Open Access Article
This Open Access Article is licensed under a Creative Commons Attribution-Non Commercial 3.0 Unported Licence

Transforming microfluidics for single-cell analysis with robotics and artificial intelligence

Jinxiong Cheng ab, Rajiv Anne ab and Yu-Chih Chen *abcd
aDepartment of Bioengineering, Swanson School of Engineering, University of Pittsburgh, 3700 O'Hara Street, Pittsburgh, PA 15260, USA
bUPMC Hillman Cancer Center, University of Pittsburgh, 5115 Centre Ave, Pittsburgh, PA 15232, USA. E-mail: cheny25@upmc.edu; Tel: +1 412 623 7701
cDepartment of Computational and Systems Biology, University of Pittsburgh, 3420 Forbes Avenue, Pittsburgh, PA 15260, USA
dCMU-Pitt Ph.D. Program in Computational Biology, University of Pittsburgh, 3420 Forbes Avenue, Pittsburgh, PA 15260, USA

Received 1st March 2025 , Accepted 13th October 2025

First published on 28th October 2025


Abstract

Single-cell analysis has advanced biomedical research by revealing cellular heterogeneity with unprecedented resolution, identifying rare subpopulations that drive disease progression and therapeutic resistance. Microfluidics is central to this advancement, enabling precise single-cell isolation, manipulation, and cellular profiling. However, limitations in automation, reliability, and technical barriers hinder the widespread adoption of microfluidic single-cell analysis. This review highlights key innovations in experimental methods and deep learning-driven data analysis to overcome these challenges. Operating microfluidics with robotic operation, digital microfluidics, or microrobots enhances experimental precision and scalability. Beyond experimental automation, deep learning revolutionizes data interpretation through label-free image processing and cell status classification and regression. Generative models further refine analysis by correcting batch effects and generating synthetic datasets, improving accuracy and reproducibility in single-cell studies. Considering the complexity of integrating these technologies, remote shared cloud labs represent a potential pathway toward standardized and high-throughput single-cell analysis, facilitating broader access to advanced experimental workflows. Overall, the convergence of robotics and artificial intelligence in single-cell analysis will change data acquisition, hypothesis testing, and model refinement, driving breakthroughs in drug discovery and personalized medicine. While implementation remains challenging, this paradigm shift is transforming biomedical research, enabling unprecedented precision, scalability, and data-driven innovation.


image file: d5lc00216h-p1.tif

Jinxiong Cheng

Jinxiong Cheng is a Ph.D. student in the Department of Bioengineering at the University of Pittsburgh, working in Dr. Yu-Chih Chen's laboratory. He received his B.S. degree in Biomedical Engineering from the Southern University of Science and Technology (SUSTech), Shenzhen, in 2023 under the mentorship of Prof. Xingyu Jiang. During his senior year, he conducted his undergraduate thesis research in Prof. Aaron Wheeler's laboratory at the University of Toronto, focusing on digital microfluidics. His current research interests center on high-throughput microfluidic technologies and their applications in cancer biology.

image file: d5lc00216h-p2.tif

Rajiv Anne

Rajiv Anne completed his undergraduate studies in Bioengineering with a concentration in Cellular Engineering at the University of Pittsburgh, where he worked in Dr. Yu-Chih Chen's laboratory for three years. He also participated in the Summer Undergraduate Research Internship (SURI) at Pitt. He will be pursuing his master's degree at the Technical University of Munich. His research expertise centers on applying convolutional neural networks (CNNs) for label-free cellular morphological analysis.

image file: d5lc00216h-p3.tif

Yu-Chih Chen

Yu-Chih Chen earned dual bachelor's degrees in Electrical Engineering and Law from National Taiwan University (2008) and a Ph.D. in Electrical & Computer Engineering from the University of Michigan, Ann Arbor (2014). He is an Assistant Professor in the Department of Computational and Systems Biology and at the UPMC Hillman Cancer Center, University of Pittsburgh. Dr. Chen has been recognized with the TSMC Outstanding Student Research Award and named an Emerging Forbes Scholar by the Forbes Institute for Cancer Discovery and Hillman Early Career Fellow. His research focuses on high-throughput single-cell analysis, microfluidics, and deep learning for cancer precision medicine.


1. Introduction

1.1. Historical convergence of microfluidics, robotics, and AI

Over the past two decades, microfluidics, robotics, and artificial intelligence (AI) have each undergone remarkable technological evolution, individually transforming the landscape of biological research. When viewed through a historical lens, these three fields reveal a converging trajectory that now positions their integration as both inevitable and impactful, especially in the realm of single-cell analysis. As illustrated in Fig. 1, the microfluidics field emerged with the advent of polydimethylsiloxane (PDMS) soft lithography1 and Quake valves2 in the early 2000s, enabling precise fluid control over cellular microenvironments.3,4 Digital microfluidics (DMF) subsequently introduced enhanced automation and programmability, with droplet volumes exhibiting coefficients of variation (CVs) as low as 1%, thereby enabling reliable single-cell manipulation.5–7 Droplet microfluidics8 enabled massively parallel single-cell encapsulation (∼44[thin space (1/6-em)]000 single cells in a single run) and laid the foundation for technologies like Drop-seq9 and CITE-seq.10 However, its implementation often involves complex fluidic control systems and surfactant management. Organ-on-a-chip platforms improved biological relevance and disease modeling fidelity through biomimetic architectures yet often lacked the spatial resolution and throughput necessary for robust single-cell-level analysis, such as spatial heterogeneity profiling.11–14 In parallel, robotics progressed from basic mobile platforms like AIBO15 and Roomba16 to sensor-rich collaborative robots (e.g., PR2 (ref. 17)), and eventually to human-assistive or autonomous systems capable of delicate biological operations. This trajectory has enabled robust sample handling, fluidic actuation, and physical automation of complex workflows, essential in biology. Meanwhile, the rise of deep learning and large language models transformed AI from static classifiers to adaptable, multi-modal systems capable of interpreting imaging, text, and command instructions. AI-guided decision-making was being integrated into real-time optimization and context-aware control in healthcare applications.18 The synergy among these domains is no longer aspirational but has become technically and conceptually aligned. In the context of single-cell research, microfluidics provides the physical interface for cell handling, robotics enables scalable and precise execution, and AI supplies the intelligence to drive adaptive control and data interpretation. This convergence is essential not just for scaling experiments, but for enabling new modes of inquiry in precision biology and clinical diagnostics.
image file: d5lc00216h-f1.tif
Fig. 1 Milestones in microfluidics, robotics and AI in two decades. This timeline highlights key milestones in microfluidics, from PDMS prototyping1 to the emergence of AI-guided microfluidics.187 Notable advances include the Quake valve,2 cell culture integration,4 digital,6 paper-based,188 and droplet microfluidics,9 along with acoustofluidics,189 magnetofluidics,190 3D printing,191 and optofluidics.192 Recent breakthroughs encompass Drop-seq,9 organ-on-a-chip,11–14 CITE-seq,10 POCT for COVID-19,193 and single-cell multi-omics.95 Parallel progress in robotics and AI includes early consumer robotics (AIBO,15 Roomba16), autonomous navigation through the DARPA Grand Challenge, and interactive systems such as PR2, ROS,17 and Kinect.194 The rise of deep learning with AlexNet195 and achievements like AlphaGo196 and GPT-2/3/4 (ref. 197) enabled smarter, perception-driven automation. By the 2020s, the introduction of RT-2 (ref. 198) and Tesla Optimus marked a shift toward intelligent robotic workflows. Key robotic scientist milestones include EXPO (2006),20 Adam (2009),21 Maholo (2017),24,25 Robot Chemist (2020),199 and ChemAgents (2024).28 Created using BioRender, based on references.1,4,6,9–17,20,21,24,25,28,95,187–199

With the advancement of robotics, the concept of a “robot scientist” was first introduced by King et al. in 2004.19 They envisioned an automated system capable of both hypothesis generation and experimental execution, integrating machine learning to acquire background knowledge and analyze results. Building on this vision, Soldatova et al. developed ontology of scientific experiments (EXPO) in 2006, the first formal ontology to digitally capture and structure every aspect of the scientific process.20 This foundation enabled the creation of Adam, a robot scientist that autonomously generated biological hypotheses and performed wet-lab experiments to test them, particularly in yeast functional genomics.21 Following this, the same group introduced Eve, a second-generation robot scientist focused on drug discovery.22 Eve combined machine learning-based activity prediction with high-throughput screening to prioritize validation of candidate compounds, an early attempt to close the loop between in silico prediction and wet-lab verification. In parallel, Savall et al. developed an automated platform for Drosophila neuroscience that integrated robotic handling, microsurgery, machine vision, and behavioral analysis into a single pipeline.23 During this period, platforms including Transcriptic (now Strateos) and Emerald Cloud Lab began offering remote-controlled, automated experimentation, which have been described as early demonstrations of the envisioned “cloud lab” concept. A significant breakthrough came with Maholo, a dual-arm humanoid robot introduced by Yachie et al. in 2017.24,25 Unlike traditional liquid-handling robots confined to custom workstations, Maholo's human-like architecture allowed it to operate standard lab tools without modification. This human-compatible design greatly enhanced its flexibility, enabling seamless integration into existing workflows and broadening its utility across diverse biological protocols. Since 2020, continued advances in robotics and AI have driven the emergence of more intelligent and autonomous robotic scientists. For example, Cooper et al. developed a mobile chemist that employed Bayesian optimization to iteratively guide experiments.26 The platform developed by Lunt et al. introduced coordinated multi-robot systems with autonomous decision-making,27 while the ChemAgents system proposed by Song et al. leveraged large language models (LLMs) to perform six complex, multi-step experimental tasks.28 While early implementations have largely centered on chemical research, recent advances in biological interfacing and intelligent control signal a growing opportunity within the life sciences, particularly in the context of single-cell analysis. In this review, we aim to explore the potential of such trio-interaction by examining how microfluidics provides the physical interface, robotics delivers precise and scalable execution, and AI enables adaptive decision-making. By synthesizing progress across these three domains, we seek to identify opportunities, challenges, and future directions toward truly intelligent biological studies.

1.2. Single-cell analysis in biomedical research

Single-cell analysis has emerged as a transformative approach in biomedical research, enabling the investigation of cellular heterogeneity with unprecedented resolution.29–34 Unlike bulk analysis, which averages signals across a population of cells, single-cell techniques dissect the molecular and functional diversity within seemingly homogeneous tissues. This is particularly critical given that cellular heterogeneity is a fundamental property of biological systems, arising from genetic, epigenetic, and environmental variations.35–38 Even within a clonal population, stochastic gene expression, differential microenvironmental interactions, and dynamic cellular states contribute to variability in function and fate. Traditional bulk measurements often obscure these crucial differences, masking rare but biologically significant subpopulations. By providing a high-dimensional view of cellular heterogeneity, single-cell analysis allows researchers to deconvolute complex biological processes, identify rare or transient cell states (<1%), and construct precise maps of tissue organization and disease progression.39–41

The impact of single-cell analysis is particularly evident in cancer and immunology research, where cellular heterogeneity directly influences disease outcomes. In oncology, single-cell transcriptomics has uncovered distinct tumor subpopulations with varying drug sensitivities, providing critical insights into therapeutic resistance.42–46 Even a small fraction of drug-resistant cells, can drive disease progression and treatment failure.47,48 Single-cell RNA sequencing (scRNA-seq) has identified stem-like subclones in high-grade serous ovarian cancer, fueling recurrence and metastasis.49,50 Likewise, single-cell epigenomic profiling has revealed non-genetic mechanisms of drug tolerance, exposing adaptive resistance strategies that evade conventional detection.51–54 Single-cell analysis has transformed immunology by revealing the functional diversity of immune cells, particularly within the tumor microenvironment. Even rare immune subsets, sometimes comprising less than 1–2% of the infiltrating population, can drive potent and divergent immune responses.55–57 Tumor-infiltrating lymphocytes (TILs) exhibit highly heterogeneous activation states, with exhausted CD8+ T-cell subsets constraining anti-tumor immunity.58–60 Single-cell technologies have mapped distinct exhaustion trajectories, guiding strategies to optimize immunotherapy. Beyond cancer, single-cell profiling of peripheral immune responses in infectious diseases has identified novel immune signatures predictive of disease severity, advancing precision diagnostics and treatment.61,62 The integration of spatial and multi-omics approaches is further reshaping biomedical research by uncovering deeper mechanistic insights, with current platforms achieving pixel sizes of 10–55 μm (capturing from single cells to small clusters) and enabling thousands of spatially resolved spots per tissue section.63–65 By dissecting cellular heterogeneity, single-cell analysis is driving precision medicine, enabling patient-specific therapies based on single-cell molecular profiles rather than bulk tissue properties.

1.3. Microfluidics for single-cell analysis

Microfluidics which precisely manipulate and control fluids at the microscale level is one of the most important technologies in single-cell analysis.66–68 This technology integrates principles from engineering, physics, chemistry, and biology to create miniaturized systems capable of performing various analytical, diagnostic, and biological assays. By leveraging the unique physical phenomena that occur at the microscale, such as laminar flow, surface tension, and capillary action, microfluidic devices enable the efficient handling of small volumes (picoliter–nanoliter) of liquids with high precision and throughput.69,70 In the realm of single-cell analysis, microfluidics offers several key advantages that make it an ideal platform for studying individual cells at high resolution. First, microfluidic devices enable the isolation and manipulation of single cells with unparalleled precision (70–90%), allowing researchers to study cellular behavior and responses in a controlled microenvironment.33,71,72 By confining cells within micro-chambers or droplets, microfluidic systems minimize cell-to-cell interactions and ensure uniform experimental conditions, enhancing the reproducibility and reliability of single-cell experiments. High-density microchambers or droplets drive parallel, high-throughput single-cell analysis, enabling rapid and precise large-scale cellular profiling (104–106 cells per run).73 Integrating microfluidics with imaging, sensors, and molecular analysis techniques, such as fluorescence microscopy, RNA sequencing, and proteomic profiling, unlocks comprehensive, high-dimensional insights from individual cells at an unprecedented scale.9,74–76 This capability is essential for characterizing cell-to-cell variability, identifying rare cell populations, and elucidating complex biological processes at the single-cell level. Moreover, microfluidic systems offer precise spatial and temporal control over cellular microenvironments, enabling researchers to mimic physiological conditions and study dynamic cellular processes in real time.77–79 By modulating fluid flow, chemical gradients, and environmental cues within microfluidic devices, researchers can investigate diverse aspects of cellular physiology, including cell migration, proliferation, differentiation, and signaling dynamics, with high spatiotemporal resolution.

In addition to facilitating functional assays and dynamic cell behavior studies, microfluidics has also profoundly advanced the field of single-cell omics by enabling the precise manipulation and processing of individual cells across multiple molecular dimensions. In single-cell genomics, Zahn et al. developed a microfluidic device comprising 192 nanoliter-scale chambers to perform direct library preparation (DLP) without the need for preamplification, enabling efficient whole-genome sequencing of hundreds of single cells.80 Building upon this approach, Laks et al. further introduced DLP+, an automated and scalable platform that integrates image-based single-cell identification and positioning, ultimately achieving processing of 51[thin space (1/6-em)]926 single-cell genomes across diverse sample types.81 Zheng et al. developed Microbe-seq, a droplet-based microfluidic platform that enables high-throughput single-microbe whole-genome sequencing by encapsulating individual microbes into droplets, followed by cell lysis, whole-genome amplification, and droplet-specific barcoding to achieve strain-level resolution in complex microbiomes.82 In transcriptomics, Bai et al. developed the dielectrophoresis (DEP)-trapping-nanowell-transfer (DEP-dTNT) platform, which utilizes electric field-driven DEP to actively trap single cells into nanowells, followed by hydrodynamic co-loading of barcoded beads.83 The paired cells and beads are then encapsulated within reaction chambers, enabling efficient mRNA capture and barcoding for downstream single-cell RNA sequencing. In proteomics, Zhu et al. developed the nanoPOTS (nanodroplet processing in one pot for trace samples) platform,84 which enables highly sensitive proteomic analysis within reaction volumes smaller than 200 nL, achieving the identification of ∼1500 to ∼3000 proteins from as few as ∼10 to 140 cells. Separately, Gebreyesus et al. introduced the single-cell integrated proteomic microfluidic chip (SciProChip),85 a fully integrated system that automates single-cell isolation, counting, imaging, and sample preparation on-chip, and is directly compatible with data-independent acquisition (DIA) mass spectrometry for high-throughput single-cell proteomics. More recently, DMF platforms such as active-matrix digital microfluidic chip for single-cell proteomics (AM-DMF-SCP)86 have demonstrated robust single-cell proteomic capabilities by enabling automated nanoliter-scale processing and label-free analysis, while addressing challenges of integration and sensitivity through active matrix control. In metabolomics analysis, researchers have developed approaches for intact single-cell metabolite profiling87–89 or targeted metabolite extraction followed by mass spectrometry (MS) analysis.90,91 More recently, microfluidic impedance cytometry has enabled one-step sample preparation for single-cell MS (>99% sorting/desalting, high purity), streamlining workflow and opening opportunities for multi-modal (electrical and metabolic) single-cell characterization.92 In lipidomics, microfluidic platforms coupled with mass spectrometry (e.g., MALDI-MS93 and DASEI-MS94) have enabled single-cell lipid profiling with high structural resolution. In addition, the digital microfluidic isolation of single cells for -omics (DISCO) platform95 leverages DMF in combination with AI-guided image processing and laser-based cell lysis to enable selective isolation and multi-omics analysis of individual cells, including their genomes, transcriptomes, and proteomes. Its tissue-resolved variant, tissue-DISCO (tDISCO),96 extends this capability to spatially contextualized cells within tissue slices, providing an integrated view of molecular profiles with spatial precision. For more detailed discussions on microfluidics-based omics strategies, readers are referred to recent reviews by Gebreyesus et al.97 and Zhang et al.,98 which comprehensively summarize microfluidic innovations in single-cell proteomics and multi-omics integration, respectively.

While microfluidics has profoundly advanced single-cell omics by enabling precise spatiotemporal control and high-throughput cellular profiling, its broader implementation outside of specialized bioengineering laboratories remains constrained by several practical limitations. The fabrication of PDMS-based devices often requires several hours to a full day of casting, aligning, bonding, and punching, that require specialized equipment, cleanroom access, and technical proficiency. These devices are frequently single-use and prone to failure, and the process is susceptible to chip-to-chip variability that can compromise reproducibility. Even after fabrication, fluid control systems (e.g., syringe pumps or pneumatic valves) and imaging or multi-omics integration require meticulous calibration and synchronization. Moreover, microfluidic experiments typically produce large, complex datasets, such as time-lapse microscopy or high-content omics, which demand sophisticated analytical pipelines and expertise in computational biology. These combined factors present significant barriers for many biology-focused labs. In this review, we not only discuss these limitations in detail but also highlight emerging solutions aimed at advancing and disseminating microfluidic technologies, such as standardized and automated platforms and data analysis tools, to make single-cell microfluidics more accessible, reproducible, and scalable.

2. Automation of microfluidics with robotics

Automating microfluidics with robotic operation seems straightforward, yet significant technical challenges remain. Microfluidic devices are predominantly fabricated using soft lithography with PDMS due to its biocompatibility, optical transparency, and gas permeability.99,100 The fabrication process involves molding PDMS on a photolithographically patterned master, curing, and manually peeling and punching to create inlets and outlets. These manual steps introduce device-device variability, compromising reproducibility and posing challenges for robotic operation. Moreover, microfluidic systems depend on tubing for precise fluid control and seamless integration with various instruments.101,102 While tubing provides adaptability, its connection and disconnection remain non-trivial for robotic handling, introducing additional complexity to fully automate workflows. Addressing these challenges requires advancements in standardized fabrication techniques and robotic dexterity for seamless microfluidic integration. Alternatively, non-PDMS microfluidic systems actuated by electrical or optical signals offer a promising solution to overcome the fabrication and operation challenges associated with PDMS-based platforms. Emerging digital microfluidics or microrobotic technologies have demonstrated initial success in achieving automation, yet there are still limitations in technical complexity, reliability, throughput, and scalability. In this section, we will highlight single-cell analysis advancements enabled by robotics.

2.1. Customized robotic systems for single-cell experiments

Despite existing challenges, several pioneering groups have developed customized robotic systems specifically designed for single-cell manipulation. As mentioned earlier, the nanoPOTS platform was built on a home-assembled robotic system comprising a high-precision syringe pump, a high-density nanowell-array chip with a droplet storage frame, an xyz translational stage, and a stereomicroscope (Fig. 2A).84,103–105 Building on this foundation, an automated, label-free nanoproteomics imaging approach was introduced to quantitatively map over 2000 proteins across mouse uterine tissue sections at a spatial resolution of 100 μm, enabling high-throughput, cell-type-specific spatial proteome analysis.106 Durrer et al. introduced the robot-assisted acoustofluidic end effector (RAEE) system,107 which merges robotics with acoustofluidics to overcome limitations in microscale liquid manipulation (Fig. 2B). Traditional lab processes rely on macroscale liquid handling, while lab-on-a-chip (LoC) technologies, despite their promise, often lack automation and adaptability. The RAEE system bridges this gap by integrating a robotic arm with an acoustofluidic end effector, enabling precise fluid pumping, particle trapping, embryo handling, and automated mixing of viscous liquids. Wang et al. developed the pick-up single-cell proteomic analysis (PiSPA) workflow,108 a probe-based single-cell proteomic platform built on the sequential operation droplet array (SODA) liquid handling system109 to enable nanoliter-scale cell capture, pretreatment, and liquid chromatography–mass spectrometer (LC–MS) injection (Fig. 2C). Target cells are aspirated individually and processed in insert tubes through in situ lysis, digestion, and other steps, minimizing sample loss and maximizing protein recovery. This workflow achieves deep proteome coverage, up to 3000 proteins per cell, and reveals cellular heterogeneity, as demonstrated in migrating HeLa cells.
image file: d5lc00216h-f2.tif
Fig. 2 Lab-built robotic systems for microfluidic operations in single-cell and microscale biological analysis. (A) The nanoPOTS platform integrates a high-precision syringe pump, nanowell chip, droplet capture frame, and stereomicroscope for picoliter-scale liquid dispensing in single-cell proteomics workflows.84 (B) A robot-assisted acoustofluidic end effector system, integrating a robotic arm, glass capillary, piezo transducer, and acoustofluidic device to enable high-precision fluid mixing, particle manipulation, and biological sample handling.107 (C) The PiSPA (pick-up single-cell proteomics analysis) system couples a capillary probe with a droplet-array-based liquid handling platform (SODA: sequential operation droplet array)105 to isolate and process individual cells through sequential in situ lysis, digestion, and LC–MS injection.108 (D) A compact robotic platform combining a pipette, syringe pump, onboard camera, and motion control system to automate microtissue handling in 384-well plates under sterile culture hood conditions.110 (E) The RoboCulture system for yeast cultivation integrates real-time optical feedback and force sensing with robotic tip exchange and liquid handling to enable fully autonomous, long-duration culture and monitoring.111 Created using BioRender, based on references.84,105,107,108,110,111

While not all robotic systems were originally designed for single-cell analysis, several recent innovations in robotic microfluidic handling demonstrate a clear trajectory toward clinical relevance and intelligent operation in the field. For example, Stepanov et al. developed a compact, low-cost robotic platform integrating a pipette, syringe pump, and onboard camera to manipulate live microtissues in 384-well plates (Fig. 2D).110 This system operates within standard tissue culture hoods without the need for pneumatic controllers or external microscopes, streamlining sterile workflows and enabling drug testing on limited patient biopsy samples. In another example, Angers et al. demonstrated RoboCulture, a fully automated platform for yeast cultivation that leverages real-time optical and mechanical feedback to guide experimental decisions over a continuous 15 hour protocol (Fig. 2E).111 While the system remains limited in throughput (5 seconds per tip change) and format (96-well plate), it exemplifies an emerging paradigm in robotic microfluidics, one where vision systems, real-time inference, and robotic learning converge to support fully autonomous biological experimentation. These precedents, though not tailored to single-cell analysis, underscore the feasibility and importance of combining robotics and AI to overcome the manual bottlenecks of microfluidic operation and pave the way for next-generation automated biological research.

2.2. Integration of commercial liquid-handling robotics with microfluidic single-cell systems

In addition to lab-built robotic systems, numerous commercialized robotic liquid handlers have been applied in the microfluidics field. Owing to their throughput and reliability, many of these systems are designed for well plate formats, such as 3D spheroid cultures using hanging drop arrays.112 For organ-on-a-chip applications, several research groups have developed their own robotic liquid handling platforms, for example, Novak et al. implemented a system for perfusing multiple vascularized organ chips,113 while Jiang et al. designed a robotic workflow for organoid-based assays.114 These platforms have significantly enhanced the throughput of tissue engineering applications and enabled the analysis of inter-organoid or inter-spheroid heterogeneity. However, they generally do not achieve single-cell resolution, as such drug screening applications often prioritize scalability over cellular-level precision.

To further extend the applicability of commercial robotic handlers in single-cell microfluidic systems, system-level optimizations are essential. One strategy involves encapsulating individual cells within water-in-oil droplets, facilitating precise reagent manipulation while preserving single-cell integrity and minimizing cross-contamination. Tran et al. demonstrated the automation of droplet microfluidics using commercial fluid-handling robotics (Freedom EVO, Tecan). Their approach, robotic operation of droplet microfluidics (RAD microfluidics), integrates key microfluidic components, such as droplet generators, mergers, and sorters, into fully automated workflows, significantly improving efficiency and scalability (Fig. 3A).115 Alternatively, employing high-precision robotic platforms such as cellenONE, which supports picoliter-to-nanoliter dispensing and integrated image-based cell selection, can improve compatibility with low-volume microfluidic environments. Ctortecka et al. developed an automated platform for multiplexed single-cell proteomics using a custom-designed microfabricated chip (proteoCHIP) coupled with the high-precision robotic system cellenONE. Their streamlined workflow achieves exceptional sensitivity with minimal sample loss (Fig. 3B).116 By eliminating manual handling and carrier proteome dependence, the platform enabled the identification of ∼2600 proteins across 170 single cells, with high reproducibility and over 90% data completeness per run. Another approach is to engineer microfluidic devices with structures such as confined migration channels or single-cell traps to ensure controlled cell passage, thereby enhancing throughput and automation across single-cell workflows. Zhou et al. developed a high-throughput microfluidic platform capable of tracking single-cell movement under multiple treatment conditions in a 384-well format, fully compatible with robotic liquid handling (Opentrons, OT-2 Liquid Handler) (Fig. 3C).117–119 This system incorporates autonomous image analysis software for real-time quantification of cell migration, encompassing image registration, quality control, channel segmentation, cell identification, and migration distance calculation in a semi-automated fashion requiring reduced human intervention, primarily for tasks such as plate handling, reagent loading, and experimental setup. Their innovative approach enabled the screening of 172 compounds for migration inhibition and toxicity, identifying promising low-toxicity regulators. With single-cell resolution, the system quantified both average migration distances and the behavior of top-ranked fast-moving cells, offering unparalleled insights into cell dynamics.


image file: d5lc00216h-f3.tif
Fig. 3 Applications of commercial liquid-handling robots in microfluidic single-cell analysis. (A) A droplet microfluidics system coupled with a commercial liquid handling robot (Freedom EVO, Tecan) and automated syringe pumps.115 (B) A nanoliter-scale single-cell proteomics workflow based on the proteoCHIP platform, integrated with the cellenONE robotic system. Single cells are dispensed into nanowells, followed by nanoliter-scale (nL) cell lysis and tryptic digestion, tandem mass tag (TMT) labeling for multiplexed quantification, and sample pooling for liquid chromatography–tandem mass spectrometry (LC–MS/MS) analysis.116 (C) A high-throughput single-cell migration platform capable of tracking tens of thousands of individual cells. The system combines a liquid handling robot (OT-2, Opentrons) with autonomous image analysis software for microfluidic single-cell analysis.117 Created using BioRender, based on references.115–117

Despite technical challenges in integrating microfluidic single-cell analysis with robotic operation, this convergence holds the potential to enhance throughput, reliability, and reproducibility. Standardization in microfluidic fabrication is crucial, with a shift toward precision-engineered components, such as those produced via injection molding, replacing labor-intensive manual soft lithography processes. Simultaneously, advancements in robotics will enable increasingly intelligent systems capable of precisely handling complex fluidic networks. While obstacles persist, the rapid evolution of both microfluidics and robotics is steadily bridging the gap.

2.3. On-chip micro-robots for sample manipulation

Beyond robotic operation of microfluidics, advances in micro-robotics now enable precise, programmable single-cell manipulation. Among these, electromagnetic field-based control has emerged as a leading strategy, offering high-speed, programmable, and reproducible fluidic operations with robust performance. Yu et al. developed a ferrobotic system using electrowetting-on-dielectric (EWOD) to achieve parallelized and sequential droplet control in digital microfluidics, drawing inspiration from automated guided vehicles.120 Lin et al. further enhanced this system by introducing a circuit board-based programmable platform, significantly improving SARS-CoV-2 amplification and detection efficiency (Fig. 4A).121 In digital microfluidics, on-chip ferrobots not only preserve but enhance the advantages of low sample consumption and high throughput, establishing an efficient, high-fidelity operational platform for point-of-care and other biomedical applications.
image file: d5lc00216h-f4.tif
Fig. 4 On-chip microrobots for single-cell manipulation and analysis. (A) A digital microfluidic ferrobotics system enables high-throughput, programmable fluidic operations for biomedical diagnostics, including SARS-CoV-2 amplification and detection.120,121 (B) An optoelectronic microrobot within an OET system achieves precise single-cell manipulation with minimal cellular damage. Light-controlled actuation allows targeted cell collection and transfer.124 (C) Optically actuated, TPP-fabricated soft microrobots enable gentle, high-precision single-cell manipulation using optical tweezers. The designs illustrate deformation under trapping beam-induced forces, leveraging wireframe structures for controlled cell manipulation.125 Created using BioRender, based on references.120,121,124,125

In addition to ferrobotic systems, Chiou et al. pioneered optoelectronic tweezer (OET), leveraging image-based DEP to modulate photoconductive materials and precisely manipulate particles and cells with an electric field.122 Zhang et al. advanced this concept with patterned OET (p-OET), integrating a continuous photoconductive layer to eliminate the need for constant illumination.123 This breakthrough enabled the development of optoelectronic micro-robots, achieving programmable, submillimeter-scale manipulation (Fig. 4B).124 These micro-robots generate larger, more uniform forces, minimize cellular damage, and offer flexible single-cell isolation for downstream applications such as clonal expansion and RNA sequencing.

Further expanding the capabilities of micro-robotic systems, soft micro-robots have emerged as an innovative approach for precise and stable control of cellular orientation under minimal forces (ranging from tens to hundreds of piconewtons). Soft micro-robots excel in collecting, sorting, transporting, and rotating individual cells with high sensitivity. Iványi et al. developed an elastic and deformable micro-robot using multiphoton polymerization, a technique that offers nanometer-scale resolution while minimizing biological damage (Fig. 4C).125 This micro-robot is capable of performing tasks such as collection, rotation, release, and pairing of single cells. Notably, its actuation mechanism is based on optical tweezers (OTs), where traction, pushing, pulling, and deformation forces are generated by two-photon polymerization (TPP)-fabricated bendable nanorods or torsion nanostrings. Compared to opto-thermal manipulation, this non-contact OT-based approach minimizes thermal effects on cells while allowing vertical cell positioning. Experimentally, these soft micro-robots have demonstrated successful operation within microfluidic chambers, significantly enhancing the sensitivity and precision of single-cell manipulation.

2.4. Combination of robotic single-cell operation with deep learning

Based on robotic single-cell operation, there are some pilot studies including deep learning in the pipeline. Jin et al. presents a label-free live-cell imaging method that leverages deep learning to predict cell phenotypes based on whole-transcriptome sequencing.126 This approach, enabled by the live imaging and cell picking system (ALPS), establishes a direct link between cell images and transcriptomic profiles, allowing real-time molecular characterization without compromising cell viability. This noninvasive and unbiased technique offers significant advantages for studying cell dynamics and functional heterogeneity. In parallel, Guo et al. introduced an artificial intelligence-assisted digital microfluidic framework (μDropAI) for multistate droplet control, which integrates semantic segmentation to recognize droplet morphology and interactions with an error rate <0.63%.127 By reducing the coefficient of variation of split droplet volumes to 2.74%, μDropAI improves precision in droplet manipulation and highlights the potential of semantic-driven DMF systems for fully automated and adaptive operation. Another breakthrough is a deep learning-based single-cell sorting platform developed by Guo et al.,128 which integrates YOLOv8, a real-time object detection system, with digital microfluidics to achieve highly efficient and pure (>96%) single-cell isolation. Unlike conventional fluorescence-activated (FACS) and magnetic-activated (MACS) cell sorting, this label-free approach enhances precision (98.5% accuracy) by utilizing safe interval path planning (SIPP) for optimized droplet trajectory control. Together, these advancements highlight the transformative potential of deep learning in noninvasive single-cell analysis and high-precision cell sorting.

2.5. Commercial platforms for single-cell analysis

In addition to academic publications (Table 1), new commercial systems provide solutions in single-cell workflows (Table 2). Despite their diversity, these systems share a common goal: to enhance precision, reproducibility, and throughput in single-cell assays. These platforms utilize core microfluidic principles, digital, droplet-based, optofluidic, or capillary-driven, to enable diverse omics workflows, including single-cell RNA sequencing, multi-omics integration, and high-sensitivity proteomics. High-throughput systems such as BOXmini SCP (ACX Instruments), Chromium (10× Genomics) and Beacon (Bruker) exemplify DMF, droplet and optofluidic solutions for multi-parameter profiling. Other platforms, such as CellenONE, F.SIGHT, Cell Handler, and DispenCell, are designed primarily for single-cell isolation and dispensing and operate in well-plate formats. These systems integrate imaging and AI-driven decision, making them well-suited for workflows that require precise cell selection and deposition into multi-well formats. Their compatibility with standard labware and omics workflows makes them practical tools for screening and targeted single-cell experiments. While most robotic liquid handling systems rely on physical pipette contact to aspirate and dispense fluids, raising concerns about cross-contamination and shear-induced cell damage, non-contact strategies have begun to emerge. In addition to DMF, Zhang et al. recently introduced an acoustofluidic liquid handling platform, PULSE, which enables precise, programmable ejection of nanodroplets and single cells via ultrasonic energy.129 Collectively, robotic and commercial systems represent two complementary directions in single-cell microfluidics: customized robotic platforms offer flexibility and depth for specialized applications, whereas commercial robotic solutions provide robust, standardized workflows suitable for broader adoption in research and clinical settings. As integration between robotics, microfluidics, and downstream omics continues to evolve, these platforms are poised to play a central role in the next generation of single-cell analysis.
Table 1 Comparison of robotic-microfluidic platforms for automated cell and omics analysis
Methods Robotics Omics/phenotypes Principle Sample type Advantages Limitations Throughput Ref
nanoPOTs Custom XYZ robotic microdispenser system Proteomics Nanowell Cells Extremely low sample loss; high sensitivity Requires custom-built platform Low 84
RAEE Dorna robotics arm Cell manipulation Acoustic streaming Cells, particles High-precision, label-free particle/cell manipulation; contactless operation High system complexity; low throughput Low 107
PiSPA Self-built SODA system Proteomics Droplet Cells High proteome coverage; precise single-cell capture Low throughput; robotic setup required Low 105, 108
Live microtissue manipulation Dobot MG400 robotic arm Drug response Liquid handling Micro dissected tissues Low cost and compact; high accuracy Not for single cell Medium 110
RoboCulture Franka Emika robot, Robotiq 2F-85 gripper Cell growth Cell culture Yeast cells Fully autonomous closed-loop system; real-time decision-making; compatible with standard labware Limited to specific cell types; not applicable to single-cell resolution Low–medium 111
RAD microfluidics Freedom EVO, Tecan Genomics; other omics with modifications Droplet Cells No user intervention theoretically Requires custom valve/pump interfacing hardware High 115
proteoCHIP CellenONE® Proteomics Nanowell Cells Low sample loss, scalable, fully automated proteomics workflow Endpoint analysis only; no cell recovery Medium–high 116
Single cell migration assay OT-2, Opentrons Cell migration Channel-based microfluidics Cells, drug/compounds High efficiency; compatible with imaging; semi-automated Difficult to retrieve individual cells; limited compatibility with multi-omics analysis Medium–high 117
Ferrobot Ferrofluid-driven magnetic microrobots MMP enzyme activity assay, nucleic acid detection DMF Human plasma, saliva Programmable; fully automated; low-cost; scalable for molecular diagnostics Moderate fabrication complexity; not yet adapted for single-cell resolution High 120, 121
OET & pOET SU8 fabricated-microrobot Transcriptome, cell–cell interaction Optoelectronic tweezers Cells, particles Programmable; non-contact; minimal cell damage Requires high-intensity light sources; limited speed and thermal effects High 124
Optically actuated soft microrobot TPP-fabricated microrobot Cell manipulation, cell–cell interaction Optical tweezers Cells, particles Low mechanical damage, high spatial precision, shape-adaptive Complex fabrication; low actuation speed limits real-time applications Low 125


Table 2 Commercial microfluidics platforms for single-cell analysis, isolation and dispensing
Category Platform Brand Principle Automation level Throughput Applications Features
Single-cell analysis BOXmini SCP ACX Instruments Digital microfluidics High High (1536 cells per run) Proteomics High-throughput single-cell proteomics sample preparation
Chromium 10× Genomics Droplet microfluidics High High (hundreds to millions of cells per run) Multi-omics High-throughput droplet-based single-cell encapsulation; supports scrna-seq, scatac-seq, multiome
Beacon Bruker Optofluidics High High (500 to 60[thin space (1/6-em)]000 cells per run) Genomics, transcriptomics Optofluidic live-cell culture and functional profiling; for antibody discovery, vaccine and t-cell screening
Single-cell isolation and dispensing CellShepherd ARRALYZE Microarray-based200 Medium Low (1 cell per minute) General cell applications Imaging-based cell tracking and isolation; AI-driven analysis
Cell Handler Yamaha Wellplate and capillary Medium/high Medium (8-channel tips; 96-/384-well plate) General cell applications Automated imaging and single-cell picking arm; Ai-integrated analysis, high resolution imaging
CellenONE SCIENION Capillary droplet generation High High (96 cells isolation/3 minutes) Multi-omics High-accuracy image-based isolation; temperature control; omics-compatible
F.SIGHT OMICS Cytena Cartridge microfluidics Medium/high High (384 cells isolation/8 minutes) Multi-omics Morphology- and fluorescence-based sorting; picoliter droplet generation
DispenCell S4 SEED Biosciences Wellplate and capillary Low/medium Medium (96 cells isolation/4 minutes) General cell applications Tip-based monoclonal cell isolation; semi-automated; for cell line development
CellRaft AIR System Cell Microsystem Microraft array Medium Low (96 cells isolation/1.5 hours) General cell applications Raft array-based clone identification; enhanced cell outgrowth
CellCelector Sartorius Wellplate and capillary Medium/high Low (1 cell isolation/20–30 seconds) General cell applications Image-guided robotic picking; monoclonal selection; well/nanowell compatibility


3. Deep learning for single-cell analysis

Deep learning has transformed single-cell analysis by enabling the extraction of meaningful insights from large-scale datasets. Convolutional neural networks (CNNs),130 generative adversarial networks (GANs),131 and variational autoencoders (VAEs)132 have emerged as key frameworks in this transformation. CNNs (Fig. 5A), which excel in image-based tasks, capture spatial hierarchies of cellular features, making them indispensable for classification and regression models. Classification models categorize cells based on morphological and molecular characteristics, while regression models predict continuous variables such as gene expression levels or phenotypic responses. Generative models, including GANs (Fig. 5B) and VAEs, further expand analytical capabilities by generating synthetic single-cell data, correcting batch effects, and simulating cellular states under diverse conditions. By integrating these advanced deep learning approaches, single-cell analysis achieves unprecedented accuracy and scalability, driving new discoveries in cellular heterogeneity and disease progression. Building on the transformative applications of deep learning in single-cell analysis, we will review key advancements in classification, regression, generative modeling, and data integration, highlighting their impact on cellular characterization and predictive modeling (Table 3).
image file: d5lc00216h-f5.tif
Fig. 5 AI models for single-cell analysis. (A) CNNs capture spatial hierarchies of cellular features, enabling precise cell classification and regression. Classification models distinguish cells by morphology and molecular traits, while regression models predict continuous variables like gene expression and phenotypic responses. (B) GANs consist of a generator that creates synthetic data and a discriminator that differentiates real from fake, engaging in an adversarial process to refine data generation. This framework enables high-fidelity single-cell data synthesis, correcting batch effects, and simulating cellular states under diverse conditions. Images were created using BioRender.
Table 3 Recent deep learning methods in single-cell analysis across segmentation, classification, regression, and generative modeling
Category Source (year) Models/methods Sample type Key advantage/findings Applications Limitations
Cell segmentation Cellpose 2.0 (2021)133 DNN + U-Net style architecture + residual blocks Fluorescence, and phase-contrast microscopy images Provides a pretrained model zoo to accommodate various microscopy modalities; uses a human-in-the-loop approach to simplify model generation Single-cell segmentation for diverse microscopy data Human-in-loop approach may be less effective for complex morphologies
LIVECell (2021)135 CNN-based segmentation model Phase-contrast microscopy images Enables label-free segmentation across diverse cell densities and morphologies Single-cell segmentation and tracking in live-cell imaging Performance depends on imaging quality; less effective for non-adherent cells
Din et al. (2021)136 Self-supervised CNN segmentation model Bright-field and fluorescence single-cell microscopy Achieves accurate segmentation without manual labels or extensive supervision Preprocessing for pipelines across modalities Sensitive to cell density and imaging conditions; cannot distinguish cells from background in certain cases
Classification Pattarone et al. (2021)139 CNN classifier (live vs. dead cells) Bright-field breast cancer cell images Achieves AUC of 0.94–0.98 without staining; suitable for live-cell analysis Label-free viability screening Specific to single cell lines; staining or capture methods may limit generalizability
Ulicna et al. (2021)140 U-Net + classifier + Bayesian tracking Time-lapse bright-field and fluorescence microscopy images Combines cell classification with tracking to improve cell lineage analysis accuracy Lineage tracking; single-cell studies Requires high-quality time-lapse imaging
Hartnett et al. (2022) – LANCE141 CNN classifier Label-free bright-field images of cell lines Provides 96.3 ± 0.5% accuracy in label-free bright-field imaging; tracks cell dynamics non-destructively Cell death monitoring; cytotoxicity assessment Sensitive to imaging conditions; may misclassify certain cell states
Regression Green et al. (2021)149 DNN + cGAN Zebrafish assay HTS chemical data Achieves AUC of 0.837 in consensus models; improves sensitivity in chemical screening Drug toxicity screening Requires accurate 3D chemical structures; cannot evaluate mixtures
Pham et al. (2021/22)150,151 GCN + feed forward neural network + interaction network + prediction network LINCS L1000, STRING, DrugBank, patient expression SARS-CoV-2 Outperforms state-of-the-art methods; suitable for drug repurposing Drug repurposing; prediction of gene expression changes Strongly influenced by input data quality
Chiang et al. (2024)153 CNN regression model Phase-contrast images of microfluidic spheroids Achieves correlation of r = 0.989 with LIVE/DEAD assays; transferable across laboratories High-throughput drug screening Imaging conditions may affect prediction
Generative modeling Palma et al. (2025)158 Generative modeling for morphology prediction Single-cell images under perturbations Predicts morphological changes under drug or genetic perturbations In silico screening of morphological effects Requires diverse perturbation training datasets
Xu et al. (2020) – scIGANs160 GAN-based imputation model Single-cell RNA-seq data Improves imputation accuracy and reduces sparsity in scRNA-seq data Enhanced scRNA-seq analysis and clustering Sensitive to input data quality; risk of over-smoothing
Lopez et al. (2018)163 VAE Single-cell RNA-seq data Learns latent structures; supports clustering and trajectory inference Unsupervised transcriptomics analysis Interpretation challenges; computationally intensive
Marouf et al. (2020)201 GAN/cscGAN Single-cell RNA-seq data Generates realistic scRNA-seq profiles; improves downstream analysis Data augmentation; rare-cell analysis Susceptible to mode collapse; high computational demand


3.1. Classification of cell states and fates

Classification models are essential for distinguishing cell states and fates in single-cell analysis. CNNs have been particularly effective in the recognition of cellular morphology with high precision. Deep learning methods like Cellpose and LIVECell have advanced cell segmentation in microscopy images. Cellpose,133,134 trained on over 70[thin space (1/6-em)]000 segmented objects, enables precise segmentation across diverse image types without retraining. LIVECell,135 a high-quality dataset of 1.6 million phase-contrast images, supports training models for accurate segmentation in variable cell morphologies and densities, improving high-throughput imaging. In addition, Din and Yu advanced single-cell segmentation by developing deep learning models capable of delineating cell boundaries with minimal human annotation.136

With segmented individual cells, there are multiple works that classify cell viability and damage severity, facilitating high-efficiency toxicity evaluation and the screening of potential anticancer drugs. For examples, Tox_(R)CNN was established by Jimenez-Carretero et al. to predict toxicity from images of DAPI-stained cells.137 Chen et al. developed a deep learning model that predicts tumorsphere formation on day 14 based on day 4 images, significantly accelerating cancer stem-like cell identification.138 Similarly, Pattarone et al. established a deep learning framework for classifying live and dead breast cancer cells without staining, expanding the applicability of label-free cell classification.139 Ulicna et al. developed a Bayesian-based single-cell tracking approach that performs lineage tree analysis, by utilizing cell state classification which in turn improves tracking of cells in both 2D and 3D environments.140 Hartnett et al. introduced live, apoptotic, and necrotic cell explorer (LANCE), a CNN-based model that classifies apoptotic and necrotic cells based on brightfield microscopy images, with an accuracy of 96.3 ± 0.5%, eliminating the need for fluorescence labeling.141 He et al. developed a deep learning model, named detector of mitosis, apoptosis, interphase, necrosis, and senescence (D-MAINS), which leverages cellular morphology in phase contrast images for accurate cell status classification.142 Li et al. developed an AI-based approach for detecting cell damage, utilizing a CNN trained on time-series fluorescence images collected before and after drug exposure, achieving an accuracy of over 93%.143 These breakthroughs underscore the power of deep learning in advancing classification tasks for single-cell analysis. However, challenges persist in dataset annotation, model interpretability, and the generalizability of predictive accuracy across diverse cell models. To address these limitations, strategies such as transfer learning144,145 and self-supervised learning146,147 are being explored to enhance model robustness across diverse experimental conditions.

3.2. Regression to assess continuous biological variables

Regression models play a pivotal role in single-cell analysis by predicting continuous biological variables, offering deeper quantitative insights beyond discrete classification. These methods are particularly valuable for high-throughput studies, where understanding heterogeneous cellular responses is essential. Zhang et al. applies random decision forest and artificial neural network to microfluidic single-cell migration data, and the model achieved high accuracy in predicting cell movement direction (over 99%) and speed (91%), identifying key morphological features that drive cell migration and metastasis.148 Green et al. leveraged deep neural networks (DNNs) and conditional generative adversarial networks (cGANs) to predict toxicological responses from large-scale screening data, improving drug candidate prioritization, achieving an area under the receiver operating characteristic of 0.837.149 Pham et al. introduced DeepCE, a deep-learning framework that predicts chemical-induced gene expression changes in a cell-type-specific manner, facilitating drug discovery and toxicology assessments and supporting drug repurposing for COVID-19 (ref. 150 and 151) Lakkis et al. introduced single cell imputation protein embedding neural network (sciPENN),152 a versatile deep learning framework designed for the integration of CITE-seq and scRNA-seq data, enabling protein expression prediction for scRNA-seq and protein expression imputation for CITE-seq. Zhang et al. (1920 tumor spheres) and Chiang et al. (12[thin space (1/6-em)]000 spheroids per chip) leveraged deep learning for label-free viability assessment of cancer spheroids in microfluidic platforms, reducing reliance on fluorescence staining while maintaining high classification accuracy.153,154 Ma et al. systematically predicted the efficacy of >6000 compounds against therapy-resistant polyploid giant cancer cells.155 Despite their promise, single-cell regression models face challenges such as biological noise, batch effects, and variability in experimental conditions. To mitigate these issues, the incorporation of more diverse single-cell datasets, along with the application of data augmentation, ensemble learning, and regularization techniques, is anticipated to improve model robustness and enhance predictive accuracy.

3.3. Generative modeling for single-cell analysis

Generative models, including GANs and VAEs, have emerged as powerful tools in single-cell analysis, enabling data augmentation, image denoising, and cellular behavior simulation. These models significantly enhance computational biology by facilitating synthetic data generation and improving analytical methodologies. In image-based single-cell analysis, Witmer and Bhanu employed GANs for dataset augmentation,156 improving classification accuracy and achieving a 2% increase in both true positive rate and F1-score compared to imbalanced datasets. Similarly, Wu et al. developed a GAN-based method to generate synthetic images of human cardiomyocytes at various maturation stages,157 enhancing classification accuracy and cellular structure analysis, outperforming conventional machine learning approaches. Palma et al. introduced the image perturbation autoencoder (IMPA)158 to predict morphological changes in response to genetic and chemical perturbations, improving high-content screening analysis by accounting for batch effects and technical variations, making it a valuable tool for drug discovery. Additionally, Ternes et al. developed a multi-encoder VAE,159 improving single-cell image analysis by extracting biologically relevant features, enhancing cell population separation, phenotypic distinctions, and correlation with other analytical methods. Beyond morphological analysis, generative models also aid in scRNA-seq, which, despite its ability to characterize transcriptomic profiles at high throughput, faces challenges such as dropout errors and technical noise. To address these limitations, GANs, including scRNA-seq imputation GANs (scIGANs)160 and conditional single-cell GANs (cscGANs),161 generate realistic imputed or augmented single-cell RNA-seq data, improving downstream analyses, rare cell population detection, and classification robustness. These models also mitigate data sparsity, enhance expression quality, and reduce reliance on biological samples, improving reproducibility and cost efficiency in biomedical research. Overall, GANs and VAEs are transforming single-cell analysis by improving data integrity, addressing technical limitations, and expanding analytical capabilities, paving the way for more accurate and scalable biomedical research.

3.4. The role of AI in enhancing single-cell data integration and analysis

AI is also playing an increasingly central role in the integration of multi-omics single-cell data by addressing challenges spanning data generation, integration, storage, analysis, and interpretation. In data generation, AI-driven experimental design and active learning methods help optimize sampling strategies, enabling researchers to prioritize informative cell populations or experimental conditions that maximize biological insight while reducing experimental costs and redundancy.162 For example, adaptive sampling algorithms can guide iterative single-cell RNA sequencing to focus on rare or transitional cell states. In the realm of data integration, cutting-edge AI models such as variational autoencoders (e.g., scVI163 and TotalVI164) and graph neural networks165,166 have proven effective at aligning and integrating diverse modalities including transcriptomics, proteomics, and epigenomics, enabling comprehensive characterization of cellular states across multiple molecular layers. These models can overcome batch effects and technical noise, harmonizing datasets collected across different platforms or time points.

Scalable data storage, transfer, and archiving infrastructures, combined with standardized metadata frameworks (such as the Human Cell Atlas metadata standards167,168), are critical to support reproducible AI-driven analyses and facilitate large-scale data sharing. To enable efficient AI applications, curated and well-annotated databases, like the Single Cell Expression Atlas169 and Tabula Muris,170 are developed with AI-friendly formats, accompanied by modular, reproducible pipelines (e.g., Scanpy171 and Seurat172) that streamline preprocessing and downstream machine learning analyses. AI-powered quality control tools automatically detect technical artifacts, low-quality cells, and batch effects; methods like DoubletFinder173 and SoupX174 are widely used to clean single-cell datasets prior to analysis.

Importantly, advances in explainable AI are making it possible to interpret complex models and extract biologically meaningful insights. For example, CellOracle175 uses machine learning to infer gene regulatory networks and predict cellular responses to perturbations, providing mechanistic insights rather than black-box predictions. Similarly, scMoMaT176 integrates multi-omics single-cell data to identify coordinated regulatory programs, facilitating hypothesis generation grounded in interpretable features. Together, these developments demonstrate AI's transformative potential to not only enhance technical aspects of data handling but also to enable deeper biological discovery through integrative and interpretable analyses in single-cell and multi-omics research.

3.5. Barriers to reliable AI in single-cell microfluidics

Despite their impressive performance, current AI pipelines for single-cell imaging and microfluidic read-outs still suffer from a number of limitations that negatively affect scientific transparency and reproducibility.177 The deeply layered, “black-box” nature of CNNs, GANs, and transformer models obscures the mechanistic links between an input image and the model's prediction, making it hard for researchers or regulators to verify AI-generated hypotheses or understand the thought process behind certain classifications or features that might be selected for training the models. Because models typically demand large, expertly annotated datasets, any bias or noise introduced during annotation, image acquisition, or batch processing can propagate through training and destabilize downstream results. Even when sizeable datasets exist (e.g., Cellpose or LIVECell), differences in microscope settings, cell lines, or microfluidic chip geometries often erode model generalizability, forcing each laboratory to perform costly re-training and hyper-parameter tuning that are rarely described in sufficient detail for others to reproduce. The heavy computational footprint of state-of-the-art networks further complicates transparency: model weights, training logs, and version-specific software dependencies are rarely saved, so a result obtained on one graphics processing unit (GPU) stack may be irrecoverable on another. Finally, the field lacks standardized benchmarks for evaluating continuous-variable regressors (e.g., viability scores or migration velocities), leaving researchers to report disparate metrics that impede cross-study comparison. Addressing these gaps will require open, modality-matched reference datasets, shareable training pipelines with locked random seeds and environment files, and the wider adoption of interpretable, self-supervised, or transfer-learning frameworks that can be audited and stress-tested across laboratories before their biological conclusions are trusted.

4. Potential role of cloud lab concepts in single-cell experimentation

Despite the transformative potential of integrating microfluidics, robotics, and AI in single-cell analysis, technical and operational challenges remain significant. These complexities have prompted interest in a new envisioned model: the cloud lab.178–180 Cloud labs (Fig. 6) have been described as centralized facilities equipped with sophisticated instrumentation and infrastructure that researchers can access remotely.181–183 Through cloud interfaces, scientists can submit standardized protocols and samples, receive real-time or asynchronous experimental outputs, and conduct downstream computational analyses, enabling a degree of experimentation without direct physical presence. This model holds promise for supporting single-cell workflows, where robotic execution of highly repetitive and sensitive steps, such as droplet generation, reagent dispensing, and high-content imaging, could benefit from automation and central standardization if realized. In practice, however, many upstream elements of single-cell experiments, such as microfluidic chip fabrication, reagent formulation, and biological sample preparation, still demand expert manual handling and cannot yet be fully automated. As such, cloud labs should currently be seen as complementary to traditional laboratories, providing infrastructure for partial automation, protocol standardization, and collaborative data analysis, rather than serving as complete replacements. Cloud labs offer advantages in enhancing reproducibility and scalability in robotic-driven workflows. By centralizing the execution of experimental protocols and integrating with cloud-based data processing pipelines, they can minimize human variability in routine tasks and improve consistency across different users and institutions. For example, robotic liquid handling and imaging systems operated within cloud lab environments can deliver consistent performance under tightly controlled conditions, reducing experimental drift over time and variations between laboratories, which is critical for deep learning data analysis.
image file: d5lc00216h-f6.tif
Fig. 6 Cloud Lab synergizing microfluidics, robotics, and AI for accelerated single-cell discovery. The Cloud Lab concept connects laboratory protocol management, single-cell microfluidics, robotic operation, and artificial intelligence into a unified workflow. Laboratory modules manage experimental protocols, while microfluidic systems enable high-throughput single-cell data collection. Robotic platforms execute automated workflows, and AI modules generate and refine hypotheses based on acquired data. Images were created using BioRender.

From a collaborative perspective, cloud labs are especially useful within structured partnerships or consortia, where institutions can share expertise and infrastructure under clearly defined governance models. In such frameworks, cloud labs act as intermediaries, allowing remote users to leverage specialized instrumentation managed by trained personnel. Commercial platforms (e.g., Strateos, Emerald Cloud Lab) have provided early demonstrations to operate this model through subscription or usage-based services, offering access to robotics and analytical infrastructure for research groups without direct access to such resources. While the financial expense of cloud lab access varies depending on experimental complexity and support needs, this shared-access model has the potential to lower entry barriers for technically demanding workflows such as single-cell assays.

Importantly, while cloud labs offer a forward-looking infrastructure, claims that they can fully eliminate physical laboratory needs or democratize all aspects of experimental biology should be considered in light of real-world constraints, including cost, technical complexity, and the need for real-time decision-making in dynamic biological experiments. A more realistic and achievable path lies in hybrid integration, in which cloud-connected systems enhance reproducibility, enable remote collaboration, and operate standardized components of experiments, while still relying on local expertise for complex, hands-on steps. Overall, cloud labs represent an envisioned framework that may, in the future, support single-cell research, particularly in the realms of standardized data acquisition, robotics, and inter-institutional collaboration. While challenges remain in areas such as device fabrication and biological sample handling, cloud labs may ultimately augment traditional laboratory workflows, acting as powerful facilitators of experimental rigor, scalability, and accessibility.

5. Conclusion and perspective: challenges and opportunities in integrating AI and robotics for advancing single-cell analysis

Currently, AI and robotics are transforming single-cell research, with their potential realized through integration across three critical domains: (1) robotic generation of large-scale datasets for deep learning, (2) rapid validation of novel hypotheses generated by AI, and (3) iterative feedback to enhance model refinement. Robotics is indispensable for creating high-quality, large-scale datasets required to train and optimize AI models. For example, microfluidic platforms can process thousands of single cells simultaneously, generating vast datasets for transcriptomics, proteomics, or metabolomics. These datasets feed into AI algorithms, sharpening their predictive capabilities and enabling the generation of increasingly complex and innovative hypotheses. Recent developments in deep visual proteomics (DVP)184 and single-cell DVP (scDVP)185 demonstrate how AI-guided image processing and robotic microdissection can link spatial cellular phenotypes to proteomic profiles at single-cell resolution, uncovering spatially regulated disease mechanisms in complex tissues. These approaches illustrate the promise of this technological convergence.

However, this vision is also constrained by both technical and conceptual limitations. One major challenge is the annotation burden: biological datasets often require expert labeling to train supervised models, which become particularly onerous in single-cell contexts where morphological, molecular, and functional heterogeneity is high. Moreover, many AI models, especially deep neural networks, function as “black boxes”, raising concerns about interpretability and the transparency of biological insights they generate. Without a clear understanding of how predictions are derived, it can be difficult to assess their scientific validity or clinical relevance. Furthermore, even with automation, reproducibility in AI-driven workflows remains a concern, particularly when predictions rely on complex models trained with non-standardized preprocessing pipelines or limited sample diversity. These issues may undermine confidence in AI-generated hypotheses unless carefully addressed through rigorous experimental design and benchmarking.

Looking ahead, the iterative feedback loop between AI and robotics represents an envisioned paradigm for biomedical research (Fig. 6). Robotic experimentation generates data that improves AI models, while AI refines hypotheses that drive further experimentation. This self-reinforcing cycle, although not yet fully realized, could accelerate discovery, enabling deeper insights and breakthroughs that would be unattainable with either technology alone. Together, AI and robotics are poised to revolutionize single-cell analysis and redefine biomedical innovation. The symbiotic relationship between AI and robotics can also extend to the development of new experimental techniques. Robotics can facilitate the design and testing of novel methodologies proposed by unsupervised AI models.186 For instance, AI might suggest unconventional ways to sort or label cells based on subtle, previously unrecognized features through unsupervised learning. Robotic platforms can implement these techniques, generating data that not only validates the methodology but also expands the boundaries of what is experimentally possible. This iterative process continuously pushes the limits of single-cell analysis, enabling breakthroughs that were previously inconceivable.

Ultimately, the convergence of microfluidics, robotics, and AI is poised to reshape single-cell analysis. While each of these technologies has independently contributed to advances in throughput, precision, and discovery, their true transformative power lies in their integration. However, the field remains fragmented, with relatively few platforms offering end-to-end solutions from sample preparation to hypothesis generation and validation. We anticipate a shift from Siloed tool development to modular, interoperable ecosystems, where standardization, cloud-based experimentation, and real-time AI feedback loops will be central. Importantly, the next phase of innovation will demand more than technical optimization; it will require frameworks for interpretability, reproducibility, and deployment of autonomous decision-making in biological research. Rather than a linear progression, we foresee the field evolving into a multidimensional space, one that combines biological insight, engineering, and responsible AI. In this light, integration should be viewed not as an accomplished reality, but as a forward-looking vision that redefines how discovery in life sciences may be achieved in the future.

Abbreviations

AM-DMF-SCPActive-matrix digital microfluidic chip for single-cell proteomics
AIArtificial intelligence
AIBOArtificial intelligence robot
ALPSAutomated live imaging and cell picking system
BERTBidirectional encoder representations from transformers
CITE-seqCellular indexing of transcriptomes and epitopes by sequencing
CCDCharge-coupled device
cscGANsConditional single-cell GANs
cGANConditional GAN
CNNConvolutional neural network
CVsCoefficients of variation
DIAData-independent acquisition
DNNDeep neural network
DVPDeep visual proteomics
DMFDigital microfluidics
DISCODigital microfluidic isolation of single cells for -omics
DEPDielectrophoresis
DEP-dTNTDielectrophoresis-trapping-nanowell-transfer
DLPDirect library preparation
DASEI-MSDroplet-assisted electrospray ionization-MS
Drop-seqDroplet-based single-cell RNA Sequencing
EWODElectrowetting-on-dielectric
FACSFluorescence-activated cell sorting
GANGenerative adversarial network
GPUGraphics processing unit
IMPAImage perturbation autoencoder
LoCLab-on-a-chip
LLMLarge language model
LC–MSLiquid chromatography–mass spectrometer
LANCELive, apoptotic, and necrotic cell explorer
MACSMagnetic-activated cell sorting
MSMass spectrometry
MALDI MSMatrix-assisted laser desorption/ionization-MS
nanoPOTSNanodroplet processing in one pot for trace samples
OTOptical tweezer
OETOptoelectronic tweezer
p-OETPatterned OET
PR2Personal robot 2
PiSPAPick-up single-cell proteomic analysis
POCTPoint-of-care testing
PDMSPolydimethylsiloxane
RAEERobot-assisted acoustofluidic end effector
RNARibonucleic acid
RAD microfluidicsRobotics automation of droplet microfluidics
ROSRobot operating system
RT-1Robotic transformer-1
SIPPSafe interval path planning
scIGANsscRNA-seq imputation GANs
scRNA-seqSingle-cell RNA sequencing
SciProChipSingle-cell integrated proteomic microfluidic chip
sciPENNSingle cell imputation protein embedding neural network
SODAsequential operation droplet array
scDVPSingle-cell DVP
TMTTandem mass tag
tDISCOTissue-DISCO
TILsTumor-infiltrating lymphocytes
TPPTwo-photon polymerization
VAEVariational autoencoder
VIVariational inference
YOLOYou only look once

Author contributions

Jinxiong Cheng: conceptualization, visualization, writing – original draft, writing – review & editing. Rajiv Anne: conceptualization, visualization, writing – original draft. Yu-Chih Chen: conceptualization, funding acquisition, project administration, resources, supervision, writing – original draft, writing – review & editing.

Conflicts of interest

The authors declare that no competing interests exist.

Data availability

No primary research results, software or code have been included and no new data were generated or analysed as part of this review.

Acknowledgements

This study was generously funded by start-up support from the UPMC Hillman Cancer Center, the Women's Cancer Research Center (WCRC) at Magee Women's Research Institute, and the US National Institutes of Health grants P30 CA047904, P50 CA272218, R35 GM150509, and R21CA293424. We acknowledge the use of BioRender (https://www.biorender.com) for creating schematic figures. The authors utilized ChatGPT 4o to enhance the readability and language of this work. Following its use, the authors thoroughly reviewed and edited the content as necessary and take full responsibility for the content of the publication.

References

  1. Y. Xia and G. M. Whitesides, Soft Lithography, Angew. Chem., Int. Ed., 1998, 37(5), 550–575 CrossRef CAS PubMed.
  2. M. A. Unger, H. P. Chou, T. Thorsen, A. Scherer and S. R. Quake, Monolithic microfabricated valves and pumps by multilayer soft lithography, Science, 2000, 288(5463), 113–116 CrossRef CAS PubMed.
  3. S. K. Dertinger, X. Jiang, Z. Li, V. N. Murthy and G. M. Whitesides, Gradients of substrate-bound laminin orient axonal specification of neurons, Proc. Natl. Acad. Sci. U. S. A., 2002, 99(20), 12542–12547 CrossRef CAS PubMed.
  4. G. M. Whitesides, E. Ostuni, S. Takayama, X. Jiang and D. E. Ingber, Soft lithography in biology and biochemistry, Annu. Rev. Biomed. Eng., 2001, 3, 335–373 CrossRef CAS PubMed.
  5. S. K. Cho, H. J. Moon and C. J. Kim, Creating, transporting, cutting, and merging liquid droplets by electrowetting-based actuation for digital microfluidic circuits, J. Microelectromech. Syst., 2003, 12(1), 70–80 CrossRef.
  6. K. Choi, A. H. Ng, R. Fobel and A. R. Wheeler, Digital microfluidics, Annu. Rev. Anal. Chem., 2012, 5, 413–440 CrossRef CAS PubMed.
  7. M. G. Pollack, R. B. Fair and A. D. Shenderov, Electrowetting-based actuation of liquid droplets for microfluidic applications, Appl. Phys. Lett., 2000, 77(11), 1725–1726 CrossRef CAS.
  8. R. Zilionis, J. Nainys, A. Veres, V. Savova, D. Zemmour, A. M. Klein and L. Mazutis, Single-cell barcoding and sequencing using droplet microfluidics, Nat. Protoc., 2017, 12(1), 44–73 CrossRef CAS PubMed.
  9. E. Z. Macosko, A. Basu, R. Satija, J. Nemesh, K. Shekhar, M. Goldman, I. Tirosh, A. R. Bialas, N. Kamitaki, E. M. Martersteck, J. J. Trombetta, D. A. Weitz, J. R. Sanes, A. K. Shalek, A. Regev and S. A. McCarroll, Highly Parallel Genome-wide Expression Profiling of Individual Cells Using Nanoliter Droplets, Cell, 2015, 161(5), 1202–1214 CrossRef CAS PubMed.
  10. M. Stoeckius, C. Hafemeister, W. Stephenson, B. Houck-Loomis, P. K. Chattopadhyay, H. Swerdlow, R. Satija and P. Smibert, Simultaneous epitope and transcriptome measurement in single cells, Nat. Methods, 2017, 14(9), 865–868 CrossRef CAS PubMed.
  11. D. Yoon No, K. H. Lee, J. Lee and S. H. Lee, 3D liver models on a microplatform: well-defined culture, engineering of liver tissue and liver-on-a-chip, Lab Chip, 2015, 15(19), 3822–3837 RSC.
  12. G. Agrawal, A. Aung and S. Varghese, Skeletal muscle-on-a-chip: an in vitro model to evaluate tissue formation and injury, Lab Chip, 2017, 17(20), 3447–3461 RSC.
  13. T. Satoh, S. Sugiura, K. Shin, R. Onuki-Nagasaki, S. Ishida, K. Kikuchi, M. Kakiki and T. Kanamori, A multi-throughput multi-organ-on-a-chip system on a plate formatted pneumatic pressure-driven medium circulation platform, Lab Chip, 2017, 18(1), 115–125 RSC.
  14. K. H. Benam, R. Villenave, C. Lucchesi, A. Varone, C. Hubeau, H. H. Lee, S. E. Alves, M. Salmon, T. C. Ferrante, J. C. Weaver, A. Bahinski, G. A. Hamilton and D. E. Ingber, Small airway-on-a-chip enables analysis of human lung inflammation and drug responses in vitro, Nat. Methods, 2016, 13(2), 151–157 CrossRef CAS PubMed.
  15. M. M. Veloso, P. E. Rybski, S. Lenser, S. Chernova and D. Vail, CMRoboBits: Creating an intelligent AIBO robot, AI Mag., 2006, 27(1), 67–82 Search PubMed.
  16. J. Forlizzi and C. DiSalvo, Service robots in the domestic environment: a study of the roomba vacuum in the home, in Proceedings of the 1st ACM SIGCHI/SIGART conference on Human-robot interaction, 2006, pp. 258–265 Search PubMed.
  17. S. Cousins, ROS on the PR2, IEEE Robot. Autom. Mag., 2010, 17(3), 23–25 Search PubMed.
  18. M. Khosravi, Z. Zare, S. M. Mojtabaeian and R. Izadi, Artificial Intelligence and Decision-Making in Healthcare: A Thematic Analysis of a Systematic Review of Reviews, Health Serv. Res. Manag. Epidemiol., 2024, 11, 23333928241234863 Search PubMed.
  19. R. D. King, K. E. Whelan, F. M. Jones, P. G. Reiser, C. H. Bryant, S. H. Muggleton, D. B. Kell and S. G. Oliver, Functional genomic hypothesis generation and experimentation by a robot scientist, Nature, 2004, 427(6971), 247–252 CrossRef CAS PubMed.
  20. L. N. Soldatova, A. Clare, A. Sparkes and R. D. King, An ontology for a Robot Scientist, Bioinformatics, 2006, 22(14), e464–e471 CrossRef CAS PubMed.
  21. R. D. King, J. Rowland, S. G. Oliver, M. Young, W. Aubrey, E. Byrne, M. Liakata, M. Markham, P. Pir, L. N. Soldatova, A. Sparkes, K. E. Whelan and A. Clare, The automation of science, Science, 2009, 324(5923), 85–89 CrossRef CAS PubMed.
  22. K. Williams, E. Bilsland, A. Sparkes, W. Aubrey, M. Young, L. N. Soldatova, K. De Grave, J. Ramon, M. de Clare, W. Sirawaraporn, S. G. Oliver and R. D. King, Cheaper faster drug development validated by the repositioning of drugs against neglected tropical diseases, J. R. Soc., Interface, 2015, 12(104), 20141289 CrossRef PubMed.
  23. J. Savall, E. T. W. Ho, C. Huang, J. R. Maxey and M. J. Schnitzer, Dexterous robotic manipulation of alert adult Drosophila for high-content experimentation, Nat. Methods, 2015, 12(7), 657–660 CrossRef CAS PubMed.
  24. N. Yachie, C. Robotic Biology and T. Natsume, Robotic crowd biology with Maholo LabDroids, Nat. Biotechnol., 2017, 35(4), 310–312 CrossRef CAS PubMed.
  25. D. W. McClymont and P. S. Freemont, With all due respect to Maholo, lab automation isn't anthropomorphic, Nat. Biotechnol., 2017, 35(4), 312–314 CrossRef CAS PubMed.
  26. S. Asche, G. J. T. Cooper, G. Keenan, C. Mathis and L. Cronin, A robotic prebiotic chemist probes long term reactions of complexifying mixtures, Nat. Commun., 2021, 12(1), 3547 CrossRef CAS PubMed.
  27. A. M. Lunt, H. Fakhruldeen, G. Pizzuto, L. Longley, A. White, N. Rankin, R. Clowes, B. Alston, L. Gigli, G. M. Day, A. I. Cooper and S. Y. Chong, Modular, multi-robot integration of laboratories: an autonomous workflow for solid-state chemistry, Chem. Sci., 2024, 15(7), 2456–2463 RSC.
  28. T. Song, M. Luo, X. Zhang, L. Chen, Y. Huang, J. Cao, Q. Zhu, D. Liu, B. Zhang, G. Zou, G. Zhang, F. Zhang, W. Shang, Y. Fu, J. Jiang and Y. Luo, A Multiagent-Driven Robotic AI Chemist Enabling Autonomous Chemical Research On Demand, J. Am. Chem. Soc., 2025, 147(15), 12534–12545 CrossRef CAS PubMed.
  29. J. M. Gudbergsson, S. Kostrikov, K. B. Johnsen, F. P. Fliedner, C. B. Stolberg, N. Humle, A. E. Hansen, B. W. Kristensen, G. Christiansen, A. Kjaer, T. L. Andresen and M. Duroux, A tumorsphere model of glioblastoma multiforme with intratumoral heterogeneity for quantitative analysis of cellular migration and drug response, Exp. Cell Res., 2019, 379(1), 73–82 CrossRef CAS PubMed.
  30. P. H. Wu, A. Giri, S. X. Sun and D. Wirtz, Three-dimensional cell migration does not follow a random walk, Proc. Natl. Acad. Sci. U. S. A., 2014, 111(11), 3949–3954 CrossRef CAS PubMed.
  31. T. Kwon, O. S. Kwon, H. J. Cha and B. J. Sung, Stochastic and Heterogeneous Cancer Cell Migration: Experiment and Theory, Sci. Rep., 2019, 9(1), 16297 CrossRef PubMed.
  32. H. Shafqat-Abbasi, J. M. Kowalewski, A. Kiss, X. Gong, P. Hernandez-Varas, U. Berge, M. Jafari-Mamaghani, J. G. Lock and S. Stromblad, An analysis toolbox to explore mesenchymal migration heterogeneity reveals adaptive switching between distinct modes, eLife, 2016, 5, e11384 CrossRef PubMed.
  33. Y. C. Chen, S. G. Allen, P. N. Ingram, R. Buckanovich, S. D. Merajver and E. Yoon, Single-cell Migration Chip for Chemotaxis-based Microfluidic Selection of Heterogeneous Cell Populations, Sci. Rep., 2015, 5, 9980 CrossRef CAS PubMed.
  34. Y. C. Chen, B. Humphries, R. Brien, A. E. Gibbons, Y. T. Chen, T. Qyli, H. R. Haley, M. E. Pirone, B. Chiang, A. Xiao, Y. H. Cheng, Y. Luan, Z. Zhang, J. Cong, K. E. Luker, G. D. Luker and E. Yoon, Functional Isolation of Tumor-Initiating Cells using Microfluidic-Based Migration Identifies Phosphatidylserine Decarboxylase as a Key Regulator, Sci. Rep., 2018, 8(1), 244 CrossRef.
  35. S. Huang, Non-genetic heterogeneity of cells in development: more than just noise, Development, 2009, 136(23), 3853–3862 CrossRef CAS PubMed.
  36. A. Raj and A. van Oudenaarden, Nature, nurture, or chance: stochastic gene expression and its consequences, Cell, 2008, 135(2), 216–226 CrossRef CAS PubMed.
  37. P. B. Gupta, C. M. Fillmore, G. Jiang, S. D. Shapira, K. Tao, C. Kuperwasser and E. S. Lander, Stochastic state transitions give rise to phenotypic equilibrium in populations of cancer cells, Cell, 2011, 146(4), 633–644 CrossRef CAS PubMed.
  38. E. H. Finn and T. Misteli, Molecular basis and biological function of variability in spatial genome organization, Science, 2019, 365(6457), eaaw9498 CrossRef CAS PubMed.
  39. B. L. Emert, C. J. Cote, E. A. Torre, I. P. Dardani, C. L. Jiang, N. Jain, S. M. Shaffer and A. Raj, Variability within rare cell states enables multiple paths toward drug resistance, Nat. Biotechnol., 2021, 39(7), 865–876 CrossRef CAS.
  40. L. Heumos, A. C. Schaar, C. Lance, A. Litinetskaya, F. Drost, L. Zappia, M. D. Lucken, D. C. Strobl, J. Henao, F. Curion, H. B. Schiller and F. J. Theis, Single-cell Best practices for single-cell analysis across modalities, Nat. Rev. Genet., 2023, 24(8), 550–572 CrossRef CAS PubMed.
  41. D. Jovic, X. Liang, H. Zeng, L. Lin, F. Xu and Y. Luo, Single-cell RNA sequencing technologies and applications: A brief overview, Clin. Transl. Med., 2022, 12(3), e694 CrossRef CAS PubMed.
  42. I. Dagogo-Jack and A. T. Shaw, Tumour heterogeneity and resistance to cancer therapies, Nat. Rev. Clin. Oncol., 2018, 15(2), 81–94 CrossRef CAS PubMed.
  43. C. E. Meacham and S. J. Morrison, Tumour heterogeneity and cancer cell plasticity, Nature, 2013, 501(7467), 328–337 CrossRef CAS PubMed.
  44. P. R. Prasetyanti and J. P. Medema, Intra-tumor heterogeneity from a cancer stem cell perspective, Mol. Cancer, 2017, 16(1), 41 CrossRef PubMed.
  45. D. G. Tang and T. Kondo, Cancer cell heterogeneity and plasticity: From molecular understanding to therapeutic targeting, Semin. Cancer Biol., 2022, 82, 1–2 CrossRef CAS PubMed.
  46. L. A. Schulte, J. C. Lopez-Gil, B. Sainz Jr and P. C. Hermann, The Cancer Stem Cell in Hepatocellular Carcinoma, Cancers, 2020, 12(3), 684 CrossRef CAS PubMed.
  47. E. Batlle and H. Clevers, Cancer stem cells revisited, Nat. Med., 2017, 23(10), 1124–1134 CrossRef CAS PubMed.
  48. Z. Yu, T. G. Pestell, M. P. Lisanti and R. G. Pestell, Cancer stem cells, Int. J. Biochem. Cell Biol., 2012, 44(12), 2144–2151 CrossRef CAS PubMed.
  49. B. Izar, I. Tirosh, E. H. Stover, I. Wakiro, M. S. Cuoco, I. Alter, C. Rodman, R. Leeson, M. J. Su, P. Shah, M. Iwanicki, S. R. Walker, A. Kanodia, J. C. Melms, S. Mei, J. R. Lin, C. B. M. Porter, M. Slyper, J. Waldman, L. Jerby-Arnon, O. Ashenberg, T. J. Brinker, C. Mills, M. Rogava, S. Vigneau, P. K. Sorger, L. A. Garraway, P. A. Konstantinopoulos, J. F. Liu, U. Matulonis, B. E. Johnson, O. Rozenblatt-Rosen, A. Rotem and A. Regev, A single-cell landscape of high-grade serous ovarian cancer, Nat. Med., 2020, 26(8), 1271–1279 CrossRef CAS PubMed.
  50. Y. Deng, Y. Tan, D. Zhou, Y. Bai, T. Cao, C. Zhong, W. Huang, Y. Ou, L. Guo, Q. Liu, D. Yin, L. Chen, X. Luo, D. Sun and X. Sheng, Single-Cell RNA-Sequencing Atlas Reveals the Tumor Microenvironment of Metastatic High-Grade Serous Ovarian Carcinoma, Front. Immunol., 2022, 13, 923194 CrossRef CAS PubMed.
  51. Y. Su, M. E. Ko, H. Cheng, R. Zhu, M. Xue, J. Wang, J. W. Lee, L. Frankiw, A. Xu, S. Wong, L. Robert, K. Takata, D. Yuan, Y. Lu, S. Huang, A. Ribas, R. Levine, G. P. Nolan, W. Wei, S. K. Plevritis, G. Li, D. Baltimore and J. R. Heath, Multi-omic single-cell snapshots reveal multiple independent trajectories to drug tolerance in a melanoma cell line, Nat. Commun., 2020, 11(1), 2345 CrossRef CAS PubMed.
  52. M. Meyer, A. Paquet, M. J. Arguel, L. Peyre, L. C. Gomes-Pereira, K. Lebrigand, B. Mograbi, P. Brest, R. Waldmann, P. Barbry, P. Hofman and J. Roux, Profiling the Non-genetic Origins of Cancer Drug Resistance with a Single-Cell Functional Genomics Approach Using Predictive Cell Dynamics, Cell Syst., 2020, 11(4), 367–374 e5 CrossRef CAS PubMed.
  53. R. Salgia and P. Kulkarni, The Genetic/Non-genetic Duality of Drug 'Resistance' in Cancer, Trends Cancer, 2018, 4(2), 110–118 CrossRef CAS PubMed.
  54. C. E. Eyler, H. Matsunaga, V. Hovestadt, S. J. Vantine, P. van Galen and B. E. Bernstein, Single-cell lineage analysis reveals genetic and epigenetic interplay in glioblastoma drug resistance, Genome Biol., 2020, 21(1), 174 CrossRef CAS PubMed.
  55. E. Papalexi and R. Satija, Single-cell RNA sequencing to explore immune cell heterogeneity, Nat. Rev. Immunol., 2018, 18(1), 35–45 CrossRef CAS PubMed.
  56. B. S. Boland, Z. He, M. S. Tsai, J. G. Olvera, K. D. Omilusik, H. G. Duong, E. S. Kim, A. E. Limary, W. Jin, J. J. Milner, B. Yu, S. A. Patel, T. L. Louis, T. Tysl, N. S. Kurd, A. Bortnick, L. K. Quezada, J. N. Kanbar, A. Miralles, D. Huylebroeck, M. A. Valasek, P. S. Dulai, S. Singh, L. F. Lu, J. D. Bui, C. Murre, W. J. Sandborn, A. W. Goldrath, G. W. Yeo and J. T. Chang, Heterogeneity and clonal relationships of adaptive immune cells in ulcerative colitis revealed by single-cell analyses, Sci. Immunol., 2020, 5(50), eabb4432 CrossRef CAS.
  57. R. Satija and A. K. Shalek, Heterogeneity in immune responses: from populations to single cells, Trends Immunol., 2014, 35(5), 219–229 CrossRef CAS PubMed.
  58. C. Sievers, M. Craveiro, J. Friedman, Y. Robbins, X. Yang, K. Bai, A. Nguyen, J. M. Redman, R. Chari, P. Soon-Shiong, J. Schlom, J. Gulley and C. T. Allen, Phenotypic plasticity and reduced tissue retention of exhausted tumor-infiltrating T cells following neoadjuvant immunotherapy in head and neck cancer, Cancer Cell, 2023, 41(5), 887–902 e5 CrossRef CAS.
  59. C. A. Egelston, W. Guo, J. Tan, C. Avalos, D. L. Simons, M. H. Lim, Y. J. Huang, M. S. Nelson, A. Chowdhury, D. B. Schmolze, J. H. Yim, L. Kruper, L. Melstrom, K. Margolin, J. E. Mortimer, Y. Yuan, J. R. Waisman and P. P. Lee, Tumor-infiltrating exhausted CD8+ T cells dictate reduced survival in premenopausal estrogen receptor-positive breast cancer, JCI Insight, 2022, 7(3), e153963 CrossRef PubMed.
  60. M. Ahmadzadeh, L. A. Johnson, B. Heemskerk, J. R. Wunderlich, M. E. Dudley, D. E. White and S. A. Rosenberg, Tumor antigen-specific CD8 T cells infiltrating the tumor express high levels of PD-1 and are functionally impaired, Blood, 2009, 114(8), 1537–1544 CrossRef CAS PubMed.
  61. A. J. Wilk, A. Rustagi, N. Q. Zhao, J. Roque, G. J. Martinez-Colon, J. L. McKechnie, G. T. Ivison, T. Ranganath, R. Vergara, T. Hollis, L. J. Simpson, P. Grant, A. Subramanian, A. J. Rogers and C. A. Blish, A single-cell atlas of the peripheral immune response in patients with severe COVID-19, Nat. Med., 2020, 26(7), 1070–1076 CrossRef CAS PubMed.
  62. W. Wen, W. Su, H. Tang, W. Le, X. Zhang, Y. Zheng, X. Liu, L. Xie, J. Li, J. Ye, L. Dong, X. Cui, Y. Miao, D. Wang, J. Dong, C. Xiao, W. Chen and H. Wang, Immune cell profiling of COVID-19 patients in the recovery stage by single-cell sequencing, Cell Discovery, 2020, 6, 31 CrossRef CAS PubMed.
  63. H. Li, S. Bao, N. Farzad, X. Qin, A. A. Fung, D. Zhang, Z. Bai, B. Tao and R. Fan, Spatially resolved genome-wide joint profiling of epigenome and transcriptome with spatial-ATAC-RNA-seq and spatial-CUT&Tag-RNA-seq, Nat. Protoc., 2025, 20(9), 2383–2417 CrossRef CAS PubMed.
  64. Y. Deng, M. Bartosovic, S. Ma, D. Zhang, P. Kukanja, Y. Xiao, G. Su, Y. Liu, X. Qin, G. B. Rosoklija, A. J. Dwork, J. J. Mann, M. L. Xu, S. Halene, J. E. Craft, K. W. Leong, M. Boldrini, G. Castelo-Branco and R. Fan, Spatial profiling of chromatin accessibility in mouse and human tissues, Nature, 2022, 609(7926), 375–383 CrossRef CAS PubMed.
  65. Y. Liu, M. Yang, Y. Deng, G. Su, A. Enninful, C. C. Guo, T. Tebaldi, D. Zhang, D. Kim, Z. Bai, E. Norris, A. Pan, J. Li, Y. Xiao, S. Halene and R. Fan, High-Spatial-Resolution Multi-Omics Sequencing via Deterministic Barcoding in Tissue, Cell, 2020, 183(6), 1665–1681 e18 CrossRef CAS PubMed.
  66. H. Yin and D. Marshall, Microfluidics for single cell analysis, Curr. Opin. Biotechnol., 2012, 23(1), 110–119 CrossRef CAS PubMed.
  67. L. Mazutis, J. Gilbert, W. L. Ung, D. A. Weitz, A. D. Griffiths and J. A. Heyman, Single-cell analysis and sorting using droplet-based microfluidics, Nat. Protoc., 2013, 8(5), 870–891 CrossRef CAS PubMed.
  68. H. C. Chen, Y. Ma, J. Cheng and Y. C. Chen, Advances in Single-Cell Techniques for Linking Phenotypes to Genotypes, Cancer Heterog. Plast., 2024, 1(1), 0004 Search PubMed.
  69. G. M. Whitesides, The origins and the future of microfluidics, Nature, 2006, 442(7101), 368–373 CrossRef CAS PubMed.
  70. D. J. Beebe, G. A. Mensing and G. M. Walker, Physics and applications of microfluidics in biology, Annu. Rev. Biomed. Eng., 2002, 4, 261–286 CrossRef CAS PubMed.
  71. W. H. Tan and S. Takeuchi, A trap-and-release integrated microfluidic system for dynamic microarray applications, Proc. Natl. Acad. Sci. U. S. A., 2007, 104(4), 1146–1151 CrossRef CAS PubMed.
  72. A. M. Skelley, O. Kirak, H. Suh, R. Jaenisch and J. Voldman, Microfluidic control of cell pairing and fusion, Nat. Methods, 2009, 6(2), 147–152 CrossRef CAS PubMed.
  73. Y. H. Cheng, Y. C. Chen, R. Brien and E. Yoon, Scaling and automation of a high-throughput single-cell-derived tumor sphere assay chip, Lab Chip, 2016, 16(19), 3708–3717 RSC.
  74. J. S. Marcus, W. F. Anderson and S. R. Quake, Microfluidic single-cell mRNA isolation and analysis, Anal. Chem., 2006, 78, 3084–3089 CrossRef CAS PubMed.
  75. T. M. Gierahn, M. H. Wadsworth, T. K. Hughes, B. D. Bryson, A. Butler, R. Satija, S. Fortune, J. C. Love and A. K. Shalek, Seq-Well: portable, low-cost RNA sequencing of single cells at high throughput, Nat. Methods, 2017, 14(4), 395–398 CrossRef CAS PubMed.
  76. Y. H. Cheng, Y. C. Chen, E. Lin, R. Brien, S. Jung, Y. T. Chen, W. Lee, Z. Hao, S. Sahoo, H. Min Kang, J. Cong, M. Burness, S. Nagrath, M. S. Wicha and E. Yoon, Hydro-Seq enables contamination-free high-throughput single-cell RNA-sequencing for circulating tumor cells, Nat. Commun., 2019, 10(1), 2163 CrossRef PubMed.
  77. B. Dura, S. K. Dougan, M. Barisa, M. M. Hoehl, C. T. Lo, H. L. Ploegh and J. Voldman, Profiling lymphocyte interactions at the single-cell level by microfluidic cell pairing, Nat. Commun., 2015, 6, 5940 CrossRef CAS.
  78. M. Rothbauer, H. Zirath and P. Ertl, Recent advances in microfluidic technologies for cell-to-cell interaction studies, Lab Chip, 2018, 18(2), 249–270 RSC.
  79. Y. C. Chen, M. E. Gonzalez, B. Burman, X. Zhao, T. Anwar, M. Tran, N. Medhora, A. B. Hiziroglu, W. Lee, Y. H. Cheng, Y. Choi, E. Yoon and C. G. Kleer, Mesenchymal Stem/Stromal Cell Engulfment Reveals Metastatic Advantage in Breast Cancer, Cell Rep., 2019, 27(13), 3916–3926 e5 CrossRef CAS PubMed.
  80. H. Zahn, A. Steif, E. Laks, P. Eirew, M. VanInsberghe, S. P. Shah, S. Aparicio and C. L. Hansen, Scalable whole-genome single-cell library preparation without preamplification, Nat. Methods, 2017, 14(2), 167–173 CrossRef CAS PubMed.
  81. E. Laks, A. McPherson, H. Zahn, D. Lai, A. Steif, J. Brimhall, J. Biele, B. Wang, T. Masud, J. Ting, D. Grewal, C. Nielsen, S. Leung, V. Bojilova, M. Smith, O. Golovko, S. Poon, P. Eirew, F. Kabeer, T. Ruiz de Algara, S. R. Lee, M. J. Taghiyar, C. Huebner, J. Ngo, T. Chan, S. Vatrt-Watts, P. Walters, N. Abrar, S. Chan, M. Wiens, L. Martin, R. W. Scott, T. M. Underhill, E. Chavez, C. Steidl, D. Da Costa, Y. Ma, R. J. N. Coope, R. Corbett, S. Pleasance, R. Moore, A. J. Mungall, C. Mar, F. Cafferty, K. Gelmon, S. Chia, C. I. G. C. Team, M. A. Marra, C. Hansen, S. P. Shah and S. Aparicio, Clonal Decomposition and DNA Replication States Defined by Scaled Single-Cell Genome Sequencing, Cell, 2019, 179(5), 1207–1221 e22 CrossRef CAS PubMed.
  82. W. Zheng, S. Zhao, Y. Yin, H. Zhang, D. M. Needham, E. D. Evans, C. L. Dai, P. J. Lu, E. J. Alm and D. A. Weitz, High-throughput, single-microbe genomics with strain resolution, applied to a human gut microbiome, Science, 2022, 376(6597), eabm1483 CrossRef CAS PubMed.
  83. Z. Bai, Y. Deng, D. Kim, Z. Chen, Y. Xiao and R. Fan, An Integrated Dielectrophoresis-Trapping and Nanowell Transfer Approach to Enable Double-Sub-Poisson Single-Cell RNA Sequencing, ACS Nano, 2020, 14(6), 7412–7424 CrossRef CAS PubMed.
  84. Y. Zhu, P. D. Piehowski, R. Zhao, J. Chen, Y. Shen, R. J. Moore, A. K. Shukla, V. A. Petyuk, M. Campbell-Thompson, C. E. Mathews, R. D. Smith, W. J. Qian and R. T. Kelly, Nanodroplet processing platform for deep and quantitative proteome profiling of 10-100 mammalian cells, Nat. Commun., 2018, 9(1), 882 CrossRef PubMed.
  85. S. T. Gebreyesus, A. A. Siyal, R. B. Kitata, E. S. Chen, B. Enkhbayar, T. Angata, K. I. Lin, Y. J. Chen and H. L. Tu, Streamlined single-cell proteomics by an integrated microfluidic chip and data-independent acquisition mass spectrometry, Nat. Commun., 2022, 13(1), 37 CrossRef CAS PubMed.
  86. Z. Yang, K. Jin, Y. Chen, Q. Liu, H. Chen, S. Hu, Y. Wang, Z. Pan, F. Feng, M. Shi, H. Xie, H. Ma and H. Zhou, AM-DMF-SCP: Integrated Single-Cell Proteomics Analysis on an Active Matrix Digital Microfluidic Chip, JACS Au, 2024, 4(5), 1811–1823 CrossRef CAS PubMed.
  87. S. Xu, M. Liu, Y. Bai and H. Liu, Multi-Dimensional Organic Mass Cytometry: Simultaneous Analysis of Proteins and Metabolites on Single Cells, Angew. Chem., 2021, 60(4), 1806–1812 CrossRef CAS PubMed.
  88. D. Feng, H. Li, T. Xu, F. Zheng, C. Hu, X. Shi and G. Xu, High-throughput single cell metabolomics and cellular heterogeneity exploration by inertial microfluidics coupled with pulsed electric field-induced electrospray ionization-high resolution mass spectrometry, Anal. Chim. Acta, 2022, 1221, 340116 CrossRef CAS PubMed.
  89. A. J. Ibanez, S. R. Fagerer, A. M. Schmidt, P. L. Urban, K. Jefimovs, P. Geiger, R. Dechant, M. Heinemann and R. Zenobi, Mass spectrometry-based metabolomics of single yeast cells, Proc. Natl. Acad. Sci. U. S. A., 2013, 110(22), 8790–8794 CrossRef CAS PubMed.
  90. J. Feng, X. Zhang, L. Huang, H. Yao, C. Yang, X. Ma, S. Zhang and X. Zhang, Quantitation of Glucose-phosphate in Single Cells by Microwell-Based Nanoliter Droplet Microextraction and Mass Spectrometry, Anal. Chem., 2019, 91(9), 5613–5620 CrossRef CAS PubMed.
  91. H. Zhu, Q. Li, T. Liao, X. Yin, Q. Chen, Z. Wang, M. Dai, L. Yi, S. Ge, C. Miao, W. Zeng, L. Qu, Z. Ju, G. Huang, C. Cang and W. Xiong, Metabolomic profiling of single enlarged lysosomes, Nat. Methods, 2021, 18(7), 788–798 CrossRef CAS PubMed.
  92. J. Zhu, S. Pan, H. Chai, P. Zhao, Y. Feng, Z. Cheng, S. Zhang and W. Wang, Microfluidic Impedance Cytometry Enabled One-Step Sample Preparation for Efficient Single-Cell Mass Spectrometry, Small, 2024, 20(26), e2310700 CrossRef PubMed.
  93. W. Xie, D. Gao, F. Jin, Y. Jiang and H. Liu, Study of Phospholipids in Single Cells Using an Integrated Microfluidic Device Combined with Matrix-Assisted Laser Desorption/Ionization Mass Spectrometry, Anal. Chem., 2015, 87(14), 7052–7059 CrossRef CAS PubMed.
  94. Z. Li, S. Cheng, Q. Lin, W. Cao, J. Yang, M. Zhang, A. Shen, W. Zhang, Y. Xia, X. Ma and Z. Ouyang, Single-cell lipidomics with high structural specificity by mass spectrometry, Nat. Commun., 2021, 12(1), 2869 CrossRef CAS PubMed.
  95. J. Lamanna, E. Y. Scott, H. S. Edwards, M. D. Chamberlain, M. D. M. Dryden, J. Peng, B. Mair, A. Lee, C. Chan, A. A. Sklavounos, A. Heffernan, F. Abbas, C. Lam, M. E. Olson, J. Moffat and A. R. Wheeler, Digital microfluidic isolation of single cells for -Omics, Nat. Commun., 2020, 11(1), 5632 CrossRef CAS PubMed.
  96. E. Y. Scott, N. Safarian, D. L. Casasbuenas, M. Dryden, T. Tockovska, S. Ali, J. Peng, E. Daniele, I. N. X. Lim, K. W. A. Bang, S. Tripathy, S. A. Yuzwa, A. R. Wheeler and M. Faiz, Integrating single-cell and spatially resolved transcriptomic strategies to survey the astrocyte response to stroke in male mice, Nat. Commun., 2024, 15(1), 1584 CrossRef CAS PubMed.
  97. S. T. Gebreyesus, G. Muneer, C. C. Huang, A. A. Siyal, M. Anand, Y. J. Chen and H. L. Tu, Recent advances in microfluidics for single-cell functional proteomics, Lab Chip, 2023, 23(7), 1726–1751 RSC.
  98. D. Zhang and L. Qiao, Microfluidics Coupled Mass Spectrometry for Single Cell Multi-Omics, Small Methods, 2024, 8(1), e2301179 CrossRef PubMed.
  99. J. M. Ng, I. Gitlin, A. D. Stroock and G. M. Whitesides, Components for integrated poly(dimethylsiloxane) microfluidic systems, Electrophoresis, 2002, 23(20), 3461–3473 CrossRef CAS PubMed.
  100. A. Folch, A. Ayon, O. Hurtado, M. A. Schmidt and M. Toner, Molding of deep polydimethylsiloxane microstructures for microfluidics and biological applications, J. Biomech. Eng., 1999, 121(1), 28–34 CrossRef CAS PubMed.
  101. W. Zheng, Z. Wang, W. Zhang and X. Jiang, A simple PDMS-based microfluidic channel design that removes bubbles for long-term on-chip culture of mammalian cells, Lab Chip, 2010, 10(21), 2906–2910 RSC.
  102. J. Wang, W. Chen, J. Sun, C. Liu, Q. Yin, L. Zhang, Y. Xianyu, X. Shi, G. Hu and X. Jiang, A microfluidic tubing method and its application for controlled synthesis of polymeric nanoparticles, Lab Chip, 2014, 14(10), 1673–1677 RSC.
  103. Y. Zhu, Y. X. Zhang, W. W. Liu, Y. Ma, Q. Fang and B. Yao, Printing 2-dimentional droplet array for single-cell reverse transcription quantitative PCR assay with a microfluidic robot, Sci. Rep., 2015, 5, 9551 CrossRef PubMed.
  104. Y. Zhu, G. Clair, W. B. Chrisler, Y. Shen, R. Zhao, A. K. Shukla, R. J. Moore, R. S. Misra, G. S. Pryhuber, R. D. Smith, C. Ansong and R. T. Kelly, Proteomic Analysis of Single Mammalian Cells Enabled by Microfluidic Nanodroplet Sample Preparation and Ultrasensitive NanoLC-MS, Angew. Chem., Int. Ed., 2018, 57(38), 12370–12374 CrossRef CAS PubMed.
  105. Y. Zhu, Y. X. Zhang, L. F. Cai and Q. Fang, Sequential operation droplet array: an automated microfluidic platform for picoliter-scale liquid handling, analysis, and screening, Anal. Chem., 2013, 85(14), 6723–6731 CrossRef CAS PubMed.
  106. Y. Zhu, L. N. Zhu, R. Guo, H. J. Cui, S. Ye and Q. Fang, Nanoliter-scale protein crystallization and screening with a microfluidic droplet robot, Sci. Rep., 2014, 4, 5046 CrossRef CAS PubMed.
  107. J. Durrer, P. Agrawal, A. Ozgul, S. C. F. Neuhauss, N. Nama and D. Ahmed, A robot-assisted acoustofluidic end effector, Nat. Commun., 2022, 13(1), 6370 CrossRef CAS PubMed.
  108. Y. Wang, Z. Y. Guan, S. W. Shi, Y. R. Jiang, J. Zhang, Y. Yang, Q. Wu, J. Wu, J. B. Chen, W. X. Ying, Q. Q. Xu, Q. X. Fan, H. F. Wang, L. Zhou, L. Wang, J. Fang, J. Z. Pan and Q. Fang, Pick-up single-cell proteomic analysis for quantifying up to 3000 proteins in a Mammalian cell, Nat. Commun., 2024, 15(1), 1279 CrossRef CAS PubMed.
  109. Y. Wang, D. F. Wang, H. F. Wang, J. W. Wang, J. Z. Pan, X. G. Guo and Q. Fang, A microfluidic robot for rare cell sorting based on machine vision identification and multi-step sorting strategy, Talanta, 2021, 226, 122136 CrossRef CAS PubMed.
  110. I. Stepanov, N. R. Gottshall, A. Ahmadianyazdi, D. Sinha, E. J. Lockhart, T. N. H. Nguyen, S. Hassan, L. F. Horowitz, R. S. Yeung, T. S. Gujral and A. Folch, Low-cost robotic manipulation of live microtissues for cancer drug testing, Sci. Adv., 2025, 11(20), eads1631 CrossRef CAS PubMed.
  111. K. Angers, K. Darvish, N. Yoshikawa, S. Okhovatian, D. Bannerman, I. Yakavets, F. Shkurti, A. Aspuru-Guzik and M. Radisic, RoboCulture: A Robotics Platform for Automated Biological Experimentation, arXiv, 2025, preprint, arXiv:2505.14941,  DOI:10.48550/arXiv.2505.14941.
  112. Y. C. Tung, A. Y. Hsiao, S. G. Allen, Y. S. Torisawa, M. Ho and S. Takayama, High-throughput 3D spheroid culture and drug testing using a 384 hanging drop array, Analyst, 2011, 136(3), 473–478 RSC.
  113. R. Novak, M. Ingram, S. Marquez, D. Das, A. Delahanty, A. Herland, B. M. Maoz, S. S. F. Jeanty, M. R. Somayaji, M. Burt, E. Calamari, A. Chalkiadaki, A. Cho, Y. Choe, D. B. Chou, M. Cronce, S. Dauth, T. Divic, J. Fernandez-Alcon, T. Ferrante, J. Ferrier, E. A. FitzGerald, R. Fleming, S. Jalili-Firoozinezhad, T. Grevesse, J. A. Goss, T. Hamkins-Indik, O. Henry, C. Hinojosa, T. Huffstater, K. J. Jang, V. Kujala, L. Leng, R. Mannix, Y. Milton, J. Nawroth, B. A. Nestor, C. F. Ng, B. O'Connor, T. E. Park, H. Sanchez, J. Sliz, A. Sontheimer-Phelps, B. Swenor, G. Thompson, G. J. Touloumes, Z. Tranchemontagne, N. Wen, M. Yadid, A. Bahinski, G. A. Hamilton, D. Levner, O. Levy, A. Przekwas, R. Prantil-Baun, K. K. Parker and D. E. Ingber, Robotic fluidic coupling and interrogation of multiple vascularized organ chips, Nat. Biomed. Eng., 2020, 4(4), 407–420 CrossRef PubMed.
  114. S. Jiang, H. Zhao, W. Zhang, J. Wang, Y. Liu, Y. Cao, H. Zheng, Z. Hu, S. Wang, Y. Zhu, W. Wang, S. Cui, P. E. Lobie, L. Huang and S. Ma, An Automated Organoid Platform with Inter-organoid Homogeneity and Inter-patient Heterogeneity, Cell Rep. Med., 2020, 1(9), 100161 CrossRef CAS PubMed.
  115. T. M. Tran, S. C. Kim, C. Modavi and A. R. Abate, Robotic automation of droplet microfluidics, Biomicrofluidics, 2022, 16(1), 014102 CrossRef CAS.
  116. C. Ctortecka, D. Hartlmayr, A. Seth, S. Mendjan, G. Tourniaire and K. Mechtler, An automated workflow for multiplexed single-cell proteomics sample preparation at unprecedented sensitivity, Mol. Cell. Proteomics, 2023, 22(12), 100665 CrossRef CAS PubMed.
  117. M. Zhou, Y. Ma, C. C. Chiang, E. C. Rock, K. E. Luker, G. D. Luker and Y. C. Chen, High-Throughput Cellular Heterogeneity Analysis in Cell Migration at the Single-Cell Level, Small, 2023, 19(6), e2206754 CrossRef PubMed.
  118. M. Zhou, Y. Ma, E. C. Rock, C. C. Chiang, K. E. Luker, G. D. Luker and Y. C. Chen, Microfluidic single-cell migration chip reveals insights into the impact of extracellular matrices on cell movement, Lab Chip, 2023, 23(21), 4619–4635 RSC.
  119. J. Cheng, E. C. Rock, M. Rao, H. C. Chen, Y. Ma, K. C. Chang and Y. C. Chen, 3D-printed plugs enhance cell usage efficiency for single-cell migration and neuron axon guidance assays, Cell Rep. Med., 2025, 101117 CAS.
  120. W. Yu, H. Lin, Y. Wang, X. He, N. Chen, K. Sun, D. Lo, B. Cheng, C. Yeung, J. Tan, D. Di Carlo and S. Emaminejad, A ferrobotic system for automated microfluidic logistics, Sci. Robot., 2020, 5(39), eaba4411 CrossRef PubMed.
  121. H. Lin, W. Yu, K. A. Sabet, M. Bogumil, Y. Zhao, J. Hambalek, S. Lin, S. Chandrasekaran, O. Garner, D. Di Carlo and S. Emaminejad, Ferrobotic swarms enable accessible and adaptable automated viral testing, Nature, 2022, 611(7936), 570–577 CrossRef CAS PubMed.
  122. P. Y. Chiou, A. T. Ohta and M. C. Wu, Massively parallel manipulation of single cells and microparticles using optical images, Nature, 2005, 436(7049), 370–372 CrossRef CAS PubMed.
  123. S. Zhang, N. Shakiba, Y. Chen, Y. Zhang, P. Tian, J. Singh, M. D. Chamberlain, M. Satkauskas, A. G. Flood, N. P. Kherani, S. Yu, P. W. Zandstra and A. R. Wheeler, Patterned Optoelectronic Tweezers: A New Scheme for Selecting, Moving, and Storing Dielectric Particles and Cells, Small, 2018, 14(45), e1803342 CrossRef PubMed.
  124. S. Zhang, E. Y. Scott, J. Singh, Y. Chen, Y. Zhang, M. Elsayed, M. D. Chamberlain, N. Shakiba, K. Adams, S. Yu, C. M. Morshead, P. W. Zandstra and A. R. Wheeler, The optoelectronic microrobot: A versatile toolbox for micromanipulation, Proc. Natl. Acad. Sci. U. S. A., 2019, 116(30), 14823–14828 CrossRef CAS PubMed.
  125. G. T. Ivanyi, B. Nemes, I. Grof, T. Fekete, J. Kubackova, Z. Tomori, G. Bano, G. Vizsnyiczai and L. Kelemen, Optically Actuated Soft Microrobot Family for Single-Cell Manipulation, Adv. Mater., 2024, 36(32), e2401115 CrossRef PubMed.
  126. J. Jin, T. Ogawa, N. Hojo, K. Kryukov, K. Shimizu, T. Ikawa, T. Imanishi, T. Okazaki and K. Shiroguchi, Robotic data acquisition with deep learning enables cell image-based prediction of transcriptomic phenotypes, Proc. Natl. Acad. Sci. U. S. A., 2023, 120(1), e2210283120 CrossRef CAS PubMed.
  127. K. Guo, Z. Song, J. Zhou, B. Shen, B. Yan, Z. Gu and H. Wang, An artificial intelligence-assisted digital microfluidic system for multistate droplet control, Microsyst. Nanoeng., 2024, 10(1), 138 CrossRef PubMed.
  128. Z. Guo, F. Li, H. Li, M. Zhao, H. Liu, H. Wang, H. Hu, R. Fu, Y. Lu, S. Hu, H. Xie, H. Ma and S. Zhang, Deep Learning-Assisted Label-Free Parallel Cell Sorting with Digital Microfluidics, Adv. Sci., 2025, 12(1), e2408353 CrossRef PubMed.
  129. P. Zhang, Z. Tian, K. Jin, K. Yang, W. Collyer, J. Rufo, N. Upreti, X. Dong, L. P. Lee and T. J. Huang, Automating life science labs at the single-cell level through precise ultrasonic liquid sample ejection: PULSE, Microsyst. Nanoeng., 2024, 10(1), 172 CrossRef CAS PubMed.
  130. Y. C. Chu and J. Huang, A neural-network method for the nonlinear servomechanism problem, IEEE Trans. Neural Netw., 1999, 10(6), 1412–1423 CrossRef CAS PubMed.
  131. I. Goodfellow, J. Pouget-Abadie, M. Mirza, B. Xu, D. Warde-Farley, S. Ozair, A. Courville and Y. Bengio, Generative Adversarial Networks, Commun. ACM, 2020, 63(11), 139–144 CrossRef.
  132. D. P. Kingma and M. Welling, An Introduction to Variational Autoencoders, Found. Trends Mach. Learn., 2019, 12(4), 307–392 CrossRef.
  133. C. Stringer, T. Wang, M. Michaelos and M. Pachitariu, Cellpose: a generalist algorithm for cellular segmentation, Nat. Methods, 2021, 18(1), 100–106 CrossRef CAS PubMed.
  134. M. Pachitariu and C. Stringer, Cellpose 2.0: how to train your own model, Nat. Methods, 2022, 19(12), 1634–1641 CrossRef CAS PubMed.
  135. C. Edlund, T. R. Jackson, N. Khalid, N. Bevan, T. Dale, A. Dengel, S. Ahmed, J. Trygg and R. Sjogren, LIVECell-A large-scale dataset for label-free live cell segmentation, Nat. Methods, 2021, 18(9), 1038–1045 CrossRef CAS PubMed.
  136. N. U. Din and J. Yu, Training a deep learning model for single-cell segmentation without manual annotation, Sci. Rep., 2021, 11(1), 23995 CrossRef CAS PubMed.
  137. D. Jimenez-Carretero, V. Abrishami, L. Fernandez-de-Manuel, I. Palacios, A. Quilez-Alvarez, A. Diez-Sanchez, M. A. Del Pozo and M. C. Montoya, Tox_(R)CNN: Deep learning-based nuclei profiling tool for drug toxicity screening, PLoS Comput. Biol., 2018, 14(11), e1006238 CrossRef PubMed.
  138. Y. C. Chen, Z. Zhang and E. Yoon, Early Prediction of Single-Cell Derived Sphere Formation Rate Using Convolutional Neural Network Image Analysis, Anal. Chem., 2020, 92(11), 7717–7724 CrossRef CAS PubMed.
  139. G. Pattarone, L. Acion, M. Simian, R. Mertelsmann, M. Follo and E. Iarussi, Learning deep features for dead and living breast cancer cell classification without staining, Sci. Rep., 2021, 11(1), 10304 CrossRef CAS PubMed.
  140. K. Ulicna, G. Vallardi, G. Charras and A. R. Lowe, Automated Deep Lineage Tree Analysis Using a Bayesian Single Cell Tracking Approach, Front. Comput. Sci., 2021, 3, 734559 CrossRef.
  141. E. B. Hartnett, M. Zhou, Y. N. Gong and Y. C. Chen, LANCE: a Label-Free Live Apoptotic and Necrotic Cell Explorer Using Convolutional Neural Network Image Analysis, Anal. Chem., 2022, 94(43), 14827–14834 CrossRef CAS PubMed.
  142. S. He, M. Sillah, A. R. Cole, A. Uboveja, K. M. Aird, Y. C. Chen and Y. N. Gong, D-MAINS: A Deep-Learning Model for the Label-Free Detection of Mitosis, Apoptosis, Interphase, Necrosis, and Senescence in Cancer Cells, Cells, 2024, 13(12), 1004 CrossRef CAS PubMed.
  143. Q. Li, J. Zhou, S. Jiang, Y. Fu and M. Su, Single-Cell Array Enhanced Cell Damage Recognition Using Artificial Intelligence for Anticancer Drug Discovery, Anal. Chem., 2025, 97(7), 4202–4208 CrossRef CAS PubMed.
  144. M. Toseef, O. Olayemi Petinrin, F. Wang, S. Rahaman, Z. Liu, X. Li and K. C. Wong, Deep transfer learning for clinical decision-making based on high-throughput data: comprehensive survey with benchmark results, Briefings Bioinf., 2023, 24(4), bbad254 CrossRef PubMed.
  145. A. Kowald, I. Barrantes, S. Moller, D. Palmer, H. Murua Escobar, A. Schwerk and G. Fuellen, Transfer learning of clinical outcomes from preclinical molecular data, principles and perspectives, Briefings Bioinf., 2022, 23(3), bbac133 CrossRef PubMed.
  146. R. Krishnan, P. Rajpurkar and E. J. Topol, Self-supervised learning in medicine and healthcare, Nat. Biomed. Eng., 2022, 6(12), 1346–1352 CrossRef PubMed.
  147. H. Wang, T. Fu, Y. Du, W. Gao, K. Huang, Z. Liu, P. Chandak, S. Liu, P. Van Katwyk, A. Deac, A. Anandkumar, K. Bergen, C. P. Gomes, S. Ho, P. Kohli, J. Lasenby, J. Leskovec, T. Y. Liu, A. Manrai, D. Marks, B. Ramsundar, L. Song, J. Sun, J. Tang, P. Velickovic, M. Welling, L. Zhang, C. W. Coley, Y. Bengio and M. Zitnik, Scientific discovery in the age of artificial intelligence, Nature, 2023, 620(7972), 47–60 CrossRef CAS PubMed.
  148. Z. Zhang, L. Chen, B. Humphries, R. Brien, M. S. Wicha, K. E. Luker, G. D. Luker, Y. C. Chen and E. Yoon, Morphology-based prediction of cancer cell migration using an artificial neural network and a random decision forest, Integr. Biol., 2018, 10(12), 758–767 CrossRef CAS PubMed.
  149. A. J. Green, M. J. Mohlenkamp, J. Das, M. Chaudhari, L. Truong, R. L. Tanguay and D. M. Reif, Leveraging high-throughput screening data, deep neural networks, and conditional generative adversarial networks to advance predictive toxicology, PLoS Comput. Biol., 2021, 17(7), e1009135 CrossRef CAS PubMed.
  150. T. H. Pham, Y. Qiu, J. Liu, S. Zimmer, E. O'Neill, L. Xie and P. Zhang, Chemical-induced gene expression ranking and its application to pancreatic cancer drug repurposing, Patterns, 2022, 3(4), 100441 CrossRef CAS PubMed.
  151. T. H. Pham, Y. Qiu, J. Zeng, L. Xie and P. Zhang, A deep learning framework for high-throughput mechanism-driven phenotype compound screening and its application to COVID-19 drug repurposing, Nat. Mach. Intell., 2021, 3(3), 247–257 CrossRef PubMed.
  152. J. Lakkis, A. Schroeder, K. Su, M. Y. Y. Lee, A. C. Bashore, M. P. Reilly and M. Li, A multi-use deep learning method for CITE-seq and single-cell RNA-seq data integration with cell surface protein prediction and imputation, Nat. Mach. Intell., 2022, 4(11), 940–952 CrossRef PubMed.
  153. C. C. Chiang, R. Anne, P. Chawla, R. M. Shaw, S. He, E. C. Rock, M. Zhou, J. Cheng, Y. N. Gong and Y. C. Chen, Deep learning unlocks label-free viability assessment of cancer spheroids in microfluidics, Lab Chip, 2024, 24(12), 3169–3182 RSC.
  154. Z. Zhang, L. Chen, Y. Wang, T. Zhang, Y. C. Chen and E. Yoon, Label-Free Estimation of Therapeutic Efficacy on 3D Cancer Spheres Using Convolutional Neural Network Image Analysis, Anal. Chem., 2019, 91(21), 14093–14100 CrossRef CAS PubMed.
  155. Y. Ma, C. H. Shih, J. Cheng, H. C. Chen, L. J. Wang, Y. Tan, Y. Zhang, D. D. Brown, S. Oesterreich, A. V. Lee, Y. C. Chiu and Y. C. Chen, High-Throughput Empirical and Virtual Screening To Discover Novel Inhibitors of Polyploid Giant Cancer Cells in Breast Cancer, Anal. Chem., 2025, 97(10), 5498–5506 CrossRef CAS PubMed.
  156. A. Witmer and B. Bhanu, Generative Adversarial Networks for Morphological-Temporal Classification of Stem Cell Images, Sensors, 2021, 22(1), 206 CrossRef PubMed.
  157. Z. Wu, J. Park, P. R. Steiner, B. Zhu and J. X. J. Zhang, Generative adversarial network model to classify human induced pluripotent stem cell-cardiomyocytes based on maturation level, Sci. Rep., 2024, 14(1), 27016 CrossRef CAS PubMed.
  158. A. Palma, F. J. Theis and M. Lotfollahi, Predicting cell morphological responses to perturbations using generative modeling, Nat. Commun., 2025, 16(1), 505 CrossRef CAS PubMed.
  159. L. Ternes, M. Dane, S. Gross, M. Labrie, G. Mills, J. Gray, L. Heiser and Y. H. Chang, A multi-encoder variational autoencoder controls multiple transformational features in single-cell image analysis, Commun. Biol., 2022, 5(1), 255 CrossRef PubMed.
  160. Y. Xu, Z. Zhang, L. You, J. Liu, Z. Fan and X. Zhou, scIGANs: single-cell RNA-seq imputation using generative adversarial networks, Nucleic Acids Res., 2020, 48(15), e85 CrossRef CAS PubMed.
  161. M. Marouf, P. Machart, V. Bansal, C. Kilian, D. S. Magruder, C. F. Krebs and S. Bonn, Realistic in silico generation and augmentation of single-cell RNA-seq data using generative adversarial networks, Nat. Commun., 2020, 11(1), 166 CrossRef CAS PubMed.
  162. P. Zhang, L. Wei, J. Li and X. Wang, Artificial intelligence-guided strategies for next-generation biological sequence design, Natl. Sci. Rev., 2024, 11(11), nwae343 CrossRef PubMed.
  163. R. Lopez, J. Regier, M. B. Cole, M. I. Jordan and N. Yosef, Deep generative modeling for single-cell transcriptomics, Nat. Methods, 2018, 15(12), 1053–1058 CrossRef CAS PubMed.
  164. A. Gayoso, Z. Steier, R. Lopez, J. Regier, K. L. Nazor, A. Streets and N. Yosef, Joint probabilistic modeling of single-cell multi-omic data with totalVI, Nat. Methods, 2021, 18(3), 272–282 CrossRef CAS PubMed.
  165. N. A. Valous, F. Popp, I. Zornig, D. Jager and P. Charoentong, Graph machine learning for integrated multi-omics analysis, Br. J. Cancer, 2024, 131(2), 205–211 CrossRef PubMed.
  166. J. Wang, A. Ma, Y. Chang, J. Gong, Y. Jiang, R. Qi, C. Wang, H. Fu, Q. Ma and D. Xu, scGNN is a novel graph neural network framework for single-cell RNA-Seq analyses, Nat. Commun., 2021, 12(1), 1882 CrossRef CAS PubMed.
  167. A. Regev, S. A. Teichmann, E. S. Lander, I. Amit, C. Benoist, E. Birney, B. Bodenmiller, P. Campbell, P. Carninci, M. Clatworthy, H. Clevers, B. Deplancke, I. Dunham, J. Eberwine, R. Eils, W. Enard, A. Farmer, L. Fugger, B. Gottgens, N. Hacohen, M. Haniffa, M. Hemberg, S. Kim, P. Klenerman, A. Kriegstein, E. Lein, S. Linnarsson, E. Lundberg, J. Lundeberg, P. Majumder, J. C. Marioni, M. Merad, M. Mhlanga, M. Nawijn, M. Netea, G. Nolan, D. Pe’er, A. Phillipakis, C. P. Ponting, S. Quake, W. Reik, O. Rozenblatt-Rosen, J. Sanes, R. Satija, T. N. Schumacher, A. Shalek, E. Shapiro, P. Sharma, J. W. Shin, O. Stegle, M. Stratton, M. J. T. Stubbington, F. J. Theis, M. Uhlen, A. van Oudenaarden, A. Wagner, F. Watt, J. Weissman, B. Wold, R. Xavier and N. Yosef, Human Cell Atlas Meeting, P., The Human Cell Atlas, eLife, 2017, 6, e27041 CrossRef PubMed.
  168. E. Kirby, A. Bernier, R. Guigo, B. Wold, F. Arzuaga, M. Kusunose, M. Zawati and B. M. Knoppers, Data sharing ethics toolkit: The Human Cell Atlas, Nat. Commun., 2024, 15(1), 9901 CrossRef CAS PubMed.
  169. P. Moreno, N. Huang, J. R. Manning, S. Mohammed, A. Solovyev, K. Polanski, W. Bacon, R. Chazarra, C. Talavera-Lopez, M. A. Doyle, G. Marnier, B. Gruning, H. Rasche, N. George, S. K. Fexova, M. Alibi, Z. Miao, Y. Perez-Riverol, M. Haeussler, A. Brazma, S. Teichmann, K. B. Meyer and I. Papatheodorou, User-friendly, scalable tools and workflows for single-cell RNA-seq analysis, Nat. Methods, 2021, 18(4), 327–328 CrossRef CAS PubMed.
  170. C. Tabula Muris, Overall, c.; Logistical, c.; Organ, c.; processing; Library, p.; sequencing; Computational data, a.; Cell type, a.; Writing, g.; Supplemental text writing, g.; Principal, i., Single-cell transcriptomics of 20 mouse organs creates a Tabula Muris, Nature, 2018, 562(7727), 367–372 CrossRef PubMed.
  171. F. A. Wolf, P. Angerer and F. J. Theis, SCANPY: large-scale single-cell gene expression data analysis, Genome Biol., 2018, 19(1), 15 CrossRef PubMed.
  172. A. Butler, P. Hoffman, P. Smibert, E. Papalexi and R. Satija, Integrating single-cell transcriptomic data across different conditions, technologies, and species, Nat. Biotechnol., 2018, 36(5), 411–420 CrossRef CAS PubMed.
  173. C. S. McGinnis, L. M. Murrow and Z. J. Gartner, DoubletFinder: Doublet Detection in Single-Cell RNA Sequencing Data Using Artificial Nearest Neighbors, Cell Syst., 2019, 8(4), 329–337 e4 CrossRef CAS PubMed.
  174. M. D. Young and S. Behjati, SoupX removes ambient RNA contamination from droplet-based single-cell RNA sequencing data, GigaScience, 2020, 9(12), giaa151 CrossRef PubMed.
  175. K. Kamimoto, B. Stringa, C. M. Hoffmann, K. Jindal, L. Solnica-Krezel and S. A. Morris, Dissecting cell identity via network inference and in silico gene perturbation, Nature, 2023, 614(7949), 742–751 CrossRef CAS PubMed.
  176. Z. Zhang, H. Sun, R. Mariappan, X. Chen, X. Chen, M. S. Jain, M. Efremova, S. A. Teichmann, V. Rajan and X. Zhang, scMoMaT jointly performs single cell mosaic integration and multi-modal bio-marker detection, Nat. Commun., 2023, 14(1), 384 CrossRef CAS PubMed.
  177. J. Zhou, J. Dong, H. Hou, L. Huang and J. Li, High-throughput microfluidic systems accelerated by artificial intelligence for biomedical applications, Lab Chip, 2024, 24(5), 1307–1326 RSC.
  178. C. Arnold, Cloud labs: where robots do the research, Nature, 2022, 606(7914), 612–613 CrossRef CAS PubMed.
  179. M. Segal, An operating system for the biology lab, Nature, 2019, 573(7775), S112–S113 CrossRef CAS PubMed.
  180. C. Armer, F. Letronne and E. DeBenedictis, Support academic access to automated cloud labs to improve reproducibility, PLoS Biol., 2023, 21(1), e3001919 CrossRef CAS PubMed.
  181. J. Li, J. Li, R. Liu, Y. Tu, Y. Li, J. Cheng, T. He and X. Zhu, Autonomous discovery of optically active chiral inorganic perovskite nanocrystals through an intelligent cloud lab, Nat. Commun., 2020, 11(1), 2046 CrossRef CAS PubMed.
  182. M. Bates, A. J. Berliner, J. Lachoff, P. R. Jaschke and E. S. Groban, Wet Lab Accelerator: A Web-Based Application Democratizing Laboratory Automation for Synthetic Biology, ACS Synth. Biol., 2017, 6(1), 167–171 CrossRef CAS PubMed.
  183. P. V. Baudin, R. E. Sacksteder, A. K. Worthington, K. Voitiuk, V. T. Ly, R. N. Hoffman, M. A. T. Elliott, D. F. Parks, R. Ward, S. Torres-Montoya, F. Amend, N. Montellano Duran, P. A. Vargas, G. Martinez, S. M. Ramirez, L. E. Alvarado-Arnez, D. Ehrlich, Y. M. Rosen, A. Breevoort, T. Schouten, S. Kurniawan, D. Haussler, M. Teodorescu and M. A. Mostajo-Radji, Cloud-controlled microscopy enables remote project-based biology education in underserved Latinx communities, Heliyon, 2022, 8(11), e11596 CrossRef PubMed.
  184. A. Mund, F. Coscia, A. Kriston, R. Hollandi, F. Kovacs, A. D. Brunner, E. Migh, L. Schweizer, A. Santos, M. Bzorek, S. Naimy, L. M. Rahbek-Gjerdrum, B. Dyring-Andersen, J. Bulkescher, C. Lukas, M. A. Eckert, E. Lengyel, C. Gnann, E. Lundberg, P. Horvath and M. Mann, Deep Visual Proteomics defines single-cell identity and heterogeneity, Nat. Biotechnol., 2022, 40(8), 1231–1240 CrossRef CAS PubMed.
  185. F. A. Rosenberger, M. Thielert, M. T. Strauss, L. Schweizer, C. Ammar, S. C. Madler, A. Metousis, P. Skowronek, M. Wahle, K. Madden, J. Gote-Schniering, A. Semenova, H. B. Schiller, E. Rodriguez, T. M. Nordmann, A. Mund and M. Mann, Spatial single-cell mass spectrometry defines zonation of the hepatocyte proteome, Nat. Methods, 2023, 20(10), 1530–1536 CrossRef CAS PubMed.
  186. A. Rives, J. Meier, T. Sercu, S. Goyal, Z. Lin, J. Liu, D. Guo, M. Ott, C. L. Zitnick, J. Ma and R. Fergus, Biological structure and function emerge from scaling unsupervised learning to 250 million protein sequences, Proc. Natl. Acad. Sci. U. S. A., 2021, 118(15), e2016239118 CrossRef CAS PubMed.
  187. Z. Yang, Z. Jiang, H. Lin, X. Fan, C. Wu, E. Y. Lam, H. K. H. So and H. C. Shum, CeyeHao: AI-driven microfluidic flow programming with hierarchically assembled obstacles and receptive field-augmented neural network, Sci. Adv., 2025, 11(31), eadx2826 CrossRef PubMed.
  188. A. W. Martinez, S. T. Phillips, M. J. Butte and G. M. Whitesides, Patterned paper as a platform for inexpensive, low-volume, portable bioassays, Angew. Chem., Int. Ed., 2007, 46(8), 1318–1320 CrossRef CAS PubMed.
  189. J. Shi, H. Huang, Z. Stratton, Y. Huang and T. J. Huang, Continuous particle separation in a microfluidic channel via standing surface acoustic waves (SSAW), Lab Chip, 2009, 9(23), 3354–3359 RSC.
  190. V. Misuk, A. Mai, K. Giannopoulos, F. Alobaid, B. Epple and H. Loewe, Micro magnetofluidics: droplet manipulation of double emulsions based on paramagnetic ionic liquids, Lab Chip, 2013, 13(23), 4542–4548 RSC.
  191. K. Kamei, Y. Mashimo, Y. Koyama, C. Fockenberg, M. Nakashima, M. Nakajima, J. Li and Y. Chen, 3D printing of soft lithography mold for rapid production of polydimethylsiloxane-based microfluidic devices for cell stimulation with concentration gradients, Biomed. Microdevices, 2015, 17(2), 36 CrossRef PubMed.
  192. H. Zhu and A. Ozcan, Opto-fluidics based microscopy and flow cytometry on a cell phone for blood analysis, Methods Mol. Biol., 2015, 1256, 171–190 CrossRef CAS PubMed.
  193. Y. Liu, Y. Yang, G. Wang, D. Wang, P. L. Shao, J. Tang, T. He, J. Zheng, R. Hu, Y. Liu, Z. Xu, D. Niu, J. Lv, J. Yang, H. Xiao, S. Wu, S. He, Z. Tang, Y. Liu, M. Tang, X. Jiang, J. Yuan, H. Dai and B. Zhang, Multiplexed discrimination of SARS-CoV-2 variants via plasmonic-enhanced fluorescence in a portable and automated device, Nat. Biomed. Eng., 2023, 7(12), 1636–1648 CrossRef CAS PubMed.
  194. Z. Zhang, Microsoft kinect sensor and its effect, IEEE Multimedia, 2012, 19(2), 4–10 Search PubMed.
  195. M. Z. Alom, T. M. Taha, C. Yakopcic, S. Westberg, P. Sidike, M. S. Nasrin, B. C. Van Esesn, A. A. S. Awwal and V. K. Asari, The history began from alexnet: A comprehensive survey on deep learning approaches, arXiv, 2018, preprint, arXiv:1803.01164,  DOI:10.48550/arXiv.1803.01164.
  196. J. X. Chen, The evolution of computing: AlphaGo, Comput. Sci. Eng., 2016, 18(4), 4–7 Search PubMed.
  197. J. Achiam, S. Adler, S. Agarwal, L. Ahmad, I. Akkaya, F. L. Aleman, D. Almeida, J. Altenschmidt, S. Altman and S. Anadkat, Gpt-4 technical report, arXiv, 2023, preprint, arXiv:2303.08774 Search PubMed.
  198. B. Zitkovich, T. Yu, S. Xu, P. Xu, T. Xiao, F. Xia, J. Wu, P. Wohlhart, S. Welker and A. Wahid, Rt-2: Vision-language-action models transfer web knowledge to robotic control, Conference on Robot Learning, PMLR, 2023, pp. 2165–2183 Search PubMed.
  199. B. Burger, P. M. Maffettone, V. V. Gusev, C. M. Aitchison, Y. Bai, X. Wang, X. Li, B. M. Alston, B. Li, R. Clowes, N. Rankin, B. Harris, R. S. Sprick and A. I. Cooper, A mobile robotic chemist, Nature, 2020, 583(7815), 237–241 CrossRef CAS PubMed.
  200. N. Sandstrom, L. Brandt, P. A. Sandoz, C. Zambarda, K. Guldevall, M. Schulz-Ruhtenberg, B. Rosener, R. A. Kruger and B. Onfelt, Live single cell imaging assays in glass microwells produced by laser-induced deep etching, Lab Chip, 2022, 22(11), 2107–2121 RSC.
  201. M. Marouf, P. Machart, V. Bansal, C. Kilian, D. S. Magruder, C. F. Krebs and S. Bonn, Realistic in silico generation and augmentation of single-cell RNA-seq data using generative adversarial networks, Nat. Commun., 2020, 11(1), 166 CrossRef CAS PubMed.

Footnote

Equal contributions.

This journal is © The Royal Society of Chemistry 2025
Click here to see how this site uses Cookies. View our privacy policy here.