1
|
Hao H, Wang H, Wang X, Ding X, Zhang S, Pan CF, Rahman MA, Ling T, Li H, Tan J, Yang JKW, Lu W, Liu J, Hu G. Single-Shot 3D Imaging Meta-Microscope. NANO LETTERS 2024; 24:13364-13373. [PMID: 39393017 DOI: 10.1021/acs.nanolett.4c03952] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/13/2024]
Abstract
Three-dimensional (3D) imaging enables high-precision and high-resolution axial positioning, which is crucial for biological imaging, semiconductor defect monitoring, and other applications. Conventional implementations rely on bulky optical elements or scanning mechanisms, resulting in low speed and complicated setups. Here, we generate the double-helix (DH) point spread function with an all-dielectric metasurface and thus innovate the 3D imaging microscope (hence dubbed meta-microscope), both in 4f and 2f imaging systems. The 4f-meta-microscope with a numerical aperture of 0.7 achieves an axial localization accuracy below 0.12 μm within a 15.47 μm detection range, while the 2f-DH meta-microscope with a numerical aperture of 0.3 shows a 1.12 μm accuracy within a 227.33 μm range. We also demonstrate single-shot and accurate 3D biological imaging of the mouse kidney tissue and peach anther, providing a comprehensive and efficient approach for 3D bioimaging and other applications through a single-shot 3D meta-microscope.
Collapse
Affiliation(s)
- Huijie Hao
- Advanced Microscopy and Instrumentation Research Center, School of Instrumentation Science and Engineering, Harbin Institute of Technology, Harbin 150080, China
| | - Hao Wang
- Engineering Product Development Pillar, Singapore University of Technology and Design, 8 Somapah Road, 487372 Singapore, Singapore
| | - Xinwei Wang
- School of Electrical and Electronic Engineering, Nanyang Technological University, 50 Nanyang Avenue Singapore, 639798 Singapore, Singapore
| | - Xumin Ding
- Advanced Microscopy and Instrumentation Research Center, School of Instrumentation Science and Engineering, Harbin Institute of Technology, Harbin 150080, China
- Key Lab of Ultra-Precision Intelligent Instrumentation (Harbin Institute of Technology), Ministry of Industry and Information Technology, Harbin 150080, China
| | - Shutao Zhang
- School of Electrical and Electronic Engineering, Nanyang Technological University, 50 Nanyang Avenue Singapore, 639798 Singapore, Singapore
- Institute of Materials Research and Engineering (IMRE), Agency for Science, Technology and Research (A*STAR), 2 Fusionopolis Way, Singapore 138634, Singapore
| | - Cheng-Feng Pan
- Engineering Product Development Pillar, Singapore University of Technology and Design, 8 Somapah Road, 487372 Singapore, Singapore
| | - Md Abdur Rahman
- Engineering Product Development Pillar, Singapore University of Technology and Design, 8 Somapah Road, 487372 Singapore, Singapore
| | - Tong Ling
- School of Electrical and Electronic Engineering, Nanyang Technological University, 50 Nanyang Avenue Singapore, 639798 Singapore, Singapore
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, Singapore 637457, Singapore
- SERI-NTU Advanced Ocular Engineering (STANCE) Program, Singapore 637553, Singapore
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore 169856, Singapore
| | - Haoyu Li
- Key Lab of Ultra-Precision Intelligent Instrumentation (Harbin Institute of Technology), Ministry of Industry and Information Technology, Harbin 150080, China
- Center of Ultra-Precision Optoelectronic Instrument Engineering, Harbin Institute of Technology, Harbin 150080, China
| | - Jiubin Tan
- Advanced Microscopy and Instrumentation Research Center, School of Instrumentation Science and Engineering, Harbin Institute of Technology, Harbin 150080, China
- Key Lab of Ultra-Precision Intelligent Instrumentation (Harbin Institute of Technology), Ministry of Industry and Information Technology, Harbin 150080, China
- Center of Ultra-Precision Optoelectronic Instrument Engineering, Harbin Institute of Technology, Harbin 150080, China
| | - Joel K W Yang
- Engineering Product Development Pillar, Singapore University of Technology and Design, 8 Somapah Road, 487372 Singapore, Singapore
- Singapore-HUJ Alliance for Research and Enterprise (SHARE), The Smart Grippers for Soft Robotics (SGSR) Programme, Campus for Research Excellence and Technological Enterprise (CREATE), Singapore 138602, Singapore
| | - Wenlong Lu
- State Key Laboratory of Digital Manufacturing Equipment and Technology, School of Mechanical Science and Engineering, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Jian Liu
- Advanced Microscopy and Instrumentation Research Center, School of Instrumentation Science and Engineering, Harbin Institute of Technology, Harbin 150080, China
- Key Lab of Ultra-Precision Intelligent Instrumentation (Harbin Institute of Technology), Ministry of Industry and Information Technology, Harbin 150080, China
| | - Guangwei Hu
- School of Electrical and Electronic Engineering, Nanyang Technological University, 50 Nanyang Avenue Singapore, 639798 Singapore, Singapore
| |
Collapse
|
2
|
Zhang Y, Wang M, Zhu Q, Guo Y, Liu B, Li J, Yao X, Kong C, Zhang Y, Huang Y, Qi H, Wu J, Guo ZV, Dai Q. Long-term mesoscale imaging of 3D intercellular dynamics across a mammalian organ. Cell 2024; 187:6104-6122.e25. [PMID: 39276776 DOI: 10.1016/j.cell.2024.08.026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Revised: 06/06/2024] [Accepted: 08/13/2024] [Indexed: 09/17/2024]
Abstract
A comprehensive understanding of physio-pathological processes necessitates non-invasive intravital three-dimensional (3D) imaging over varying spatial and temporal scales. However, huge data throughput, optical heterogeneity, surface irregularity, and phototoxicity pose great challenges, leading to an inevitable trade-off between volume size, resolution, speed, sample health, and system complexity. Here, we introduce a compact real-time, ultra-large-scale, high-resolution 3D mesoscope (RUSH3D), achieving uniform resolutions of 2.6 × 2.6 × 6 μm3 across a volume of 8,000 × 6,000 × 400 μm3 at 20 Hz with low phototoxicity. Through the integration of multiple computational imaging techniques, RUSH3D facilitates a 13-fold improvement in data throughput and an orders-of-magnitude reduction in system size and cost. With these advantages, we observed premovement neural activity and cross-day visual representational drift across the mouse cortex, the formation and progression of multiple germinal centers in mouse inguinal lymph nodes, and heterogeneous immune responses following traumatic brain injury-all at single-cell resolution, opening up a horizon for intravital mesoscale study of large-scale intercellular interactions at the organ level.
Collapse
Affiliation(s)
- Yuanlong Zhang
- Department of Automation, Tsinghua University, Beijing 100084, China; Institute for Brain and Cognitive Sciences, Tsinghua University, Beijing 100084, China; Beijing Key Laboratory of Multi-dimension & Multi-scale Computational Photography (MMCP), Tsinghua University, Beijing 100084, China; IDG/McGovern Institute for Brain Research, Tsinghua University, Beijing 100084, China
| | - Mingrui Wang
- Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen 518071, China
| | - Qiyu Zhu
- IDG/McGovern Institute for Brain Research, Tsinghua University, Beijing 100084, China; School of Basic Medical Sciences, Tsinghua University, Beijing 100084, China; Tsinghua-Peking Center for Life Sciences, Beijing 100084, China
| | - Yuduo Guo
- Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen 518071, China
| | - Bo Liu
- School of Basic Medical Sciences, Tsinghua University, Beijing 100084, China; Tsinghua-Peking Center for Life Sciences, Beijing 100084, China; Laboratory of Dynamic Immunobiology, Institute for Immunology, Tsinghua University, Beijing 100084, China
| | - Jiamin Li
- IDG/McGovern Institute for Brain Research, Tsinghua University, Beijing 100084, China; School of Basic Medical Sciences, Tsinghua University, Beijing 100084, China; Tsinghua-Peking Center for Life Sciences, Beijing 100084, China
| | - Xiao Yao
- IDG/McGovern Institute for Brain Research, Tsinghua University, Beijing 100084, China; School of Basic Medical Sciences, Tsinghua University, Beijing 100084, China; Tsinghua-Peking Center for Life Sciences, Beijing 100084, China
| | - Chui Kong
- School of Information Science and Technology, Fudan University, Shanghai 200433, China
| | - Yi Zhang
- Department of Automation, Tsinghua University, Beijing 100084, China; Institute for Brain and Cognitive Sciences, Tsinghua University, Beijing 100084, China; Beijing Key Laboratory of Multi-dimension & Multi-scale Computational Photography (MMCP), Tsinghua University, Beijing 100084, China; IDG/McGovern Institute for Brain Research, Tsinghua University, Beijing 100084, China
| | - Yuchao Huang
- IDG/McGovern Institute for Brain Research, Tsinghua University, Beijing 100084, China; School of Basic Medical Sciences, Tsinghua University, Beijing 100084, China; Tsinghua-Peking Center for Life Sciences, Beijing 100084, China
| | - Hai Qi
- School of Basic Medical Sciences, Tsinghua University, Beijing 100084, China; Tsinghua-Peking Center for Life Sciences, Beijing 100084, China; Laboratory of Dynamic Immunobiology, Institute for Immunology, Tsinghua University, Beijing 100084, China
| | - Jiamin Wu
- Department of Automation, Tsinghua University, Beijing 100084, China; Institute for Brain and Cognitive Sciences, Tsinghua University, Beijing 100084, China; Beijing Key Laboratory of Multi-dimension & Multi-scale Computational Photography (MMCP), Tsinghua University, Beijing 100084, China; IDG/McGovern Institute for Brain Research, Tsinghua University, Beijing 100084, China.
| | - Zengcai V Guo
- IDG/McGovern Institute for Brain Research, Tsinghua University, Beijing 100084, China; School of Basic Medical Sciences, Tsinghua University, Beijing 100084, China; Tsinghua-Peking Center for Life Sciences, Beijing 100084, China.
| | - Qionghai Dai
- Department of Automation, Tsinghua University, Beijing 100084, China; Institute for Brain and Cognitive Sciences, Tsinghua University, Beijing 100084, China; Beijing Key Laboratory of Multi-dimension & Multi-scale Computational Photography (MMCP), Tsinghua University, Beijing 100084, China; IDG/McGovern Institute for Brain Research, Tsinghua University, Beijing 100084, China.
| |
Collapse
|
3
|
Ruan X, Mueller M, Liu G, Görlitz F, Fu TM, Milkie DE, Lillvis JL, Kuhn A, Gan Chong J, Hong JL, Herr CYA, Hercule W, Nienhaus M, Killilea AN, Betzig E, Upadhyayula S. Image processing tools for petabyte-scale light sheet microscopy data. Nat Methods 2024:10.1038/s41592-024-02475-4. [PMID: 39420143 DOI: 10.1038/s41592-024-02475-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Accepted: 09/16/2024] [Indexed: 10/19/2024]
Abstract
Light sheet microscopy is a powerful technique for high-speed three-dimensional imaging of subcellular dynamics and large biological specimens. However, it often generates datasets ranging from hundreds of gigabytes to petabytes in size for a single experiment. Conventional computational tools process such images far slower than the time to acquire them and often fail outright due to memory limitations. To address these challenges, we present PetaKit5D, a scalable software solution for efficient petabyte-scale light sheet image processing. This software incorporates a suite of commonly used processing tools that are optimized for memory and performance. Notable advancements include rapid image readers and writers, fast and memory-efficient geometric transformations, high-performance Richardson-Lucy deconvolution and scalable Zarr-based stitching. These features outperform state-of-the-art methods by over one order of magnitude, enabling the processing of petabyte-scale image data at the full teravoxel rates of modern imaging cameras. The software opens new avenues for biological discoveries through large-scale imaging experiments.
Collapse
Affiliation(s)
- Xiongtao Ruan
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA, US.
| | - Matthew Mueller
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA, US
- Howard Hughes Medical Institute, Berkeley, CA, US
| | - Gaoxiang Liu
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA, US
| | - Frederik Görlitz
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA, US
- Department of Microsystems Engineering, University of Freiburg, Freiburg, Germany
| | - Tian-Ming Fu
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA, US
- Department of Electrical and Computer Engineering, Princeton University, Princeton, NJ, US
| | - Daniel E Milkie
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA, US
| | - Joshua L Lillvis
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA, US
| | | | - Johnny Gan Chong
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA, US
| | - Jason Li Hong
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA, US
| | - Chu Yi Aaron Herr
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA, US
| | - Wilmene Hercule
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA, US
| | | | - Alison N Killilea
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA, US
| | - Eric Betzig
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA, US.
- Howard Hughes Medical Institute, Berkeley, CA, US.
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA, US.
- Department of Physics, Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, CA, US.
| | - Srigokul Upadhyayula
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA, US.
- Molecular Biophysics and Integrated Bioimaging Division, Lawrence Berkeley National Laboratory, Berkeley, CA, US.
- Chan Zuckerberg Biohub, San Francisco, CA, US.
| |
Collapse
|
4
|
Ortkrass H, Müller M, Engdahl AK, Holst G, Huser T. High sensitivity cameras can lower spatial resolution in high-resolution optical microscopy. Nat Commun 2024; 15:8886. [PMID: 39406700 PMCID: PMC11480200 DOI: 10.1038/s41467-024-53198-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Accepted: 10/07/2024] [Indexed: 10/19/2024] Open
Abstract
High-resolution optical fluorescence microscopies and, in particular, super-resolution fluorescence microscopy, are rapidly adopting highly sensitive cameras as their preferred photodetectors. Camera-based parallel detection facilitates high-speed live cell imaging with the highest spatial resolution. Here, we show that the drive to use ever more sensitive, photon-counting image sensors in cameras can, however, have detrimental effects on the spatial resolution of the resulting images. This is particularly noticeable in applications that demand a high space-bandwidth product, where the image magnification is close to the Nyquist sampling limit of the sensor. Most scientists will often select image sensors based on parameters such as pixel size, quantum efficiency, signal-to-noise performance, dynamic range, and frame rate of the sensor. A parameter that is, however, typically overlooked is the sensor's modulation transfer function (MTF). We have determined the wavelength-specific MTF of front- and back-illuminated image sensors and evaluated how it affects the spatial resolution that can be achieved in high-resolution fluorescence microscopy modalities. We find significant differences in image sensor performance that cause the resulting spatial resolution to vary by up to 28%. This result shows that the choice of image sensor has a significant impact on the imaging performance of all camera-based optical microscopy modalities.
Collapse
Affiliation(s)
- Henning Ortkrass
- Biomolecular Photonics, Faculty of Physics, Bielefeld University, Bielefeld, Germany.
| | - Marcel Müller
- Biomolecular Photonics, Faculty of Physics, Bielefeld University, Bielefeld, Germany
| | | | | | - Thomas Huser
- Biomolecular Photonics, Faculty of Physics, Bielefeld University, Bielefeld, Germany.
| |
Collapse
|
5
|
Cunha I, Latron E, Bauer S, Sage D, Griffié J. Machine learning in microscopy - insights, opportunities and challenges. J Cell Sci 2024; 137:jcs262095. [PMID: 39465533 DOI: 10.1242/jcs.262095] [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] [Indexed: 10/29/2024] Open
Abstract
Machine learning (ML) is transforming the field of image processing and analysis, from automation of laborious tasks to open-ended exploration of visual patterns. This has striking implications for image-driven life science research, particularly microscopy. In this Review, we focus on the opportunities and challenges associated with applying ML-based pipelines for microscopy datasets from a user point of view. We investigate the significance of different data characteristics - quantity, transferability and content - and how this determines which ML model(s) to use, as well as their output(s). Within the context of cell biological questions and applications, we further discuss ML utility range, namely data curation, exploration, prediction and explanation, and what they entail and translate to in the context of microscopy. Finally, we explore the challenges, common artefacts and risks associated with ML in microscopy. Building on insights from other fields, we propose how these pitfalls might be mitigated for in microscopy.
Collapse
Affiliation(s)
- Inês Cunha
- Science for Life Laboratory, Department of Biochemistry and Biophysics, Stockholm University, Tomtebodavägen 23, 171 65 Solna, Sweden
| | - Emma Latron
- Science for Life Laboratory, Department of Biochemistry and Biophysics, Stockholm University, Tomtebodavägen 23, 171 65 Solna, Sweden
| | - Sebastian Bauer
- Science for Life Laboratory, Department of Biochemistry and Biophysics, Stockholm University, Tomtebodavägen 23, 171 65 Solna, Sweden
| | - Daniel Sage
- Biomedical Imaging Group and EPFL Center for Imaging, École Polytechnique, Rte Cantonale, 1015 Lausanne, Switzerland
| | - Juliette Griffié
- Science for Life Laboratory, Department of Biochemistry and Biophysics, Stockholm University, Tomtebodavägen 23, 171 65 Solna, Sweden
| |
Collapse
|
6
|
Yan G, Zhou J, Yin J, Gao D, Zhong X, Deng X, Kang H, Sun A. Membrane Ruffles: Composition, Function, Formation and Visualization. Int J Mol Sci 2024; 25:10971. [PMID: 39456754 PMCID: PMC11507850 DOI: 10.3390/ijms252010971] [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: 09/05/2024] [Revised: 10/09/2024] [Accepted: 10/10/2024] [Indexed: 10/28/2024] Open
Abstract
Membrane ruffles are cell actin-based membrane protrusions that have distinct structural characteristics. Linear ruffles with columnar spike-like and veil-like structures assemble at the leading edge of cell membranes. Circular dorsal ruffles (CDRs) have no supporting columnar structures but their veil-like structures, connecting from end to end, present an enclosed ring-shaped circular outline. Membrane ruffles are involved in multiple cell functions such as cell motility, macropinocytosis, receptor internalization, fluid viscosity sensing in a two-dimensional culture environment, and protecting cells from death in response to physiologically compressive loads. Herein, we review the state-of-the-art knowledge on membrane ruffle structure and function, the growth factor-induced membrane ruffling process, and the growth factor-independent ruffling mode triggered by calcium and other stimulating factors, together with the respective underlying mechanisms. We also summarize the inhibitors used in ruffle formation studies and their specificity. In the last part, an overview is given of the various techniques in which the membrane ruffles have been visualized up to now.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Hongyan Kang
- Key Laboratory of Biomechanics and Mechanobiology (Beihang University), Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing 100083, China; (G.Y.); (J.Z.); (J.Y.); (D.G.); (X.Z.); (X.D.)
| | - Anqiang Sun
- Key Laboratory of Biomechanics and Mechanobiology (Beihang University), Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing 100083, China; (G.Y.); (J.Z.); (J.Y.); (D.G.); (X.Z.); (X.D.)
| |
Collapse
|
7
|
Liu S, Alexander KD, Francis MM. Neural Circuit Remodeling: Mechanistic Insights from Invertebrates. J Dev Biol 2024; 12:27. [PMID: 39449319 PMCID: PMC11503349 DOI: 10.3390/jdb12040027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Revised: 10/07/2024] [Accepted: 10/10/2024] [Indexed: 10/26/2024] Open
Abstract
As nervous systems mature, neural circuit connections are reorganized to optimize the performance of specific functions in adults. This reorganization of connections is achieved through a remarkably conserved phase of developmental circuit remodeling that engages neuron-intrinsic and neuron-extrinsic molecular mechanisms to establish mature circuitry. Abnormalities in circuit remodeling and maturation are broadly linked with a variety of neurodevelopmental disorders, including autism spectrum disorders and schizophrenia. Here, we aim to provide an overview of recent advances in our understanding of the molecular processes that govern neural circuit remodeling and maturation. In particular, we focus on intriguing mechanistic insights gained from invertebrate systems, such as the nematode Caenorhabditis elegans and the fruit fly Drosophila melanogaster. We discuss how transcriptional control mechanisms, synaptic activity, and glial engulfment shape specific aspects of circuit remodeling in worms and flies. Finally, we highlight mechanistic parallels across invertebrate and mammalian systems, and prospects for further advances in each.
Collapse
Affiliation(s)
- Samuel Liu
- Department of Neurobiology, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
- Program in Neuroscience, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Kellianne D. Alexander
- Department of Neurobiology, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
- Program in Neuroscience, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Michael M. Francis
- Department of Neurobiology, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
- Program in Neuroscience, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| |
Collapse
|
8
|
Yildiz A. Mechanism and regulation of kinesin motors. Nat Rev Mol Cell Biol 2024:10.1038/s41580-024-00780-6. [PMID: 39394463 DOI: 10.1038/s41580-024-00780-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/04/2024] [Indexed: 10/13/2024]
Abstract
Kinesins are a diverse superfamily of microtubule-based motors that perform fundamental roles in intracellular transport, cytoskeletal dynamics and cell division. These motors share a characteristic motor domain that powers unidirectional motility and force generation along microtubules, and they possess unique tail domains that recruit accessory proteins and facilitate oligomerization, regulation and cargo recognition. The location, direction and timing of kinesin-driven processes are tightly regulated by various cofactors, adaptors, microtubule tracks and microtubule-associated proteins. This Review focuses on recent structural and functional studies that reveal how members of the kinesin superfamily use the energy of ATP hydrolysis to transport cargoes, depolymerize microtubules and regulate microtubule dynamics. I also survey how accessory proteins and post-translational modifications regulate the autoinhibition, cargo binding and motility of some of the best-studied kinesins. Despite much progress, the mechanism and regulation of kinesins are still emerging, and unresolved questions can now be tackled using newly developed approaches in biophysics and structural biology.
Collapse
Affiliation(s)
- Ahmet Yildiz
- Physics Department, University of California at Berkeley, Berkeley, CA, USA.
- Department of Molecular and Cellular Biology, University of California at Berkeley, Berkeley, CA, USA.
| |
Collapse
|
9
|
Kumar P, Mondal PP. Multicolor iLIFE (m-iLIFE) volume cytometry for high-throughput imaging of multiple organelles. Sci Rep 2024; 14:23798. [PMID: 39394224 PMCID: PMC11470118 DOI: 10.1038/s41598-024-73667-3] [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: 02/14/2024] [Accepted: 09/19/2024] [Indexed: 10/13/2024] Open
Abstract
To be able to resolve multiple organelles at high throughput is an incredible achievement. This will have immediate implications in a range of fields ranging from fundamental cell biology to translational medicine. To realize such a high-throughput multicolor interrogation modality, we have developed a light-sheet based flow imaging system that is capable of visualizing multiple sub-cellular components with organelle-level resolution. This is possible due to the unique optical design that combines an illumination system comprising two collinear light sheets illuminating the flowing cells and a dedicated dual-color 4f-detection, enabling simultaneous recording of multiple organelles. The system PSF sections up to 4 parallel microfluidic channels through which cells are flowing, and multicolor images of cell cross-sections are recorded. The data is then computationally processed (filtered using ML algorithm, shift-corrected, and merged) and combined to reconstruct the 3D multicolor volume. System testing is conducted using multicolor fluorescent nano-beads (size ∼ 175 nm) and flow-based imaging parameters (PSF size, motion-blur, flow rate, frame rate, and number of cell-sections) are determined for quality imaging. Drug treatment studies were carried out for healthy and cancerous HeLa cells to check the performance of the proposed system. The cells were treated with a drug (Vincristine, which is known to promote mitochondrial fission in cells), and the same is compared with untreated control cells. The proposed multicolor iLIFE system could screen ∼ 800 cells/min (at a flow speed of 2490 μ m/s), and the drug treatment studies were carried out up to 24 h. Studies showed the disintegration of mitochondrial network and dysfunctional lysosomes and their accumulation at the cell membrane, which is a clear indication of cell apoptosis. Compared to control cells (untreated), the mortality is highest at a concentration of 500 nM post 12 h of drug treatment. With the capability of multiorganelle interrogation and organelle-level resolution, the multicolor iLIFE cytometry system is suitably placed to assist optical imaging and biomedical research.
Collapse
Affiliation(s)
- Prashant Kumar
- Mondal Lab, Department of Instrumentation and Applied Physics, Indian Institute of Science, Bangalore, 560012, India
| | - Partha Pratim Mondal
- Mondal Lab, Department of Instrumentation and Applied Physics, Indian Institute of Science, Bangalore, 560012, India.
- Centre for Cryogenic Technology, Indian Institute of Science, Bangalore, 560012, India.
| |
Collapse
|
10
|
Mazzella L, Mangeat T, Giroussens G, Rogez B, Li H, Creff J, Saadaoui M, Martins C, Bouzignac R, Labouesse S, Idier J, Galland F, Allain M, Sentenac A, LeGoff L. Extended-depth of field random illumination microscopy, EDF-RIM, provides super-resolved projective imaging. LIGHT, SCIENCE & APPLICATIONS 2024; 13:285. [PMID: 39384765 PMCID: PMC11479626 DOI: 10.1038/s41377-024-01612-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 08/15/2024] [Accepted: 08/27/2024] [Indexed: 10/11/2024]
Abstract
The ultimate aim of fluorescence microscopy is to achieve high-resolution imaging of increasingly larger biological samples. Extended depth of field presents a potential solution to accelerate imaging of large samples when compression of information along the optical axis is not detrimental to the interpretation of images. We have implemented an extended depth of field (EDF) approach in a random illumination microscope (RIM). RIM uses multiple speckled illuminations and variance data processing to double the resolution. It is particularly adapted to the imaging of thick samples as it does not require the knowledge of illumination patterns. We demonstrate highly-resolved projective images of biological tissues and cells. Compared to a sequential scan of the imaged volume with conventional 2D-RIM, EDF-RIM allows an order of magnitude improvement in speed and light dose reduction, with comparable resolution. As the axial information is lost in an EDF modality, we propose a method to retrieve the sample topography for samples that are organized in cell sheets.
Collapse
Affiliation(s)
- Lorry Mazzella
- Aix Marseille Université, CNRS, Centrale Med, Institut Fresnel UMR7249, Turing Center for Living Systems, Marseille, France
| | - Thomas Mangeat
- LITC Core Facility, Centre de Biologie Integrative (CBI), CNRS, Université de Toulouse, UT3, Toulouse, France
| | - Guillaume Giroussens
- Aix Marseille Université, CNRS, Centrale Med, Institut Fresnel UMR7249, Turing Center for Living Systems, Marseille, France
| | - Benoit Rogez
- Aix Marseille Université, CNRS, Centrale Med, Institut Fresnel UMR7249, Turing Center for Living Systems, Marseille, France
| | - Hao Li
- MCD, Centre de Biologie Intégrative (CBI), CNRS, Université de Toulouse, UT3, Toulouse, France
| | - Justine Creff
- MCD, Centre de Biologie Intégrative (CBI), CNRS, Université de Toulouse, UT3, Toulouse, France
| | - Mehdi Saadaoui
- Aix Marseille University, CNRS, IBDM UMR7288, Turing Centre for Living Systems, Marseille, France
| | - Carla Martins
- Aix Marseille Université, CNRS, Centrale Med, Institut Fresnel UMR7249, Turing Center for Living Systems, Marseille, France
| | - Ronan Bouzignac
- MCD, Centre de Biologie Intégrative (CBI), CNRS, Université de Toulouse, UT3, Toulouse, France
| | - Simon Labouesse
- LITC Core Facility, Centre de Biologie Integrative (CBI), CNRS, Université de Toulouse, UT3, Toulouse, France
| | - Jérome Idier
- LS2N, CNRS UMR 6004, F44321, Nantes Cedex 3, France
| | - Frédéric Galland
- Aix Marseille Université, CNRS, Centrale Med, Institut Fresnel UMR7249, Turing Center for Living Systems, Marseille, France
| | - Marc Allain
- Aix Marseille Université, CNRS, Centrale Med, Institut Fresnel UMR7249, Turing Center for Living Systems, Marseille, France
| | - Anne Sentenac
- Aix Marseille Université, CNRS, Centrale Med, Institut Fresnel UMR7249, Turing Center for Living Systems, Marseille, France.
| | - Loïc LeGoff
- Aix Marseille Université, CNRS, Centrale Med, Institut Fresnel UMR7249, Turing Center for Living Systems, Marseille, France.
| |
Collapse
|
11
|
Wang Z, Hakozaki H, McMahon G, Medina-Carbonero M, Schöneberg J. LiveLattice: Real-time visualisation of tilted light-sheet microscopy data using a memory-efficient transformation algorithm. J Microsc 2024. [PMID: 39360400 DOI: 10.1111/jmi.13358] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Revised: 09/02/2024] [Accepted: 09/03/2024] [Indexed: 10/04/2024]
Abstract
Light-sheet fluorescence microscopy (LSFM), a prominent fluorescence microscopy technique, offers enhanced temporal resolution for imaging biological samples in four dimensions (4D; x, y, z, time). Some of the most recent implementations, including inverted selective plane illumination microscopy (iSPIM) and lattice light-sheet microscopy (LLSM), move the sample substrate at an oblique angle relative to the detection objective's optical axis. Data from such tilted-sample-scan LSFMs require subsequent deskewing and rotation for proper visualisation and analysis. Such data preprocessing operations currently demand substantial memory allocation and pose significant computational challenges for large 4D dataset. The consequence is prolonged data preprocessing time compared to data acquisition time, which limits the ability for live-viewing the data as it is being captured by the microscope. To enable the fast preprocessing of large light-sheet microscopy datasets without significant hardware demand, we have developed WH-Transform, a memory-efficient transformation algorithm for deskewing and rotating the raw dataset, significantly reducing memory usage and the run time by more than 10-fold for large image stacks. Benchmarked against the conventional method and existing software, our approach demonstrates linear runtime compared to the cubic and quadratic runtime of the other approaches. Preprocessing a raw 3D volume of 2 GB (512 × 1536 × 600 pixels) can be accomplished in 3 s using a GPU with 24 GB of memory on a single workstation. Applied to 4D LLSM datasets of human hepatocytes, lung organoid tissue and brain organoid tissue, our method provided rapid and accurate preprocessing within seconds. Importantly, such preprocessing speeds now allow visualisation of the raw microscope data stream in real time, significantly improving the usability of LLSM in biology. In summary, this advancement holds transformative potential for light-sheet microscopy, enabling real-time, on-the-fly data preprocessing, visualisation, and analysis on standard workstations, thereby revolutionising biological imaging applications for LLSM and similar microscopes.
Collapse
Affiliation(s)
- Zichen Wang
- Department of Pharmacology, University of California, San Diego, California, USA
- Department of Chemistry and Biochemistry, University of California, San Diego, California, USA
| | - Hiroyuki Hakozaki
- Department of Pharmacology, University of California, San Diego, California, USA
- Department of Chemistry and Biochemistry, University of California, San Diego, California, USA
| | - Gillian McMahon
- Department of Pharmacology, University of California, San Diego, California, USA
- Department of Chemistry and Biochemistry, University of California, San Diego, California, USA
| | - Marta Medina-Carbonero
- Department of Pharmacology, University of California, San Diego, California, USA
- Department of Chemistry and Biochemistry, University of California, San Diego, California, USA
| | - Johannes Schöneberg
- Department of Pharmacology, University of California, San Diego, California, USA
- Department of Chemistry and Biochemistry, University of California, San Diego, California, USA
| |
Collapse
|
12
|
Hockenberry MA, Daugird TA, Legant WR. Cell dynamics revealed by microscopy advances. Curr Opin Cell Biol 2024; 90:102418. [PMID: 39159598 PMCID: PMC11392612 DOI: 10.1016/j.ceb.2024.102418] [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: 05/01/2024] [Revised: 07/23/2024] [Accepted: 07/24/2024] [Indexed: 08/21/2024]
Abstract
Cell biology emerges from spatiotemporally coordinated molecular processes. Recent advances in live-cell microscopy, fueled by a surge in optical, molecular, and computational technologies, have enabled dynamic observations from single molecules to whole organisms. Despite technological leaps, there is still an untapped opportunity to fully leverage their capabilities toward biological insight. We highlight how single-molecule imaging has transformed our understanding of biological processes, with a focus on chromatin organization and transcription in the nucleus. We describe how this was enabled by the close integration of new imaging techniques with analysis tools and discuss the challenges to make a comparable impact at larger scales from organelles to organisms. By highlighting recent successful examples, we describe an outlook of ever-increasing data and the need for seamless integration between dataset visualization and quantification to realize the full potential warranted by advances in new imaging technologies.
Collapse
Affiliation(s)
- Max A Hockenberry
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, Chapel Hill, NC, USA
| | - Timothy A Daugird
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Wesley R Legant
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA; Joint Department of Biomedical Engineering, North Carolina State University, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
| |
Collapse
|
13
|
Rudinskiy M, Morone D, Molinari M. Fluorescent Reporters, Imaging, and Artificial Intelligence Toolkits to Monitor and Quantify Autophagy, Heterophagy, and Lysosomal Trafficking Fluxes. Traffic 2024; 25:e12957. [PMID: 39450581 DOI: 10.1111/tra.12957] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 08/21/2024] [Accepted: 10/03/2024] [Indexed: 10/26/2024]
Abstract
Lysosomal compartments control the clearance of cell-own material (autophagy) or of material that cells endocytose from the external environment (heterophagy) to warrant supply of nutrients, to eliminate macromolecules or parts of organelles present in excess, aged, or containing toxic material. Inherited or sporadic mutations in lysosomal proteins and enzymes may hamper their folding in the endoplasmic reticulum (ER) and their lysosomal transport via the Golgi compartment, resulting in lysosomal dysfunction and storage disorders. Defective cargo delivery to lysosomal compartments is harmful to cells and organs since it causes accumulation of toxic compounds and defective organellar homeostasis. Assessment of resident proteins and cargo fluxes to the lysosomal compartments is crucial for the mechanistic dissection of intracellular transport and catabolic events. It might be combined with high-throughput screenings to identify cellular, chemical, or pharmacological modulators of these events that may find therapeutic use for autophagy-related and lysosomal storage disorders. Here, discuss qualitative, quantitative and chronologic monitoring of autophagic, heterophagic and lysosomal protein trafficking in fixed and live cells, which relies on fluorescent single and tandem reporters used in combination with biochemical, flow cytometry, light and electron microscopy approaches implemented by artificial intelligence-based technology.
Collapse
Affiliation(s)
- Mikhail Rudinskiy
- Università della Svizzera italiana, Lugano, Switzerland
- Institute for Research in Biomedicine, Bellinzona, Switzerland
- Department of Biology, Swiss Federal Institute of Technology, Zurich, Switzerland
| | - Diego Morone
- Università della Svizzera italiana, Lugano, Switzerland
- Institute for Research in Biomedicine, Bellinzona, Switzerland
- Graduate School for Cellular and Biomedical Sciences, University of Bern, Bern, Switzerland
| | - Maurizio Molinari
- Università della Svizzera italiana, Lugano, Switzerland
- Institute for Research in Biomedicine, Bellinzona, Switzerland
- École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| |
Collapse
|
14
|
Jiao Z, Pan M, Yousaf K, Doveiko D, Maclean M, Griffin D, Chen Y, Li DDU. Smartphone-based optical sectioning (SOS) microscopy with a telecentric design for fluorescence imaging. J Microsc 2024; 296:10-23. [PMID: 38808665 DOI: 10.1111/jmi.13334] [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: 10/17/2023] [Revised: 04/15/2024] [Accepted: 05/17/2024] [Indexed: 05/30/2024]
Abstract
We propose a smartphone-based optical sectioning (SOS) microscope based on the HiLo technique, with a single smartphone replacing a high-cost illumination source and a camera sensor. We built our SOS with off-the-shelf optical, mechanical cage systems with 3D-printed adapters to seamlessly integrate the smartphone with the SOS main body. The liquid light guide can be integrated with the adapter, guiding the smartphone's LED light to the digital mirror device (DMD) with neglectable loss. We used an electrically tuneable lens (ETL) instead of a mechanical translation stage to realise low-cost axial scanning. The ETL was conjugated to the objective lens's back pupil plane (BPP) to construct a telecentric design by a 4f configuration to maintain the lateral magnification for different axial positions. SOS has a 571.5 µm telecentric scanning range and an 11.7 µm axial resolution. The broadband smartphone LED torch can effectively excite fluorescent polystyrene (PS) beads. We successfully used SOS for high-contrast fluorescent PS beads imaging with different wavelengths and optical sectioning imaging of multilayer fluorescent PS beads. To our knowledge, the proposed SOS is the first smartphone-based HiLo optical sectioning microscopy (£1965), which can save around £7035 compared with a traditional HiLo system (£9000). It is a powerful tool for biomedical research in resource-limited areas.
Collapse
Affiliation(s)
- Ziao Jiao
- Department of Biomedical Engineering, University of Strathclyde, Glasgow, Scotland, UK
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, Scotland, UK
| | - Mingliang Pan
- Department of Biomedical Engineering, University of Strathclyde, Glasgow, Scotland, UK
| | - Khadija Yousaf
- Department of Physics, University of Strathclyde, Glasgow, Scotland, UK
| | - Daniel Doveiko
- Department of Physics, University of Strathclyde, Glasgow, Scotland, UK
| | - Michelle Maclean
- Department of Biomedical Engineering, University of Strathclyde, Glasgow, Scotland, UK
- Department of Electronic & Electrical Engineering, The Robertson Trust Laboratory for Electronic Sterilisation Technologies (ROLEST), University of Strathclyde, Glasgow, UK
| | - David Griffin
- Department of Biomedical Engineering, University of Strathclyde, Glasgow, Scotland, UK
| | - Yu Chen
- Department of Physics, University of Strathclyde, Glasgow, Scotland, UK
| | - David Day Uei Li
- Department of Biomedical Engineering, University of Strathclyde, Glasgow, Scotland, UK
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, Scotland, UK
| |
Collapse
|
15
|
Pesce L, Ricci P, Sportelli G, Belcari N, Sancataldo G. Expansion and Light-Sheet Microscopy for Nanoscale 3D Imaging. SMALL METHODS 2024; 8:e2301715. [PMID: 38461540 DOI: 10.1002/smtd.202301715] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Revised: 02/10/2024] [Indexed: 03/12/2024]
Abstract
Expansion Microscopy (ExM) and Light-Sheet Fluorescence Microscopy (LSFM) are forefront imaging techniques that enable high-resolution visualization of biological specimens. ExM enhances nanoscale investigation using conventional fluorescence microscopes, while LSFM offers rapid, minimally invasive imaging over large volumes. This review explores the joint advancements of ExM and LSFM, focusing on the excellent performance of the integrated modality obtained from the combination of the two, which is refer to as ExLSFM. In doing so, the chemical processes required for ExM, the tailored optical setup of LSFM for examining expanded samples, and the adjustments in sample preparation for accurate data collection are emphasized. It is delve into various specimen types studied using this integrated method and assess its potential for future applications. The goal of this literature review is to enrich the comprehension of ExM and LSFM, encouraging their wider use and ongoing development, looking forward to the upcoming challenges, and anticipating innovations in these imaging techniques.
Collapse
Affiliation(s)
- Luca Pesce
- Department of Physics - Enrico Fermi, University of Pisa, Largo Pontecorvo, 3, Pisa, 56127, Italy
| | - Pietro Ricci
- Department of Applied Physics, University of Barcelona, C/Martí i Franquès, 1, Barcelona, 08028, Spain
| | - Giancarlo Sportelli
- Department of Physics - Enrico Fermi, University of Pisa, Largo Pontecorvo, 3, Pisa, 56127, Italy
| | - Nicola Belcari
- Department of Physics - Enrico Fermi, University of Pisa, Largo Pontecorvo, 3, Pisa, 56127, Italy
| | - Giuseppe Sancataldo
- Department of Physics - Emilio Segrè, University of Palermo, Viale delle Scienze, 18, Palermo, 90128, Italy
| |
Collapse
|
16
|
Takáč T, Kuběnová L, Šamajová O, Dvořák P, Řehák J, Haberland J, Bundschuh ST, Pechan T, Tomančák P, Ovečka M, Šamaj J. Actin cytoskeleton and plasma membrane aquaporins are involved in different drought response of Arabidopsis rhd2 and der1 root hair mutants. PLANT PHYSIOLOGY AND BIOCHEMISTRY : PPB 2024; 216:109137. [PMID: 39357201 DOI: 10.1016/j.plaphy.2024.109137] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Revised: 09/17/2024] [Accepted: 09/19/2024] [Indexed: 10/04/2024]
Abstract
Actin cytoskeleton and reactive oxygen species are principal determinants of root hair polarity and tip growth. Loss of function in RESPIRATORY BURST OXIDASE HOMOLOG C/ROOT HAIR DEFECTIVE 2 (AtRBOHC/RHD2), an NADPH oxidase emitting superoxide to the apoplast, and in ACTIN 2, a vegetative actin isovariant, in rhd2-1 and der1-3 mutants, respectively, lead to similar defects in root hair formation and elongation Since early endosome-mediated polar localization of AtRBOHC/RHD2 depends on actin cytoskeleton, comparing the proteome-wide consequences of both mutations might be of eminent interest. Therefore, we employed a differential proteomic analysis of Arabidopsis rhd2-1 and der1-3 mutants. Both mutants exhibited substantial alterations in abundances of stress-related proteins. Notably, plasma membrane (PM)-localized PIP aquaporins showed contrasting abundance patterns in the mutants compared to wild-types. Drought-responsive proteins were mostly downregulated in rhd2-1 but upregulated in der1-3. Proteomic data suggest that opposite to der1-3, altered vesicular transport in rhd2-1 mutant likely contributes to the deregulation of PM-localized proteins, including PIPs. Moreover, lattice light sheet microscopy revealed reduced actin dynamics in rhd2-1 roots, a finding contrasting with previous reports on der1-3 mutant. Phenotypic experiments demonstrated a drought stress susceptibility in rhd2-1 and resistance in der1-3. Thus, mutations in AtRBOHC/RHD2 and ACTIN2 cause similar root hair defects, but they differently affect the actin cytoskeleton and vesicular transport. Reduced actin dynamics in rhd2-1 mutant is accompanied by alteration of vesicular transport proteins abundance, likely leading to altered protein delivery to PM, including aquaporins, thereby significantly affecting drought stress responses.
Collapse
Affiliation(s)
- Tomáš Takáč
- Department of Biotechnology, Faculty of Science, Palacký University Olomouc, Olomouc, Czech Republic
| | - Lenka Kuběnová
- Department of Biotechnology, Faculty of Science, Palacký University Olomouc, Olomouc, Czech Republic
| | - Olga Šamajová
- Department of Biotechnology, Faculty of Science, Palacký University Olomouc, Olomouc, Czech Republic
| | - Petr Dvořák
- Department of Biotechnology, Faculty of Science, Palacký University Olomouc, Olomouc, Czech Republic
| | - Jan Řehák
- Department of Biotechnology, Faculty of Science, Palacký University Olomouc, Olomouc, Czech Republic
| | - Jan Haberland
- Department of Biotechnology, Faculty of Science, Palacký University Olomouc, Olomouc, Czech Republic
| | | | - Tibor Pechan
- Institute for Genomics, Biocomputing and Biotechnology, Mississippi Agricultural and Forestry Experiment Station, Mississippi State University, Starkville, MS, United States
| | - Pavel Tomančák
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | - Miroslav Ovečka
- Department of Biotechnology, Faculty of Science, Palacký University Olomouc, Olomouc, Czech Republic
| | - Jozef Šamaj
- Department of Biotechnology, Faculty of Science, Palacký University Olomouc, Olomouc, Czech Republic.
| |
Collapse
|
17
|
You Y, Lin S, Tang C, Li Y, Yan D, Wang D, Chen X. Dual-/multi-organelle-targeted AIE probes associated with oxidative stress for biomedical applications. J Mater Chem B 2024; 12:8812-8824. [PMID: 39150370 DOI: 10.1039/d4tb01440e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/17/2024]
Abstract
In situ monitoring of biological processes between different organelles upon oxidative stress is one of the most important research hotspots. Fluorescence imaging is especially suitable for biomedical applications due to its distinct advantages of high spatiotemporal resolution, high sensitivity, non-invasiveness, and in situ monitoring capabilities. However, most fluorescent probes can only achieve light-up imaging of single organelles, thus the combined use of two or more probes is usually required for monitoring biological processes between organelles, which can suffer from tedious staining and washing procedures, increased cytotoxicity and poor photostability. Exogenetic oxidants can affect broad-spectrum subcellular organelles, which are not conducive to in situ monitoring of biological processes between specific organelles. To tackle these challenges, a series of dual-/multi-organelle-targeted aggregation-induced emission (AIE) probes associated with oxidative stress have been designed and developed in the past few years. Herein, the recent progress of these AIE probes is summarized in biomedical applications, such as apoptosis monitoring, interplay between organelles, microenvironmental changes of organelles, organelle morphology tracking, precise cancer therapy, and so forth. Moreover, the further outlook for dual-/multi-organelle-targeted AIE probes is discussed, aiming to promote innovative research in biomedical applications.
Collapse
Affiliation(s)
- Yuanyuan You
- School of Pharmacy, Guangdong Medical University, Dongguan, 523808, China.
| | - Songling Lin
- School of Pharmacy, Guangdong Medical University, Dongguan, 523808, China.
| | - Chengwei Tang
- Institute of Laboratory Medicine, School of Medical Technology, Guangdong Medical University, Dongguan, 523808, China.
| | - Yuchao Li
- Institute of Laboratory Medicine, School of Medical Technology, Guangdong Medical University, Dongguan, 523808, China.
| | - Dingyuan Yan
- Center for AIE Research, College of Materials Science and Engineering, Shenzhen University, Shenzhen 518060, China
| | - Dong Wang
- Center for AIE Research, College of Materials Science and Engineering, Shenzhen University, Shenzhen 518060, China
| | - Xiaohui Chen
- Institute of Laboratory Medicine, School of Medical Technology, Guangdong Medical University, Dongguan, 523808, China.
| |
Collapse
|
18
|
Saliba N, Gagliano G, Gustavsson AK. Whole-cell multi-target single-molecule super-resolution imaging in 3D with microfluidics and a single-objective tilted light sheet. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.09.27.559876. [PMID: 37808751 PMCID: PMC10557638 DOI: 10.1101/2023.09.27.559876] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/10/2023]
Abstract
Multi-target single-molecule super-resolution fluorescence microscopy offers a powerful means of understanding the distributions and interplay between multiple subcellular structures at the nanoscale. However, single-molecule super-resolution imaging of whole mammalian cells is often hampered by high fluorescence background and slow acquisition speeds, especially when imaging multiple targets in 3D. In this work, we have mitigated these issues by developing a steerable, dithered, single-objective tilted light sheet for optical sectioning to reduce fluorescence background and a pipeline for 3D nanoprinting microfluidic systems for reflection of the light sheet into the sample. This easily adaptable novel microfluidic fabrication pipeline allows for the incorporation of reflective optics into microfluidic channels without disrupting efficient and automated solution exchange. By combining these innovations with point spread function engineering for nanoscale localization of individual molecules in 3D, deep learning for analysis of overlapping emitters, active 3D stabilization for drift correction and long-term imaging, and Exchange-PAINT for sequential multi-target imaging without chromatic offsets, we demonstrate whole-cell multi-target 3D single-molecule super-resolution imaging with improved precision and imaging speed.
Collapse
Affiliation(s)
- Nahima Saliba
- Department of Chemistry, Rice University, Houston, TX, 77005
| | - Gabriella Gagliano
- Department of Chemistry, Rice University, Houston, TX, 77005
- Smalley-Curl Institute, Rice University, Houston, TX, 77005
- Applied Physics Program, Rice University, Houston, TX, 77005
| | - Anna-Karin Gustavsson
- Department of Chemistry, Rice University, Houston, TX, 77005
- Smalley-Curl Institute, Rice University, Houston, TX, 77005
- Department of BioSciences, Rice University, Houston, TX, 77005
- Department of Electrical and Computer Engineering, Rice University, Houston, TX, 77005
- Center for Nanoscale Imaging Sciences, Rice University, Houston, TX, 77005
- Department of Cancer Biology, University of Texas MD Anderson Cancer Center, Houston, TX, 77030
| |
Collapse
|
19
|
Lamon S, Yu H, Zhang Q, Gu M. Lanthanide ion-doped upconversion nanoparticles for low-energy super-resolution applications. LIGHT, SCIENCE & APPLICATIONS 2024; 13:252. [PMID: 39277593 PMCID: PMC11401911 DOI: 10.1038/s41377-024-01547-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Revised: 05/31/2024] [Accepted: 07/22/2024] [Indexed: 09/17/2024]
Abstract
Energy-intensive technologies and high-precision research require energy-efficient techniques and materials. Lens-based optical microscopy technology is useful for low-energy applications in the life sciences and other fields of technology, but standard techniques cannot achieve applications at the nanoscale because of light diffraction. Far-field super-resolution techniques have broken beyond the light diffraction limit, enabling 3D applications down to the molecular scale and striving to reduce energy use. Typically targeted super-resolution techniques have achieved high resolution, but the high light intensity needed to outperform competing optical transitions in nanomaterials may result in photo-damage and high energy consumption. Great efforts have been made in the development of nanomaterials to improve the resolution and efficiency of these techniques toward low-energy super-resolution applications. Lanthanide ion-doped upconversion nanoparticles that exhibit multiple long-lived excited energy states and emit upconversion luminescence have enabled the development of targeted super-resolution techniques that need low-intensity light. The use of lanthanide ion-doped upconversion nanoparticles in these techniques for emerging low-energy super-resolution applications will have a significant impact on life sciences and other areas of technology. In this review, we describe the dynamics of lanthanide ion-doped upconversion nanoparticles for super-resolution under low-intensity light and their use in targeted super-resolution techniques. We highlight low-energy super-resolution applications of lanthanide ion-doped upconversion nanoparticles, as well as the related research directions and challenges. Our aim is to analyze targeted super-resolution techniques using lanthanide ion-doped upconversion nanoparticles, emphasizing fundamental mechanisms governing transitions in lanthanide ions to surpass the diffraction limit with low-intensity light, and exploring their implications for low-energy nanoscale applications.
Collapse
Affiliation(s)
- Simone Lamon
- School of Artificial Intelligence Science and Technology, University of Shanghai for Science and Technology, 200093, Shanghai, China.
- Institute of Photonic Chips, University of Shanghai for Science and Technology, 200093, Shanghai, China.
| | - Haoyi Yu
- School of Artificial Intelligence Science and Technology, University of Shanghai for Science and Technology, 200093, Shanghai, China
- Institute of Photonic Chips, University of Shanghai for Science and Technology, 200093, Shanghai, China
| | - Qiming Zhang
- School of Artificial Intelligence Science and Technology, University of Shanghai for Science and Technology, 200093, Shanghai, China
- Institute of Photonic Chips, University of Shanghai for Science and Technology, 200093, Shanghai, China
| | - Min Gu
- School of Artificial Intelligence Science and Technology, University of Shanghai for Science and Technology, 200093, Shanghai, China.
- Institute of Photonic Chips, University of Shanghai for Science and Technology, 200093, Shanghai, China.
| |
Collapse
|
20
|
Kramer SN, Antarasen J, Reinholt CR, Kisley L. A practical guide to light-sheet microscopy for nanoscale imaging: Looking beyond the cell. JOURNAL OF APPLIED PHYSICS 2024; 136:091101. [PMID: 39247785 PMCID: PMC11380115 DOI: 10.1063/5.0218262] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Accepted: 08/12/2024] [Indexed: 09/10/2024]
Abstract
We present a comprehensive guide to light-sheet microscopy (LSM) to assist scientists in navigating the practical implementation of this microscopy technique. Emphasizing the applicability of LSM to image both static microscale and nanoscale features, as well as diffusion dynamics, we present the fundamental concepts of microscopy, progressing through beam profile considerations, to image reconstruction. We outline key practical decisions in constructing a home-built system and provide insight into the alignment and calibration processes. We briefly discuss the conditions necessary for constructing a continuous 3D image and introduce our home-built code for data analysis. By providing this guide, we aim to alleviate the challenges associated with designing and constructing LSM systems and offer scientists new to LSM a valuable resource in navigating this complex field.
Collapse
Affiliation(s)
- Stephanie N Kramer
- Department of Physics, Case Western Reserve University, Rockefeller Building, 2076 Adelbert Road, Cleveland, Ohio 44106, USA
| | - Jeanpun Antarasen
- Department of Physics, Case Western Reserve University, Rockefeller Building, 2076 Adelbert Road, Cleveland, Ohio 44106, USA
| | - Cole R Reinholt
- Department of Physics, Case Western Reserve University, Rockefeller Building, 2076 Adelbert Road, Cleveland, Ohio 44106, USA
| | | |
Collapse
|
21
|
Shih CP, Tang WC, Chen P, Chen BC. Applications of Lightsheet Fluorescence Microscopy by High Numerical Aperture Detection Lens. J Phys Chem B 2024; 128:8273-8289. [PMID: 39177503 PMCID: PMC11382282 DOI: 10.1021/acs.jpcb.4c01721] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/24/2024]
Abstract
This Review explores the evolution, improvements, and recent applications of Light Sheet Fluorescence Microscopy (LSFM) in biological research using a high numerical aperture detection objective (lens) for imaging subcellular structures. The Review begins with an overview of the development of LSFM, tracing its evolution from its inception to its current state and emphasizing key milestones and technological advancements over the years. Subsequently, we will discuss various improvements of LSFM techniques, covering advancements in hardware such as illumination strategies, optical designs, and sample preparation methods that have enhanced imaging capabilities and resolution. The advancements in data acquisition and processing are also included, which provides a brief overview of the recent development of artificial intelligence. Fluorescence probes that were commonly used in LSFM will be highlighted, together with some insights regarding the selection of potential probe candidates for future LSFM development. Furthermore, we also discuss recent advances in the application of LSFM with a focus on high numerical aperture detection objectives for various biological studies. For sample preparation techniques, there are discussions regarding fluorescence probe selection, tissue clearing protocols, and some insights into expansion microscopy. Integrated setups such as adaptive optics, single objective modification, and microfluidics will also be some of the key discussion points in this Review. We hope that this comprehensive Review will provide a holistic perspective on the historical development, technical enhancements, and cutting-edge applications of LSFM, showcasing its pivotal role and future potential in advancing biological research.
Collapse
Affiliation(s)
- Chun-Pei Shih
- Institute of Physics, Academia Sinica, Taipei 11529, Taiwan
- Department of Chemistry, National Taiwan University, Taipei 106319, Taiwan
- Nano Science and Technology Program, Taiwan International Graduate Program, Academia Sinica and National Taiwan University, Taipei 11529, Taiwan
| | - Wei-Chun Tang
- Research Center for Applied Sciences, Academia Sinica, Taipei 11529, Taiwan
| | - Peilin Chen
- Institute of Physics, Academia Sinica, Taipei 11529, Taiwan
- Research Center for Applied Sciences, Academia Sinica, Taipei 11529, Taiwan
| | - Bi-Chang Chen
- Research Center for Applied Sciences, Academia Sinica, Taipei 11529, Taiwan
| |
Collapse
|
22
|
Chow DJX, Schartner EP, Corsetti S, Upadhya A, Morizet J, Gunn-Moore FJ, Dunning KR, Dholakia K. Quantifying DNA damage following light sheet and confocal imaging of the mammalian embryo. Sci Rep 2024; 14:20760. [PMID: 39237572 PMCID: PMC11377761 DOI: 10.1038/s41598-024-71443-x] [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: 12/22/2023] [Accepted: 08/28/2024] [Indexed: 09/07/2024] Open
Abstract
Embryo quality assessment by optical imaging is increasing in popularity. Among available optical techniques, light sheet microscopy has emerged as a superior alternative to confocal microscopy due to its geometry, enabling faster image acquisition with reduced photodamage to the sample. However, previous assessments of photodamage induced by imaging may have failed to measure more subtle impacts. In this study, we employed DNA damage as a sensitive indicator of photodamage. We use light sheet microscopy with excitation at a wavelength of 405 nm for imaging embryo autofluorescence and compare its performance to laser scanning confocal microscopy. At an equivalent signal-to-noise ratio for images acquired with both modalities, light sheet microscopy reduced image acquisition time by ten-fold, and did not induce DNA damage when compared to non-imaged embryos. In contrast, imaging with confocal microscopy led to significantly higher levels of DNA damage within embryos and had a higher photobleaching rate. Light sheet imaging is also capable of inducing DNA damage within the embryo but requires multiple cycles of volumetric imaging. Collectively, this study confirms that light sheet microscopy is faster and safer than confocal microscopy for imaging live embryos, indicating its potential as a label-free diagnostic for embryo quality.
Collapse
Affiliation(s)
- Darren J X Chow
- Robinson Research Institute, School of Biomedicine, The University of Adelaide, Adelaide, Australia
- Institute for Photonics and Advanced Sensing, The University of Adelaide, Adelaide, Australia
- Centre of Light for Life, The University of Adelaide, Adelaide, Australia
| | - Erik P Schartner
- Institute for Photonics and Advanced Sensing, The University of Adelaide, Adelaide, Australia
| | - Stella Corsetti
- SUPA, School of Physics and Astronomy, University of St Andrews, North Haugh, St Andrews, Fife, UK.
| | - Avinash Upadhya
- Institute for Photonics and Advanced Sensing, The University of Adelaide, Adelaide, Australia
- School of Biological Sciences, The University of Adelaide, Adelaide, Australia
- Centre of Light for Life, The University of Adelaide, Adelaide, Australia
| | - Josephine Morizet
- SUPA, School of Physics and Astronomy, University of St Andrews, North Haugh, St Andrews, Fife, UK
| | - Frank J Gunn-Moore
- School of Biology, University of St Andrews, North Haugh, St Andrews, Fife, UK
| | - Kylie R Dunning
- Robinson Research Institute, School of Biomedicine, The University of Adelaide, Adelaide, Australia
- Institute for Photonics and Advanced Sensing, The University of Adelaide, Adelaide, Australia
- Centre of Light for Life, The University of Adelaide, Adelaide, Australia
| | - Kishan Dholakia
- School of Biological Sciences, The University of Adelaide, Adelaide, Australia.
- Centre of Light for Life, The University of Adelaide, Adelaide, Australia.
- SUPA, School of Physics and Astronomy, University of St Andrews, North Haugh, St Andrews, Fife, UK.
| |
Collapse
|
23
|
Bragantini J, Theodoro I, Zhao X, Huijben TAPM, Hirata-Miyasaki E, VijayKumar S, Balasubramanian A, Lao T, Agrawal R, Xiao S, Lammerding J, Mehta S, Falcão AX, Jacobo A, Lange M, Royer LA. Ultrack: pushing the limits of cell tracking across biological scales. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.02.610652. [PMID: 39282368 PMCID: PMC11398427 DOI: 10.1101/2024.09.02.610652] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 09/22/2024]
Abstract
Tracking live cells across 2D, 3D, and multi-channel time-lapse recordings is crucial for understanding tissue-scale biological processes. Despite advancements in imaging technology, achieving accurate cell tracking remains challenging, particularly in complex and crowded tissues where cell segmentation is often ambiguous. We present Ultrack, a versatile and scalable cell-tracking method that tackles this challenge by considering candidate segmentations derived from multiple algorithms and parameter sets. Ultrack employs temporal consistency to select optimal segments, ensuring robust performance even under segmentation uncertainty. We validate our method on diverse datasets, including terabyte-scale developmental time-lapses of zebrafish, fruit fly, and nematode embryos, as well as multi-color and label-free cellular imaging. We show that Ultrack achieves state-of-the-art performance on the Cell Tracking Challenge and demonstrates superior accuracy in tracking densely packed embryonic cells over extended periods. Moreover, we propose an approach to tracking validation via dual-channel sparse labeling that enables high-fidelity ground truth generation, pushing the boundaries of long-term cell tracking assessment. Our method is freely available as a Python package with Fiji and napari plugins and can be deployed in a high-performance computing environment, facilitating widespread adoption by the research community.
Collapse
Affiliation(s)
| | - Ilan Theodoro
- Chan Zuckerberg Biohub, San Francisco, United States
- Institute of Computing - State University of Campinas, Campinas, Brazil
| | - Xiang Zhao
- Chan Zuckerberg Biohub, San Francisco, United States
| | | | | | | | | | - Tiger Lao
- Chan Zuckerberg Biohub, San Francisco, United States
| | - Richa Agrawal
- Weill Institute for Cell and Molecular Biology - Cornell University, Ithaca, United States
| | - Sheng Xiao
- Chan Zuckerberg Biohub, San Francisco, United States
| | - Jan Lammerding
- Weill Institute for Cell and Molecular Biology - Cornell University, Ithaca, United States
- Meinig School of Biomedical Engineering - Cornell University, Ithaca, United States
| | - Shalin Mehta
- Chan Zuckerberg Biohub, San Francisco, United States
| | | | - Adrian Jacobo
- Chan Zuckerberg Biohub, San Francisco, United States
| | - Merlin Lange
- Chan Zuckerberg Biohub, San Francisco, United States
| | - Loïc A Royer
- Chan Zuckerberg Biohub, San Francisco, United States
| |
Collapse
|
24
|
Lin J, Mehra D, Marin Z, Wang X, Borges HM, Shen Q, Gałecki S, Haug J, Abbott DH, Dean KM. Mechanically sheared axially swept light-sheet microscopy. BIOMEDICAL OPTICS EXPRESS 2024; 15:5314-5327. [PMID: 39296406 PMCID: PMC11407235 DOI: 10.1364/boe.526145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Revised: 07/06/2024] [Accepted: 07/23/2024] [Indexed: 09/21/2024]
Abstract
We present a mechanically sheared image acquisition format for upright and open-top light-sheet microscopes that automatically places data in its proper spatial context. This approach, which reduces computational post-processing and eliminates unnecessary interpolation or duplication of the data, is demonstrated on an upright variant of axially swept light-sheet microscopy (ASLM) that achieves a field of view, measuring 774 × 435 microns, that is 3.2-fold larger than previous models and a raw and isotropic resolution of ∼460 nm. Combined, we demonstrate the power of this approach by imaging sub-diffraction beads, cleared biological tissues, and expanded specimens.
Collapse
Affiliation(s)
- Jinlong Lin
- Lyda Hill Department of Bioinformatics, UT Southwestern Medical Center, 6000 Harry Hines Blvd, Dallas, TX 75390, USA
- Cecil H. and Ida Green Center for Systems Biology, UT Southwestern Medical Center, 6000 Harry Hines Blvd, Dallas, TX 75390, USA
| | - Dushyant Mehra
- Lyda Hill Department of Bioinformatics, UT Southwestern Medical Center, 6000 Harry Hines Blvd, Dallas, TX 75390, USA
- Cecil H. and Ida Green Center for Systems Biology, UT Southwestern Medical Center, 6000 Harry Hines Blvd, Dallas, TX 75390, USA
| | - Zach Marin
- Lyda Hill Department of Bioinformatics, UT Southwestern Medical Center, 6000 Harry Hines Blvd, Dallas, TX 75390, USA
- Cecil H. and Ida Green Center for Systems Biology, UT Southwestern Medical Center, 6000 Harry Hines Blvd, Dallas, TX 75390, USA
- Max Perutz Labs, Department of Structural and Computational Biology, University of Vienna, Dr. Bohr-Gasse 9, 1030 Vienna, Austria
| | - Xiaoding Wang
- Lyda Hill Department of Bioinformatics, UT Southwestern Medical Center, 6000 Harry Hines Blvd, Dallas, TX 75390, USA
- Cecil H. and Ida Green Center for Systems Biology, UT Southwestern Medical Center, 6000 Harry Hines Blvd, Dallas, TX 75390, USA
| | - Hazel M. Borges
- Lyda Hill Department of Bioinformatics, UT Southwestern Medical Center, 6000 Harry Hines Blvd, Dallas, TX 75390, USA
- Cecil H. and Ida Green Center for Systems Biology, UT Southwestern Medical Center, 6000 Harry Hines Blvd, Dallas, TX 75390, USA
| | - Qionghua Shen
- Lyda Hill Department of Bioinformatics, UT Southwestern Medical Center, 6000 Harry Hines Blvd, Dallas, TX 75390, USA
- Cecil H. and Ida Green Center for Systems Biology, UT Southwestern Medical Center, 6000 Harry Hines Blvd, Dallas, TX 75390, USA
| | - Seweryn Gałecki
- Lyda Hill Department of Bioinformatics, UT Southwestern Medical Center, 6000 Harry Hines Blvd, Dallas, TX 75390, USA
- Cecil H. and Ida Green Center for Systems Biology, UT Southwestern Medical Center, 6000 Harry Hines Blvd, Dallas, TX 75390, USA
- Department of Systems Biology and Engineering, Silesian University of Technology, Akademicka 16, 44-100 Gliwice, Poland
| | - John Haug
- Lyda Hill Department of Bioinformatics, UT Southwestern Medical Center, 6000 Harry Hines Blvd, Dallas, TX 75390, USA
- Cecil H. and Ida Green Center for Systems Biology, UT Southwestern Medical Center, 6000 Harry Hines Blvd, Dallas, TX 75390, USA
| | - Derek H. Abbott
- Lyda Hill Department of Bioinformatics, UT Southwestern Medical Center, 6000 Harry Hines Blvd, Dallas, TX 75390, USA
- Cecil H. and Ida Green Center for Systems Biology, UT Southwestern Medical Center, 6000 Harry Hines Blvd, Dallas, TX 75390, USA
| | - Kevin M. Dean
- Lyda Hill Department of Bioinformatics, UT Southwestern Medical Center, 6000 Harry Hines Blvd, Dallas, TX 75390, USA
- Cecil H. and Ida Green Center for Systems Biology, UT Southwestern Medical Center, 6000 Harry Hines Blvd, Dallas, TX 75390, USA
| |
Collapse
|
25
|
Chen Y, Chauhan S, Gong C, Dayton H, Xu C, De La Cruz ED, Tsai YYW, Datta MS, Rosoklija GB, Dwork AJ, Mann JJ, Boldrini M, Leong KW, Dietrich LEP, Tomer R. Low-cost and scalable projected light-sheet microscopy for the high-resolution imaging of cleared tissue and living samples. Nat Biomed Eng 2024; 8:1109-1123. [PMID: 39209948 DOI: 10.1038/s41551-024-01249-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2023] [Accepted: 08/02/2024] [Indexed: 09/04/2024]
Abstract
Light-sheet fluorescence microscopy (LSFM) is a widely used technique for imaging cleared tissue and living samples. However, high-performance LSFM systems are typically expensive and not easily scalable. Here we introduce a low-cost, scalable and versatile LSFM framework, which we named 'projected light-sheet microscopy' (pLSM), with high imaging performance and small device and computational footprints. We characterized the capabilities of pLSM, which repurposes readily available consumer-grade components, optimized optics, over-network control architecture and software-driven light-sheet modulation, by performing high-resolution mapping of cleared mouse brains and of post-mortem pathological human brain samples, and via the molecular phenotyping of brain and blood-vessel organoids derived from human induced pluripotent stem cells. We also report a method that leverages pLSM for the live imaging of the dynamics of sparsely labelled multi-layered bacterial pellicle biofilms at an air-liquid interface. pLSM can make high-resolution LSFM for biomedical applications more accessible, affordable and scalable.
Collapse
Affiliation(s)
- Yannan Chen
- Department of Biological Sciences, Columbia University, New York, NY, USA
- Department of Biomedical Engineering, Columbia University, New York, NY, USA
| | - Shradha Chauhan
- Department of Biological Sciences, Columbia University, New York, NY, USA
| | - Cheng Gong
- Department of Biological Sciences, Columbia University, New York, NY, USA
- Department of Biomedical Engineering, Columbia University, New York, NY, USA
| | - Hannah Dayton
- Department of Biological Sciences, Columbia University, New York, NY, USA
| | - Cong Xu
- Department of Biomedical Engineering, Columbia University, New York, NY, USA
| | | | - Yu-Young Wesley Tsai
- Department of Biological Sciences, Columbia University, New York, NY, USA
- Mortimer B. Zuckerman Mind Brain and Behavior Institute, Columbia University, New York, NY, USA
| | - Malika S Datta
- Department of Biological Sciences, Columbia University, New York, NY, USA
- Mortimer B. Zuckerman Mind Brain and Behavior Institute, Columbia University, New York, NY, USA
| | - Gorazd B Rosoklija
- Department of Psychiatry, Columbia University Irving Medical Center, New York State Psychiatric Institute, New York, NY, USA
| | - Andrew J Dwork
- Department of Psychiatry, Columbia University Irving Medical Center, New York State Psychiatric Institute, New York, NY, USA
- Department of Pathology and Cell Biology, Columbia University, New York, NY, USA
| | - J John Mann
- Department of Psychiatry, Columbia University Irving Medical Center, New York State Psychiatric Institute, New York, NY, USA
| | - Maura Boldrini
- Department of Psychiatry, Columbia University Irving Medical Center, New York State Psychiatric Institute, New York, NY, USA
| | - Kam W Leong
- Department of Biomedical Engineering, Columbia University, New York, NY, USA
| | - Lars E P Dietrich
- Department of Biological Sciences, Columbia University, New York, NY, USA
| | - Raju Tomer
- Department of Biological Sciences, Columbia University, New York, NY, USA.
- Department of Biomedical Engineering, Columbia University, New York, NY, USA.
- Mortimer B. Zuckerman Mind Brain and Behavior Institute, Columbia University, New York, NY, USA.
| |
Collapse
|
26
|
Kay RR, Lutton JE, King JS, Bretschneider T. Making cups and rings: the 'stalled-wave' model for macropinocytosis. Biochem Soc Trans 2024; 52:1785-1794. [PMID: 38934501 DOI: 10.1042/bst20231426] [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: 03/13/2024] [Revised: 05/29/2024] [Accepted: 06/07/2024] [Indexed: 06/28/2024]
Abstract
Macropinocytosis is a broadly conserved endocytic process discovered nearly 100 years ago, yet still poorly understood. It is prominent in cancer cell feeding, immune surveillance, uptake of RNA vaccines and as an invasion route for pathogens. Macropinocytic cells extend large cups or flaps from their plasma membrane to engulf droplets of medium and trap them in micron-sized vesicles. Here they are digested and the products absorbed. A major problem - discussed here - is to understand how cups are shaped and closed. Recently, lattice light-sheet microscopy has given a detailed description of this process in Dictyostelium amoebae, leading to the 'stalled-wave' model for cup formation and closure. This is based on membrane domains of PIP3 and active Ras and Rac that occupy the inner face of macropinocytic cups and are readily visible with suitable reporters. These domains attract activators of dendritic actin polymerization to their periphery, creating a ring of protrusive F-actin around themselves, thus shaping the walls of the cup. As domains grow, they drive a wave of actin polymerization across the plasma membrane that expands the cup. When domains stall, continued actin polymerization under the membrane, combined with increasing membrane tension in the cup, drives closure at lip or base. Modelling supports the feasibility of this scheme. No specialist coat proteins or contractile activities are required to shape and close cups: rings of actin polymerization formed around PIP3 domains that expand and stall seem sufficient. This scheme may be widely applicable and begs many biochemical questions.
Collapse
Affiliation(s)
- Robert R Kay
- MRC Laboratory of Molecular Biology, Cambridge CB2 0QH, U.K
| | - Judith E Lutton
- Department of Computer Science, University of Warwick, Coventry CV4 7AL, U.K
| | - Jason S King
- Department of Biomedical Sciences, Western Bank, Sheffield S10 2TN, U.K
| | - Till Bretschneider
- Department of Computer Science, University of Warwick, Coventry CV4 7AL, U.K
| |
Collapse
|
27
|
Bhattacharya S, Ettela A, Haydak J, Hobson CM, Stern A, Yoo M, Chew TL, Gusella GL, Gallagher EJ, Hone JC, Azeloglu EU. A high-throughput microfabricated platform for rapid quantification of metastatic potential. SCIENCE ADVANCES 2024; 10:eadk0015. [PMID: 39151003 PMCID: PMC11328906 DOI: 10.1126/sciadv.adk0015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Accepted: 07/12/2024] [Indexed: 08/18/2024]
Abstract
Assays that measure morphology, proliferation, motility, deformability, and migration are used to study the invasiveness of cancer cells. However, native invasive potential of cells may be hidden from these contextual metrics because they depend on culture conditions. We created a micropatterned chip that mimics the native environmental conditions, quantifies the invasive potential of tumor cells, and improves our understanding of the malignancy signatures. Unlike conventional assays, which rely on indirect measurements of metastatic potential, our method uses three-dimensional microchannels to measure the basal native invasiveness without chemoattractants or microfluidics. No change in cell death or proliferation is observed on our chips. Using six cancer cell lines, we show that our system is more sensitive than other motility-based assays, measures of nuclear deformability, or cell morphometrics. In addition to quantifying metastatic potential, our platform can distinguish between motility and invasiveness, help study molecular mechanisms of invasion, and screen for targeted therapeutics.
Collapse
Affiliation(s)
- Smiti Bhattacharya
- Barbara T. Murphy Division of Nephrology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Mechanical Engineering, Columbia University, New York, NY, USA
| | - Abora Ettela
- Division of Endocrinology, Diabetes and Bone Disease, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Jonathan Haydak
- Barbara T. Murphy Division of Nephrology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Chad M Hobson
- Advanced Imaging Center, Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA, USA
| | - Alan Stern
- Barbara T. Murphy Division of Nephrology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Miran Yoo
- Barbara T. Murphy Division of Nephrology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Teng-Leong Chew
- Advanced Imaging Center, Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA, USA
| | - G Luca Gusella
- Barbara T. Murphy Division of Nephrology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Emily J Gallagher
- Division of Endocrinology, Diabetes and Bone Disease, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Tisch Cancer Institute at Mount Sinai, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - James C Hone
- Department of Mechanical Engineering, Columbia University, New York, NY, USA
| | - Evren U Azeloglu
- Barbara T. Murphy Division of Nephrology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|
28
|
Illibauer J, Clodi-Seitz T, Zoufaly A, Aberle JH, Weninger WJ, Foedinger M, Elsayad K. Diagnostic potential of blood plasma longitudinal viscosity measured using Brillouin light scattering. Proc Natl Acad Sci U S A 2024; 121:e2323016121. [PMID: 39088388 PMCID: PMC11331083 DOI: 10.1073/pnas.2323016121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2023] [Accepted: 07/01/2024] [Indexed: 08/03/2024] Open
Abstract
Blood plasma viscosity (PV) is an established biomarker for numerous diseases. Measurement of the shear PV using conventional rheological techniques is, however, time consuming and requires significant plasma volumes. Here, we show that Brillouin light scattering (BLS) and angle-resolved spectroscopy measurements of the longitudinal PV from microliter-sized plasma volumes can serve as a proxy for the shear PV measured using conventional viscometers. This is not trivial given the distinct frequency regime probed and the longitudinal viscosity, a combination of the shear and bulk viscosity, representing a unique material property on account of the latter. We demonstrate this for plasma from healthy persons and patients suffering from different severities of COVID-19 (CoV), which has been associated with an increased shear PV. We further show that the additional information contained in the BLS-measured effective longitudinal PV and its temperature scaling can provide unique insight into the chemical constituents and physical properties of plasma that can be of diagnostic value. In particular, we find that changes in the effective longitudinal viscosity are consistent with an increased suspension concentration in CoV patient samples at elevated temperatures that is correlated with disease severity and progression. This is supported by results from rapid BLS spatial-mapping, angle-resolved BLS measurements, changes in the elastic scattering, and anomalies in the temperature scaling of the shear viscosity. Finally, we introduce a compact BLS probe to rapidly perform measurements in plastic transport tubes. Our results open a broad avenue for PV diagnostics based on the high-frequency effective longitudinal PV and show that BLS can provide a means for its implementation.
Collapse
Affiliation(s)
- Jennifer Illibauer
- Division of Anatomy, Center for Anatomy & Cell Biology, Medical University of Vienna, ViennaA-1090, Austria
- Medical Imaging Cluster, Medical University of Vienna, ViennaA-1090, Austria
| | | | - Alexander Zoufaly
- Department of Medicine, Klinik Favoriten, ViennaA-1100, Austria
- Sigmund Freud Private University, ViennaA-1020, Austria
| | - Judith H. Aberle
- Center for Virology, Medical University of Vienna, ViennaA-1090, Austria
| | - Wolfgang J. Weninger
- Division of Anatomy, Center for Anatomy & Cell Biology, Medical University of Vienna, ViennaA-1090, Austria
- Medical Imaging Cluster, Medical University of Vienna, ViennaA-1090, Austria
| | - Manuela Foedinger
- Sigmund Freud Private University, ViennaA-1020, Austria
- Institute of Laboratory Diagnostics, Klinik Favoriten, ViennaA-1100, Austria
| | - Kareem Elsayad
- Division of Anatomy, Center for Anatomy & Cell Biology, Medical University of Vienna, ViennaA-1090, Austria
- Medical Imaging Cluster, Medical University of Vienna, ViennaA-1090, Austria
- Advanced Microscopy, Vienna Biocenter Core Facilities, ViennaA-1030, Austria
| |
Collapse
|
29
|
Jo S, Fischer BR, Cronin NM, Nurmalasari NPD, Loyd YM, Kerkvliet JG, Bailey EM, Anderson RB, Scott BL, Hoppe AD. Antibody surface mobility amplifies FcγR signaling via Arp2/3 during phagocytosis. Biophys J 2024; 123:2312-2327. [PMID: 38321740 PMCID: PMC11331046 DOI: 10.1016/j.bpj.2024.01.036] [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: 06/30/2023] [Revised: 11/07/2023] [Accepted: 01/29/2024] [Indexed: 02/08/2024] Open
Abstract
We report herein that the anti-CD20 therapeutic antibody, rituximab, is rearranged into microclusters within the phagocytic synapse by macrophage Fcγ receptors (FcγR) during antibody-dependent cellular phagocytosis. These microclusters were observed to potently recruit Syk and to undergo rearrangements that were limited by the cytoskeleton of the target cell, with depolymerization of target-cell actin filaments leading to modest increases in phagocytic efficiency. Total internal reflection fluorescence analysis revealed that FcγR total phosphorylation, Syk phosphorylation, and Syk recruitment were enhanced when IgG-FcγR microclustering was enabled on fluid bilayers relative to immobile bilayers in a process that required Arp2/3. We conclude that on fluid surfaces, IgG-FcγR microclustering promotes signaling through Syk that is amplified by Arp2/3-driven actin rearrangements. Thus, the surface mobility of antigens bound by IgG shapes the signaling of FcγR with an unrecognized complexity beyond the zipper and trigger models of phagocytosis.
Collapse
Affiliation(s)
- Seongwan Jo
- Department of Chemistry and Biochemistry, South Dakota State University, Brookings, South Dakota; BioSNTRii, South Dakota State University, Brookings, South Dakota
| | - Brady R Fischer
- Department of Chemistry and Biochemistry, South Dakota State University, Brookings, South Dakota; BioSNTRii, South Dakota State University, Brookings, South Dakota
| | - Nicholas M Cronin
- Department of Chemistry and Biochemistry, South Dakota State University, Brookings, South Dakota; BioSNTRii, South Dakota State University, Brookings, South Dakota
| | - Ni Putu Dewi Nurmalasari
- Department of Nanoscience & Biomedical Engineering, South Dakota School of Mines and Technology, Rapid City, South Dakota; BioSNTRii, South Dakota School of Mines and Technology, Rapid City, South Dakota
| | - Yoseph M Loyd
- Department of Nanoscience & Biomedical Engineering, South Dakota School of Mines and Technology, Rapid City, South Dakota; BioSNTRii, South Dakota School of Mines and Technology, Rapid City, South Dakota
| | - Jason G Kerkvliet
- Department of Chemistry and Biochemistry, South Dakota State University, Brookings, South Dakota; BioSNTRii, South Dakota State University, Brookings, South Dakota
| | - Elizabeth M Bailey
- Department of Chemistry and Biochemistry, South Dakota State University, Brookings, South Dakota; BioSNTRii, South Dakota State University, Brookings, South Dakota
| | - Robert B Anderson
- Department of Nanoscience & Biomedical Engineering, South Dakota School of Mines and Technology, Rapid City, South Dakota; BioSNTRii, South Dakota School of Mines and Technology, Rapid City, South Dakota
| | - Brandon L Scott
- Department of Nanoscience & Biomedical Engineering, South Dakota School of Mines and Technology, Rapid City, South Dakota; BioSNTRii, South Dakota School of Mines and Technology, Rapid City, South Dakota
| | - Adam D Hoppe
- Department of Chemistry and Biochemistry, South Dakota State University, Brookings, South Dakota; BioSNTRii, South Dakota State University, Brookings, South Dakota.
| |
Collapse
|
30
|
Mazalo JK, Tay SS, Kempe D, Biro M. Chemokine receptor distribution on the surface of repolarizing T cells. Biophys J 2024; 123:2328-2342. [PMID: 38327056 PMCID: PMC11331047 DOI: 10.1016/j.bpj.2024.02.001] [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: 09/22/2023] [Revised: 12/17/2023] [Accepted: 02/02/2024] [Indexed: 02/09/2024] Open
Abstract
T cells migrate constitutively with a polarized morphology, underpinned by signaling compartmentalization and discrete cytoskeletal organizations, giving rise to a dynamic and expansive leading edge, distinct from the stable and constricted uropod at the rear. In vivo, the motion and function of T cells at various stages of differentiation is highly directed by chemokine gradients. When cognate ligands bind chemokine receptors on their surface, T cells respond by reorientating their polarity axis and migrating toward the source of the chemokine signal. Despite the significance of such chemotactic repolarization to the accurate navigation and function of T cells, the precise signaling mechanisms that underlie it remain elusive. Notably, it remained unclear whether the distribution of chemokine receptors on the T cell surface is altered during repolarization. Here, we developed parallel cell-secreted and microfluidics-based chemokine gradient delivery methods and employed both fixed imaging and live lattice light-sheet microscopy to investigate the dynamics of chemokine receptor CCR5 on the surface of primary murine CD8+ T cells. Our findings show that, during constitutive migration, chemokine receptor distribution is largely isotropic on the T cell surface. However, upon exposure to a CCL3 gradient, surface chemokine receptor distributions exhibit a transient bias toward the uropod. The chemokine receptors then progressively redistribute from the uropod to cover the T cell surface uniformly. This study sheds new light on the dynamics of surface chemokine receptor distribution during T cell repolarization, advancing our understanding of the signaling of immune cells in the complex chemokine landscapes they navigate.
Collapse
Affiliation(s)
- Jessica K Mazalo
- EMBL Australia, Single Molecule Science Node, School of Biomedical Sciences, University of New South Wales, Sydney, NSW, Australia
| | - Szun S Tay
- EMBL Australia, Single Molecule Science Node, School of Biomedical Sciences, University of New South Wales, Sydney, NSW, Australia
| | - Daryan Kempe
- EMBL Australia, Single Molecule Science Node, School of Biomedical Sciences, University of New South Wales, Sydney, NSW, Australia.
| | - Maté Biro
- EMBL Australia, Single Molecule Science Node, School of Biomedical Sciences, University of New South Wales, Sydney, NSW, Australia.
| |
Collapse
|
31
|
Polak R, Zhang ET, Kuo CJ. Cancer organoids 2.0: modelling the complexity of the tumour immune microenvironment. Nat Rev Cancer 2024; 24:523-539. [PMID: 38977835 DOI: 10.1038/s41568-024-00706-6] [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] [Accepted: 05/09/2024] [Indexed: 07/10/2024]
Abstract
The development of neoplasia involves a complex and continuous interplay between malignantly transformed cells and the tumour microenvironment (TME). Cancer immunotherapies targeting the immune TME have been increasingly validated in clinical trials but response rates vary substantially between tumour histologies and are often transient, idiosyncratic and confounded by resistance. Faithful experimental models of the patient-specific tumour immune microenvironment, capable of recapitulating tumour biology and immunotherapy effects, would greatly improve patient selection, target identification and definition of resistance mechanisms for immuno-oncology therapeutics. In this Review, we discuss currently available and rapidly evolving 3D tumour organoid models that capture important immune features of the TME. We highlight diverse opportunities for organoid-based investigations of tumour immunity, drug development and precision medicine.
Collapse
Affiliation(s)
- Roel Polak
- Department of Medicine, Division of Hematology, Stanford University School of Medicine, Stanford, CA, USA
- Princess Máxima Center for Pediatric Oncology, Utrecht, Netherlands
| | - Elisa T Zhang
- Department of Medicine, Division of Hematology, Stanford University School of Medicine, Stanford, CA, USA
| | - Calvin J Kuo
- Department of Medicine, Division of Hematology, Stanford University School of Medicine, Stanford, CA, USA.
| |
Collapse
|
32
|
Shi Y, Daugird TA, Legant WR. Posterior approach to correct for focal plane offsets in lattice light-sheet structured illumination microscopy. JOURNAL OF BIOMEDICAL OPTICS 2024; 29:086502. [PMID: 39086928 PMCID: PMC11289499 DOI: 10.1117/1.jbo.29.8.086502] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Revised: 06/20/2024] [Accepted: 06/24/2024] [Indexed: 08/02/2024]
Abstract
Significance Lattice light-sheet structured illumination microscopy (latticeSIM) has proven highly effective in producing three-dimensional images with super resolution rapidly and with minimal photobleaching. However, due to the use of two separate objectives, sample-induced aberrations can result in an offset between the planes of excitation and detection, causing artifacts in the reconstructed images. Aim We introduce a posterior approach to detect and correct the axial offset between the excitation and detection focal planes in latticeSIM and provide a method to minimize artifacts in the reconstructed images. Approach We utilized the residual phase information within the overlap regions of the laterally shifted structured illumination microscopy information components in frequency space to retrieve the axial offset between the excitation and the detection focal planes in latticeSIM. Results We validated our technique through simulations and experiments, encompassing a range of samples from fluorescent beads to subcellular structures of adherent cells. We also show that using transfer functions with the same axial offset as the one present during data acquisition results in reconstructed images with minimal artifacts and salvages otherwise unusable data. Conclusion We envision that our method will be a valuable addition to restore image quality in latticeSIM datasets even for those acquired under non-ideal experimental conditions.
Collapse
Affiliation(s)
- Yu Shi
- University of North Carolina at Chapel Hill and North Carolina State University, Joint Department of Biomedical Engineering, Chapel Hill, North Carolina, United States
| | - Tim A. Daugird
- University of North Carolina at Chapel Hill, Department of Pharmacology, Chapel Hill, North Carolina, United States
| | - Wesley R. Legant
- University of North Carolina at Chapel Hill and North Carolina State University, Joint Department of Biomedical Engineering, Chapel Hill, North Carolina, United States
- University of North Carolina at Chapel Hill, Department of Pharmacology, Chapel Hill, North Carolina, United States
| |
Collapse
|
33
|
Dong P, Zhang S, Gandin V, Xie L, Wang L, Lemire AL, Li W, Otsuna H, Kawase T, Lander AD, Chang HY, Liu ZJ. Cohesin prevents cross-domain gene coactivation. Nat Genet 2024; 56:1654-1664. [PMID: 39048795 PMCID: PMC11319207 DOI: 10.1038/s41588-024-01852-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Accepted: 06/27/2024] [Indexed: 07/27/2024]
Abstract
The contrast between the disruption of genome topology after cohesin loss and the lack of downstream gene expression changes instigates intense debates regarding the structure-function relationship between genome and gene regulation. Here, by analyzing transcriptome and chromatin accessibility at the single-cell level, we discover that, instead of dictating population-wide gene expression levels, cohesin supplies a general function to neutralize stochastic coexpression tendencies of cis-linked genes in single cells. Notably, cohesin loss induces widespread gene coactivation and chromatin co-opening tens of million bases apart in cis. Spatial genome and protein imaging reveals that cohesin prevents gene co-bursting along the chromosome and blocks spatial mixing of transcriptional hubs. Single-molecule imaging shows that cohesin confines the exploration of diverse enhancer and core promoter binding transcriptional regulators. Together, these results support that cohesin arranges nuclear topology to control gene coexpression in single cells.
Collapse
Affiliation(s)
- Peng Dong
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA, USA.
- Institute of Biomedical and Health Engineering, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China.
| | - Shu Zhang
- Center for Personal Dynamic Regulomes and Howard Hughes Medical Institute, Stanford University, Stanford, CA, USA
| | - Valentina Gandin
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA, USA
| | - Liangqi Xie
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA, USA
- Cancer Biology and Infection Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Lihua Wang
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA, USA
| | - Andrew L Lemire
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA, USA
| | - Wenhong Li
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA, USA
| | - Hideo Otsuna
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA, USA
| | - Takashi Kawase
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA, USA
| | - Arthur D Lander
- Department of Developmental and Cell Biology, Center for Complex Biological Systems, University of California, Irvine, CA, USA
| | - Howard Y Chang
- Center for Personal Dynamic Regulomes and Howard Hughes Medical Institute, Stanford University, Stanford, CA, USA
| | - Zhe J Liu
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA, USA.
| |
Collapse
|
34
|
Heiles B, Nelissen F, Terwiel D, Park BM, Munoz Ibarra E, Matalliotakis A, Waasdorp R, Ara T, Barturen-Larrea P, Duan M, Shapiro MG, Gazzola V, Maresca D. Nonlinear sound-sheet microscopy: imaging opaque organs at the capillary and cellular scale. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.31.605825. [PMID: 39211282 PMCID: PMC11361007 DOI: 10.1101/2024.07.31.605825] [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
Light-sheet fluorescence microscopy has revolutionized biology by visualizing dynamic cellular processes in three dimensions. However, light scattering in thick tissue and photobleaching of fluorescent reporters limit this method to studying thin or translucent specimens. Here we show that non-diffractive ultrasonic beams used in conjunction with a cross-amplitude modulation sequence and nonlinear acoustic reporters enable fast and volumetric imaging of targeted biological functions. We report volumetric imaging of tumor gene expression at the cm 3 scale using genetically encoded gas vesicles, and localization microscopy of currently uncharted cerebral capillary networks using intravascular microbubble contrast agents. Nonlinear sound-sheet microscopy provides a ∼64x acceleration in imaging speed, ∼35x increase in imaged volume and ∼4x increase in classical imaging resolution compared to the state-of-the-art in biomolecular ultrasound.
Collapse
|
35
|
Vanderleest TE, Xie Y, Budhathoki R, Linvill K, Hobson C, Heddleston J, Loerke D, Blankenship JT. Lattice light sheet microscopy reveals 4D force propagation dynamics and leading-edge behaviors in an embryonic epithelium in Drosophila. Curr Biol 2024; 34:3165-3177.e3. [PMID: 38959881 DOI: 10.1016/j.cub.2024.06.017] [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: 12/14/2023] [Revised: 04/04/2024] [Accepted: 06/07/2024] [Indexed: 07/05/2024]
Abstract
How pulsed contractile dynamics drive the remodeling of cell and tissue topologies in epithelial sheets has been a key question in development and disease. Due to constraints in imaging and analysis technologies, studies that have described the in vivo mechanisms underlying changes in cell and neighbor relationships have largely been confined to analyses of planar apical regions. Thus, how the volumetric nature of epithelial cells affects force propagation and remodeling of the cell surface in three dimensions, including especially the apical-basal axis, is unclear. Here, we perform lattice light sheet microscopy (LLSM)-based analysis to determine how far and fast forces propagate across different apical-basal layers, as well as where topological changes initiate from in a columnar epithelium. These datasets are highly time- and depth-resolved and reveal that topology-changing forces are spatially entangled, with contractile force generation occurring across the observed apical-basal axis in a pulsed fashion, while the conservation of cell volumes constrains instantaneous cell deformations. Leading layer behaviors occur opportunistically in response to favorable phasic conditions, with lagging layers "zippering" to catch up as new contractile pulses propel further changes in cell topologies. These results argue against specific zones of topological initiation and demonstrate the importance of systematic 4D-based analysis in understanding how forces and deformations in cell dimensions propagate in a three-dimensional environment.
Collapse
Affiliation(s)
- Timothy E Vanderleest
- Department of Biological Sciences, University of Denver, Denver, CO 80208, USA; Department of Physics and Astronomy, University of Denver, Denver, CO 80208, USA
| | - Yi Xie
- Department of Biological Sciences, University of Denver, Denver, CO 80208, USA
| | - Rashmi Budhathoki
- Department of Biological Sciences, University of Denver, Denver, CO 80208, USA
| | - Katie Linvill
- Department of Physics and Astronomy, University of Denver, Denver, CO 80208, USA
| | - Chad Hobson
- Advanced Imaging Center, Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA 20147, USA
| | - John Heddleston
- Cleveland Clinic Florida Research & Innovation Center, Port St. Lucie, FL, USA
| | - Dinah Loerke
- Department of Physics and Astronomy, University of Denver, Denver, CO 80208, USA.
| | - J Todd Blankenship
- Department of Biological Sciences, University of Denver, Denver, CO 80208, USA.
| |
Collapse
|
36
|
Munshi R. How Transcription Factor Clusters Shape the Transcriptional Landscape. Biomolecules 2024; 14:875. [PMID: 39062589 PMCID: PMC11274464 DOI: 10.3390/biom14070875] [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: 06/01/2024] [Revised: 07/14/2024] [Accepted: 07/16/2024] [Indexed: 07/28/2024] Open
Abstract
In eukaryotic cells, gene transcription typically occurs in discrete periods of promoter activity, interspersed with intervals of inactivity. This pattern deviates from simple stochastic events and warrants a closer examination of the molecular interactions that activate the promoter. Recent studies have identified transcription factor (TF) clusters as key precursors to transcriptional bursting. Often, these TF clusters form at chromatin segments that are physically distant from the promoter, making changes in chromatin conformation crucial for promoter-TF cluster interactions. In this review, I explore the formation and constituents of TF clusters, examining how the dynamic interplay between chromatin architecture and TF clustering influences transcriptional bursting. Additionally, I discuss techniques for visualizing TF clusters and provide an outlook on understanding the remaining gaps in this field.
Collapse
Affiliation(s)
- Rahul Munshi
- Joseph Henry Laboratories of Physics and Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ 08544, USA
| |
Collapse
|
37
|
Thaprawat P, Zhang Z, Rentchler EC, Wang F, Chalasani S, Giuliano CJ, Lourido S, Di Cristina M, Klionsky DJ, Carruthers VB. TgATG9 is required for autophagosome biogenesis and maintenance of chronic infection in Toxoplasma gondii. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.08.602581. [PMID: 39026823 PMCID: PMC11257638 DOI: 10.1101/2024.07.08.602581] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/20/2024]
Abstract
Toxoplasma gondii is a ubiquitous protozoan parasite that can reside long-term within hosts as intracellular tissue cysts comprised of chronic stage bradyzoites. To perturb chronic infection requires a better understanding of the cellular processes that mediate parasite persistence. Macroautophagy/autophagy is a catabolic and homeostatic pathway that is required for T. gondii chronic infection, although the molecular details of this process remain poorly understood. A key step in autophagy is the initial formation of the phagophore that sequesters cytoplasmic components and matures into a double-membraned autophagosome for delivery of the cargo to a cell's digestive organelle for degradative recycling. While T. gondii appears to have a reduced repertoire of autophagy proteins, it possesses a putative phospholipid scramblase, TgATG9. Through structural modeling and complementation assays, we show herein that TgATG9 can partially rescue bulk autophagy in atg9Δ yeast. We demonstrated the importance of TgATG9 for proper autophagosome dynamics at the subcellular level using three-dimensional live cell lattice light sheet microscopy. Conditional knockdown of TgATG9 in T. gondii after bradyzoite differentiation resulted in markedly reduced parasite viability. Together, our findings provide insights into the molecular dynamics of autophagosome biogenesis within an early-branching eukaryote and pinpoint the indispensable role of autophagy in maintaining T. gondii chronic infection.
Collapse
|
38
|
Yang R, Xiao T, Cheng Y, Li A, Qu J, Liang R, Bao S, Wang X, Wang J, Suo J, Luo Q, Dai Q. Sharing massive biomedical data at magnitudes lower bandwidth using implicit neural function. Proc Natl Acad Sci U S A 2024; 121:e2320870121. [PMID: 38959033 PMCID: PMC11252806 DOI: 10.1073/pnas.2320870121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Accepted: 05/21/2024] [Indexed: 07/04/2024] Open
Abstract
Efficient storage and sharing of massive biomedical data would open up their wide accessibility to different institutions and disciplines. However, compressors tailored for natural photos/videos are rapidly limited for biomedical data, while emerging deep learning-based methods demand huge training data and are difficult to generalize. Here, we propose to conduct Biomedical data compRession with Implicit nEural Function (BRIEF) by representing the target data with compact neural networks, which are data specific and thus have no generalization issues. Benefiting from the strong representation capability of implicit neural function, BRIEF achieves 2[Formula: see text]3 orders of magnitude compression on diverse biomedical data at significantly higher fidelity than existing techniques. Besides, BRIEF is of consistent performance across the whole data volume, and supports customized spatially varying fidelity. BRIEF's multifold advantageous features also serve reliable downstream tasks at low bandwidth. Our approach will facilitate low-bandwidth data sharing and promote collaboration and progress in the biomedical field.
Collapse
Affiliation(s)
- Runzhao Yang
- Department of Automation, Tsinghua University, Beijing100084, China
- Institute of Brain and Cognitive Sciences, Tsinghua University, Beijing100084, China
- Shanghai Artificial Intelligence Laboratory, Shanghai200232, China
| | - Tingxiong Xiao
- Department of Automation, Tsinghua University, Beijing100084, China
| | - Yuxiao Cheng
- Department of Automation, Tsinghua University, Beijing100084, China
| | - Anan Li
- Britton Chance Center and MoE Key Laboratory for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology, Wuhan430074, China
- Huazhong University of Science and Technology-Suzhou Institute for Brainsmatics, Jiangsu Industrial Technology Research Institute, Suzhou215123, China
| | - Jinyuan Qu
- Department of Automation, Tsinghua University, Beijing100084, China
| | - Rui Liang
- School of Biomedical Engineering, Hainan University, Haikou570228, China
| | - Shengda Bao
- Britton Chance Center and MoE Key Laboratory for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology, Wuhan430074, China
| | - Xiaofeng Wang
- Britton Chance Center and MoE Key Laboratory for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology, Wuhan430074, China
| | - Jue Wang
- Department of Automation, Tsinghua University, Beijing100084, China
| | - Jinli Suo
- Department of Automation, Tsinghua University, Beijing100084, China
- Institute of Brain and Cognitive Sciences, Tsinghua University, Beijing100084, China
- Shanghai Artificial Intelligence Laboratory, Shanghai200232, China
| | - Qingming Luo
- School of Biomedical Engineering, Hainan University, Haikou570228, China
| | - Qionghai Dai
- Department of Automation, Tsinghua University, Beijing100084, China
- Institute of Brain and Cognitive Sciences, Tsinghua University, Beijing100084, China
| |
Collapse
|
39
|
Tian Y, Wu Z, Lui H, Zhao J, Kalia S, Seo I, Ou-Yang H, Zeng H. Precise Serial Microregistration Enables Quantitative Microscopy Imaging Tracking of Human Skin Cells In Vivo. Cells 2024; 13:1158. [PMID: 38995009 PMCID: PMC11240548 DOI: 10.3390/cells13131158] [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: 06/05/2024] [Revised: 07/04/2024] [Accepted: 07/04/2024] [Indexed: 07/13/2024] Open
Abstract
We developed an automated microregistration method that enables repeated in vivo skin microscopy imaging of the same tissue microlocation and specific cells over a long period of days and weeks with unprecedented precision. Applying this method in conjunction with an in vivo multimodality multiphoton microscope, the behavior of human skin cells such as cell proliferation, melanin upward migration, blood flow dynamics, and epidermal thickness adaptation can be recorded over time, facilitating quantitative cellular dynamics analysis. We demonstrated the usefulness of this method in a skin biology study by successfully monitoring skin cellular responses for a period of two weeks following an acute exposure to ultraviolet light.
Collapse
Affiliation(s)
- Yunxian Tian
- Imaging Unit, Integrative Oncology Department, BC Cancer Research Centre, Vancouver, BC V5Z 1L3, Canada
- Photomedicine Institute, Department of Dermatology and Skin Science, University of British Columbia, Vancouver, BC V5Z 4E8, Canada
- Vancouver Coastal Health Research Institute, Vancouver, BC V5Z 1M9, Canada
| | - Zhenguo Wu
- Imaging Unit, Integrative Oncology Department, BC Cancer Research Centre, Vancouver, BC V5Z 1L3, Canada
- Photomedicine Institute, Department of Dermatology and Skin Science, University of British Columbia, Vancouver, BC V5Z 4E8, Canada
- Vancouver Coastal Health Research Institute, Vancouver, BC V5Z 1M9, Canada
| | - Harvey Lui
- Imaging Unit, Integrative Oncology Department, BC Cancer Research Centre, Vancouver, BC V5Z 1L3, Canada
- Photomedicine Institute, Department of Dermatology and Skin Science, University of British Columbia, Vancouver, BC V5Z 4E8, Canada
- Vancouver Coastal Health Research Institute, Vancouver, BC V5Z 1M9, Canada
| | - Jianhua Zhao
- Imaging Unit, Integrative Oncology Department, BC Cancer Research Centre, Vancouver, BC V5Z 1L3, Canada
- Photomedicine Institute, Department of Dermatology and Skin Science, University of British Columbia, Vancouver, BC V5Z 4E8, Canada
- Vancouver Coastal Health Research Institute, Vancouver, BC V5Z 1M9, Canada
| | - Sunil Kalia
- Photomedicine Institute, Department of Dermatology and Skin Science, University of British Columbia, Vancouver, BC V5Z 4E8, Canada
- Vancouver Coastal Health Research Institute, Vancouver, BC V5Z 1M9, Canada
| | - InSeok Seo
- Johnson and Johnson Consumer Inc., Skillman, NJ 08558, USA
| | - Hao Ou-Yang
- Johnson and Johnson Consumer Inc., Skillman, NJ 08558, USA
| | - Haishan Zeng
- Imaging Unit, Integrative Oncology Department, BC Cancer Research Centre, Vancouver, BC V5Z 1L3, Canada
- Photomedicine Institute, Department of Dermatology and Skin Science, University of British Columbia, Vancouver, BC V5Z 4E8, Canada
- Vancouver Coastal Health Research Institute, Vancouver, BC V5Z 1M9, Canada
| |
Collapse
|
40
|
Kuchimaru T. Emerging Synthetic Bioluminescent Reactions for Non-Invasive Imaging of Freely Moving Animals. Int J Mol Sci 2024; 25:7338. [PMID: 39000448 PMCID: PMC11242611 DOI: 10.3390/ijms25137338] [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: 05/18/2024] [Revised: 07/02/2024] [Accepted: 07/03/2024] [Indexed: 07/16/2024] Open
Abstract
Bioluminescence imaging (BLI) is an indispensable technique for visualizing the dynamics of diverse biological processes in mammalian animal models, including cancer, viral infections, and immune responses. However, a critical scientific challenge remains: non-invasively visualizing homeostatic and disease mechanisms in freely moving animals to understand the molecular basis of exercises, social behavior, and other phenomena. Classical BLI relies on prolonged camera exposure to accumulate the limited number of photons that traveled from deep tissues in anesthetized or constrained animals. Recent advancements in synthetic bioluminescence reactions, utilizing artificial luciferin-luciferase pairs, have considerably increased the number of detectable photons from deep tissues, facilitating high-speed BLI to capture moving objects. In this review, I provide an overview of emerging synthetic bioluminescence reactions that enable the non-invasive imaging of freely moving animals. This approach holds the potential to uncover unique physiological processes that are inaccessible with current methodologies.
Collapse
Affiliation(s)
- Takahiro Kuchimaru
- Center for Molecular Medicine, Jichi Medical University, Tochigi 329-0498, Japan
| |
Collapse
|
41
|
Feng R, Xie J, Gao L. EDTP enhances and protects the fluorescent signal of GFP in cleared and expanded tissues. Sci Rep 2024; 14:15279. [PMID: 38961181 PMCID: PMC11222453 DOI: 10.1038/s41598-024-66398-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Accepted: 07/01/2024] [Indexed: 07/05/2024] Open
Abstract
Advanced 3D high-resolution imaging techniques are essential for investigating biological challenges, such as neural circuit analysis and tumor microenvironment in intact tissues. However, the fluorescence signal emitted by endogenous fluorescent proteins in cleared or expanded biological samples gradually diminishes with repeated irradiation and prolonged imaging, compromising its ability to accurately depict the underlying scientific problem. We have developed a strategy to preserve fluorescence in cleared and expanded tissue samples during prolonged high-resolution three-dimensional imaging. We evaluated various compounds at different concentrations to determine their ability to enhance fluorescence intensity and resistance to photobleaching while maintaining the structural integrity of the tissue. Specifically, we investigated the impact of EDTP utilization on GFP, as it has been observed to significantly improve fluorescence intensity, resistance to photobleaching, and maintain fluorescence during extended room temperature storage. This breakthrough will facilitate extended hydrophilic and hydrogel-based clearing and expansion methods for achieving long-term high-resolution 3D imaging of cleared biological tissues by effectively safeguarding fluorescent proteins within the tissue.
Collapse
Affiliation(s)
- Ruili Feng
- Fudan University, Shanghai, 200433, China.
- Key Laboratory of Structural Biology of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, 310024, Zhejiang, China.
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, 310024, Zhejiang, China.
| | - Jiongfang Xie
- Fudan University, Shanghai, 200433, China
- Key Laboratory of Structural Biology of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, 310024, Zhejiang, China
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, 310024, Zhejiang, China
| | - Liang Gao
- Fudan University, Shanghai, 200433, China
- Key Laboratory of Structural Biology of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, 310024, Zhejiang, China
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, 310024, Zhejiang, China
| |
Collapse
|
42
|
Presman DM, Benítez B, Lafuente AL, Vázquez Lareu A. Chromatin structure and dynamics: one nucleosome at a time. Histochem Cell Biol 2024; 162:79-90. [PMID: 38607419 DOI: 10.1007/s00418-024-02281-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/19/2024] [Indexed: 04/13/2024]
Abstract
Eukaryotic genomes store information on many levels, including their linear DNA sequence, the posttranslational modifications of its constituents (epigenetic modifications), and its three-dimensional folding. Understanding how this information is stored and read requires multidisciplinary collaborations from many branches of science beyond biology, including physics, chemistry, and computer science. Concurrent recent developments in all these areas have enabled researchers to image the genome with unprecedented spatial and temporal resolution. In this review, we focus on what single-molecule imaging and tracking of individual proteins in live cells have taught us about chromatin structure and dynamics. Starting with the basics of single-molecule tracking (SMT), we describe some advantages over in situ imaging techniques and its current limitations. Next, we focus on single-nucleosome studies and what they have added to our current understanding of the relationship between chromatin dynamics and transcription. In celebration of Robert Feulgen's ground-breaking discovery that allowed us to start seeing the genome, we discuss current models of chromatin structure and future challenges ahead.
Collapse
Affiliation(s)
- Diego M Presman
- Instituto de Fisiología, Biología Molecular y Neurociencias (IFIBYNE), Facultad de Ciencias Exactas y Naturales, CONICET-Universidad de Buenos Aires, C1428EGA, Buenos Aires, Argentina.
- Departamento de Fisiología, Biología Molecular y Celular, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, C1428EGA, Buenos Aires, Argentina.
| | - Belén Benítez
- Instituto de Fisiología, Biología Molecular y Neurociencias (IFIBYNE), Facultad de Ciencias Exactas y Naturales, CONICET-Universidad de Buenos Aires, C1428EGA, Buenos Aires, Argentina
- Instituto de Química Biológica (IQUIBICEN), Facultad de Ciencias Exactas y Naturales, CONICET-Universidad de Buenos Aires, C1428EGA, Buenos Aires, Argentina
| | - Agustina L Lafuente
- Instituto de Fisiología, Biología Molecular y Neurociencias (IFIBYNE), Facultad de Ciencias Exactas y Naturales, CONICET-Universidad de Buenos Aires, C1428EGA, Buenos Aires, Argentina
| | - Alejo Vázquez Lareu
- Instituto de Fisiología, Biología Molecular y Neurociencias (IFIBYNE), Facultad de Ciencias Exactas y Naturales, CONICET-Universidad de Buenos Aires, C1428EGA, Buenos Aires, Argentina
- Instituto de Química Biológica (IQUIBICEN), Facultad de Ciencias Exactas y Naturales, CONICET-Universidad de Buenos Aires, C1428EGA, Buenos Aires, Argentina
| |
Collapse
|
43
|
Joshi K, York HM, Wright CS, Biswas RR, Arumugam S, Iyer-Biswas S. Emergent Spatiotemporal Organization in Stochastic Intracellular Transport Dynamics. Annu Rev Biophys 2024; 53:193-220. [PMID: 38346244 DOI: 10.1146/annurev-biophys-030422-044448] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/18/2024]
Abstract
The interior of a living cell is an active, fluctuating, and crowded environment, yet it maintains a high level of coherent organization. This dichotomy is readily apparent in the intracellular transport system of the cell. Membrane-bound compartments called endosomes play a key role in carrying cargo, in conjunction with myriad components including cargo adaptor proteins, membrane sculptors, motor proteins, and the cytoskeleton. These components coordinate to effectively navigate the crowded cell interior and transport cargo to specific intracellular locations, even though the underlying protein interactions and enzymatic reactions exhibit stochastic behavior. A major challenge is to measure, analyze, and understand how, despite the inherent stochasticity of the constituent processes, the collective outcomes show an emergent spatiotemporal order that is precise and robust. This review focuses on this intriguing dichotomy, providing insights into the known mechanisms of noise suppression and noise utilization in intracellular transport processes, and also identifies opportunities for future inquiry.
Collapse
Affiliation(s)
- Kunaal Joshi
- Department of Physics and Astronomy, Purdue University, West Lafayette, Indiana, USA;
| | - Harrison M York
- Monash Biomedicine Discovery Institute, Faculty of Medicine, Nursing and Health Sciences, Monash University, Clayton, Victoria, Australia;
| | - Charles S Wright
- Department of Physics and Astronomy, Purdue University, West Lafayette, Indiana, USA;
- Monash Biomedicine Discovery Institute, Faculty of Medicine, Nursing and Health Sciences, Monash University, Clayton, Victoria, Australia;
| | - Rudro R Biswas
- Department of Physics and Astronomy, Purdue University, West Lafayette, Indiana, USA;
| | - Senthil Arumugam
- ARC Centre of Excellence in Advanced Molecular Imaging, Monash University, Melbourne, Victoria, Australia
- Single Molecule Science, University of New South Wales, Sydney, New South Wales, Australia
- Monash Biomedicine Discovery Institute, Faculty of Medicine, Nursing and Health Sciences, Monash University, Clayton, Victoria, Australia;
- European Molecular Biological Laboratory Australia (EMBL Australia), Monash University, Melbourne, Victoria, Australia
| | - Srividya Iyer-Biswas
- Department of Physics and Astronomy, Purdue University, West Lafayette, Indiana, USA;
- Santa Fe Institute, Santa Fe, New Mexico, USA
| |
Collapse
|
44
|
Linsley JW, Reisine T, Finkbeiner S. Three dimensional and four dimensional live imaging to study mechanisms of progressive neurodegeneration. J Biol Chem 2024; 300:107433. [PMID: 38825007 PMCID: PMC11261153 DOI: 10.1016/j.jbc.2024.107433] [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: 08/30/2023] [Revised: 05/20/2024] [Accepted: 05/26/2024] [Indexed: 06/04/2024] Open
Abstract
Neurodegenerative diseases are complex and progressive, posing challenges to their study and understanding. Recent advances in microscopy imaging technologies have enabled the exploration of neurons in three spatial dimensions (3D) over time (4D). When applied to 3D cultures, tissues, or animals, these technologies can provide valuable insights into the dynamic and spatial nature of neurodegenerative diseases. This review focuses on the use of imaging techniques and neurodegenerative disease models to study neurodegeneration in 4D. Imaging techniques such as confocal microscopy, two-photon microscopy, miniscope imaging, light sheet microscopy, and robotic microscopy offer powerful tools to visualize and analyze neuronal changes over time in 3D tissue. Application of these technologies to in vitro models of neurodegeneration such as mouse organotypic culture systems and human organoid models provide versatile platforms to study neurodegeneration in a physiologically relevant context. Additionally, use of 4D imaging in vivo, including in mouse and zebrafish models of neurodegenerative diseases, allows for the investigation of early dysfunction and behavioral changes associated with neurodegeneration. We propose that these studies have the power to overcome the limitations of two-dimensional monolayer neuronal cultures and pave the way for improved understanding of the dynamics of neurodegenerative diseases and the development of effective therapeutic strategies.
Collapse
Affiliation(s)
- Jeremy W Linsley
- Center for Systems and Therapeutics, Gladstone Institutes, San Francisco, California, USA; Operant Biopharma, San Francisco, California, USA
| | - Terry Reisine
- Independent Scientific Consultant, Santa Cruz, California, USA
| | - Steven Finkbeiner
- Center for Systems and Therapeutics, Gladstone Institutes, San Francisco, California, USA; Operant Biopharma, San Francisco, California, USA; Taube/Koret Center for Neurodegenerative Disease, Gladstone Institutes, San Francisco, California, USA; Departments of Neurology and Physiology, University of California, San Francisco, California, USA; Neuroscience Graduate Program, University of California, San Francisco, California, USA; Biomedical Sciences Graduate Program, University of California, San Francisco, California, USA.
| |
Collapse
|
45
|
Soumier A, Lio G, Demily C. Current and future applications of light-sheet imaging for identifying molecular and developmental processes in autism spectrum disorders. Mol Psychiatry 2024; 29:2274-2284. [PMID: 38443634 DOI: 10.1038/s41380-024-02487-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Revised: 02/09/2024] [Accepted: 02/13/2024] [Indexed: 03/07/2024]
Abstract
Autism spectrum disorder (ASD) is identified by a set of neurodevelopmental divergences that typically affect the social communication domain. ASD is also characterized by heterogeneous cognitive impairments and is associated with cooccurring physical and medical conditions. As behaviors emerge as the brain matures, it is particularly essential to identify any gaps in neurodevelopmental trajectories during early perinatal life. Here, we introduce the potential of light-sheet imaging for studying developmental biology and cross-scale interactions among genetic, cellular, molecular and macroscale levels of circuitry and connectivity. We first report the core principles of light-sheet imaging and the recent progress in studying brain development in preclinical animal models and human organoids. We also present studies using light-sheet imaging to understand the development and function of other organs, such as the skin and gastrointestinal tract. We also provide information on the potential of light-sheet imaging in preclinical drug development. Finally, we speculate on the translational benefits of light-sheet imaging for studying individual brain-body interactions in advancing ASD research and creating personalized interventions.
Collapse
Affiliation(s)
- Amelie Soumier
- Le Vinatier Hospital Center, 95 boulevard Pinel, 69675, Bron cedex, France.
- iMIND, Center of Excellence for Autism, 95 boulevard Pinel, 69675, Bron cedex, France.
- Institute of Cognitive Science Marc Jeannerod, CNRS, UMR 5229, 67 boulevard Pinel, 69675, Bron cedex, France.
- University Claude Bernard Lyon 1, 43 boulevard du 11 Novembre 1918, 69622, Villeurbanne cedex, France.
| | - Guillaume Lio
- Le Vinatier Hospital Center, 95 boulevard Pinel, 69675, Bron cedex, France
- iMIND, Center of Excellence for Autism, 95 boulevard Pinel, 69675, Bron cedex, France
- Institute of Cognitive Science Marc Jeannerod, CNRS, UMR 5229, 67 boulevard Pinel, 69675, Bron cedex, France
| | - Caroline Demily
- Le Vinatier Hospital Center, 95 boulevard Pinel, 69675, Bron cedex, France
- iMIND, Center of Excellence for Autism, 95 boulevard Pinel, 69675, Bron cedex, France
- Institute of Cognitive Science Marc Jeannerod, CNRS, UMR 5229, 67 boulevard Pinel, 69675, Bron cedex, France
- University Claude Bernard Lyon 1, 43 boulevard du 11 Novembre 1918, 69622, Villeurbanne cedex, France
| |
Collapse
|
46
|
Pérez-Mitta G, Sezgin Y, Wang W, MacKinnon R. Freestanding bilayer microscope for single-molecule imaging of membrane proteins. SCIENCE ADVANCES 2024; 10:eado4722. [PMID: 38905330 PMCID: PMC11192074 DOI: 10.1126/sciadv.ado4722] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Accepted: 05/16/2024] [Indexed: 06/23/2024]
Abstract
Integral membrane proteins (IMPs) constitute a large fraction of organismal proteomes, playing fundamental roles in physiology and disease. Despite their importance, the mechanisms underlying dynamic features of IMPs, such as anomalous diffusion, protein-protein interactions, and protein clustering, remain largely unknown due to the high complexity of cell membrane environments. Available methods for in vitro studies are insufficient to study IMP dynamics systematically. This publication introduces the freestanding bilayer microscope (FBM), which combines the advantages of freestanding bilayers with single-particle tracking. The FBM, based on planar lipid bilayers, enables the study of IMP dynamics with single-molecule resolution and unconstrained diffusion. This paper benchmarks the FBM against total internal reflection fluorescence imaging on supported bilayers and is used here to estimate ion channel open probability and to examine the diffusion behavior of an ion channel in phase-separated bilayers. The FBM emerges as a powerful tool to examine membrane protein/lipid organization and dynamics to understand cell membrane processes.
Collapse
Affiliation(s)
- Gonzalo Pérez-Mitta
- Laboratory of Molecular Neurobiology and Biophysics, Howard Hughes Medical Institute, The Rockefeller University, New York, NY, USA
| | - Yeliz Sezgin
- Laboratory of Molecular Neurobiology and Biophysics, Howard Hughes Medical Institute, The Rockefeller University, New York, NY, USA
| | | | - Roderick MacKinnon
- Laboratory of Molecular Neurobiology and Biophysics, Howard Hughes Medical Institute, The Rockefeller University, New York, NY, USA
| |
Collapse
|
47
|
Otomo K, Omura T, Nozawa Y, Edwards SJ, Sato Y, Saito Y, Yagishita S, Uchida H, Watakabe Y, Naitou K, Yanai R, Sahara N, Takagi S, Katayama R, Iwata Y, Shiokawa T, Hayakawa Y, Otsuka K, Watanabe-Takano H, Haneda Y, Fukuhara S, Fujiwara M, Nii T, Meno C, Takeshita N, Yashiro K, Rosales Rocabado JM, Kaku M, Yamada T, Oishi Y, Koike H, Cheng Y, Sekine K, Koga JI, Sugiyama K, Kimura K, Karube F, Kim H, Manabe I, Nemoto T, Tainaka K, Hamada A, Brismar H, Susaki EA. descSPIM: an affordable and easy-to-build light-sheet microscope optimized for tissue clearing techniques. Nat Commun 2024; 15:4941. [PMID: 38866781 PMCID: PMC11169475 DOI: 10.1038/s41467-024-49131-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Accepted: 05/24/2024] [Indexed: 06/14/2024] Open
Abstract
Despite widespread adoption of tissue clearing techniques in recent years, poor access to suitable light-sheet fluorescence microscopes remains a major obstacle for biomedical end-users. Here, we present descSPIM (desktop-equipped SPIM for cleared specimens), a low-cost ($20,000-50,000), low-expertise (one-day installation by a non-expert), yet practical do-it-yourself light-sheet microscope as a solution for this bottleneck. Even the most fundamental configuration of descSPIM enables multi-color imaging of whole mouse brains and a cancer cell line-derived xenograft tumor mass for the visualization of neurocircuitry, assessment of drug distribution, and pathological examination by false-colored hematoxylin and eosin staining in a three-dimensional manner. Academically open-sourced ( https://github.com/dbsb-juntendo/descSPIM ), descSPIM allows routine three-dimensional imaging of cleared samples in minutes. Thus, the dissemination of descSPIM will accelerate biomedical discoveries driven by tissue clearing technologies.
Collapse
Affiliation(s)
- Kohei Otomo
- Department of Biochemistry and Systems Biomedicine, Juntendo University Graduate School of Medicine, Tokyo, Japan
- Biochemistry II, Juntendo University School of Medicine, Tokyo, Japan
- Nakatani Biomedical Spatialomics Hub, Juntendo University Graduate School of Medicine, Tokyo, Japan
- Division of Biophotonics, National Institute for Physiological Sciences, National Institutes of Natural Sciences, Okazaki, Japan
- Biophotonics Research Group, Exploratory Research Center on Life and Living Systems, National Institutes of Natural Sciences, Okazaki, Japan
| | - Takaki Omura
- Department of Biochemistry and Systems Biomedicine, Juntendo University Graduate School of Medicine, Tokyo, Japan
- Nakatani Biomedical Spatialomics Hub, Juntendo University Graduate School of Medicine, Tokyo, Japan
- Department of Neurosurgery, University of Tokyo, Tokyo, Japan
- Department of Neurosurgery and Neuro-Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Yuki Nozawa
- Biochemistry II, Juntendo University School of Medicine, Tokyo, Japan
| | - Steven J Edwards
- Science for Life Laboratory, Department of Applied Physics, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Yukihiko Sato
- Department of Biochemistry and Systems Biomedicine, Juntendo University Graduate School of Medicine, Tokyo, Japan
- Nakatani Biomedical Spatialomics Hub, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Yuri Saito
- Department of Biochemistry and Systems Biomedicine, Juntendo University Graduate School of Medicine, Tokyo, Japan
- Nakatani Biomedical Spatialomics Hub, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Shigehiro Yagishita
- Department of Pharmacology and Therapeutics, Fundamental Innovative Oncology Core, National Cancer Center Research Institute, Tokyo, Japan
- Division of Molecular Pharmacology, National Cancer Center Research Institute, Tokyo, Japan
| | - Hitoshi Uchida
- Department of System Pathology for Neurological Disorders, Brain Research Institute, Niigata University, Niigata, Japan
| | - Yuki Watakabe
- Division of Biophotonics, National Institute for Physiological Sciences, National Institutes of Natural Sciences, Okazaki, Japan
- Biophotonics Research Group, Exploratory Research Center on Life and Living Systems, National Institutes of Natural Sciences, Okazaki, Japan
| | - Kiyotada Naitou
- Department of Basic Veterinary Science, Joint Faculty of Veterinary Medicine, Kagoshima University, Kagoshima, Japan
| | - Rin Yanai
- Advanced Neuroimaging Center, Institute for Quantum Medical Sciences, National Institutes for Quantum Science and Technology, Chiba, Japan
| | - Naruhiko Sahara
- Advanced Neuroimaging Center, Institute for Quantum Medical Sciences, National Institutes for Quantum Science and Technology, Chiba, Japan
| | - Satoshi Takagi
- Division of Experimental Chemotherapy, Cancer Chemotherapy Center, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Ryohei Katayama
- Division of Experimental Chemotherapy, Cancer Chemotherapy Center, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Yusuke Iwata
- Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Toshiro Shiokawa
- Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Yoku Hayakawa
- Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Kensuke Otsuka
- Biology and Environmental Chemistry Division, Sustainable System Research Laboratory, Central Research Institute of Electric Power Industry, Chiba, Japan
| | - Haruko Watanabe-Takano
- Department of Molecular Pathophysiology, Institute for Advanced Medical Sciences, Nippon Medical School, Tokyo, Japan
| | - Yuka Haneda
- Department of Molecular Pathophysiology, Institute for Advanced Medical Sciences, Nippon Medical School, Tokyo, Japan
| | - Shigetomo Fukuhara
- Department of Molecular Pathophysiology, Institute for Advanced Medical Sciences, Nippon Medical School, Tokyo, Japan
| | - Miku Fujiwara
- Department of Developmental Biology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Takenobu Nii
- Department of Developmental Biology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Chikara Meno
- Department of Developmental Biology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Naoki Takeshita
- Anatomy and Developmental Biology, Kyoto Prefectural University of Medicine, Kyoto, Japan
- Department of Pediatrics, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Kenta Yashiro
- Anatomy and Developmental Biology, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Juan Marcelo Rosales Rocabado
- Division of Bio-prosthodontics, Faculty of Dentistry & Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
| | - Masaru Kaku
- Division of Bio-prosthodontics, Faculty of Dentistry & Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
| | - Tatsuya Yamada
- Department of Biochemistry, University of Nebraska-Lincoln, Lincoln, USA
| | - Yumiko Oishi
- Department of Meidical Biochemistry, Tokyo Medical and Dental University, Tokyo, Japan
| | - Hiroyuki Koike
- Department of Meidical Biochemistry, Tokyo Medical and Dental University, Tokyo, Japan
| | - Yinglan Cheng
- Department of Meidical Biochemistry, Tokyo Medical and Dental University, Tokyo, Japan
| | - Keisuke Sekine
- Laboratory of Cancer Cell Systems, National Cancer Center Research Institute, Tokyo, Japan
| | - Jun-Ichiro Koga
- The Second Department of Internal Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Kaori Sugiyama
- Institute for Advanced Research of Biosystem Dynamics, Research Institute for Science and Engineering, Waseda University, Tokyo, Japan
| | - Kenichi Kimura
- Life Science Center for Survival Dynamics, Tsukuba Advanced Research Alliance (TARA), University of Tsukuba, Tsukuba, Japan
| | - Fuyuki Karube
- Lab of Histology and Cytology, Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Hyeree Kim
- Department of Systems Medicine, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Ichiro Manabe
- Department of Systems Medicine, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Tomomi Nemoto
- Division of Biophotonics, National Institute for Physiological Sciences, National Institutes of Natural Sciences, Okazaki, Japan
- Biophotonics Research Group, Exploratory Research Center on Life and Living Systems, National Institutes of Natural Sciences, Okazaki, Japan
| | - Kazuki Tainaka
- Department of System Pathology for Neurological Disorders, Brain Research Institute, Niigata University, Niigata, Japan
| | - Akinobu Hamada
- Department of Pharmacology and Therapeutics, Fundamental Innovative Oncology Core, National Cancer Center Research Institute, Tokyo, Japan
- Division of Molecular Pharmacology, National Cancer Center Research Institute, Tokyo, Japan
| | - Hjalmar Brismar
- Science for Life Laboratory, Department of Applied Physics, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Etsuo A Susaki
- Department of Biochemistry and Systems Biomedicine, Juntendo University Graduate School of Medicine, Tokyo, Japan.
- Biochemistry II, Juntendo University School of Medicine, Tokyo, Japan.
- Nakatani Biomedical Spatialomics Hub, Juntendo University Graduate School of Medicine, Tokyo, Japan.
| |
Collapse
|
48
|
Katoh TA, Fukai YT, Ishibashi T. Optical microscopic imaging, manipulation, and analysis methods for morphogenesis research. Microscopy (Oxf) 2024; 73:226-242. [PMID: 38102756 PMCID: PMC11154147 DOI: 10.1093/jmicro/dfad059] [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: 06/30/2023] [Revised: 11/20/2023] [Accepted: 03/22/2024] [Indexed: 12/17/2023] Open
Abstract
Morphogenesis is a developmental process of organisms being shaped through complex and cooperative cellular movements. To understand the interplay between genetic programs and the resulting multicellular morphogenesis, it is essential to characterize the morphologies and dynamics at the single-cell level and to understand how physical forces serve as both signaling components and driving forces of tissue deformations. In recent years, advances in microscopy techniques have led to improvements in imaging speed, resolution and depth. Concurrently, the development of various software packages has supported large-scale, analyses of challenging images at the single-cell resolution. While these tools have enhanced our ability to examine dynamics of cells and mechanical processes during morphogenesis, their effective integration requires specialized expertise. With this background, this review provides a practical overview of those techniques. First, we introduce microscopic techniques for multicellular imaging and image analysis software tools with a focus on cell segmentation and tracking. Second, we provide an overview of cutting-edge techniques for mechanical manipulation of cells and tissues. Finally, we introduce recent findings on morphogenetic mechanisms and mechanosensations that have been achieved by effectively combining microscopy, image analysis tools and mechanical manipulation techniques.
Collapse
Affiliation(s)
- Takanobu A Katoh
- Department of Cell Biology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Yohsuke T Fukai
- Nonequilibrium Physics of Living Matter RIKEN Hakubi Research Team, RIKEN Center for Biosystems Dynamics Research, 2-2-3 Minatojima-minamimachi, Chuo-ku, Kobe, Hyogo 650-0047, Japan
| | - Tomoki Ishibashi
- Laboratory for Physical Biology, RIKEN Center for Biosystems Dynamics Research, 2-2-3 Minatojima-minamimachi, Chuo-ku, Kobe, Hyogo 650-0047, Japan
| |
Collapse
|
49
|
Tveriakhina L, Scanavachi G, Egan ED, Da Cunha Correia RB, Martin AP, Rogers JM, Yodh JS, Aster JC, Kirchhausen T, Blacklow SC. Temporal dynamics and stoichiometry in human Notch signaling from Notch synaptic complex formation to nuclear entry of the Notch intracellular domain. Dev Cell 2024; 59:1425-1438.e8. [PMID: 38574735 DOI: 10.1016/j.devcel.2024.03.021] [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: 10/05/2023] [Revised: 01/10/2024] [Accepted: 03/11/2024] [Indexed: 04/06/2024]
Abstract
Mammalian Notch signaling occurs when the binding of Delta or Jagged to Notch stimulates the proteolytic release of the Notch intracellular domain (NICD), which enters the nucleus to control target gene expression. To determine the temporal dynamics of events associated with Notch signaling under native conditions, we fluorescently tagged Notch and Delta at their endogenous genomic loci and visualized them upon pairing of receiver (Notch) and sender (Delta) cells as a function of time after cell contact. At contact sites, Notch and Delta immediately accumulated at 1:1 stoichiometry in synapses, which resolved by 15-20 min after contact. Synapse formation preceded the entrance of the Notch extracellular domain into the sender cell and accumulation of NICD in the nucleus of the receiver cell, which approached a maximum after ∼45 min and was prevented by chemical and genetic inhibitors of signaling. These findings directly link Notch-Delta synapse dynamics to NICD production with spatiotemporal precision.
Collapse
Affiliation(s)
- Lena Tveriakhina
- Department of Biological Chemistry and Molecular Pharmacology, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Gustavo Scanavachi
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA; Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA 02115, USA
| | - Emily D Egan
- Department of Biological Chemistry and Molecular Pharmacology, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Ricardo Bango Da Cunha Correia
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA; Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA 02115, USA
| | - Alexandre P Martin
- Department of Biological Chemistry and Molecular Pharmacology, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Julia M Rogers
- Department of Biological Chemistry and Molecular Pharmacology, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Jeremy S Yodh
- Department of Physics, Harvard University, Cambridge, MA 02138, USA
| | - Jon C Aster
- Department of Pathology, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Tom Kirchhausen
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA; Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA 02115, USA; Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA.
| | - Stephen C Blacklow
- Department of Biological Chemistry and Molecular Pharmacology, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA; Department of Cancer Biology, Dana Farber Cancer Institute, Boston, MA 02215, USA.
| |
Collapse
|
50
|
Teixeira P, Galland R, Chevrollier A. Super-resolution microscopies, technological breakthrough to decipher mitochondrial structure and dynamic. Semin Cell Dev Biol 2024; 159-160:38-51. [PMID: 38310707 DOI: 10.1016/j.semcdb.2024.01.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Revised: 01/08/2024] [Accepted: 01/25/2024] [Indexed: 02/06/2024]
Abstract
Mitochondria are complex organelles with an outer membrane enveloping a second inner membrane that creates a vast matrix space partitioned by pockets or cristae that join the peripheral inner membrane with several thin junctions. Several micrometres long, mitochondria are generally close to 300 nm in diameter, with membrane layers separated by a few tens of nanometres. Ultrastructural data from electron microscopy revealed the structure of these mitochondria, while conventional optical microscopy revealed their extraordinary dynamics through fusion, fission, and migration processes but its limited resolution power restricted the possibility to go further. By overcoming the limits of light diffraction, Super-Resolution Microscopy (SRM) now offers the potential to establish the links between the ultrastructure and remodelling of mitochondrial membranes, leading to major advances in our understanding of mitochondria's structure-function. Here we review the contributions of SRM imaging to our understanding of the relationship between mitochondrial structure and function. What are the hopes for these new imaging approaches which are particularly important for mitochondrial pathologies?
Collapse
Affiliation(s)
- Pauline Teixeira
- Univ. Angers, INSERM, CNRS, MITOVASC, Equipe MITOLAB, SFR ICAT, F-49000 Angers, France
| | - Rémi Galland
- Univ. Bordeaux, CNRS, Interdisciplinary Institute for Neuroscience, IINS, UMR 5297, F-33000 Bordeaux, France
| | - Arnaud Chevrollier
- Univ. Angers, INSERM, CNRS, MITOVASC, Equipe MITOLAB, SFR ICAT, F-49000 Angers, France.
| |
Collapse
|