1
|
Li G, Nichols EK, Browning VE, Longhi NJ, Sanchez-Forman M, Camplisson CK, Beliveau BJ, Noble WS. Predicting cell cycle stage from 3D single-cell nuclear-stained images. Life Sci Alliance 2025; 8:e202403067. [PMID: 40180577 PMCID: PMC11969383 DOI: 10.26508/lsa.202403067] [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: 09/27/2024] [Revised: 03/16/2025] [Accepted: 03/17/2025] [Indexed: 04/05/2025] Open
Abstract
The cell cycle governs the proliferation of all eukaryotic cells. Profiling cell cycle dynamics is therefore central to basic and biomedical research. However, current approaches to cell cycle profiling involve complex interventions that may confound experimental interpretation. We developed CellCycleNet, a machine learning (ML) workflow, to simplify cell cycle staging from fluorescent microscopy data with minimal experimenter intervention and cost. CellCycleNet accurately predicts cell cycle phase using only a fluorescent nuclear stain (DAPI) in fixed interphase cells. Using the Fucci2a cell cycle reporter system as ground truth, we collected two benchmarking image datasets and trained 2D and 3D ML models-of support vector machine and deep neural network architecture-to classify nuclei in the G1 or S/G2 phases. Our results show that 3D CellCycleNet outperforms support vector machine models on each dataset. When trained on two image datasets simultaneously, CellCycleNet achieves the highest classification accuracy (AUROC of 0.94-0.95). Overall, we found that using 3D features, rather than 2D features alone, significantly improves classification performance for all model architectures. We released our image data, models, and software as a community resource.
Collapse
Affiliation(s)
- Gang Li
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
- eScience Institute, University of Washington, Seattle, WA, USA
| | - Eva K Nichols
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
| | | | - Nicolas J Longhi
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
| | | | - Conor K Camplisson
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
| | - Brian J Beliveau
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
- Brotman Baty Institute for Precision Medicine, Seattle, WA, USA
| | - William Stafford Noble
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
- Paul G. Allen School of Computer Science and Engineering, University of Washington, Seattle, WA, USA
| |
Collapse
|
2
|
Ahmed EH, Farrag SM, El-Latif NA. Evaluating the effects of L-carnitine on albino rat's gingiva-derived stem cells (In-Vitro Study). Arch Oral Biol 2025; 173:106192. [PMID: 39954495 DOI: 10.1016/j.archoralbio.2025.106192] [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: 11/08/2024] [Revised: 12/24/2024] [Accepted: 02/08/2025] [Indexed: 02/17/2025]
Abstract
OBJECTIVE Stem cells as therapy is currently a well-established scientific research topic. Poor maintenance and survival of cells supplied to the damaged tissue are barriers to improving the efficacy of regenerative medicine. Antioxidants such as L-carnitine are used to promote cell survival and maintenance properties. This study aims to assess the effects of L-carnitine on albino rat gingiva-derived mesenchymal stem cells proliferation. DESIGN Rat gingiva-derived mesenchymal stem cells were isolated and exposed to 0, 1, 3, and 10 Mm of L-carnitine. Flow cytometry was then utilized to measure gene and protein expression levels for CD90, CD105, CD45, and CD19. The MTT test was used to examine the proliferation of cells. The proportion of apoptosis was determined using the Annexin V/PI technique. Cell cycle investigations to assess cells and identify the percentages of cells in the G0/G1, S, and G2/M phases. Expression of TGF-β gene has been evaluated using Real time‑PCR analysis. RESULTS The results showed that gingiva-derived mesenchymal stem cells, including CD90 and CD105, consistently showed positive immunostaining, whereas CD45 and CD19 were weakly positive or negative. Concentration-dependent increase of growth proliferation, more rapid proliferation of the cells treated with the highest L-carnitine concentration (10 mM) after 72 h (0.934 ± 0.063). Cells treated with 10 mM L-carnitine showed considerably decreased percentages of necrotic (2.38 ± 0.55), late (1.23 ± 0.90), early apoptotic cells (1.18 ± 0.13), and increased the percentage of viable cells (95.13 ± 1.61). CONCLUSION Our findings suggest that adding L-carnitine to gingiva-derived mesenchymal stem cells during expansion enables efficient and viable cell production.
Collapse
Affiliation(s)
- Elham H Ahmed
- Lecturer of Oral Biology, Faculty of Dentistry, Mansoura University, Mansoura, Egypt.
| | - Sara Mohamed Farrag
- Lab specialist, Medical Experimental Research Center (MERC), Mansoura University, Egypt.
| | - Noura Abd El-Latif
- Lecturer of Oral Biology, Faculty of Dentistry, Mansoura University, Mansoura, Egypt.
| |
Collapse
|
3
|
Hang R, Yao X, Bai L, Hang R. Evolving biomaterials design from trial and error to intelligent innovation. Acta Biomater 2025; 197:29-47. [PMID: 40081552 DOI: 10.1016/j.actbio.2025.03.013] [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: 11/20/2024] [Revised: 01/20/2025] [Accepted: 03/06/2025] [Indexed: 03/16/2025]
Abstract
The design and exploration of biomaterials plays a pivotal role in many fields, including medical and engineering. The prevailing approach to biomaterials discovery relies on orthogonal experiments, with repeated attempts to optimize experimental conditions. This method has proven invaluable in gaining experience, but it is also inefficient and challenging to predict the behavior of complex systems. The advent of high-throughput screening (HTS) techniques has led to a notable enhancement in the efficiency of biomaterials development, enabling researchers to assess a vast array of material combinations within a relatively short timeframe. Nevertheless, the emergence of artificial intelligence (AI) has been the catalyst for a new era in biomaterials design. AI has markedly accelerated the development of new materials by enabling the prediction of material properties through machine learning (ML) and deep learning models, as well as optimizing the design pipeline. This review will present a systematic overview of the development of biomaterials design technology. It will also explore the integration of AI with HTS technology and envisage the potential of AI-driven materials design in biomaterials for the future. STATEMENT OF SIGNIFICANCE: The design and synthesis of biomaterials have undergone substantial shifts, reflecting evolving research paradigms. High-throughput screening has emerged as a broad and efficient alternative to traditional free-form combination methods in biomaterial design. The advent of artificial intelligence (AI) enables personalized biomaterial design and, as a transformative tool in biomaterial development, is poised to redefine the field and offer long-term solutions for its advancement. Building on these advancements, this review systematically summarizes the evolution of biomaterial design, offering insights into the future trajectory of the field.
Collapse
Affiliation(s)
- Ruiyue Hang
- Shanxi Key Laboratory of Biomedical Metal Materials, College of Materials Science and Engineering, Taiyuan University of Technology, Taiyuan 030024, PR China
| | - Xiaohong Yao
- Shanxi Key Laboratory of Biomedical Metal Materials, College of Materials Science and Engineering, Taiyuan University of Technology, Taiyuan 030024, PR China
| | - Long Bai
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, PR China.
| | - Ruiqiang Hang
- Shanxi Key Laboratory of Biomedical Metal Materials, College of Materials Science and Engineering, Taiyuan University of Technology, Taiyuan 030024, PR China.
| |
Collapse
|
4
|
Zhang Z, Zhou X, Fang Y, Xiong Z, Zhang T. AI-driven 3D bioprinting for regenerative medicine: From bench to bedside. Bioact Mater 2025; 45:201-230. [PMID: 39651398 PMCID: PMC11625302 DOI: 10.1016/j.bioactmat.2024.11.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Revised: 11/01/2024] [Accepted: 11/16/2024] [Indexed: 12/11/2024] Open
Abstract
In recent decades, 3D bioprinting has garnered significant research attention due to its ability to manipulate biomaterials and cells to create complex structures precisely. However, due to technological and cost constraints, the clinical translation of 3D bioprinted products (BPPs) from bench to bedside has been hindered by challenges in terms of personalization of design and scaling up of production. Recently, the emerging applications of artificial intelligence (AI) technologies have significantly improved the performance of 3D bioprinting. However, the existing literature remains deficient in a methodological exploration of AI technologies' potential to overcome these challenges in advancing 3D bioprinting toward clinical application. This paper aims to present a systematic methodology for AI-driven 3D bioprinting, structured within the theoretical framework of Quality by Design (QbD). This paper commences by introducing the QbD theory into 3D bioprinting, followed by summarizing the technology roadmap of AI integration in 3D bioprinting, including multi-scale and multi-modal sensing, data-driven design, and in-line process control. This paper further describes specific AI applications in 3D bioprinting's key elements, including bioink formulation, model structure, printing process, and function regulation. Finally, the paper discusses current prospects and challenges associated with AI technologies to further advance the clinical translation of 3D bioprinting.
Collapse
Affiliation(s)
- Zhenrui Zhang
- Biomanufacturing Center, Department of Mechanical Engineering, Tsinghua University, Beijing, 100084, PR China
- Biomanufacturing and Rapid Forming Technology Key Laboratory of Beijing, Beijing, 100084, PR China
- “Biomanufacturing and Engineering Living Systems” Innovation International Talents Base (111 Base), Beijing, 100084, PR China
| | - Xianhao Zhou
- Biomanufacturing Center, Department of Mechanical Engineering, Tsinghua University, Beijing, 100084, PR China
- Biomanufacturing and Rapid Forming Technology Key Laboratory of Beijing, Beijing, 100084, PR China
- “Biomanufacturing and Engineering Living Systems” Innovation International Talents Base (111 Base), Beijing, 100084, PR China
| | - Yongcong Fang
- Biomanufacturing Center, Department of Mechanical Engineering, Tsinghua University, Beijing, 100084, PR China
- Biomanufacturing and Rapid Forming Technology Key Laboratory of Beijing, Beijing, 100084, PR China
- “Biomanufacturing and Engineering Living Systems” Innovation International Talents Base (111 Base), Beijing, 100084, PR China
- State Key Laboratory of Tribology in Advanced Equipment, Tsinghua University, Beijing, 100084, PR China
| | - Zhuo Xiong
- Biomanufacturing Center, Department of Mechanical Engineering, Tsinghua University, Beijing, 100084, PR China
- Biomanufacturing and Rapid Forming Technology Key Laboratory of Beijing, Beijing, 100084, PR China
- “Biomanufacturing and Engineering Living Systems” Innovation International Talents Base (111 Base), Beijing, 100084, PR China
| | - Ting Zhang
- Biomanufacturing Center, Department of Mechanical Engineering, Tsinghua University, Beijing, 100084, PR China
- Biomanufacturing and Rapid Forming Technology Key Laboratory of Beijing, Beijing, 100084, PR China
- “Biomanufacturing and Engineering Living Systems” Innovation International Talents Base (111 Base), Beijing, 100084, PR China
- State Key Laboratory of Tribology in Advanced Equipment, Tsinghua University, Beijing, 100084, PR China
| |
Collapse
|
5
|
Jeong IJ, Hong JK, Bae YJ, Lee TK. Enhancing Bacterial Phenotype Classification Through the Integration of Autogating and Automated Machine Learning in Flow Cytometric Analysis. Cytometry A 2025; 107:203-213. [PMID: 40062709 DOI: 10.1002/cyto.a.24923] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Revised: 12/17/2024] [Accepted: 02/27/2025] [Indexed: 04/11/2025]
Abstract
Although flow cytometry produces reliable results, the data processing from gating to fingerprinting is prone to subjective bias. Here, we integrated autogating with Automated Machine Learning in flow cytometry to enhance the classification of bacterial phenotypes. We analyzed six bacterial strains prevalent in the soil and groundwater- Bacillus subtilis , Burkholderia thailandensis , Corynebacterium glutamicum , Escherichia coli , Pseudomonas putida , and Pseudomonas stutzeri . Using the H2O-AutoML framework, we applied gradient-boosting machine (GBM) models to classify bacteria across different metabolic phases. Our results demonstrated an overall classification accuracy of 82.34% for GBM. Notably, accuracy varied across metabolic phases, with the highest observed during the late log (88.06%), lag (88.43%), and early log phases (89.37%), whereas the stationary phase showed a slightly lower accuracy of 80.73%. P. stutzeri exhibited consistently high sensitivity and specificity across all the phases, which indicated that it was the most distinctly identifiable strain. In contrast, E. coli showed low sensitivity, particularly in the stationary phase, which indicated challenges in its classification. Overall, this study with incorporating autogating and the AutoML framework, substantially reduces subjective biases and enhances the reproducibility and accuracy of microbial classification. Our methodology offers a robust framework for microbial classification in flow cytometric analysis, paving the way for more precise and comprehensive analyses of microbial ecology.
Collapse
Affiliation(s)
- In Jae Jeong
- Department of Environmental and Energy Engineering, Yonsei University, Wonju, Republic of Korea
| | - Jin-Kyung Hong
- Department of Environmental and Energy Engineering, Yonsei University, Wonju, Republic of Korea
| | - Young Jun Bae
- Department of Environmental and Energy Engineering, Yonsei University, Wonju, Republic of Korea
| | - Tea Kwon Lee
- Department of Environmental and Energy Engineering, Yonsei University, Wonju, Republic of Korea
| |
Collapse
|
6
|
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
|
7
|
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
|
8
|
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
|
9
|
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 PMCID: PMC11685671 DOI: 10.1038/s41467-024-55094-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [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
|
10
|
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
|
11
|
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
|
12
|
Zhou C, Liu C, Liao Z, Pang Y, Sun W. AI for biofabrication. Biofabrication 2024; 17:012004. [PMID: 39433065 DOI: 10.1088/1758-5090/ad8966] [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: 05/05/2024] [Accepted: 10/21/2024] [Indexed: 10/23/2024]
Abstract
Biofabrication is an advanced technology that holds great promise for constructing highly biomimeticin vitrothree-dimensional human organs. Such technology would help address the issues of immune rejection and organ donor shortage in organ transplantation, aiding doctors in formulating personalized treatments for clinical patients and replacing animal experiments. Biofabrication typically involves the interdisciplinary application of biology, materials science, mechanical engineering, and medicine to generate large amounts of data and correlations that require processing and analysis. Artificial intelligence (AI), with its excellent capabilities in big data processing and analysis, can play a crucial role in handling and processing interdisciplinary data and relationships and in better integrating and applying them in biofabrication. In recent years, the development of the semiconductor and integrated circuit industries has propelled the rapid advancement of computer processing power. An AI program can learn and iterate multiple times within a short period, thereby gaining strong automation capabilities for a specific research content or issue. To date, numerous AI programs have been applied to various processes around biofabrication, such as extracting biological information, designing and optimizing structures, intelligent cell sorting, optimizing biomaterials and processes, real-time monitoring and evaluation of models, accelerating the transformation and development of these technologies, and even changing traditional research patterns. This article reviews and summarizes the significant changes and advancements brought about by AI in biofabrication, and discusses its future application value and direction.
Collapse
Affiliation(s)
- Chang Zhou
- Biomanufacturing Center, Department of Mechanical Engineering, Tsinghua University, Beijing 100084, People's Republic of China
- Biomanufacturing and Rapid Forming Technology Key Laboratory of Beijing, Beijing 100084, People's Republic of China
| | - Changru Liu
- Biomanufacturing Center, Department of Mechanical Engineering, Tsinghua University, Beijing 100084, People's Republic of China
- Biomanufacturing and Rapid Forming Technology Key Laboratory of Beijing, Beijing 100084, People's Republic of China
| | - Zhendong Liao
- Biomanufacturing Center, Department of Mechanical Engineering, Tsinghua University, Beijing 100084, People's Republic of China
- Biomanufacturing and Rapid Forming Technology Key Laboratory of Beijing, Beijing 100084, People's Republic of China
| | - Yuan Pang
- Biomanufacturing Center, Department of Mechanical Engineering, Tsinghua University, Beijing 100084, People's Republic of China
- Biomanufacturing and Rapid Forming Technology Key Laboratory of Beijing, Beijing 100084, People's Republic of China
| | - Wei Sun
- Biomanufacturing Center, Department of Mechanical Engineering, Tsinghua University, Beijing 100084, People's Republic of China
- Biomanufacturing and Rapid Forming Technology Key Laboratory of Beijing, Beijing 100084, People's Republic of China
- Department of Mechanical Engineering, Drexel University, Philadelphia, PA 19104, United States of America
| |
Collapse
|
13
|
Lippeveld M, Peralta D, Filby A, Saeys Y. SCIP: A scalable, reproducible and open-source pipeline for morphological profiling of image cytometry and microscopy data. Cytometry A 2024; 105:816-828. [PMID: 39351999 DOI: 10.1002/cyto.a.24896] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Revised: 06/17/2024] [Accepted: 08/23/2024] [Indexed: 11/16/2024]
Abstract
Imaging flow cytometry (IFC) provides single-cell imaging data at a high acquisition rate. It is increasingly used in image-based profiling experiments consisting of hundreds of thousands of multi-channel images of cells. Currently available software solutions for processing microscopy data can provide good results in downstream analysis, but are limited in efficiency and scalability, and often ill-adapted to IFC data. In this work, we propose Scalable Cytometry Image Processing (SCIP), a Python software that efficiently processes images from IFC and standard microscopy datasets. We also propose a file format for efficiently storing IFC data. We showcase our contributions on two large-scale microscopy and one IFC datasets, all of which are publicly available. Our results show that SCIP can extract the same kind of information as other tools, in a much shorter time and in a more scalable manner.
Collapse
Affiliation(s)
- Maxim Lippeveld
- Data Mining and Modelling for Biomedicine, VIB Center for Inflammation Research, Ghent, Belgium
- Department of Applied Mathematics, Computer Science and Statistics, Ghent University, Ghent, Belgium
| | - Daniel Peralta
- IDLab, Department of Information Technology, Ghent University - imec, Ghent, Belgium
| | - Andrew Filby
- Biosciences Institute and Innovation Methodology and Application Research Theme, Newcastle University, Newcastle upon Tyne, UK
| | - Yvan Saeys
- Data Mining and Modelling for Biomedicine, VIB Center for Inflammation Research, Ghent, Belgium
- Department of Applied Mathematics, Computer Science and Statistics, Ghent University, Ghent, Belgium
| |
Collapse
|
14
|
Tsilikas I, Tsirigotis A, Sarantoglou G, Deligiannidis S, Bogris A, Posch C, Van den Branden G, Mesaritakis C. Photonic neuromorphic accelerators for event-based imaging flow cytometry. Sci Rep 2024; 14:24179. [PMID: 39406898 PMCID: PMC11480101 DOI: 10.1038/s41598-024-75667-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Accepted: 10/05/2024] [Indexed: 10/19/2024] Open
Abstract
In this work, we present experimental results of a high-speed label-free imaging cytometry system that seamlessly merges the high-capturing rate and data sparsity of an event-based CMOS camera with lightweight photonic neuromorphic processing. This combination offers high classification accuracy and a massive reduction in the number of trainable parameters of the digital machine-learning back-end. The event-based camera is capable of capturing 1 Gevents/sec, where events correspond to pixel contrast changes, similar to the retina's ganglion cell function. The photonic neuromorphic accelerator is based on a hardware-friendly passive optical spectrum slicing technique that is able to extract meaningful features from the generated spike-trains using a purely analogue version of the convolutional operation. The experimental scenario comprises the discrimination of artificial polymethyl methacrylate calibrated beads, having different diameters, flowing at a mean speed of 0.1 m/sec. Classification accuracy, using only lightweight digital machine-learning schemes has topped at 98.2%. On the other hand, by experimentally pre-processing the raw spike data through the proposed photonic neuromorphic spectrum slicer at a rate of 3 × 106 images per second, we achieved an accuracy of 98.6%. This performance was accompanied by a reduction in the number of trainable parameters at the classification back-end by a factor ranging from 8 to 22, depending on the configuration of the digital neural network. These results confirm that neuromorphic sensing and neuromorphic computing can be efficiently merged to a unified bio-inspired system, offering a holistic enhancement in emerging bio-imaging applications.
Collapse
Affiliation(s)
- I Tsilikas
- Department of Information and Communication Systems Engineering, University of the Aegean, Palama 2, 83100, Samos, Greece
- Department of Physics, School of Applied Mathematical and Physical Sciences, Zografou Campus, 157 80, Athens, Greece
| | - A Tsirigotis
- Department of Information and Communication Systems Engineering, University of the Aegean, Palama 2, 83100, Samos, Greece
| | - G Sarantoglou
- Department of Information and Communication Systems Engineering, University of the Aegean, Palama 2, 83100, Samos, Greece
| | - S Deligiannidis
- Department of Informatics and Computer Engineering, University of West Attica, Ag. Spyridonos, Egaleo, Greece
| | - A Bogris
- Department of Informatics and Computer Engineering, University of West Attica, Ag. Spyridonos, Egaleo, Greece
| | - C Posch
- Prophesee Metavision, Rue du Faubourg Saint-Antoine 74, 75012, Paris, France
| | - G Van den Branden
- Prophesee Metavision, Rue du Faubourg Saint-Antoine 74, 75012, Paris, France
| | - C Mesaritakis
- Department of Information and Communication Systems Engineering, University of the Aegean, Palama 2, 83100, Samos, Greece.
| |
Collapse
|
15
|
Howell J, Omwenga S, Jimenez M, Hammarton TC. Analysis of the Leishmania mexicana promastigote cell cycle using imaging flow cytometry provides new insights into cell cycle flexibility and events of short duration. PLoS One 2024; 19:e0311367. [PMID: 39361666 PMCID: PMC11449296 DOI: 10.1371/journal.pone.0311367] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Accepted: 09/17/2024] [Indexed: 10/05/2024] Open
Abstract
Promastigote Leishmania mexicana have a complex cell division cycle characterised by the ordered replication of several single-copy organelles, a prolonged S phase and rapid G2 and cytokinesis phases, accompanied by cell cycle stage-associated morphological changes. Here we exploit these morphological changes to develop a high-throughput and semi-automated imaging flow cytometry (IFC) pipeline to analyse the cell cycle in live L. mexicana. Firstly, we demonstrate that, unlike several other DNA stains, Vybrant™ DyeCycle™ Orange (DCO) is non-toxic and enables quantitative DNA imaging in live promastigotes. Secondly, by tagging the orphan spindle kinesin, KINF, with mNeonGreen, we describe KINF's cell cycle-dependent expression and localisation. Then, by combining manual gating of DCO DNA intensity profiles with automated masking and morphological measurements of parasite images, visual determination of the number of flagella per cell, and automated masking and analysis of mNG:KINF fluorescence, we provide a newly detailed description of L. mexicana promastigote cell cycle events that, for the first time, includes the durations of individual G2, mitosis and post-mitosis phases, and identifies G1 cells within the first 12 minutes of the new cell cycle. Our custom-developed masking and gating scheme allowed us to identify elusive G2 cells and to demonstrate that the CDK-inhibitor, flavopiridol, arrests cells in G2 phase, rather than mitosis, providing proof-of-principle of the utility of IFC for drug mechanism-of-action studies. Further, the high-throughput nature of IFC allowed the close examination of promastigote cytokinesis, revealing considerable flexibility in both the timing of cytokinesis initiation and the direction of furrowing, in contrast to the related kinetoplastid parasite, Trypanosoma brucei and many other cell types. Our new pipeline offers many advantages over traditional methods of cell cycle analysis such as fluorescence microscopy and flow cytometry and paves the way for novel high-throughput analysis of Leishmania cell division.
Collapse
Affiliation(s)
- Jessie Howell
- James Watt School of Engineering, University of Glasgow, Glasgow, United Kingdom
| | - Sulochana Omwenga
- School of Infection and Immunity, University of Glasgow, Glasgow, United Kingdom
| | - Melanie Jimenez
- Biomedical Engineering Department, University of Strathclyde, Glasgow, United Kingdom
| | - Tansy C. Hammarton
- School of Infection and Immunity, University of Glasgow, Glasgow, United Kingdom
| |
Collapse
|
16
|
Wang Y, Zhou S, Quan Y, Liu Y, Zhou B, Chen X, Ma Z, Zhou Y. Label-free spatiotemporal decoding of single-cell fate via acoustic driven 3D tomography. Mater Today Bio 2024; 28:101201. [PMID: 39221213 PMCID: PMC11364901 DOI: 10.1016/j.mtbio.2024.101201] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Revised: 08/08/2024] [Accepted: 08/11/2024] [Indexed: 09/04/2024] Open
Abstract
Label-free three-dimensional imaging plays a crucial role in unraveling the complexities of cellular functions and interactions in biomedical research. Conventional single-cell optical tomography techniques offer affordability and the convenience of bypassing laborious cell labelling protocols. However, these methods are encumbered by restricted illumination scanning ranges on abaxial plane, resulting in the loss of intricate cellular imaging details. The ability to fully control cellular rotation across all angles has emerged as an optimal solution for capturing comprehensive structural details of cells. Here, we introduce a label-free, cost-effective, and readily fabricated contactless acoustic-induced vibration system, specifically designed to enable multi-degree-of-freedom rotation of cells, ultimately attaining stable in-situ rotation. Furthermore, by integrating this system with advanced deep learning technologies, we perform 3D reconstruction and morphological analysis on diverse cell types, thus validating groups of high-precision cell identification. Notably, long-term observation of cells reveals distinct features associated with drug-induced apoptosis in both cancerous and normal cells populations. This methodology, based on deep learning-enabled cell 3D reconstruction, charts a novel trajectory for groups of real-time cellular visualization, offering promising advancements in the realms of drug screening and post-single-cell analysis, thereby addressing potential clinical requisites.
Collapse
Affiliation(s)
- Yuxin Wang
- Joint Key Laboratory of the Ministry of Education, Institute of Applied Physics and Materials Engineering, University of Macau, Avenida da Universidade, Taipa, Macau, 999078, China
| | - Shizheng Zhou
- Joint Key Laboratory of the Ministry of Education, Institute of Applied Physics and Materials Engineering, University of Macau, Avenida da Universidade, Taipa, Macau, 999078, China
| | - Yue Quan
- Joint Key Laboratory of the Ministry of Education, Institute of Applied Physics and Materials Engineering, University of Macau, Avenida da Universidade, Taipa, Macau, 999078, China
| | - Yu Liu
- Joint Key Laboratory of the Ministry of Education, Institute of Applied Physics and Materials Engineering, University of Macau, Avenida da Universidade, Taipa, Macau, 999078, China
| | - Bingpu Zhou
- Joint Key Laboratory of the Ministry of Education, Institute of Applied Physics and Materials Engineering, University of Macau, Avenida da Universidade, Taipa, Macau, 999078, China
| | - Xiuping Chen
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Avenida da Universidade, Taipa, Macau, 999078, China
| | - Zhichao Ma
- Institute of Medical Robotics, School of Biomedical Engineering, Shanghai Jiao Tong University, No.800 Dongchuan Road, Shanghai, 200240, China
| | - Yinning Zhou
- Joint Key Laboratory of the Ministry of Education, Institute of Applied Physics and Materials Engineering, University of Macau, Avenida da Universidade, Taipa, Macau, 999078, China
| |
Collapse
|
17
|
Wang Z, Liu Q, Zhou J, Su X. Single-detector multiplex imaging flow cytometry for cancer cell classification with deep learning. Cytometry A 2024; 105:666-676. [PMID: 39101554 DOI: 10.1002/cyto.a.24890] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 06/27/2024] [Accepted: 07/22/2024] [Indexed: 08/06/2024]
Abstract
Imaging flow cytometry, which combines the advantages of flow cytometry and microscopy, has emerged as a powerful tool for cell analysis in various biomedical fields such as cancer detection. In this study, we develop multiplex imaging flow cytometry (mIFC) by employing a spatial wavelength division multiplexing technique. Our mIFC can simultaneously obtain brightfield and multi-color fluorescence images of individual cells in flow, which are excited by a metal halide lamp and measured by a single detector. Statistical analysis results of multiplex imaging experiments with resolution test lens, magnification test lens, and fluorescent microspheres validate the operation of the mIFC with good imaging channel consistency and micron-scale differentiation capabilities. A deep learning method is designed for multiplex image processing that consists of three deep learning networks (U-net, very deep super resolution, and visual geometry group 19). It is demonstrated that the cluster of differentiation 24 (CD24) imaging channel is more sensitive than the brightfield, nucleus, or cancer antigen 125 (CA125) imaging channel in classifying the three types of ovarian cell lines (IOSE80 normal cell, A2780, and OVCAR3 cancer cells). An average accuracy rate of 97.1% is achieved for the classification of these three types of cells by deep learning analysis when all four imaging channels are considered. Our single-detector mIFC is promising for the development of future imaging flow cytometers and for the automatic single-cell analysis with deep learning in various biomedical fields.
Collapse
Affiliation(s)
- Zhiwen Wang
- School of Integrated Circuits, Shandong University, Jinan, China
- Institute of Biomedical Engineering, School of Control Science and Engineering, Shandong University, Jinan, China
| | - Qiao Liu
- Department of Molecular Medicine and Genetics, School of Basic Medicine Sciences, Shandong University, Jinan, China
| | - Jie Zhou
- School of Integrated Circuits, Shandong University, Jinan, China
| | - Xuantao Su
- School of Integrated Circuits, Shandong University, Jinan, China
| |
Collapse
|
18
|
Li G, Nichols EK, Browning VE, Longhi NJ, Camplisson C, Beliveau BJ, Noble WS. Predicting cell cycle stage from 3D single-cell nuclear-stained images. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.30.610553. [PMID: 39257739 PMCID: PMC11383680 DOI: 10.1101/2024.08.30.610553] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/12/2024]
Abstract
The cell cycle governs the proliferation, differentiation, and regeneration of all eukaryotic cells. Profiling cell cycle dynamics is therefore central to basic and biomedical research spanning development, health, aging, and disease. However, current approaches to cell cycle profiling involve complex interventions that may confound experimental interpretation. To facilitate more efficient cell cycle annotation of microscopy data, we developed CellCycleNet, a machine learning (ML) workflow designed to simplify cell cycle staging with minimal experimenter intervention and cost. CellCycleNet accurately predicts cell cycle phase using only a fluorescent nuclear stain (DAPI) in fixed interphase cells. Using the Fucci2a cell cycle reporter system as ground truth, we collected two benchmarking image datasets and trained two ML models-a support vector machine (SVM) and a deep neural network-to classify nuclei as being in either the G1 or S/G2 phases of the cell cycle. Our results suggest that CellCycleNet outperforms state-of-the-art SVM models on each dataset individually. When trained on two image datasets simultaneously, CellCycleNet achieves the highest classification accuracy, with an improvement in AUROC of 0.08-0.09. The model also demonstrates excellent generalization across different microscopes, achieving an AUROC of 0.95. Overall, using features derived from 3D images, rather than 2D projections of those same images, significantly improves classification performance. We have released our image data, trained models, and software as a community resource.
Collapse
Affiliation(s)
- Gang Li
- Department of Genome Sciences, University of Washington
- eScience Institute, University of Washington
| | | | | | | | | | - Brian J. Beliveau
- Department of Genome Sciences, University of Washington
- Brotman Baty Institute for Precision Medicine, Seattle, WA 98195, USA
| | - William Stafford Noble
- Department of Genome Sciences, University of Washington
- Paul G. Allen School of Computer Science and Engineering, University of Washington
| |
Collapse
|
19
|
Fang G, Qiao Z, Huang L, Zhu H, Xie J, Zhou T, Xiong Z, Su IH, Jin D, Chen YC. Single-cell laser emitting cytometry for label-free nucleolus fingerprinting. Nat Commun 2024; 15:7332. [PMID: 39187494 PMCID: PMC11347630 DOI: 10.1038/s41467-024-51574-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2024] [Accepted: 08/09/2024] [Indexed: 08/28/2024] Open
Abstract
The nucleolus, a recognized biomolecular condensate, serves as the hub for ribosome biogenesis within the cell nucleus. Its quantity and morphology are discernible indicators of cellular functional states. However, precise identification and quantification of nucleoli remain challenging without specific labeling, particularly for suspended cells, tissue-level analysis and high-throughput applications. Here we introduce a single-cell laser emitting cytometry (SLEC) for label-free nucleolus differentiation through light-matter interactions within a Fabry-Perot resonator. The separated gain medium enhances the threshold difference by 36-fold between nucleolus and its surroundings, enabling selective laser emissions at nucleolar area while maintaining lower-order mode. The laser emission image provides insights into structural inhomogeneity, temporal fluid-like dynamics, and pathological application. Lasing spectral fingerprint depicts the quantity and size of nucleoli within a single cell, showcasing the label-free flow cytometry for nucleolus. This approach holds promise for nucleolus-guided cell screening and drug evaluation, advancing the study of diseases such as cancer and neurodegenerative disorders.
Collapse
Affiliation(s)
- Guocheng Fang
- School of Electrical and Electronics Engineering, Nanyang Technological University, Singapore, Singapore
| | - Zhen Qiao
- School of Optical-Electrical and Computer Engineering, University of Shanghai for Science and Technology, Shanghai, China
| | - Luqi Huang
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
| | - Hui Zhu
- School of Electrical and Electronics Engineering, Nanyang Technological University, Singapore, Singapore
| | - Jun Xie
- School of Electrical and Electronics Engineering, Nanyang Technological University, Singapore, Singapore
| | - Tian Zhou
- School of Electrical and Electronics Engineering, Nanyang Technological University, Singapore, Singapore
| | - Zhongshu Xiong
- School of Electrical and Electronics Engineering, Nanyang Technological University, Singapore, Singapore
| | - I-Hsin Su
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
| | - Dayong Jin
- Institute for Biomedical Materials and Devices (IBMD), Faculty of Science, University of Technology Sydney, Sydney, NSW, Australia
| | - Yu-Cheng Chen
- School of Electrical and Electronics Engineering, Nanyang Technological University, Singapore, Singapore.
| |
Collapse
|
20
|
Thite NG, Tuberty-Vaughan E, Wilcox P, Wallace N, Calderon CP, Randolph TW. Stain-Free Approach to Determine and Monitor Cell Heath Using Supervised and Unsupervised Image-Based Deep Learning. J Pharm Sci 2024; 113:2114-2127. [PMID: 38710387 PMCID: PMC11670887 DOI: 10.1016/j.xphs.2024.05.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Revised: 05/01/2024] [Accepted: 05/01/2024] [Indexed: 05/08/2024]
Abstract
Cell-based medicinal products (CBMPs) are a growing class of therapeutics that promise new treatments for complex and rare diseases. Given the inherent complexity of the whole human cells comprising CBMPs, there is a need for robust and fast analytical methods for characterization, process monitoring, and quality control (QC) testing during their manufacture. Existing techniques to evaluate and monitor cell quality typically constitute labor-intensive, expensive, and highly specific staining assays. In this work, we combine image-based deep learning with flow imaging microscopy (FIM) to predict cell health metrics using cellular morphology "fingerprints" extracted from images of unstained Jurkat cells (immortalized human T-lymphocyte cells). A supervised (i.e., algorithm trained with human-generated labels for images) fingerprinting algorithm, trained on images of unstained healthy and dead cells, provides a robust stain-free, non-invasive, and non-destructive method for determining cell viability. Results from the stain-free method are in good agreement with traditional stain-based cytometric viability measurements. Additionally, when trained with images of healthy cells, dead cells and cells undergoing chemically induced apoptosis, the supervised fingerprinting algorithm is able to distinguish between the three cell states, and the results are independent of specific treatments or signaling pathways. We then show that an unsupervised variational autoencoder (VAE) algorithm trained on the same images, but without human-generated labels, is able to distinguish between samples of healthy, dead and apoptotic cells along with cellular debris based on learned morphological features and without human input. With this, we demonstrate that VAEs are a powerful exploratory technique that can be used as a process monitoring analytical tool.
Collapse
Affiliation(s)
- Nidhi G Thite
- Department of Chemical and Biological Engineering, University of Colorado Boulder, Boulder, CO 80309, USA
| | - Emma Tuberty-Vaughan
- Dosage Form Design & Development (DFDD), BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, USA
| | - Paige Wilcox
- Dosage Form Design & Development (DFDD), BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, USA
| | - Nicole Wallace
- Dosage Form Design & Development (DFDD), BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, USA
| | - Christopher P Calderon
- Department of Chemical and Biological Engineering, University of Colorado Boulder, Boulder, CO 80309, USA; Ursa Analytics, Denver, CO 80212, USA
| | - Theodore W Randolph
- Department of Chemical and Biological Engineering, University of Colorado Boulder, Boulder, CO 80309, USA
| |
Collapse
|
21
|
Chen HC, Ma Y, Cheng J, Chen YC. Advances in Single-Cell Techniques for Linking Phenotypes to Genotypes. CANCER HETEROGENEITY AND PLASTICITY 2024; 1:0004. [PMID: 39156821 PMCID: PMC11328949 DOI: 10.47248/chp2401010004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/20/2024]
Abstract
Single-cell analysis has become an essential tool in modern biological research, providing unprecedented insights into cellular behavior and heterogeneity. By examining individual cells, this approach surpasses conventional population-based methods, revealing critical variations in cellular states, responses to environmental cues, and molecular signatures. In the context of cancer, with its diverse cell populations, single-cell analysis is critical for investigating tumor evolution, metastasis, and therapy resistance. Understanding the phenotype-genotype relationship at the single-cell level is crucial for deciphering the molecular mechanisms driving tumor development and progression. This review highlights innovative strategies for selective cell isolation based on desired phenotypes, including robotic aspiration, laser detachment, microraft arrays, optical traps, and droplet-based microfluidic systems. These advanced tools facilitate high-throughput single-cell phenotypic analysis and sorting, enabling the identification and characterization of specific cell subsets, thereby advancing therapeutic innovations in cancer and other diseases.
Collapse
Affiliation(s)
- Hsiao-Chun Chen
- UPMC Hillman Cancer Center, University of Pittsburgh, 5115 Centre Ave, Pittsburgh, PA 15232, USA
- Department of Computational and Systems Biology, University of Pittsburgh, 3420 Forbes Avenue, Pittsburgh, PA 15260, USA
| | - Yushu Ma
- UPMC Hillman Cancer Center, University of Pittsburgh, 5115 Centre Ave, Pittsburgh, PA 15232, USA
- Department of Computational and Systems Biology, University of Pittsburgh, 3420 Forbes Avenue, Pittsburgh, PA 15260, USA
| | - Jinxiong Cheng
- UPMC Hillman Cancer Center, University of Pittsburgh, 5115 Centre Ave, Pittsburgh, PA 15232, USA
- Department of Bioengineering, Swanson School of Engineering, University of Pittsburgh, 3700 O'Hara Street, Pittsburgh, PA 15260, USA
| | - Yu-Chih Chen
- UPMC Hillman Cancer Center, University of Pittsburgh, 5115 Centre Ave, Pittsburgh, PA 15232, USA
- Department of Computational and Systems Biology, University of Pittsburgh, 3420 Forbes Avenue, Pittsburgh, PA 15260, USA
- Department of Bioengineering, Swanson School of Engineering, University of Pittsburgh, 3700 O'Hara Street, Pittsburgh, PA 15260, USA
- CMU-Pitt Ph.D. Program in Computational Biology, University of Pittsburgh, 3420 Forbes Avenue, Pittsburgh, PA 15260, USA
| |
Collapse
|
22
|
Chiang CC, Anne R, Chawla P, Shaw RM, He S, Rock EC, Zhou M, Cheng J, Gong YN, Chen YC. Deep learning unlocks label-free viability assessment of cancer spheroids in microfluidics. LAB ON A CHIP 2024; 24:3169-3182. [PMID: 38804084 PMCID: PMC11165951 DOI: 10.1039/d4lc00197d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/02/2024] [Accepted: 05/22/2024] [Indexed: 05/29/2024]
Abstract
Despite recent advances in cancer treatment, refining therapeutic agents remains a critical task for oncologists. Precise evaluation of drug effectiveness necessitates the use of 3D cell culture instead of traditional 2D monolayers. Microfluidic platforms have enabled high-throughput drug screening with 3D models, but current viability assays for 3D cancer spheroids have limitations in reliability and cytotoxicity. This study introduces a deep learning model for non-destructive, label-free viability estimation based on phase-contrast images, providing a cost-effective, high-throughput solution for continuous spheroid monitoring in microfluidics. Microfluidic technology facilitated the creation of a high-throughput cancer spheroid platform with approximately 12 000 spheroids per chip for drug screening. Validation involved tests with eight conventional chemotherapeutic drugs, revealing a strong correlation between viability assessed via LIVE/DEAD staining and phase-contrast morphology. Extending the model's application to novel compounds and cell lines not in the training dataset yielded promising results, implying the potential for a universal viability estimation model. Experiments with an alternative microscopy setup supported the model's transferability across different laboratories. Using this method, we also tracked the dynamic changes in spheroid viability during the course of drug administration. In summary, this research integrates a robust platform with high-throughput microfluidic cancer spheroid assays and deep learning-based viability estimation, with broad applicability to various cell lines, compounds, and research settings.
Collapse
Affiliation(s)
- Chun-Cheng Chiang
- UPMC Hillman Cancer Center, University of Pittsburgh, 5115 Centre Ave, Pittsburgh, PA 15232, USA.
- Department of Computational and Systems Biology, University of Pittsburgh, 3420 Forbes Avenue, Pittsburgh, PA 15260, USA
| | - Rajiv Anne
- UPMC Hillman Cancer Center, University of Pittsburgh, 5115 Centre Ave, Pittsburgh, PA 15232, USA.
- Department of Bioengineering, Swanson School of Engineering, University of Pittsburgh, 3700 O'Hara Street, Pittsburgh, PA 15260, USA
| | - Pooja Chawla
- Department of Bioengineering, Swanson School of Engineering, University of Pittsburgh, 3700 O'Hara Street, Pittsburgh, PA 15260, USA
| | - Rachel M Shaw
- Department of Bioengineering, Swanson School of Engineering, University of Pittsburgh, 3700 O'Hara Street, Pittsburgh, PA 15260, USA
| | - Sarah He
- UPMC Hillman Cancer Center, University of Pittsburgh, 5115 Centre Ave, Pittsburgh, PA 15232, USA.
- Carnegie Mellon University, Department of Biological Sciences, 5000 Forbes Avenue, Pittsburgh, PA, 15213, USA
| | - Edwin C Rock
- Department of Bioengineering, Swanson School of Engineering, University of Pittsburgh, 3700 O'Hara Street, Pittsburgh, PA 15260, USA
| | - Mengli Zhou
- UPMC Hillman Cancer Center, University of Pittsburgh, 5115 Centre Ave, Pittsburgh, PA 15232, USA.
- Department of Computational and Systems Biology, University of Pittsburgh, 3420 Forbes Avenue, Pittsburgh, PA 15260, USA
- Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
| | - Jinxiong Cheng
- UPMC Hillman Cancer Center, University of Pittsburgh, 5115 Centre Ave, Pittsburgh, PA 15232, USA.
- Department of Bioengineering, Swanson School of Engineering, University of Pittsburgh, 3700 O'Hara Street, Pittsburgh, PA 15260, USA
| | - Yi-Nan Gong
- UPMC Hillman Cancer Center, University of Pittsburgh, 5115 Centre Ave, Pittsburgh, PA 15232, USA.
- Department of Immunology, University of Pittsburgh School of Medicine, 3420 Forbes Avenue, Pittsburgh, PA, 15260, USA
| | - Yu-Chih Chen
- UPMC Hillman Cancer Center, University of Pittsburgh, 5115 Centre Ave, Pittsburgh, PA 15232, USA.
- Department of Computational and Systems Biology, University of Pittsburgh, 3420 Forbes Avenue, Pittsburgh, PA 15260, USA
- Department of Bioengineering, Swanson School of Engineering, University of Pittsburgh, 3700 O'Hara Street, Pittsburgh, PA 15260, USA
- CMU-Pitt Ph.D. Program in Computational Biology, University of Pittsburgh, 3420 Forbes Avenue, Pittsburgh, PA 15260, USA
| |
Collapse
|
23
|
Wang W, Yang L, Sun H, Peng X, Yuan J, Zhong W, Chen J, He X, Ye L, Zeng Y, Gao Z, Li Y, Qu X. Cellular nucleus image-based smarter microscope system for single cell analysis. Biosens Bioelectron 2024; 250:116052. [PMID: 38266616 DOI: 10.1016/j.bios.2024.116052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2023] [Revised: 12/31/2023] [Accepted: 01/18/2024] [Indexed: 01/26/2024]
Abstract
Cell imaging technology is undoubtedly a powerful tool for studying single-cell heterogeneity due to its non-invasive and visual advantages. It covers microscope hardware, software, and image analysis techniques, which are hindered by low throughput owing to abundant hands-on time and expertise. Herein, a cellular nucleus image-based smarter microscope system for single-cell analysis is reported to achieve high-throughput analysis and high-content detection of cells. By combining the hardware of an automatic fluorescence microscope and multi-object recognition/acquisition software, we have achieved more advanced process automation with the assistance of Robotic Process Automation (RPA), which realizes a high-throughput collection of single-cell images. Automated acquisition of single-cell images has benefits beyond ease and throughout and can lead to uniform standard and higher quality images. We further constructed a single-cell image database-based convolutional neural network (Efficient Convolutional Neural Network, E-CNN) exceeding 20618 single-cell nucleus images. Computational analysis of large and complex data sets enhances the content and efficiency of single-cell analysis with the assistance of Artificial Intelligence (AI), which breaks through the super-resolution microscope's hardware limitation, such as specialized light sources with specific wavelengths, advanced optical components, and high-performance graphics cards. Our system can identify single-cell nucleus images that cannot be artificially distinguished with an accuracy of 95.3%. Overall, we build an ordinary microscope into a high-throughput analysis and high-content smarter microscope system, making it a candidate tool for Imaging cytology.
Collapse
Affiliation(s)
- Wentao Wang
- Key Laboratory of Sensing Technology and Biomedical Instruments of Guangdong Province, School of Biomedical Engineering, Sun Yat-Sen University, Shenzhen, Guangdong Province, 518017, China
| | - Lin Yang
- Key Laboratory of Sensing Technology and Biomedical Instruments of Guangdong Province, School of Biomedical Engineering, Sun Yat-Sen University, Shenzhen, Guangdong Province, 518017, China
| | - Hang Sun
- Key Laboratory of Sensing Technology and Biomedical Instruments of Guangdong Province, School of Biomedical Engineering, Sun Yat-Sen University, Shenzhen, Guangdong Province, 518017, China
| | - Xiaohong Peng
- YueYang Central Hospital, YueYang, Hunan Province, 414000, China
| | - Junjie Yuan
- Key Laboratory of Sensing Technology and Biomedical Instruments of Guangdong Province, School of Biomedical Engineering, Sun Yat-Sen University, Shenzhen, Guangdong Province, 518017, China
| | - Wenhao Zhong
- Key Laboratory of Sensing Technology and Biomedical Instruments of Guangdong Province, School of Biomedical Engineering, Sun Yat-Sen University, Shenzhen, Guangdong Province, 518017, China
| | - Jinqi Chen
- Key Laboratory of Sensing Technology and Biomedical Instruments of Guangdong Province, School of Biomedical Engineering, Sun Yat-Sen University, Shenzhen, Guangdong Province, 518017, China
| | - Xin He
- Key Laboratory of Sensing Technology and Biomedical Instruments of Guangdong Province, School of Biomedical Engineering, Sun Yat-Sen University, Shenzhen, Guangdong Province, 518017, China
| | - Lingzhi Ye
- Key Laboratory of Sensing Technology and Biomedical Instruments of Guangdong Province, School of Biomedical Engineering, Sun Yat-Sen University, Shenzhen, Guangdong Province, 518017, China
| | - Yi Zeng
- College of Chemistry and Chemical Engineering, Huanggang Normal University, Huanggang, 438000, China
| | - Zhifan Gao
- Key Laboratory of Sensing Technology and Biomedical Instruments of Guangdong Province, School of Biomedical Engineering, Sun Yat-Sen University, Shenzhen, Guangdong Province, 518017, China.
| | - Yunhui Li
- Department of Laboratory Medical Center, General Hospital of Northern Theater Command, No.83, Wenhua Road, Shenhe District, Shenyang, Liaoning Province, 110016, China.
| | - Xiangmeng Qu
- Key Laboratory of Sensing Technology and Biomedical Instruments of Guangdong Province, School of Biomedical Engineering, Sun Yat-Sen University, Shenzhen, Guangdong Province, 518017, China.
| |
Collapse
|
24
|
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
|
25
|
Shetab Boushehri S, Kornivetc A, Winter DJE, Kazeminia S, Essig K, Schmich F, Marr C. PXPermute reveals staining importance in multichannel imaging flow cytometry. CELL REPORTS METHODS 2024; 4:100715. [PMID: 38412831 PMCID: PMC10921034 DOI: 10.1016/j.crmeth.2024.100715] [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: 05/28/2023] [Revised: 11/08/2023] [Accepted: 01/29/2024] [Indexed: 02/29/2024]
Abstract
Imaging flow cytometry (IFC) allows rapid acquisition of numerous single-cell images per second, capturing information from multiple fluorescent channels. However, the traditional process of staining cells with fluorescently labeled conjugated antibodies for IFC analysis is time consuming, expensive, and potentially harmful to cell viability. To streamline experimental workflows and reduce costs, it is crucial to identify the most relevant channels for downstream analysis. In this study, we introduce PXPermute, a user-friendly and powerful method for assessing the significance of IFC channels, particularly for cell profiling. Our approach evaluates channel importance by permuting pixel values within each channel and analyzing the resulting impact on machine learning or deep learning models. Through rigorous evaluation of three multichannel IFC image datasets, we demonstrate PXPermute's potential in accurately identifying the most informative channels, aligning with established biological knowledge. PXPermute can assist biologists with systematic channel analysis, experimental design optimization, and biomarker identification.
Collapse
Affiliation(s)
- Sayedali Shetab Boushehri
- Institute of AI for Health, Helmholtz Zentrum München - German Research Center for Environmental Health, 85764 Neuherberg, Germany; Institute of Computational Biology, Helmholtz Zentrum MünchenMunich - Helmholtz Munich - German Research Center for Environmental Health, 85764 Neuherberg, Germany; Technical University of Munich, Department of Mathematics, 85748 Munich, Germany; Data & Analytics (D&A), Roche Pharma Research and Early Development (pRED), Roche Innovation Center Munich, 82377 Penzberg, Germany
| | - Aleksandra Kornivetc
- Institute of AI for Health, Helmholtz Zentrum München - German Research Center for Environmental Health, 85764 Neuherberg, Germany; Institute of Computational Biology, Helmholtz Zentrum MünchenMunich - Helmholtz Munich - German Research Center for Environmental Health, 85764 Neuherberg, Germany; University of Hamburg, Department of Informatics, 22527 Hamburg, Germany
| | - Domink J E Winter
- Institute of AI for Health, Helmholtz Zentrum München - German Research Center for Environmental Health, 85764 Neuherberg, Germany; Institute of Computational Biology, Helmholtz Zentrum MünchenMunich - Helmholtz Munich - German Research Center for Environmental Health, 85764 Neuherberg, Germany; Technical University of Munich, School of Life Sciences, 85354 Weihenstephan, Germany
| | - Salome Kazeminia
- Institute of AI for Health, Helmholtz Zentrum München - German Research Center for Environmental Health, 85764 Neuherberg, Germany; Technical University of Munich, Department of Mathematics, 85748 Munich, Germany
| | - Katharina Essig
- Large Molecule Research (LMR), Roche Pharma Research and Early Development (pRED), Roche Innovation Center Munich, 82377 Penzberg, Germany
| | - Fabian Schmich
- Data & Analytics (D&A), Roche Pharma Research and Early Development (pRED), Roche Innovation Center Munich, 82377 Penzberg, Germany
| | - Carsten Marr
- Institute of AI for Health, Helmholtz Zentrum München - German Research Center for Environmental Health, 85764 Neuherberg, Germany.
| |
Collapse
|
26
|
Ciaparrone G, Pirone D, Fiore P, Xin L, Xiao W, Li X, Bardozzo F, Bianco V, Miccio L, Pan F, Memmolo P, Tagliaferri R, Ferraro P. Label-free cell classification in holographic flow cytometry through an unbiased learning strategy. LAB ON A CHIP 2024; 24:924-932. [PMID: 38264771 DOI: 10.1039/d3lc00385j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/25/2024]
Abstract
Nowadays, label-free imaging flow cytometry at the single-cell level is considered the stepforward lab-on-a-chip technology to address challenges in clinical diagnostics, biology, life sciences and healthcare. In this framework, digital holography in microscopy promises to be a powerful imaging modality thanks to its multi-refocusing and label-free quantitative phase imaging capabilities, along with the encoding of the highest information content within the imaged samples. Moreover, the recent achievements of new data analysis tools for cell classification based on deep/machine learning, combined with holographic imaging, are urging these systems toward the effective implementation of point of care devices. However, the generalization capabilities of learning-based models may be limited from biases caused by data obtained from other holographic imaging settings and/or different processing approaches. In this paper, we propose a combination of a Mask R-CNN to detect the cells, a convolutional auto-encoder, used to the image feature extraction and operating on unlabelled data, thus overcoming the bias due to data coming from different experimental settings, and a feedforward neural network for single cell classification, that operates on the above extracted features. We demonstrate the proposed approach in the challenging classification task related to the identification of drug-resistant endometrial cancer cells.
Collapse
Affiliation(s)
- Gioele Ciaparrone
- Neurone Lab, Department of Management and Innovation Systems (DISA-MIS), University of Salerno, Fisciano, Italy.
| | - Daniele Pirone
- CNR - Institute of Applied Sciences and Intelligent Systems "Eduardo Caianiello", Pozzuoli, Italy.
| | - Pierpaolo Fiore
- Neurone Lab, Department of Management and Innovation Systems (DISA-MIS), University of Salerno, Fisciano, Italy.
| | - Lu Xin
- Key Laboratory of Precision Opto-Mechatronics Technology of Ministry of Education, School of Instrumentation Science & Optoelectronics Engineering, Beihang University, 100191 Beijing, China.
| | - Wen Xiao
- Key Laboratory of Precision Opto-Mechatronics Technology of Ministry of Education, School of Instrumentation Science & Optoelectronics Engineering, Beihang University, 100191 Beijing, China.
| | - Xiaoping Li
- Department of Obstetrics and Gynecology, Peking University People's Hospital, Beijing 100044, China
| | - Francesco Bardozzo
- Neurone Lab, Department of Management and Innovation Systems (DISA-MIS), University of Salerno, Fisciano, Italy.
- CNR - Institute of Applied Sciences and Intelligent Systems "Eduardo Caianiello", Pozzuoli, Italy.
| | - Vittorio Bianco
- CNR - Institute of Applied Sciences and Intelligent Systems "Eduardo Caianiello", Pozzuoli, Italy.
| | - Lisa Miccio
- CNR - Institute of Applied Sciences and Intelligent Systems "Eduardo Caianiello", Pozzuoli, Italy.
| | - Feng Pan
- Key Laboratory of Precision Opto-Mechatronics Technology of Ministry of Education, School of Instrumentation Science & Optoelectronics Engineering, Beihang University, 100191 Beijing, China.
| | - Pasquale Memmolo
- CNR - Institute of Applied Sciences and Intelligent Systems "Eduardo Caianiello", Pozzuoli, Italy.
| | - Roberto Tagliaferri
- Neurone Lab, Department of Management and Innovation Systems (DISA-MIS), University of Salerno, Fisciano, Italy.
- CNR - Institute of Applied Sciences and Intelligent Systems "Eduardo Caianiello", Pozzuoli, Italy.
| | - Pietro Ferraro
- CNR - Institute of Applied Sciences and Intelligent Systems "Eduardo Caianiello", Pozzuoli, Italy.
| |
Collapse
|
27
|
Marchese A, Ricci P, Saggau P, Duocastella M. Scan-less microscopy based on acousto-optic encoded illumination. NANOPHOTONICS 2024; 13:63-73. [PMID: 38235070 PMCID: PMC10790963 DOI: 10.1515/nanoph-2023-0616] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Accepted: 12/04/2023] [Indexed: 01/19/2024]
Abstract
Several optical microscopy methods are now available for characterizing scientific and industrial processes at sub-micron resolution. However, they are often ill-suited for imaging rapid events. Limited by the trade-off between camera frame-rate and sensitivity, or the need for mechanical scanning, current microscopes are optimized for imaging at hundreds of frames-per-second (fps), well-below what is needed in processes such as neuronal signaling or moving parts in manufacturing lines. Here, we present a scan-less technology that allows sub-micrometric imaging at thousands of fps. It is based on combining a single-pixel camera with parallelized encoded illumination. We use two acousto-optic deflectors (AODs) placed in a Mach-Zehnder interferometer and drive them simultaneously with multiple and unique acoustic frequencies. As a result, orthogonal light stripes are obtained that interfere with the sample plane, forming a two-dimensional array of flickering spots - each with its modulation frequency. The light from the sample is collected with a single photodiode that, after spectrum analysis, allows for image reconstruction at speeds only limited by the AOD's bandwidth and laser power. We describe the working principle of our approach, characterize its imaging performance as a function of the number of pixels - up to 400 × 400 - and characterize dynamic events at 5000 fps.
Collapse
Affiliation(s)
- Andrea Marchese
- Department of Applied Physics, Universitat de Barcelona, Martí i Franquès, 1, 08028Barcelona, Spain
| | - Pietro Ricci
- Department of Applied Physics, Universitat de Barcelona, Martí i Franquès, 1, 08028Barcelona, Spain
| | - Peter Saggau
- Department of Neuroscience, Baylor College of Medicine, One Baylor Plaza, S640, 77030Houston, TX, USA
| | - Martí Duocastella
- Department of Applied Physics, Universitat de Barcelona, Martí i Franquès, 1, 08028Barcelona, Spain
| |
Collapse
|
28
|
Tang M, Zhang M, Fu Y, Chen L, Li D, Zhang H, Yang Z, Wang C, Xiu P, Wilksch JJ, Luo Y, Han J, Yang H, Wang H. Terahertz label-free detection of nicotine-induced neural cell changes and the underlying mechanisms. Biosens Bioelectron 2023; 241:115697. [PMID: 37751650 DOI: 10.1016/j.bios.2023.115697] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Revised: 08/28/2023] [Accepted: 09/16/2023] [Indexed: 09/28/2023]
Abstract
Nicotine exposure can lead to neurological impairments and brain tumors, and a label-free and nondestructive detection technique is urgently required by the scientific community to assess the effects of nicotine on neural cells. Herein, a terahertz (THz) time-domain attenuated total reflection (TD-ATR) spectroscopy approach is reported, by which the effects of nicotine on normal and cancerous neural cells, i.e., HEB and U87 cells, are successfully investigated in a label/stain-free and nondestructive manner. The obtained THz absorption coefficients of HEB cells exposed to low-dose nicotine and high-dose nicotine are smaller and larger, respectively, than the untreated cells. In contrast, the THz absorption coefficients of U87 cells treated by nicotine are always smaller than the untreated cells. The THz absorption coefficients can be well related to the proliferation properties (cell number and compositional changes) and morphological changes of neural cells, by which different types of neural cells are differentiated and the viabilities of neural cells treated by nicotine are reliably assessed. Collectively, this work sheds new insights on the effects of nicotine on neural cells, and provides a useful tool (THz TD-ATR spectroscopy) for the study of chemical-cell interactions.
Collapse
Affiliation(s)
- Mingjie Tang
- Research Center of Super-Resolution Optics & Chongqing Engineering Research Center of High-Resolution and Three-Dimensional Dynamic Imaging Technology, Chongqing Institute of Green and Intelligent Technology, Chinese Academy of Sciences, Chongqing, 400714, China; Chongqing School, University of Chinese Academy of Sciences, Chongqing, 400714, China
| | - Mingkun Zhang
- Research Center of Super-Resolution Optics & Chongqing Engineering Research Center of High-Resolution and Three-Dimensional Dynamic Imaging Technology, Chongqing Institute of Green and Intelligent Technology, Chinese Academy of Sciences, Chongqing, 400714, China; Chongqing School, University of Chinese Academy of Sciences, Chongqing, 400714, China
| | - Ying Fu
- Research Center of Super-Resolution Optics & Chongqing Engineering Research Center of High-Resolution and Three-Dimensional Dynamic Imaging Technology, Chongqing Institute of Green and Intelligent Technology, Chinese Academy of Sciences, Chongqing, 400714, China; Chongqing School, University of Chinese Academy of Sciences, Chongqing, 400714, China
| | - Ligang Chen
- Research Center of Super-Resolution Optics & Chongqing Engineering Research Center of High-Resolution and Three-Dimensional Dynamic Imaging Technology, Chongqing Institute of Green and Intelligent Technology, Chinese Academy of Sciences, Chongqing, 400714, China; Chongqing School, University of Chinese Academy of Sciences, Chongqing, 400714, China
| | - Dandan Li
- Research Center of Super-Resolution Optics & Chongqing Engineering Research Center of High-Resolution and Three-Dimensional Dynamic Imaging Technology, Chongqing Institute of Green and Intelligent Technology, Chinese Academy of Sciences, Chongqing, 400714, China; Chongqing School, University of Chinese Academy of Sciences, Chongqing, 400714, China
| | - Hua Zhang
- Research Center of Super-Resolution Optics & Chongqing Engineering Research Center of High-Resolution and Three-Dimensional Dynamic Imaging Technology, Chongqing Institute of Green and Intelligent Technology, Chinese Academy of Sciences, Chongqing, 400714, China; Chongqing School, University of Chinese Academy of Sciences, Chongqing, 400714, China
| | - Zhongbo Yang
- Research Center of Super-Resolution Optics & Chongqing Engineering Research Center of High-Resolution and Three-Dimensional Dynamic Imaging Technology, Chongqing Institute of Green and Intelligent Technology, Chinese Academy of Sciences, Chongqing, 400714, China; Chongqing School, University of Chinese Academy of Sciences, Chongqing, 400714, China
| | - Chunlei Wang
- Department of Chemistry, Shanghai University, Shanghai, 200444, China
| | - Peng Xiu
- Department of Engineering Mechanics, Zhejiang University, Hangzhou, 310027, China
| | - Jonathan J Wilksch
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, Victoria, Australia
| | - Yang Luo
- Center of Smart Laboratory and Molecular Medicine, School of Medicine, Chongqing University, Chongqing, 400044, China
| | - Jiaguang Han
- Center for Terahertz Waves and College of Precision Instrument and Optoelectronics Engineering, Tianjin University, Tianjin, 300072, China
| | - Haijun Yang
- Shanghai Synchrotron Radiation Facility, Shanghai Advanced Research Institute, Chinese Academy of Sciences, Shanghai, 201204, China.
| | - Huabin Wang
- Research Center of Super-Resolution Optics & Chongqing Engineering Research Center of High-Resolution and Three-Dimensional Dynamic Imaging Technology, Chongqing Institute of Green and Intelligent Technology, Chinese Academy of Sciences, Chongqing, 400714, China; Chongqing School, University of Chinese Academy of Sciences, Chongqing, 400714, China.
| |
Collapse
|
29
|
Timonen VA, Kerkelä E, Impola U, Penna L, Partanen J, Kilpivaara O, Arvas M, Pitkänen E. DeepIFC: Virtual fluorescent labeling of blood cells in imaging flow cytometry data with deep learning. Cytometry A 2023; 103:807-817. [PMID: 37276178 DOI: 10.1002/cyto.a.24770] [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: 09/30/2022] [Revised: 05/16/2023] [Accepted: 06/02/2023] [Indexed: 06/07/2023]
Abstract
Imaging flow cytometry (IFC) combines flow cytometry with microscopy, allowing rapid characterization of cellular and molecular properties via high-throughput single-cell fluorescent imaging. However, fluorescent labeling is costly and time-consuming. We present a computational method called DeepIFC based on the Inception U-Net neural network architecture, able to generate fluorescent marker images and learn morphological features from IFC brightfield and darkfield images. Furthermore, the DeepIFC workflow identifies cell types from the generated fluorescent images and visualizes the single-cell features generated in a 2D space. We demonstrate that rarer cell types are predicted well when a balanced data set is used to train the model, and the model is able to recognize red blood cells not seen during model training as a distinct entity. In summary, DeepIFC allows accurate cell reconstruction, typing and recognition of unseen cell types from brightfield and darkfield images via virtual fluorescent labeling.
Collapse
Affiliation(s)
- Veera A Timonen
- Institute for Molecular Medicine Finland (FIMM), Helsinki Institute of Life Science (HiLIFE), University of Helsinki, Helsinki, Finland
- Applied Tumor Genomics Research Program, Research Programs Unit, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Erja Kerkelä
- Advanced Cell Therapy Centre, Finnish Red Cross Blood Service, Vantaa, Finland
| | - Ulla Impola
- Research and Development, Finnish Red Cross Blood Service, Helsinki, Finland
| | - Leena Penna
- Research and Development, Finnish Red Cross Blood Service, Helsinki, Finland
| | - Jukka Partanen
- Research and Development, Finnish Red Cross Blood Service, Helsinki, Finland
| | - Outi Kilpivaara
- Applied Tumor Genomics Research Program, Research Programs Unit, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Department of Medical and Clinical Genetics, Medicum, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- HUSLAB Laboratory of Genetics, HUS Diagnostic Center, Helsinki University Hospital, Helsinki, Finland
- iCAN Digital Precision Cancer Medicine Flagship, Helsinki, Finland
| | - Mikko Arvas
- Research and Development, Finnish Red Cross Blood Service, Helsinki, Finland
| | - Esa Pitkänen
- Institute for Molecular Medicine Finland (FIMM), Helsinki Institute of Life Science (HiLIFE), University of Helsinki, Helsinki, Finland
- Applied Tumor Genomics Research Program, Research Programs Unit, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- iCAN Digital Precision Cancer Medicine Flagship, Helsinki, Finland
| |
Collapse
|
30
|
Cioffi G, Dannhauser D, Rossi D, Netti PA, Causa F. Unknown cell class distinction via neural network based scattering snapshot recognition. BIOMEDICAL OPTICS EXPRESS 2023; 14:5060-5074. [PMID: 37854558 PMCID: PMC10581789 DOI: 10.1364/boe.492028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 07/25/2023] [Accepted: 07/31/2023] [Indexed: 10/20/2023]
Abstract
Neural network-based image classification is widely used in life science applications. However, it is essential to extrapolate a correct classification method for unknown images, where no prior knowledge can be utilised. Under a closed set assumption, unknown images will be inevitably misclassified, but this can be genuinely overcome choosing an open-set classification approach, which first generates an in-distribution of identified images to successively discriminate out-of-distribution images. The testing of such image classification for single cell applications in life science scenarios has yet to be done but could broaden our expertise in quantifying the influence of prediction uncertainty in deep learning. In this framework, we implemented the open-set concept on scattering snapshots of living cells to distinguish between unknown and known cell classes, targeting four different known monoblast cell classes and a single tumoral unknown monoblast cell line. We also investigated the influence on experimental sample errors and optimised neural network hyperparameters to obtain a high unknown cell class detection accuracy. We discovered that our open-set approach exhibits robustness against sample noise, a crucial aspect for its application in life science. Moreover, the presented open-set based neural network reveals measurement uncertainty out of the cell prediction, which can be applied to a wide range of single cell classifications.
Collapse
Affiliation(s)
- Gaia Cioffi
- Interdisciplinary Research Centre on Biomaterials (CRIB) and Dipartimento di Ingegneria Chimica, dei Materiali e della Produzione Industriale, Università degli Studi di Napoli “Federico II”, Piazzale Tecchio 80, 80125 Naples, Italy
| | - David Dannhauser
- Interdisciplinary Research Centre on Biomaterials (CRIB) and Dipartimento di Ingegneria Chimica, dei Materiali e della Produzione Industriale, Università degli Studi di Napoli “Federico II”, Piazzale Tecchio 80, 80125 Naples, Italy
| | - Domenico Rossi
- Center for Advanced Biomaterials for Healthcare@CRIB, Istituto Italiano di Tecnologia, Largo Barsanti e Matteucci 53, 80125 Naples, Italy
| | - Paolo A. Netti
- Interdisciplinary Research Centre on Biomaterials (CRIB) and Dipartimento di Ingegneria Chimica, dei Materiali e della Produzione Industriale, Università degli Studi di Napoli “Federico II”, Piazzale Tecchio 80, 80125 Naples, Italy
- Center for Advanced Biomaterials for Healthcare@CRIB, Istituto Italiano di Tecnologia, Largo Barsanti e Matteucci 53, 80125 Naples, Italy
| | - Filippo Causa
- Interdisciplinary Research Centre on Biomaterials (CRIB) and Dipartimento di Ingegneria Chimica, dei Materiali e della Produzione Industriale, Università degli Studi di Napoli “Federico II”, Piazzale Tecchio 80, 80125 Naples, Italy
| |
Collapse
|
31
|
Hayashi M, Ohnuki S, Tsai Y, Kondo N, Zhou Y, Zhang H, Ishii NT, Ding T, Herbig M, Isozaki A, Ohya Y, Goda K. Is AI essential? Examining the need for deep learning in image-activated sorting of Saccharomyces cerevisiae. LAB ON A CHIP 2023; 23:4232-4244. [PMID: 37650583 DOI: 10.1039/d3lc00556a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/01/2023]
Abstract
Artificial intelligence (AI) has become a focal point across a multitude of societal sectors, with science not being an exception. Particularly in the life sciences, imaging flow cytometry has increasingly integrated AI for automated management and categorization of extensive cell image data. However, the necessity of AI over traditional classification methods when extending imaging flow cytometry to include cell sorting remains uncertain, primarily due to the time constraints between image acquisition and sorting actuation. AI-enabled image-activated cell sorting (IACS) methods remain substantially limited, even as recent advancements in IACS have found success while largely relying on traditional feature gating strategies. Here we assess the necessity of AI for image classification in IACS by contrasting the performance of feature gating, classical machine learning (ML), and deep learning (DL) with convolutional neural networks (CNNs) in the differentiation of Saccharomyces cerevisiae mutant images. We show that classical ML could only yield a 2.8-fold enhancement in target enrichment capability, albeit at the cost of a 13.7-fold increase in processing time. Conversely, a CNN could offer an 11.0-fold improvement in enrichment capability at an 11.5-fold increase in processing time. We further executed IACS on mixed mutant populations and quantified target strain enrichment via downstream DNA sequencing to substantiate the applicability of DL for the proposed study. Our findings validate the feasibility and value of employing DL in IACS for morphology-based genetic screening of S. cerevisiae, encouraging its incorporation in future advancements of similar technologies.
Collapse
Affiliation(s)
- Mika Hayashi
- Department of Chemistry, Graduate School of Science, The University of Tokyo, Tokyo 113-0033, Japan.
| | - Shinsuke Ohnuki
- Department of Integrated Biosciences, Graduate School of Frontier Sciences, The University of Tokyo, Chiba 277-8562, Japan.
| | - Yating Tsai
- Department of Integrated Biosciences, Graduate School of Frontier Sciences, The University of Tokyo, Chiba 277-8562, Japan.
| | - Naoko Kondo
- Department of Integrated Biosciences, Graduate School of Frontier Sciences, The University of Tokyo, Chiba 277-8562, Japan.
| | - Yuqi Zhou
- Department of Chemistry, Graduate School of Science, The University of Tokyo, Tokyo 113-0033, Japan.
| | - Hongqian Zhang
- Department of Chemistry, Graduate School of Science, The University of Tokyo, Tokyo 113-0033, Japan.
| | - Natsumi Tiffany Ishii
- Department of Chemistry, Graduate School of Science, The University of Tokyo, Tokyo 113-0033, Japan.
| | - Tianben Ding
- Department of Chemistry, Graduate School of Science, The University of Tokyo, Tokyo 113-0033, Japan.
| | - Maik Herbig
- Department of Chemistry, Graduate School of Science, The University of Tokyo, Tokyo 113-0033, Japan.
| | - Akihiro Isozaki
- Department of Chemistry, Graduate School of Science, The University of Tokyo, Tokyo 113-0033, Japan.
- Department of Mechanical Engineering, College of Science and Engineering, Ritsumeikan University, Shiga 525-8577, Japan.
| | - Yoshikazu Ohya
- Department of Integrated Biosciences, Graduate School of Frontier Sciences, The University of Tokyo, Chiba 277-8562, Japan.
- Collaborative Research Institute for Innovative Microbiology, The University of Tokyo, Tokyo 113-8654, Japan
| | - Keisuke Goda
- Department of Chemistry, Graduate School of Science, The University of Tokyo, Tokyo 113-0033, Japan.
- Department of Bioengineering, University of California, Los Angeles, California 90095, USA
- Institute of Technological Sciences, Wuhan University, Wuhan 430072, China
- CYBO, Tokyo 135-0064, Japan
| |
Collapse
|
32
|
Rani S, Sahoo RK, Mahale A, Panchal K, Chaurasiya A, Kulkarni O, Kuche K, Jain S, Nakhate KT, Ajazuddin, Gupta U. Sialic Acid Engineered Prodrug Nanoparticles for Codelivery of Bortezomib and Selenium in Tumor Bearing Mice. Bioconjug Chem 2023; 34:1528-1552. [PMID: 37603704 DOI: 10.1021/acs.bioconjchem.3c00210] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/23/2023]
Abstract
Most cancer patients rarely benefit from monodrug therapy because of both cancer complexity and tumor environment. One of the main reasons for this failure is insufficient accumulation of the optimal dose at the tumorous site. Our investigation implies a promising strategy to engineer prodrug nanoparticles (NPs) of bortezomib (BTZ) and selenium (Se) using sialic acid (SAL) as a ligand to improve breast cancer therapy. BTZ was conjugated with SAL and HPMA (N-2-hydroxypropyl methacrylamide) to prepare a prodrug conjugate; BTZ-SAL-HPMA (BSAL-HP) and then fabricated into prodrug NPs with Se (Se_BSAL-HP prodrug NPs). The self-assembly of prodrug NPs functionalized with Se showed size (204.13 ± 0.02 nm) and zeta potential (-31.0 ± 0.11 mV) in dynamic light scattering (DLS) experiments and spherical shape in TEM and SEM analysis. Good stability and low pH drug release profile were characterized by Se_BSAL-HP prodrug NPs. The tumor-selective boronate-ester-based prodrug NPs of BTZ in combination with Se endowed a synergistic effect against cancer cells. Compared to prodrug conjugate, Se_BSAL-HP prodrug NPs exhibited higher cell cytotoxicity and enhanced cellular internalization with significant changes in mitochondria membrane potential (MMP). Elevated apoptosis was observed in the (G2/M) phase of the cell cycle for Se_BSAL-HP prodrug NPs (2.7-fold) higher than BTZ. In vivo studies were performed on Sprague-Dawley rats and resulted in positive trends. The increased therapeutic activity of Se_BSAL-HP prodrug NPs inhibited primary tumor growth and showed 43.05 fold decrease in tumor volume than the control in 4T1 tumor bearing mice. The surprising and remarkable outcomes for Se_BSAL-HP prodrug NPs were probably due to the ROS triggering effect of boronate ester and selenium given together.
Collapse
Affiliation(s)
- Sarita Rani
- Nanopolymeric Drug Delivery Lab, Department of Pharmacy, School of Chemical Sciences and Pharmacy, Central University of Rajasthan, Bandarsindri, Ajmer, Rajasthan 305817, India
| | - Rakesh K Sahoo
- Nanopolymeric Drug Delivery Lab, Department of Pharmacy, School of Chemical Sciences and Pharmacy, Central University of Rajasthan, Bandarsindri, Ajmer, Rajasthan 305817, India
| | - Ashutosh Mahale
- Department of Pharmacy, Birla Institute of Technology & Science, Pilani, Hyderabad Campus, Jawahar Nagar, Kapra Mandal Medchal District, Telangana 500078, India
| | - Kanan Panchal
- Department of Pharmacy, Birla Institute of Technology & Science, Pilani, Hyderabad Campus, Jawahar Nagar, Kapra Mandal Medchal District, Telangana 500078, India
| | - Akash Chaurasiya
- Department of Pharmacy, Birla Institute of Technology & Science, Pilani, Hyderabad Campus, Jawahar Nagar, Kapra Mandal Medchal District, Telangana 500078, India
| | - Onkar Kulkarni
- Department of Pharmacy, Birla Institute of Technology & Science, Pilani, Hyderabad Campus, Jawahar Nagar, Kapra Mandal Medchal District, Telangana 500078, India
| | - Kaushik Kuche
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), SAS Nagar Campus, Sector-67, Punjab 160062, India
| | - Sanyog Jain
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), SAS Nagar Campus, Sector-67, Punjab 160062, India
| | - Kartik T Nakhate
- Department of Pharmacology, Shri Vile Parle Kelavani Mandal's Institute of Pharmacy, Dhule, Maharashtra 424001, India
| | - Ajazuddin
- Rungta College of Pharmaceutical Sciences and Research, Kohka-Kurud Road, Bhilai, Chhattisgarh 490024, India
| | - Umesh Gupta
- Nanopolymeric Drug Delivery Lab, Department of Pharmacy, School of Chemical Sciences and Pharmacy, Central University of Rajasthan, Bandarsindri, Ajmer, Rajasthan 305817, India
| |
Collapse
|
33
|
Haddad M, Frickenstein A, Wilhelm S. High-Throughput Single-Cell Analysis of Nanoparticle-Cell Interactions. Trends Analyt Chem 2023; 166:117172. [PMID: 37520860 PMCID: PMC10373476 DOI: 10.1016/j.trac.2023.117172] [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] [Indexed: 08/01/2023]
Abstract
Understanding nanoparticle-cell interactions at single-nanoparticle and single-cell resolutions is crucial to improving the design of next-generation nanoparticles for safer, more effective, and more efficient applications in nanomedicine. This review focuses on recent advances in the continuous high-throughput analysis of nanoparticle-cell interactions at the single-cell level. We highlight and discuss the current trends in continual flow high-throughput methods for analyzing single cells, such as advanced flow cytometry techniques and inductively coupled plasma mass spectrometry methods, as well as their intersection in the form of mass cytometry. This review further discusses the challenges and opportunities with current single-cell analysis approaches and provides proposed directions for innovation in the high-throughput analysis of nanoparticle-cell interactions.
Collapse
Affiliation(s)
- Majood Haddad
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, Oklahoma, 73019, USA
| | - Alex Frickenstein
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, Oklahoma, 73019, USA
| | - Stefan Wilhelm
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, Oklahoma, 73019, USA
- Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, 73104, USA
- Institute for Biomedical Engineering, Science, and Technology (IBEST), University of Oklahoma, Norman, Oklahoma, 73019, USA
| |
Collapse
|
34
|
Julian T, Tang T, Hosokawa Y, Yalikun Y. Machine learning implementation strategy in imaging and impedance flow cytometry. BIOMICROFLUIDICS 2023; 17:051506. [PMID: 37900052 PMCID: PMC10613093 DOI: 10.1063/5.0166595] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Accepted: 10/06/2023] [Indexed: 10/31/2023]
Abstract
Imaging and impedance flow cytometry is a label-free technique that has shown promise as a potential replacement for standard flow cytometry. This is due to its ability to provide rich information and archive high-throughput analysis. Recently, significant efforts have been made to leverage machine learning for processing the abundant data generated by those techniques, enabling rapid and accurate analysis. Harnessing the power of machine learning, imaging and impedance flow cytometry has demonstrated its capability to address various complex phenotyping scenarios. Herein, we present a comprehensive overview of the detailed strategies for implementing machine learning in imaging and impedance flow cytometry. We initiate the discussion by outlining the commonly employed setup to acquire the data (i.e., image or signal) from the cell. Subsequently, we delve into the necessary processes for extracting features from the acquired image or signal data. Finally, we discuss how these features can be utilized for cell phenotyping through the application of machine learning algorithms. Furthermore, we discuss the existing challenges and provide insights for future perspectives of intelligent imaging and impedance flow cytometry.
Collapse
Affiliation(s)
- Trisna Julian
- Division of Materials Science, Nara Institute of Science and Technology, 8916-5 Takayamacho, Ikoma, Nara 630-0192, Japan
| | - Tao Tang
- Department of Biomedical Engineering, National University of Singapore, 4 Engineering Drive 3, Singapore 117583, Singapore
| | - Yoichiroh Hosokawa
- Division of Materials Science, Nara Institute of Science and Technology, 8916-5 Takayamacho, Ikoma, Nara 630-0192, Japan
| | | |
Collapse
|
35
|
Raj M K, Priyadarshani J, Karan P, Bandyopadhyay S, Bhattacharya S, Chakraborty S. Bio-inspired microfluidics: A review. BIOMICROFLUIDICS 2023; 17:051503. [PMID: 37781135 PMCID: PMC10539033 DOI: 10.1063/5.0161809] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Accepted: 09/01/2023] [Indexed: 10/03/2023]
Abstract
Biomicrofluidics, a subdomain of microfluidics, has been inspired by several ideas from nature. However, while the basic inspiration for the same may be drawn from the living world, the translation of all relevant essential functionalities to an artificially engineered framework does not remain trivial. Here, we review the recent progress in bio-inspired microfluidic systems via harnessing the integration of experimental and simulation tools delving into the interface of engineering and biology. Development of "on-chip" technologies as well as their multifarious applications is subsequently discussed, accompanying the relevant advancements in materials and fabrication technology. Pointers toward new directions in research, including an amalgamated fusion of data-driven modeling (such as artificial intelligence and machine learning) and physics-based paradigm, to come up with a human physiological replica on a synthetic bio-chip with due accounting of personalized features, are suggested. These are likely to facilitate physiologically replicating disease modeling on an artificially engineered biochip as well as advance drug development and screening in an expedited route with the minimization of animal and human trials.
Collapse
Affiliation(s)
- Kiran Raj M
- Department of Applied Mechanics and Biomedical Engineering, Indian Institute of Technology Madras, Chennai, Tamil Nadu 600036, India
| | - Jyotsana Priyadarshani
- Department of Mechanical Engineering, Biomechanics Section (BMe), KU Leuven, Celestijnenlaan 300, 3001 Louvain, Belgium
| | - Pratyaksh Karan
- Géosciences Rennes Univ Rennes, CNRS, Géosciences Rennes, UMR 6118, 35000 Rennes, France
| | - Saumyadwip Bandyopadhyay
- Advanced Technology Development Centre, Indian Institute of Technology Kharagpur, Kharagpur, West Bengal 721302, India
| | - Soumya Bhattacharya
- Achira Labs Private Limited, 66b, 13th Cross Rd., Dollar Layout, 3–Phase, JP Nagar, Bangalore, Karnataka 560078, India
| | - Suman Chakraborty
- Department of Mechanical Engineering, Indian Institute of Technology Kharagpur, Kharagpur, West Bengal 721302, India
| |
Collapse
|
36
|
Pang K, Dong S, Zhu Y, Zhu X, Zhou Q, Gu B, Jin W, Zhang R, Fu Y, Yu B, Sun D, Duanmu Z, Wei X. Advanced flow cytometry for biomedical applications. JOURNAL OF BIOPHOTONICS 2023; 16:e202300135. [PMID: 37263969 DOI: 10.1002/jbio.202300135] [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: 04/24/2023] [Revised: 05/29/2023] [Accepted: 05/30/2023] [Indexed: 06/03/2023]
Abstract
Flow cytometry (FC) is a versatile tool with excellent capabilities to detect and measure multiple characteristics of a population of cells or particles. Notable advancements in in vivo photoacoustic FC, coherent Raman FC, microfluidic FC, and so on, have been achieved in the last two decades, which endows FC with new functions and expands its applications in basic research and clinical practice. Advanced FC broadens the tools available to researchers to conduct research involving cancer detection, microbiology (COVID-19, HIV, bacteria, etc.), and nucleic acid analysis. This review presents an overall picture of advanced flow cytometers and provides not only a clear understanding of their mechanisms but also new insights into their practical applications. We identify the latest trends in this area and aim to raise awareness of advanced techniques of FC. We hope this review expands the applications of FC and accelerates its clinical translation.
Collapse
Affiliation(s)
- Kai Pang
- School of Instrument Science and Opto-Electronics Engineering of Beijing Information Science & Technology University, Beijing, China
| | - Sihan Dong
- Institute of Medical Technology, Peking University Health Science Center, Beijing, China
| | - Yuxi Zhu
- School of Instrument Science and Opto-Electronics Engineering of Beijing Information Science & Technology University, Beijing, China
| | - Xi Zhu
- Med-X Research Institute and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Quanyu Zhou
- Med-X Research Institute and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Bobo Gu
- Med-X Research Institute and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Wei Jin
- International Cancer Institute, Peking University, Beijing, China
| | - Rui Zhang
- Institute of Medical Technology, Peking University Health Science Center, Beijing, China
| | - Yuting Fu
- Institute of Medical Technology, Peking University Health Science Center, Beijing, China
| | - Bingchen Yu
- School of Instrument Science and Opto-Electronics Engineering of Beijing Information Science & Technology University, Beijing, China
| | - Da Sun
- School of Instrument Science and Opto-Electronics Engineering of Beijing Information Science & Technology University, Beijing, China
| | - Zheng Duanmu
- School of Instrument Science and Opto-Electronics Engineering of Beijing Information Science & Technology University, Beijing, China
| | - Xunbin Wei
- International Cancer Institute, Peking University, Beijing, China
| |
Collapse
|
37
|
Isiksacan Z, D’Alessandro A, Wolf SM, McKenna DH, Tessier SN, Kucukal E, Gokaltun AA, William N, Sandlin RD, Bischof J, Mohandas N, Busch MP, Elbuken C, Gurkan UA, Toner M, Acker JP, Yarmush ML, Usta OB. Assessment of stored red blood cells through lab-on-a-chip technologies for precision transfusion medicine. Proc Natl Acad Sci U S A 2023; 120:e2115616120. [PMID: 37494421 PMCID: PMC10410732 DOI: 10.1073/pnas.2115616120] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/28/2023] Open
Abstract
Transfusion of red blood cells (RBCs) is one of the most valuable and widespread treatments in modern medicine. Lifesaving RBC transfusions are facilitated by the cold storage of RBC units in blood banks worldwide. Currently, RBC storage and subsequent transfusion practices are performed using simplistic workflows. More specifically, most blood banks follow the "first-in-first-out" principle to avoid wastage, whereas most healthcare providers prefer the "last-in-first-out" approach simply favoring chronologically younger RBCs. Neither approach addresses recent advances through -omics showing that stored RBC quality is highly variable depending on donor-, time-, and processing-specific factors. Thus, it is time to rethink our workflows in transfusion medicine taking advantage of novel technologies to perform RBC quality assessment. We imagine a future where lab-on-a-chip technologies utilize novel predictive markers of RBC quality identified by -omics and machine learning to usher in a new era of safer and precise transfusion medicine.
Collapse
Affiliation(s)
- Ziya Isiksacan
- Center for Engineering in Medicine and Surgery, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA02114
- Shriners Children’s, Boston, MA02114
| | - Angelo D’Alessandro
- Department of Biochemistry and Molecular Genetics, University of Colorado Denver – Anschutz Medical Campus, Aurora, CO80045
| | - Susan M. Wolf
- Law School, Medical School, Consortium on Law and Values in Health, Environment & the Life Sciences, University of Minnesota, Minneapolis, MN55455
| | - David H. McKenna
- Division of Transfusion Medicine, Department of Laboratory Medicine and Pathology, University of Minnesota Medical School, Minneapolis, MN55455
| | - Shannon N. Tessier
- Center for Engineering in Medicine and Surgery, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA02114
- Shriners Children’s, Boston, MA02114
| | | | - A. Aslihan Gokaltun
- Center for Engineering in Medicine and Surgery, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA02114
- Shriners Children’s, Boston, MA02114
- Department of Chemical Engineering, Hacettepe University, Ankara06532, Turkey
| | - Nishaka William
- Laboratory Medicine and Pathology, University of Alberta, Edmonton, ABT6G 2R8, Canada
| | - Rebecca D. Sandlin
- Center for Engineering in Medicine and Surgery, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA02114
| | - John Bischof
- Department of Mechanical Engineering, University of Minnesota, Minneapolis, MN55455
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, MN55455
| | | | - Michael P. Busch
- Vitalant Research Institute, San Francisco, CA94105
- Department of Laboratory Medicine, University of California San Francisco, San Francisco, CA94105
| | - Caglar Elbuken
- Institute of Materials Science and Nanotechnology, National Nanotechnology Research Center, Bilkent University, Ankara06800, Turkey
- Faculty of Biochemistry and Molecular Medicine, Faculty of Medicine, University of Oulu, 90014Oulu, Finland
- Valtion Teknillinen Tutkimuskeskus Technical Research Centre of Finland Ltd., 90570Oulu, Finland
| | - Umut A. Gurkan
- Department of Mechanical and Aerospace Engineering, Case Western Reserve University, Cleveland, OH44106
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH44106
- Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH44106
| | - Mehmet Toner
- Center for Engineering in Medicine and Surgery, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA02114
- Shriners Children’s, Boston, MA02114
| | - Jason P. Acker
- Laboratory Medicine and Pathology, University of Alberta, Edmonton, ABT6G 2R8, Canada
- Innovation and Portfolio Management, Canadian Blood Services, Edmonton, ABT6G 2R8, Canada
| | - Martin L. Yarmush
- Center for Engineering in Medicine and Surgery, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA02114
- Shriners Children’s, Boston, MA02114
- Department of Biomedical Engineering, Rutgers University, Piscataway, NJ08854
| | - O. Berk Usta
- Center for Engineering in Medicine and Surgery, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA02114
- Shriners Children’s, Boston, MA02114
| |
Collapse
|
38
|
Wang Y, Huang Z, Wang X, Yang F, Yao X, Pan T, Li B, Chu J. Real-time fluorescence imaging flow cytometry enabled by motion deblurring and deep learning algorithms. LAB ON A CHIP 2023; 23:3615-3627. [PMID: 37458395 DOI: 10.1039/d3lc00194f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/09/2023]
Abstract
Fluorescence imaging flow cytometry (IFC) has been demonstrated as a crucial biomedical technique for analyzing specific cell subpopulations from heterogeneous cellular populations. However, the high-speed flow of fluorescent cells leads to motion blur in cell images, making it challenging to identify cell types from the raw images. In this study, we present a real-time single-cell imaging and classification system based on a fluorescence microscope and deep learning algorithm, which is able to directly identify cell types from motion-blur images. To obtain annotated datasets of blurred images for deep learning model training, we developed a motion deblurring algorithm for the reconstruction of blur-free images. To demonstrate the ability of this system, deblurred images of HeLa cells with various fluorescent labels and HeLa cells at different cell cycle stages were acquired. The trained ResNet achieved a high accuracy of 96.6% for single-cell classification of HeLa cells in three different mitotic stages, with a short processing time of only 2 ms. This technology provides a simple way to realize single-cell fluorescence IFC and real-time cell classification, offering significant potential in various biological and medical applications.
Collapse
Affiliation(s)
- Yiming Wang
- Department of Precision Machinery and Precision Instrumentation, University of Science and Technology of China, Hefei, 230027, China.
- Key Laboratory of Precision Scientific Instrumentation of Anhui Higher Education Institutes, University of Science and Technology of China, Hefei, 230027, China
| | - Ziwei Huang
- Department of Precision Machinery and Precision Instrumentation, University of Science and Technology of China, Hefei, 230027, China.
- Key Laboratory of Precision Scientific Instrumentation of Anhui Higher Education Institutes, University of Science and Technology of China, Hefei, 230027, China
| | - Xiaojie Wang
- Department of Precision Machinery and Precision Instrumentation, University of Science and Technology of China, Hefei, 230027, China.
- Key Laboratory of Precision Scientific Instrumentation of Anhui Higher Education Institutes, University of Science and Technology of China, Hefei, 230027, China
| | - Fengrui Yang
- MOE Key Laboratory for Membraneless Organelles and Cellular Dynamics, University of Science and Technology of China School of Life Sciences, Hefei, 230026, China
| | - Xuebiao Yao
- MOE Key Laboratory for Membraneless Organelles and Cellular Dynamics, University of Science and Technology of China School of Life Sciences, Hefei, 230026, China
| | - Tingrui Pan
- Department of Precision Machinery and Precision Instrumentation, University of Science and Technology of China, Hefei, 230027, China.
- Suzhou Institute for Advanced Research, University of Science and Technology of China, Suzhou, 215123, China
| | - Baoqing Li
- Department of Precision Machinery and Precision Instrumentation, University of Science and Technology of China, Hefei, 230027, China.
- Key Laboratory of Precision Scientific Instrumentation of Anhui Higher Education Institutes, University of Science and Technology of China, Hefei, 230027, China
| | - Jiaru Chu
- Department of Precision Machinery and Precision Instrumentation, University of Science and Technology of China, Hefei, 230027, China.
- Key Laboratory of Precision Scientific Instrumentation of Anhui Higher Education Institutes, University of Science and Technology of China, Hefei, 230027, China
| |
Collapse
|
39
|
Sreekumar SP, Palanisamy R, Swaminathan R. Semantic Segmentation of Cell Painted Organelles using DeepLabv3plus Model. ANNUAL INTERNATIONAL CONFERENCE OF THE IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. ANNUAL INTERNATIONAL CONFERENCE 2023; 2023:1-4. [PMID: 38082807 DOI: 10.1109/embc40787.2023.10340728] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2023]
Abstract
Cell painting based high content fluorescence imaging technique offers deep insight into the functional and biological changes in subcellular structures. However, advanced instrumentation and the limited availability of suitable fluorescent dyes restricts the tool to comprehensively characterize the cell morphology. Therefore, generating fluorescent specific organelle images using transmitted light microscopy provides an alternative solution for clinical applications. In this work, the utility of semantic segmentation deep network for predicting the Endoplasmic Reticulum (ER), cytoplasm and nuclei from a composite image is investigated. To perform this study, a public dataset consisting of 3456 composite images are considered from Broad Bioimage Benchmark collection. The pixel wise labeling is carried out with the generated binary masks for ER, cytoplasm and nuclei. DeepLabv3plus architecture with Atrous Spatial Pyramid Pooling (ASPP) and depth wise separable convolution is used as a learning model to perform semantic segmentation. The accuracy and loss function at different learning rates are analyzed and the segmentation results are validated using Jaccard index, mean Boundary F (BF) score and dice index. The trained model achieved 97.86% accuracy with a loss of 0.07 at the learning rate of 0.01. Mean BF score, dice index and Jaccard index for nuclei, ER and cytoplasm are (0.98, 0.94, 0.88), (0.97, 0.82, 0.7) and (0.95, 0.88, 0.66) respectively. The obtained results indicate that the adopted methodology could delineate the subcellular structures by accurately detecting sharp object boundaries. Therefore, this study could be useful for predicting the cell painted images from transmitted light microscopy without the requirement of fluorescent labeling.
Collapse
|
40
|
Yu HQ, Li F, Xiong H, Fang L, Zhang J, Bie P, Xie CM. Elevated FBXL18 promotes RPS15A ubiquitination and SMAD3 activation to drive HCC. Hepatol Commun 2023; 7:e00198. [PMID: 37378633 DOI: 10.1097/hc9.0000000000000198] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Accepted: 05/12/2023] [Indexed: 06/29/2023] Open
Abstract
BACKGROUND F-box and leucine-rich repeat protein 18 (FBXL18) is an E3 ubiquitin ligase that is reported to be involved in the tumorigenesis of various types of cancer. However, it remains unknown whether FBXL18 is correlated with hepatocarcinogenesis. METHODS AND RESULTS In the current study, we found that FBXL18 was highly expressed in HCC tissues and positively associated with poor overall survival of HCC patients. FBXL18 was an independent risk factor for HCC patients. We observed that FBXL18 drove HCC in FBXL18 transgenic mice. Mechanistically, FBXL18 promoted the K63-linked ubiquitination of small-subunit ribosomal protein S15A (RPS15A) and enhanced its stability, increasing SMAD family member 3 (SMAD3) levels and translocation to the nucleus and promoting HCC cell proliferation. Moreover, the knockdown of RPS15A or SMAD3 significantly suppressed FBXL18-mediated HCC proliferation. In clinical samples, elevated FBXL18 expression was positively associated with RPS15A expression. CONCLUSION FBXL18 promotes RPS15A ubiquitination and upregulates SMAD3 expression, leading to hepatocellular carcinogenesis, and this study provides a novel therapeutic strategy for HCC treatment by targeting the FBXL18/RPS15A/SMAD3 pathway.
Collapse
Affiliation(s)
- Hong-Qiang Yu
- Key Laboratory of Hepatobiliary and Pancreatic Surgery, Institute of Hepatobiliary Surgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, P.R. China
| | - Feng Li
- Department of Hepatobiliary Surgery, The Third Affiliated hospital of Chongqing Medical University, Chongqing, P.R. China
| | - HaoJun Xiong
- Key Laboratory of Hepatobiliary and Pancreatic Surgery, Institute of Hepatobiliary Surgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, P.R. China
| | - Lei Fang
- Key Laboratory of Hepatobiliary and Pancreatic Surgery, Institute of Hepatobiliary Surgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, P.R. China
| | - Jie Zhang
- Key Laboratory of Hepatobiliary and Pancreatic Surgery, Institute of Hepatobiliary Surgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, P.R. China
| | - Ping Bie
- Department of Hepatobiliary Surgery, The Third Affiliated hospital of Chongqing Medical University, Chongqing, P.R. China
| | - Chuan-Ming Xie
- Key Laboratory of Hepatobiliary and Pancreatic Surgery, Institute of Hepatobiliary Surgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, P.R. China
| |
Collapse
|
41
|
Pfisterer F, Godino N, Gerling T, Kirschbaum M. Continuous microfluidic flow-through protocol for selective and image-activated electroporation of single cells. RSC Adv 2023; 13:19379-19387. [PMID: 37383687 PMCID: PMC10294288 DOI: 10.1039/d3ra03100d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Accepted: 05/31/2023] [Indexed: 06/30/2023] Open
Abstract
Electroporation of cells is a widely-used tool to transport molecules such as proteins or nucleic acids into cells or to extract cellular material. However, bulk methods for electroporation do not offer the possibility to selectively porate subpopulations or single cells in heterogeneous cell samples. To achieve this, either presorting or complex single-cell technologies are required currently. In this work, we present a microfluidic flow protocol for selective electroporation of predefined target cells identified in real-time by high-quality microscopic image analysis of fluorescence and transmitted light. While traveling through the microchannel, the cells are focused by dielectrophoretic forces into the microscopic detection area, where they are classified based on image analysis techniques. Finally, the cells are forwarded to a poration electrode and only the target cells are pulsed. By processing a heterogenically stained cell sample, we were able to selectively porate only target cells (green-fluorescent) while non-target cells (blue-fluorescent) remained unaffected. We achieved highly selective poration with >90% specificity at average poration rates of >50% and throughputs of up to 7200 cells per hour.
Collapse
Affiliation(s)
- Felix Pfisterer
- Fraunhofer Institute for Cell Therapy and Immunology IZI, Branch Bioanalytics and Bioprocesses IZI-BB Am Muehlenberg 13 14476 Potsdam Germany
| | - Neus Godino
- Fraunhofer Institute for Cell Therapy and Immunology IZI, Branch Bioanalytics and Bioprocesses IZI-BB Am Muehlenberg 13 14476 Potsdam Germany
| | - Tobias Gerling
- Fraunhofer Institute for Cell Therapy and Immunology IZI, Branch Bioanalytics and Bioprocesses IZI-BB Am Muehlenberg 13 14476 Potsdam Germany
| | - Michael Kirschbaum
- Fraunhofer Institute for Cell Therapy and Immunology IZI, Branch Bioanalytics and Bioprocesses IZI-BB Am Muehlenberg 13 14476 Potsdam Germany
| |
Collapse
|
42
|
Gerling T, Godino N, Pfisterer F, Hupf N, Kirschbaum M. High-precision, low-complexity, high-resolution microscopy-based cell sorting. LAB ON A CHIP 2023. [PMID: 37314345 DOI: 10.1039/d3lc00242j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Continuous flow cell sorting based on image analysis is a powerful concept that exploits spatially-resolved features in cells, such as subcellular protein localisation or cell and organelle morphology, to isolate highly specialised cell types that were previously inaccessible to biomedical research, biotechnology, and medicine. Recently, sorting protocols have been proposed that achieve impressive throughput by combining ultra-high flow rates with sophisticated imaging and data processing protocols. However, moderate image quality and high complex experimental setups still prevent the full potential of image-activated cell sorting from being a general-purpose tool. Here, we present a new low-complexity microfluidic approach based on high numerical aperture wide-field microscopy and precise dielectrophoretic cell handling. It provides high-quality images with unprecedented resolution in image-activated cell sorting (i.e., 216 nm). In addition, it also allows long image processing times of several hundred milliseconds for thorough image analysis, while ensuring reliable and low-loss cell processing. Using our approach, we sorted live T cells based on subcellular localisation of fluorescence signals and demonstrated that purities above 80% are possible while targeting maximum yields and sample volume throughputs in the range of μl min-1. We were able to recover 85% of the target cells analysed. Finally, we ensure and quantify the full vitality of the sorted cells cultivating the cells for a period of time and through colorimetric viability tests.
Collapse
Affiliation(s)
- Tobias Gerling
- Fraunhofer Institute for Cell Therapy and Immunology, Branch Bioanalytics and Bioprocesses IZI-BB, Am Muehlenberg 13, 14476 Potsdam, Germany.
- Institute of Biochemistry and Biology, University of Potsdam, Karl-Liebknecht-Str. 24/25, 14476 Potsdam, Germany
| | - Neus Godino
- Fraunhofer Institute for Cell Therapy and Immunology, Branch Bioanalytics and Bioprocesses IZI-BB, Am Muehlenberg 13, 14476 Potsdam, Germany.
| | - Felix Pfisterer
- Fraunhofer Institute for Cell Therapy and Immunology, Branch Bioanalytics and Bioprocesses IZI-BB, Am Muehlenberg 13, 14476 Potsdam, Germany.
| | - Nina Hupf
- Fraunhofer Institute for Cell Therapy and Immunology, Branch Bioanalytics and Bioprocesses IZI-BB, Am Muehlenberg 13, 14476 Potsdam, Germany.
| | - Michael Kirschbaum
- Fraunhofer Institute for Cell Therapy and Immunology, Branch Bioanalytics and Bioprocesses IZI-BB, Am Muehlenberg 13, 14476 Potsdam, Germany.
| |
Collapse
|
43
|
Song T, He N, Hao Z, Yang Y. Upregulation of ENKD1 disrupts cellular homeostasis to promote lymphoma development. J Cell Physiol 2023; 238:1308-1323. [PMID: 36960713 DOI: 10.1002/jcp.31012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Revised: 03/09/2023] [Accepted: 03/14/2023] [Indexed: 03/25/2023]
Abstract
Diffuse large B cell lymphoma (DLBCL) is a common and aggressive form of B cell lymphoma. Approximately 40% of DLBCL patients are incurable despite modern therapeutic approaches. To explore the molecular mechanisms driving the growth and progression of DLBCL, we analyzed genes with differential expression in DLBCL using the Gene Expression Profiling Interactive Analysis database. Enkurin domain-containing protein 1 (ENKD1), a centrosomal protein-encoding gene, was found to be highly expressed in DLBCL samples compared with normal samples. The phylogenetic analysis revealed that ENKD1 is evolutionarily conserved. Depletion of ENKD1 in cultured DLBCL cells induced apoptosis, suppressed cell proliferation, and blocked cell cycle progression in the G2/M phase. Moreover, ENKD1 expression positively correlates with the expression levels of a number of cellular homeostatic regulators, including Sperm-associated antigen 5, a gene encoding an important mitotic regulator. These findings thus demonstrate a critical function for ENKD1 in regulating the cellular homeostasis and suggest a potential value of targeting ENKD1 for the treatment of DLBCL.
Collapse
Affiliation(s)
- Ting Song
- Department of Cell Biology, School of Basic Medical Sciences, Cheeloo Medical College, Shandong University, Jinan, China
| | - Na He
- Department of Hematology, Qilu Hospital of Shandong University, Jinan, China
| | - Ziqian Hao
- College of Artificial Intelligence and Big Data for Medical Sciences, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| | - Yunfan Yang
- Department of Cell Biology, School of Basic Medical Sciences, Cheeloo Medical College, Shandong University, Jinan, China
| |
Collapse
|
44
|
Zhang XW, Yang YF, Qi GX, Zhai FH, Fei T, Wang JH, Yu YL, Chen S. Rapid and Accurate Identification of Cell Phenotypes of Different Drug Mechanisms by Using Single-Cell Fluorescence Images Via Deep Learning. Anal Chem 2023; 95:8113-8120. [PMID: 37162406 DOI: 10.1021/acs.analchem.3c01140] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/11/2023]
Abstract
Identification of a drug mechanism is vital for drug development. However, it often resorts to the expensive and cumbersome omics methods along with complex data analysis. Herein, we developed a methodology to analyze organelle staining images of single cells using a deep learning algorithm (TL-ResNet50) for rapid and accurate identification of different drug mechanisms. Based on the organelle-related cell morphological changes caused by drug action, the constructed deep learning model can fast predict the drug mechanism with a high accuracy of 92%. Further analysis reveals that drug combination at different ratios can enhance a certain mechanism or generate a new mechanism. This work would highly facilitate clinical medication and drug screening.
Collapse
Affiliation(s)
- Xue-Wei Zhang
- Research Center for Analytical Sciences, Department of Chemistry, College of Sciences, Northeastern University, Box 332, Shenyang 110819, China
| | - Yan-Fei Yang
- Research Center for Analytical Sciences, Department of Chemistry, College of Sciences, Northeastern University, Box 332, Shenyang 110819, China
| | - Gong-Xiang Qi
- Research Center for Analytical Sciences, Department of Chemistry, College of Sciences, Northeastern University, Box 332, Shenyang 110819, China
| | - Fu-Heng Zhai
- Research Center for Analytical Sciences, Department of Chemistry, College of Sciences, Northeastern University, Box 332, Shenyang 110819, China
| | - Teng Fei
- Research Center for Analytical Sciences, Department of Chemistry, College of Sciences, Northeastern University, Box 332, Shenyang 110819, China
| | - Jian-Hua Wang
- Research Center for Analytical Sciences, Department of Chemistry, College of Sciences, Northeastern University, Box 332, Shenyang 110819, China
| | - Yong-Liang Yu
- Research Center for Analytical Sciences, Department of Chemistry, College of Sciences, Northeastern University, Box 332, Shenyang 110819, China
| | - Shuai Chen
- Research Center for Analytical Sciences, Department of Chemistry, College of Sciences, Northeastern University, Box 332, Shenyang 110819, China
| |
Collapse
|
45
|
Godino E, Restrepo Sierra AM, Danelon C. Imaging Flow Cytometry for High-Throughput Phenotyping of Synthetic Cells. ACS Synth Biol 2023. [PMID: 37155828 PMCID: PMC10367129 DOI: 10.1021/acssynbio.3c00074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/10/2023]
Abstract
The reconstitution of basic cellular functions in micrometer-sized liposomes has led to a surge of interest in the construction of synthetic cells. Microscopy and flow cytometry are powerful tools for characterizing biological processes in liposomes with fluorescence readouts. However, applying each method separately leads to a compromise between information-rich imaging by microscopy and statistical population analysis by flow cytometry. To address this shortcoming, we here introduce imaging flow cytometry (IFC) for high-throughput, microscopy-based screening of gene-expressing liposomes in laminar flow. We developed a comprehensive pipeline and analysis toolset based on a commercial IFC instrument and software. About 60 thousands of liposome events were collected per run starting from one microliter of the stock liposome solution. Robust population statistics from individual liposome images was performed based on fluorescence and morphological parameters. This allowed us to quantify complex phenotypes covering a wide range of liposomal states that are relevant for building a synthetic cell. The general applicability, current workflow limitations, and future prospects of IFC in synthetic cell research are finally discussed.
Collapse
Affiliation(s)
- Elisa Godino
- Department of Bionanoscience, Kavli Institute of Nanoscience, Delft University of Technology, 2629HZ Delft, The Netherlands
| | - Ana Maria Restrepo Sierra
- Department of Bionanoscience, Kavli Institute of Nanoscience, Delft University of Technology, 2629HZ Delft, The Netherlands
| | - Christophe Danelon
- Department of Bionanoscience, Kavli Institute of Nanoscience, Delft University of Technology, 2629HZ Delft, The Netherlands
- Toulouse Biotechnology Institute (TBI), Université de Toulouse, CNRS, INRAE, INSA, 31077 Toulouse, France
| |
Collapse
|
46
|
Zhong J, Liang M, Tang Q, Ai Y. Selectable encapsulated cell quantity in droplets via label-free electrical screening and impedance-activated sorting. Mater Today Bio 2023; 19:100594. [PMID: 36910274 PMCID: PMC9999206 DOI: 10.1016/j.mtbio.2023.100594] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2023] [Revised: 02/21/2023] [Accepted: 02/22/2023] [Indexed: 03/02/2023] Open
Abstract
Single-cell encapsulation in droplets has become a powerful tool in immunotherapy, medicine discovery, and single-cell analysis, thanks to its capability for cell confinement in picoliter volumes. However, the purity and throughput of single-cell droplets are limited by random encapsulation process, which resuts in a majority of empty and multi-cells droplets. Herein we introduce the first label-free selectable cell quantity encapsulation in droplets sorting system to overcome this problem. The system utilizes a simple and reliable electrical impedance based screening (98.9% of accuracy) integrated with biocompatible acoustic sorting to select single-cell droplets, achieving 90.3% of efficiency and up to 200 Hz of throughput, by removing multi-cells (∼60% of rejection) and empty droplets (∼90% of rejection). We demonstrate the use of the droplet sorting to improve the throughput of single-cell encapsulation by ∼9-fold compared to the conventional random encapsulation process.
Collapse
Affiliation(s)
- Jianwei Zhong
- Pillar of Engineering Product Development, Singapore University of Technology and Design, 8 Somapah Road, Singapore, 487372, Singapore
| | - Minhui Liang
- Pillar of Engineering Product Development, Singapore University of Technology and Design, 8 Somapah Road, Singapore, 487372, Singapore
| | - Qiang Tang
- Jiangsu Provincial Engineering Research Center for Biomedical Materials and Advanced Medical Devices, Faculty of Mechanical and Material Engineering, Huaiyin Institute of Technology, Huaian, 223003, China
| | - Ye Ai
- Pillar of Engineering Product Development, Singapore University of Technology and Design, 8 Somapah Road, Singapore, 487372, Singapore
| |
Collapse
|
47
|
Wills JW, Robertson J, Tourlomousis P, Gillis CM, Barnes CM, Miniter M, Hewitt RE, Bryant CE, Summers HD, Powell JJ, Rees P. Label-free cell segmentation of diverse lymphoid tissues in 2D and 3D. CELL REPORTS METHODS 2023; 3:100398. [PMID: 36936072 PMCID: PMC10014308 DOI: 10.1016/j.crmeth.2023.100398] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Revised: 10/14/2022] [Accepted: 01/11/2023] [Indexed: 02/05/2023]
Abstract
Unlocking and quantifying fundamental biological processes through tissue microscopy requires accurate, in situ segmentation of all cells imaged. Currently, achieving this is complex and requires exogenous fluorescent labels that occupy significant spectral bandwidth, increasing the duration and complexity of imaging experiments while limiting the number of channels remaining to address the study's objectives. We demonstrate that the excitation light reflected during routine confocal microscopy contains sufficient information to achieve accurate, label-free cell segmentation in 2D and 3D. This is achieved using a simple convolutional neural network trained to predict the probability that reflected light pixels belong to either nucleus, cytoskeleton, or background classifications. We demonstrate the approach across diverse lymphoid tissues and provide video tutorials demonstrating deployment in Python and MATLAB or via standalone software for Windows.
Collapse
Affiliation(s)
- John W. Wills
- Department of Veterinary Medicine, Cambridge University, Madingley Road, Cambridge CB3 0ES, UK
- Department of Biomedical Engineering, Swansea University, Fabian Way, Crymlyn Burrows, Swansea SA1 8EN, Wales, UK
| | - Jack Robertson
- Department of Veterinary Medicine, Cambridge University, Madingley Road, Cambridge CB3 0ES, UK
| | - Pani Tourlomousis
- Department of Veterinary Medicine, Cambridge University, Madingley Road, Cambridge CB3 0ES, UK
| | - Clare M.C. Gillis
- Department of Veterinary Medicine, Cambridge University, Madingley Road, Cambridge CB3 0ES, UK
| | - Claire M. Barnes
- Department of Biomedical Engineering, Swansea University, Fabian Way, Crymlyn Burrows, Swansea SA1 8EN, Wales, UK
| | - Michelle Miniter
- Department of Veterinary Medicine, Cambridge University, Madingley Road, Cambridge CB3 0ES, UK
| | - Rachel E. Hewitt
- Department of Veterinary Medicine, Cambridge University, Madingley Road, Cambridge CB3 0ES, UK
| | - Clare E. Bryant
- Department of Veterinary Medicine, Cambridge University, Madingley Road, Cambridge CB3 0ES, UK
| | - Huw D. Summers
- Department of Biomedical Engineering, Swansea University, Fabian Way, Crymlyn Burrows, Swansea SA1 8EN, Wales, UK
| | - Jonathan J. Powell
- Department of Veterinary Medicine, Cambridge University, Madingley Road, Cambridge CB3 0ES, UK
| | - Paul Rees
- Department of Biomedical Engineering, Swansea University, Fabian Way, Crymlyn Burrows, Swansea SA1 8EN, Wales, UK
- Imaging Platform, Broad Institute of MIT and Harvard, 415 Main Street, Boston, Cambridge, MA 02142, USA
| |
Collapse
|
48
|
Kinegawa R, Gala de Pablo J, Wang Y, Hiramatsu K, Goda K. Label-free multiphoton imaging flow cytometry. Cytometry A 2023. [PMID: 36799568 DOI: 10.1002/cyto.a.24723] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Revised: 01/31/2023] [Accepted: 02/13/2023] [Indexed: 02/18/2023]
Abstract
Label-free imaging flow cytometry is a powerful tool for biological and medical research as it overcomes technical challenges in conventional fluorescence-based imaging flow cytometry that predominantly relies on fluorescent labeling. To date, two distinct types of label-free imaging flow cytometry have been developed, namely optofluidic time-stretch quantitative phase imaging flow cytometry and stimulated Raman scattering (SRS) imaging flow cytometry. Unfortunately, these two methods are incapable of probing some important molecules such as starch and collagen. Here, we present another type of label-free imaging flow cytometry, namely multiphoton imaging flow cytometry, for visualizing starch and collagen in live cells with high throughput. Our multiphoton imaging flow cytometer is based on nonlinear optical imaging whose image contrast is provided by two optical nonlinear effects: four-wave mixing (FWM) and second-harmonic generation (SHG). It is composed of a microfluidic chip with an acoustic focuser, a lab-made laser scanning SHG-FWM microscope, and a high-speed image acquisition circuit to simultaneously acquire FWM and SHG images of flowing cells. As a result, it acquires FWM and SHG images (100 × 100 pixels) with a spatial resolution of 500 nm and a field of view of 50 μm × 50 μm at a high event rate of four to five events per second, corresponding to a high throughput of 560-700 kb/s, where the event is defined by the passage of a cell or a cell-like particle. To show the utility of our multiphoton imaging flow cytometer, we used it to characterize Chromochloris zofingiensis (NIES-2175), a unicellular green alga that has recently attracted attention from the industrial sector for its ability to efficiently produce valuable materials for bioplastics, food, and biofuel. Our statistical image analysis found that starch was distributed at the center of the cells at the early cell cycle stage and became delocalized at the later stage. Multiphoton imaging flow cytometry is expected to be an effective tool for statistical high-content studies of biological functions and optimizing the evolution of highly productive cell strains.
Collapse
Affiliation(s)
- Ryo Kinegawa
- Department of Chemistry, The University of Tokyo, Tokyo, Japan
| | | | - Yi Wang
- Department of Chemistry, Renmin University of China, Beijing, China
| | - Kotaro Hiramatsu
- Department of Chemistry, The University of Tokyo, Tokyo, Japan.,Research Centre for Spectrochemistry, The University of Tokyo, Tokyo, Japan.,PRESTO, Japan Science and Technology Agency, Saitama, 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, California, USA.,CYBO, Inc., Tokyo, Japan
| |
Collapse
|
49
|
Recent advances in non-optical microfluidic platforms for bioparticle detection. Biosens Bioelectron 2023; 222:114944. [PMID: 36470061 DOI: 10.1016/j.bios.2022.114944] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Revised: 11/17/2022] [Accepted: 11/22/2022] [Indexed: 12/03/2022]
Abstract
The effective analysis of the basic structure and functional information of bioparticles are of great significance for the early diagnosis of diseases. The synergism between microfluidics and particle manipulation/detection technologies offers enhanced system integration capability and test accuracy for the detection of various bioparticles. Most microfluidic detection platforms are based on optical strategies such as fluorescence, absorbance, and image recognition. Although optical microfluidic platforms have proven their capabilities in the practical clinical detection of bioparticles, shortcomings such as expensive components and whole bulky devices have limited their practicality in the development of point-of-care testing (POCT) systems to be used in remote and underdeveloped areas. Therefore, there is an urgent need to develop cost-effective non-optical microfluidic platforms for bioparticle detection that can act as alternatives to optical counterparts. In this review, we first briefly summarise passive and active methods for bioparticle manipulation in microfluidics. Then, we survey the latest progress in non-optical microfluidic strategies based on electrical, magnetic, and acoustic techniques for bioparticle detection. Finally, a perspective is offered, clarifying challenges faced by current non-optical platforms in developing practical POCT devices and clinical applications.
Collapse
|
50
|
Hsieh PH, Phal Y, Prasanth KV, Bhargava R. Cell Phase Identification in a Three-Dimensional Engineered Tumor Model by Infrared Spectroscopic Imaging. Anal Chem 2023; 95:3349-3357. [PMID: 36574385 PMCID: PMC10214899 DOI: 10.1021/acs.analchem.2c04554] [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] [Indexed: 12/28/2022]
Abstract
Cell cycle progression plays a vital role in regulating proliferation, metabolism, and apoptosis. Three-dimensional (3D) cell cultures have emerged as an important class of in vitro disease models, and incorporating the variation occurring from cell cycle progression in these systems is critical. Here, we report the use of Fourier transform infrared (FT-IR) spectroscopic imaging to identify subtle biochemical changes within cells, indicative of the G1/S and G2/M phases of the cell cycle. Following previous studies, we first synchronized samples from two-dimensional (2D) cell cultures, confirmed their states by flow cytometry and DNA quantification, and recorded spectra. We determined two critical wavenumbers (1059 and 1219 cm-1) as spectral indicators of the cell cycle for a set of isogenic breast cancer cell lines (MCF10AT series). These two simple spectral markers were then applied to distinguish cell cycle stages in a 3D cell culture model using four cell lines that represent the main stages of cancer progression from normal cells to metastatic disease. Temporal dependence of spectral biomarkers during acini maturation validated the hypothesis that the cells are more proliferative in the early stages of acini development; later stages of the culture showed stability in the overall composition but unique spatial differences in cells in the two phases. Altogether, this study presents a computational and quantitative approach for cell phase analysis in tissue-like 3D structures without any biomarker staining and provides a means to characterize the impact of the cell cycle on 3D biological systems and disease diagnostic studies using IR imaging.
Collapse
Affiliation(s)
- Pei-Hsuan Hsieh
- Department of Bioengineering and Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, United States
| | - Yamuna Phal
- Department of Electrical and Computer Engineering and Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, United States
| | - Kannanganattu V Prasanth
- Department of Cell and Developmental Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, United States
| | - Rohit Bhargava
- Departments of Bioengineering, Electrical and Computer Engineering, Mechanical Science and Engineering, Chemical and Biomolecular Engineering, and Chemistry, Beckman Institute for Advanced Science and Technology, Cancer Center at Illinois, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, United States
| |
Collapse
|