, College of Medicine, University of Cincinnati, Cincinnati, OH 45229, USA; Pediatric Pain Research Center, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA.
| |
Collapse
|
27
|
Lee EJ, Suh M, Choi H, Choi Y, Hwang DW, Bae S, Lee DS. Spatial transcriptomic brain imaging reveals the effects of immunomodulation therapy on specific regional brain cells in a mouse dementia model. BMC Genomics 2024; 25:516. [PMID: 38796425 PMCID: PMC11128132 DOI: 10.1186/s12864-024-10434-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2024] [Accepted: 05/20/2024] [Indexed: 05/28/2024] Open
Abstract
Increasing evidence of brain-immune crosstalk raises expectations for the efficacy of novel immunotherapies in Alzheimer's disease (AD), but the lack of methods to examine brain tissues makes it difficult to evaluate therapeutics. Here, we investigated the changes in spatial transcriptomic signatures and brain cell types using the 10x Genomics Visium platform in immune-modulated AD models after various treatments. To proceed with an analysis suitable for barcode-based spatial transcriptomics, we first organized a workflow for segmentation of neuroanatomical regions, establishment of appropriate gene combinations, and comprehensive review of altered brain cell signatures. Ultimately, we investigated spatial transcriptomic changes following administration of immunomodulators, NK cell supplements and an anti-CD4 antibody, which ameliorated behavior impairment, and designated brain cells and regions showing probable associations with behavior changes. We provided the customized analytic pipeline into an application named STquantool. Thus, we anticipate that our approach can help researchers interpret the real action of drug candidates by simultaneously investigating the dynamics of all transcripts for the development of novel AD therapeutics.
Collapse
Affiliation(s)
- Eun Ji Lee
- Department of Nuclear Medicine, Seoul National University College of Medicine, 101 Daehak-ro, Jongno-gu, Seoul, 03080, Republic of Korea
- Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, Seoul National University, Seoul, Republic of Korea
| | - Minseok Suh
- Department of Nuclear Medicine, Seoul National University College of Medicine, 101 Daehak-ro, Jongno-gu, Seoul, 03080, Republic of Korea
- Department of Nuclear Medicine, Seoul National University Hospital, Seoul, Republic of Korea
- Institute of Radiation Medicine, Medical Research Center, Seoul National University, 103 Daehak-ro, Jongno-gu, Seoul, 03080, Republic of Korea
| | - Hongyoon Choi
- Department of Nuclear Medicine, Seoul National University College of Medicine, 101 Daehak-ro, Jongno-gu, Seoul, 03080, Republic of Korea
- Department of Nuclear Medicine, Seoul National University Hospital, Seoul, Republic of Korea
| | - Yoori Choi
- Department of Nuclear Medicine, Seoul National University College of Medicine, 101 Daehak-ro, Jongno-gu, Seoul, 03080, Republic of Korea
- Cliniclal Research Institute, Seoul National University Hospital, Seoul, Republic of Korea
| | - Do Won Hwang
- Research and Development Center, THERABEST Inc., Seocho-daero 40-gil, Seoul, 06657, Republic of Korea
| | - Sungwoo Bae
- Department of Nuclear Medicine, Seoul National University College of Medicine, 101 Daehak-ro, Jongno-gu, Seoul, 03080, Republic of Korea.
- Department of Nuclear Medicine, Seoul National University Hospital, Seoul, Republic of Korea.
- Institute of Radiation Medicine, Medical Research Center, Seoul National University, 103 Daehak-ro, Jongno-gu, Seoul, 03080, Republic of Korea.
| | - Dong Soo Lee
- Department of Nuclear Medicine, Seoul National University College of Medicine, 101 Daehak-ro, Jongno-gu, Seoul, 03080, Republic of Korea.
- Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, Seoul National University, Seoul, Republic of Korea.
- Department of Nuclear Medicine, Seoul National University Hospital, Seoul, Republic of Korea.
- Institute of Radiation Medicine, Medical Research Center, Seoul National University, 103 Daehak-ro, Jongno-gu, Seoul, 03080, Republic of Korea.
- Medical Science and Engineering, School of Convergence Science and Technology, POSTECH, Pohang, Republic of Korea.
| |
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
|
Xie DF, Crouzet C, LoPresti K, Wang Y, Robinson C, Jones W, Muqolli F, Fang C, Cribbs DH, Fisher M, Choi B. Semi-automated protocol to quantify and characterize fluorescent three-dimensional vascular images. PLoS One 2024; 19:e0289109. [PMID: 38753706 PMCID: PMC11098357 DOI: 10.1371/journal.pone.0289109] [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: 08/30/2022] [Accepted: 07/11/2023] [Indexed: 05/18/2024] Open
Abstract
The microvasculature facilitates gas exchange, provides nutrients to cells, and regulates blood flow in response to stimuli. Vascular abnormalities are an indicator of pathology for various conditions, such as compromised vessel integrity in small vessel disease and angiogenesis in tumors. Traditional immunohistochemistry enables the visualization of tissue cross-sections containing exogenously labeled vasculature. Although this approach can be utilized to quantify vascular changes within small fields of view, it is not a practical way to study the vasculature on the scale of whole organs. Three-dimensional (3D) imaging presents a more appropriate method to visualize the vascular architecture in tissue. Here we describe the complete protocol that we use to characterize the vasculature of different organs in mice encompassing the methods to fluorescently label vessels, optically clear tissue, collect 3D vascular images, and quantify these vascular images with a semi-automated approach. To validate the automated segmentation of vascular images, one user manually segmented one hundred random regions of interest across different vascular images. The automated segmentation results had an average sensitivity of 83±11% and an average specificity of 91±6% when compared to manual segmentation. Applying this procedure of image analysis presents a method to reliably quantify and characterize vascular networks in a timely fashion. This procedure is also applicable to other methods of tissue clearing and vascular labels that generate 3D images of microvasculature.
Collapse
Affiliation(s)
- Danny F. Xie
- Beckman Laser Institute and Medical Clinic, University of California-Irvine, Irvine, CA, United States of America
- Department of Biomedical Engineering, University of California-Irvine, Irvine, CA, United States of America
| | - Christian Crouzet
- Beckman Laser Institute and Medical Clinic, University of California-Irvine, Irvine, CA, United States of America
- Department of Biomedical Engineering, University of California-Irvine, Irvine, CA, United States of America
| | - Krystal LoPresti
- Beckman Laser Institute and Medical Clinic, University of California-Irvine, Irvine, CA, United States of America
- Department of Biomedical Engineering, University of California-Irvine, Irvine, CA, United States of America
| | - Yuke Wang
- Beckman Laser Institute and Medical Clinic, University of California-Irvine, Irvine, CA, United States of America
- Department of Biomedical Engineering, University of California-Irvine, Irvine, CA, United States of America
| | - Christopher Robinson
- Beckman Laser Institute and Medical Clinic, University of California-Irvine, Irvine, CA, United States of America
- Department of Biomedical Engineering, University of California-Irvine, Irvine, CA, United States of America
| | - William Jones
- Beckman Laser Institute and Medical Clinic, University of California-Irvine, Irvine, CA, United States of America
| | - Fjolla Muqolli
- Beckman Laser Institute and Medical Clinic, University of California-Irvine, Irvine, CA, United States of America
| | - Chuo Fang
- Department of Neurology, University of California-Irvine, Irvine, CA, United States of America
| | - David H. Cribbs
- Institute for Memory Impairments and Neurological Disorders, University of California-Irvine, Irvine, CA, United States of America
| | - Mark Fisher
- Beckman Laser Institute and Medical Clinic, University of California-Irvine, Irvine, CA, United States of America
- Department of Neurology, University of California-Irvine, Irvine, CA, United States of America
- Institute for Memory Impairments and Neurological Disorders, University of California-Irvine, Irvine, CA, United States of America
- Department of Pathology & Laboratory Medicine, University of California-Irvine, Irvine, CA, United States of America
| | - Bernard Choi
- Beckman Laser Institute and Medical Clinic, University of California-Irvine, Irvine, CA, United States of America
- Department of Biomedical Engineering, University of California-Irvine, Irvine, CA, United States of America
| |
Collapse
|
30
|
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
|
31
|
Da Mesquita S, Rua R. Brain border-associated macrophages: common denominators in infection, aging, and Alzheimer's disease? Trends Immunol 2024; 45:346-357. [PMID: 38632001 PMCID: PMC11088519 DOI: 10.1016/j.it.2024.03.007] [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: 01/24/2024] [Revised: 03/18/2024] [Accepted: 03/20/2024] [Indexed: 04/19/2024]
Abstract
Mammalian brain border-associated macrophages (BAMs) are strategically positioned to support vital properties and processes: for example, the composition of the brain's perivascular extracellular matrix and cerebrospinal fluid flow via the glymphatic pathway. BAMs also effectively restrict the spread of infectious microbes into the brain. However, while fighting infections, BAMs sustain long-term transcriptomic changes and can be replaced by inflammatory monocytes, potentially leading to a gradual loss of their beneficial homeostatic functions. We hypothesize that by expediting the deterioration of BAMs, multiple infection episodes might be associated with accelerated brain aging and the putative development of neurodegenerative diseases. Our viewpoint is supported by recent studies suggesting that rejuvenating aged BAMs, and counterbalancing their detrimental inflammatory signatures during infections, might hold promise in treating aging-related neurological disorders, including Alzheimer's disease (AD).
Collapse
Affiliation(s)
| | - Rejane Rua
- Centre d'Immunologie de Marseille-Luminy, Aix Marseille Université, Inserm, CNRS, Marseille, France.
| |
Collapse
|
32
|
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
|
33
|
Goodman GW, Devlin P, West BE, Ritzel RM. The emerging importance of skull-brain interactions in traumatic brain injury. Front Immunol 2024; 15:1353513. [PMID: 38680490 PMCID: PMC11047125 DOI: 10.3389/fimmu.2024.1353513] [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: 12/10/2023] [Accepted: 04/01/2024] [Indexed: 05/01/2024] Open
Abstract
The recent identification of skull bone marrow as a reactive hematopoietic niche that can contribute to and direct leukocyte trafficking into the meninges and brain has transformed our view of this bone structure from a solid, protective casing to a living, dynamic tissue poised to modulate brain homeostasis and neuroinflammation. This emerging concept may be highly relevant to injuries that directly impact the skull such as in traumatic brain injury (TBI). From mild concussion to severe contusion with skull fracturing, the bone marrow response of this local myeloid cell reservoir has the potential to impact not just the acute inflammatory response in the brain, but also the remodeling of the calvarium itself, influencing its response to future head impacts. If we borrow understanding from recent discoveries in other CNS immunological niches and extend them to this nascent, but growing, subfield of neuroimmunology, it is not unreasonable to consider the hematopoietic compartment in the skull may similarly play an important role in health, aging, and neurodegenerative disease following TBI. This literature review briefly summarizes the traditional role of the skull in TBI and offers some additional insights into skull-brain interactions and their potential role in affecting secondary neuroinflammation and injury outcomes.
Collapse
Affiliation(s)
| | | | | | - Rodney M. Ritzel
- Department of Neurology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, United States
| |
Collapse
|
34
|
Francis VI, Liddle C, Camacho E, Kulkarni M, Junior SRS, Harvey JA, Ballou ER, Thomson DD, Brown GD, Hardwick JM, Casadevall A, Witton J, Coelho C. Cryptococcus neoformans rapidly invades the murine brain by sequential breaching of airway and endothelial tissues barriers, followed by engulfment by microglia. mBio 2024; 15:e0307823. [PMID: 38511961 PMCID: PMC11005363 DOI: 10.1128/mbio.03078-23] [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: 11/14/2023] [Accepted: 02/29/2024] [Indexed: 03/22/2024] Open
Abstract
Cryptococcus neoformans causes lethal meningitis and accounts for approximately 10%-15% of AIDS-associated deaths worldwide. There are major gaps in our understanding of how this fungus invades the mammalian brain. To investigate the dynamics of C. neoformans tissue invasion, we mapped fungal localization and host cell interactions in infected brain, lung, and upper airways using mouse models of systemic and airway infection. To enable this, we developed an in situ imaging pipeline capable of measuring large volumes of tissue while preserving anatomical and cellular information by combining thick tissue sections, tissue clarification, and confocal imaging. We confirm high fungal burden in mouse upper airway after nasal inoculation. Yeast in turbinates were frequently titan cells, with faster kinetics than reported in mouse lungs. Importantly, we observed one instance of fungal cells enmeshed in lamina propria of the upper airways, suggesting penetration of airway mucosa as a possible route of tissue invasion and dissemination to the bloodstream. We extend previous literature positing bloodstream dissemination of C. neoformans, by finding viable fungi in the bloodstream of mice a few days after intranasal infection. As early as 24 h post systemic infection, the majority of C. neoformans cells traversed the blood-brain barrier, and were engulfed or in close proximity to microglia. Our work presents a new method for investigating microbial invasion, establishes that C. neoformans can breach multiple tissue barriers within the first days of infection, and demonstrates microglia as the first cells responding to C. neoformans invasion of the brain.IMPORTANCECryptococcal meningitis causes 10%-15% of AIDS-associated deaths globally. Still, brain-specific immunity to cryptococci is a conundrum. By employing innovative imaging, this study reveals what occurs during the first days of infection in brain and in airways. We found that titan cells predominate in upper airways and that cryptococci breach the upper airway mucosa, which implies that, at least in mice, the upper airways are a site for fungal dissemination. This would signify that mucosal immunity of the upper airway needs to be better understood. Importantly, we also show that microglia, the brain-resident macrophages, are the first responders to infection, and microglia clusters are formed surrounding cryptococci. This study opens the field to detailed molecular investigations on airway immune response, how fungus traverses the blood-brain barrier, how microglia respond to infection, and ultimately how microglia monitor the blood-brain barrier to preserve brain function.
Collapse
Affiliation(s)
- Vanessa I. Francis
- MRC Centre for Medical Mycology at University of Exeter, University of Exeter, Exeter, United Kingdom
- Faculty of Health and Life Sciences, University of Exeter, Exeter, United Kingdom
| | - Corin Liddle
- Bioimaging Facility, University of Exeter, Exeter, United Kingdom
| | - Emma Camacho
- Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Madhura Kulkarni
- Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | | | - Jamie A. Harvey
- MRC Centre for Medical Mycology at University of Exeter, University of Exeter, Exeter, United Kingdom
| | - Elizabeth R. Ballou
- MRC Centre for Medical Mycology at University of Exeter, University of Exeter, Exeter, United Kingdom
| | - Darren D. Thomson
- MRC Centre for Medical Mycology at University of Exeter, University of Exeter, Exeter, United Kingdom
| | - Gordon D. Brown
- MRC Centre for Medical Mycology at University of Exeter, University of Exeter, Exeter, United Kingdom
- Faculty of Health and Life Sciences, University of Exeter, Exeter, United Kingdom
| | - J. Marie Hardwick
- Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Arturo Casadevall
- Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Jonathan Witton
- Faculty of Health and Life Sciences, University of Exeter, Exeter, United Kingdom
| | - Carolina Coelho
- MRC Centre for Medical Mycology at University of Exeter, University of Exeter, Exeter, United Kingdom
- Faculty of Health and Life Sciences, University of Exeter, Exeter, United Kingdom
| |
Collapse
|
35
|
Takahashi-Yamashiro K, Miyazono K. Tissue clearing method in visualization of cancer progression and metastasis. Ups J Med Sci 2024; 129:10634. [PMID: 38716075 PMCID: PMC11075440 DOI: 10.48101/ujms.v129.10634] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2024] [Accepted: 03/07/2024] [Indexed: 05/24/2024] Open
Abstract
Since various imaging modalities have been developed, cancer metastasis can be detected from an early stage. However, limitations still exist, especially in terms of spatial resolution. Tissue-clearing technology has emerged as a new imaging modality in cancer research, which has been developed and utilized for a long time mainly in neuroscience field. This method enables us to detect cancer metastatic foci with single-cell resolution at whole mouse body/organ level. On top of that, 3D images of cancer metastasis of whole mouse organs make it easy to understand their characteristics. Recently, further applications of tissue clearing methods were reported in combination with reporter systems, labeling, and machine learning. In this review, we would like to provide an overview of this technique and current applications in cancer research and discuss their potentials and limitations.
Collapse
Affiliation(s)
- Kei Takahashi-Yamashiro
- Department of Chemistry, Faculty of Science, University of Alberta, Edmonton, Alberta, Canada
| | - Kohei Miyazono
- Department of Applied Pathology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
- Laboratory for Cancer Invasion and Metastasis, Institute for Medical Sciences, RIKEN, Yokohama City, Kanagawa, Japan
| |
Collapse
|
36
|
Breaking boundaries in whole-body imaging and disease understanding with wildDISCO. Nat Biotechnol 2024; 42:576-577. [PMID: 37430075 DOI: 10.1038/s41587-023-01864-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/12/2023]
|
37
|
Freret ME, Boire A. The anatomic basis of leptomeningeal metastasis. J Exp Med 2024; 221:e20212121. [PMID: 38451255 PMCID: PMC10919154 DOI: 10.1084/jem.20212121] [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: 03/30/2022] [Revised: 09/20/2022] [Accepted: 02/08/2024] [Indexed: 03/08/2024] Open
Abstract
Leptomeningeal metastasis (LM), or spread of cancer to the cerebrospinal fluid (CSF)-filled space surrounding the central nervous system, is a fatal complication of cancer. Entry into this space poses an anatomical challenge for cancer cells; movement of cells between the blood and CSF is tightly regulated by the blood-CSF barriers. Anatomical understanding of the leptomeninges provides a roadmap of corridors for cancer entry. This Review describes the anatomy of the leptomeninges and routes of cancer spread to the CSF. Granular understanding of LM by route of entry may inform strategies for novel diagnostic and preventive strategies as well as therapies.
Collapse
Affiliation(s)
- Morgan E. Freret
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Adrienne Boire
- Department of Neurology, Human Oncology and Pathogenesis Program, Brain Tumor Center, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| |
Collapse
|
38
|
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: 23] [Impact Index Per Article: 23.0] [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
|
39
|
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
|
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
|
Mesa H, Meade J, Gajewski-Kurdziel P, Blakely RD, Zhang Q. Simple Rescue of Opaque Tissue Previously Cleared by iDISCO. Bio Protoc 2024; 14:e4948. [PMID: 38464943 PMCID: PMC10917697 DOI: 10.21769/bioprotoc.4948] [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: 11/14/2023] [Revised: 01/04/2024] [Accepted: 01/30/2024] [Indexed: 03/12/2024] Open
Abstract
Recent advancements in tissue-clearing techniques and volumetric imaging have greatly facilitated visualization and quantification of biomolecules, organelles, and cells in intact organs or even entire organisms. Generally, there are two types of clearing methods: hydrophobic and hydrophilic (i.e., clearing with organic or aqueous solvents, respectively). The popular iDISCO approach and its modifications are hydrophobic methods that involve dehydration, delipidation, decolorization (optional), decalcification (optional), and refractive-index (RI) matching steps. Cleared samples are often stored for a relatively long period of time and imaged repeatedly. However, cleared tissues can become opaque over time, which prevents accurate reimaging. We reasoned that the resurgent haziness is likely due to rehydration, residual lipids, and uneven RI deep inside those tissue samples. For rescue, we have developed a simple procedure based on iDISCO. Beginning with a methanol dehydration, samples are delipidated using dichloromethane, followed by RI matching with dibenzyl ether (DBE). This simple method effectively re-clears mouse brains that have turned opaque during months of storage, allowing the user to effectively image immunolabeled samples over longer periods of time. Key features • This simple protocol rescues previously cleared tissue that has turned opaque. • The method does not cause detectable loss of immunofluorescence from previously stained samples. Graphical overview.
Collapse
Affiliation(s)
- Haylee Mesa
- Department of Chemistry and Biochemistry, Charles E.
Schmidt College of Science, Florida Atlantic University, Boca Raton, FL, USA
| | - Jonathan Meade
- Department of Chemistry and Biochemistry, Charles E.
Schmidt College of Science, Florida Atlantic University, Boca Raton, FL, USA
| | | | - Randy D. Blakely
- Department of Biomedical Science, Charles E. Schmidt
College of Medicine, Jupiter, FL, USA
- Stiles-Nicholson Brain Institute, Florida Atlantic
University, Jupiter, FL, USA
| | - Qi Zhang
- Department of Chemistry and Biochemistry, Charles E.
Schmidt College of Science, Florida Atlantic University, Boca Raton, FL, USA
- Stiles-Nicholson Brain Institute, Florida Atlantic
University, Jupiter, FL, USA
| |
Collapse
|
42
|
Ren H, Liu Q. Skull and vertebral bone marrow in central nervous system inflammation. FUNDAMENTAL RESEARCH 2024; 4:246-250. [PMID: 38933518 PMCID: PMC11197483 DOI: 10.1016/j.fmre.2023.01.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Revised: 01/02/2023] [Accepted: 01/06/2023] [Indexed: 03/07/2023] Open
Abstract
Emerging evidence has highlighted the capacity of hematogenous cells in skull and vertebral bone marrow to enter the meningeal borders via ossified vascular channels and maintain immune homeostasis in the central nervous system (CNS). CNS-adjacent skull and vertebral bone marrow comprises hematopoietic niches that can sense CNS injury and supply specialized immune cells to fine-tune inflammatory responses. Here, we review recent advances in our understanding of skull and vertebral bone marrow-derived immune cells in homeostasis and inflammatory CNS diseases. Further, we discuss the implications for future development of therapies to mitigate CNS inflammation and its detrimental sequelae in neurological disorders.
Collapse
Affiliation(s)
- Honglei Ren
- Department of Neurology, Tianjin Neurological Institute, Tianjin Institute of Immunology, State Key Laboratory of Experimental Hematology, International Joint Laboratory of Ocular Diseases, Ministry of Education, Haihe Laboratory of Cell Ecosystem, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Qiang Liu
- Department of Neurology, Tianjin Neurological Institute, Tianjin Institute of Immunology, State Key Laboratory of Experimental Hematology, International Joint Laboratory of Ocular Diseases, Ministry of Education, Haihe Laboratory of Cell Ecosystem, Tianjin Medical University General Hospital, Tianjin 300052, China
| |
Collapse
|
43
|
Mertens TF, Liebheit AT, Ehl J, Köhler R, Rakhymzhan A, Woehler A, Katthän L, Ebel G, Liublin W, Kasapi A, Triantafyllopoulou A, Schulz TJ, Niesner RA, Hauser AE. MarShie: a clearing protocol for 3D analysis of single cells throughout the bone marrow at subcellular resolution. Nat Commun 2024; 15:1764. [PMID: 38409121 PMCID: PMC10897183 DOI: 10.1038/s41467-024-45827-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Accepted: 02/01/2024] [Indexed: 02/28/2024] Open
Abstract
Analyzing immune cell interactions in the bone marrow is vital for understanding hematopoiesis and bone homeostasis. Three-dimensional analysis of the complete, intact bone marrow within the cortex of whole long bones remains a challenge, especially at subcellular resolution. We present a method that stabilizes the marrow and provides subcellular resolution of fluorescent signals throughout the murine femur, enabling identification and spatial characterization of hematopoietic and stromal cell subsets. By combining a pre-processing algorithm for stripe artifact removal with a machine-learning approach, we demonstrate reliable cell segmentation down to the deepest bone marrow regions. This reveals age-related changes in the marrow. It highlights the interaction between CX3CR1+ cells and the vascular system in homeostasis, in contrast to other myeloid cell types, and reveals their spatial characteristics after injury. The broad applicability of this method will contribute to a better understanding of bone marrow biology.
Collapse
Affiliation(s)
- Till Fabian Mertens
- Department of Rheumatology and Clinical Immunology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 10117, Berlin, Germany
- Immune Dynamics, Deutsches Rheuma-Forschungszentrum (DRFZ), a Leibniz Institute, Charitéplatz 1, 10117, Berlin, Germany
| | - Alina Tabea Liebheit
- Department of Rheumatology and Clinical Immunology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 10117, Berlin, Germany
- Immune Dynamics, Deutsches Rheuma-Forschungszentrum (DRFZ), a Leibniz Institute, Charitéplatz 1, 10117, Berlin, Germany
- Institute of Chemistry and Biochemistry, Department of Biology, Chemistry and Pharmacy, Freie Universität Berlin, Berlin, Germany
| | - Johanna Ehl
- Department of Rheumatology and Clinical Immunology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 10117, Berlin, Germany
- Immune Dynamics, Deutsches Rheuma-Forschungszentrum (DRFZ), a Leibniz Institute, Charitéplatz 1, 10117, Berlin, Germany
| | - Ralf Köhler
- Immune Dynamics, Deutsches Rheuma-Forschungszentrum (DRFZ), a Leibniz Institute, Charitéplatz 1, 10117, Berlin, Germany
| | - Asylkhan Rakhymzhan
- Department of Rheumatology and Clinical Immunology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 10117, Berlin, Germany
- Biophysical Analytics, Deutsches Rheuma-Forschungszentrum (DRFZ), a Leibniz Institute, Charitéplatz 1, 10117, Berlin, Germany
| | - Andrew Woehler
- Berlin Institute for Medical Systems Biology, Max Delbrück Center for Molecular Medicine, 10115, Berlin, Germany
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA, 20147, USA
| | - Lukas Katthän
- Miltenyi Biotec B.V. and Co. Bertha-von-Suttner-Straße 5, 37085, Göttingen, Germany
| | - Gernot Ebel
- Miltenyi Biotec B.V. and Co. Bertha-von-Suttner-Straße 5, 37085, Göttingen, Germany
| | - Wjatscheslaw Liublin
- Biophysical Analytics, Deutsches Rheuma-Forschungszentrum (DRFZ), a Leibniz Institute, Charitéplatz 1, 10117, Berlin, Germany
| | - Ana Kasapi
- Department of Rheumatology and Clinical Immunology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 10117, Berlin, Germany
- Innate Immunity in Rheumatic Diseases, Deutsches Rheuma-Forschungszentrum (DRFZ), a Leibniz Institute, Charitéplatz 1, 10117, Berlin, Germany
| | - Antigoni Triantafyllopoulou
- Department of Rheumatology and Clinical Immunology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 10117, Berlin, Germany
- Innate Immunity in Rheumatic Diseases, Deutsches Rheuma-Forschungszentrum (DRFZ), a Leibniz Institute, Charitéplatz 1, 10117, Berlin, Germany
| | - Tim Julius Schulz
- Department of Adipocyte Development and Nutrition, German Institute of Human Nutrition (DIfE) Potsdam-Rehbruecke, 14558, Nuthetal, Germany
- German Center for Diabetes Research (DZD), 85764, Munich-Neuherberg, Germany
| | - Raluca Aura Niesner
- Biophysical Analytics, Deutsches Rheuma-Forschungszentrum (DRFZ), a Leibniz Institute, Charitéplatz 1, 10117, Berlin, Germany
- Dynamic and Functional in vivo Imaging, Veterinary Medicine, Freie Universität Berlin, Berlin, Germany
| | - Anja Erika Hauser
- Department of Rheumatology and Clinical Immunology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 10117, Berlin, Germany.
- Immune Dynamics, Deutsches Rheuma-Forschungszentrum (DRFZ), a Leibniz Institute, Charitéplatz 1, 10117, Berlin, Germany.
| |
Collapse
|
44
|
Wang Y, Ye L. TESOS: an integrated approach for uniform mesoscale imaging. Cell Res 2024; 34:93-94. [PMID: 38168641 PMCID: PMC10837420 DOI: 10.1038/s41422-023-00888-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2024] Open
Affiliation(s)
- Yu Wang
- Department of Neuroscience and Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA, USA
| | - Li Ye
- Department of Neuroscience and Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA, USA.
| |
Collapse
|
45
|
Yi Y, Li Y, Zhang S, Men Y, Wang Y, Jing D, Ding J, Zhu Q, Chen Z, Chen X, Li JL, Wang Y, Wang J, Peng H, Zhang L, Luo W, Feng JQ, He Y, Ge WP, Zhao H. Mapping of individual sensory nerve axons from digits to spinal cord with the transparent embedding solvent system. Cell Res 2024; 34:124-139. [PMID: 38168640 PMCID: PMC10837210 DOI: 10.1038/s41422-023-00867-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Accepted: 08/07/2023] [Indexed: 01/05/2024] Open
Abstract
Achieving uniform optical resolution for a large tissue sample is a major challenge for deep imaging. For conventional tissue clearing methods, loss of resolution and quality in deep regions is inevitable due to limited transparency. Here we describe the Transparent Embedding Solvent System (TESOS) method, which combines tissue clearing, transparent embedding, sectioning and block-face imaging. We used TESOS to acquire volumetric images of uniform resolution for an adult mouse whole-body sample. The TESOS method is highly versatile and can be combined with different microscopy systems to achieve uniformly high resolution. With a light sheet microscope, we imaged the whole body of an adult mouse, including skin, at a uniform 0.8 × 0.8 × 3.5 μm3 voxel resolution within 120 h. With a confocal microscope and a 40×/1.3 numerical aperture objective, we achieved a uniform sub-micron resolution in the whole sample to reveal a complete projection of individual nerve axons within the central or peripheral nervous system. Furthermore, TESOS allowed the first mesoscale connectome mapping of individual sensory neuron axons spanning 5 cm from adult mouse digits to the spinal cord at a uniform sub-micron resolution.
Collapse
Affiliation(s)
- Yating Yi
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
- Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
- Chinese Institute for Brain Research, Beijing, China
| | - Youqi Li
- Chinese Institute for Brain Research, Beijing, China
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Shiwen Zhang
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
- Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Yi Men
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
- Department of Head and Neck Oncology, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Yuhong Wang
- West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Dian Jing
- Department of Orthodontics, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine; College of Stomatology, Shanghai Jiao Tong University; National Center for Stomatology; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology; Shanghai Research Institute of Stomatology, Shanghai, China
| | - Jiayi Ding
- Chinese Institute for Brain Research, Beijing, China
- Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, China
| | - Qingjie Zhu
- Chinese Institute for Brain Research, Beijing, China
| | - Zexi Chen
- Chinese Institute for Brain Research, Beijing, China
| | - Xingjun Chen
- Chinese Institute for Brain Research, Beijing, China
- Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, China
| | - Jun-Liszt Li
- Chinese Institute for Brain Research, Beijing, China
- Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, China
| | - Yilong Wang
- Chinese Institute for Brain Research, Beijing, China
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Jun Wang
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
- Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Hanchuan Peng
- SEU-ALLEN Joint Center, Institute for Brain and Intelligence, Southeast University, Nanjing, Jiangsu, China
| | - Li Zhang
- Chinese Institute for Brain Research, Beijing, China
| | | | - Jian Q Feng
- Texas A&M University, College of Dentistry, Dallas, TX, USA
| | - Yongwen He
- Qujing Medical College, Qujing, Yunnan, China.
| | - Woo-Ping Ge
- Chinese Institute for Brain Research, Beijing, China.
| | - Hu Zhao
- Chinese Institute for Brain Research, Beijing, China.
| |
Collapse
|
46
|
Gritti N, Power RM, Graves A, Huisken J. Image restoration of degraded time-lapse microscopy data mediated by near-infrared imaging. Nat Methods 2024; 21:311-321. [PMID: 38177507 PMCID: PMC10864180 DOI: 10.1038/s41592-023-02127-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Accepted: 11/10/2023] [Indexed: 01/06/2024]
Abstract
Time-lapse fluorescence microscopy is key to unraveling biological development and function; however, living systems, by their nature, permit only limited interrogation and contain untapped information that can only be captured by more invasive methods. Deep-tissue live imaging presents a particular challenge owing to the spectral range of live-cell imaging probes/fluorescent proteins, which offer only modest optical penetration into scattering tissues. Herein, we employ convolutional neural networks to augment live-imaging data with deep-tissue images taken on fixed samples. We demonstrate that convolutional neural networks may be used to restore deep-tissue contrast in GFP-based time-lapse imaging using paired final-state datasets acquired using near-infrared dyes, an approach termed InfraRed-mediated Image Restoration (IR2). Notably, the networks are remarkably robust over a wide range of developmental times. We employ IR2 to enhance the information content of green fluorescent protein time-lapse images of zebrafish and Drosophila embryo/larval development and demonstrate its quantitative potential in increasing the fidelity of cell tracking/lineaging in developing pescoids. Thus, IR2 is poised to extend live imaging to depths otherwise inaccessible.
Collapse
Affiliation(s)
- Nicola Gritti
- Morgridge Institute for Research, Madison, WI, USA
- Mesoscopic Imaging Facility, European Molecular Biology Laboratory Barcelona, Barcelona, Spain
| | - Rory M Power
- Morgridge Institute for Research, Madison, WI, USA
- EMBL Imaging Center, European Molecular Biology Laboratory Heidelberg, Heidelberg, Germany
| | | | - Jan Huisken
- Morgridge Institute for Research, Madison, WI, USA.
- Department of Integrative Biology, University of Wisconsin Madison, Madison, WI, USA.
- Department of Biology and Psychology, Georg-August-University Göttingen, Göttingen, Germany.
- Cluster of Excellence 'Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells' (MBExC), University of Göttingen, Göttingen, Germany.
| |
Collapse
|
47
|
Rebejac J, Eme-Scolan E, Rua R. Role of meningeal immunity in brain function and protection against pathogens. J Inflamm (Lond) 2024; 21:3. [PMID: 38291415 PMCID: PMC10829400 DOI: 10.1186/s12950-023-00374-7] [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: 10/31/2023] [Accepted: 12/22/2023] [Indexed: 02/01/2024] Open
Abstract
The brain and spinal cord collectively referred to as the Central Nervous System (CNS) are protected by the blood-brain barrier that limits molecular, microbial and immunological trafficking. However, in the last decade, many studies have emphasized the protective role of 'border regions' at the surface of the CNS which are highly immunologically active, in contrast with the CNS parenchyma. In the steady-state, lymphoid and myeloid cells residing in the cranial meninges can affect brain function and behavior. Upon infection, they provide a first layer of protection against microbial neuroinvasion. The maturation of border sites over time enables more effective brain protection in adults as compared to neonates. Here, we provide a comprehensive update on the meningeal immune system and its role in physiological brain function and protection against infectious agents.
Collapse
Affiliation(s)
- Julie Rebejac
- Centre d'Immunologie de Marseille-Luminy, Aix Marseille Université, Inserm, CNRS, Marseille, France
| | - Elisa Eme-Scolan
- Centre d'Immunologie de Marseille-Luminy, Aix Marseille Université, Inserm, CNRS, Marseille, France
| | - Rejane Rua
- Centre d'Immunologie de Marseille-Luminy, Aix Marseille Université, Inserm, CNRS, Marseille, France.
| |
Collapse
|
48
|
Gold L, Barci E, Brendel M, Orth M, Cheng J, Kirchleitner SV, Bartos LM, Pötter D, Kirchner MA, Unterrainer LM, Kaiser L, Ziegler S, Weidner L, Riemenschneider MJ, Unterrainer M, Belka C, Tonn JC, Bartenstein P, Niyazi M, von Baumgarten L, Kälin RE, Glass R, Lauber K, Albert NL, Holzgreve A. The Traumatic Inoculation Process Affects TSPO Radioligand Uptake in Experimental Orthotopic Glioblastoma. Biomedicines 2024; 12:188. [PMID: 38255293 PMCID: PMC10813339 DOI: 10.3390/biomedicines12010188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Revised: 01/10/2024] [Accepted: 01/11/2024] [Indexed: 01/24/2024] Open
Abstract
BACKGROUND The translocator protein (TSPO) has been proven to have great potential as a target for the positron emission tomography (PET) imaging of glioblastoma. However, there is an ongoing debate about the potential various sources of the TSPO PET signal. This work investigates the impact of the inoculation-driven immune response on the PET signal in experimental orthotopic glioblastoma. METHODS Serial [18F]GE-180 and O-(2-[18F]fluoroethyl)-L-tyrosine ([18F]FET) PET scans were performed at day 7/8 and day 14/15 after the inoculation of GL261 mouse glioblastoma cells (n = 24) or saline (sham, n = 6) into the right striatum of immunocompetent C57BL/6 mice. An additional n = 25 sham mice underwent [18F]GE-180 PET and/or autoradiography (ARG) at days 7, 14, 21, 28, 35, 50 and 90 in order to monitor potential reactive processes that were solely related to the inoculation procedure. In vivo imaging results were directly compared to tissue-based analyses including ARG and immunohistochemistry. RESULTS We found that the inoculation process represents an immunogenic event, which significantly contributes to TSPO radioligand uptake. [18F]GE-180 uptake in GL261-bearing mice surpassed [18F]FET uptake both in the extent and the intensity, e.g., mean target-to-background ratio (TBRmean) in PET at day 7/8: 1.22 for [18F]GE-180 vs. 1.04 for [18F]FET, p < 0.001. Sham mice showed increased [18F]GE-180 uptake at the inoculation channel, which, however, continuously decreased over time (e.g., TBRmean in PET: 1.20 at day 7 vs. 1.09 at day 35, p = 0.04). At the inoculation channel, the percentage of TSPO/IBA1 co-staining decreased, whereas TSPO/GFAP (glial fibrillary acidic protein) co-staining increased over time (p < 0.001). CONCLUSION We identify the inoculation-driven immune response to be a relevant contributor to the PET signal and add a new aspect to consider for planning PET imaging studies in orthotopic glioblastoma models.
Collapse
Affiliation(s)
- Lukas Gold
- Department of Nuclear Medicine, LMU University Hospital, LMU Munich, Marchioninistr. 15, 81377 Munich, Germany; (L.G.)
| | - Enio Barci
- Department of Nuclear Medicine, LMU University Hospital, LMU Munich, Marchioninistr. 15, 81377 Munich, Germany; (L.G.)
- Neurosurgical Research, Department of Neurosurgery, LMU University Hospital, LMU Munich, Marchioninistr. 15, 81377 Munich, Germany
| | - Matthias Brendel
- Department of Nuclear Medicine, LMU University Hospital, LMU Munich, Marchioninistr. 15, 81377 Munich, Germany; (L.G.)
- Munich Cluster for Systems Neurology (SyNergy), LMU Munich, 81377 Munich, Germany
| | - Michael Orth
- Department of Radiation Oncology, LMU University Hospital, LMU Munich, Marchioninistr. 15, 81377 Munich, Germany
- Department of Radiation Oncology, University Hospital Tübingen, 72076 Tübingen, Germany
| | - Jiying Cheng
- Neurosurgical Research, Department of Neurosurgery, LMU University Hospital, LMU Munich, Marchioninistr. 15, 81377 Munich, Germany
| | - Sabrina V. Kirchleitner
- Department of Neurosurgery, LMU University Hospital, LMU Munich, Marchioninistr 15, 81377 Munich, Germany
| | - Laura M. Bartos
- Department of Nuclear Medicine, LMU University Hospital, LMU Munich, Marchioninistr. 15, 81377 Munich, Germany; (L.G.)
| | - Dennis Pötter
- Department of Nuclear Medicine, LMU University Hospital, LMU Munich, Marchioninistr. 15, 81377 Munich, Germany; (L.G.)
| | - Maximilian A. Kirchner
- Department of Nuclear Medicine, LMU University Hospital, LMU Munich, Marchioninistr. 15, 81377 Munich, Germany; (L.G.)
| | - Lena M. Unterrainer
- Department of Nuclear Medicine, LMU University Hospital, LMU Munich, Marchioninistr. 15, 81377 Munich, Germany; (L.G.)
| | - Lena Kaiser
- Department of Nuclear Medicine, LMU University Hospital, LMU Munich, Marchioninistr. 15, 81377 Munich, Germany; (L.G.)
| | - Sibylle Ziegler
- Department of Nuclear Medicine, LMU University Hospital, LMU Munich, Marchioninistr. 15, 81377 Munich, Germany; (L.G.)
| | - Lorraine Weidner
- Department of Neuropathology, Regensburg University Hospital, 93053 Regensburg, Germany
| | | | - Marcus Unterrainer
- Department of Nuclear Medicine, LMU University Hospital, LMU Munich, Marchioninistr. 15, 81377 Munich, Germany; (L.G.)
- DIE RADIOLOGIE, 80331 Munich, Germany
| | - Claus Belka
- Department of Radiation Oncology, LMU University Hospital, LMU Munich, Marchioninistr. 15, 81377 Munich, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, 81377 Munich, Germany
- Bavarian Cancer Research Center (BZKF), 81377 Munich, Germany
| | - Joerg-Christian Tonn
- Department of Neurosurgery, LMU University Hospital, LMU Munich, Marchioninistr 15, 81377 Munich, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, 81377 Munich, Germany
| | - Peter Bartenstein
- Department of Nuclear Medicine, LMU University Hospital, LMU Munich, Marchioninistr. 15, 81377 Munich, Germany; (L.G.)
- Munich Cluster for Systems Neurology (SyNergy), LMU Munich, 81377 Munich, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, 81377 Munich, Germany
| | - Maximilian Niyazi
- Department of Radiation Oncology, LMU University Hospital, LMU Munich, Marchioninistr. 15, 81377 Munich, Germany
- Department of Radiation Oncology, University Hospital Tübingen, 72076 Tübingen, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, 81377 Munich, Germany
- Bavarian Cancer Research Center (BZKF), 81377 Munich, Germany
| | - Louisa von Baumgarten
- Department of Neurosurgery, LMU University Hospital, LMU Munich, Marchioninistr 15, 81377 Munich, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, 81377 Munich, Germany
- Bavarian Cancer Research Center (BZKF), 81377 Munich, Germany
| | - Roland E. Kälin
- Neurosurgical Research, Department of Neurosurgery, LMU University Hospital, LMU Munich, Marchioninistr. 15, 81377 Munich, Germany
| | - Rainer Glass
- Neurosurgical Research, Department of Neurosurgery, LMU University Hospital, LMU Munich, Marchioninistr. 15, 81377 Munich, Germany
| | - Kirsten Lauber
- Department of Radiation Oncology, LMU University Hospital, LMU Munich, Marchioninistr. 15, 81377 Munich, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, 81377 Munich, Germany
| | - Nathalie L. Albert
- Department of Nuclear Medicine, LMU University Hospital, LMU Munich, Marchioninistr. 15, 81377 Munich, Germany; (L.G.)
- German Cancer Consortium (DKTK), Partner Site Munich, 81377 Munich, Germany
- Bavarian Cancer Research Center (BZKF), 81377 Munich, Germany
| | - Adrien Holzgreve
- Department of Nuclear Medicine, LMU University Hospital, LMU Munich, Marchioninistr. 15, 81377 Munich, Germany; (L.G.)
| |
Collapse
|
49
|
Okar SV, Fagiani F, Absinta M, Reich DS. Imaging of brain barrier inflammation and brain fluid drainage in human neurological diseases. Cell Mol Life Sci 2024; 81:31. [PMID: 38212566 PMCID: PMC10838199 DOI: 10.1007/s00018-023-05073-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2023] [Revised: 11/22/2023] [Accepted: 11/29/2023] [Indexed: 01/13/2024]
Abstract
The intricate relationship between the central nervous system (CNS) and the immune system plays a crucial role in the pathogenesis of various neurological diseases. Understanding the interactions among the immunopathological processes at the brain borders is essential for advancing our knowledge of disease mechanisms and developing novel diagnostic and therapeutic approaches. In this review, we explore the emerging role of neuroimaging in providing valuable insights into brain barrier inflammation and brain fluid drainage in human neurological diseases. Neuroimaging techniques have enabled us not only to visualize and assess brain structures, but also to study the dynamics of the CNS in health and disease in vivo. By analyzing imaging findings, we can gain a deeper understanding of the immunopathology observed at the brain-immune interface barriers, which serve as critical gatekeepers that regulate immune cell trafficking, cytokine release, and clearance of waste products from the brain. This review explores the integration of neuroimaging data with immunopathological findings, providing valuable insights into brain barrier integrity and immune responses in neurological diseases. Such integration may lead to the development of novel diagnostic markers and targeted therapeutic approaches that can benefit patients with neurological disorders.
Collapse
Affiliation(s)
- Serhat V Okar
- Translational Neuroradiology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Francesca Fagiani
- Translational Neuropathology Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, 20132, Milan, Italy
| | - Martina Absinta
- Translational Neuropathology Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, 20132, Milan, Italy.
- Division of Neuroscience, Vita-Salute San Raffaele University, 20132, Milan, Italy.
| | - Daniel S Reich
- Translational Neuroradiology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, 20892, USA.
| |
Collapse
|
50
|
Voigt FF, Reuss AM, Naert T, Hildebrand S, Schaettin M, Hotz AL, Whitehead L, Bahl A, Neuhauss SCF, Roebroeck A, Stoeckli ET, Lienkamp SS, Aguzzi A, Helmchen F. Reflective multi-immersion microscope objectives inspired by the Schmidt telescope. Nat Biotechnol 2024; 42:65-71. [PMID: 36997681 PMCID: PMC10791577 DOI: 10.1038/s41587-023-01717-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Accepted: 02/20/2023] [Indexed: 04/03/2023]
Abstract
Imaging large, cleared samples requires microscope objectives that combine a large field of view (FOV) with a long working distance (WD) and a high numerical aperture (NA). Ideally, such objectives should be compatible with a wide range of immersion media, which is challenging to achieve with conventional lens-based objective designs. Here we introduce the multi-immersion 'Schmidt objective' consisting of a spherical mirror and an aspherical correction plate as a solution to this problem. We demonstrate that a multi-photon variant of the Schmidt objective is compatible with all homogeneous immersion media and achieves an NA of 1.08 at a refractive index of 1.56, 1.1-mm FOV and 11-mm WD. We highlight its versatility by imaging cleared samples in various media ranging from air and water to benzyl alcohol/benzyl benzoate, dibenzyl ether and ethyl cinnamate and by imaging of neuronal activity in larval zebrafish in vivo. In principle, the concept can be extended to any imaging modality, including wide-field, confocal and light-sheet microscopy.
Collapse
Affiliation(s)
- Fabian F Voigt
- Brain Research Institute, University of Zurich, Zurich, Switzerland.
- Neuroscience Center Zurich, University of Zurich, Zurich, Switzerland.
- Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA, USA.
| | - Anna Maria Reuss
- Neuroscience Center Zurich, University of Zurich, Zurich, Switzerland
- Institute of Neuropathology, University Hospital Zurich, Zurich, Switzerland
| | - Thomas Naert
- Institute of Anatomy, University of Zurich, Zurich, Switzerland
| | - Sven Hildebrand
- Department of Cognitive Neuroscience, Faculty of Psychology & Neuroscience, Maastricht University, Maastricht, the Netherlands
| | - Martina Schaettin
- Neuroscience Center Zurich, University of Zurich, Zurich, Switzerland
- Department of Molecular Life Sciences, University of Zurich, Zurich, Switzerland
| | - Adriana L Hotz
- Neuroscience Center Zurich, University of Zurich, Zurich, Switzerland
- Department of Molecular Life Sciences, University of Zurich, Zurich, Switzerland
| | - Lachlan Whitehead
- Walter and Eliza Hall Institute of Medical Research, Melbourne, Victoria, Australia
- Department of Medical Biology, University of Melbourne, Melbourne, Victoria, Australia
| | - Armin Bahl
- Centre for the Advanced Study of Collective Behaviour, University of Konstanz, Konstanz, Germany
| | - Stephan C F Neuhauss
- Neuroscience Center Zurich, University of Zurich, Zurich, Switzerland
- Department of Molecular Life Sciences, University of Zurich, Zurich, Switzerland
| | - Alard Roebroeck
- Department of Cognitive Neuroscience, Faculty of Psychology & Neuroscience, Maastricht University, Maastricht, the Netherlands
| | - Esther T Stoeckli
- Neuroscience Center Zurich, University of Zurich, Zurich, Switzerland
- Department of Molecular Life Sciences, University of Zurich, Zurich, Switzerland
- University Research Priority Program (URPP), Adaptive Brain Circuits in Development and Learning (AdaBD), University of Zürich, Zurich, Switzerland
| | | | - Adriano Aguzzi
- Neuroscience Center Zurich, University of Zurich, Zurich, Switzerland
- Institute of Neuropathology, University Hospital Zurich, Zurich, Switzerland
| | - Fritjof Helmchen
- Brain Research Institute, University of Zurich, Zurich, Switzerland
- Neuroscience Center Zurich, University of Zurich, Zurich, Switzerland
- University Research Priority Program (URPP), Adaptive Brain Circuits in Development and Learning (AdaBD), University of Zürich, Zurich, Switzerland
| |
Collapse
|