1
|
Li Z, Zhang X, Li G, Peng J, Su X. Light scattering imaging modal expansion cytometry for label-free single-cell analysis with deep learning. COMPUTER METHODS AND PROGRAMS IN BIOMEDICINE 2025; 264:108726. [PMID: 40112688 DOI: 10.1016/j.cmpb.2025.108726] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/14/2025] [Revised: 03/02/2025] [Accepted: 03/14/2025] [Indexed: 03/22/2025]
Abstract
BACKGROUND AND OBJECTIVE Single-cell imaging plays a key role in various fields, including drug development, disease diagnosis, and personalized medicine. To obtain multi-modal information from a single-cell image, especially for label-free cells, this study develops modal expansion cytometry for label-free single-cell analysis. METHODS The study utilizes a deep learning-based architecture to expand single-mode light scattering images into multi-modality images, including bright-field (non-fluorescent) and fluorescence images, for label-free single-cell analysis. By combining adversarial loss, L1 distance loss, and VGG perceptual loss, a new network optimization method is proposed. The effectiveness of this method is verified by experiments on simulated images, standard spheres of different sizes, and multiple cell types (such as cervical cancer and leukemia cells). Additionally, the capability of this method in single-cell analysis is assessed through multi-modal cell classification experiments, such as cervical cancer subtypes. RESULTS This is demonstrated by using both cervical cancer cells and leukemia cells. The expanded bright-field and fluorescence images derived from the light scattering images align closely with those obtained through conventional microscopy, showing a contour ratio near 1 for both the whole cell and its nucleus. Using machine learning, the subtyping of cervical cancer cells achieved 92.85 % accuracy with the modal expansion images, which represents an improvement of nearly 20 % over single-mode light scattering images. CONCLUSIONS This study demonstrates the light scattering imaging modal expansion cytometry with deep learning has the capability to expand the single-mode light scattering image into the artificial multimodal images of label-free single cells, which not only provides the visualization of cells but also helps for the cell classification, showing great potential in the field of single-cell analysis such as cancer cell diagnosis.
Collapse
Affiliation(s)
- Zhi Li
- School of Integrated Circuits, Shandong University, Jinan 250101, China; Institute of Biomedical Engineering, School of Control Science and Engineering, Shandong University, Jinan 250061, China
| | - Xiaoyu Zhang
- Department of Hematology, Qilu Hospital of Shandong University, Jinan 250012, China
| | - Guosheng Li
- Department of Hematology, Qilu Hospital of Shandong University, Jinan 250012, China
| | - Jun Peng
- Department of Hematology, Qilu Hospital of Shandong University, Jinan 250012, China
| | - Xuantao Su
- School of Integrated Circuits, Shandong University, Jinan 250101, China.
| |
Collapse
|
2
|
Brandi C, De Ninno A, Ruggiero F, Mussi V, Nanni M, Caselli F. Analysis of Single Nuclei in a Microfluidic Cytometer Towards Metaphase Enrichment. Electrophoresis 2025. [PMID: 40347085 DOI: 10.1002/elps.8152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Revised: 04/03/2025] [Accepted: 04/28/2025] [Indexed: 05/12/2025]
Abstract
Identifying analyzable metaphase chromosomes is crucial for karyotyping, a common procedure used by clinicians to diagnose genetic disorders and some forms of cancer. This task is often laborious and time-consuming, making it essential to develop automated, efficient, and reliable methods to assist clinical technicians. In this work, an original label-free microfluidic approach to identify potential metaphases is developed that uses impedance-based detection of individual flowing nuclei and machine-learning-based processing of synchronized high-speed videos. Specifically, impedance signals are used to identify nucleus-containing frames, which are then processed to extract the contour of each nucleus. Feature extraction is then performed, and both unsupervised and supervised classification approaches are implemented to identify potential metaphases from those features. The proposed framework is tested on K562 cells, and the highest classification accuracy is obtained with the supervised approach coupled with a feature selection procedure and the Synthetic Minority Over-sampling Technique (SMOTE). Overall, this study encourages future developments aimed at integrating a sorting functionality in the device, thus achieving an effective microfluidic system for metaphase enrichment.
Collapse
Affiliation(s)
- Cristian Brandi
- Department of Civil Engineering and Computer Science, University of Rome Tor Vergata, Rome, Italy
| | - Adele De Ninno
- Institute for Photonics and Nanotechnology, Italian National Research Council, Rome, Italy
| | - Filippo Ruggiero
- Institute for Photonics and Nanotechnology, Italian National Research Council, Rome, Italy
| | - Valentina Mussi
- Institute for Microelectronics and Microsystems, Italian National Research Council, Rome, Italy
| | - Mauro Nanni
- Department of Translational and Precision Medicine, Sapienza University, Rome, Italy
| | - Federica Caselli
- Department of Civil Engineering and Computer Science, University of Rome Tor Vergata, Rome, Italy
| |
Collapse
|
3
|
Liu Z, Zhang Y, Li J, Chen S, Zhao H, Zhao X, Sun D. Gray-Level Guided Image-Activated Droplet Sorter for Label-Free, High-Accuracy Screening of Single-Cell on Demand. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2025:e2500520. [PMID: 40342217 DOI: 10.1002/smll.202500520] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/14/2025] [Revised: 04/03/2025] [Indexed: 05/11/2025]
Abstract
Single-cell encapsulation in droplet microfluidics has become a powerful tool in precision medicine, single-cell analysis, and immunotherapy. However, droplet generation with a single-cell encapsulation is a random process, which also results in a large number of empty and multi-cell droplets. Current microfluidics sorting technologies suffer from drawbacks such as fluorescent labeling, inability to remove multi-cell droplets, or low throughput. This paper presents a gray-level guided image-activated droplet sorter (GL-IADS), which enables label-free, high-accuracy screening of single-cell droplets by rejecting empty and multi-cell droplets. The gray-level based recognition method can accurately classify droplet images (empty, single-cell, and multi-cell droplets), especially in differentiating empty and cell-laden droplets (accuracy of 100%). Crucially, this method reduces the image processing time to ≈300 µs, which makes the GL-IADS possible to reach an ultra-high sorting throughput up to hundreds or even KHz. The GL-IADS integrates the novel recognition method with a detachable acoustofluidic system, achieving sorting purity of 97.9%, 97.4%, and >99% for single-cell, multi-cell, and cell-laden droplets, respectively, with a throughput of 43 Hz. The GL-IADS holds promise for numerous biological applications that are previously difficult with fluorescence-based technologies.
Collapse
Affiliation(s)
- Zhen Liu
- Department of Biomedical Engineering, City University of Hong Kong, Hong Kong SAR, China
| | - Yidi Zhang
- Department of Biomedical Engineering, City University of Hong Kong, Hong Kong SAR, China
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, Engineering Research Center of Trusted Behavior Intelligence, Ministry of Education, Tianjin Key Laboratory of Intelligent Robotic (tjKLIR), Institute of Robotics and Automatic Information System (IRAIS), Nankai University, Tianjin, 300350, China
| | - Jianing Li
- Department of Biomedical Engineering, City University of Hong Kong, Hong Kong SAR, China
| | - Shuxun Chen
- Department of Biomedical Engineering, City University of Hong Kong, Hong Kong SAR, China
| | - Han Zhao
- Department of Biomedical Engineering, City University of Hong Kong, Hong Kong SAR, China
| | - Xin Zhao
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, Engineering Research Center of Trusted Behavior Intelligence, Ministry of Education, Tianjin Key Laboratory of Intelligent Robotic (tjKLIR), Institute of Robotics and Automatic Information System (IRAIS), Nankai University, Tianjin, 300350, China
- Institute of Intelligence Technology and Robotic Systems, Shenzhen Research Institute of Nankai University, Shenzhen, 518083, China
| | - Dong Sun
- Department of Biomedical Engineering, City University of Hong Kong, Hong Kong SAR, China
| |
Collapse
|
4
|
Gao L, Yuan J, Hong K, Ma NL, Liu S, Wu X. Technological advancement spurs Komagataella phaffii as a next-generation platform for sustainable biomanufacturing. Biotechnol Adv 2025; 82:108593. [PMID: 40339766 DOI: 10.1016/j.biotechadv.2025.108593] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2025] [Revised: 04/11/2025] [Accepted: 05/01/2025] [Indexed: 05/10/2025]
Abstract
Biomanufacturing stands as a cornerstone of sustainable industrial development, necessitating a shift toward non-food carbon feedstocks to alleviate agricultural resource competition and advance a circular bioeconomy. Methanol, a renewable one‑carbon substrate, has emerged as a pivotal candidate due to its abundance, cost-effectiveness, and high reduction potential, further bolstered by breakthroughs in CO₂ hydrogenation-based synthesis. Capitalizing on this momentum, the methylotrophic yeast Komagataella phaffii has undergone transformative technological upgrades, evolving from a conventional protein expression workhorse into an intelligent bioproduction chassis. This paradigm shift is fundamentally driven by converging innovations across CRISPR-empowered advancement in genome editing and AI-powered metabolic pathway design in K. phaffii. The integration of CRISPR systems with droplet microfluidics high-throughput screening has redefined strain engineering efficiency, achieving much higher editing precision than traditional homologous recombination while compressing the "design-build-test-learn" cycle. Concurrently, machine learning-enhanced genome-scale metabolic models facilitate dynamic flux balancing, enabling simultaneous improvements in product titers, carbon yields, and volumetric productivity. Finally, technological advancement promotes the application of K. phaffii, including directing more efficiently metabolic flux toward nutrient products, and strengthening efficient synthesis of excreted proteins. As DNA synthesis automation and robotic experimentation platforms mature, next-generation breakthroughs in genome modification, cofactor engineering, and AI-guided autonomous evolution will further cement K. phaffii as a next-generation platform for decarbonizing global manufacturing paradigms. This technological trajectory positions methanol-based biomanufacturing as a cornerstone of the low-carbon circular economy.
Collapse
Affiliation(s)
- Le Gao
- Key Laboratory of Engineering Biology for Low-Carbon Manufacturing, Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences, National Center of Technology Innovation for Synthetic Biology, No. 32, Xiqi Road, Tianjin Airport Economic Park, Tianjin 300308, China.
| | - Jie Yuan
- Key Laboratory of Engineering Biology for Low-Carbon Manufacturing, Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences, National Center of Technology Innovation for Synthetic Biology, No. 32, Xiqi Road, Tianjin Airport Economic Park, Tianjin 300308, China
| | - Kai Hong
- Key Laboratory of Engineering Biology for Low-Carbon Manufacturing, Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences, National Center of Technology Innovation for Synthetic Biology, No. 32, Xiqi Road, Tianjin Airport Economic Park, Tianjin 300308, China
| | - Nyuk Ling Ma
- Institute of Tropical Biodiversity and Sustainable Development, University Malaysia Terengganu, Malaysia
| | - Shuguang Liu
- Beijing Chasing future Biotechnology Co., Ltd, Beijing, China
| | - Xin Wu
- Key Laboratory of Engineering Biology for Low-Carbon Manufacturing, Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences, National Center of Technology Innovation for Synthetic Biology, No. 32, Xiqi Road, Tianjin Airport Economic Park, Tianjin 300308, China.
| |
Collapse
|
5
|
Gao G, Shao T, Li T, Wang S. Harnessing optical forces with advanced nanophotonic structures: principles and applications. DISCOVER NANO 2025; 20:76. [PMID: 40317364 PMCID: PMC12049358 DOI: 10.1186/s11671-025-04252-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/26/2025] [Accepted: 04/09/2025] [Indexed: 05/07/2025]
Abstract
Non-contact mechanical control of light has given rise to optical manipulation, facilitating diverse light-matter interactions and enabling pioneering applications like optical tweezers. However, the practical adoption of versatile optical tweezing systems remains constrained by the complexity and bulkiness of their optical setups, underscoring the urgent requirement for advancements in miniaturization and functional integration. In this paper, we present innovations in optical manipulation within the nanophotonic domain, including fiber-based and metamaterial tweezers, as well as their emerging applications in manipulating cells and artificial micro-nano robots. Furthermore, we explore interdisciplinary on-chip devices that integrate photonic crystals and optofluidics. By merging optical manipulation with the dynamism of nanophotonics and metamaterials, this work seeks to chart a transformative pathway for the future of optomechanics and beyond.
Collapse
Affiliation(s)
- Geze Gao
- National Laboratory of Solid-State Microstructures, Collaborative Innovation Center of Advanced Microstructures, School of Physics, Nanjing University, Nanjing, 210093, China
| | - Tianhua Shao
- National Laboratory of Solid-State Microstructures, Collaborative Innovation Center of Advanced Microstructures, School of Physics, Nanjing University, Nanjing, 210093, China
| | - Tianyue Li
- National Laboratory of Solid-State Microstructures, Collaborative Innovation Center of Advanced Microstructures, School of Physics, Nanjing University, Nanjing, 210093, China.
| | - Shuming Wang
- National Laboratory of Solid-State Microstructures, Collaborative Innovation Center of Advanced Microstructures, School of Physics, Nanjing University, Nanjing, 210093, China.
| |
Collapse
|
6
|
Rembotte L, Beneyton T, Buisson L, Badon A, Boyreau A, Douillet C, Hermant L, Jana A, Nassoy P, Baret J. Pheno-Morphological Screening and Acoustic Sorting of 3D Multicellular Aggregates Using Drop Millifluidics. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2410677. [PMID: 39792815 PMCID: PMC11884609 DOI: 10.1002/advs.202410677] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Revised: 12/20/2024] [Indexed: 01/12/2025]
Abstract
Three-dimensional multicellular aggregates (MCAs) like organoids and spheroids have become essential tools to study the biological mechanisms involved in the progression of diseases. In cancer research, they are now widely used as in vitro models for drug testing. However, their analysis still relies on tedious manual procedures, which hinders their routine use in large-scale biological assays. Here, a novel drop millifluidic approach is introduced to screen and sort large populations containing over one thousand MCAs: ImOCAS (Image-based Organoid Cytometry and Acoustic Sorting). This system utilizes real-time image processing to detect pheno-morphological traits in MCAs. They are then encapsulated in millimetric drops, actuated on-demand using the acoustic radiation force. The performance of ImOCAS is demonstrated by sorting spheroids with uniform sizes from a heterogeneous population, and by isolating organoids from spheroids with different phenotypes. This approach lays the groundwork for high-throughput screening and high-content analysis of MCAs with controlled morphological and phenotypical properties, which promises accelerated progress in biomedical research.
Collapse
Affiliation(s)
- Leon Rembotte
- CNRSUniv. BordeauxCRPPUMR 5031PessacF‐33600France
- LP2NUniv. BordeauxTalenceF‐33400France
| | | | | | - Amaury Badon
- LP2NUniv. BordeauxTalenceF‐33400France
- IOGSCNRSUMR5298TalenceF‐33400France
| | | | - Camille Douillet
- LP2NUniv. BordeauxTalenceF‐33400France
- IOGSCNRSUMR5298TalenceF‐33400France
| | - Loic Hermant
- LP2NUniv. BordeauxTalenceF‐33400France
- IOGSCNRSUMR5298TalenceF‐33400France
| | - Anirban Jana
- LP2NUniv. BordeauxTalenceF‐33400France
- TreeFrog TherapeuticsPessacF‐33600France
| | - Pierre Nassoy
- LP2NUniv. BordeauxTalenceF‐33400France
- IOGSCNRSUMR5298TalenceF‐33400France
| | | |
Collapse
|
7
|
Yang M, Shi Y, Song Q, Wei Z, Dun X, Wang Z, Wang Z, Qiu CW, Zhang H, Cheng X. Optical sorting: past, present and future. LIGHT, SCIENCE & APPLICATIONS 2025; 14:103. [PMID: 40011460 DOI: 10.1038/s41377-024-01734-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Revised: 12/02/2024] [Accepted: 12/24/2024] [Indexed: 02/28/2025]
Abstract
Optical sorting combines optical tweezers with diverse techniques, including optical spectrum, artificial intelligence (AI) and immunoassay, to endow unprecedented capabilities in particle sorting. In comparison to other methods such as microfluidics, acoustics and electrophoresis, optical sorting offers appreciable advantages in nanoscale precision, high resolution, non-invasiveness, and is becoming increasingly indispensable in fields of biophysics, chemistry, and materials science. This review aims to offer a comprehensive overview of the history, development, and perspectives of various optical sorting techniques, categorised as passive and active sorting methods. To begin, we elucidate the fundamental physics and attributes of both conventional and exotic optical forces. We then explore sorting capabilities of active optical sorting, which fuses optical tweezers with a diversity of techniques, including Raman spectroscopy and machine learning. Afterwards, we reveal the essential roles played by deterministic light fields, configured with lens systems or metasurfaces, in the passive sorting of particles based on their varying sizes and shapes, sorting resolutions and speeds. We conclude with our vision of the most promising and futuristic directions, including AI-facilitated ultrafast and bio-morphology-selective sorting. It can be envisioned that optical sorting will inevitably become a revolutionary tool in scientific research and practical biomedical applications.
Collapse
Affiliation(s)
- Meng Yang
- Institute of Precision Optical Engineering, School of Physics Science and Engineering, Tongji University, Shanghai, 200092, China
- MOE Key Laboratory of Advanced Micro-Structured Materials, Shanghai, 200092, China
- Shanghai Institute of Intelligent Science and Technology, Tongji University, Shanghai, 200092, China
- Shanghai Frontiers Science Center of Digital Optics, Shanghai, 200092, China
| | - Yuzhi Shi
- Institute of Precision Optical Engineering, School of Physics Science and Engineering, Tongji University, Shanghai, 200092, China.
- MOE Key Laboratory of Advanced Micro-Structured Materials, Shanghai, 200092, China.
- Shanghai Institute of Intelligent Science and Technology, Tongji University, Shanghai, 200092, China.
- Shanghai Frontiers Science Center of Digital Optics, Shanghai, 200092, China.
| | - Qinghua Song
- Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen, 518055, China
| | - Zeyong Wei
- Institute of Precision Optical Engineering, School of Physics Science and Engineering, Tongji University, Shanghai, 200092, China
- MOE Key Laboratory of Advanced Micro-Structured Materials, Shanghai, 200092, China
- Shanghai Institute of Intelligent Science and Technology, Tongji University, Shanghai, 200092, China
- Shanghai Frontiers Science Center of Digital Optics, Shanghai, 200092, China
| | - Xiong Dun
- Institute of Precision Optical Engineering, School of Physics Science and Engineering, Tongji University, Shanghai, 200092, China
- MOE Key Laboratory of Advanced Micro-Structured Materials, Shanghai, 200092, China
- Shanghai Institute of Intelligent Science and Technology, Tongji University, Shanghai, 200092, China
- Shanghai Frontiers Science Center of Digital Optics, Shanghai, 200092, China
| | - Zhiming Wang
- Institute of Fundamental and Frontier Sciences, University of Electronic Science and Technology of China, Chengdu, 610054, China
| | - Zhanshan Wang
- Institute of Precision Optical Engineering, School of Physics Science and Engineering, Tongji University, Shanghai, 200092, China
- MOE Key Laboratory of Advanced Micro-Structured Materials, Shanghai, 200092, China
- Shanghai Institute of Intelligent Science and Technology, Tongji University, Shanghai, 200092, China
- Shanghai Frontiers Science Center of Digital Optics, Shanghai, 200092, China
| | - Cheng-Wei Qiu
- Department of Electrical and Computer Engineering, National University of Singapore, Singapore, 117583, Singapore.
| | - Hui Zhang
- Institute of Precision Optical Engineering, School of Physics Science and Engineering, Tongji University, Shanghai, 200092, China.
- MOE Key Laboratory of Advanced Micro-Structured Materials, Shanghai, 200092, China.
- Shanghai Institute of Intelligent Science and Technology, Tongji University, Shanghai, 200092, China.
- Shanghai Frontiers Science Center of Digital Optics, Shanghai, 200092, China.
| | - Xinbin Cheng
- Institute of Precision Optical Engineering, School of Physics Science and Engineering, Tongji University, Shanghai, 200092, China.
- MOE Key Laboratory of Advanced Micro-Structured Materials, Shanghai, 200092, China.
- Shanghai Institute of Intelligent Science and Technology, Tongji University, Shanghai, 200092, China.
- Shanghai Frontiers Science Center of Digital Optics, Shanghai, 200092, China.
| |
Collapse
|
8
|
Venugopal Menon N, Lee J, Tang T, Lim CT. Microfluidics for morpholomics and spatial omics applications. LAB ON A CHIP 2025; 25:752-763. [PMID: 39865877 DOI: 10.1039/d4lc00869c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/28/2025]
Abstract
Creative designs, precise fluidic manipulation, and automation have supported the development of microfluidics for single-cell applications. Together with the advancements in detection technologies and artificial intelligence (AI), microfluidic-assisted platforms have been increasingly used for new modalities of single-cell investigations and in spatial omics applications. This review explores the use of microfluidic technologies for morpholomics and spatial omics with a focus on single-cell and tissue characterization. We emphasize how various fluid dynamic principles and unique design integrations enable highly precise fluid manipulation, enhancing sample handling in morpholomics. Additionally, we examine the use of microfluidics-assisted spatial barcoding with micrometer resolutions for the spatial profiling of tissue specimens. Finally, we discuss how microfluidics can serve as a bridge for integrating multiple unique fields in omics research and outline key challenges that these technologies may face in practical translation.
Collapse
Affiliation(s)
- Nishanth Venugopal Menon
- Mechanobiology Institute, National University of Singapore, Singapore, 117411 Singapore
- Institute for Digital Molecular Analytics and Science, Nanyang Technological University, 636921, Singapore
| | - Jeeyeon Lee
- Institute for Health Innovation and Technology (iHealthtech), National University of Singapore, Singapore, 117599 Singapore
| | - Tao Tang
- Department of Biomedical Engineering, National University of Singapore, 117583, Singapore
| | - Chwee Teck Lim
- Mechanobiology Institute, National University of Singapore, Singapore, 117411 Singapore
- Institute for Health Innovation and Technology (iHealthtech), National University of Singapore, Singapore, 117599 Singapore
- Department of Biomedical Engineering, National University of Singapore, 117583, Singapore
- Institute for Digital Molecular Analytics and Science, Nanyang Technological University, 636921, Singapore
| |
Collapse
|
9
|
Zhang Z, Zhu Y, Lai Z, Zhou M, Chen X, Tang R, Alaynick W, Cho SH, Lo YH. Predicting cell properties with AI from 3D imaging flow cytometer data. Sci Rep 2025; 15:5715. [PMID: 39962067 PMCID: PMC11833109 DOI: 10.1038/s41598-024-80722-6] [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: 08/10/2024] [Accepted: 11/21/2024] [Indexed: 02/20/2025] Open
Abstract
Predicting the properties of tissues or organisms from the genomics data is widely accepted by the medical community. Here we ask a question: can we predict the properties of each individual cell? Single-cell genomics does not work because the RNA sequencing process destroys the cell, not allowing us to verify our predictions. To test the hypothesis, we investigate the approach of using AI to analyze single-cell images obtained from a 3D imaging flow cytometer. We analyze the cell image at day zero and make the AI-assisted cell property prediction. The prediction is then examined later when the cells continue to live and develop. Our preliminary results are promising, showing 88% accuracy in predicting cells that will have a high protein expression level. The technique can have strong ramifications and impact on preventive medicine, drug development, cell therapy, and fundamental biomedical research.
Collapse
Affiliation(s)
- Zunming Zhang
- Department of Electrical and Computer Engineering, University of California, San Diego, La Jolla, CA, 92093, USA
| | - Yuxuan Zhu
- Department of Electrical and Computer Engineering, University of California, San Diego, La Jolla, CA, 92093, USA
| | - Zhaoyu Lai
- Department of Electrical and Computer Engineering, University of California, San Diego, La Jolla, CA, 92093, USA
| | - Minhong Zhou
- Department of Electrical and Computer Engineering, University of California, San Diego, La Jolla, CA, 92093, USA
| | - Xinyu Chen
- Department of Electrical and Computer Engineering, University of California, San Diego, La Jolla, CA, 92093, USA
| | - Rui Tang
- NanoCellect Biomedical Inc., San Diego, CA, 92121, USA
| | | | - Sung Hwan Cho
- NanoCellect Biomedical Inc., San Diego, CA, 92121, USA
| | - Yu-Hwa Lo
- Department of Electrical and Computer Engineering, University of California, San Diego, La Jolla, CA, 92093, USA.
| |
Collapse
|
10
|
Wang Z, Kelley SO. Microfluidic technologies for enhancing the potency, predictability and affordability of adoptive cell therapies. Nat Biomed Eng 2025:10.1038/s41551-024-01315-2. [PMID: 39953325 DOI: 10.1038/s41551-024-01315-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2024] [Accepted: 10/31/2024] [Indexed: 02/17/2025]
Abstract
The development and wider adoption of adoptive cell therapies is constrained by complex and costly manufacturing processes and by inconsistent efficacy across patients. Here we discuss how microfluidic and other fluidic devices can be implemented at each stage of cell manufacturing for adoptive cell therapies, from the harvesting and isolation of the cells to their editing, culturing and functional selection. We suggest that precise and controllable microfluidic systems can streamline the development of these therapies by offering scalability in cell production, bolstering the efficacy and predictability of the therapies and improving their cost-effectiveness and accessibility for broader populations of patients with cancer.
Collapse
Affiliation(s)
- Zongjie Wang
- Chan Zuckerberg Biohub Chicago, Chicago, IL, USA
- Department of Biomedical Engineering, McCormick School of Engineering, Northwestern University, Evanston, IL, USA
| | - Shana O Kelley
- Chan Zuckerberg Biohub Chicago, Chicago, IL, USA.
- Department of Biomedical Engineering, McCormick School of Engineering, Northwestern University, Evanston, IL, USA.
- Department of Chemistry, Weinberg College of Arts & Sciences, Northwestern University, Evanston, IL, USA.
- Department of Biochemistry, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA.
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, IL, USA.
- International Institute for Nanotechnology, Northwestern University, Evanston, IL, USA.
- Simpson Querrey Institute, Northwestern University, Chicago, IL, USA.
| |
Collapse
|
11
|
Yang C, He C, Zhuo H, Wang J, Yong T, Gan L, Yang X, Nie L, Xi S, Liu Z, Liao G, Shi T. Cost-effective microfluidic flow cytometry for precise and gentle cell sorting. LAB ON A CHIP 2025; 25:698-713. [PMID: 39895391 DOI: 10.1039/d4lc00900b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/04/2025]
Abstract
Microfluidic flow cytometry (MFCM) is considered to be an effective substitute for traditional flow cytometry, because of its advantages in terms of higher integration, smaller device size, lower cost, and higher cell sorting activity. However, MFCM still faces challenges in balancing parameters such as sorting throughput, viability, sorting efficiency, and cost. Here, we demonstrate a cost-effective and high-performance microfluidic cytometry cell sorting system, along with a customized microfluidic chip that integrates hydrodynamic focusing, droplet encapsulation, and sorting for precise cell manipulation. An innovative photon incremental counting-based fluorescence detection method is proposed, which requires only one-fiftieth of the data compared to traditional methods. This significantly simplifies the structure of the system and substantially reduces costs. The system exhibits detection recoveries exceeding 95% across sample solution flow rates ranging from 10 to 80 μL min-1. Moreover, it accurately achieves individual droplet deflections at a droplet generation frequency of 1600 Hz. Ultimately, our cell sorting system offers an impressive sorting efficiency of 90.7% and a high cell viability of 94.3% when operating at a droplet generation frequency of 1316 Hz, highlighting its accuracy and gentleness throughout the entire process. Our work will enhance advances in the life sciences, thereby creating a boom in great applications in single-cell cloning, single-cell analysis, drug screening, etc.
Collapse
Affiliation(s)
- Canfeng Yang
- State Key Laboratory of Digital Manufacturing Equipment and Technology, School of Mechanical Science & Engineering, Huazhong University of Science and Technology, Wuhan 430074, China.
| | - Chunhua He
- State Key Laboratory of Digital Manufacturing Equipment and Technology, School of Mechanical Science & Engineering, Huazhong University of Science and Technology, Wuhan 430074, China.
| | - Huasheng Zhuo
- State Key Laboratory of Digital Manufacturing Equipment and Technology, School of Mechanical Science & Engineering, Huazhong University of Science and Technology, Wuhan 430074, China.
| | - Jianxin Wang
- State Key Laboratory of Digital Manufacturing Equipment and Technology, School of Mechanical Science & Engineering, Huazhong University of Science and Technology, Wuhan 430074, China.
| | - Tuying Yong
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Lu Gan
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Xiangliang Yang
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Lei Nie
- School of Mechanical Engineering, Hubei University of Technology, Wuhan 430068, China
| | - Shuang Xi
- College of Mechanical and Electronic Engineering, Nanjing Forestry University, Nanjing, 210037, China
| | - Zhiyong Liu
- State Key Laboratory of Digital Manufacturing Equipment and Technology, School of Mechanical Science & Engineering, Huazhong University of Science and Technology, Wuhan 430074, China.
| | - Guanglan Liao
- State Key Laboratory of Digital Manufacturing Equipment and Technology, School of Mechanical Science & Engineering, Huazhong University of Science and Technology, Wuhan 430074, China.
| | - Tielin Shi
- State Key Laboratory of Digital Manufacturing Equipment and Technology, School of Mechanical Science & Engineering, Huazhong University of Science and Technology, Wuhan 430074, China.
| |
Collapse
|
12
|
Zhou J, Mei L, Yu M, Ma X, Hou D, Yin Z, Liu X, Ding Y, Yang K, Xiao R, Yuan X, Weng Y, Long M, Hu T, Hou J, Xu Y, Tao L, Mei S, Shen H, Yalikun Y, Zhou F, Wang L, Wang D, Liu S, Lei C. Imaging flow cytometry with a real-time throughput beyond 1,000,000 events per second. LIGHT, SCIENCE & APPLICATIONS 2025; 14:76. [PMID: 39924500 PMCID: PMC11808109 DOI: 10.1038/s41377-025-01754-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Revised: 12/09/2024] [Accepted: 01/08/2025] [Indexed: 02/11/2025]
Abstract
Imaging flow cytometry (IFC) combines the imaging capabilities of microscopy with the high throughput of flow cytometry, offering a promising solution for high-precision and high-throughput cell analysis in fields such as biomedicine, green energy, and environmental monitoring. However, due to limitations in imaging framerate and real-time data processing, the real-time throughput of existing IFC systems has been restricted to approximately 1000-10,000 events per second (eps), which is insufficient for large-scale cell analysis. In this work, we demonstrate IFC with real-time throughput exceeding 1,000,000 eps by integrating optical time-stretch (OTS) imaging, microfluidic-based cell manipulation, and online image processing. Cells flowing at speeds up to 15 m/s are clearly imaged with a spatial resolution of 780 nm, and images of each individual cell are captured, stored, and analyzed. The capabilities and performance of our system are validated through the identification of malignancies in clinical colorectal samples. This work sets a new record for throughput in imaging flow cytometry, and we believe it has the potential to revolutionize cell analysis by enabling highly efficient, accurate, and intelligent measurement.
Collapse
Affiliation(s)
- Jiehua Zhou
- The Institute of Technological Sciences, Wuhan University, Wuhan, 430072, China
| | - Liye Mei
- The Institute of Technological Sciences, Wuhan University, Wuhan, 430072, China
- School of Computer Science, Hubei University of Technology, Wuhan, 430068, China
| | - Mingjie Yu
- The Institute of Technological Sciences, Wuhan University, Wuhan, 430072, China
| | - Xiao Ma
- The Institute of Technological Sciences, Wuhan University, Wuhan, 430072, China
| | - Dan Hou
- The Institute of Technological Sciences, Wuhan University, Wuhan, 430072, China
| | - Zhuo Yin
- The Institute of Technological Sciences, Wuhan University, Wuhan, 430072, China
| | - Xun Liu
- The Institute of Technological Sciences, Wuhan University, Wuhan, 430072, China
- Division of Materials Science, Nara Institute of Science and Technology, Takayama-cho, 8916-5, Japan
| | - Yan Ding
- The Institute of Technological Sciences, Wuhan University, Wuhan, 430072, China
| | - Kaining Yang
- The Institute of Technological Sciences, Wuhan University, Wuhan, 430072, China
| | - Ruidong Xiao
- The Institute of Technological Sciences, Wuhan University, Wuhan, 430072, China
| | - Xiandan Yuan
- The Institute of Technological Sciences, Wuhan University, Wuhan, 430072, China
- School of Science, Hubei University of Technology, Wuhan, 430068, China
| | - Yueyun Weng
- The Institute of Technological Sciences, Wuhan University, Wuhan, 430072, China
| | - Mengping Long
- The Institute of Technological Sciences, Wuhan University, Wuhan, 430072, China
- Department of Pathology, Peking University Cancer Hospital, Beijing, 100142, China
| | - Taobo Hu
- The Institute of Technological Sciences, Wuhan University, Wuhan, 430072, China
- Department of Breast Surgery, Peking University People's Hospital, Beijing, 100044, China
| | - Jinxuan Hou
- Department of Thyroid and Breast Surgery, Zhongnan Hospital, Wuhan University, Wuhan, 430071, China
| | - Yu Xu
- Department of Radiation and Medical Oncology, Zhongnan Hospital, Wuhan University, Wuhan, 430071, China
| | - Liang Tao
- People's Hospital of Anshun City Guizhou Province, Anshun, 561000, China
| | - Sisi Mei
- People's Hospital of Anshun City Guizhou Province, Anshun, 561000, China
| | - Hui Shen
- Department of Hematology, Zhongnan Hospital, Wuhan University, Wuhan, 430071, China
| | - Yaxiaer Yalikun
- Division of Materials Science, Nara Institute of Science and Technology, Takayama-cho, 8916-5, Japan
| | - Fuling Zhou
- Department of Hematology, Zhongnan Hospital, Wuhan University, Wuhan, 430071, China
| | - Liang Wang
- National Engineering Laboratory for Next Generation Internet Access System, School of Optics and Electronic Information, Huazhong University of Science and Technology, Wuhan, 430074, China.
| | - Du Wang
- The Institute of Technological Sciences, Wuhan University, Wuhan, 430072, China.
| | - Sheng Liu
- The Institute of Technological Sciences, Wuhan University, Wuhan, 430072, China
| | - Cheng Lei
- The Institute of Technological Sciences, Wuhan University, Wuhan, 430072, China.
- Suzhou Institute of Wuhan University, Suzhou, 215000, China.
- Shenzhen Institute of Wuhan University, Shenzhen, 518057, China.
| |
Collapse
|
13
|
Jo J, Hugonnet H, Lee MJ, Park Y. Digital Cytometry: Extraction of Forward and Side Scattering Signals From Holotomography. JOURNAL OF BIOPHOTONICS 2025:e202400387. [PMID: 39906965 DOI: 10.1002/jbio.202400387] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Revised: 01/11/2025] [Accepted: 01/12/2025] [Indexed: 02/06/2025]
Abstract
Flow cytometry is a cornerstone technique in medical and biological research, providing crucial information about cell size and granularity through forward scatter (FSC) and side scatter (SSC) signals. Despite its widespread use, the precise relationship between these scatter signals and corresponding microscopic images remains underexplored. Here, we investigate this intrinsic relationship by utilizing scattering theory and holotomography, a three-dimensional quantitative phase imaging (QPI) technique. We demonstrate the extraction of FSC and SSC signals from individual, unlabeled cells by analyzing their three-dimensional refractive index distributions obtained through holotomography. Additionally, we introduce a method for digital windowing of SSC signals to facilitate effective segmentation and morphology-based cell type classification. Our approach bridges the gap between flow cytometry and microscopic imaging, offering a new perspective on analyzing cellular characteristics with high accuracy and without the need for labeling.
Collapse
Affiliation(s)
- Jaepil Jo
- Department of Physics, Korea Advanced Institute of Science and Technology, Daejeon, Republic of Korea
- KAIST Institute for Health Science and Technology, KAIST, Daejeon, Republic of Korea
- Semiconductor R&D Center, Samsung Electronics Co. Ltd., Hwaseong-si, Gyeonggi-do, Republic of Korea
| | - Herve Hugonnet
- Department of Physics, Korea Advanced Institute of Science and Technology, Daejeon, Republic of Korea
- KAIST Institute for Health Science and Technology, KAIST, Daejeon, Republic of Korea
| | - Mahn Jae Lee
- KAIST Institute for Health Science and Technology, KAIST, Daejeon, Republic of Korea
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, South Korea
| | - YongKeun Park
- Department of Physics, Korea Advanced Institute of Science and Technology, Daejeon, Republic of Korea
- KAIST Institute for Health Science and Technology, KAIST, Daejeon, Republic of Korea
- Tomocube Inc., Daejeon, South Korea
| |
Collapse
|
14
|
Carré J, Demont Y, Mouton C, Vayne C, Guéry E, Voyer A, Garçon L, Le Guyader M, Demagny J. Imaging flow cytometry as a novel approach for the diagnosis of heparin-induced thrombocytopenia. Br J Haematol 2025; 206:666-674. [PMID: 39658032 PMCID: PMC11829136 DOI: 10.1111/bjh.19945] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2024] [Accepted: 11/29/2024] [Indexed: 12/12/2024]
Abstract
Heparin-induced thrombocytopenia (HIT) is an adverse reaction characterized by anti-PF4-heparin antibody generation and hypercoagulability. Imaging flow cytometry (IFC) provides a detailed morphological analysis of platelets, which change upon activation. We evaluated IFC-derived morphometric features to detect platelet activation and developed a functional assay for HIT diagnosis. We analysed blood samples from 42 patients with suspected HIT and extracted platelet size, shape and texture features using IFC. The morphological features were compared with CD62P expression, light transmission aggregometry (LTA) and a serotonin release assay (SRA) in terms of their ability to predict a HIT diagnosis. Five IFC-derived morphological features (area, circularity, contrast, diameter and major axis) significantly distinguished resting from activated platelets. The major axis feature performed best for HIT diagnosis, with a sensitivity of 89.3% and a specificity of 92.9% versus functional assays (LTA/SRA); this diagnostic performance was similar to that of CD62P expression on the same platelet donors. The area and diameter had similar specificity (92.9%) and a slightly lower sensitivity (85.7%). The morphological features associated with platelet activation might be effective markers for the diagnosis of HIT, matching platelet CD62P expression assay performance. The high-throughput IFC exploration of platelet activation offers new perspectives in label-free analysis and time-saving.
Collapse
Affiliation(s)
- Julie Carré
- Service d'Hématologie BiologiqueCHU Amiens‐PicardieAmiensFrance
| | - Yohann Demont
- Service d'Hématologie BiologiqueCHU Amiens‐PicardieAmiensFrance
| | - Christine Mouton
- Laboratoire d'HématologieHôpital Haut‐Lévêque, CHU BordeauxBordeauxFrance
| | - Caroline Vayne
- Service d'Hématologie‐HémostaseCHRU ToursToursFrance
- INSERM UMR1327 Ischemia, Université de ToursToursFrance
| | | | - Annelise Voyer
- Service d'Hématologie BiologiqueCHU Amiens‐PicardieAmiensFrance
| | - Loïc Garçon
- Service d'Hématologie BiologiqueCHU Amiens‐PicardieAmiensFrance
- HEMATIM UR666, Jules Verne University of PicardieAmiensFrance
| | | | - Julien Demagny
- Service d'Hématologie BiologiqueCHU Amiens‐PicardieAmiensFrance
- HEMATIM UR666, Jules Verne University of PicardieAmiensFrance
| |
Collapse
|
15
|
Isozaki A, Kita K, Tiffany Ishii N, Oka Y, Herbig M, Yamagishi M, Wakamiya T, Araki T, Matsumura H, Harmon J, Shirasaki Y, Huang K, Zhao Y, Yuan D, Hayashi M, Ding T, Okamoto Y, Kishimoto A, Ishii M, Yanagida M, Goda K. Investigating T-Cell Receptor Dynamics Under In Vitro Antibody-Based Stimulation Using Imaging Flow Cytometry. Cytometry A 2025; 107:88-97. [PMID: 39982013 DOI: 10.1002/cyto.a.24916] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Revised: 11/14/2024] [Accepted: 02/05/2025] [Indexed: 02/22/2025]
Abstract
T cells play a pivotal role in the immune system's response to various conditions. They are activated by antigen-presenting cells (APCs) via T-cell surface receptors, resulting in cytokine production and T-cell proliferation. These interactions occur through the formation of immunological synapses. The advent of imaging flow cytometry has enabled detailed statistical analyses of these cellular interactions. However, the dynamics of T-cell receptors in response to in vitro stimulation are yet to receive attention, despite it being a crucial aspect of understanding T-cell behavior. In this article, we explore the responses of T cells to in vitro antibody-based stimulation without APCs. Specifically, we established a Th1 cell clone, subjected it to a combination of centrifugation-induced mechanical stress and anti-human CD3 and anti-human CD28 antibody stimulation as the in vitro antibody-based stimulation, and captured and analyzed bright-field and fluorescence images of single cells various hours after stimulation using an imaging flow cytometer. Our results indicate distinct temporal dynamics of CD3 and CD28. Notably, CD3 and CD28 relocated on the T-cell surface immediately after stimulation, with CD3 receptors dispersing after 3.5 h, whereas CD28 remained clustered for 7.5 h. These receptor morphological changes precede cytokine production, suggesting their potential as early indicators of T-cell activation.
Collapse
Affiliation(s)
- Akihiro Isozaki
- Department of Mechanical Engineering, Ritsumeikan University, Kusatsu, Shiga, Japan
- Department of Chemistry, The University of Tokyo, Tokyo, Japan
| | - Kazuma Kita
- Central Research Laboratories, Sysmex Corporation, Kobe, Japan
| | | | - Yuma Oka
- Central Research Laboratories, Sysmex Corporation, Kobe, Japan
| | - Maik Herbig
- Department of Chemistry, The University of Tokyo, Tokyo, Japan
| | | | | | - Taketo Araki
- Department of Chemistry, The University of Tokyo, Tokyo, Japan
| | | | - Jeffrey Harmon
- Department of Chemistry, The University of Tokyo, Tokyo, Japan
| | - Yoshitaka Shirasaki
- Research Center for Advanced Science and Technology, The University of Tokyo, Tokyo, Japan
| | - Kangrui Huang
- Department of Chemistry, The University of Tokyo, Tokyo, Japan
| | - Yaqi Zhao
- Department of Chemistry, The University of Tokyo, Tokyo, Japan
| | - Dan Yuan
- Department of Chemistry, The University of Tokyo, Tokyo, Japan
| | - Mika Hayashi
- Department of Chemistry, The University of Tokyo, Tokyo, Japan
| | - Tianben Ding
- Department of Chemistry, The University of Tokyo, Tokyo, Japan
| | - Yuji Okamoto
- Central Research Laboratories, Sysmex Corporation, Kobe, Japan
| | - Ayuko Kishimoto
- Central Research Laboratories, Sysmex Corporation, Kobe, Japan
| | - Masaru Ishii
- Department of Immunology and Cell Biology, Osaka University, Osaka, Japan
| | | | - Keisuke Goda
- Department of Chemistry, The University of Tokyo, Tokyo, Japan
- CYBO, Tokyo, Japan
- Department of Bioengineering, University of California, Los Angeles, California, USA
- Institute of Technological Sciences, Wuhan University, Wuhan, China
| |
Collapse
|
16
|
Jarmoshti J, Siddique A, Rane A, Mirhosseini S, Adair SJ, Bauer TW, Caselli F, Swami NS. Neural Network-Enabled Multiparametric Impedance Signal Templating for High throughput Single-Cell Deformability Cytometry Under Viscoelastic Extensional Flows. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2025; 21:e2407212. [PMID: 39439143 PMCID: PMC11798358 DOI: 10.1002/smll.202407212] [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: 08/17/2024] [Revised: 10/08/2024] [Indexed: 10/25/2024]
Abstract
Cellular biophysical metrics exhibit systematic alterations during processes, such as metastasis and immune cell activation, which can be used to identify and separate live cell subpopulations for targeting drug screening. Image-based biophysical cytometry under extensional flows can accurately quantify cell deformability based on cell shape alterations but needs extensive image reconstruction, which limits its inline utilization to activate cell sorting. Impedance cytometry can measure these cell shape alterations based on electric field screening, while its frequency response offers functional information on cell viability and interior structure, which are difficult to discern by imaging. Furthermore, 1-D temporal impedance signal trains exhibit characteristic shapes that can be rapidly templated in near real-time to extract single-cell biophysical metrics to activate sorting. We present a multilayer perceptron neural network signal templating approach that utilizes raw impedance signals from cells under extensional flow, alongside its training with image metrics from corresponding cells to derive net electrical anisotropy metrics that quantify cell deformability over wide anisotropy ranges and with minimal errors from cell size distributions. Deformability and electrical physiology metrics are applied in conjunction on the same cell for multiparametric classification of live pancreatic cancer cells versus cancer associated fibroblasts using the support vector machine model.
Collapse
Affiliation(s)
- Javad Jarmoshti
- Electrical & Computer EngineeringUniversity of VirginiaCharlottesvilleVA22904USA
| | | | - Aditya Rane
- Chemistry, University of VirginiaUniversity of VirginiaCharlottesvilleVA22904USA
| | | | - Sara J. Adair
- Surgery, School of MedicineUniversity of VirginiaCharlottesvilleVA22903USA
| | - Todd W. Bauer
- Surgery, School of MedicineUniversity of VirginiaCharlottesvilleVA22903USA
| | - Federica Caselli
- Civil Engineering and Computer ScienceUniversity of Rome Tor VergataRome00133Italy
| | - Nathan S. Swami
- Electrical & Computer EngineeringUniversity of VirginiaCharlottesvilleVA22904USA
- Chemistry, University of VirginiaUniversity of VirginiaCharlottesvilleVA22904USA
| |
Collapse
|
17
|
McIntyre D, Arguijo D, Kawata K, Densmore D. Component library creation and pixel array generation with micromilled droplet microfluidics. MICROSYSTEMS & NANOENGINEERING 2025; 11:6. [PMID: 39809750 PMCID: PMC11733136 DOI: 10.1038/s41378-024-00839-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Accepted: 10/19/2024] [Indexed: 01/16/2025]
Abstract
Droplet microfluidics enable high-throughput screening, sequencing, and formulation of biological and chemical systems at the microscale. Such devices are generally fabricated in a soft polymer such as polydimethylsiloxane (PDMS). However, developing design masks for PDMS devices can be a slow and expensive process, requiring an internal cleanroom facility or using an external vendor. Here, we present the first complete droplet-based component library using low-cost rapid prototyping and electrode integration. This fabrication method for droplet microfluidic devices costs less than $12 per device and a full design-build-test cycle can be completed within a day. Discrete microfluidic components for droplet generation, re-injection, picoinjection, anchoring, fluorescence sensing, and sorting were built and characterized. These devices are biocompatible, low-cost, and high-throughput. To show its ability to perform multistep workflows, these components were used to assemble droplet "pixel" arrays, where droplets were generated, sensed, sorted, and anchored onto a grid to produce images.
Collapse
Affiliation(s)
- David McIntyre
- Biomedical Engineering Department, Boston University, Boston, MA, USA
- Biological Design Center, Boston University, Boston, MA, USA
| | - Diana Arguijo
- Biomedical Engineering Department, Boston University, Boston, MA, USA
- Biological Design Center, Boston University, Boston, MA, USA
| | - Kaede Kawata
- Biological Design Center, Boston University, Boston, MA, USA
- Electrical and Computer Engineering Department, Boston University, Boston, MA, USA
| | - Douglas Densmore
- Biological Design Center, Boston University, Boston, MA, USA.
- Electrical and Computer Engineering Department, Boston University, Boston, MA, USA.
| |
Collapse
|
18
|
Guo Z, Li F, Li H, Zhao M, Liu H, Wang H, Hu H, Fu R, Lu Y, Hu S, Xie H, Ma H, Zhang S. Deep Learning-Assisted Label-Free Parallel Cell Sorting with Digital Microfluidics. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2408353. [PMID: 39497614 PMCID: PMC11906218 DOI: 10.1002/advs.202408353] [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: 07/21/2024] [Revised: 10/03/2024] [Indexed: 01/11/2025]
Abstract
Sorting specific cells from heterogeneous samples is important for research and clinical applications. In this work, a novel label-free cell sorting method is presented that integrates deep learning image recognition with microfluidic manipulation to differentiate cells based on morphology. Using an Active-Matrix Digital Microfluidics (AM-DMF) platform, the YOLOv8 object detection model ensures precise droplet classification, and the Safe Interval Path Planning algorithm manages multi-target, collision-free droplet path planning. Simulations and experiments revealed that detection model precision, concentration ratios, and sorting cycles significantly affect recovery rates and purity. With HeLa cells and polystyrene beads as samples, the method achieved 98.5% sorting precision, 96.49% purity, and an 80% recovery over three cycles. After a series of experimental validations, this method can also be used to sort HeLa cells from red blood cells, cancer cells from white blood cells (represented by HeLa and Jurkat cells), and differentiate white blood cell subtypes (represented by HL-60 cells and Jurkat cells). Cells sorted using this method can be lysed directly on chip within their hosting droplets, ensuring minimal sample loss and suitability for downstream bioanalysis. This innovative AM-DMF cell sorting technique holds significant potential to advance diagnostics, therapeutics, and fundamental research in cell biology.
Collapse
Affiliation(s)
- Zongliang Guo
- Beijing Advanced Innovation Center for Intelligent Robots and Systems, School of Mechatronical Engineering, Beijing Institute of Technology, Beijing, 100081, China
| | - Fenggang Li
- Beijing Advanced Innovation Center for Intelligent Robots and Systems, School of Mechatronical Engineering, Beijing Institute of Technology, Beijing, 100081, China
| | - Hang Li
- School of Medical Technology, Beijing Institute of Technology, Beijing, 100081, China
| | - Menglei Zhao
- Beijing Advanced Innovation Center for Intelligent Robots and Systems, School of Mechatronical Engineering, Beijing Institute of Technology, Beijing, 100081, China
| | - Haobing Liu
- Beijing Advanced Innovation Center for Intelligent Robots and Systems, School of Mechatronical Engineering, Beijing Institute of Technology, Beijing, 100081, China
| | - Haopu Wang
- School of Integrated Circuits and Electronics, Engineering Research Center of Integrated Acousto-Opto-Electronic Microsystems (Ministry of Education of China), Beijing Institute of Technology, Beijing, 100081, China
| | - Hanqi Hu
- School of Medical Technology, Beijing Institute of Technology, Beijing, 100081, China
| | - Rongxin Fu
- School of Medical Technology, Beijing Institute of Technology, Beijing, 100081, China
| | - Yao Lu
- School of Integrated Circuits and Electronics, Engineering Research Center of Integrated Acousto-Opto-Electronic Microsystems (Ministry of Education of China), Beijing Institute of Technology, Beijing, 100081, China
| | - Siyi Hu
- CAS Key Laboratory of Bio-Medical Diagnostics, Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou, 215163, China
- ACX Instruments Ltd, St John's Innovation Centre, Cambridge, CB40WS, UK
| | - Huikai Xie
- School of Integrated Circuits and Electronics, Engineering Research Center of Integrated Acousto-Opto-Electronic Microsystems (Ministry of Education of China), Beijing Institute of Technology, Beijing, 100081, China
| | - Hanbin Ma
- CAS Key Laboratory of Bio-Medical Diagnostics, Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou, 215163, China
- ACX Instruments Ltd, St John's Innovation Centre, Cambridge, CB40WS, UK
| | - Shuailong Zhang
- Beijing Advanced Innovation Center for Intelligent Robots and Systems, School of Mechatronical Engineering, Beijing Institute of Technology, Beijing, 100081, China
- School of Medical Technology, Beijing Institute of Technology, Beijing, 100081, China
- School of Integrated Circuits and Electronics, Engineering Research Center of Integrated Acousto-Opto-Electronic Microsystems (Ministry of Education of China), Beijing Institute of Technology, Beijing, 100081, China
| |
Collapse
|
19
|
He W, Zhu J, Feng Y, Liang F, You K, Chai H, Sui Z, Hao H, Li G, Zhao J, Deng L, Zhao R, Wang W. Neuromorphic-enabled video-activated cell sorting. Nat Commun 2024; 15:10792. [PMID: 39737963 DOI: 10.1038/s41467-024-55094-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Accepted: 11/29/2024] [Indexed: 01/01/2025] Open
Abstract
Imaging flow cytometry allows image-activated cell sorting (IACS) with enhanced feature dimensions in cellular morphology, structure, and composition. However, existing IACS frameworks suffer from the challenges of 3D information loss and processing latency dilemma in real-time sorting operation. Herein, we establish a neuromorphic-enabled video-activated cell sorter (NEVACS) framework, designed to achieve high-dimensional spatiotemporal characterization content alongside high-throughput sorting of particles in wide field of view. NEVACS adopts event camera, CPU, spiking neural networks deployed on a neuromorphic chip, and achieves sorting throughput of 1000 cells/s with relatively economic hybrid hardware solution (~$10 K for control) and simple-to-make-and-use microfluidic infrastructures. Particularly, the application of NEVACS in classifying regular red blood cells and blood-disease-relevant spherocytes highlights the accuracy of using video over a single frame (i.e., average error of 0.99% vs 19.93%), indicating NEVACS' potential in cell morphology screening and disease diagnosis.
Collapse
Affiliation(s)
- Weihua He
- State Key Laboratory of Precision Measurement Technology and Instrument, Department of Precision Instrument, Tsinghua University, Beijing, China
| | - Junwen Zhu
- State Key Laboratory of Precision Measurement Technology and Instrument, Department of Precision Instrument, Tsinghua University, Beijing, China
| | - Yongxiang Feng
- State Key Laboratory of Precision Measurement Technology and Instrument, Department of Precision Instrument, Tsinghua University, Beijing, China
| | - Fei Liang
- State Key Laboratory of Precision Measurement Technology and Instrument, Department of Precision Instrument, Tsinghua University, Beijing, China
| | - Kaichao You
- Software School, Tsinghua University, Beijing, China
| | - Huichao Chai
- State Key Laboratory of Precision Measurement Technology and Instrument, Department of Precision Instrument, Tsinghua University, Beijing, China
| | - Zhipeng Sui
- State Key Laboratory of Precision Measurement Technology and Instrument, Department of Precision Instrument, Tsinghua University, Beijing, China
| | - Haiqing Hao
- State Key Laboratory of Precision Measurement Technology and Instrument, Department of Precision Instrument, Tsinghua University, Beijing, China
| | - Guoqi Li
- Institute of Automation, Chinese Academy of Sciences, Beijing, China
| | - Jingjing Zhao
- Institute of Medical Equipment Science and Engineering, Huazhong University of Science and Technology, Wuhan, China
| | - Lei Deng
- Center for Brain-Inspired Computing Research (CBICR), Beijing Advanced Innovation Center for Integrated Circuits, Optical Memory National Engineering Research Center, & Department of Precision Instrument, Tsinghua University, Beijing, China
| | - Rong Zhao
- Center for Brain-Inspired Computing Research (CBICR), Beijing Advanced Innovation Center for Integrated Circuits, Optical Memory National Engineering Research Center, & Department of Precision Instrument, Tsinghua University, Beijing, China
| | - Wenhui Wang
- State Key Laboratory of Precision Measurement Technology and Instrument, Department of Precision Instrument, Tsinghua University, Beijing, China.
| |
Collapse
|
20
|
Wang J, Zhao X, Wang Y, Li D. Color-multiplexed 3D differential phase contrast microscopy with optimal annular illumination. OPTICS EXPRESS 2024; 32:49135-49152. [PMID: 39876200 DOI: 10.1364/oe.545480] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Accepted: 12/12/2024] [Indexed: 01/30/2025]
Abstract
Quantitative phase imaging (QPI) has become a valuable tool in the field of biomedical research due to its ability to quantify refractive index variations of live cells and tissues. For example, three-dimensional differential phase contrast (3D DPC) imaging uses through-focus images captured under different illumination patterns deconvoluted with a computed 3D phase transfer function (PTF) to reconstruct the 3D refractive index. In conventional 3D DPC with semi-circular illumination, partially spatially coherent illumination often diminishes phase contrast, exacerbating inherent noise, and can lead to a large number of zero values in the 3D PTF, resulting in strong low-frequency artifacts and deteriorating imaging resolution. To overcome the above drawbacks, we obtain the conditions for acquiring the optimal 3D PTF based on the analysis of the 3D imaging model and the derivation of the 3D PTF calculation process and propose a 3D DPC microscopy based on optimal annular illumination. The proposed optimal annular illumination pattern minimizes the missing frequency components in the 3D Fourier space, resulting in the best noise-robustness and significantly increased phase contrast. To expedite imaging speed, we utilize a 1/2 annular multiplexed illumination, reducing data acquisition volume by 75%. The 3D refractive index tomography of a simulated 3D phase object, unstained tongue sections, and oral epithelial cells demonstrates that our proposed method achieves the above advantages. In conclusion, we demonstrate a novel 3D DPC microscope that only requires replacing the illumination of a commercial microscope with a programmable LED array. The accurate 3D refractive index tomography and the compactness of the system setup allow the method to play a significant role in the biomedical field.
Collapse
|
21
|
Ugawa M, Ota S. Recent Technologies on 2D and 3D Imaging Flow Cytometry. Cells 2024; 13:2073. [PMID: 39768164 PMCID: PMC11674929 DOI: 10.3390/cells13242073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Revised: 12/11/2024] [Accepted: 12/13/2024] [Indexed: 01/11/2025] Open
Abstract
Imaging flow cytometry is a technology that performs microscopy image analysis of cells within flow cytometry and allows high-throughput, high-content cell analysis based on their intracellular molecular distribution and/or cellular morphology. While the technology has been available for a couple of decades, it has recently gained significant attention as technical limitations for higher throughput, sorting capability, and additional imaging dimensions have been overcome with various approaches. These evolutions have enabled imaging flow cytometry to offer a variety of solutions for life science and medicine that are not possible with conventional flow cytometry or microscopy-based screening. It is anticipated that the extent of applications will expand in the upcoming years as the technology becomes more accessible through dissemination. In this review, we will cover the technical advances that have led to this new generation of imaging flow cytometry, focusing on the advantages and limitations of each technique.
Collapse
Affiliation(s)
- Masashi Ugawa
- Research Center for Advanced Science and Technology, University of Tokyo, Tokyo 153-8904, Japan
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, CA 94143, USA
| | - Sadao Ota
- Research Center for Advanced Science and Technology, University of Tokyo, Tokyo 153-8904, Japan
- ThinkCyte, Inc., Tokyo 113-0033, Japan
| |
Collapse
|
22
|
Ghanegolmohammadi F, Eslami M, Ohya Y. Systematic data analysis pipeline for quantitative morphological cell phenotyping. Comput Struct Biotechnol J 2024; 23:2949-2962. [PMID: 39104709 PMCID: PMC11298594 DOI: 10.1016/j.csbj.2024.07.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Revised: 07/09/2024] [Accepted: 07/10/2024] [Indexed: 08/07/2024] Open
Abstract
Quantitative morphological phenotyping (QMP) is an image-based method used to capture morphological features at both the cellular and population level. Its interdisciplinary nature, spanning from data collection to result analysis and interpretation, can lead to uncertainties, particularly among those new to this actively growing field. High analytical specificity for a typical QMP is achieved through sophisticated approaches that can leverage subtle cellular morphological changes. Here, we outline a systematic workflow to refine the QMP methodology. For a practical review, we describe the main steps of a typical QMP; in each step, we discuss the available methods, their applications, advantages, and disadvantages, along with the R functions and packages for easy implementation. This review does not cover theoretical backgrounds, but provides several references for interested researchers. It aims to broaden the horizons for future phenome studies and demonstrate how to exploit years of endeavors to achieve more with less.
Collapse
Affiliation(s)
- Farzan Ghanegolmohammadi
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
- Department of Integrated Biosciences, Graduate School of Frontier Sciences, The University of Tokyo, Kashiwa, Chiba, Japan
| | - Mohammad Eslami
- Harvard Ophthalmology AI Lab, Schepen’s Eye Research Institute of Massachusetts Eye and Ear Infirmary, Harvard Medical School, Boston, USA
| | - Yoshikazu Ohya
- Department of Integrated Biosciences, Graduate School of Frontier Sciences, The University of Tokyo, Kashiwa, Chiba, Japan
| |
Collapse
|
23
|
Chai B, Efstathiou C, Yue H, Draviam VM. Opportunities and challenges for deep learning in cell dynamics research. Trends Cell Biol 2024; 34:955-967. [PMID: 38030542 DOI: 10.1016/j.tcb.2023.10.010] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 09/30/2023] [Accepted: 10/13/2023] [Indexed: 12/01/2023]
Abstract
The growth of artificial intelligence (AI) has led to an increase in the adoption of computer vision and deep learning (DL) techniques for the evaluation of microscopy images and movies. This adoption has not only addressed hurdles in quantitative analysis of dynamic cell biological processes but has also started to support advances in drug development, precision medicine, and genome-phenome mapping. We survey existing AI-based techniques and tools, as well as open-source datasets, with a specific focus on the computational tasks of segmentation, classification, and tracking of cellular and subcellular structures and dynamics. We summarise long-standing challenges in microscopy video analysis from a computational perspective and review emerging research frontiers and innovative applications for DL-guided automation in cell dynamics research.
Collapse
Affiliation(s)
- Binghao Chai
- School of Biological and Behavioural Sciences, Queen Mary University of London (QMUL), London E1 4NS, UK
| | - Christoforos Efstathiou
- School of Biological and Behavioural Sciences, Queen Mary University of London (QMUL), London E1 4NS, UK
| | - Haoran Yue
- School of Biological and Behavioural Sciences, Queen Mary University of London (QMUL), London E1 4NS, UK
| | - Viji M Draviam
- School of Biological and Behavioural Sciences, Queen Mary University of London (QMUL), London E1 4NS, UK; The Alan Turing Institute, London NW1 2DB, UK.
| |
Collapse
|
24
|
Findinier J, Joubert LM, Fakhimi N, Schmid MF, Malkovskiy AV, Chiu W, Burlacot A, Grossman AR. Dramatic changes in mitochondrial subcellular location and morphology accompany activation of the CO 2 concentrating mechanism. Proc Natl Acad Sci U S A 2024; 121:e2407548121. [PMID: 39405346 PMCID: PMC11513932 DOI: 10.1073/pnas.2407548121] [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: 04/15/2024] [Accepted: 09/06/2024] [Indexed: 10/23/2024] Open
Abstract
Dynamic changes in intracellular ultrastructure can be critical for the ability of organisms to acclimate to environmental conditions. Microalgae, which are responsible for ~50% of global photosynthesis, compartmentalize their Ribulose 1,5 Bisphosphate Carboxylase/Oxygenase (Rubisco) into a specialized structure known as the pyrenoid when the cells experience limiting CO2 conditions; this compartmentalization is a component of the CO2 Concentrating Mechanism (CCM), which facilitates photosynthetic CO2 fixation as environmental levels of inorganic carbon (Ci) decline. Changes in the spatial distribution of mitochondria in green algae have also been observed under CO2 limitation, although a role for this reorganization in CCM function remains unclear. We used the green microalga Chlamydomonas reinhardtii to monitor changes in mitochondrial position and ultrastructure as cells transition between high CO2 and Low/Very Low CO2 (LC/VLC). Upon transferring cells to VLC, the mitochondria move from a central to a peripheral cell location and orient in parallel tubular arrays that extend along the cell's apico-basal axis. We show that these ultrastructural changes correlate with CCM induction and are regulated by the CCM master regulator CIA5. The apico-basal orientation of the mitochondrial membranes, but not the movement of the mitochondrion to the cell periphery, is dependent on microtubules and the MIRO1 protein, with the latter involved in membrane-microtubule interactions. Furthermore, blocking mitochondrial respiration in VLC-acclimated cells reduces the affinity of the cells for Ci. Overall, our results suggest that mitochondrial repositioning functions in integrating cellular architecture and energetics with CCM activities and invite further exploration of how intracellular architecture can impact fitness under dynamic environmental conditions.
Collapse
Affiliation(s)
- Justin Findinier
- The Carnegie Institution for Science, Biosphere Sciences and Engineering, Stanford, CA94305
| | - Lydia-Marie Joubert
- SLAC National Accelerator Laboratory, Division of CryoElectron Microscopy and Bioimaging, Menlo Park, CA94025
| | - Neda Fakhimi
- The Carnegie Institution for Science, Biosphere Sciences and Engineering, Stanford, CA94305
| | - Michael F. Schmid
- SLAC National Accelerator Laboratory, Division of CryoElectron Microscopy and Bioimaging, Menlo Park, CA94025
| | - Andrey V. Malkovskiy
- The Carnegie Institution for Science, Biosphere Sciences and Engineering, Stanford, CA94305
| | - Wah Chiu
- SLAC National Accelerator Laboratory, Division of CryoElectron Microscopy and Bioimaging, Menlo Park, CA94025
- Department of Bioengineering, Stanford University, Stanford, CA94305
| | - Adrien Burlacot
- The Carnegie Institution for Science, Biosphere Sciences and Engineering, Stanford, CA94305
- Biology Department, Stanford University, Stanford, CA94305
| | - Arthur R. Grossman
- The Carnegie Institution for Science, Biosphere Sciences and Engineering, Stanford, CA94305
- Biology Department, Stanford University, Stanford, CA94305
| |
Collapse
|
25
|
Zhang K, Xia Z, Wang Y, Zheng L, Li B, Chu J. Label-free high-throughput impedance-activated cell sorting. LAB ON A CHIP 2024; 24:4918-4929. [PMID: 39315634 DOI: 10.1039/d4lc00487f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/25/2024]
Abstract
Cell sorting holds broad applications in fields such as early cancer diagnosis, cell differentiation studies, drug screening, and single-cell sequencing. However, achieving high-throughput and high-purity in label-free single-cell sorting is challenging. To overcome this issue, we propose a label-free, high-throughput, and high-accuracy impedance-activated cell sorting system based on impedance detection and dual membrane pumps. Leveraging the low-latency characteristics of FPGA, the system facilitates real-time dual-frequency single-cell impedance detection with high-throughput (5 × 104 cells per s) for HeLa, MDA-MB-231, and Jurkat cells. Furthermore, the system accomplishes low-latency (less than 0.3 ms), label-free, high-throughput (1000 particles per s) and high-accuracy (almost 99%) single-particle sorting using FPGA-based high-precision sort-timing prediction. In experiments with Jurkat and MDA-MB-231 cells, the system achieved a throughput of up to 1000 cells per s, maintaining a pre-sorting purity of 28.57% and increasing post-sorting purity to 97.09%. These findings indicate that our system holds significant potential for applications in label-free, high-throughput cell sorting.
Collapse
Affiliation(s)
- Kui Zhang
- Department of Precision Machinery and Precision Instrumentation, University of Science and Technology of China, Hefei, Anhui, 230027, China.
- Key Laboratory of Precision Scientific Instrumentation of Anhui Higher Education Institutes, University of Science and Technology of China, Hefei, Anhui, 230027, China
| | - Ziyang Xia
- Department of Precision Machinery and Precision Instrumentation, University of Science and Technology of China, Hefei, Anhui, 230027, China.
- Key Laboratory of Precision Scientific Instrumentation of Anhui Higher Education Institutes, University of Science and Technology of China, Hefei, Anhui, 230027, China
| | - Yiming Wang
- Department of Precision Machinery and Precision Instrumentation, University of Science and Technology of China, Hefei, Anhui, 230027, China.
- Key Laboratory of Precision Scientific Instrumentation of Anhui Higher Education Institutes, University of Science and Technology of China, Hefei, Anhui, 230027, China
- Biomedical Robotics Laboratory, School of Biomedical Engineering, Anhui Medical University, Hefei, Anhui, 230032, China
| | - Lisheng Zheng
- Department of Precision Machinery and Precision Instrumentation, University of Science and Technology of China, Hefei, Anhui, 230027, China.
- Key Laboratory of Precision Scientific Instrumentation of Anhui Higher Education Institutes, University of Science and Technology of China, Hefei, Anhui, 230027, China
| | - Baoqing Li
- Department of Precision Machinery and Precision Instrumentation, University of Science and Technology of China, Hefei, Anhui, 230027, China.
- Key Laboratory of Precision Scientific Instrumentation of Anhui Higher Education Institutes, University of Science and Technology of China, Hefei, Anhui, 230027, China
| | - Jiaru Chu
- Department of Precision Machinery and Precision Instrumentation, University of Science and Technology of China, Hefei, Anhui, 230027, China.
- Key Laboratory of Precision Scientific Instrumentation of Anhui Higher Education Institutes, University of Science and Technology of China, Hefei, Anhui, 230027, China
| |
Collapse
|
26
|
Kanno H, Hiramatsu K, Mikami H, Nakayashiki A, Yamashita S, Nagai A, Okabe K, Li F, Yin F, Tominaga K, Bicer OF, Noma R, Kiani B, Efa O, Büscher M, Wazawa T, Sonoshita M, Shintaku H, Nagai T, Braun S, Houston JP, Rashad S, Niizuma K, Goda K. High-throughput fluorescence lifetime imaging flow cytometry. Nat Commun 2024; 15:7376. [PMID: 39231964 PMCID: PMC11375057 DOI: 10.1038/s41467-024-51125-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: 01/06/2024] [Accepted: 07/31/2024] [Indexed: 09/06/2024] Open
Abstract
Flow cytometry is a vital tool in biomedical research and laboratory medicine. However, its accuracy is often compromised by undesired fluctuations in fluorescence intensity. While fluorescence lifetime imaging microscopy (FLIM) bypasses this challenge as fluorescence lifetime remains unaffected by such fluctuations, the full integration of FLIM into flow cytometry has yet to be demonstrated due to speed limitations. Here we overcome the speed limitations in FLIM, thereby enabling high-throughput FLIM flow cytometry at a high rate of over 10,000 cells per second. This is made possible by using dual intensity-modulated continuous-wave beam arrays with complementary modulation frequency pairs for fluorophore excitation and acquiring fluorescence lifetime images of rapidly flowing cells. Moreover, our FLIM system distinguishes subpopulations in male rat glioma and captures dynamic changes in the cell nucleus induced by an anti-cancer drug. FLIM flow cytometry significantly enhances cellular analysis capabilities, providing detailed insights into cellular functions, interactions, and environments.
Collapse
Grants
- R35 GM152076 NIGMS NIH HHS
- This work was supported by JSPS Core-to-Core Program (K. G.), JSPS KAKENHI Grant Numbers 19H05633 and 20H00317 (K. G.), Ogasawara Foundation (K. G.), Nakatani Foundation (K. G.), Konica Minolta Foundation (K. G.), Philipp Franz von Siebold Award (K. G.), Humboldt Association of Japan (K. G.), Precise Measurement Technology Promotion Foundation (H. M.), JST PRESTO (JPMJPR1878) (K. H.), JST FOREST (21470594) (K. H.), JSPS Gran-in-Aid for Scientific Research (B) (22538379) (K. H.), JSPS Grant-in-Aid for Young Scientists (20K15227) (K. H.), Research Foundation for Opto-Science and Technology (K. H.), JSPS KAKENHI Grant Numbers 21J10600 and 24K18149 (H. K.), Konica Minolta Light Future Incentive Award (H. K.). We thank Mayu Sehara for her help with the cell sample preparation. The manuscript underwent editing with the assistance of a large language model (LLM).
Collapse
Affiliation(s)
- Hiroshi Kanno
- Department of Chemistry, The University of Tokyo, Tokyo, Japan.
- Department of Neurosurgical Engineering and Translational Neuroscience, Tohoku University Graduate School of Medicine, Miyagi, Japan.
| | - Kotaro Hiramatsu
- Department of Chemistry, The University of Tokyo, Tokyo, Japan
- Department of Chemistry, Kyushu University, Fukuoka, Japan
| | - Hideharu Mikami
- Department of Chemistry, The University of Tokyo, Tokyo, Japan
- Research Institute for Electronic Science, Hokkaido University, Hokkaido, Japan
| | - Atsushi Nakayashiki
- Department of Neurosurgery, Tohoku University Graduate School of Medicine, Miyagi, Japan
| | - Shota Yamashita
- Department of Neurosurgery, Tohoku University Graduate School of Medicine, Miyagi, Japan
| | - Arata Nagai
- Department of Neurosurgery, Tohoku University Graduate School of Medicine, Miyagi, Japan
| | - Kohki Okabe
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| | - Fan Li
- Department of Chemistry, The University of Tokyo, Tokyo, Japan
| | - Fei Yin
- Department of Neurosurgical Engineering and Translational Neuroscience, Tohoku University Graduate School of Medicine, Miyagi, Japan
| | - Keita Tominaga
- Department of Neurosurgery, Tohoku University Graduate School of Medicine, Miyagi, Japan
| | | | - Ryohei Noma
- SANKEN (The Institute of Scientific and Industrial Research), Osaka University, Osaka, Japan
| | - Bahareh Kiani
- Miltenyi Biotec B.V. & Co. KG, Bergisch Gladbach, Germany
| | - Olga Efa
- Miltenyi Biotec B.V. & Co. KG, Bergisch Gladbach, Germany
| | - Martin Büscher
- Miltenyi Biotec B.V. & Co. KG, Bergisch Gladbach, Germany
| | - Tetsuichi Wazawa
- SANKEN (The Institute of Scientific and Industrial Research), Osaka University, Osaka, Japan
| | | | - Hirofumi Shintaku
- Institute for Life and Medical Sciences, Kyoto University, Kyoto, Japan
| | - Takeharu Nagai
- SANKEN (The Institute of Scientific and Industrial Research), Osaka University, Osaka, Japan
| | - Sigurd Braun
- Institute for Genetics, Justus-Liebig-University Giessen, Giessen, Germany
| | - Jessica P Houston
- Department of Chemical and Materials Engineering, New Mexico State University, Las Cruces, NM, USA
| | - Sherif Rashad
- Department of Neurosurgical Engineering and Translational Neuroscience, Tohoku University Graduate School of Medicine, Miyagi, Japan
- Department of Neurosurgical Engineering and Translational Neuroscience Graduate School of Biomedical Engineering, Tohoku University, Miyagi, Japan
| | - Kuniyasu Niizuma
- Department of Neurosurgical Engineering and Translational Neuroscience, Tohoku University Graduate School of Medicine, Miyagi, Japan
- Department of Neurosurgery, Tohoku University Graduate School of Medicine, Miyagi, Japan
- Department of Neurosurgical Engineering and Translational Neuroscience Graduate School of Biomedical Engineering, Tohoku University, Miyagi, Japan
| | - Keisuke Goda
- Department of Chemistry, The University of Tokyo, Tokyo, Japan.
- Institute of Technological Sciences, Wuhan University, Hubei, China.
- Department of Bioengineering, University of California, Los Angeles, CA, USA.
| |
Collapse
|
27
|
Saito M, Arai F, Yamanishi Y, Sakuma S. Spatiotemporally controlled microvortices provide advanced microfluidic components. Proc Natl Acad Sci U S A 2024; 121:e2306182121. [PMID: 39102543 PMCID: PMC11331141 DOI: 10.1073/pnas.2306182121] [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: 04/17/2023] [Accepted: 06/27/2024] [Indexed: 08/07/2024] Open
Abstract
Microvortices are emerging components that impart functionality to microchannels by exploiting inertia effects such as high shear stress, effective fluid diffusion, and large pressure loss. Exploring the dynamic generation of vortices further expands the scope of microfluidic applications, including cell stimulation, fluid mixing, and transport. Despite the crucial role of vortices' development within sub-millisecond timescales, previous studies in microfluidics did not explore the modulation of the Reynolds number (Re) in the range of several hundred. In this study, we modulated high-speed flows (54 < [Formula: see text] < 456) within sub-millisecond timescales using a piezo-driven on-chip membrane pump. By applying this method to microchannels with asymmetric geometries, we successfully controlled the spatiotemporal development of vortices, adjusting their behavior in response to oscillatory flow directions. These different vortices induced different pressure losses, imparting the microchannels with direction-dependent flow resistance, mimicking a diode-like behavior. Through precise control of vortex development, we managed to regulate this direction-dependent resistance, enabling the rectification of oscillatory flow resembling a diode and the ability to switch its rectification direction. This component facilitated bidirectional flow control without the need for mechanical valves. Moreover, we demonstrated its application in microfluidic cell pipetting, enabling the isolation of single cells. Consequently, based on modulating high-speed flow, our approach offers precise control over the spatiotemporal development of vortices in microstructures, thereby introducing innovative microfluidic functionalities.
Collapse
Affiliation(s)
- Makoto Saito
- Department of Mechanical Engineering, Faculty of Engineering, Kyushu University, Fukuoka819-0395, Japan
| | - Fumihito Arai
- Department of Mechanical Engineering, Graduate School of Engineering, The University of Tokyo, Bunkyo-ku113-8656, Japan
| | - Yoko Yamanishi
- Department of Mechanical Engineering, Faculty of Engineering, Kyushu University, Fukuoka819-0395, Japan
| | - Shinya Sakuma
- Department of Mechanical Engineering, Faculty of Engineering, Kyushu University, Fukuoka819-0395, Japan
| |
Collapse
|
28
|
Lo MCK, Siu DMD, Lee KCM, Wong JSJ, Yeung MCF, Hsin MKY, Ho JCM, Tsia KK. Information-Distilled Generative Label-Free Morphological Profiling Encodes Cellular Heterogeneity. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2307591. [PMID: 38864546 PMCID: PMC11304271 DOI: 10.1002/advs.202307591] [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: 10/11/2023] [Revised: 05/17/2024] [Indexed: 06/13/2024]
Abstract
Image-based cytometry faces challenges due to technical variations arising from different experimental batches and conditions, such as differences in instrument configurations or image acquisition protocols, impeding genuine biological interpretation of cell morphology. Existing solutions, often necessitating extensive pre-existing data knowledge or control samples across batches, have proved limited, especially with complex cell image data. To overcome this, "Cyto-Morphology Adversarial Distillation" (CytoMAD), a self-supervised multi-task learning strategy that distills biologically relevant cellular morphological information from batch variations, is introduced to enable integrated analysis across multiple data batches without complex data assumptions or extensive manual annotation. Unique to CytoMAD is its "morphology distillation", symbiotically paired with deep-learning image-contrast translation-offering additional interpretable insights into label-free cell morphology. The versatile efficacy of CytoMAD is demonstrated in augmenting the power of biophysical imaging cytometry. It allows integrated label-free classification of human lung cancer cell types and accurately recapitulates their progressive drug responses, even when trained without the drug concentration information. CytoMAD also allows joint analysis of tumor biophysical cellular heterogeneity, linked to epithelial-mesenchymal plasticity, that standard fluorescence markers overlook. CytoMAD can substantiate the wide adoption of biophysical cytometry for cost-effective diagnosis and screening.
Collapse
Affiliation(s)
- Michelle C. K. Lo
- Department of Electrical and Electronic EngineeringThe University of Hong KongHong Kong000000Hong Kong
- Advanced Biomedical Instrumentation CentreHong Kong Science Park, New TerritoriesHong Kong000000Hong Kong
| | - Dickson M. D. Siu
- Department of Electrical and Electronic EngineeringThe University of Hong KongHong Kong000000Hong Kong
- Advanced Biomedical Instrumentation CentreHong Kong Science Park, New TerritoriesHong Kong000000Hong Kong
| | - Kelvin C. M. Lee
- Department of Electrical and Electronic EngineeringThe University of Hong KongHong Kong000000Hong Kong
- Advanced Biomedical Instrumentation CentreHong Kong Science Park, New TerritoriesHong Kong000000Hong Kong
| | - Justin S. J. Wong
- Conzeb LimitedHong Kong Science Park, New TerritoriesHong Kong000000Hong Kong
| | - Maximus C. F. Yeung
- Department of Pathology, Li Ka Shing Faculty of MedicineThe University of Hong KongPokfulam RoadHong Kong000000Hong Kong
| | - Michael K. Y. Hsin
- Department of Surgery, Li Ka Shing Faculty of MedicineThe University of Hong KongPokfulam RoadHong Kong000000Hong Kong
| | - James C. M. Ho
- Department of Medicine, Li Ka Shing Faculty of MedicineThe University of Hong KongPokfulam RoadHong Kong000000Hong Kong
| | - Kevin K. Tsia
- Department of Electrical and Electronic EngineeringThe University of Hong KongHong Kong000000Hong Kong
- Advanced Biomedical Instrumentation CentreHong Kong Science Park, New TerritoriesHong Kong000000Hong Kong
| |
Collapse
|
29
|
Sonmez ME, Gumus NE, Eczacioglu N, Develi EE, Yücel K, Yildiz HB. Enhancing microalgae classification accuracy in marine ecosystems through convolutional neural networks and support vector machines. MARINE POLLUTION BULLETIN 2024; 205:116616. [PMID: 38936001 DOI: 10.1016/j.marpolbul.2024.116616] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Revised: 06/13/2024] [Accepted: 06/16/2024] [Indexed: 06/29/2024]
Abstract
Accurately classifying microalgae species is vital for monitoring marine ecosystems and managing the emergence of marine mucilage, which is crucial for monitoring mucilage phenomena in marine environments. Traditional methods have been inadequate due to time-consuming processes and the need for expert knowledge. The purpose of this article is to employ convolutional neural networks (CNNs) and support vector machines (SVMs) to improve classification accuracy and efficiency. By employing advanced computational techniques, including MobileNet and GoogleNet models, alongside SVM classification, the study demonstrates significant advancements over conventional identification methods. In the classification of a dataset consisting of 7820 images using four different SVM kernel functions, the linear kernel achieved the highest success rate at 98.79 %. It is followed by the RBF kernel at 98.73 %, the polynomial kernel at 97.84 %, and the sigmoid kernel at 97.20 %. This research not only provides a methodological framework for future studies in marine biodiversity monitoring but also highlights the potential for real-time applications in ecological conservation and understanding mucilage dynamics amidst climate change and environmental pollution.
Collapse
Affiliation(s)
- Mesut Ersin Sonmez
- Department of Bioengineering, Faculty of Engineering, Karamanoglu Mehmetbey University, Karaman, Türkiye
| | - Numan Emre Gumus
- Department of Environmental Protection Technology, Kazım Karabekir Vocational School, Karamanoglu Mehmetbey University, Karaman, Türkiye.
| | - Numan Eczacioglu
- Department of Bioengineering, Faculty of Engineering, Karamanoglu Mehmetbey University, Karaman, Türkiye
| | | | - Kamile Yücel
- Department of Medical Biochemistry, Faculty of Medicine, KTO, Karatay University, Konya, Türkiye
| | | |
Collapse
|
30
|
Kuhn TM, Paulsen M, Cuylen-Haering S. Accessible high-speed image-activated cell sorting. Trends Cell Biol 2024; 34:657-670. [PMID: 38789300 DOI: 10.1016/j.tcb.2024.04.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Revised: 04/15/2024] [Accepted: 04/23/2024] [Indexed: 05/26/2024]
Abstract
Over the past six decades, fluorescence-activated cell sorting (FACS) has become an essential technology for basic and clinical research by enabling the isolation of cells of interest in high throughput. Recent technological advancements have started a new era of flow cytometry. By combining the spatial resolution of microscopy with high-speed cell sorting, new instruments allow cell sorting based on simple image-derived parameters or sophisticated image analysis algorithms, thereby greatly expanding the scope of applications. In this review, we discuss the systems that are commercially available or have been described in enough methodological and engineering detail to allow their replication. We summarize their strengths and limitations and highlight applications that have the potential to transform various fields in basic life science research and clinical settings.
Collapse
Affiliation(s)
- Terra M Kuhn
- Cell Biology and Biophysics Unit, European Molecular Biology Laboratory (EMBL), Heidelberg, Germany
| | - Malte Paulsen
- Novo Nordisk Foundation Center for Stem Cell Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.
| | - Sara Cuylen-Haering
- Cell Biology and Biophysics Unit, European Molecular Biology Laboratory (EMBL), Heidelberg, Germany.
| |
Collapse
|
31
|
Zhu S, Zhu Z, Ni C, Zhou Z, Chen Y, Tang D, Guo K, Yang S, Liu K, Ni Z, Xiang N. Liquid Biopsy Instrument for Ultra-Fast and Label-Free Detection of Circulating Tumor Cells. RESEARCH (WASHINGTON, D.C.) 2024; 7:0431. [PMID: 39050821 PMCID: PMC11266806 DOI: 10.34133/research.0431] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Accepted: 06/27/2024] [Indexed: 07/27/2024]
Abstract
Rapid diagnosis and real-time monitoring are of great important in the fight against cancer. However, most available diagnostic technologies are time-consuming and labor-intensive and are commonly invasive. Here, we describe CytoExam, an automatic liquid biopsy instrument designed based on inertial microfluidics and impedance cytometry, which uses a deep learning algorithm for the analysis of circulating tumor cells (CTCs). In silico and in vitro experiments demonstrated that CytoExam could achieve label-free detection of CTCs in the peripheral blood of cancer patients within 15 min. The clinical applicability of CytoExam was also verified using peripheral blood samples from 10 healthy donors and >50 patients with breast, colorectal, or lung cancer. Significant differences in the number of collected cells and predicted CTCs were observed between the 2 groups, with variations in the dielectric properties of the collected cells from cancer patients also being observed. The ultra-fast and minimally invasive features of CytoExam may pave the way for new paths for cancer diagnosis and scientific research.
Collapse
Affiliation(s)
- Shu Zhu
- School of Mechanical Engineering, and Jiangsu Key Laboratory for Design and Manufacture of Micro-Nano Biomedical Instruments,
Southeast University, Nanjing 211189, China
- School of Electrical and Automation Engineering, and Jiangsu Key Laboratory of 3D Printing Equipment and Manufacturing,
Nanjing Normal University, Nanjing 210023, China
| | - Zhixian Zhu
- School of Mechanical Engineering, and Jiangsu Key Laboratory for Design and Manufacture of Micro-Nano Biomedical Instruments,
Southeast University, Nanjing 211189, China
| | - Chen Ni
- School of Mechanical Engineering, and Jiangsu Key Laboratory for Design and Manufacture of Micro-Nano Biomedical Instruments,
Southeast University, Nanjing 211189, China
| | - Zheng Zhou
- School of Mechanical Engineering, and Jiangsu Key Laboratory for Design and Manufacture of Micro-Nano Biomedical Instruments,
Southeast University, Nanjing 211189, China
| | - Yao Chen
- School of Mechanical Engineering, and Jiangsu Key Laboratory for Design and Manufacture of Micro-Nano Biomedical Instruments,
Southeast University, Nanjing 211189, China
| | - Dezhi Tang
- School of Mechanical Engineering, and Jiangsu Key Laboratory for Design and Manufacture of Micro-Nano Biomedical Instruments,
Southeast University, Nanjing 211189, China
| | - Kefan Guo
- School of Mechanical Engineering, and Jiangsu Key Laboratory for Design and Manufacture of Micro-Nano Biomedical Instruments,
Southeast University, Nanjing 211189, China
| | - Shuai Yang
- School of Mechanical Engineering, and Jiangsu Key Laboratory for Design and Manufacture of Micro-Nano Biomedical Instruments,
Southeast University, Nanjing 211189, China
| | - Kang Liu
- School of Mechanical Engineering, and Jiangsu Key Laboratory for Design and Manufacture of Micro-Nano Biomedical Instruments,
Southeast University, Nanjing 211189, China
| | - Zhonghua Ni
- School of Mechanical Engineering, and Jiangsu Key Laboratory for Design and Manufacture of Micro-Nano Biomedical Instruments,
Southeast University, Nanjing 211189, China
| | - Nan Xiang
- School of Mechanical Engineering, and Jiangsu Key Laboratory for Design and Manufacture of Micro-Nano Biomedical Instruments,
Southeast University, Nanjing 211189, China
| |
Collapse
|
32
|
Mika T, Kalnins M, Spalvins K. The use of droplet-based microfluidic technologies for accelerated selection of Yarrowia lipolytica and Phaffia rhodozyma yeast mutants. Biol Methods Protoc 2024; 9:bpae049. [PMID: 39114747 PMCID: PMC11303513 DOI: 10.1093/biomethods/bpae049] [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: 05/11/2024] [Revised: 06/24/2024] [Accepted: 07/09/2024] [Indexed: 08/10/2024] Open
Abstract
Microorganisms are widely used for the industrial production of various valuable products, such as pharmaceuticals, food and beverages, biofuels, enzymes, amino acids, vaccines, etc. Research is constantly carried out to improve their properties, mainly to increase their productivity and efficiency and reduce the cost of the processes. The selection of microorganisms with improved qualities takes a lot of time and resources (both human and material); therefore, this process itself needs optimization. In the last two decades, microfluidics technology appeared in bioengineering, which allows for manipulating small particles (from tens of microns to nanometre scale) in the flow of liquid in microchannels. The technology is based on small-volume objects (microdroplets from nano to femtolitres), which are manipulated using a microchip. The chip is made of an optically transparent inert to liquid medium material and contains a series of channels of small size (<1 mm) of certain geometry. Based on the physical and chemical properties of microparticles (like size, weight, optical density, dielectric constant, etc.), they are separated using microsensors. The idea of accelerated selection of microorganisms is the application of microfluidic technologies to separate mutants with improved qualities after mutagenesis. This article discusses the possible application and practical implementation of microfluidic separation of mutants, including yeasts like Yarrowia lipolytica and Phaffia rhodozyma after chemical mutagenesis will be discussed.
Collapse
Affiliation(s)
- Taras Mika
- Institute of Energy Systems and Environment, Riga Technical University, 12 – K1 Āzene street, Riga, LV-1048, Latvia
| | - Martins Kalnins
- Institute of Energy Systems and Environment, Riga Technical University, 12 – K1 Āzene street, Riga, LV-1048, Latvia
| | - Kriss Spalvins
- Institute of Energy Systems and Environment, Riga Technical University, 12 – K1 Āzene street, Riga, LV-1048, Latvia
| |
Collapse
|
33
|
Wei Y, Forelli RF, Hansen C, Levesque JP, Tran N, Agar JC, Di Guglielmo G, Mauel ME, Navratil GA. Low latency optical-based mode tracking with machine learning deployed on FPGAs on a tokamak. THE REVIEW OF SCIENTIFIC INSTRUMENTS 2024; 95:073509. [PMID: 38980128 DOI: 10.1063/5.0190354] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Accepted: 06/18/2024] [Indexed: 07/10/2024]
Abstract
Active feedback control in magnetic confinement fusion devices is desirable to mitigate plasma instabilities and enable robust operation. Optical high-speed cameras provide a powerful, non-invasive diagnostic and can be suitable for these applications. In this study, we process high-speed camera data, at rates exceeding 100 kfps, on in situ field-programmable gate array (FPGA) hardware to track magnetohydrodynamic (MHD) mode evolution and generate control signals in real time. Our system utilizes a convolutional neural network (CNN) model, which predicts the n = 1 MHD mode amplitude and phase using camera images with better accuracy than other tested non-deep-learning-based methods. By implementing this model directly within the standard FPGA readout hardware of the high-speed camera diagnostic, our mode tracking system achieves a total trigger-to-output latency of 17.6 μs and a throughput of up to 120 kfps. This study at the High Beta Tokamak-Extended Pulse (HBT-EP) experiment demonstrates an FPGA-based high-speed camera data acquisition and processing system, enabling application in real-time machine-learning-based tokamak diagnostic and control as well as potential applications in other scientific domains.
Collapse
Affiliation(s)
- Y Wei
- Department of Applied Physics and Applied Mathematics, Columbia University, New York, New York 10027, USA
| | - R F Forelli
- Real-time Processing Systems Division, Fermi National Accelerator Laboratory, Batavia, Illinois 60510, USA
- Department of Electrical and Computer Engineering, Lehigh University, Bethlehem, Pennsylvania 18015, USA
| | - C Hansen
- Department of Applied Physics and Applied Mathematics, Columbia University, New York, New York 10027, USA
| | - J P Levesque
- Department of Applied Physics and Applied Mathematics, Columbia University, New York, New York 10027, USA
| | - N Tran
- Real-time Processing Systems Division, Fermi National Accelerator Laboratory, Batavia, Illinois 60510, USA
- Department of Electrical and Computer Engineering, Northwestern University, Evanston, Illinois 60208, USA
| | - J C Agar
- Department of Mechanical Engineering and Mechanics, Drexel University, Philadelphia, Pennsylvania 19104, USA
| | - G Di Guglielmo
- Department of Electrical and Computer Engineering, Northwestern University, Evanston, Illinois 60208, USA
- Microelectronics Division, Fermi National Accelerator Laboratory, Batavia, Illinois 60510, USA
| | - M E Mauel
- Department of Applied Physics and Applied Mathematics, Columbia University, New York, New York 10027, USA
| | - G A Navratil
- Department of Applied Physics and Applied Mathematics, Columbia University, New York, New York 10027, USA
| |
Collapse
|
34
|
Greatbatch CJ, Lu Q, Hung S, Tran SN, Wing K, Liang H, Han X, Zhou T, Siggs OM, Mackey DA, Liu GS, Cook AL, Powell JE, Craig JE, MacGregor S, Hewitt AW. Deep Learning-Based Identification of Intraocular Pressure-Associated Genes Influencing Trabecular Meshwork Cell Morphology. OPHTHALMOLOGY SCIENCE 2024; 4:100504. [PMID: 38682030 PMCID: PMC11046128 DOI: 10.1016/j.xops.2024.100504] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Revised: 02/27/2024] [Accepted: 02/27/2024] [Indexed: 05/01/2024]
Abstract
Purpose Genome-wide association studies have recently uncovered many loci associated with variation in intraocular pressure (IOP). Artificial intelligence (AI) can be used to interrogate the effect of specific genetic knockouts on the morphology of trabecular meshwork cells (TMCs) and thus, IOP regulation. Design Experimental study. Subjects Primary TMCs collected from human donors. Methods Sixty-two genes at 55 loci associated with IOP variation were knocked out in primary TMC lines. All cells underwent high-throughput microscopy imaging after being stained with a 5-channel fluorescent cell staining protocol. A convolutional neural network was trained to distinguish between gene knockout and normal control cell images. The area under the receiver operator curve (AUC) metric was used to quantify morphological variation in gene knockouts to identify potential pathological perturbations. Main Outcome Measures Degree of morphological variation as measured by deep learning algorithm accuracy of differentiation from normal controls. Results Cells where LTBP2 or BCAS3 had been perturbed demonstrated the greatest morphological variation from normal TMCs (AUC 0.851, standard deviation [SD] 0.030; and AUC 0.845, SD 0.020, respectively). Of 7 multigene loci, 5 had statistically significant differences in AUC (P < 0.05) between genes, allowing for pathological gene prioritization. The mitochondrial channel most frequently showed the greatest degree of morphological variation (33.9% of cell lines). Conclusions We demonstrate a robust method for functionally interrogating genome-wide association signals using high-throughput microscopy and AI. Genetic variations inducing marked morphological variation can be readily identified, allowing for the gene-based dissection of loci associated with complex traits. Financial Disclosures Proprietary or commercial disclosure may be found in the Footnotes and Disclosures at the end of this article.
Collapse
Affiliation(s)
- Connor J. Greatbatch
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Tasmania, Australia
| | - Qinyi Lu
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Tasmania, Australia
| | - Sandy Hung
- Centre for Eye Research Australia, University of Melbourne, Melbourne, Victoria, Australia
| | - Son N. Tran
- Department of Information and Communication Technology, University of Tasmania, Hobart, Tasmania, Australia
| | - Kristof Wing
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Tasmania, Australia
| | - Helena Liang
- Centre for Eye Research Australia, University of Melbourne, Melbourne, Victoria, Australia
| | - Xikun Han
- Statistical Genetics Laboratory, QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | - Tiger Zhou
- Department of Ophthalmology, Flinders Medical Centre, Flinders University, Bedford Park, Australia
| | - Owen M. Siggs
- Cellular Genomics Group, Garvan Institute of Medical Research, Sydney, New South Wales, Australia
- Faculty of Medicine and Health, School of Clinical Medicine, UNSW, Sydney, New South Wales, Australia
| | - David A. Mackey
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Tasmania, Australia
- Lions Eye Institute, Centre for Ophthalmology and Visual Science, University of Western Australia, Perth, Western Australia, Australia
| | - Guei-Sheung Liu
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Tasmania, Australia
- Centre for Eye Research Australia, University of Melbourne, Melbourne, Victoria, Australia
| | - Anthony L. Cook
- Wicking Dementia Research and Education Centre, University of Tasmania, Hobart, Tasmania, Australia
| | - Joseph E. Powell
- Garvan-Weizmann Centre for Cellular Genomics, Garvan Institute of Medical Research, Sydney, New South Wales, Australia
- UNSW Cellular Genomics Futures Institute, UNSW, Sydney, New South Wales, Australia
| | - Jamie E. Craig
- Department of Ophthalmology, Flinders Medical Centre, Flinders University, Bedford Park, Australia
| | - Stuart MacGregor
- Statistical Genetics Laboratory, QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | - Alex W. Hewitt
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Tasmania, Australia
- Centre for Eye Research Australia, University of Melbourne, Melbourne, Victoria, Australia
| |
Collapse
|
35
|
Zhu J, Pan S, Chai H, Zhao P, Feng Y, Cheng Z, Zhang S, Wang W. Microfluidic Impedance Cytometry Enabled One-Step Sample Preparation for Efficient Single-Cell Mass Spectrometry. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2310700. [PMID: 38483007 DOI: 10.1002/smll.202310700] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Revised: 03/05/2024] [Indexed: 06/27/2024]
Abstract
Single-cell mass spectrometry (MS) is significant in biochemical analysis and holds great potential in biomedical applications. Efficient sample preparation like sorting (i.e., separating target cells from the mixed population) and desalting (i.e., moving the cells off non-volatile salt solution) is urgently required in single-cell MS. However, traditional sample preparation methods suffer from complicated operation with various apparatus, or insufficient performance. Herein, a one-step sample preparation strategy by leveraging label-free impedance flow cytometry (IFC) based microfluidics is proposed. Specifically, the IFC framework to characterize and sort single-cells is adopted. Simultaneously with sorting, the target cell is transferred from the local high-salinity buffer to the MS-compatible solution. In this way, one-step sorting and desalting are achieved and the collected cells can be directly fed for MS analysis. A high sorting efficiency (>99%), cancer cell purity (≈87%), and desalting efficiency (>99%), and the whole workflow of impedance-based separation and MS analysis of normal cells (MCF-10A) and cancer cells (MDA-MB-468) are verified. As a standalone sample preparation module, the microfluidic chip is compatible with a variety of MS analysis methods, and envisioned to provide a new paradigm in efficient MS sample preparation, and further in multi-modal (i.e., electrical and metabolic) characterization of single-cells.
Collapse
Affiliation(s)
- Junwen Zhu
- State Key Laboratory of Precision Measurement Technology and Instrument, Department of Precision Instrument, Tsinghua University, Beijing, 100084, China
| | - Siyuan Pan
- Department of Chemistry, Tsinghua University, Beijing, 100084, China
| | - Huichao Chai
- State Key Laboratory of Precision Measurement Technology and Instrument, Department of Precision Instrument, Tsinghua University, Beijing, 100084, China
| | - Peng Zhao
- State Key Laboratory of Precision Measurement Technology and Instrument, Department of Precision Instrument, Tsinghua University, Beijing, 100084, China
| | - Yongxiang Feng
- State Key Laboratory of Precision Measurement Technology and Instrument, Department of Precision Instrument, Tsinghua University, Beijing, 100084, China
| | - Zhen Cheng
- Department of Automation, Tsinghua University, Beijing, 100084, China
| | - Sichun Zhang
- Department of Chemistry, Tsinghua University, Beijing, 100084, China
| | - Wenhui Wang
- State Key Laboratory of Precision Measurement Technology and Instrument, Department of Precision Instrument, Tsinghua University, Beijing, 100084, China
| |
Collapse
|
36
|
Mandracchia B, Zheng C, Rajendran S, Liu W, Forghani P, Xu C, Jia S. High-speed optical imaging with sCMOS pixel reassignment. Nat Commun 2024; 15:4598. [PMID: 38816394 PMCID: PMC11139943 DOI: 10.1038/s41467-024-48987-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Accepted: 05/13/2024] [Indexed: 06/01/2024] Open
Abstract
Fluorescence microscopy has undergone rapid advancements, offering unprecedented visualization of biological events and shedding light on the intricate mechanisms governing living organisms. However, the exploration of rapid biological dynamics still poses a significant challenge due to the limitations of current digital camera architectures and the inherent compromise between imaging speed and other capabilities. Here, we introduce sHAPR, a high-speed acquisition technique that leverages the operating principles of sCMOS cameras to capture fast cellular and subcellular processes. sHAPR harnesses custom fiber optics to convert microscopy images into one-dimensional recordings, enabling acquisition at the maximum camera readout rate, typically between 25 and 250 kHz. We have demonstrated the utility of sHAPR with a variety of phantom and dynamic systems, including high-throughput flow cytometry, cardiomyocyte contraction, and neuronal calcium waves, using a standard epi-fluorescence microscope. sHAPR is highly adaptable and can be integrated into existing microscopy systems without requiring extensive platform modifications. This method pushes the boundaries of current fluorescence imaging capabilities, opening up new avenues for investigating high-speed biological phenomena.
Collapse
Affiliation(s)
- Biagio Mandracchia
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
- E.T.S.I. Telecomunicación, Universidad de Valladolid, Valladolid, Spain
| | - Corey Zheng
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Suraj Rajendran
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Wenhao Liu
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Parvin Forghani
- Department of Pediatrics, School of Medicine, Emory University, Atlanta, GA, USA
| | - Chunhui Xu
- Department of Pediatrics, School of Medicine, Emory University, Atlanta, GA, USA
- Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, USA
| | - Shu Jia
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA.
- Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, USA.
| |
Collapse
|
37
|
Ghosh R, Arnheim A, van Zee M, Shang L, Soemardy C, Tang RC, Mellody M, Baghdasarian S, Sanchez Ochoa E, Ye S, Chen S, Williamson C, Karunaratne A, Di Carlo D. Lab on a Particle Technologies. Anal Chem 2024; 96:7817-7839. [PMID: 38650433 PMCID: PMC11112544 DOI: 10.1021/acs.analchem.4c01510] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Revised: 04/14/2024] [Accepted: 04/16/2024] [Indexed: 04/25/2024]
Affiliation(s)
- Rajesh Ghosh
- Department
of Bioengineering, University of California,
Los Angeles, Los Angeles, California 90095, United States
| | - Alyssa Arnheim
- Department
of Bioengineering, University of California,
Los Angeles, Los Angeles, California 90095, United States
| | - Mark van Zee
- Department
of Bioengineering, University of California,
Los Angeles, Los Angeles, California 90095, United States
| | - Lily Shang
- Department
of Bioengineering, University of California,
Los Angeles, Los Angeles, California 90095, United States
| | - Citradewi Soemardy
- Department
of Bioengineering, University of California,
Los Angeles, Los Angeles, California 90095, United States
| | - Rui-Chian Tang
- Department
of Bioengineering, University of California,
Los Angeles, Los Angeles, California 90095, United States
| | - Michael Mellody
- Department
of Bioengineering, University of California,
Los Angeles, Los Angeles, California 90095, United States
| | - Sevana Baghdasarian
- Department
of Bioengineering, University of California,
Los Angeles, Los Angeles, California 90095, United States
| | - Edwin Sanchez Ochoa
- Department
of Bioengineering, University of California,
Los Angeles, Los Angeles, California 90095, United States
| | - Shun Ye
- Department
of Bioengineering, University of California,
Los Angeles, Los Angeles, California 90095, United States
| | - Siyu Chen
- Department
of Bioengineering, University of California,
Los Angeles, Los Angeles, California 90095, United States
| | - Cayden Williamson
- Department
of Bioengineering, University of California,
Los Angeles, Los Angeles, California 90095, United States
| | - Amrith Karunaratne
- Department
of Bioengineering, University of California,
Los Angeles, Los Angeles, California 90095, United States
| | - Dino Di Carlo
- Department
of Bioengineering, University of California,
Los Angeles, Los Angeles, California 90095, United States
- Jonsson
Comprehensive Cancer Center, University
of California, Los Angeles, Los Angeles, California 90095, United States
- Department
of Mechanical and Aerospace Engineering, University of California, Los Angeles, Los Angeles, California 90095, United States
- California
NanoSystems Institute, Los Angeles, California 90095, United States
| |
Collapse
|
38
|
Ferreira EKGD, Silveira GF. Classification and counting of cells in brightfield microscopy images: an application of convolutional neural networks. Sci Rep 2024; 14:9031. [PMID: 38641688 PMCID: PMC11031575 DOI: 10.1038/s41598-024-59625-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Accepted: 04/12/2024] [Indexed: 04/21/2024] Open
Abstract
Microscopy is integral to medical research, facilitating the exploration of various biological questions, notably cell quantification. However, this process's time-consuming and error-prone nature, attributed to human intervention or automated methods usually applied to fluorescent images, presents challenges. In response, machine learning algorithms have been integrated into microscopy, automating tasks and constructing predictive models from vast datasets. These models adeptly learn representations for object detection, image segmentation, and target classification. An advantageous strategy involves utilizing unstained images, preserving cell integrity and enabling morphology-based classification-something hindered when fluorescent markers are used. The aim is to introduce a model proficient in classifying distinct cell lineages in digital contrast microscopy images. Additionally, the goal is to create a predictive model identifying lineage and determining optimal quantification of cell numbers. Employing a CNN machine learning algorithm, a classification model predicting cellular lineage achieved a remarkable accuracy of 93%, with ROC curve results nearing 1.0, showcasing robust performance. However, some lineages, namely SH-SY5Y (78%), HUH7_mayv (85%), and A549 (88%), exhibited slightly lower accuracies. These outcomes not only underscore the model's quality but also emphasize CNNs' potential in addressing the inherent complexities of microscopic images.
Collapse
Affiliation(s)
| | - G F Silveira
- Carlos Chagas Institute, Curitiba, PR, CEP 81310-020, Brazil.
| |
Collapse
|
39
|
Findinier J, Joubert LM, Schmid MF, Malkovskiy A, Chiu W, Burlacot A, Grossman AR. Dramatic Changes in Mitochondrial Subcellular Location and Morphology Accompany Activation of the CO 2 Concentrating Mechanism. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.25.586705. [PMID: 38585955 PMCID: PMC10996633 DOI: 10.1101/2024.03.25.586705] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/09/2024]
Abstract
Dynamic changes in intracellular ultrastructure can be critical for the ability of organisms to acclimate to environmental conditions. Microalgae, which are responsible for ~50% of global photosynthesis, compartmentalize their Rubisco into a specialized structure known as the pyrenoid when the cells experience limiting CO2 conditions; this compartmentalization appears to be a component of the CO2 Concentrating Mechanism (CCM), which facilitates photosynthetic CO2 fixation as environmental levels of inorganic carbon (Ci) decline. Changes in the spatial distribution of mitochondria in green algae have also been observed under CO2 limiting conditions, although a role for this reorganization in CCM function remains unclear. We used the green microalgae Chlamydomonas reinhardtii to monitor changes in the position and ultrastructure of mitochondrial membranes as cells transition between high CO2 (HC) and Low/Very Low CO2 (LC/VLC). Upon transferring cells to VLC, the mitochondria move from a central to a peripheral location, become wedged between the plasma membrane and chloroplast envelope, and mitochondrial membranes orient in parallel tubular arrays that extend from the cell's apex to its base. We show that these ultrastructural changes require protein and RNA synthesis, occur within 90 min of shifting cells to VLC conditions, correlate with CCM induction and are regulated by the CCM master regulator CIA5. The apico-basal orientation of the mitochondrial membrane, but not the movement of the mitochondrion to the cell periphery, is dependent on microtubules and the MIRO1 protein, which is involved in membrane-microtubule interactions. Furthermore, blocking mitochondrial electron transport in VLC acclimated cells reduces the cell's affinity for inorganic carbon. Overall, our results suggest that CIA5-dependent mitochondrial repositioning/reorientation functions in integrating cellular architecture and energetics with CCM activities and invite further exploration of how intracellular architecture can impact fitness under dynamic environmental conditions.
Collapse
Affiliation(s)
- Justin Findinier
- The Carnegie Institution for Science, Biosphere Sciences & Engineering, Stanford, CA 94305, USA
| | - Lydia-Marie Joubert
- SLAC National Accelerator Laboratory, Division of CryoEM and Bioimaging, Menlo Park, CA 94025, USA
| | - Michael F. Schmid
- SLAC National Accelerator Laboratory, Division of CryoEM and Bioimaging, Menlo Park, CA 94025, USA
| | - Andrey Malkovskiy
- The Carnegie Institution for Science, Biosphere Sciences & Engineering, Stanford, CA 94305, USA
| | - Wah Chiu
- SLAC National Accelerator Laboratory, Division of CryoEM and Bioimaging, Menlo Park, CA 94025, USA
- Stanford University, Department of Bioengineering, Stanford, CA 94305, USA
| | - Adrien Burlacot
- The Carnegie Institution for Science, Biosphere Sciences & Engineering, Stanford, CA 94305, USA
- Stanford University, Biology Department, Stanford, CA 94305, USA
| | - Arthur R. Grossman
- The Carnegie Institution for Science, Biosphere Sciences & Engineering, Stanford, CA 94305, USA
- Stanford University, Biology Department, Stanford, CA 94305, USA
| |
Collapse
|
40
|
Ichimura T, Kakizuka T, Sato Y, Fujioka Y, Ohba Y, Horikawa K, Nagai T. Strength in numbers: Unleashing the potential of trans-scale scope AMATERAS for massive cell quantification. Biophys Physicobiol 2024; 21:e211017. [PMID: 39175860 PMCID: PMC11338690 DOI: 10.2142/biophysico.bppb-v21.s017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Accepted: 03/22/2024] [Indexed: 08/24/2024] Open
Abstract
Singularity biology is a scientific field that targets drastic state changes in multicellular systems, aiming to discover the key cells that induce the state change and investigate the mechanisms behind them. To achieve this goal, we developed a trans-scale optical imaging system (trans-scale scope), that is capable of capturing both macroscale changes across the entire system and the micro-scale behavior of individual cells, surpassing the cell observation capabilities of traditional microscopes. We developed two units of the trans-scale scope, named AMATERAS-1 and -2, which demonstrated the ability to observe multicellular systems consisting of over one million cells in a single field of view with sub-cellular resolution. This flagship instrument has been used to observe the dynamics of various cell species, with the advantage of being able to observe a large number of cells, allowing the detection and analysis of rare events and cells such as leader cells in multicellular pattern formation and cells that spontaneously initiate calcium waves. In this paper, we present the design concept of AMATERAS, the optical configuration, and several examples of observations, and demonstrate how the strength-in-numbers works in life sciences.
Collapse
Affiliation(s)
- Taro Ichimura
- Transdimensional Life Imaging Division, Institute for Open and Transdisciplinary Research Initiatives, Osaka University, Suita, Osaka 565-0871, Japan
| | - Taishi Kakizuka
- Department of Biomolecular Science and Engineering, SANKEN, Osaka University, Ibaraki, Osaka 567-0047, Japan
| | - Yuki Sato
- Department of Anatomy and Cell Biology, Graduate School of Medical Sciences, Kyushu University, Higashi-ku, Fukuoka 812-8582, Japan
| | - Yoichiro Fujioka
- Department of Cell Physiology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Hokkaido 060-8638, Japan
| | - Yusuke Ohba
- Department of Cell Physiology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Hokkaido 060-8638, Japan
| | - Kazuki Horikawa
- Department of Optical Imaging, Advanced Research Promotion Center, Tokushima University, Tokushima 770-8503, Japan
| | - Takeharu Nagai
- Transdimensional Life Imaging Division, Institute for Open and Transdisciplinary Research Initiatives, Osaka University, Suita, Osaka 565-0871, Japan
- Department of Biomolecular Science and Engineering, SANKEN, Osaka University, Ibaraki, Osaka 567-0047, Japan
- Research Institute for Electronic Science, Hokkaido University, Sapporo, Hokkaido 001-0020, Japan
| |
Collapse
|
41
|
Dimitriadis S, Dova L, Kotsianidis I, Hatzimichael E, Kapsali E, Markopoulos GS. Imaging Flow Cytometry: Development, Present Applications, and Future Challenges. Methods Protoc 2024; 7:28. [PMID: 38668136 PMCID: PMC11054958 DOI: 10.3390/mps7020028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Revised: 03/13/2024] [Accepted: 03/21/2024] [Indexed: 04/29/2024] Open
Abstract
Imaging flow cytometry (ImFC) represents a significant technological advancement in the field of cytometry, effectively merging the high-throughput capabilities of flow analysis with the detailed imaging characteristics of microscopy. In our comprehensive review, we adopt a historical perspective to chart the development of ImFC, highlighting its origins and current state of the art and forecasting potential future advancements. The genesis of ImFC stemmed from merging the hydraulic system of a flow cytometer with advanced camera technology. This synergistic coupling facilitates the morphological analysis of cell populations at a high-throughput scale, effectively evolving the landscape of cytometry. Nevertheless, ImFC's implementation has encountered hurdles, particularly in developing software capable of managing its sophisticated data acquisition and analysis needs. The scale and complexity of the data generated by ImFC necessitate the creation of novel analytical tools that can effectively manage and interpret these data, thus allowing us to unlock the full potential of ImFC. Notably, artificial intelligence (AI) algorithms have begun to be applied to ImFC, offering promise for enhancing its analytical capabilities. The adaptability and learning capacity of AI may prove to be essential in knowledge mining from the high-dimensional data produced by ImFC, potentially enabling more accurate analyses. Looking forward, we project that ImFC may become an indispensable tool, not only in research laboratories, but also in clinical settings. Given the unique combination of high-throughput cytometry and detailed imaging offered by ImFC, we foresee a critical role for this technology in the next generation of scientific research and diagnostics. As such, we encourage both current and future scientists to consider the integration of ImFC as an addition to their research toolkit and clinical diagnostic routine.
Collapse
Affiliation(s)
- Savvas Dimitriadis
- Hematology Laboratory, Unit of Molecular Biology and Translational Flow Cytometry, University Hospital of Ioannina, 45100 Ioannina, Greece; (S.D.); (L.D.)
| | - Lefkothea Dova
- Hematology Laboratory, Unit of Molecular Biology and Translational Flow Cytometry, University Hospital of Ioannina, 45100 Ioannina, Greece; (S.D.); (L.D.)
| | - Ioannis Kotsianidis
- Department of Hematology, University Hospital of Alexandroupolis, Democritus University of Thrace, 69100 Alexandroupolis, Greece;
| | - Eleftheria Hatzimichael
- Department of Hematology, Faculty of Medicine, University of Ioannina, 45110 Ioannina, Greece; (E.H.); (E.K.)
| | - Eleni Kapsali
- Department of Hematology, Faculty of Medicine, University of Ioannina, 45110 Ioannina, Greece; (E.H.); (E.K.)
| | - Georgios S. Markopoulos
- Hematology Laboratory, Unit of Molecular Biology and Translational Flow Cytometry, University Hospital of Ioannina, 45100 Ioannina, Greece; (S.D.); (L.D.)
- Department of Surgery, Faculty of Medicine, University of Ioannina, 45110 Ioannina, Greece
| |
Collapse
|
42
|
Tabata K, Kawagoe H, Taylor JN, Mochizuki K, Kubo T, Clement JE, Kumamoto Y, Harada Y, Nakamura A, Fujita K, Komatsuzaki T. On-the-fly Raman microscopy guaranteeing the accuracy of discrimination. Proc Natl Acad Sci U S A 2024; 121:e2304866121. [PMID: 38483992 PMCID: PMC10962959 DOI: 10.1073/pnas.2304866121] [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: 04/03/2023] [Accepted: 12/15/2023] [Indexed: 03/19/2024] Open
Abstract
Accelerating the measurement for discrimination of samples, such as classification of cell phenotype, is crucial when faced with significant time and cost constraints. Spontaneous Raman microscopy offers label-free, rich chemical information but suffers from long acquisition time due to extremely small scattering cross-sections. One possible approach to accelerate the measurement is by measuring necessary parts with a suitable number of illumination points. However, how to design these points during measurement remains a challenge. To address this, we developed an imaging technique based on a reinforcement learning in machine learning (ML). This ML approach adaptively feeds back "optimal" illumination pattern during the measurement to detect the existence of specific characteristics of interest, allowing faster measurements while guaranteeing discrimination accuracy. Using a set of Raman images of human follicular thyroid and follicular thyroid carcinoma cells, we showed that our technique requires 3,333 to 31,683 times smaller number of illuminations for discriminating the phenotypes than raster scanning. To quantitatively evaluate the number of illuminations depending on the requisite discrimination accuracy, we prepared a set of polymer bead mixture samples to model anomalous and normal tissues. We then applied a home-built programmable-illumination microscope equipped with our algorithm, and confirmed that the system can discriminate the sample conditions with 104 to 4,350 times smaller number of illuminations compared to standard point illumination Raman microscopy. The proposed algorithm can be applied to other types of microscopy that can control measurement condition on the fly, offering an approach for the acceleration of accurate measurements in various applications including medical diagnosis.
Collapse
Affiliation(s)
- Koji Tabata
- Research Center of Mathematics for Social Creativity, Research Institute for Electronic Science, Hokkaido University, Sapporo001–0020, Hokkaido, Japan
- Institute for Chemical Reaction Design and Discovery, Hokkaido University, Sapporo001–0021, Hokkaido, Japan
| | - Hiroyuki Kawagoe
- Department of Applied Physics, Osaka University, Suita565–0871, Osaka, Japan
| | - J. Nicholas Taylor
- Research Center of Mathematics for Social Creativity, Research Institute for Electronic Science, Hokkaido University, Sapporo001–0020, Hokkaido, Japan
| | - Kentaro Mochizuki
- Department of Pathology and Cell Regulation, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto602–8566, Kyoto, Japan
| | - Toshiki Kubo
- Department of Applied Physics, Osaka University, Suita565–0871, Osaka, Japan
| | - Jean-Emmanuel Clement
- Institute for Chemical Reaction Design and Discovery, Hokkaido University, Sapporo001–0021, Hokkaido, Japan
| | - Yasuaki Kumamoto
- Department of Applied Physics, Osaka University, Suita565–0871, Osaka, Japan
- Institute for Open and Transdisciplinary Research Initiatives, Osaka University, Suita565–0871, Osaka, Japan
| | - Yoshinori Harada
- Department of Pathology and Cell Regulation, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto602–8566, Kyoto, Japan
| | - Atsuyoshi Nakamura
- Graduate School of Information Science and Technology, Hokkaido University, Sapporo060–0814, Hokkaido, Japan
| | - Katsumasa Fujita
- Department of Applied Physics, Osaka University, Suita565–0871, Osaka, Japan
- Institute for Open and Transdisciplinary Research Initiatives, Osaka University, Suita565–0871, Osaka, Japan
- Advanced Photonics and Biosensing Open Innovation Laboratory, AIST-Osaka University, Suita565–0871, Osaka, Japan
| | - Tamiki Komatsuzaki
- Research Center of Mathematics for Social Creativity, Research Institute for Electronic Science, Hokkaido University, Sapporo001–0020, Hokkaido, Japan
- Institute for Chemical Reaction Design and Discovery, Hokkaido University, Sapporo001–0021, Hokkaido, Japan
- Institute for Open and Transdisciplinary Research Initiatives, Osaka University, Suita565–0871, Osaka, Japan
- Graduate School of Chemical Sciences and Engineering Materials Chemistry, and Engineering Course, Hokkaido University, Sapporo060–0812, Hokkaido, Japan
- The Institute of Scientific and Industrial Research, Osaka University, Ibaraki567-0047, Osaka, Japan
| |
Collapse
|
43
|
Lugagne JB, Blassick CM, Dunlop MJ. Deep model predictive control of gene expression in thousands of single cells. Nat Commun 2024; 15:2148. [PMID: 38459057 PMCID: PMC10923782 DOI: 10.1038/s41467-024-46361-1] [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: 08/09/2023] [Accepted: 02/26/2024] [Indexed: 03/10/2024] Open
Abstract
Gene expression is inherently dynamic, due to complex regulation and stochastic biochemical events. However, the effects of these dynamics on cell phenotypes can be difficult to determine. Researchers have historically been limited to passive observations of natural dynamics, which can preclude studies of elusive and noisy cellular events where large amounts of data are required to reveal statistically significant effects. Here, using recent advances in the fields of machine learning and control theory, we train a deep neural network to accurately predict the response of an optogenetic system in Escherichia coli cells. We then use the network in a deep model predictive control framework to impose arbitrary and cell-specific gene expression dynamics on thousands of single cells in real time, applying the framework to generate complex time-varying patterns. We also showcase the framework's ability to link expression patterns to dynamic functional outcomes by controlling expression of the tetA antibiotic resistance gene. This study highlights how deep learning-enabled feedback control can be used to tailor distributions of gene expression dynamics with high accuracy and throughput without expert knowledge of the biological system.
Collapse
Affiliation(s)
- Jean-Baptiste Lugagne
- Department of Biomedical Engineering, Boston University, Boston, Massachusetts, 02215, USA.
- Biological Design Center, Boston University, Boston, Massachusetts, 02215, USA.
| | - Caroline M Blassick
- Department of Biomedical Engineering, Boston University, Boston, Massachusetts, 02215, USA
- Biological Design Center, Boston University, Boston, Massachusetts, 02215, USA
| | - Mary J Dunlop
- Department of Biomedical Engineering, Boston University, Boston, Massachusetts, 02215, USA.
- Biological Design Center, Boston University, Boston, Massachusetts, 02215, USA.
| |
Collapse
|
44
|
Hua X, Han K, Mandracchia B, Radmand A, Liu W, Kim H, Yuan Z, Ehrlich SM, Li K, Zheng C, Son J, Silva Trenkle AD, Kwong GA, Zhu C, Dahlman JE, Jia S. Light-field flow cytometry for high-resolution, volumetric and multiparametric 3D single-cell analysis. Nat Commun 2024; 15:1975. [PMID: 38438356 PMCID: PMC10912605 DOI: 10.1038/s41467-024-46250-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Accepted: 02/15/2024] [Indexed: 03/06/2024] Open
Abstract
Imaging flow cytometry (IFC) combines flow cytometry and fluorescence microscopy to enable high-throughput, multiparametric single-cell analysis with rich spatial details. However, current IFC techniques remain limited in their ability to reveal subcellular information with a high 3D resolution, throughput, sensitivity, and instrumental simplicity. In this study, we introduce a light-field flow cytometer (LFC), an IFC system capable of high-content, single-shot, and multi-color acquisition of up to 5,750 cells per second with a near-diffraction-limited resolution of 400-600 nm in all three dimensions. The LFC system integrates optical, microfluidic, and computational strategies to facilitate the volumetric visualization of various 3D subcellular characteristics through convenient access to commonly used epi-fluorescence platforms. We demonstrate the effectiveness of LFC in assaying, analyzing, and enumerating intricate subcellular morphology, function, and heterogeneity using various phantoms and biological specimens. The advancement offered by the LFC system presents a promising methodological pathway for broad cell biological and translational discoveries, with the potential for widespread adoption in biomedical research.
Collapse
Affiliation(s)
- Xuanwen Hua
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Keyi Han
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Biagio Mandracchia
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Afsane Radmand
- Parker H. Petit Institute for Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, GA, USA
- Department of Chemical Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| | - Wenhao Liu
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Hyejin Kim
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Zhou Yuan
- Parker H. Petit Institute for Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, GA, USA
- Georgia W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| | - Samuel M Ehrlich
- Parker H. Petit Institute for Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, GA, USA
- Georgia W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| | - Kaitao Li
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Corey Zheng
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Jeonghwan Son
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Aaron D Silva Trenkle
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Gabriel A Kwong
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
- Parker H. Petit Institute for Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, GA, USA
| | - Cheng Zhu
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
- Parker H. Petit Institute for Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, GA, USA
| | - James E Dahlman
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
- Parker H. Petit Institute for Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, GA, USA
| | - Shu Jia
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA.
- Parker H. Petit Institute for Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, GA, USA.
| |
Collapse
|
45
|
Zhang T, Di Carlo D, Lim CT, Zhou T, Tian G, Tang T, Shen AQ, Li W, Li M, Yang Y, Goda K, Yan R, Lei C, Hosokawa Y, Yalikun Y. Passive microfluidic devices for cell separation. Biotechnol Adv 2024; 71:108317. [PMID: 38220118 DOI: 10.1016/j.biotechadv.2024.108317] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Revised: 12/27/2023] [Accepted: 01/06/2024] [Indexed: 01/16/2024]
Abstract
The separation of specific cell populations is instrumental in gaining insights into cellular processes, elucidating disease mechanisms, and advancing applications in tissue engineering, regenerative medicine, diagnostics, and cell therapies. Microfluidic methods for cell separation have propelled the field forward, benefitting from miniaturization, advanced fabrication technologies, a profound understanding of fluid dynamics governing particle separation mechanisms, and a surge in interdisciplinary investigations focused on diverse applications. Cell separation methodologies can be categorized according to their underlying separation mechanisms. Passive microfluidic separation systems rely on channel structures and fluidic rheology, obviating the necessity for external force fields to facilitate label-free cell separation. These passive approaches offer a compelling combination of cost-effectiveness and scalability when compared to active methods that depend on external fields to manipulate cells. This review delves into the extensive utilization of passive microfluidic techniques for cell separation, encompassing various strategies such as filtration, sedimentation, adhesion-based techniques, pinched flow fractionation (PFF), deterministic lateral displacement (DLD), inertial microfluidics, hydrophoresis, viscoelastic microfluidics, and hybrid microfluidics. Besides, the review provides an in-depth discussion concerning cell types, separation markers, and the commercialization of these technologies. Subsequently, it outlines the current challenges faced in the field and presents a forward-looking perspective on potential future developments. This work hopes to aid in facilitating the dissemination of knowledge in cell separation, guiding future research, and informing practical applications across diverse scientific disciplines.
Collapse
Affiliation(s)
- Tianlong Zhang
- College of Mechanical Engineering, Jiangsu University of Science and Technology, Zhenjiang 212100, China
| | - Dino Di Carlo
- Department of Bioengineering, University of California, Los Angeles, CA 90095, USA
| | - Chwee Teck Lim
- Department of Biomedical Engineering, National University of Singapore, Singapore 117583, Singapore
| | - Tianyuan Zhou
- College of Mechanical Engineering, Jiangsu University of Science and Technology, Zhenjiang 212100, China
| | - Guizhong Tian
- College of Mechanical Engineering, Jiangsu University of Science and Technology, Zhenjiang 212100, China.
| | - Tao Tang
- Department of Biomedical Engineering, National University of Singapore, Singapore 117583, Singapore
| | - Amy Q Shen
- Micro/Bio/Nanofluidics Unit, Okinawa Institute of Science and Technology Graduate University, Onna-son, Okinawa 904-0495, Japan
| | - Weihua Li
- School of Mechanical, Materials, Mechatronic and Biomedical Engineering, University of Wollongong, Wollongong, NSW 2522, Australia
| | - Ming Li
- School of Mechanical and Manufacturing Engineering, University of New South Wales, Sydney, NSW 2052, Australia
| | - Yang Yang
- Institute of Deep-Sea Science and Engineering, Chinese Academy of Sciences, Sanya, Hainan 572000, China
| | - Keisuke Goda
- Department of Bioengineering, University of California, Los Angeles, CA 90095, USA; Department of Chemistry, University of Tokyo, Tokyo 113-0033, Japan; The Institute of Technological Sciences, Wuhan University, Wuhan 430072, China
| | - Ruopeng Yan
- The Institute of Technological Sciences, Wuhan University, Wuhan 430072, China
| | - Cheng Lei
- The Institute of Technological Sciences, Wuhan University, Wuhan 430072, China
| | - Yoichiroh Hosokawa
- Division of Materials Science, Nara Institute of Science and Technology, Nara 630-0192, Japan
| | - Yaxiaer Yalikun
- Division of Materials Science, Nara Institute of Science and Technology, Nara 630-0192, Japan.
| |
Collapse
|
46
|
Zhou J, Dong J, Hou H, Huang L, Li J. High-throughput microfluidic systems accelerated by artificial intelligence for biomedical applications. LAB ON A CHIP 2024; 24:1307-1326. [PMID: 38247405 DOI: 10.1039/d3lc01012k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/23/2024]
Abstract
High-throughput microfluidic systems are widely used in biomedical fields for tasks like disease detection, drug testing, and material discovery. Despite the great advances in automation and throughput, the large amounts of data generated by the high-throughput microfluidic systems generally outpace the abilities of manual analysis. Recently, the convergence of microfluidic systems and artificial intelligence (AI) has been promising in solving the issue by significantly accelerating the process of data analysis as well as improving the capability of intelligent decision. This review offers a comprehensive introduction on AI methods and outlines the current advances of high-throughput microfluidic systems accelerated by AI, covering biomedical detection, drug screening, and automated system control and design. Furthermore, the challenges and opportunities in this field are critically discussed as well.
Collapse
Affiliation(s)
- Jianhua Zhou
- School of Biomedical Engineering, Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, China.
- Key Laboratory of Sensing Technology and Biomedical Instruments of Guangdong Province, School of Biomedical Engineering, Sun Yat-sen University, Shenzhen 518107, China
| | - Jianpei Dong
- School of Biomedical Engineering, Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, China.
- Key Laboratory of Sensing Technology and Biomedical Instruments of Guangdong Province, School of Biomedical Engineering, Sun Yat-sen University, Shenzhen 518107, China
| | - Hongwei Hou
- Beijing Life Science Academy, Beijing 102209, China
| | - Lu Huang
- School of Biomedical Engineering, Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, China.
- Key Laboratory of Sensing Technology and Biomedical Instruments of Guangdong Province, School of Biomedical Engineering, Sun Yat-sen University, Shenzhen 518107, China
| | - Jinghong Li
- Department of Chemistry, Center for BioAnalytical Chemistry, Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Tsinghua University, Beijing 100084, China.
- New Cornerstone Science Laboratory, Shenzhen 518054, China
- Beijing Life Science Academy, Beijing 102209, China
- Center for BioAnalytical Chemistry, Hefei National Laboratory of Physical Science at Microscale, University of Science and Technology of China, Hefei 230026, China
| |
Collapse
|
47
|
Camunas-Soler J. Integrating single-cell transcriptomics with cellular phenotypes: cell morphology, Ca 2+ imaging and electrophysiology. Biophys Rev 2024; 16:89-107. [PMID: 38495444 PMCID: PMC10937895 DOI: 10.1007/s12551-023-01174-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Accepted: 11/29/2023] [Indexed: 03/19/2024] Open
Abstract
I review recent technological advancements in coupling single-cell transcriptomics with cellular phenotypes including morphology, calcium signaling, and electrophysiology. Single-cell RNA sequencing (scRNAseq) has revolutionized cell type classifications by capturing the transcriptional diversity of cells. A new wave of methods to integrate scRNAseq and biophysical measurements is facilitating the linkage of transcriptomic data to cellular function, which provides physiological insight into cellular states. I briefly discuss critical factors of these phenotypical characterizations such as timescales, information content, and analytical tools. Dedicated sections focus on the integration with cell morphology, calcium imaging, and electrophysiology (patch-seq), emphasizing their complementary roles. I discuss their application in elucidating cellular states, refining cell type classifications, and uncovering functional differences in cell subtypes. To illustrate the practical applications and benefits of these methods, I highlight their use in tissues with excitable cell-types such as the brain, pancreatic islets, and the retina. The potential of combining functional phenotyping with spatial transcriptomics for a detailed mapping of cell phenotypes in situ is explored. Finally, I discuss open questions and future perspectives, emphasizing the need for a shift towards broader accessibility through increased throughput.
Collapse
Affiliation(s)
- Joan Camunas-Soler
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, University of Gothenburg, 405 30 Gothenburg, Sweden
- Wallenberg Centre for Molecular and Translational Medicine, Sahlgrenska Academy, University of Gothenburg, 405 30 Gothenburg, Sweden
| |
Collapse
|
48
|
Lee S, Kim G, Lee J, Lee AC, Kwon S. Mapping cancer biology in space: applications and perspectives on spatial omics for oncology. Mol Cancer 2024; 23:26. [PMID: 38291400 PMCID: PMC10826015 DOI: 10.1186/s12943-024-01941-z] [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: 06/19/2023] [Accepted: 01/12/2024] [Indexed: 02/01/2024] Open
Abstract
Technologies to decipher cellular biology, such as bulk sequencing technologies and single-cell sequencing technologies, have greatly assisted novel findings in tumor biology. Recent findings in tumor biology suggest that tumors construct architectures that influence the underlying cancerous mechanisms. Increasing research has reported novel techniques to map the tissue in a spatial context or targeted sampling-based characterization and has introduced such technologies to solve oncology regarding tumor heterogeneity, tumor microenvironment, and spatially located biomarkers. In this study, we address spatial technologies that can delineate the omics profile in a spatial context, novel findings discovered via spatial technologies in oncology, and suggest perspectives regarding therapeutic approaches and further technological developments.
Collapse
Affiliation(s)
- Sumin Lee
- Department of Electrical and Computer Engineering, Seoul National University, Seoul, 08826, Republic of Korea
- Meteor Biotech,, Co. Ltd, Seoul, 08826, Republic of Korea
| | - Gyeongjun Kim
- Interdisciplinary Program in Bioengineering, Seoul National University, Seoul, 08826, Republic of Korea
| | - JinYoung Lee
- Division of Engineering Science, University of Toronto, Toronto, Ontario, ON, M5S 3H6, Canada
| | - Amos C Lee
- Meteor Biotech,, Co. Ltd, Seoul, 08826, Republic of Korea.
- Bio-MAX Institute, Seoul National University, Seoul, 08826, Republic of Korea.
| | - Sunghoon Kwon
- Department of Electrical and Computer Engineering, Seoul National University, Seoul, 08826, Republic of Korea.
- Interdisciplinary Program in Bioengineering, Seoul National University, Seoul, 08826, Republic of Korea.
- Bio-MAX Institute, Seoul National University, Seoul, 08826, Republic of Korea.
- Institutes of Entrepreneurial BioConvergence, Seoul National University, Seoul, 08826, Republic of Korea.
- Cancer Research Institute, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea.
| |
Collapse
|
49
|
Xu Z, Chen Z, Yang S, Chen S, Guo T, Chen H. Passive Focusing of Single Cells Using Microwell Arrays for High-Accuracy Image-Activated Sorting. Anal Chem 2024; 96:347-354. [PMID: 38153415 DOI: 10.1021/acs.analchem.3c04195] [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: 12/29/2023]
Abstract
Sorting single cells from a population was of critical importance in areas such as cell line development and cell therapy. Image-based sorting is becoming a promising technique for the nonlabeling isolation of cells due to the capability of providing the details of cell morphology. This study reported the focusing of cells using microwell arrays and the following automatic size sorting based on the real-time recognition of cells. The simulation first demonstrated the converged streamlines to the symmetrical plane contributed to the focusing effect. Then, the influence of connecting microchannel, flowing length, particle size, and the sample flow rate on the focusing effect was experimentally analyzed. Both microspheres and cells could be aligned in a straight line at the Reynolds number (Re) of 0.027-0.187 and 0.027-0.08, respectively. The connecting channel was proved to drastically improve the focusing performance. Afterward, a tapered microwell array was utilized to focus sphere/cell spreading in a wide channel to a straight line. Finally, a custom algorithm was employed to identify and sort the size of microspheres/K562 cells with a throughput of 1 event/s and an accuracy of 97.8/97.1%. The proposed technique aligned cells to a straight line at low Reynolds numbers and greatly facilitated the image-activated sorting without the need for a high-speed camera or flow control components with high frequency. Therefore, it is of enormous application potential in the field of nonlabeled separation of single cells.
Collapse
Affiliation(s)
- Zheng Xu
- School of Mechanical Engineering and Automation, Harbin Institute of Technology, Shenzhen, Shenzhen 518055, China
| | - Zhenlin Chen
- Department of Biomedical Engineering, College of Engineering, Kowloon, City University of Hong Kong, Hong Kong SAR, China
| | - Shiming Yang
- School of Mechanical Engineering and Automation, Harbin Institute of Technology, Shenzhen, Shenzhen 518055, China
| | - Siyuan Chen
- School of Mechanical Engineering and Automation, Harbin Institute of Technology, Shenzhen, Shenzhen 518055, China
| | - Tianruo Guo
- Graduate School of Biomedical Engineering, The University of New South Wales, Sydney, New South Wales 2052, Australia
| | - Huaying Chen
- School of Mechanical Engineering and Automation, Harbin Institute of Technology, Shenzhen, Shenzhen 518055, China
| |
Collapse
|
50
|
Bai Y, Camargo CM, Glasauer SMK, Gifford R, Tian X, Longhini AP, Kosik KS. Single-cell mapping of lipid metabolites using an infrared probe in human-derived model systems. Nat Commun 2024; 15:350. [PMID: 38191490 PMCID: PMC10774263 DOI: 10.1038/s41467-023-44675-0] [Citation(s) in RCA: 18] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Accepted: 12/20/2023] [Indexed: 01/10/2024] Open
Abstract
Understanding metabolic heterogeneity is the key to uncovering the underlying mechanisms of metabolic-related diseases. Current metabolic imaging studies suffer from limitations including low resolution and specificity, and the model systems utilized often lack human relevance. Here, we present a single-cell metabolic imaging platform to enable direct imaging of lipid metabolism with high specificity in various human-derived 2D and 3D culture systems. Through the incorporation of an azide-tagged infrared probe, selective detection of newly synthesized lipids in cells and tissue became possible, while simultaneous fluorescence imaging enabled cell-type identification in complex tissues. In proof-of-concept experiments, newly synthesized lipids were directly visualized in human-relevant model systems among different cell types, mutation status, differentiation stages, and over time. We identified upregulated lipid metabolism in progranulin-knockdown human induced pluripotent stem cells and in their differentiated microglia cells. Furthermore, we observed that neurons in brain organoids exhibited a significantly lower lipid metabolism compared to astrocytes.
Collapse
Affiliation(s)
- Yeran Bai
- Neuroscience Research Institute, Department of Molecular, Cellular, and Developmental Biology, University of California, Santa Barbara, CA, USA.
- Photothermal Spectroscopy Corp., Santa Barbara, CA, USA.
| | - Carolina M Camargo
- Neuroscience Research Institute, Department of Molecular, Cellular, and Developmental Biology, University of California, Santa Barbara, CA, USA
| | - Stella M K Glasauer
- Neuroscience Research Institute, Department of Molecular, Cellular, and Developmental Biology, University of California, Santa Barbara, CA, USA
| | - Raymond Gifford
- Neuroscience Research Institute, Department of Molecular, Cellular, and Developmental Biology, University of California, Santa Barbara, CA, USA
| | - Xinran Tian
- Neuroscience Research Institute, Department of Molecular, Cellular, and Developmental Biology, University of California, Santa Barbara, CA, USA
| | - Andrew P Longhini
- Neuroscience Research Institute, Department of Molecular, Cellular, and Developmental Biology, University of California, Santa Barbara, CA, USA
| | - Kenneth S Kosik
- Neuroscience Research Institute, Department of Molecular, Cellular, and Developmental Biology, University of California, Santa Barbara, CA, USA.
| |
Collapse
|