1
|
Kumar P, Mondal PP. Multicolor iLIFE (m-iLIFE) volume cytometry for high-throughput imaging of multiple organelles. Sci Rep 2024; 14:23798. [PMID: 39394224 PMCID: PMC11470118 DOI: 10.1038/s41598-024-73667-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Accepted: 09/19/2024] [Indexed: 10/13/2024] Open
Abstract
To be able to resolve multiple organelles at high throughput is an incredible achievement. This will have immediate implications in a range of fields ranging from fundamental cell biology to translational medicine. To realize such a high-throughput multicolor interrogation modality, we have developed a light-sheet based flow imaging system that is capable of visualizing multiple sub-cellular components with organelle-level resolution. This is possible due to the unique optical design that combines an illumination system comprising two collinear light sheets illuminating the flowing cells and a dedicated dual-color 4f-detection, enabling simultaneous recording of multiple organelles. The system PSF sections up to 4 parallel microfluidic channels through which cells are flowing, and multicolor images of cell cross-sections are recorded. The data is then computationally processed (filtered using ML algorithm, shift-corrected, and merged) and combined to reconstruct the 3D multicolor volume. System testing is conducted using multicolor fluorescent nano-beads (size ∼ 175 nm) and flow-based imaging parameters (PSF size, motion-blur, flow rate, frame rate, and number of cell-sections) are determined for quality imaging. Drug treatment studies were carried out for healthy and cancerous HeLa cells to check the performance of the proposed system. The cells were treated with a drug (Vincristine, which is known to promote mitochondrial fission in cells), and the same is compared with untreated control cells. The proposed multicolor iLIFE system could screen ∼ 800 cells/min (at a flow speed of 2490 μ m/s), and the drug treatment studies were carried out up to 24 h. Studies showed the disintegration of mitochondrial network and dysfunctional lysosomes and their accumulation at the cell membrane, which is a clear indication of cell apoptosis. Compared to control cells (untreated), the mortality is highest at a concentration of 500 nM post 12 h of drug treatment. With the capability of multiorganelle interrogation and organelle-level resolution, the multicolor iLIFE cytometry system is suitably placed to assist optical imaging and biomedical research.
Collapse
Affiliation(s)
- Prashant Kumar
- Mondal Lab, Department of Instrumentation and Applied Physics, Indian Institute of Science, Bangalore, 560012, India
| | - Partha Pratim Mondal
- Mondal Lab, Department of Instrumentation and Applied Physics, Indian Institute of Science, Bangalore, 560012, India.
- Centre for Cryogenic Technology, Indian Institute of Science, Bangalore, 560012, India.
| |
Collapse
|
2
|
Kalasin S, Surareungchai W. Artificial intelligence-aiding lab-on-a-chip workforce designed oral [3.1.0] bi and [4.2.0] tricyclic catalytic interceptors inhibiting multiple SARS-CoV-2 protomers assisted by double-shell deep learning. RSC Adv 2024; 14:26897-26910. [PMID: 39193274 PMCID: PMC11347926 DOI: 10.1039/d4ra03965c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Accepted: 08/20/2024] [Indexed: 08/29/2024] Open
Abstract
While each massive pandemic has claimed the lives of millions of vulnerable populations over the centuries, one limitation exists: that the Edisonian approach (human-directed with trial errors) relies on repurposing pharmaceuticals, designing drugs, and herbal remedies with the violation of Lipinski's rule of five druglikeness. It may lead to adverse health effects with long-term health multimorbidity. Nevertheless, declining birth rates and aging populations will likely cause a shift in society due to a shortage of a scientific workforce to defend against the next pandemic incursion. The challenge of combating the ongoing post-COVID-19 pandemic has been exacerbated by the lack of gold standard drugs to deactivate multiple SARS-CoV-2 protein targets. Meanwhile, there are three FDA-approved antivirals, Remdesivir, Molnupiravir, and Paxlovid, with moderate clinical efficacy and drug resistance. There is a pressing need for additional antivirals and prepared omics technology to combat the current and future devastating coronavirus pandemics. While there is a limitation of existing contemporary inhibitors to deactivate viral RNA replication with minimal rotational bonds, one strategy is to create Lipinski inhibitors with less than 10 rotational bonds and precise halogen bond placement to destabilize multiple viral protomers. This work describes the efforts to design gold-standard oral inhibitors of bi- and tri-cyclic catalytic interceptors with electrophilic heads using double-shell deep learning. Here, KS1 with and KS2 compounds designed by lab-on-a-chip technology attain 5-fold novel filtered-Lipinski, GHOSE, VEBER, EGAN, and MUEGGE druglikeness. The graph neural network (GNN) relies on module-initiation, expansion, relabeling atom index, and termination (METORITE) iterations, while the deep neural network (DNN) engages pinning, extraction, convolution, pooling, and flattening (PROOF) operations. The cyclic compound's specific halogen atom location enhances the nitrile catalytic head, which deactivates several viral protein targets. Initiating this lab-on-a-chip that is not susceptible to the aging process for creating clinical compounds can leverage a new path to many valuable drugs with speedy oral drug discovery, especially to defend the loss of vulnerable population and prevent multimorbidity that is susceptible to hidden viral persistence in the continuing aging times.
Collapse
Affiliation(s)
- Surachate Kalasin
- Faculty of Science and Nanoscience & Nanotechnology Graduate Program, King Mongkut's University of Technology Thonburi 10140 Thailand
| | - Werasak Surareungchai
- Faculty of Science and Nanoscience & Nanotechnology Graduate Program, King Mongkut's University of Technology Thonburi 10140 Thailand
- Pilot Plant Research and Development Laboratory, King Mongkut's University of Technology Thonburi 10150 Bangkok Thailand
- School of Bioresource and Technology, King Mongkut's University of Technology Thonburi 10150 Bangkok Thailand
- Analytical Sciences and National Doping Test Institute, Mahidol University Bangkok 10400 Thailand
| |
Collapse
|
3
|
Rodoplu Solovchuk D. Advances in AI-assisted biochip technology for biomedicine. Biomed Pharmacother 2024; 177:116997. [PMID: 38943990 DOI: 10.1016/j.biopha.2024.116997] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Revised: 06/13/2024] [Accepted: 06/15/2024] [Indexed: 07/01/2024] Open
Abstract
The integration of biochips with AI opened up new possibilities and is expected to revolutionize smart healthcare tools within the next five years. The combination of miniaturized, multi-functional, rapid, high-throughput sample processing and sensing capabilities of biochips, with the computational data processing and predictive power of AI, allows medical professionals to collect and analyze vast amounts of data quickly and efficiently, leading to more accurate and timely diagnoses and prognostic evaluations. Biochips, as smart healthcare devices, offer continuous monitoring of patient symptoms. Integrated virtual assistants have the potential to send predictive feedback to users and healthcare practitioners, paving the way for personalized and predictive medicine. This review explores the current state-of-the-art biochip technologies including gene-chips, organ-on-a-chips, and neural implants, and the diagnostic and therapeutic utility of AI-assisted biochips in medical practices such as cancer, diabetes, infectious diseases, and neurological disorders. Choosing the appropriate AI model for a specific biomedical application, and possible solutions to the current challenges are explored. Surveying advances in machine learning models for biochip functionality, this paper offers a review of biochips for the future of biomedicine, an essential guide for keeping up with trends in healthcare, while inspiring cross-disciplinary collaboration among biomedical engineering, medicine, and machine learning fields.
Collapse
Affiliation(s)
- Didem Rodoplu Solovchuk
- Institute of Biomedical Engineering and Nanomedicine, National Health Research Institutes, Zhunan, Miaoli 35053, Taiwan.
| |
Collapse
|
4
|
Zhang P, Liu C, Modavi C, Abate A, Chen H. Printhead on a chip: empowering droplet-based bioprinting with microfluidics. Trends Biotechnol 2024; 42:353-368. [PMID: 37777352 DOI: 10.1016/j.tibtech.2023.09.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2023] [Revised: 09/02/2023] [Accepted: 09/11/2023] [Indexed: 10/02/2023]
Abstract
Droplet-based bioprinting has long struggled with the manipulation and dispensation of individual cells from a printhead, hindering the fabrication of artificial cellular structures with high precision. The integration of modern microfluidic modules into the printhead of a bioprinter is emerging as one approach to overcome this bottleneck. This convergence allows for high-accuracy manipulation and spatial control over placement of cells during printing, and enables the fabrication of cell arrays and hierarchical heterogenous microtissues, opening new applications in bioanalysis and high-throughput screening. In this review, we summarize recent developments in the use of microfluidics in droplet printing systems, with consideration of the working principles; present applications extended through microfluidic features; and discuss the future of this technology.
Collapse
Affiliation(s)
- Pengfei Zhang
- School of Mechanical Engineering and Automation, Beihang University, Beijing, China.
| | - Congying Liu
- School of Mechanical Engineering and Automation, Beihang University, Beijing, China
| | - Cyrus Modavi
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, San Francisco, CA, USA
| | - Adam Abate
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, San Francisco, CA, USA; California Institute for Quantitative Biosciences, University of California, San Francisco, San Francisco, CA, USA.
| | - Huawei Chen
- School of Mechanical Engineering and Automation, Beihang University, Beijing, China
| |
Collapse
|
5
|
Lin M, Zhang Z, Gao X, Bian Y, Wu RS, Park G, Lou Z, Zhang Z, Xu X, Chen X, Kang A, Yang X, Yue W, Yin L, Wang C, Qi B, Zhou S, Hu H, Huang H, Li M, Gu Y, Mu J, Yang A, Yaghi A, Chen Y, Lei Y, Lu C, Wang R, Wang J, Xiang S, Kistler EB, Vasconcelos N, Xu S. A fully integrated wearable ultrasound system to monitor deep tissues in moving subjects. Nat Biotechnol 2024; 42:448-457. [PMID: 37217752 DOI: 10.1038/s41587-023-01800-0] [Citation(s) in RCA: 39] [Impact Index Per Article: 39.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Accepted: 04/21/2023] [Indexed: 05/24/2023]
Abstract
Recent advances in wearable ultrasound technologies have demonstrated the potential for hands-free data acquisition, but technical barriers remain as these probes require wire connections, can lose track of moving targets and create data-interpretation challenges. Here we report a fully integrated autonomous wearable ultrasonic-system-on-patch (USoP). A miniaturized flexible control circuit is designed to interface with an ultrasound transducer array for signal pre-conditioning and wireless data communication. Machine learning is used to track moving tissue targets and assist the data interpretation. We demonstrate that the USoP allows continuous tracking of physiological signals from tissues as deep as 164 mm. On mobile subjects, the USoP can continuously monitor physiological signals, including central blood pressure, heart rate and cardiac output, for as long as 12 h. This result enables continuous autonomous surveillance of deep tissue signals toward the internet-of-medical-things.
Collapse
Affiliation(s)
- Muyang Lin
- Department of Nanoengineering, University of California San Diego, La Jolla, CA, USA
| | - Ziyang Zhang
- Department of Computer Science Engineering, University of California San Diego, La Jolla, CA, USA
| | - Xiaoxiang Gao
- Department of Nanoengineering, University of California San Diego, La Jolla, CA, USA
| | - Yizhou Bian
- Department of Nanoengineering, University of California San Diego, La Jolla, CA, USA
| | - Ray S Wu
- Department of Nanoengineering, University of California San Diego, La Jolla, CA, USA
| | - Geonho Park
- Department of Nanoengineering, University of California San Diego, La Jolla, CA, USA
| | - Zhiyuan Lou
- Department of Nanoengineering, University of California San Diego, La Jolla, CA, USA
| | - Zhuorui Zhang
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Xiangchen Xu
- Department of Nanoengineering, University of California San Diego, La Jolla, CA, USA
| | - Xiangjun Chen
- Materials Science and Engineering Program, University of California San Diego, La Jolla, CA, USA
| | - Andrea Kang
- Department of Electrical and Computer Engineering, University of California San Diego, La Jolla, CA, USA
| | - Xinyi Yang
- Materials Science and Engineering Program, University of California San Diego, La Jolla, CA, USA
| | - Wentong Yue
- Department of Nanoengineering, University of California San Diego, La Jolla, CA, USA
| | - Lu Yin
- Department of Nanoengineering, University of California San Diego, La Jolla, CA, USA
| | - Chonghe Wang
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Baiyan Qi
- Materials Science and Engineering Program, University of California San Diego, La Jolla, CA, USA
| | - Sai Zhou
- Materials Science and Engineering Program, University of California San Diego, La Jolla, CA, USA
| | - Hongjie Hu
- Department of Nanoengineering, University of California San Diego, La Jolla, CA, USA
| | - Hao Huang
- Department of Nanoengineering, University of California San Diego, La Jolla, CA, USA
| | - Mohan Li
- Department of Electrical and Computer Engineering, University of California San Diego, La Jolla, CA, USA
| | - Yue Gu
- Materials Science and Engineering Program, University of California San Diego, La Jolla, CA, USA
- Department of Neurosurgery, Yale University, New Haven, CT, USA
| | - Jing Mu
- Materials Science and Engineering Program, University of California San Diego, La Jolla, CA, USA
| | - Albert Yang
- Department of Bioengineering, University of California San Diego, La Jolla, CA, USA
| | - Amer Yaghi
- Department of Nanoengineering, University of California San Diego, La Jolla, CA, USA
| | - Yimu Chen
- Department of Nanoengineering, University of California San Diego, La Jolla, CA, USA
| | - Yusheng Lei
- Department of Nanoengineering, University of California San Diego, La Jolla, CA, USA
- Department of Chemical Engineering, Stanford University, Stanford, CA, USA
| | - Chengchangfeng Lu
- Department of Electrical and Computer Engineering, University of California San Diego, La Jolla, CA, USA
| | - Ruotao Wang
- Department of Nanoengineering, University of California San Diego, La Jolla, CA, USA
| | - Joseph Wang
- Department of Nanoengineering, University of California San Diego, La Jolla, CA, USA
| | | | - Erik B Kistler
- Department of Bioengineering, University of California San Diego, La Jolla, CA, USA
- Department of Anesthesiology and Critical Care, University of California San Diego, La Jolla, CA, USA
| | - Nuno Vasconcelos
- Department of Electrical and Computer Engineering, University of California San Diego, La Jolla, CA, USA
| | - Sheng Xu
- Department of Nanoengineering, University of California San Diego, La Jolla, CA, USA.
- Materials Science and Engineering Program, University of California San Diego, La Jolla, CA, USA.
- Department of Electrical and Computer Engineering, University of California San Diego, La Jolla, CA, USA.
- Department of Bioengineering, University of California San Diego, La Jolla, CA, USA.
- Department of Radiology, School of Medicine, University of California San Diego, La Jolla, CA, USA.
| |
Collapse
|
6
|
Zhou J, Dong J, Hou H, Huang L, Li J. High-throughput microfluidic systems accelerated by artificial intelligence for biomedical applications. LAB ON A CHIP 2024; 24:1307-1326. [PMID: 38247405 DOI: 10.1039/d3lc01012k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/23/2024]
Abstract
High-throughput microfluidic systems are widely used in biomedical fields for tasks like disease detection, drug testing, and material discovery. Despite the great advances in automation and throughput, the large amounts of data generated by the high-throughput microfluidic systems generally outpace the abilities of manual analysis. Recently, the convergence of microfluidic systems and artificial intelligence (AI) has been promising in solving the issue by significantly accelerating the process of data analysis as well as improving the capability of intelligent decision. This review offers a comprehensive introduction on AI methods and outlines the current advances of high-throughput microfluidic systems accelerated by AI, covering biomedical detection, drug screening, and automated system control and design. Furthermore, the challenges and opportunities in this field are critically discussed as well.
Collapse
Affiliation(s)
- Jianhua Zhou
- School of Biomedical Engineering, Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, China.
- Key Laboratory of Sensing Technology and Biomedical Instruments of Guangdong Province, School of Biomedical Engineering, Sun Yat-sen University, Shenzhen 518107, China
| | - Jianpei Dong
- School of Biomedical Engineering, Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, China.
- Key Laboratory of Sensing Technology and Biomedical Instruments of Guangdong Province, School of Biomedical Engineering, Sun Yat-sen University, Shenzhen 518107, China
| | - Hongwei Hou
- Beijing Life Science Academy, Beijing 102209, China
| | - Lu Huang
- School of Biomedical Engineering, Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, China.
- Key Laboratory of Sensing Technology and Biomedical Instruments of Guangdong Province, School of Biomedical Engineering, Sun Yat-sen University, Shenzhen 518107, China
| | - Jinghong Li
- Department of Chemistry, Center for BioAnalytical Chemistry, Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Tsinghua University, Beijing 100084, China.
- New Cornerstone Science Laboratory, Shenzhen 518054, China
- Beijing Life Science Academy, Beijing 102209, China
- Center for BioAnalytical Chemistry, Hefei National Laboratory of Physical Science at Microscale, University of Science and Technology of China, Hefei 230026, China
| |
Collapse
|
7
|
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
|
8
|
Betts R, Dierking I. Machine learning classification of polar sub-phases in liquid crystal MHPOBC. SOFT MATTER 2023; 19:7502-7512. [PMID: 37646209 DOI: 10.1039/d3sm00902e] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/01/2023]
Abstract
Experimental polarising microscopy texture images of the fluid smectic phases and sub-phases of the classic liquid crystal MHPOBC were classified as paraelectric (SmA*), ferroelectric (SmC*), ferrielectric (SmC1/3*), and antiferroelectric (SmCA*) using convolutional neural networks, CNNs. Two neural network architectures were tested, a sequential convolutional neural network with varying numbers of layers and a simplified inception model with varying number of inception blocks. Both models are successful in binary classifications between different phases as well as classification between all four phases. Optimised architectures for the multi-phase classification achieved accuracies of (84 ± 2)% and (93 ± 1)% for sequential convolutional and inception networks, respectively. The results of this study contribute to the understanding of how CNNs may be used in classifying liquid crystal phases. Especially the inception model is of sufficient accuracy to allow automated characterization of liquid crystal phase sequences and thus opens a path towards an additional method to determine the phases of novel liquid crystals for applications in electro-optics, photonics or sensors. The outlined procedure of supervised machine learning can be applied to practically all liquid crystal phases and materials, provided the infrastructure of training data and computational power is provided.
Collapse
Affiliation(s)
- Rebecca Betts
- Department of Physics and Astronomy, University of Manchester, Oxford Road, Manchester M139PL, UK.
| | - Ingo Dierking
- Department of Physics and Astronomy, University of Manchester, Oxford Road, Manchester M139PL, UK.
| |
Collapse
|
9
|
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
|
10
|
Gao Z, Li Y. Enhancing single-cell biology through advanced AI-powered microfluidics. BIOMICROFLUIDICS 2023; 17:051301. [PMID: 37799809 PMCID: PMC10550334 DOI: 10.1063/5.0170050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Accepted: 09/23/2023] [Indexed: 10/07/2023]
Abstract
Microfluidic technology has largely benefited both fundamental biological research and translational clinical diagnosis with its advantages in high-throughput, single-cell resolution, high integrity, and wide-accessibility. Despite the merits we obtained from microfluidics in the last two decades, the current requirement of intelligence in biomedicine urges the microfluidic technology to process biological big data more efficiently and intelligently. Thus, the current readout technology based on the direct detection of the signals in either optics or electrics was not able to meet the requirement. The implementation of artificial intelligence (AI) in microfluidic technology matches up with the large-scale data usually obtained in the high-throughput assays of microfluidics. At the same time, AI is able to process the multimodal datasets obtained from versatile microfluidic devices, including images, videos, electric signals, and sequences. Moreover, AI provides the microfluidic technology with the capability to understand and decipher the obtained datasets rather than simply obtaining, which eventually facilitates fundamental and translational research in many areas, including cell type discovery, cell signaling, single-cell genetics, and diagnosis. In this Perspective, we will highlight the recent advances in employing AI for single-cell biology and present an outlook on the future direction with more advanced AI algorithms.
Collapse
Affiliation(s)
- Zhaolong Gao
- The Key Laboratory for Biomedical Photonics of MOE at Wuhan National Laboratory for Optoelectronics—Hubei Bioinformatics and Molecular Imaging Key Laboratory, Department of Biomedical Engineering, Systems Biology Theme, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Yiwei Li
- The Key Laboratory for Biomedical Photonics of MOE at Wuhan National Laboratory for Optoelectronics—Hubei Bioinformatics and Molecular Imaging Key Laboratory, Department of Biomedical Engineering, Systems Biology Theme, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| |
Collapse
|
11
|
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
|
12
|
Goda K, Lu H, Fei P, Guck J. Revolutionizing microfluidics with artificial intelligence: a new dawn for lab-on-a-chip technologies. LAB ON A CHIP 2023; 23:3737-3740. [PMID: 37503818 DOI: 10.1039/d3lc90061d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/29/2023]
Abstract
Keisuke Goda, Hang Lu, Peng Fei, and Jochen Guck introduce the AI in Microfluidics themed collection, on revolutionizing microfluidics with artificial intelligence: a new dawn for lab-on-a-chip technologies.
Collapse
Affiliation(s)
- Keisuke Goda
- Department of Chemistry, 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
| | - Hang Lu
- School of Chemical and Biomolecular Engineering, Georgia Institute of Technology, Atlanta, Georgia 30332, USA
- Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, Georgia 30332, USA
| | - Peng Fei
- School of Optical and Electronic Information, Wuhan National Laboratory for Optoelectronics, Advanced Biomedical Imaging Facility, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Jochen Guck
- Max Planck Institute for the Science of Light and Max-Planck-Zentrum für Physik und Medizin, Erlangen, Germany
| |
Collapse
|
13
|
Mazuryk J, Klepacka K, Kutner W, Sharma PS. Glyphosate Separating and Sensing for Precision Agriculture and Environmental Protection in the Era of Smart Materials. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2023. [PMID: 37384557 DOI: 10.1021/acs.est.3c01269] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/01/2023]
Abstract
The present article critically and comprehensively reviews the most recent reports on smart sensors for determining glyphosate (GLP), an active agent of GLP-based herbicides (GBHs) traditionally used in agriculture over the past decades. Commercialized in 1974, GBHs have now reached 350 million hectares of crops in over 140 countries with an annual turnover of 11 billion USD worldwide. However, rolling exploitation of GLP and GBHs in the last decades has led to environmental pollution, animal intoxication, bacterial resistance, and sustained occupational exposure of the herbicide of farm and companies' workers. Intoxication with these herbicides dysregulates the microbiome-gut-brain axis, cholinergic neurotransmission, and endocrine system, causing paralytic ileus, hyperkalemia, oliguria, pulmonary edema, and cardiogenic shock. Precision agriculture, i.e., an (information technology)-enhanced approach to crop management, including a site-specific determination of agrochemicals, derives from the benefits of smart materials (SMs), data science, and nanosensors. Those typically feature fluorescent molecularly imprinted polymers or immunochemical aptamer artificial receptors integrated with electrochemical transducers. Fabricated as portable or wearable lab-on-chips, smartphones, and soft robotics and connected with SM-based devices that provide machine learning algorithms and online databases, they integrate, process, analyze, and interpret massive amounts of spatiotemporal data in a user-friendly and decision-making manner. Exploited for the ultrasensitive determination of toxins, including GLP, they will become practical tools in farmlands and point-of-care testing. Expectedly, smart sensors can be used for personalized diagnostics, real-time water, food, soil, and air quality monitoring, site-specific herbicide management, and crop control.
Collapse
Affiliation(s)
- Jarosław Mazuryk
- Department of Electrode Processes, Institute of Physical Chemistry, Polish Academy of Sciences, 01-224 Warsaw, Poland
- Bio & Soft Matter, Institute of Condensed Matter and Nanosciences, Université catholique de Louvain, 1 Place Louis Pasteur, 1348 Louvain-la-Neuve, Belgium
| | - Katarzyna Klepacka
- Functional Polymers Research Team, Institute of Physical Chemistry, Polish Academy of Sciences, 01-224 Warsaw, Poland
- ENSEMBLE3 sp. z o. o., 01-919 Warsaw, Poland
- Faculty of Mathematics and Natural Sciences. School of Sciences, Cardinal Stefan Wyszynski University in Warsaw, 01-938 Warsaw, Poland
| | - Włodzimierz Kutner
- Faculty of Mathematics and Natural Sciences. School of Sciences, Cardinal Stefan Wyszynski University in Warsaw, 01-938 Warsaw, Poland
- Modified Electrodes for Potential Application in Sensors and Cells Research Team, Institute of Physical Chemistry, Polish Academy of Sciences, 01-224 Warsaw, Poland
| | - Piyush Sindhu Sharma
- Functional Polymers Research Team, Institute of Physical Chemistry, Polish Academy of Sciences, 01-224 Warsaw, Poland
| |
Collapse
|
14
|
Ferguson C, Zhang Y, Palego C, Cheng X. Recent Approaches to Design and Analysis of Electrical Impedance Systems for Single Cells Using Machine Learning. SENSORS (BASEL, SWITZERLAND) 2023; 23:5990. [PMID: 37447838 DOI: 10.3390/s23135990] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Revised: 06/17/2023] [Accepted: 06/26/2023] [Indexed: 07/15/2023]
Abstract
Individual cells have many unique properties that can be quantified to develop a holistic understanding of a population. This can include understanding population characteristics, identifying subpopulations, or elucidating outlier characteristics that may be indicators of disease. Electrical impedance measurements are rapid and label-free for the monitoring of single cells and generate large datasets of many cells at single or multiple frequencies. To increase the accuracy and sensitivity of measurements and define the relationships between impedance and biological features, many electrical measurement systems have incorporated machine learning (ML) paradigms for control and analysis. Considering the difficulty capturing complex relationships using traditional modelling and statistical methods due to population heterogeneity, ML offers an exciting approach to the systemic collection and analysis of electrical properties in a data-driven way. In this work, we discuss incorporation of ML to improve the field of electrical single cell analysis by addressing the design challenges to manipulate single cells and sophisticated analysis of electrical properties that distinguish cellular changes. Looking forward, we emphasize the opportunity to build on integrated systems to address common challenges in data quality and generalizability to save time and resources at every step in electrical measurement of single cells.
Collapse
Affiliation(s)
- Caroline Ferguson
- Department of Bioengineering, Lehigh University, Bethlehem, PA 18015, USA
| | - Yu Zhang
- Department of Bioengineering, Lehigh University, Bethlehem, PA 18015, USA
| | - Cristiano Palego
- Department of Computer Science and Electronic Engineering, Bangor University, Bangor LL57 2DG, UK
| | - Xuanhong Cheng
- Department of Bioengineering, Lehigh University, Bethlehem, PA 18015, USA
- Department of Materials Science and Engineering, Lehigh University, Bethlehem, PA 18015, USA
| |
Collapse
|
15
|
Sun H, Xie W, Mo J, Huang Y, Dong H. Deep learning with microfluidics for on-chip droplet generation, control, and analysis. Front Bioeng Biotechnol 2023; 11:1208648. [PMID: 37351472 PMCID: PMC10282949 DOI: 10.3389/fbioe.2023.1208648] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Accepted: 05/25/2023] [Indexed: 06/24/2023] Open
Abstract
Droplet microfluidics has gained widespread attention in recent years due to its advantages of high throughput, high integration, high sensitivity and low power consumption in droplet-based micro-reaction. Meanwhile, with the rapid development of computer technology over the past decade, deep learning architectures have been able to process vast amounts of data from various research fields. Nowadays, interdisciplinarity plays an increasingly important role in modern research, and deep learning has contributed greatly to the advancement of many professions. Consequently, intelligent microfluidics has emerged as the times require, and possesses broad prospects in the development of automated and intelligent devices for integrating the merits of microfluidic technology and artificial intelligence. In this article, we provide a general review of the evolution of intelligent microfluidics and some applications related to deep learning, mainly in droplet generation, control, and analysis. We also present the challenges and emerging opportunities in this field.
Collapse
Affiliation(s)
- Hao Sun
- School of Mechanical Engineering and Automation, Fuzhou University, Fuzhou, China
- Fujian Provincial Collaborative Innovation Center of High-End Equipment Manufacturing, Fuzhou, China
| | - Wantao Xie
- School of Mechanical Engineering and Automation, Fuzhou University, Fuzhou, China
- Fujian Provincial Collaborative Innovation Center of High-End Equipment Manufacturing, Fuzhou, China
| | - Jin Mo
- School of Mechanical Engineering and Automation, Fuzhou University, Fuzhou, China
- Fujian Provincial Collaborative Innovation Center of High-End Equipment Manufacturing, Fuzhou, China
| | - Yi Huang
- Centre for Experimental Research in Clinical Medicine, Fujian Provincial Hospital, Fuzhou, China
| | - Hui Dong
- School of Mechanical Engineering and Automation, Fuzhou University, Fuzhou, China
- Fujian Provincial Collaborative Innovation Center of High-End Equipment Manufacturing, Fuzhou, China
| |
Collapse
|
16
|
Sabaté Del Río J, Ro J, Yoon H, Park TE, Cho YK. Integrated technologies for continuous monitoring of organs-on-chips: Current challenges and potential solutions. Biosens Bioelectron 2023; 224:115057. [PMID: 36640548 DOI: 10.1016/j.bios.2022.115057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 12/29/2022] [Accepted: 12/30/2022] [Indexed: 01/03/2023]
Abstract
Organs-on-chips (OoCs) are biomimetic in vitro systems based on microfluidic cell cultures that recapitulate the in vivo physicochemical microenvironments and the physiologies and key functional units of specific human organs. These systems are versatile and can be customized to investigate organ-specific physiology, pathology, or pharmacology. They are more physiologically relevant than traditional two-dimensional cultures, can potentially replace the animal models or reduce the use of these models, and represent a unique opportunity for the development of personalized medicine when combined with human induced pluripotent stem cells. Continuous monitoring of important quality parameters of OoCs via a label-free, non-destructive, reliable, high-throughput, and multiplex method is critical for assessing the conditions of these systems and generating relevant analytical data; moreover, elaboration of quality predictive models is required for clinical trials of OoCs. Presently, these analytical data are obtained by manual or automatic sampling and analyzed using single-point, off-chip traditional methods. In this review, we describe recent efforts to integrate biosensing technologies into OoCs for monitoring the physiologies, functions, and physicochemical microenvironments of OoCs. Furthermore, we present potential alternative solutions to current challenges and future directions for the application of artificial intelligence in the development of OoCs and cyber-physical systems. These "smart" OoCs can learn and make autonomous decisions for process optimization, self-regulation, and data analysis.
Collapse
Affiliation(s)
- Jonathan Sabaté Del Río
- Center for Soft and Living Matter, Institute for Basic Science (IBS), Ulsan, 44919, Republic of Korea
| | - Jooyoung Ro
- Center for Soft and Living Matter, Institute for Basic Science (IBS), Ulsan, 44919, Republic of Korea; Department of Biomedical Engineering, Ulsan National Institute of Science and Technology (UNIST), Ulsan, 44919, Republic of Korea
| | - Heejeong Yoon
- Department of Biomedical Engineering, Ulsan National Institute of Science and Technology (UNIST), Ulsan, 44919, Republic of Korea
| | - Tae-Eun Park
- Department of Biomedical Engineering, Ulsan National Institute of Science and Technology (UNIST), Ulsan, 44919, Republic of Korea.
| | - Yoon-Kyoung Cho
- Center for Soft and Living Matter, Institute for Basic Science (IBS), Ulsan, 44919, Republic of Korea; Department of Biomedical Engineering, Ulsan National Institute of Science and Technology (UNIST), Ulsan, 44919, Republic of Korea.
| |
Collapse
|
17
|
Wu J, Fang H, Zhang J, Yan S. Modular microfluidics for life sciences. J Nanobiotechnology 2023; 21:85. [PMID: 36906553 PMCID: PMC10008080 DOI: 10.1186/s12951-023-01846-x] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Accepted: 03/06/2023] [Indexed: 03/13/2023] Open
Abstract
The advancement of microfluidics has enabled numerous discoveries and technologies in life sciences. However, due to the lack of industry standards and configurability, the design and fabrication of microfluidic devices require highly skilled technicians. The diversity of microfluidic devices discourages biologists and chemists from applying this technique in their laboratories. Modular microfluidics, which integrates the standardized microfluidic modules into a whole, complex platform, brings the capability of configurability to conventional microfluidics. The exciting features, including portability, on-site deployability, and high customization motivate us to review the state-of-the-art modular microfluidics and discuss future perspectives. In this review, we first introduce the working mechanisms of the basic microfluidic modules and evaluate their feasibility as modular microfluidic components. Next, we explain the connection approaches among these microfluidic modules, and summarize the advantages of modular microfluidics over integrated microfluidics in biological applications. Finally, we discuss the challenge and future perspectives of modular microfluidics.
Collapse
Affiliation(s)
- Jialin Wu
- Institute for Advanced Study, Shenzhen University, Shenzhen, 518060, China
- Nanophotonics Research Center, Institute of Microscale Optoelectronics, Shenzhen University, Shenzhen, China
| | - Hui Fang
- Nanophotonics Research Center, Institute of Microscale Optoelectronics, Shenzhen University, Shenzhen, China
| | - Jun Zhang
- Queensland Micro and Nanotechnology Centre, Griffith University, Brisbane, QLD, 4111, Australia
| | - Sheng Yan
- Institute for Advanced Study, Shenzhen University, Shenzhen, 518060, China.
| |
Collapse
|
18
|
Siu DMD, Lee KCM, Chung BMF, Wong JSJ, Zheng G, Tsia KK. Optofluidic imaging meets deep learning: from merging to emerging. LAB ON A CHIP 2023; 23:1011-1033. [PMID: 36601812 DOI: 10.1039/d2lc00813k] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
Propelled by the striking advances in optical microscopy and deep learning (DL), the role of imaging in lab-on-a-chip has dramatically been transformed from a silo inspection tool to a quantitative "smart" engine. A suite of advanced optical microscopes now enables imaging over a range of spatial scales (from molecules to organisms) and temporal window (from microseconds to hours). On the other hand, the staggering diversity of DL algorithms has revolutionized image processing and analysis at the scale and complexity that were once inconceivable. Recognizing these exciting but overwhelming developments, we provide a timely review of their latest trends in the context of lab-on-a-chip imaging, or coined optofluidic imaging. More importantly, here we discuss the strengths and caveats of how to adopt, reinvent, and integrate these imaging techniques and DL algorithms in order to tailor different lab-on-a-chip applications. In particular, we highlight three areas where the latest advances in lab-on-a-chip imaging and DL can form unique synergisms: image formation, image analytics and intelligent image-guided autonomous lab-on-a-chip. Despite the on-going challenges, we anticipate that they will represent the next frontiers in lab-on-a-chip imaging that will spearhead new capabilities in advancing analytical chemistry research, accelerating biological discovery, and empowering new intelligent clinical applications.
Collapse
Affiliation(s)
- Dickson M D Siu
- Department of Electrical and Electronic Engineering, The University of Hong Kong, Hong Kong, Hong Kong.
| | - Kelvin C M Lee
- Department of Electrical and Electronic Engineering, The University of Hong Kong, Hong Kong, Hong Kong.
| | - Bob M F Chung
- Advanced Biomedical Instrumentation Centre, Hong Kong Science Park, Shatin, New Territories, Hong Kong
| | - Justin S J Wong
- Conzeb Limited, Hong Kong Science Park, Shatin, New Territories, Hong Kong
| | - Guoan Zheng
- Department of Biomedical Engineering, University of Connecticut, Storrs, CT, USA
| | - Kevin K Tsia
- Department of Electrical and Electronic Engineering, The University of Hong Kong, Hong Kong, Hong Kong.
- Advanced Biomedical Instrumentation Centre, Hong Kong Science Park, Shatin, New Territories, Hong Kong
| |
Collapse
|
19
|
Lu N, Tay HM, Petchakup C, He L, Gong L, Maw KK, Leong SY, Lok WW, Ong HB, Guo R, Li KHH, Hou HW. Label-free microfluidic cell sorting and detection for rapid blood analysis. LAB ON A CHIP 2023; 23:1226-1257. [PMID: 36655549 DOI: 10.1039/d2lc00904h] [Citation(s) in RCA: 27] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
Blood tests are considered as standard clinical procedures to screen for markers of diseases and health conditions. However, the complex cellular background (>99.9% RBCs) and biomolecular composition often pose significant technical challenges for accurate blood analysis. An emerging approach for point-of-care blood diagnostics is utilizing "label-free" microfluidic technologies that rely on intrinsic cell properties for blood fractionation and disease detection without any antibody binding. A growing body of clinical evidence has also reported that cellular dysfunction and their biophysical phenotypes are complementary to standard hematoanalyzer analysis (complete blood count) and can provide a more comprehensive health profiling. In this review, we will summarize recent advances in microfluidic label-free separation of different blood cell components including circulating tumor cells, leukocytes, platelets and nanoscale extracellular vesicles. Label-free single cell analysis of intrinsic cell morphology, spectrochemical properties, dielectric parameters and biophysical characteristics as novel blood-based biomarkers will also be presented. Next, we will highlight research efforts that combine label-free microfluidics with machine learning approaches to enhance detection sensitivity and specificity in clinical studies, as well as innovative microfluidic solutions which are capable of fully integrated and label-free blood cell sorting and analysis. Lastly, we will envisage the current challenges and future outlook of label-free microfluidics platforms for high throughput multi-dimensional blood cell analysis to identify non-traditional circulating biomarkers for clinical diagnostics.
Collapse
Affiliation(s)
- Nan Lu
- School of Mechanical and Aerospace Engineering, Nanyang Technological University, 50 Nanyang Avenue, Blk N3, Level 2, Room 86 (N3-02c-86), 639798, Singapore.
- HP-NTU Digital Manufacturing Corporate Lab, Nanyang Technological University, 65 Nanyang Drive, Block N3, 637460, Singapore
| | - Hui Min Tay
- School of Mechanical and Aerospace Engineering, Nanyang Technological University, 50 Nanyang Avenue, Blk N3, Level 2, Room 86 (N3-02c-86), 639798, Singapore.
| | - Chayakorn Petchakup
- School of Mechanical and Aerospace Engineering, Nanyang Technological University, 50 Nanyang Avenue, Blk N3, Level 2, Room 86 (N3-02c-86), 639798, Singapore.
| | - Linwei He
- School of Mechanical and Aerospace Engineering, Nanyang Technological University, 50 Nanyang Avenue, Blk N3, Level 2, Room 86 (N3-02c-86), 639798, Singapore.
| | - Lingyan Gong
- School of Mechanical and Aerospace Engineering, Nanyang Technological University, 50 Nanyang Avenue, Blk N3, Level 2, Room 86 (N3-02c-86), 639798, Singapore.
| | - Kay Khine Maw
- School of Mechanical and Aerospace Engineering, Nanyang Technological University, 50 Nanyang Avenue, Blk N3, Level 2, Room 86 (N3-02c-86), 639798, Singapore.
| | - Sheng Yuan Leong
- School of Mechanical and Aerospace Engineering, Nanyang Technological University, 50 Nanyang Avenue, Blk N3, Level 2, Room 86 (N3-02c-86), 639798, Singapore.
| | - Wan Wei Lok
- School of Mechanical and Aerospace Engineering, Nanyang Technological University, 50 Nanyang Avenue, Blk N3, Level 2, Room 86 (N3-02c-86), 639798, Singapore.
| | - Hong Boon Ong
- School of Mechanical and Aerospace Engineering, Nanyang Technological University, 50 Nanyang Avenue, Blk N3, Level 2, Room 86 (N3-02c-86), 639798, Singapore.
| | - Ruya Guo
- Key Laboratory of Agricultural Information Acquisition Technology, Ministry of Agriculture and Rural Affairs, China Agricultural University, Beijing, 100083, China
| | - King Ho Holden Li
- School of Mechanical and Aerospace Engineering, Nanyang Technological University, 50 Nanyang Avenue, Blk N3, Level 2, Room 86 (N3-02c-86), 639798, Singapore.
- HP-NTU Digital Manufacturing Corporate Lab, Nanyang Technological University, 65 Nanyang Drive, Block N3, 637460, Singapore
| | - Han Wei Hou
- School of Mechanical and Aerospace Engineering, Nanyang Technological University, 50 Nanyang Avenue, Blk N3, Level 2, Room 86 (N3-02c-86), 639798, Singapore.
- HP-NTU Digital Manufacturing Corporate Lab, Nanyang Technological University, 65 Nanyang Drive, Block N3, 637460, Singapore
- Lee Kong Chian School of Medicine, Nanyang Technological University, 11 Mandalay Road, Clinical Sciences Building, 308232, Singapore
| |
Collapse
|
20
|
Zhao Y, Isozaki A, Herbig M, Hayashi M, Hiramatsu K, Yamazaki S, Kondo N, Ohnuki S, Ohya Y, Nitta N, Goda K. Intelligent sort-timing prediction for image-activated cell sorting. Cytometry A 2023; 103:88-97. [PMID: 35766305 DOI: 10.1002/cyto.a.24664] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Revised: 05/20/2022] [Accepted: 06/11/2022] [Indexed: 02/07/2023]
Abstract
Intelligent image-activated cell sorting (iIACS) has enabled high-throughput image-based sorting of single cells with artificial intelligence (AI) algorithms. This AI-on-a-chip technology combines fluorescence microscopy, AI-based image processing, sort-timing prediction, and cell sorting. Sort-timing prediction is particularly essential due to the latency on the order of milliseconds between image acquisition and sort actuation, during which image processing is performed. The long latency amplifies the effects of the fluctuations in the flow speed of cells, leading to fluctuation and uncertainty in the arrival time of cells at the sort point on the microfluidic chip. To compensate for this fluctuation, iIACS measures the flow speed of each cell upstream, predicts the arrival timing of the cell at the sort point, and activates the actuation of the cell sorter appropriately. Here, we propose and demonstrate a machine learning technique to increase the accuracy of the sort-timing prediction that would allow for the improvement of sort event rate, yield, and purity. Specifically, we trained an algorithm to predict the sort timing for morphologically heterogeneous budding yeast cells. The algorithm we developed used cell morphology, position, and flow speed as inputs for prediction and achieved 41.5% lower prediction error compared to the previously employed method based solely on flow speed. As a result, our technique would allow for an increase in the sort event rate of iIACS by a factor of ~2.
Collapse
Affiliation(s)
- Yaqi Zhao
- Department of Chemistry, Graduate School of Science, The University of Tokyo, Tokyo, Japan
| | - Akihiro Isozaki
- Department of Chemistry, Graduate School of Science, The University of Tokyo, Tokyo, Japan
| | - Maik Herbig
- Department of Chemistry, Graduate School of Science, The University of Tokyo, Tokyo, Japan
| | - Mika Hayashi
- Department of Chemistry, Graduate School of Science, The University of Tokyo, Tokyo, Japan
| | - Kotaro Hiramatsu
- Department of Chemistry, Graduate School of Science, The University of Tokyo, Tokyo, Japan
| | - Sota Yamazaki
- Department of Chemistry, Graduate School of Science, The University of Tokyo, Tokyo, Japan
| | - Naoko Kondo
- Department of Integrated Biosciences, Graduate School of Frontier Sciences, The University of Tokyo, Chiba, Japan
| | - Shinsuke Ohnuki
- Department of Integrated Biosciences, Graduate School of Frontier Sciences, The University of Tokyo, Chiba, Japan
| | - Yoshikazu Ohya
- Department of Integrated Biosciences, Graduate School of Frontier Sciences, The University of Tokyo, Chiba, Japan.,Collaborative Research Institute for Innovative Microbiology, The University of Tokyo, Tokyo, Japan
| | | | - Keisuke Goda
- Department of Chemistry, Graduate School of Science, The University of Tokyo, Tokyo, Japan.,CYBO, Tokyo, Japan.,Department of Bioengineering, University of California, California, Los Angeles, USA.,Institute of Technological Sciences, Wuhan University, Hubei, China
| |
Collapse
|
21
|
Tang R, Xia L, Gutierrez B, Gagne I, Munoz A, Eribez K, Jagnandan N, Chen X, Zhang Z, Waller L, Alaynick W, Cho SH, An C, Lo YH. Low-latency label-free image-activated cell sorting using fast deep learning and AI inferencing. Biosens Bioelectron 2023; 220:114865. [DOI: 10.1016/j.bios.2022.114865] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 09/27/2022] [Accepted: 10/25/2022] [Indexed: 11/09/2022]
|
22
|
Harmon J, Findinier J, Ishii NT, Herbig M, Isozaki A, Grossman A, Goda K. Intelligent image-activated sorting of Chlamydomonas reinhardtii by mitochondrial localization. Cytometry A 2022; 101:1027-1034. [PMID: 35643943 DOI: 10.1002/cyto.a.24661] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Revised: 05/22/2022] [Accepted: 05/24/2022] [Indexed: 01/27/2023]
Abstract
Organelle positioning in cells is associated with various metabolic functions and signaling in unicellular organisms. Specifically, the microalga Chlamydomonas reinhardtii repositions its mitochondria, depending on the levels of inorganic carbon. Mitochondria are typically randomly distributed in the Chlamydomonas cytoplasm, but relocate toward the cell periphery at low inorganic carbon levels. This mitochondrial relocation is linked with the carbon-concentrating mechanism, but its significance is not yet thoroughly understood. A genotypic understanding of this relocation would require a high-throughput method to isolate rare mutant cells not exhibiting this relocation. However, this task is technically challenging due to the complex intracellular morphological difference between mutant and wild-type cells, rendering conventional non-image-based high-event-rate methods unsuitable. Here, we report our demonstration of intelligent image-activated cell sorting by mitochondrial localization. Specifically, we applied an intelligent image-activated cell sorting system to sort for C. reinhardtii cells displaying no mitochondrial relocation. We trained a convolutional neural network (CNN) to distinguish the cell types based on the complex morphology of their mitochondria. The CNN was employed to perform image-activated sorting for the mutant cell type at 180 events per second, which is 1-2 orders of magnitude faster than automated microscopy with robotic pipetting, resulting in an enhancement of the concentration from 5% to 56.5% corresponding to an enrichment factor of 11.3. These results show the potential of image-activated cell sorting for connecting genotype-phenotype relations for rare-cell populations, which require a high throughput and could lead to a better understanding of metabolic functions in cells.
Collapse
Affiliation(s)
- Jeffrey Harmon
- Department of Chemistry, The University of Tokyo, Tokyo, Japan
| | - Justin Findinier
- Department of Plant Biology, Carnegie Institution for Science, Stanford, California, USA
| | | | - Maik Herbig
- Department of Chemistry, The University of Tokyo, Tokyo, Japan
| | - Akihiro Isozaki
- Department of Chemistry, The University of Tokyo, Tokyo, Japan
| | - Arthur Grossman
- Department of Plant Biology, Carnegie Institution for Science, Stanford, California, USA.,Department of Biology, Stanford University, Stanford, California, USA
| | - Keisuke Goda
- Department of Chemistry, The University of Tokyo, Tokyo, Japan.,Department of Bioengineering, University of California, California, Los Angeles, USA.,Institute of Technological Sciences, Wuhan University, Hubei, China
| |
Collapse
|
23
|
Sinha Mahapatra P, Ganguly R, Ghosh A, Chatterjee S, Lowrey S, Sommers AD, Megaridis CM. Patterning Wettability for Open-Surface Fluidic Manipulation: Fundamentals and Applications. Chem Rev 2022; 122:16752-16801. [PMID: 36195098 DOI: 10.1021/acs.chemrev.2c00045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Effective manipulation of liquids on open surfaces without external energy input is indispensable for the advancement of point-of-care diagnostic devices. Open-surface microfluidics has the potential to benefit health care, especially in the developing world. This review highlights the prospects for harnessing capillary forces on surface-microfluidic platforms, chiefly by inducing smooth gradients or sharp steps of wettability on substrates, to elicit passive liquid transport and higher-order fluidic manipulations without off-the-chip energy sources. A broad spectrum of the recent progress in the emerging field of passive surface microfluidics is highlighted, and its promise for developing facile, low-cost, easy-to-operate microfluidic devices is discussed in light of recent applications, not only in the domain of biomedical microfluidics but also in the general areas of energy and water conservation.
Collapse
Affiliation(s)
- Pallab Sinha Mahapatra
- Micro Nano Bio-Fluidics group, Department of Mechanical Engineering, Indian Institute of Technology Madras, Chennai600036, India
| | - Ranjan Ganguly
- Department of Power Engineering, Jadavpur University, Kolkata700098, India
| | - Aritra Ghosh
- Department of Mechanical and Industrial Engineering, University of Illinois at Chicago, Chicago, Illinois60607, United States
| | - Souvick Chatterjee
- Department of Mechanical and Industrial Engineering, University of Illinois at Chicago, Chicago, Illinois60607, United States
| | - Sam Lowrey
- Department of Physics, University of Otago, Dunedin9016, New Zealand
| | - Andrew D Sommers
- Department of Mechanical and Manufacturing Engineering, Miami University, Oxford, Ohio45056, United States
| | - Constantine M Megaridis
- Department of Mechanical and Industrial Engineering, University of Illinois at Chicago, Chicago, Illinois60607, United States
| |
Collapse
|
24
|
Zare Harofte S, Soltani M, Siavashy S, Raahemifar K. Recent Advances of Utilizing Artificial Intelligence in Lab on a Chip for Diagnosis and Treatment. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2022; 18:e2203169. [PMID: 36026569 DOI: 10.1002/smll.202203169] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/21/2022] [Revised: 07/16/2022] [Indexed: 05/14/2023]
Abstract
Nowadays, artificial intelligence (AI) creates numerous promising opportunities in the life sciences. AI methods can be significantly advantageous for analyzing the massive datasets provided by biotechnology systems for biological and biomedical applications. Microfluidics, with the developments in controlled reaction chambers, high-throughput arrays, and positioning systems, generate big data that is not necessarily analyzed successfully. Integrating AI and microfluidics can pave the way for both experimental and analytical throughputs in biotechnology research. Microfluidics enhances the experimental methods and reduces the cost and scale, while AI methods significantly improve the analysis of huge datasets obtained from high-throughput and multiplexed microfluidics. This review briefly presents a survey of the role of AI and microfluidics in biotechnology. Also, the incorporation of AI with microfluidics is comprehensively investigated. Specifically, recent studies that perform flow cytometry cell classification, cell isolation, and a combination of them by gaining from both AI methods and microfluidic techniques are covered. Despite all current challenges, various fields of biotechnology can be remarkably affected by the combination of AI and microfluidic technologies. Some of these fields include point-of-care systems, precision, personalized medicine, regenerative medicine, prognostics, diagnostics, and treatment of oncology and non-oncology-related diseases.
Collapse
Affiliation(s)
- Samaneh Zare Harofte
- Department of Mechanical Engineering, K. N. Toosi University of Technology, Tehran, 19967-15433, Iran
| | - Madjid Soltani
- Department of Mechanical Engineering, K. N. Toosi University of Technology, Tehran, 19967-15433, Iran
- Department of Electrical and Computer Engineering, Faculty of Engineering, University of Waterloo, Waterloo, ON, N2L 3G1, Canada
- Centre for Biotechnology and Bioengineering (CBB), University of Waterloo, Waterloo, ON, N2L 3G1, Canada
- Advanced Bioengineering Initiative Center, Multidisciplinary International Complex, K. N. Toosi University of Technology, Tehran, 14176-14411, Iran
- Cancer Biology Research Center, Cancer Institute of Iran, Tehran University of Medical Sciences, Tehran, 14197-33141, Iran
| | - Saeed Siavashy
- Department of Mechanical Engineering, K. N. Toosi University of Technology, Tehran, 19967-15433, Iran
| | - Kaamran Raahemifar
- Data Science and Artificial Intelligence Program, College of Information Sciences and Technology (IST), Penn State University, State College, PA, 16801, USA
- School of Optometry and Vision Science, Faculty of Science, University of Waterloo, 200 University Ave. W, Waterloo, ON, N2L 3G1, Canada
- Department of Chemical Engineering, Faculty of Engineering, University of Waterloo, 200 University Ave. W, Waterloo, ON, N2L 3G1, Canada
| |
Collapse
|
25
|
McIntyre D, Lashkaripour A, Fordyce P, Densmore D. Machine learning for microfluidic design and control. LAB ON A CHIP 2022; 22:2925-2937. [PMID: 35904162 PMCID: PMC9361804 DOI: 10.1039/d2lc00254j] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/19/2022] [Accepted: 06/28/2022] [Indexed: 05/24/2023]
Abstract
Microfluidics has developed into a mature field with applications across science and engineering, having particular commercial success in molecular diagnostics, next-generation sequencing, and bench-top analysis. Despite its ubiquity, the complexity of designing and controlling custom microfluidic devices present major barriers to adoption, requiring intuitive knowledge gained from years of experience. If these barriers were overcome, microfluidics could miniaturize biological and chemical research for non-experts through fully-automated platform development and operation. The intuition of microfluidic experts can be captured through machine learning, where complex statistical models are trained for pattern recognition and subsequently used for event prediction. Integration of machine learning with microfluidics could significantly expand its adoption and impact. Here, we present the current state of machine learning for the design and control of microfluidic devices, its possible applications, and current limitations.
Collapse
Affiliation(s)
- David McIntyre
- Biomedical Engineering Department, Boston University, MA, USA
- Biological Design Center, Boston University, Boston, MA, USA.
| | - Ali Lashkaripour
- Department of Bioengineering, Stanford University, Stanford, CA, USA
- Department of Genetics, Stanford University, Stanford, CA, USA
| | - Polly Fordyce
- Department of Bioengineering, Stanford University, Stanford, CA, USA
- Department of Genetics, Stanford University, Stanford, CA, USA
- Chan-Zuckerberg Biohub, San Francisco, CA, USA
| | - Douglas Densmore
- Biological Design Center, Boston University, Boston, MA, USA.
- Electrical & Computer Engineering Department, Boston University, Boston, MA, USA
| |
Collapse
|
26
|
Zhang Y, Zhao Y, Cole T, Zheng J, Bayinqiaoge, Guo J, Tang SY. Microfluidic flow cytometry for blood-based biomarker analysis. Analyst 2022; 147:2895-2917. [PMID: 35611964 DOI: 10.1039/d2an00283c] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Flow cytometry has proven its capability for rapid and quantitative analysis of individual cells and the separation of targeted biological samples from others. The emerging microfluidics technology makes it possible to develop portable microfluidic diagnostic devices for point-of-care testing (POCT) applications. Microfluidic flow cytometry (MFCM), where flow cytometry and microfluidics are combined to achieve similar or even superior functionalities on microfluidic chips, provides a powerful single-cell characterisation and sorting tool for various biological samples. In recent years, researchers have made great progress in the development of the MFCM including focusing, detecting, and sorting subsystems, and its unique capabilities have been demonstrated in various biological applications. Moreover, liquid biopsy using blood can provide various physiological and pathological information. Thus, biomarkers from blood are regarded as meaningful circulating transporters of signal molecules or particles and have great potential to be used as non (or minimally)-invasive diagnostic tools. In this review, we summarise the recent progress of the key subsystems for MFCM and its achievements in blood-based biomarker analysis. Finally, foresight is offered to highlight the research challenges faced by MFCM in expanding into blood-based POCT applications, potentially yielding commercialisation opportunities.
Collapse
Affiliation(s)
- Yuxin Zhang
- Department of Electronic, Electrical and Systems Engineering, University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK.
| | - Ying Zhao
- National Chengdu Centre of Safety Evaluation of Drugs, West China Hospital of Sichuan University, Chengdu, China
| | - Tim Cole
- Department of Electronic, Electrical and Systems Engineering, University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK.
| | - Jiahao Zheng
- Department of Electronic, Electrical and Systems Engineering, University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK.
| | - Bayinqiaoge
- Department of Electronic, Electrical and Systems Engineering, University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK.
| | - Jinhong Guo
- The M.O.E. Key Laboratory of Laboratory Medical Diagnostics, The College of Laboratory Medicine, Chongqing Medical University, #1 Yixueyuan Road, Yuzhong District, Chongqing, 400016, China.
| | - Shi-Yang Tang
- Department of Electronic, Electrical and Systems Engineering, University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK.
| |
Collapse
|
27
|
Caselli F, Reale R, De Ninno A, Spencer D, Morgan H, Bisegna P. Deciphering impedance cytometry signals with neural networks. LAB ON A CHIP 2022; 22:1714-1722. [PMID: 35353108 DOI: 10.1039/d2lc00028h] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/10/2023]
Abstract
Microfluidic impedance cytometry is a label-free technique for high-throughput single-cell analysis. Multi-frequency impedance measurements provide data that allows full characterisation of cells, linking electrical phenotype to individual biophysical properties. To efficiently extract the information embedded in the electrical signals, potentially in real-time, tailored signal processing is needed. Artificial intelligence approaches provide a promising new direction. Here we demonstrate the ability of neural networks to decipher impedance cytometry signals in two challenging scenarios: (i) to determine the intrinsic dielectric properties of single cells directly from raw impedance data streams, (ii) to capture single-cell signals that are hidden in the measured signals of coincident cells. The accuracy of the results and the high processing speed (fractions of ms per cell) demonstrate that neural networks can have an important role in impedance-based single-cell analysis.
Collapse
Affiliation(s)
- Federica Caselli
- Department of Civil Engineering and Computer Science, University of Rome Tor Vergata, Rome, Italy.
| | - Riccardo Reale
- Center for Life Nano Science@Sapienza, Italian Institute of Technology (IIT), Rome, Italy
| | - Adele De Ninno
- Italian National Research Council - Institute for Photonics and Nanotechnologies (CNR - IFN), Rome, Italy
| | - Daniel Spencer
- School of Electronics and Computing Science, and, Institute for Life Sciences, University of Southampton, Highfield, Southampton, UK
| | - Hywel Morgan
- School of Electronics and Computing Science, and, Institute for Life Sciences, University of Southampton, Highfield, Southampton, UK
| | - Paolo Bisegna
- Department of Civil Engineering and Computer Science, University of Rome Tor Vergata, Rome, Italy.
| |
Collapse
|
28
|
Acharyya S, Nag S, Guha PK. Ultra-selective tin oxide-based chemiresistive gas sensor employing signal transform and machine learning techniques. Anal Chim Acta 2022; 1217:339996. [DOI: 10.1016/j.aca.2022.339996] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Accepted: 05/24/2022] [Indexed: 11/15/2022]
|
29
|
Serov N, Vinogradov V. Artificial intelligence to bring nanomedicine to life. Adv Drug Deliv Rev 2022; 184:114194. [PMID: 35283223 DOI: 10.1016/j.addr.2022.114194] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 03/04/2022] [Accepted: 03/07/2022] [Indexed: 12/13/2022]
Abstract
The technology of drug delivery systems (DDSs) has demonstrated an outstanding performance and effectiveness in production of pharmaceuticals, as it is proved by many FDA-approved nanomedicines that have an enhanced selectivity, manageable drug release kinetics and synergistic therapeutic actions. Nonetheless, to date, the rational design and high-throughput development of nanomaterial-based DDSs for specific purposes is far from a routine practice and is still in its infancy, mainly due to the limitations in scientists' capabilities to effectively acquire, analyze, manage, and comprehend complex and ever-growing sets of experimental data, which is vital to develop DDSs with a set of desired functionalities. At the same time, this task is feasible for the data-driven approaches, high throughput experimentation techniques, process automatization, artificial intelligence (AI) technology, and machine learning (ML) approaches, which is referred to as The Fourth Paradigm of scientific research. Therefore, an integration of these approaches with nanomedicine and nanotechnology can potentially accelerate the rational design and high-throughput development of highly efficient nanoformulated drugs and smart materials with pre-defined functionalities. In this Review, we survey the important results and milestones achieved to date in the application of data science, high throughput, as well as automatization approaches, combined with AI and ML to design and optimize DDSs and related nanomaterials. This manuscript mission is not only to reflect the state-of-art in data-driven nanomedicine, but also show how recent findings in the related fields can transform the nanomedicine's image. We discuss how all these results can be used to boost nanomedicine translation to the clinic, as well as highlight the future directions for the development, data-driven, high throughput experimentation-, and AI-assisted design, as well as the production of nanoformulated drugs and smart materials with pre-defined properties and behavior. This Review will be of high interest to the chemists involved in materials science, nanotechnology, and DDSs development for biomedical applications, although the general nature of the presented approaches enables knowledge translation to many other fields of science.
Collapse
Affiliation(s)
- Nikita Serov
- International Institute "Solution Chemistry of Advanced Materials and Technologies", ITMO University, Saint-Petersburg 191002, Russian Federation
| | - Vladimir Vinogradov
- International Institute "Solution Chemistry of Advanced Materials and Technologies", ITMO University, Saint-Petersburg 191002, Russian Federation.
| |
Collapse
|
30
|
Huang K, Matsumura H, Zhao Y, Herbig M, Yuan D, Mineharu Y, Harmon J, Findinier J, Yamagishi M, Ohnuki S, Nitta N, Grossman AR, Ohya Y, Mikami H, Isozaki A, Goda K. Deep imaging flow cytometry. LAB ON A CHIP 2022; 22:876-889. [PMID: 35142325 DOI: 10.1039/d1lc01043c] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Imaging flow cytometry (IFC) has become a powerful tool for diverse biomedical applications by virtue of its ability to image single cells in a high-throughput manner. However, there remains a challenge posed by the fundamental trade-off between throughput, sensitivity, and spatial resolution. Here we present deep-learning-enhanced imaging flow cytometry (dIFC) that circumvents this trade-off by implementing an image restoration algorithm on a virtual-freezing fluorescence imaging (VIFFI) flow cytometry platform, enabling higher throughput without sacrificing sensitivity and spatial resolution. A key component of dIFC is a high-resolution (HR) image generator that synthesizes "virtual" HR images from the corresponding low-resolution (LR) images acquired with a low-magnification lens (10×/0.4-NA). For IFC, a low-magnification lens is favorable because of reduced image blur of cells flowing at a higher speed, which allows higher throughput. We trained and developed the HR image generator with an architecture containing two generative adversarial networks (GANs). Furthermore, we developed dIFC as a method by combining the trained generator and IFC. We characterized dIFC using Chlamydomonas reinhardtii cell images, fluorescence in situ hybridization (FISH) images of Jurkat cells, and Saccharomyces cerevisiae (budding yeast) cell images, showing high similarities of dIFC images to images obtained with a high-magnification lens (40×/0.95-NA), at a high flow speed of 2 m s-1. We lastly employed dIFC to show enhancements in the accuracy of FISH-spot counting and neck-width measurement of budding yeast cells. These results pave the way for statistical analysis of cells with high-dimensional spatial information.
Collapse
Affiliation(s)
- Kangrui Huang
- Department of Chemistry, The University of Tokyo, Tokyo 113-0033, Japan.
| | - Hiroki Matsumura
- Department of Chemistry, The University of Tokyo, Tokyo 113-0033, Japan.
| | - Yaqi Zhao
- Department of Chemistry, The University of Tokyo, Tokyo 113-0033, Japan.
| | - Maik Herbig
- Department of Chemistry, The University of Tokyo, Tokyo 113-0033, Japan.
| | - Dan Yuan
- Department of Chemistry, The University of Tokyo, Tokyo 113-0033, Japan.
| | - Yohei Mineharu
- Department of Neurosurgery, Kyoto University, Kyoto 606-8507, Japan
- Department of Artificial Intelligence in Healthcare and Medicine, Kyoto University Graduate School of Medicine, Kyoto 606-8507, Japan
| | - Jeffrey Harmon
- Department of Chemistry, The University of Tokyo, Tokyo 113-0033, Japan.
| | - Justin Findinier
- Department of Plant Biology, The Carnegie Institution for Science, Stanford, California 94305, USA
| | - Mai Yamagishi
- Department of Biological Sciences, 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
| | | | - Arthur R Grossman
- Department of Plant Biology, The Carnegie Institution for Science, Stanford, California 94305, USA
- Department of Biology, Stanford University, Stanford, California 94305, USA
| | - 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
| | - Hideharu Mikami
- Department of Chemistry, The University of Tokyo, Tokyo 113-0033, Japan.
- PRESTO, Japan Science and Technology Agency, Saitama 332-0012, Japan
| | - Akihiro Isozaki
- Department of Chemistry, The University of Tokyo, Tokyo 113-0033, Japan.
| | - Keisuke Goda
- Department of Chemistry, 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, Hubei 430072, China
| |
Collapse
|
31
|
Bhuiyan NH, Hong JH, Uddin MJ, Shim JS. Artificial Intelligence-Controlled Microfluidic Device for Fluid Automation and Bubble Removal of Immunoassay Operated by a Smartphone. Anal Chem 2022; 94:3872-3880. [PMID: 35179372 DOI: 10.1021/acs.analchem.1c04827] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
There have been tremendous innovations in microfluidic clinical diagnostics to facilitate novel point-of-care testing (POCT) over the past decades. However, the automatic operation of microfluidic devices that minimize user intervention still lacks reliability and repeatability because microfluidic errors such as bubbles and incomplete filling pose a major bottleneck in commercializing the microfluidic devices for clinical testing. In this work, for the first time, various states of microfluid were recognized to control immunodiagnostics by artificial intelligence (AI) technology. The developed AI-controlled microfluidic platform was operated via an Android smartphone, along with a low-cost polymer device to effectuate enzyme-linked immunosorbent assay (ELISA). To overcome the limited machine-learning capability of smartphones, the region-of-interest (ROI) cascading and conditional activation algorithms were utilized herein. The developed microfluidic chip was incorporated with a bubble trap to remove any bubbles detected by AI, which helps in preventing false signals during immunoassay, as well as controlling the reagents' movement with an on-chip micropump and valve. Subsequently, the developed immunosensing platform was tested for conducting real ELISA using a single microplate from the 96-well to detect the Human Cardiac Troponin I (cTnI) biomarker, with a detection limit as low as 0.98 pg/mL. As a result, the developed platform can be envisaged as an AI-based revolution in microfluidics for point-of-care clinical diagnosis.
Collapse
Affiliation(s)
- Nabil H Bhuiyan
- Bio-IT Convergence Laboratory, Department of Electronic Convergence Engineering, KwangWoon University, Seoul 01897, South Korea
| | - Jun H Hong
- Bio-IT Convergence Laboratory, Department of Electronic Convergence Engineering, KwangWoon University, Seoul 01897, South Korea
| | - M Jalal Uddin
- Bio-IT Convergence Laboratory, Department of Electronic Convergence Engineering, KwangWoon University, Seoul 01897, South Korea.,BioGeneSys Inc., 20 Kwangwoon-ro, Nowon-gu, Seoul 01897, South Korea
| | - Joon S Shim
- Bio-IT Convergence Laboratory, Department of Electronic Convergence Engineering, KwangWoon University, Seoul 01897, South Korea.,BioGeneSys Inc., 20 Kwangwoon-ro, Nowon-gu, Seoul 01897, South Korea
| |
Collapse
|
32
|
Li W, Zhou Y, Deng Y, Khoo BL. Early Predictor Tool of Disease Using Label-Free Liquid Biopsy-Based Platforms for Patient-Centric Healthcare. Cancers (Basel) 2022. [PMID: 35159085 DOI: 10.3390/cancfers14030818] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/20/2023] Open
Abstract
Cancer cells undergo phenotypic changes or mutations during treatment, making detecting protein-based or gene-based biomarkers challenging. Here, we used algorithmic analysis combined with patient-derived tumor models to derive an early prediction tool using patient-derived cell clusters from liquid biopsy (LIQBP) for cancer prognosis in a label-free manner. The LIQBP platform incorporated a customized microfluidic biochip that mimicked the tumor microenvironment to establish patient clusters, and extracted physical parameters from images of each sample, including size, thickness, roughness, and thickness per area (n = 31). Samples from healthy volunteers (n = 5) and cancer patients (pretreatment; n = 4) could be easily distinguished with high sensitivity (91.16 ± 1.56%) and specificity (71.01 ± 9.95%). Furthermore, we demonstrated that the multiple unique quantitative parameters reflected patient responses. Among these, the ratio of normalized gray value to cluster size (RGVS) was the most significant parameter correlated with cancer stage and treatment duration. Overall, our work presented a novel and less invasive approach for the label-free prediction of disease prognosis to identify patients who require adjustments to their treatment regime. We envisioned that such efforts would promote the management of personalized patient care conveniently and cost effectively.
Collapse
Affiliation(s)
- Wei Li
- Department of Biomedical Engineering, City University of Hong Kong, Hong Kong 999077, China
- Hong Kong Center for Cerebro-Cardiovascular Health Engineering (COCHE), Hong Kong 999077, China
| | - Yunlan Zhou
- Department of Clinical Laboratory, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200092, China
| | - Yanlin Deng
- Department of Biomedical Engineering, City University of Hong Kong, Hong Kong 999077, China
| | - Bee Luan Khoo
- Department of Biomedical Engineering, City University of Hong Kong, Hong Kong 999077, China
- Hong Kong Center for Cerebro-Cardiovascular Health Engineering (COCHE), Hong Kong 999077, China
- Department of Precision Diagnostic and Therapeutic Technology, City University of Hong Kong Shenzhen-Futian Research Institute, Shenzhen 518057, China
| |
Collapse
|
33
|
Early Predictor Tool of Disease Using Label-Free Liquid Biopsy-Based Platforms for Patient-Centric Healthcare. Cancers (Basel) 2022; 14:cancers14030818. [PMID: 35159085 PMCID: PMC8834418 DOI: 10.3390/cancers14030818] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Revised: 01/29/2022] [Accepted: 01/31/2022] [Indexed: 12/12/2022] Open
Abstract
Simple Summary We proposed a comprehensive early prediction tool based on liquid biopsy for the label-free phenotypic analysis of cell clusters from clinical samples (n = 31). Our custom algorithm analysis, combined with a microfluidic-based tumor model, was designed to assess and stratify cancer patients in a label-free, cost-effective, and user-friendly way. Multiple quantitative phenotypic parameters (cluster size, thickness, roughness, and thickness per area) were derived from the profiling of the patient-derived cell clusters. Our platform could distinguish healthy donors from pretreatment cancer patients with high sensitivity (91.16 ± 1.56%) and specificity (71.01 ± 9.95%). In addition, the ratio of normalized gray value to cluster size (RGVS) parameter was significantly correlated to treatment duration and cancer stage. In conclusion, our patient-centric, early prediction tool will allow the prognosis of patients in a relatively less invasive manner, which can help clinicians identify diseases or indicate the need for new treatment strategies. Abstract Cancer cells undergo phenotypic changes or mutations during treatment, making detecting protein-based or gene-based biomarkers challenging. Here, we used algorithmic analysis combined with patient-derived tumor models to derive an early prediction tool using patient-derived cell clusters from liquid biopsy (LIQBP) for cancer prognosis in a label-free manner. The LIQBP platform incorporated a customized microfluidic biochip that mimicked the tumor microenvironment to establish patient clusters, and extracted physical parameters from images of each sample, including size, thickness, roughness, and thickness per area (n = 31). Samples from healthy volunteers (n = 5) and cancer patients (pretreatment; n = 4) could be easily distinguished with high sensitivity (91.16 ± 1.56%) and specificity (71.01 ± 9.95%). Furthermore, we demonstrated that the multiple unique quantitative parameters reflected patient responses. Among these, the ratio of normalized gray value to cluster size (RGVS) was the most significant parameter correlated with cancer stage and treatment duration. Overall, our work presented a novel and less invasive approach for the label-free prediction of disease prognosis to identify patients who require adjustments to their treatment regime. We envisioned that such efforts would promote the management of personalized patient care conveniently and cost effectively.
Collapse
|
34
|
Yin D, Li Y, Xia L, Li W, Chu W, Yu J, Wu M, Cheng Y, Hu M. Automated synthesis of gadopentetate dimeglumine through solid-liquid reaction in femtosecond laser fabricated microfluidic chips. CHINESE CHEM LETT 2022. [DOI: 10.1016/j.cclet.2021.05.073] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
35
|
Dittrich PG, Kraus D, Ehrhardt E, Henkel T, Notni G. Multispectral Imaging Flow Cytometry with Spatially and Spectrally Resolving Snapshot-Mosaic Cameras for the Characterization and Classification of Bioparticles. MICROMACHINES 2022; 13:mi13020238. [PMID: 35208362 PMCID: PMC8879709 DOI: 10.3390/mi13020238] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Revised: 01/28/2022] [Accepted: 01/29/2022] [Indexed: 11/26/2022]
Abstract
In the development and optimization of biotechnological cultivation processes the continuous monitoring through the acquisition and interpretation of spectral and morphological properties of bioparticles are challenging. There is therefore a need for the parallel acquisition and interpretation of spatially and spectrally resolved measurements with which particles can be characterized and classified in-flow with high throughput. Therefore, in this paper we investigated the scientific and technological connectivity of standard imaging flow cytometry (IFC) with filter-on-chip based spatially and spectrally resolving snapshot-mosaic cameras for photonic sensing and control in a smart and innovative microfluidic device. For the investigations presented here we used the microalgae Haematococcus pluvialis (HP). These microalgae are used commercially to produce the antioxidant keto-carotenoid astaxanthin. Therefore, HP is relevant to practically demonstrate the usability of the developed system for Multispectral Imaging Flow Cytometry (MIFC) platform. The extension of standard IFC with snapshot-mosaic cameras and multivariate data processing is an innovative approach for the in-flow characterization and derived classification of bioparticles. Finally, the multispectral data acquisition and the therefore developed methodology is generalizable and enables further applications far beyond the here characterized population of HP cells.
Collapse
Affiliation(s)
- Paul-Gerald Dittrich
- Department of Mechanical Engineering, Group for Quality Assurance and Industrial Image Processing, Technische Universität Ilmenau, Gustav-Kirchhoff-Platz 2, 98693 Ilmenau, Germany;
- Correspondence:
| | - Daniel Kraus
- Department of Nanobiophotonics, Leibniz Institute of Photonic Technology, Albert-Einstein-Straße 9, 07745 Jena, Germany; (D.K.); (T.H.)
| | - Enrico Ehrhardt
- Gesellschaft zur Förderung von Medizin-, Bio- und Umwelttechnologien e. V., Erich-Neuß-Weg 5, 06120 Halle (Saale), Germany;
| | - Thomas Henkel
- Department of Nanobiophotonics, Leibniz Institute of Photonic Technology, Albert-Einstein-Straße 9, 07745 Jena, Germany; (D.K.); (T.H.)
| | - Gunther Notni
- Department of Mechanical Engineering, Group for Quality Assurance and Industrial Image Processing, Technische Universität Ilmenau, Gustav-Kirchhoff-Platz 2, 98693 Ilmenau, Germany;
| |
Collapse
|
36
|
Del Giudice F. A Review of Microfluidic Devices for Rheological Characterisation. MICROMACHINES 2022; 13:167. [PMID: 35208292 PMCID: PMC8877273 DOI: 10.3390/mi13020167] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 01/19/2022] [Accepted: 01/20/2022] [Indexed: 12/20/2022]
Abstract
The rheological characterisation of liquids finds application in several fields ranging from industrial production to the medical practice. Conventional rheometers are the gold standard for the rheological characterisation; however, they are affected by several limitations, including high costs, large volumes required and difficult integration to other systems. By contrast, microfluidic devices emerged as inexpensive platforms, requiring a little sample to operate and fashioning a very easy integration into other systems. Such advantages have prompted the development of microfluidic devices to measure rheological properties such as viscosity and longest relaxation time, using a finger-prick of volumes. This review highlights some of the microfluidic platforms introduced so far, describing their advantages and limitations, while also offering some prospective for future works.
Collapse
Affiliation(s)
- Francesco Del Giudice
- Department of Chemical Engineering, Faculty of Science and Engineering, School of Engineering and Applied Sciences, Swansea University, Swansea SA1 8EN, UK
| |
Collapse
|
37
|
Feng Y, Cheng Z, Chai H, He W, Huang L, Wang W. Neural network-enhanced real-time impedance flow cytometry for single-cell intrinsic characterization. LAB ON A CHIP 2022; 22:240-249. [PMID: 34849522 DOI: 10.1039/d1lc00755f] [Citation(s) in RCA: 43] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/10/2023]
Abstract
Single-cell impedance flow cytometry (IFC) is emerging as a label-free and non-invasive method for characterizing the electrical properties and revealing sample heterogeneity. At present, most IFC studies utilize phenomenological parameters (e.g., impedance amplitude, phase and opacity) to characterize single cells instead of intrinsic biophysical metrics (e.g., radius r, cytoplasm conductivity σi and specific membrane capacitance Csm). Intrinsic parameters are normally calculated off-line by time-consuming model-fitting methods. Here, we propose to employ neural network (NN)-enhanced IFC to achieve both real-time single-cell intrinsic characterization and intrinsic parameter-based cell classification at high throughput. Three intrinsic parameters (r, σi and Csm) can be obtained online and in real-time via a trained NN at 0.3 ms per single-cell event, achieving significant improvement in calculation speed. Experiments involving four cancer cells and one lymphocyte cell demonstrated 91.5% classification accuracy in the cell type for a test group of 9751 cell samples. By performing a viability assay, we provide evidence that the IFC test per se would not substantially affect the cell property. We envision that the NN-enhanced real-time IFC will provide a new platform for high-throughput, real-time and online cell intrinsic electrical characterization.
Collapse
Affiliation(s)
- Yongxiang Feng
- State Key Laboratory of Precision Measurement Technology and Instrument, Department of Precision Instrument, Tsinghua University, Beijing, China.
| | - Zhen Cheng
- Department of Automation, Tsinghua University, Beijing, China
| | - Huichao Chai
- State Key Laboratory of Precision Measurement Technology and Instrument, Department of Precision Instrument, Tsinghua University, Beijing, China.
| | - Weihua He
- State Key Laboratory of Precision Measurement Technology and Instrument, Department of Precision Instrument, Tsinghua University, Beijing, China.
| | - Liang Huang
- Anhui Province Key Laboratory of Measuring Theory and Precision Instrument, School of Instrument Science and Optoelectronics Engineering, Hefei University of Technology, Hefei, Anhui, China
| | - Wenhui Wang
- State Key Laboratory of Precision Measurement Technology and Instrument, Department of Precision Instrument, Tsinghua University, Beijing, China.
| |
Collapse
|
38
|
Anandakumaran PN, Ayers AG, Muranski P, Creusot RJ, Sia SK. Rapid video-based deep learning of cognate versus non-cognate T cell-dendritic cell interactions. Sci Rep 2022; 12:559. [PMID: 35017558 PMCID: PMC8752671 DOI: 10.1038/s41598-021-04286-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Accepted: 12/17/2021] [Indexed: 12/11/2022] Open
Abstract
Identification of cognate interactions between antigen-specific T cells and dendritic cells (DCs) is essential to understanding immunity and tolerance, and for developing therapies for cancer and autoimmune diseases. Conventional techniques for selecting antigen-specific T cells are time-consuming and limited to pre-defined antigenic peptide sequences. Here, we demonstrate the ability to use deep learning to rapidly classify videos of antigen-specific CD8+ T cells. The trained model distinguishes distinct interaction dynamics (in motility and morphology) between cognate and non-cognate T cells and DCs over 20 to 80 min. The model classified high affinity antigen-specific CD8+ T cells from OT-I mice with an area under the curve (AUC) of 0.91, and generalized well to other types of high and low affinity CD8+ T cells. The classification accuracy achieved by the model was consistently higher than simple image analysis techniques, and conventional metrics used to differentiate between cognate and non-cognate T cells, such as speed. Also, we demonstrated that experimental addition of anti-CD40 antibodies improved model prediction. Overall, this method demonstrates the potential of video-based deep learning to rapidly classify cognate T cell-DC interactions, which may also be potentially integrated into high-throughput methods for selecting antigen-specific T cells in the future.
Collapse
Affiliation(s)
| | - Abigail G Ayers
- Department of Biomedical Engineering, Columbia University, New York, NY, 10027, USA
| | - Pawel Muranski
- Department of Medicine, Division of Hematology/Oncology, Columbia University Irving Medical Center, New York, NY, 10032, USA
- Columbia Center for Translational Immunology, Columbia University Irving Medical Center, New York, NY, 10032, USA
| | - Remi J Creusot
- Columbia Center for Translational Immunology, Columbia University Irving Medical Center, New York, NY, 10032, USA
- Department of Medicine and Naomi Berrie Diabetes Center, Columbia University Irving Medical Center, New York, NY, 10032, USA
| | - Samuel K Sia
- Department of Biomedical Engineering, Columbia University, New York, NY, 10027, USA.
| |
Collapse
|
39
|
Wilson S, Steele S, Adeli K. Innovative technological advancements in laboratory medicine: Predicting the lab of the future. BIOTECHNOL BIOTEC EQ 2022. [DOI: 10.1080/13102818.2021.2011413] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/09/2022] Open
Affiliation(s)
- Siobhan Wilson
- Clinical Biochemistry, Pediatric Laboratory Medicine and Molecular Medicine, Research Institute, The Hospital for Sick Children, Toronto, ON, Canada
- Department of Laboratory Medicine & Pathobiology, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Shannon Steele
- Clinical Biochemistry, Pediatric Laboratory Medicine and Molecular Medicine, Research Institute, The Hospital for Sick Children, Toronto, ON, Canada
| | - Khosrow Adeli
- Clinical Biochemistry, Pediatric Laboratory Medicine and Molecular Medicine, Research Institute, The Hospital for Sick Children, Toronto, ON, Canada
- Department of Laboratory Medicine & Pathobiology, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
40
|
Zheng J, Cole T, Zhang Y, Kim J, Tang SY. Exploiting machine learning for bestowing intelligence to microfluidics. Biosens Bioelectron 2021; 194:113666. [PMID: 34600338 DOI: 10.1016/j.bios.2021.113666] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Revised: 09/18/2021] [Accepted: 09/21/2021] [Indexed: 02/06/2023]
Abstract
Intelligent microfluidics is an emerging cross-discipline research area formed by combining microfluidics with machine learning. It uses the advantages of microfluidics, such as high throughput and controllability, and the powerful data processing capabilities of machine learning, resulting in improved systems in biotechnology and chemistry. Compared to traditional microfluidics using manual analysis methods, intelligent microfluidics needs less human intervention, and results in a more user-friendly experience with faster processing. There is a paucity of literature reviewing this burgeoning and highly promising cross-discipline. Therefore, we herein comprehensively and systematically summarize several aspects of microfluidic applications enabled by machine learning. We list the types of microfluidics used in intelligent microfluidic applications over the last five years, as well as the machine learning algorithms and the hardware used for training. We also present the most recent advances in key technologies, developments, challenges, and the emerging opportunities created by intelligent microfluidics.
Collapse
Affiliation(s)
- Jiahao Zheng
- Department of Electronic, Electrical and Systems Engineering, University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK
| | - Tim Cole
- Department of Electronic, Electrical and Systems Engineering, University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK
| | - Yuxin Zhang
- Department of Electronic, Electrical and Systems Engineering, University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK
| | - Jeeson Kim
- Department of Intelligent Mechatronics Engineering, Sejong University, Seoul, 05006, South Korea.
| | - Shi-Yang Tang
- Department of Electronic, Electrical and Systems Engineering, University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK.
| |
Collapse
|
41
|
Nishikawa M, Kanno H, Zhou Y, Xiao TH, Suzuki T, Ibayashi Y, Harmon J, Takizawa S, Hiramatsu K, Nitta N, Kameyama R, Peterson W, Takiguchi J, Shifat-E-Rabbi M, Zhuang Y, Yin X, Rubaiyat AHM, Deng Y, Zhang H, Miyata S, Rohde GK, Iwasaki W, Yatomi Y, Goda K. Massive image-based single-cell profiling reveals high levels of circulating platelet aggregates in patients with COVID-19. Nat Commun 2021; 12:7135. [PMID: 34887400 PMCID: PMC8660840 DOI: 10.1038/s41467-021-27378-2] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2021] [Accepted: 11/16/2021] [Indexed: 12/19/2022] Open
Abstract
A characteristic clinical feature of COVID-19 is the frequent incidence of microvascular thrombosis. In fact, COVID-19 autopsy reports have shown widespread thrombotic microangiopathy characterized by extensive diffuse microthrombi within peripheral capillaries and arterioles in lungs, hearts, and other organs, resulting in multiorgan failure. However, the underlying process of COVID-19-associated microvascular thrombosis remains elusive due to the lack of tools to statistically examine platelet aggregation (i.e., the initiation of microthrombus formation) in detail. Here we report the landscape of circulating platelet aggregates in COVID-19 obtained by massive single-cell image-based profiling and temporal monitoring of the blood of COVID-19 patients (n = 110). Surprisingly, our analysis of the big image data shows the anomalous presence of excessive platelet aggregates in nearly 90% of all COVID-19 patients. Furthermore, results indicate strong links between the concentration of platelet aggregates and the severity, mortality, respiratory condition, and vascular endothelial dysfunction level of COVID-19 patients.
Collapse
Affiliation(s)
- Masako Nishikawa
- Department of Clinical Laboratory Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, 113-0033, Japan
| | - Hiroshi Kanno
- Department of Chemistry, The University of Tokyo, Tokyo, 113-0033, Japan
| | - Yuqi Zhou
- Department of Chemistry, The University of Tokyo, Tokyo, 113-0033, Japan
| | - Ting-Hui Xiao
- Department of Chemistry, The University of Tokyo, Tokyo, 113-0033, Japan.
| | - Takuma Suzuki
- Department of Computational Biology and Medical Sciences, The University of Tokyo, Chiba, 277-8562, Japan
| | - Yuma Ibayashi
- Department of Chemistry, The University of Tokyo, Tokyo, 113-0033, Japan
| | - Jeffrey Harmon
- Department of Chemistry, The University of Tokyo, Tokyo, 113-0033, Japan
| | - Shigekazu Takizawa
- Department of Chemistry, The University of Tokyo, Tokyo, 113-0033, Japan
| | - Kotaro Hiramatsu
- Department of Chemistry, The University of Tokyo, Tokyo, 113-0033, Japan
- Research Center for Spectrochemistry, The University of Tokyo, Tokyo, 113-0033, Japan
| | | | - Risako Kameyama
- Department of Chemistry, The University of Tokyo, Tokyo, 113-0033, Japan
| | - Walker Peterson
- Department of Chemistry, The University of Tokyo, Tokyo, 113-0033, Japan
| | - Jun Takiguchi
- Department of Clinical Laboratory Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, 113-0033, Japan
| | | | - Yan Zhuang
- Department of Electrical and Computer Engineering, University of Virginia, Virginia, 22908, USA
| | - Xuwang Yin
- Department of Electrical and Computer Engineering, University of Virginia, Virginia, 22908, USA
| | | | - Yunjie Deng
- Department of Chemistry, The University of Tokyo, Tokyo, 113-0033, Japan
| | - Hongqian Zhang
- Department of Chemistry, The University of Tokyo, Tokyo, 113-0033, Japan
| | - Shigeki Miyata
- Research and Development Department, Central Blood Institute, Japanese Red Cross Society, Tokyo, 135-8521, Japan
| | - Gustavo K Rohde
- Department of Biomedical Engineering, University of Virginia, Virginia, 22908, USA
- Department of Electrical and Computer Engineering, University of Virginia, Virginia, 22908, USA
| | - Wataru Iwasaki
- Department of Computational Biology and Medical Sciences, The University of Tokyo, Chiba, 277-8562, Japan
- Department of Biological Sciences, The University of Tokyo, Tokyo, 113-0033, Japan
- Department of Integrated Biosciences, The University of Tokyo, Chiba, 277-8562, Japan
| | - Yutaka Yatomi
- Department of Clinical Laboratory Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, 113-0033, Japan.
| | - Keisuke Goda
- Department of Chemistry, The University of Tokyo, Tokyo, 113-0033, Japan.
- Institute of Technological Sciences, Wuhan University, 430072, Hubei, China.
- Department of Bioengineering, University of California, Los Angeles, California, 90095, USA.
| |
Collapse
|
42
|
Liu L, Bi M, Wang Y, Liu J, Jiang X, Xu Z, Zhang X. Artificial intelligence-powered microfluidics for nanomedicine and materials synthesis. NANOSCALE 2021; 13:19352-19366. [PMID: 34812823 DOI: 10.1039/d1nr06195j] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/28/2023]
Abstract
Artificial intelligence (AI) is an emerging technology with great potential, and its robust calculation and analysis capabilities are unmatched by traditional calculation tools. With the promotion of deep learning and open-source platforms, the threshold of AI has also become lower. Combining artificial intelligence with traditional fields to create new fields of high research and application value has become a trend. AI has been involved in many disciplines, such as medicine, materials, energy, and economics. The development of AI requires the support of many kinds of data, and microfluidic systems can often mine object data on a large scale to support AI. Due to the excellent synergy between the two technologies, excellent research results have emerged in many fields. In this review, we briefly review AI and microfluidics and introduce some applications of their combination, mainly in nanomedicine and material synthesis. Finally, we discuss the development trend of the combination of the two technologies.
Collapse
Affiliation(s)
- Linbo Liu
- John A. Paulson School of Engineering and Applied Science, Harvard University, Cambridge, MA 02138, USA
| | - Mingcheng Bi
- Institute of Process Equipment, College of Energy Engineering, Zhejiang University, Hangzhou 310027, P.R. China
| | - Yunhua Wang
- John A. Paulson School of Engineering and Applied Science, Harvard University, Cambridge, MA 02138, USA
| | - Junfeng Liu
- Institute of Process Equipment, College of Energy Engineering, Zhejiang University, Hangzhou 310027, P.R. China
| | - Xiwen Jiang
- College of Biological Science and Engineering, Fuzhou university, Fuzhou 350108, P.R. China
| | - Zhongbin Xu
- Institute of Process Equipment, College of Energy Engineering, Zhejiang University, Hangzhou 310027, P.R. China
| | - Xingcai Zhang
- John A. Paulson School of Engineering and Applied Science, Harvard University, Cambridge, MA 02138, USA
- School of Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA.
| |
Collapse
|
43
|
Lee SM, Balakrishnan HK, Yuan D, Nai YH, Guijt RM. Perspective - what constitutes a quality analytical paper: Microfluidics and Flow analysis. TALANTA OPEN 2021. [DOI: 10.1016/j.talo.2021.100055] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
|
44
|
Song P, Guo C, Jiang S, Wang T, Hu P, Hu D, Zhang Z, Feng B, Zheng G. Optofluidic ptychography on a chip. LAB ON A CHIP 2021; 21:4549-4556. [PMID: 34726219 DOI: 10.1039/d1lc00719j] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
We report the implementation of a fully on-chip, lensless microscopy technique termed optofluidic ptychography. This imaging modality complements the miniaturization provided by microfluidics and allows the integration of ptychographic microscopy into various lab-on-a-chip devices. In our prototype, we place a microfluidic channel on the top surface of a coverslip and coat the bottom surface with a scattering layer. The channel and the coated coverslip substrate are then placed on top of an image sensor for diffraction data acquisition. Similar to the operation of a flow cytometer, the device utilizes microfluidic flow to deliver specimens across the channel. The diffracted light from the flowing objects is modulated by the scattering layer and recorded by the image sensor for ptychographic reconstruction, where high-resolution quantitative complex images are recovered from the diffraction measurements. By using an image sensor with a 1.85 μm pixel size, our device can resolve the 550 nm linewidth on the resolution target. We validate the device by imaging different types of biospecimens, including C. elegans, yeast cells, paramecium, and closterium sp. We also demonstrate a high-resolution ptychographic reconstruction at a video framerate of 30 frames per second. The reported technique can address a wide range of biomedical needs and engenders new ptychographic imaging innovations in a flow cytometer configuration.
Collapse
Affiliation(s)
- Pengming Song
- Department of Biomedical Engineering, University of Connecticut, Storrs, CT, 06269, USA.
| | - Chengfei Guo
- Department of Biomedical Engineering, University of Connecticut, Storrs, CT, 06269, USA.
| | - Shaowei Jiang
- Department of Biomedical Engineering, University of Connecticut, Storrs, CT, 06269, USA.
| | - Tianbo Wang
- Department of Biomedical Engineering, University of Connecticut, Storrs, CT, 06269, USA.
| | - Patrick Hu
- Department of Computer Science, University of California Irvine, Irvine, CA, 92697, USA
| | - Derek Hu
- Amador Valley High School, Pleasanton, CA, 94566, USA
| | - Zibang Zhang
- Department of Biomedical Engineering, University of Connecticut, Storrs, CT, 06269, USA.
| | - Bin Feng
- Department of Biomedical Engineering, University of Connecticut, Storrs, CT, 06269, USA.
| | - Guoan Zheng
- Department of Biomedical Engineering, University of Connecticut, Storrs, CT, 06269, USA.
| |
Collapse
|
45
|
Xin L, Xiao W, Che L, Liu J, Miccio L, Bianco V, Memmolo P, Ferraro P, Li X, Pan F. Label-Free Assessment of the Drug Resistance of Epithelial Ovarian Cancer Cells in a Microfluidic Holographic Flow Cytometer Boosted through Machine Learning. ACS OMEGA 2021; 6:31046-31057. [PMID: 34841147 PMCID: PMC8613806 DOI: 10.1021/acsomega.1c04204] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Accepted: 10/29/2021] [Indexed: 05/13/2023]
Abstract
About 75% of epithelial ovarian cancer (EOC) patients suffer from relapsing and develop drug resistance after primary chemotherapy. The commonly used clinical examinations and biological tumor tissue models for chemotherapeutic sensitivity are time-consuming and expensive. Research studies showed that the cell morphology-based method is promising to be a new route for chemotherapeutic sensitivity evaluation. Here, we offer how the drug resistance of EOC cells can be assessed through a label-free and high-throughput microfluidic flow cytometer equipped with a digital holographic microscope reinforced by machine learning. It is the first time that such type of assessment is performed to the best of our knowledge. Several morphologic and texture features at a single-cell level have been extracted from the quantitative phase images. In addition, we compared four common machine learning algorithms, including naive Bayes, decision tree, K-nearest neighbors, support vector machine (SVM), and fully connected network. The result shows that the SVM classifier achieves the optimal performance with an accuracy of 92.2% and an area under the curve of 0.96. This study demonstrates that the proposed method achieves high-accuracy, high-throughput, and label-free assessment of the drug resistance of EOC cells. Furthermore, it reflects strong potentialities to develop data-driven individualized chemotherapy treatments in the future.
Collapse
Affiliation(s)
- Lu Xin
- Key
Laboratory of Precision Opto-mechatronics Technology, School of Instrumentation
& Optoelectronic Engineering, Beihang
University, Beijing 100191, China
| | - Wen Xiao
- Key
Laboratory of Precision Opto-mechatronics Technology, School of Instrumentation
& Optoelectronic Engineering, Beihang
University, Beijing 100191, China
| | - Leiping Che
- Key
Laboratory of Precision Opto-mechatronics Technology, School of Instrumentation
& Optoelectronic Engineering, Beihang
University, Beijing 100191, China
| | - JinJin Liu
- Department
of Obstetrics and Gynecology, Peking University
People’s Hospital, Beijing 100044, China
| | - Lisa Miccio
- CNR,
Institute of Applied Sciences & Intelligent Systems (ISASI) “E.
Caianiello”, via
Campi Flegrei 34, 80078 Pozzuoli, Italy
| | - Vittorio Bianco
- CNR,
Institute of Applied Sciences & Intelligent Systems (ISASI) “E.
Caianiello”, via
Campi Flegrei 34, 80078 Pozzuoli, Italy
| | - Pasquale Memmolo
- CNR,
Institute of Applied Sciences & Intelligent Systems (ISASI) “E.
Caianiello”, via
Campi Flegrei 34, 80078 Pozzuoli, Italy
| | - Pietro Ferraro
- CNR,
Institute of Applied Sciences & Intelligent Systems (ISASI) “E.
Caianiello”, via
Campi Flegrei 34, 80078 Pozzuoli, Italy
| | - Xiaoping Li
- Department
of Obstetrics and Gynecology, Peking University
People’s Hospital, Beijing 100044, China
| | - Feng Pan
- Key
Laboratory of Precision Opto-mechatronics Technology, School of Instrumentation
& Optoelectronic Engineering, Beihang
University, Beijing 100191, China
| |
Collapse
|
46
|
Manteca A, Gadea A, Van Assche D, Cossard P, Gillard-Bocquet M, Beneyton T, Innis CA, Baret JC. Directed Evolution in Drops: Molecular Aspects and Applications. ACS Synth Biol 2021; 10:2772-2783. [PMID: 34677942 PMCID: PMC8609573 DOI: 10.1021/acssynbio.1c00313] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Indexed: 11/29/2022]
Abstract
The process of optimizing the properties of biological molecules is paramount for many industrial and medical applications. Directed evolution is a powerful technique for modifying and improving biomolecules such as proteins or nucleic acids (DNA or RNA). Mimicking the mechanism of natural evolution, one can enhance a desired property by applying a suitable selection pressure and sorting improved variants. Droplet-based microfluidic systems offer a high-throughput solution to this approach by helping to overcome the limiting screening steps and allowing the analysis of variants within increasingly complex libraries. Here, we review cases where successful evolution of biomolecules was achieved using droplet-based microfluidics, focusing on the molecular processes involved and the incorporation of microfluidics to the workflow. We highlight the advantages and limitations of these microfluidic systems compared to low-throughput methods and show how the integration of these systems into directed evolution workflows can open new avenues to discover or improve biomolecules according to user-defined conditions.
Collapse
Affiliation(s)
- Aitor Manteca
- Univ.
Bordeaux, Institut National de la Santé et de la Recherche
Médicale, Centre National de la Recherche Scientifique, ARNA,
U1212, UMR 5320, Institut Européen de Chimie et Biologie, F-33600 Pessac, France
| | - Alejandra Gadea
- Univ.
Bordeaux, CNRS, CRPP, UMR 5031, F-33610, Pessac, France
| | | | - Pauline Cossard
- Univ.
Bordeaux, Institut National de la Santé et de la Recherche
Médicale, Centre National de la Recherche Scientifique, ARNA,
U1212, UMR 5320, Institut Européen de Chimie et Biologie, F-33600 Pessac, France
| | - Mélanie Gillard-Bocquet
- Univ.
Bordeaux, Institut National de la Santé et de la Recherche
Médicale, Centre National de la Recherche Scientifique, ARNA,
U1212, UMR 5320, Institut Européen de Chimie et Biologie, F-33600 Pessac, France
| | - Thomas Beneyton
- Univ.
Bordeaux, CNRS, CRPP, UMR 5031, F-33610, Pessac, France
| | - C. Axel Innis
- Univ.
Bordeaux, Institut National de la Santé et de la Recherche
Médicale, Centre National de la Recherche Scientifique, ARNA,
U1212, UMR 5320, Institut Européen de Chimie et Biologie, F-33600 Pessac, France
| | - Jean-Christophe Baret
- Univ.
Bordeaux, CNRS, CRPP, UMR 5031, F-33610, Pessac, France
- Institut
Universitaire de France, F-75231 Paris, France
| |
Collapse
|
47
|
Dannhauser D, Rossi D, Palatucci AT, Rubino V, Carriero F, Ruggiero G, Ripaldi M, Toriello M, Maisto G, Netti PA, Terrazzano G, Causa F. Non-invasive and label-free identification of human natural killer cell subclasses by biophysical single-cell features in microfluidic flow. LAB ON A CHIP 2021; 21:4144-4154. [PMID: 34515262 DOI: 10.1039/d1lc00651g] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Natural killer (NK) cells are indicated as favorite candidates for innovative therapeutic treatment and are divided into two subclasses: immature regulatory NK CD56bright and mature cytotoxic NK CD56dim. Therefore, the ability to discriminate CD56dim from CD56bright could be very useful because of their higher cytotoxicity. Nowadays, NK cell classification is routinely performed by cytometric analysis based on surface receptor expression. Here, we present an in-flow, label-free and non-invasive biophysical analysis of NK cells through a combination of light scattering and machine learning (ML) for NK cell subclass classification. In this respect, to identify relevant biophysical cell features, we stimulated NK cells with interleukine-15 inducing a subclass transition from CD56bright to CD56dim. We trained our ML algorithm with sorted NK cell subclasses (≥86% accuracy). Next, we applied our NK cell classification algorithm to cells stimulated over time, to investigate the transition of CD56bright to CD56dim and their biophysical feature changes. Finally, we tested our approach on several proband samples, highlighting the potential of our measurement approach. We show a label-free way for the robust identification of NK cell subclasses based on biophysical features, which can be applied in both cell biology and cell therapy.
Collapse
Affiliation(s)
- 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
| | - Anna Teresa Palatucci
- Dipartimento di Scienze (DiS), Università della Basilicata, Via dell'Ateneo Lucano 10, 85100 Potenza, Italy
| | - Valentina Rubino
- Dipartimento di Scienze Mediche Traslazionali, Università degli Studi di Napoli "Federico II", Naples, Italy
| | - Flavia Carriero
- Dipartimento di Scienze Mediche Traslazionali, Università degli Studi di Napoli "Federico II", Naples, Italy
| | - Giuseppina Ruggiero
- Dipartimento di Scienze Mediche Traslazionali, Università degli Studi di Napoli "Federico II", Naples, Italy
| | - Mimmo Ripaldi
- Dipartimento Oncologia AORN Santobono Pausilipon Hospital, Via Posillipo, 226, 80123, Naples, Italy
| | - Mario Toriello
- Dipartimento Oncologia AORN Santobono Pausilipon Hospital, Via Posillipo, 226, 80123, Naples, Italy
| | - Giovanna Maisto
- Dipartimento Oncologia AORN Santobono Pausilipon Hospital, Via Posillipo, 226, 80123, Naples, Italy
| | - Paolo Antonio 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
| | - Giuseppe Terrazzano
- Dipartimento di Scienze (DiS), Università della Basilicata, Via dell'Ateneo Lucano 10, 85100 Potenza, 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
|
48
|
Puthongkham P, Wirojsaengthong S, Suea-Ngam A. Machine learning and chemometrics for electrochemical sensors: moving forward to the future of analytical chemistry. Analyst 2021; 146:6351-6364. [PMID: 34585185 DOI: 10.1039/d1an01148k] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Electrochemical sensors and biosensors have been successfully used in a wide range of applications, but systematic optimization and nonlinear relationships have been compromised for electrode fabrication and data analysis. Machine learning and experimental designs are chemometric tools that have been proved to be useful in method development and data analysis. This minireview summarizes recent applications of machine learning and experimental designs in electroanalytical chemistry. First, experimental designs, e.g., full factorial, central composite, and Box-Behnken are discussed as systematic approaches to optimize electrode fabrication to consider the effects from individual variables and their interactions. Then, the principles of machine learning algorithms, including linear and logistic regressions, neural network, and support vector machine, are introduced. These machine learning models have been implemented to extract complex relationships between chemical structures and their electrochemical properties and to analyze complicated electrochemical data to improve calibration and analyte classification, such as in electronic tongues. Lastly, the future of machine learning and experimental designs in electrochemical sensors is outlined. These chemometric strategies will accelerate the development and enhance the performance of electrochemical devices for point-of-care diagnostics and commercialization.
Collapse
Affiliation(s)
- Pumidech Puthongkham
- Department of Chemistry, Faculty of Science, Chulalongkorn University, Bangkok 10330, Thailand. .,Electrochemistry and Optical Spectroscopy Center of Excellence (EOSCE), Chulalongkorn University, Bangkok 10330, Thailand.,Center of Excellence in Responsive Wearable Materials, Chulalongkorn University, Bangkok 10330, Thailand
| | - Supacha Wirojsaengthong
- Department of Chemistry, Faculty of Science, Chulalongkorn University, Bangkok 10330, Thailand.
| | - Akkapol Suea-Ngam
- Department of Materials, Department of Bioengineering, and Institute of Biomedical Engineering, Imperial College London, London, SW7 2AZ, UK
| |
Collapse
|
49
|
Loo MH, Nakagawa Y, Kim SH, Isozaki A, Goda K. High-throughput sorting of nanoliter droplets enabled by a sequentially addressable dielectrophoretic array. Electrophoresis 2021; 43:477-486. [PMID: 34599837 DOI: 10.1002/elps.202100057] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 09/14/2021] [Accepted: 09/20/2021] [Indexed: 11/08/2022]
Abstract
Droplet microfluidics has emerged as a powerful tool for a diverse range of biomedical and industrial applications such as single-cell analysis, directed evolution, and metabolic engineering. In these applications, droplet sorting has been effective for isolating small droplets encapsulating molecules, cells, or crystals of interest. Recently, there is an increased interest in extending the applicability of droplet sorting to larger droplets to utilize their size advantage. However, sorting throughputs of large droplets have been limited, hampering their wide adoption. Here, we report our demonstration of high-throughput fluorescence-activated droplet sorting of 1 nL droplets using an upgraded version of the sequentially addressable dielectrophoretic array (SADA), which we reported previously. The SADA is an array of electrodes that are individually and sequentially activated/deactivated according to the speed and position of a droplet passing nearby the array. We upgraded the SADA by increasing the number of driving electrodes constituting the SADA and incorporating a slanted microchannel. By using a ten-electrode SADA with the slanted microchannel, we achieved fluorescence-activated droplet sorting of 1 nL droplets at a record high throughput of 1752 droplets/s, twice as high as the previously reported maximum sorting throughput of 1 nL droplets.
Collapse
Affiliation(s)
- Mun Hong Loo
- Department of Chemistry, The University of Tokyo, Tokyo, Japan
| | - Yuta Nakagawa
- Department of Chemistry, The University of Tokyo, Tokyo, Japan
| | - Soo Hyeon Kim
- Institute of Industrial Science, The University of Tokyo, Tokyo, Japan
| | - Akihiro Isozaki
- Department of Chemistry, The University of Tokyo, Tokyo, Japan.,Kanagawa Institute of Industrial Science and Technology, Kanagawa, Japan
| | - Keisuke Goda
- Department of Chemistry, The University of Tokyo, Tokyo, Japan.,Department of Bioengineering, University of California, Los Angeles, CA, USA.,Institute of Technological Sciences, Wuhan University, Hubei, P. R. China
| |
Collapse
|
50
|
Abstract
Sepsis is a life-threatening condition caused by the extreme release of inflammatory mediators into the blood in response to infection (e.g., bacterial infection, COVID-19), resulting in the dysfunction of multiple organs. Currently, there is no direct treatment for sepsis. Here we report an abiotic hydrogel nanoparticle (HNP) as a potential therapeutic agent for late-stage sepsis. The HNP captures and neutralizes all variants of histones, a major inflammatory mediator released during sepsis. The highly optimized HNP has high capacity and long-term circulation capability for the selective sequestration and neutralization of histones. Intravenous injection of the HNP protects mice against a lethal dose of histones through the inhibition of platelet aggregation and migration into the lungs. In vivo administration in murine sepsis model mice results in near complete survival. These results establish the potential for synthetic, nonbiological polymer hydrogel sequestrants as a new intervention strategy for sepsis therapy and adds to our understanding of the importance of histones to this condition. Sepsis caused by the release of inflammatory mediators into the blood is a life threatening disease. Here, the authors report on the development of hydrogel nanoparticles for the capture and neutralisation of histones, major inflammatory mediators, and demonstrate sepsis treatment in a murine model.
Collapse
|