Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 63
Filter
Add more filters










Publication year range
1.
Adv Sci (Weinh) ; : e2307591, 2024 Jun 12.
Article in English | MEDLINE | ID: mdl-38864546

ABSTRACT

Image-based cytometry faces challenges due to technical variations arising from different experimental batches and conditions, such as differences in instrument configurations or image acquisition protocols, impeding genuine biological interpretation of cell morphology. Existing solutions, often necessitating extensive pre-existing data knowledge or control samples across batches, have proved limited, especially with complex cell image data. To overcome this, "Cyto-Morphology Adversarial Distillation" (CytoMAD), a self-supervised multi-task learning strategy that distills biologically relevant cellular morphological information from batch variations, is introduced to enable integrated analysis across multiple data batches without complex data assumptions or extensive manual annotation. Unique to CytoMAD is its "morphology distillation", symbiotically paired with deep-learning image-contrast translation-offering additional interpretable insights into label-free cell morphology. The versatile efficacy of CytoMAD is demonstrated in augmenting the power of biophysical imaging cytometry. It allows integrated label-free classification of human lung cancer cell types and accurately recapitulates their progressive drug responses, even when trained without the drug concentration information. CytoMAD  also allows joint analysis of tumor biophysical cellular heterogeneity, linked to epithelial-mesenchymal plasticity, that standard fluorescence markers overlook. CytoMAD can substantiate the wide adoption of biophysical cytometry for cost-effective diagnosis and screening.

2.
Commun Biol ; 6(1): 449, 2023 04 24.
Article in English | MEDLINE | ID: mdl-37095203

ABSTRACT

Complex and irregular cell architecture is known to statistically exhibit fractal geometry, i.e., a pattern resembles a smaller part of itself. Although fractal variations in cells are proven to be closely associated with the disease-related phenotypes that are otherwise obscured in the standard cell-based assays, fractal analysis with single-cell precision remains largely unexplored. To close this gap, here we develop an image-based approach that quantifies a multitude of single-cell biophysical fractal-related properties at subcellular resolution. Taking together with its high-throughput single-cell imaging performance (~10,000 cells/sec), this technique, termed single-cell biophysical fractometry, offers sufficient statistical power for delineating the cellular heterogeneity, in the context of lung-cancer cell subtype classification, drug response assays and cell-cycle progression tracking. Further correlative fractal analysis shows that single-cell biophysical fractometry can enrich the standard morphological profiling depth and spearhead systematic fractal analysis of how cell morphology encodes cellular health and pathological conditions.


Subject(s)
Lung Neoplasms , Humans
3.
Comput Struct Biotechnol J ; 21: 1598-1605, 2023.
Article in English | MEDLINE | ID: mdl-36874160

ABSTRACT

Current single-cell visualisation techniques project high dimensional data into 'map' views to identify high-level structures such as cell clusters and trajectories. New tools are needed to allow the transversal through the high dimensionality of single-cell data to explore the single-cell local neighbourhood. StarmapVis is a convenient web application displaying an interactive downstream analysis of single-cell expression or spatial transcriptomic data. The concise user interface is powered by modern web browsers to explore the variety of viewing angles unavailable to 2D media. Interactive scatter plots display clustering information, while the trajectory and cross-comparison among different coordinates are displayed in connectivity networks. Automated animation of camera view is a unique feature of our tool. StarmapVis also offers a useful animated transition between two-dimensional spatial omic data to three-dimensional single cell coordinates. The usability of StarmapVis is demonstrated by four data sets, showcasing its practical usability. StarmapVis is available at: https://holab-hku.github.io/starmapVis.

4.
Lab Chip ; 23(5): 1011-1033, 2023 03 01.
Article in English | MEDLINE | ID: mdl-36601812

ABSTRACT

Propelled by the striking advances in optical microscopy and deep learning (DL), the role of imaging in lab-on-a-chip has dramatically been transformed from a silo inspection tool to a quantitative "smart" engine. A suite of advanced optical microscopes now enables imaging over a range of spatial scales (from molecules to organisms) and temporal window (from microseconds to hours). On the other hand, the staggering diversity of DL algorithms has revolutionized image processing and analysis at the scale and complexity that were once inconceivable. Recognizing these exciting but overwhelming developments, we provide a timely review of their latest trends in the context of lab-on-a-chip imaging, or coined optofluidic imaging. More importantly, here we discuss the strengths and caveats of how to adopt, reinvent, and integrate these imaging techniques and DL algorithms in order to tailor different lab-on-a-chip applications. In particular, we highlight three areas where the latest advances in lab-on-a-chip imaging and DL can form unique synergisms: image formation, image analytics and intelligent image-guided autonomous lab-on-a-chip. Despite the on-going challenges, we anticipate that they will represent the next frontiers in lab-on-a-chip imaging that will spearhead new capabilities in advancing analytical chemistry research, accelerating biological discovery, and empowering new intelligent clinical applications.


Subject(s)
Deep Learning , Microscopy/methods , Lab-On-A-Chip Devices , Image Processing, Computer-Assisted , Oligonucleotide Array Sequence Analysis
5.
IEEE Open J Eng Med Biol ; 4: 204-211, 2023.
Article in English | MEDLINE | ID: mdl-38274779

ABSTRACT

Microgravity is proven to impact a wide range of human physiology, from stimulating stem cell differentiation to confounding cell health in bones, skeletal muscles, and blood cells. The research in this arena is progressively intensified by the increasing promises of human spaceflights. Considering the limited access to spaceflight, ground-based microgravity-simulating platforms have been indispensable for microgravity-biology research. However, they are generally complex, costly, hard to replicate and reconfigure - hampering the broad adoption of microgravity biology and astrobiology. To address these limitations, we developed a low-cost reconfigurable 3D-printed microscope coined EuniceScope to allow the democratization of astrobiology, especially for educational use. EuniceScope is a compact 2D clinostat system integrated with a modularized brightfield microscope, built upon 3D-printed toolbox. We demonstrated that this compact system offers plausible imaging quality and microgravity-simulating performance. Its high degree of reconfigurability thus holds great promise in the wide dissemination of microgravity-cell-biology research in the broader community, including Science, technology, engineering, and mathematics (STEM) educational and scientific community in the future.

6.
Proc Natl Acad Sci U S A ; 119(23): e2117346119, 2022 06 07.
Article in English | MEDLINE | ID: mdl-35648820

ABSTRACT

Characterizing blood flow dynamics in vivo is critical to understanding the function of the vascular network under physiological and pathological conditions. Existing methods for hemodynamic imaging have insufficient spatial and temporal resolution to monitor blood flow at the cellular level in large blood vessels. By using an ultrafast line-scanning module based on free-space angular chirped enhanced delay, we achieved two-photon fluorescence imaging of cortical blood flow at 1,000 two-dimensional (2D) frames and 1,000,000 one-dimensional line scans per second in the awake mouse. This orders-of-magnitude increase in temporal resolution allowed us to measure cerebral blood flow at up to 49 mm/s and observe pulsatile blood flow at harmonics of heart rate. Directly visualizing red blood cell (RBC) flow through vessels down to >800 µm in depth, we characterized cortical layer­dependent flow velocity distributions of capillaries, obtained radial velocity profiles and kilohertz 2D velocity mapping of multifile blood flow, and performed RBC flux measurements from penetrating blood vessels.


Subject(s)
Brain , Cerebrovascular Circulation , Animals , Brain/blood supply , Brain/diagnostic imaging , Erythrocytes , Heart Rate , Mice , Microscopy, Fluorescence/methods , Optical Imaging , Photons
7.
Opt Lett ; 47(11): 2710-2713, 2022 Jun 01.
Article in English | MEDLINE | ID: mdl-35648911

ABSTRACT

We demonstrate second-harmonic generation (SHG) microscopy excited by the ∼890-nm light frequency-doubled from a 137-fs, 19.4-MHz, and 300-mW all-fiber mode-locked laser centered at 1780 nm. The mode-locking at the 1.7-µm window is realized by controlling the emission peak of the gain fiber, and uses the dispersion management technique to broaden the optical spectrum up to 30 nm. The spectrum is maintained during the amplification and the pulse is compressed by single-mode fibers. The SHG imaging performance is showcased on a mouse skull, leg, and tail. Two-photon fluorescence imaging is also demonstrated on C. elegans labeled with green and red fluorescent proteins. The frequency-doubled all-fiber laser system provides a compact and efficient tool for SHG and fluorescence microscopy.


Subject(s)
Caenorhabditis elegans , Lasers , Animals , Mice , Microscopy, Fluorescence , Optical Imaging , Photons
8.
IEEE Trans Neural Netw Learn Syst ; 33(7): 2853-2866, 2022 07.
Article in English | MEDLINE | ID: mdl-33434136

ABSTRACT

Real-time in situ image analytics impose stringent latency requirements on intelligent neural network inference operations. While conventional software-based implementations on the graphic processing unit (GPU)-accelerated platforms are flexible and have achieved very high inference throughput, they are not suitable for latency-sensitive applications where real-time feedback is needed. Here, we demonstrate that high-performance reconfigurable computing platforms based on field-programmable gate array (FPGA) processing can successfully bridge the gap between low-level hardware processing and high-level intelligent image analytics algorithm deployment within a unified system. The proposed design performs inference operations on a stream of individual images as they are produced and has a deeply pipelined hardware design that allows all layers of a quantized convolutional neural network (QCNN) to compute concurrently with partial image inputs. Using the case of label-free classification of human peripheral blood mononuclear cell (PBMC) subtypes as a proof-of-concept illustration, our system achieves an ultralow classification latency of 34.2 [Formula: see text] with over 95% end-to-end accuracy by using a QCNN, while the cells are imaged at throughput exceeding 29 200 cells/s. Our QCNN design is modular and is readily adaptable to other QCNNs with different latency and resource requirements.


Subject(s)
Leukocytes, Mononuclear , Neural Networks, Computer , Algorithms , Humans , Image Processing, Computer-Assisted/methods , Software
9.
Nat Commun ; 12(1): 5528, 2021 09 20.
Article in English | MEDLINE | ID: mdl-34545085

ABSTRACT

Inferring cellular trajectories using a variety of omic data is a critical task in single-cell data science. However, accurate prediction of cell fates, and thereby biologically meaningful discovery, is challenged by the sheer size of single-cell data, the diversity of omic data types, and the complexity of their topologies. We present VIA, a scalable trajectory inference algorithm that overcomes these limitations by using lazy-teleporting random walks to accurately reconstruct complex cellular trajectories beyond tree-like pathways (e.g., cyclic or disconnected structures). We show that VIA robustly and efficiently unravels the fine-grained sub-trajectories in a 1.3-million-cell transcriptomic mouse atlas without losing the global connectivity at such a high cell count. We further apply VIA to discovering elusive lineages and less populous cell fates missed by other methods across a variety of data types, including single-cell proteomic, epigenomic, multi-omics datasets, and a new in-house single-cell morphological dataset.


Subject(s)
Algorithms , Genomics , Single-Cell Analysis , Animals , Cell Cycle , Cell Differentiation , Cell Line, Tumor , Cell Shape , Hematopoiesis , Humans , Islets of Langerhans/cytology , LIM-Homeodomain Proteins/metabolism , Mesoderm/cytology , Mice , Mouse Embryonic Stem Cells/cytology , Organogenesis , Transcription Factors/metabolism
10.
Nat Protoc ; 16(9): 4227-4264, 2021 09.
Article in English | MEDLINE | ID: mdl-34341580

ABSTRACT

Laser scanning is used in advanced biological microscopy to deliver superior imaging contrast, resolution and sensitivity. However, it is challenging to scale up the scanning speed required for interrogating a large and heterogeneous population of biological specimens or capturing highly dynamic biological processes at high spatiotemporal resolution. Bypassing the speed limitation of traditional mechanical methods, free-space angular-chirp-enhanced delay (FACED) is an all-optical, passive and reconfigurable laser-scanning approach that has been successfully applied in different microscopy modalities at an ultrafast line-scan rate of 1-80 MHz. Optimal FACED imaging performance requires optimized experimental design and implementation to enable specific high-speed applications. In this protocol, we aim to disseminate information allowing FACED to be applied to a broader range of imaging modalities. We provide (i) a comprehensive guide and design specifications for the FACED hardware; (ii) step-by-step optical implementations of the FACED module including the key custom components; and (iii) the overall image acquisition and reconstruction pipeline. We illustrate two practical imaging configurations: multimodal FACED imaging flow cytometry (bright-field, fluorescence and second-harmonic generation) and kHz 2D two-photon fluorescence microscopy. Users with basic experience in optical microscope operation and software engineering should be able to complete the setup of the FACED imaging hardware and software in ~2-3 months.


Subject(s)
Microscopy, Confocal/methods , Optical Imaging/methods , Flow Cytometry , Microscopy, Confocal/instrumentation , Microscopy, Fluorescence, Multiphoton , Optical Imaging/instrumentation
11.
Trends Biotechnol ; 39(12): 1249-1262, 2021 12.
Article in English | MEDLINE | ID: mdl-33895013

ABSTRACT

The biophysical properties of cells reflect their identities, underpin their homeostatic state in health, and define the pathogenesis of disease. Recent leapfrogging advances in biophysical cytometry now give access to this information, which is obscured in molecular assays, with a discriminative power that was once inconceivable. However, biophysical cytometry should go 'deeper' in terms of exploiting the information-rich cellular biophysical content, generating a molecular knowledge base of cellular biophysical properties, and standardizing the protocols for wider dissemination. Overcoming these barriers, which requires concurrent innovations in microfluidics, optical imaging, and computer vision, could unleash the enormous potential of biophysical cytometry not only for gaining a new mechanistic understanding of biological systems but also for identifying new cost-effective biomarkers of disease.


Subject(s)
Microfluidics , Optical Imaging , Biomarkers , Biophysics , Flow Cytometry/methods , Microfluidics/methods , Optical Imaging/methods
12.
Lab Chip ; 20(20): 3696-3708, 2020 10 21.
Article in English | MEDLINE | ID: mdl-32935707

ABSTRACT

The association of the intrinsic optical and biophysical properties of cells to homeostasis and pathogenesis has long been acknowledged. Defining these label-free cellular features obviates the need for costly and time-consuming labelling protocols that perturb the living cells. However, wide-ranging applicability of such label-free cell-based assays requires sufficient throughput, statistical power and sensitivity that are unattainable with current technologies. To close this gap, we present a large-scale, integrative imaging flow cytometry platform and strategy that allows hierarchical analysis of intrinsic morphological descriptors of single-cell optical and mass density within a population of millions of cells. The optofluidic cytometry system also enables the synchronous single-cell acquisition of and correlation with fluorescently labeled biochemical markers. Combined with deep neural network and transfer learning, this massive single-cell profiling strategy demonstrates the label-free power to delineate the biophysical signatures of the cancer subtypes, to detect rare populations of cells in the heterogeneous samples (10-5), and to assess the efficacy of targeted therapeutics. This technique could spearhead the development of optofluidic imaging cell-based assays that stratify the underlying physiological and pathological processes based on the information-rich biophysical cellular phenotypes.


Subject(s)
Deep Learning , Biophysics , Flow Cytometry , Image Cytometry , Phenotype
13.
Opt Lett ; 45(11): 3054-3057, 2020 Jun 01.
Article in English | MEDLINE | ID: mdl-32479457

ABSTRACT

The resolution enhancement over the extended depth of field (DOF) in the volumetric two-photon microscopy (TPM) is demonstrated by utilizing multiple orders of Bessel beams. Here the conventional method of switching laser modes (SLAM) in 2D is introduced to 3D, denoted as the volumetric SLAM (V-SLAM). The equivalent scanning beam in the TPM is a thin needle-like beam, which is generated from the subtraction between the needle-like 0th-order and the straw-like 1st-order Bessel beams. Compared with the 0th-order Bessel beam, the lateral resolution of the V-SLAM is increased by 28.6% and maintains over the axial depth of 56 µm. The V-SLAM performance is evaluated by employing fluorescent beads and a mouse brain slice. The V-SLAM approach provides a promising solution to improve the lateral resolutions for fast volumetric imaging on sparsely distributed samples.

14.
Light Sci Appl ; 9: 25, 2020.
Article in English | MEDLINE | ID: mdl-32133128

ABSTRACT

Coherent Raman scattering (CRS) microscopy is widely recognized as a powerful tool for tackling biomedical problems based on its chemically specific label-free contrast, high spatial and spectral resolution, and high sensitivity. However, the clinical translation of CRS imaging technologies has long been hindered by traditional solid-state lasers with environmentally sensitive operations and large footprints. Ultrafast fibre lasers can potentially overcome these shortcomings but have not yet been fully exploited for CRS imaging, as previous implementations have suffered from high intensity noise, a narrow tuning range and low power, resulting in low image qualities and slow imaging speeds. Here, we present a novel high-power self-synchronized two-colour pulsed fibre laser that achieves excellent performance in terms of intensity stability (improved by 50 dB), timing jitter (24.3 fs), average power fluctuation (<0.5%), modulation depth (>20 dB) and pulse width variation (<1.8%) over an extended wavenumber range (2700-3550 cm-1). The versatility of the laser source enables, for the first time, high-contrast, fast CRS imaging without complicated noise reduction via balanced detection schemes. These capabilities are demonstrated in this work by imaging a wide range of species such as living human cells and mouse arterial tissues and performing multimodal nonlinear imaging of mouse tail, kidney and brain tissue sections by utilizing second-harmonic generation and two-photon excited fluorescence, which provides multiple optical contrast mechanisms simultaneously and maximizes the gathered information content for biological visualization and medical diagnosis. This work also establishes a general scenario for remodelling existing lasers into synchronized two-colour lasers and thus promotes a wider popularization and application of CRS imaging technologies.

15.
Nat Methods ; 17(3): 287-290, 2020 03.
Article in English | MEDLINE | ID: mdl-32123392

ABSTRACT

Understanding information processing in the brain requires monitoring neuronal activity at high spatiotemporal resolution. Using an ultrafast two-photon fluorescence microscope empowered by all-optical laser scanning, we imaged neuronal activity in vivo at up to 3,000 frames per second and submicrometer spatial resolution. This imaging method enabled monitoring of both supra- and subthreshold electrical activity down to 345 µm below the brain surface in head-fixed awake mice.


Subject(s)
Brain/diagnostic imaging , Microscopy, Fluorescence, Multiphoton/methods , Neurons/physiology , Photons , Animals , Calcium/metabolism , Cells, Cultured , Computational Biology , Female , Glutamic Acid/metabolism , Lasers , Male , Membrane Potentials , Mice , Mice, Transgenic , Optics and Photonics , Rats , Software
16.
Bioinformatics ; 36(9): 2778-2786, 2020 05 01.
Article in English | MEDLINE | ID: mdl-31971583

ABSTRACT

MOTIVATION: New single-cell technologies continue to fuel the explosive growth in the scale of heterogeneous single-cell data. However, existing computational methods are inadequately scalable to large datasets and therefore cannot uncover the complex cellular heterogeneity. RESULTS: We introduce a highly scalable graph-based clustering algorithm PARC-Phenotyping by Accelerated Refined Community-partitioning-for large-scale, high-dimensional single-cell data (>1 million cells). Using large single-cell flow and mass cytometry, RNA-seq and imaging-based biophysical data, we demonstrate that PARC consistently outperforms state-of-the-art clustering algorithms without subsampling of cells, including Phenograph, FlowSOM and Flock, in terms of both speed and ability to robustly detect rare cell populations. For example, PARC can cluster a single-cell dataset of 1.1 million cells within 13 min, compared with >2 h for the next fastest graph-clustering algorithm. Our work presents a scalable algorithm to cope with increasingly large-scale single-cell analysis. AVAILABILITY AND IMPLEMENTATION: https://github.com/ShobiStassen/PARC. SUPPLEMENTARY INFORMATION: Supplementary data are available at Bioinformatics online.


Subject(s)
Algorithms , Single-Cell Analysis , Cluster Analysis , RNA-Seq , Software , Exome Sequencing
17.
Light Sci Appl ; 9: 8, 2020.
Article in English | MEDLINE | ID: mdl-31993126

ABSTRACT

Parallelized fluorescence imaging has been a long-standing pursuit that can address the unmet need for a comprehensive three-dimensional (3D) visualization of dynamical biological processes with minimal photodamage. However, the available approaches are limited to incomplete parallelization in only two dimensions or sparse sampling in three dimensions. We hereby develop a novel fluorescence imaging approach, called coded light-sheet array microscopy (CLAM), which allows complete parallelized 3D imaging without mechanical scanning. Harnessing the concept of an "infinity mirror", CLAM generates a light-sheet array with controllable sheet density and degree of coherence. Thus, CLAM circumvents the common complications of multiple coherent light-sheet generation in terms of dedicated wavefront engineering and mechanical dithering/scanning. Moreover, the encoding of multiplexed optical sections in CLAM allows the synchronous capture of all sectioned images within the imaged volume. We demonstrate the utility of CLAM in different imaging scenarios, including a light-scattering medium, an optically cleared tissue, and microparticles in fluidic flow. CLAM can maximize the signal-to-noise ratio and the spatial duty cycle, and also provides a further reduction in photobleaching compared to the major scanning-based 3D imaging systems. The flexible implementation of CLAM regarding both hardware and software ensures compatibility with any light-sheet imaging modality and could thus be instrumental in a multitude of areas in biological research.

18.
Opt Lett ; 44(21): 5238-5241, 2019 Nov 01.
Article in English | MEDLINE | ID: mdl-31674977

ABSTRACT

We demonstrate dual-Airy-beam-scanning-based volumetric two-photon microscopy (TPM) with depth-resolving capability. A pair of Airy beams with opposite acceleration is used as the excitation lights to sequentially illuminate the sample, and depth information can be resolved based on the deflection of the Airy beam. The depth-resolving range of the volumetric TPM is up to 32 µm. The advantages of the depth-resolved volumetric TPM are the depth-resolving capability over Bessel-beam-based TPM and less scanning times over traditional Gaussian-beam-based TPM. The depth-resolved volumetric TPM provides a promising fast imaging tool to study the dynamics in neural biology.

19.
IEEE Trans Biomed Circuits Syst ; 13(4): 781-792, 2019 08.
Article in English | MEDLINE | ID: mdl-31059454

ABSTRACT

A fundamental technical challenge for ultra-fast cell microscopy is the tradeoff between imaging throughput and resolution. In addition to throughput, real-time applications such as image-based cell sorting further requires ultra-low imaging latency to facilitate rapid decision making on a single-cell level. Using a novel coprime line scan sampling scheme, a real-time low-latency hardware super-resolution system for ultra-fast time-stretch microscopy is presented. The proposed scheme utilizes analog-to-digital converter with a carefully tuned sampling pattern (shifted sampling grid) to enable super-resolution image reconstruction using line scan input from an optical front-end. A fully pipelined FPGA-based system is built to efficiently handle the real-time high-resolution image reconstruction process with the input subpixel samples while achieving minimal output latency. The proposed super-resolution sampling and reconstruction scheme is parametrizable and is readily applicable to different line scan imaging systems. In our experiments, an imaging latency of 0.29 µs has been achieved based on a pixel-stream throughput of 4.123 giga pixels per second, which translates into imaging throughput of approximately 120000 cells per second.


Subject(s)
Algorithms , Microscopy/methods , Cell Line, Tumor , Humans , Image Processing, Computer-Assisted
20.
Cytometry A ; 95(5): 510-520, 2019 05.
Article in English | MEDLINE | ID: mdl-31012276

ABSTRACT

Cellular biophysical properties are the effective label-free phenotypes indicative of differences in cell types, states, and functions. However, current biophysical phenotyping methods largely lack the throughput and specificity required in the majority of cell-based assays that involve large-scale single-cell characterization for inquiring the inherently complex heterogeneity in many biological systems. Further confounded by the lack of reported robust reproducibility and quality control, widespread adoption of single-cell biophysical phenotyping in mainstream cytometry remains elusive. To address this challenge, here we present a label-free imaging flow cytometer built upon a recently developed ultrafast quantitative phase imaging (QPI) technique, coined multi-ATOM, that enables label-free single-cell QPI, from which a multitude of subcellularly resolvable biophysical phenotypes can be parametrized, at an experimentally recorded throughput of >10,000 cells/s-a capability that is otherwise inaccessible in current QPI. With the aim to translate multi-ATOM into mainstream cytometry, we report robust system calibration and validation (from image acquisition to phenotyping reproducibility) and thus demonstrate its ability to establish high-dimensional single-cell biophysical phenotypic profiles at ultra-large-scale (>1,000,000 cells). Such a combination of throughput and content offers sufficiently high label-free statistical power to classify multiple human leukemic cell types at high accuracy (~92-97%). This system could substantiate the significance of high-throughput QPI flow cytometry in enabling next frontier in large-scale image-derived single-cell analysis applied in biological discovery and cost-effective clinical diagnostics. © 2019 International Society for Advancement of Cytometry.


Subject(s)
Biophysical Phenomena , Flow Cytometry/methods , Image Processing, Computer-Assisted , Single-Cell Analysis , Blood Cells/pathology , Calibration , Cell Line, Tumor , Humans , Leukemia/pathology , Multivariate Analysis , Phenotype , Reproducibility of Results
SELECTION OF CITATIONS
SEARCH DETAIL
...