1
|
Kim GD, Liu G, Qiu D, De Leo MG, Gopaldass N, Hermes J, Timmer J, Saiardi A, Mayer A, Jessen HJ. Pools of Independently Cycling Inositol Phosphates Revealed by Pulse Labeling with 18O-Water. J Am Chem Soc 2025; 147:17626-17641. [PMID: 40372010 PMCID: PMC12123611 DOI: 10.1021/jacs.4c16206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2024] [Revised: 05/08/2025] [Accepted: 05/08/2025] [Indexed: 05/16/2025]
Abstract
Inositol phosphates control many central processes in eukaryotic cells including nutrient availability, growth, and motility. Kinetic resolution of a key modulator of their signaling functions, the turnover of the phosphate groups on the inositol ring, has been hampered by slow uptake, high dilution, and constraining growth conditions in radioactive pulse-labeling approaches. Here, we demonstrate a rapid (seconds to minutes) and nonradioactive labeling strategy of inositol polyphosphates through 18O-water in yeast, human cells, and amoeba, which can be applied in any media. In combination with capillary electrophoresis and mass spectrometry, 18O-water labeling simultaneously dissects the in vivo phosphate group dynamics of a broad spectrum of even rare inositol phosphates. The good temporal resolution allowed us to discover vigorous phosphate group exchanges in some inositol polyphosphates and pyrophosphates, whereas others remain remarkably inert. We propose a model in which the biosynthetic pathway of inositol polyphosphates and pyrophosphates is organized in distinct, kinetically separated pools. While transfer of compounds between those pools is slow, each pool undergoes rapid internal phosphate cycling. This might enable the pools to perform distinct signaling functions while being metabolically connected.
Collapse
Affiliation(s)
- Geun-Don Kim
- Département
d’immunobiologie, Université
de Lausanne, CH-1066Epalinges, Switzerland
| | - Guizhen Liu
- Institute
of Organic Chemistry, University of Freiburg, 79104Freiburg, Germany
- CIBSSCentre
for Integrative Biological Signaling Studies, University of Freiburg, 79104Freiburg, Germany
| | - Danye Qiu
- Institute
of Organic Chemistry, University of Freiburg, 79104Freiburg, Germany
| | | | - Navin Gopaldass
- Département
d’immunobiologie, Université
de Lausanne, CH-1066Epalinges, Switzerland
| | - Jacques Hermes
- CIBSSCentre
for Integrative Biological Signaling Studies, University of Freiburg, 79104Freiburg, Germany
- Institute
of Physics, University of Freiburg, 79104Freiburg, Germany
| | - Jens Timmer
- CIBSSCentre
for Integrative Biological Signaling Studies, University of Freiburg, 79104Freiburg, Germany
- Institute
of Physics, University of Freiburg, 79104Freiburg, Germany
| | - Adolfo Saiardi
- Medical
Research Council, Laboratory for Molecular Cell Biology, University College London, WC1E 6BTLondon, U.K.
| | - Andreas Mayer
- Département
d’immunobiologie, Université
de Lausanne, CH-1066Epalinges, Switzerland
| | - Henning Jacob Jessen
- Institute
of Organic Chemistry, University of Freiburg, 79104Freiburg, Germany
- CIBSSCentre
for Integrative Biological Signaling Studies, University of Freiburg, 79104Freiburg, Germany
| |
Collapse
|
2
|
Zhang J, Liu L, Zhou Q, Wang D, Liang Y, Liu N, Guo Y, Yin Y, He B, Hu L, Jiang G. Accurate Quantification of Metals in Individual Synechocystis sp. PCC 6803 Cells by Single-Cell ICP-MS: Dual-Calibration and Sample Stabilization Strategies. Anal Chem 2025; 97:10867-10876. [PMID: 40361305 DOI: 10.1021/acs.analchem.5c01304] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/15/2025]
Abstract
Single-cell inductively coupled plasma mass spectrometry (SC-ICP-MS) is an emerging technique to investigate metal heterogeneity in individual cells. However, due to the absence of consistent calibration and suitable stabilization strategy for cells, accurate quantification of cellular heterogeneity and the content of metals remains a challenge. Herein, an accurate quantification method for the content and heterogeneous distribution of metals among individual microalgae cells was developed based on SC-ICP-MS using dual-calibration strategies and robust pretreatment methods. Gold nanoparticles (AuNPs) were used as calibration for measuring metal contents in single cells, but it would lead to a 13.6-63.1% underestimation of cell numbers due to inaccurate detection of cells' transport efficiency. To avoid this inaccuracy, we proposed an additional calibration strategy to measure cellular transport efficiency and cell numbers using endogenous Mg, enabling a more accurate assessment of cell heterogeneity. Then, an effective pretreatment method was optimized through fixation of cells with glutaraldehyde for 1 h to maintain the cellular stability and obtain accurate results, with satisfactory recoveries for cell number (98.4%) and Mg contents (91.7%), even after long-time storage. After optimization, the proposed method showed high sensitivity and repeatability in both cellular metal contents (Mg, Hg, Cd, and Co) and cell number, with detection limits (LODs) to be 0.14-0.53 fg/cell and 5.5 × 103 cells/mL, respectively. Finally, the proposed method was successfully used for detecting various metals and their heterogeneity in Synechocystis sp. PCC 6803 cells provided an accurate and robust tool for investigating the uptake and heterogeneous distribution of metals in microalgae.
Collapse
Affiliation(s)
- Junhui Zhang
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China
- College of Resources and Environment, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Lihong Liu
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China
- College of Resources and Environment, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Qinfei Zhou
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China
- College of Resources and Environment, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Dingyi Wang
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China
| | - Yuan Liang
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China
- School of Environment, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310024, China
| | - Nian Liu
- School of Environment, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310024, China
| | - Yingying Guo
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China
| | - Yongguang Yin
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China
- College of Resources and Environment, University of Chinese Academy of Sciences, Beijing 100049, China
- School of Environment, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310024, China
| | - Bin He
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China
- College of Resources and Environment, University of Chinese Academy of Sciences, Beijing 100049, China
- School of Environment, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310024, China
| | - Ligang Hu
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China
- College of Resources and Environment, University of Chinese Academy of Sciences, Beijing 100049, China
- School of Environment, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310024, China
- Hubei Key Laboratory of Environmental and Health Effects of Persistent Toxic Substances, School of Environment and Health, Jianghan University, Wuhan 430056, China
| | - Guibin Jiang
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China
- College of Resources and Environment, University of Chinese Academy of Sciences, Beijing 100049, China
- School of Environment, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310024, China
| |
Collapse
|
3
|
Muhammed Y, De Sabatino M, Lazenby RA. The Heterogeneity in the Response of A549 Cells to Toyocamycin Observed Using Hopping Scanning Ion Conductance Microscopy. J Phys Chem B 2025; 129:4904-4916. [PMID: 40338629 DOI: 10.1021/acs.jpcb.4c08793] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/09/2025]
Abstract
Scanning ion conductance microscopy (SICM) is a noninvasive topographic mapping technique used in imaging live cells, unlike electron microscopy and certain applications of fluorescence microscopy, which can disrupt cell integrity. In this study, we used SICM to track the morphological changes of the same A549 cells to uncover the cell-to-cell heterogeneity in their response to the drug. We found that toyocamycin (TOY) induced rapid reorganization of the actin cytoskeleton in A549 cells, causing them to become circular, irregular, or ellipsoidal in shape. Mapping of the dynamic changes in morphology revealed membrane blebbing and a significant decrease in volume over time. Using high-throughput SICM, we mapped the morphology of multiple single cells treated with TOY, which revealed that A549 showed characteristics of apoptosis and necrosis. The drug treatment does not significantly change the average root-mean-square (RMS) roughness of the cells. However, the drug leads to an increase in membrane height, possibly indicating early apoptotic changes. Plotting the individual RMS roughness of the cells showed a cell with an increase in roughness and the presence of pores, which is also an indication of necrosis behavior. Our results demonstrate that SICM is an effective technique for revealing the evolution of heterogeneity in single cells in their responses to anticancer drugs over time.
Collapse
Affiliation(s)
- Yusuf Muhammed
- Department of Chemistry and Biochemistry, Florida State University, Tallahassee, Florida 32306, United States
| | - Mia De Sabatino
- Department of Chemistry and Biochemistry, Florida State University, Tallahassee, Florida 32306, United States
| | - Robert A Lazenby
- Department of Chemistry and Biochemistry, Florida State University, Tallahassee, Florida 32306, United States
| |
Collapse
|
4
|
Panagopoulos A, Stout M, Kilic S, Leary P, Vornberger J, Pasti V, Galarreta A, Lezaja A, Kirschenbühler K, Imhof R, Rehrauer H, Ziegler U, Altmeyer M. Multigenerational cell tracking of DNA replication and heritable DNA damage. Nature 2025:10.1038/s41586-025-08986-0. [PMID: 40399682 DOI: 10.1038/s41586-025-08986-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Accepted: 04/04/2025] [Indexed: 05/23/2025]
Abstract
Cell heterogeneity is a universal feature of life. Although biological processes affected by cell-to-cell variation are manifold, from developmental plasticity to tumour heterogeneity and differential drug responses, the sources of cell heterogeneity remain largely unclear1,2. Mutational and epigenetic signatures from cancer (epi)genomics are powerful for deducing processes that shaped cancer genome evolution3-5. However, retrospective analyses face difficulties in resolving how cellular heterogeneity emerges and is propagated to subsequent cell generations. Here, we used multigenerational single-cell tracking based on endogenously labelled proteins and custom-designed computational tools to elucidate how oncogenic perturbations induce sister cell asymmetry and phenotypic heterogeneity. Dual CRISPR-based genome editing enabled simultaneous tracking of DNA replication patterns and heritable endogenous DNA lesions. Cell lineage trees of up to four generations were tracked in asynchronously growing cells, and time-resolved lineage analyses were combined with end-point measurements of cell cycle and DNA damage markers through iterative staining. Besides revealing replication and repair dynamics, damage inheritance and emergence of sister cell heterogeneity across multiple cell generations, through combination with single-cell transcriptomics, we delineate how common oncogenic events trigger multiple routes towards polyploidization with distinct outcomes for genome integrity. Our study provides a framework to dissect phenotypic plasticity at the single-cell level and sheds light onto cellular processes that may resemble early events during cancer development.
Collapse
Affiliation(s)
- Andreas Panagopoulos
- Department of Molecular Mechanisms of Disease, University of Zurich, Zurich, Switzerland
| | - Merula Stout
- Department of Molecular Mechanisms of Disease, University of Zurich, Zurich, Switzerland
| | - Sinan Kilic
- Department of Molecular Mechanisms of Disease, University of Zurich, Zurich, Switzerland
- The Novo Nordisk Foundation Center for Protein Research, University of Copenhagen, Copenhagen, Denmark
| | - Peter Leary
- Functional Genomics Center Zurich, ETH Zurich and University of Zurich, Zurich, Switzerland
| | - Julia Vornberger
- Department of Molecular Mechanisms of Disease, University of Zurich, Zurich, Switzerland
| | - Virginia Pasti
- Department of Molecular Mechanisms of Disease, University of Zurich, Zurich, Switzerland
| | - Antonio Galarreta
- Department of Molecular Mechanisms of Disease, University of Zurich, Zurich, Switzerland
| | - Aleksandra Lezaja
- Department of Molecular Mechanisms of Disease, University of Zurich, Zurich, Switzerland
| | - Kyra Kirschenbühler
- Department of Molecular Mechanisms of Disease, University of Zurich, Zurich, Switzerland
- NEXUS Personalized Health, ETH Zurich, Schlieren, Switzerland
| | - Ralph Imhof
- Department of Molecular Mechanisms of Disease, University of Zurich, Zurich, Switzerland
| | - Hubert Rehrauer
- Functional Genomics Center Zurich, ETH Zurich and University of Zurich, Zurich, Switzerland
| | - Urs Ziegler
- Center for Microscopy and Image Analysis, University of Zurich, Zurich, Switzerland
| | - Matthias Altmeyer
- Department of Molecular Mechanisms of Disease, University of Zurich, Zurich, Switzerland.
| |
Collapse
|
5
|
Nachtigall PG, Hamilton BR, Kazandjian TD, Stincone P, Petras D, Casewell NR, Undheim EAB. The gene regulatory mechanisms shaping the heterogeneity of venom production in the Cape coral snake. Genome Biol 2025; 26:130. [PMID: 40390047 PMCID: PMC12087220 DOI: 10.1186/s13059-025-03602-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Accepted: 05/02/2025] [Indexed: 05/21/2025] Open
Abstract
BACKGROUND Venoms and their associated glands and delivery structures have evolved numerous times among animals. Within these venom systems, the molecular, cellular, and morphological components interact and co-evolve to generate distinct, venom phenotypes that are increasingly recognized as models for studying adaptive evolution. However, toxins are often unevenly distributed across venom-producing tissues in patterns that are not necessarily adaptive but instead likely result from constraints associated with protein secretion. RESULTS We generate a high-quality draft genome of the Cape coral snake (Aspidelaps lubricus) and combine analyses of venom gland single-cell RNA-seq data with spatial venom gland in situ toxin distributions. Our results reveal that while different toxin families are produced by distinct populations of cells, toxin expression is fine-tuned by regulatory modules that result in further specialization of toxin production within each cell population. We also find that the evolution of regulatory elements closely mirrors the evolution of their associated toxin genes, resulting in spatial association of closely related and functionally similar toxins in the venom gland. While this compartmentalization is non-adaptive, the modularity of the underlying regulatory network likely facilitated the repeated evolution of defensive venom in spitting cobras. CONCLUSIONS Our results provide new insight into the variability of toxin regulation across snakes, reveal the molecular mechanisms underlying the heterogeneous toxin production in snake venom glands, and provide an example of how constraints can result in non-adaptive character states that appear to be adaptive, which may nevertheless facilitate evolutionary innovation and novelty.
Collapse
Affiliation(s)
- Pedro G Nachtigall
- Centre for Ecological and Evolutionary Synthesis, Department of Biosciences, University of Oslo, PO Box 1066 Blindern, Oslo, 0316, Norway.
| | - Brett R Hamilton
- Centre for Microscopy and Microanalysis, University of Queensland, St Lucia, Brisbane, QLD, 4072, Australia
| | - Taline D Kazandjian
- Centre for Snakebite Research & Interventions, Liverpool School of Tropical Medicine, Pembroke Place, Liverpool, L3 5QA, UK
| | - Paolo Stincone
- Interfaculty Institute of Microbiology and Infection Medicine, University of Tübingen, Auf der Morgenstelle 28, Tübingen, 72076, Germany
| | - Daniel Petras
- Interfaculty Institute of Microbiology and Infection Medicine, University of Tübingen, Auf der Morgenstelle 28, Tübingen, 72076, Germany
- Department of Biochemistry, University of California Riverside, Riverside, 92507, CA, USA
| | - Nicholas R Casewell
- Centre for Snakebite Research & Interventions, Liverpool School of Tropical Medicine, Pembroke Place, Liverpool, L3 5QA, UK
| | - Eivind A B Undheim
- Centre for Ecological and Evolutionary Synthesis, Department of Biosciences, University of Oslo, PO Box 1066 Blindern, Oslo, 0316, Norway.
| |
Collapse
|
6
|
Raj P, Gupta H, Anantha P, Barman I. Cell-TIMP: Cellular Trajectory Inference Based on Morphological Parameters. NANO LETTERS 2025; 25:7845-7852. [PMID: 40317256 DOI: 10.1021/acs.nanolett.5c01009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/07/2025]
Abstract
Traditional approaches to studying cellular morphology rely on geometric metrics from stained images. However, staining processes can disrupt the cell's natural state and diminish accuracy due to photobleaching, while conventional analysis techniques, which categorize cells based on shape to discern pathophysiological conditions, often fail to capture the continuous and asynchronous nature of biological processes such as cell differentiation, immune responses, and cancer progression. In this work, we propose the use of quantitative phase imaging for morphological assessment due to its label-free nature. For analysis, we repurposed the genomic analysis toolbox to perform trajectory inference analysis purely based on morphology information. We applied the developed framework to study the progression of leukemia and breast cancer metastasis. Applying this framework to leukemia and breast cancer metastasis, we identified key shape changes linked to disease progression, highlighting the method's potential to enhance understanding of complex biological dynamics.
Collapse
Affiliation(s)
- Piyush Raj
- Department of Mechanical Engineering, Johns Hopkins University, Baltimore, Maryland 21218, United States
| | - Himanshu Gupta
- Centre for Applied Autonomous Sensor Systems (AASS), Örebro University, Örebro 70182, Sweden
| | - Pooja Anantha
- Department of Mechanical Engineering, Johns Hopkins University, Baltimore, Maryland 21218, United States
| | - Ishan Barman
- Department of Mechanical Engineering, Johns Hopkins University, Baltimore, Maryland 21218, United States
- The Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University, School of Medicine, Baltimore, Maryland 21205, United States
- The Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, Maryland 21287, United States
| |
Collapse
|
7
|
Liao GQ, Tang HM, Yu YD, Fu LZ, Li SJ, Zhu MX. Mass spectrometry-based metabolomic as a powerful tool to unravel the component and mechanism in TCM. Chin Med 2025; 20:62. [PMID: 40355943 PMCID: PMC12067679 DOI: 10.1186/s13020-025-01112-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2025] [Accepted: 04/21/2025] [Indexed: 05/15/2025] Open
Abstract
Mass spectrometry (MS)-based metabolomics has emerged as a transformative tool to unraveling components and their mechanisms in traditional Chinese medicine (TCM). The integration of advanced analytical platforms, such as LC-MS and GC-MS, coupled with metabolomics, has propelled the qualitative and quantitative characterization of TCM's complex components. This review comprehensively examines the applications of MS-based metabolomics in elucidating TCM efficacy, spanning chemical composition analysis, molecular target identification, mechanism-of-action studies, and syndrome differentiation. Recent innovations in functional metabolomics, spatial metabolomics, single-cell metabolomics, and metabolic flux analysis have further expanded TCM research horizons. Artificial intelligence (AI) and bioinformatics integration offer promising avenues for overcoming analytical bottlenecks, enhancing database standardization, and driving interdisciplinary breakthroughs. However, challenges remain, including the need for improved data processing standardization, database expansion, and understanding of metabolite-gene-protein interactions. By addressing these gaps, metabolomics can bridge traditional practices and modern biomedical research, fostering global acceptance of TCM. This review highlights the synergy of advanced MS techniques, computational tools, and TCM's holistic philosophy, presenting a forward-looking perspective on its clinical translation and internationalization.
Collapse
Affiliation(s)
- Guang-Qin Liao
- Chongqing Academy of Animal Sciences, Chongqing, 402460, China
- National Center of Technology Innovation for Pigs, Chongqing, 402460, China
| | - Hong-Mei Tang
- Chongqing Academy of Animal Sciences, Chongqing, 402460, China
- National Animal Disease-Chongqing Monitoring Station, Chongqing, 402460, China
| | - Yuan-Di Yu
- National Center of Technology Innovation for Pigs, Chongqing, 402460, China
- National Animal Disease-Chongqing Monitoring Station, Chongqing, 402460, China
| | - Li-Zhi Fu
- Chongqing Academy of Animal Sciences, Chongqing, 402460, China
- Chongqing Research Center of Veterinary Biologicals Engineering and Technology, Chongqing, 402460, China
| | - Shuang-Jiao Li
- Chinese Academy of Agricultural Sciences, Beijing, 100061, China
| | - Mai-Xun Zhu
- Chongqing Academy of Animal Sciences, Chongqing, 402460, China.
- National Center of Technology Innovation for Pigs, Chongqing, 402460, China.
| |
Collapse
|
8
|
Hu H, Fan Y, Wang J, Zhang J, Lyu Y, Hou X, Cui J, Zhang Y, Gao J, Zhang T, Nan K. Single-cell technology for cell-based drug delivery and pharmaceutical research. J Control Release 2025; 381:113587. [PMID: 40032008 DOI: 10.1016/j.jconrel.2025.113587] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Revised: 02/25/2025] [Accepted: 02/26/2025] [Indexed: 03/05/2025]
Abstract
Leveraging the capacity to precisely manipulate and analyze individual cells, single-cell technology has rapidly become an indispensable tool in the advancement of cell-based drug delivery systems and innovative cell therapies. This technology offers powerful means to address cellular heterogeneity and significantly enhance therapeutic efficacy. Recent breakthroughs in techniques such as single-cell electroporation, mechanical perforation, and encapsulation, particularly when integrated with microfluidics and bioelectronics, have led to remarkable improvements in drug delivery efficiency, reductions in cytotoxicity, and more precise targeting of therapeutic effects. Moreover, single-cell analyses, including advanced sequencing and high-resolution sensing, offer profound insights into complex disease mechanisms, the development of drug resistance, and the intricate processes of stem cell differentiation. This review summarizes the most significant applications of these single-cell technologies, highlighting their impact on the landscape of modern biomedicine. Furthermore, it provides a forward-looking perspective on future research directions aimed at further optimizing drug delivery strategies and enhancing therapeutic outcomes in the treatment of various diseases.
Collapse
Affiliation(s)
- Huihui Hu
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310000, China
| | - Yunlong Fan
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310000, China; MicroTech Medical (Hangzhou) Co., Hangzhou 311100, China
| | - Jiawen Wang
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310000, China
| | - Jialu Zhang
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310000, China
| | - Yidan Lyu
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310000, China
| | - Xiaoqi Hou
- School of Chemistry and Materials Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310024, China
| | - Jizhai Cui
- Department of Materials Science, Fudan University, Shanghai 200438, China; International Institute of Intelligent Nanorobots and Nanosystems, Fudan University, Shanghai 200438, China
| | - Yamin Zhang
- Department of Chemical and Biomolecular Engineering, National University of Singapore, 117585, Singapore
| | - Jianqing Gao
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310000, China
| | - Tianyuan Zhang
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310000, China.
| | - Kewang Nan
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310000, China.
| |
Collapse
|
9
|
Zeng J, Zhou H, Wan H, Yang J. Single-cell omics: moving towards a new era in ischemic stroke research. Eur J Pharmacol 2025; 1000:177725. [PMID: 40350018 DOI: 10.1016/j.ejphar.2025.177725] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Revised: 05/08/2025] [Accepted: 05/09/2025] [Indexed: 05/14/2025]
Abstract
Ischemic stroke (IS) is a highly complex and heterogeneous disease involving multiple pathophysiological events. A better understanding of the pathophysiology of IS will enhance preventive, diagnostic and therapeutic strategies. Despite significant advances in modern medicine, the molecular mechanisms of IS are still largely unknown. The high-throughput omics approach opens new avenues for identifying IS biomarkers and elucidating disease pathogenesis mechanisms. Single-cell omics enables a more thorough and in-depth analysis of the cellular interactions and properties in IS. This will lead to a better understanding of the onset, treatment and prognosis of IS. In this paper, we first reviewed the disease signatures and mechanisms research of IS. Subsequently, the use of single-cell omics to comprehend the mechanisms of IS was discussed, along with some recent developments in the field. To further delineate the upstream pathogenic alterations and downstream molecular impacts of IS, we also discussed the current use of machine learning approaches to single-cell omics data analysis. Particularly, single-cell omics is being used to inform risk assessment, early patient diagnosis and treatment strategies, and their potential impact on precision medicine. Thus, we summarized the role of single-cell omics in precision medicine. Despite the relative youth of the field, the development of single-cell omics promises to provide a powerful tool for elucidating the pathogenesis of IS.
Collapse
Affiliation(s)
- Jieqiong Zeng
- School of Basic Medicine Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China; School of Ecological and Environmental, Hubei Industrial Polytechnic, Shiyan, 442000, China
| | - Huifen Zhou
- School of Basic Medicine Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Haitong Wan
- School of Basic Medicine Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China.
| | - Jiehong Yang
- School of Basic Medicine Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China.
| |
Collapse
|
10
|
Nitz A, Giraldez Chavez JH, Eliason ZG, Payne SH. Are We There Yet? Assessing the Readiness of Single-Cell Proteomics to Answer Biological Hypotheses. J Proteome Res 2025; 24:1482-1492. [PMID: 38981598 PMCID: PMC11976870 DOI: 10.1021/acs.jproteome.4c00091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Revised: 05/02/2024] [Accepted: 06/13/2024] [Indexed: 07/11/2024]
Abstract
Single-cell analysis is an active area of research in many fields of biology. Measurements at single-cell resolution allow researchers to study diverse populations without losing biologically meaningful information to sample averages. Many technologies have been used to study single cells, including mass spectrometry-based single-cell proteomics (SCP). SCP has seen a lot of growth over the past couple of years through improvements in data acquisition and analysis, leading to greater proteomic depth. Because method development has been the main focus in SCP, biological applications have been sprinkled in only as proof-of-concept. However, SCP methods now provide significant coverage of the proteome and have been implemented in many laboratories. Thus, a primary question to address in our community is whether the current state of technology is ready for widespread adoption for biological inquiry. In this Perspective, we examine the potential for SCP in three thematic areas of biological investigation: cell annotation, developmental trajectories, and spatial mapping. We identify that the primary limitation of SCP is sample throughput. As proteome depth has been the primary target for method development to date, we advocate for a change in focus to facilitate measuring tens of thousands of single-cell proteomes to enable biological applications beyond proof-of-concept.
Collapse
Affiliation(s)
- Alyssa
A. Nitz
- Biology Department, Brigham Young University, Provo, Utah 84602, United States
| | | | - Zachary G. Eliason
- Biology Department, Brigham Young University, Provo, Utah 84602, United States
| | - Samuel H. Payne
- Biology Department, Brigham Young University, Provo, Utah 84602, United States
| |
Collapse
|
11
|
Kim J, Mun DG, Ding H, Forsberg EM, Meyer SW, Barsch A, Pandey A, Byeon SK. Single Cell Untargeted Lipidomics Using Liquid Chromatography Ion Mobility-Mass Spectrometry. J Proteome Res 2025; 24:1579-1585. [PMID: 39950635 DOI: 10.1021/acs.jproteome.4c00658] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/05/2025]
Abstract
Advancements in technology over the years have propelled omics analysis to the level of single cell resolution. Following the breakthroughs in single cell transcriptomics and genomics, single cell proteomics has recently rapidly progressed, aided by highly sensitive mass spectrometry instrumentation. However, there is currently a paucity of studies and methodologies for single cell lipidomics, aside from imaging-based approaches. Profiling lipids at the single cell level holds promise for providing novel insights into the complex heterogeneity of cells in various human disorders. Further, by integrating single cell lipidomics with other single cell omics including proteomics, it becomes possible to achieve single cell multiomics, enabling the discovery of novel molecular signatures. We developed untargeted single cell lipidomics using nanoflow liquid chromatography-ion mobility spectrometry-mass spectrometry. To enhance lipid coverage at the single cell level, the method was conducted in both positive and negative ion modes. We identified an average of 161 lipids spanning phospholipids, sphingolipids, cholesteryl esters, and glycerides in positive ion mode from single cells of human cholangiocarcinoma cells based on a rule-based lipid annotation. Additionally, an average of 20 species of phospholipids was identified in the negative ion mode. These preliminary data demonstrate a new methodology to profile lipids at a single or low input of cells.
Collapse
Affiliation(s)
- Jinyong Kim
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota 55905, United States
| | - Dong-Gi Mun
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota 55905, United States
| | - Husheng Ding
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota 55905, United States
| | | | - Sven W Meyer
- Bruker Daltonics GmbH & Co. KG, Bremen 28359, Germany
| | - Aiko Barsch
- Bruker Daltonics GmbH & Co. KG, Bremen 28359, Germany
| | - Akhilesh Pandey
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota 55905, United States
- Manipal Academy of Higher Education, Manipal, Karnataka 576104, India
- Center for Individualized Medicine, Mayo Clinic, Rochester, Minnesota 55905, United States
| | - Seul Kee Byeon
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota 55905, United States
| |
Collapse
|
12
|
Bozich ER, Guo X, Wilson JL, Hoffmann A. A computational workflow for assessing drug effects on temporal signaling dynamics reveals robustness in stimulus-specific NFκB signaling. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.31.645599. [PMID: 40236106 PMCID: PMC11996442 DOI: 10.1101/2025.03.31.645599] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/17/2025]
Abstract
Single-cell studies of signal transduction have revealed complex temporal dynamics that determine downstream biological function. For example, the stimulus-specific dynamics of the transcription factor NFκB specify stimulus-specific gene expression programs, and loss of specificity leads to disease. Thus, it is intriguing to consider drugs that may restore signaling specificity in disease contexts, or reduce activity but maintain signaling specificity to avoid unwanted side effects. However, while steady-state dose-response relationships have been the focus of pharmacological studies, there are no established methods for quantifying drug impact on stimulus-response signaling dynamics. Here we evaluated how drug treatments affect the stimulus-specificity of NFκB activation dynamics and its ability to accurately code ligand identity and dose. Specifically, we simulated the dynamic NFκB trajectories in response to 15 stimuli representing various immune threats under treatment of 10 representative drugs across 20 dosage levels. To quantify the effects on coding capacity, we introduced a Stimulus Response Specificity (SRS) score and a stimulus confusion score. We constructed stimulus confusion maps by employing epsilon network clustering in the trajectory space and in various dimensionally reduced spaces: canonical polyadic decomposition (CPD), functional principal component analysis (fPCA), and NFκB signaling codons (i.e. established, informative dynamic features). Our results indicated that the SRS score and the stimulus confusion map based on signaling codons are best-suited to quantify stimulus-specific NFκB dynamics confusion under pharmacological perturbations. Using these tools we found that temporal coding capacity of the NFκB signaling network is generally robust to a variety of pharmacological perturbations, thereby enabling the targeting of stimulus-specific dynamics without causing broad side-effects.
Collapse
|
13
|
Pires-Santos M, Carreira M, Morais BP, Perfeito FG, Oliveira MB, Monteiro CF, Nadine S, Mano JF. Single-Cell Liquid-Core Microcapsules for Biomedical Applications. Adv Healthc Mater 2025; 14:e2403808. [PMID: 39989098 DOI: 10.1002/adhm.202403808] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Revised: 01/29/2025] [Indexed: 02/25/2025]
Abstract
More recently, single-cell encapsulation emerged as a promising field in biomedicine due to its potential applications, in cell analysis and therapy. Traditional techniques involve embedding cells in crosslinked polymers to create continuous microgels, suitable mainly for adherent cells, or encapsulating them in droplets for only short-term analysis, due to their instability. In this study, we developed a method for encapsulating single cells in liquid-core microcapsules to address these limitations. The liquid encapsulation system is generated in an all aqueous environment through polymeric electrostatic interactions. Additionally, we design an innovative and low cost sorting system utilizing magnetic nanoparticles (MNPs) to efficiently select single-cell encapsulated units for further analysis and applications. This system is tested with both suspension and adherent cell types, demonstrating cytocompatibility and no abnormal effects on cell behavior. The MNP-based sorting achieved nearly 80% purity of the single-cell population. Overall, this technology provides a highly efficient method for single-cell applications, such as cell screening, by enabling precise short to medium-term analysis, real time monitoring, and high resolution imaging of cellular behavior. Furthermore, the semipermeable membrane unlocks new potential for advancing cell therapy by offering protection for encapsulated cells while ensuring the efficient diffusion of therapeutic factors, paving the way for innovative therapeutic strategies.
Collapse
Affiliation(s)
- Manuel Pires-Santos
- CICECO-Aveiro Institute of Materials, Department of Chemistry, Campus Universitário de Santiago, University of Aveiro, Aveiro, 3810-193, Portugal
| | - Mariana Carreira
- CICECO-Aveiro Institute of Materials, Department of Chemistry, Campus Universitário de Santiago, University of Aveiro, Aveiro, 3810-193, Portugal
| | - Bruno P Morais
- CICECO-Aveiro Institute of Materials, Department of Chemistry, Campus Universitário de Santiago, University of Aveiro, Aveiro, 3810-193, Portugal
| | - Francisca G Perfeito
- CICECO-Aveiro Institute of Materials, Department of Chemistry, Campus Universitário de Santiago, University of Aveiro, Aveiro, 3810-193, Portugal
| | - Mariana B Oliveira
- CICECO-Aveiro Institute of Materials, Department of Chemistry, Campus Universitário de Santiago, University of Aveiro, Aveiro, 3810-193, Portugal
| | - Cátia F Monteiro
- CICECO-Aveiro Institute of Materials, Department of Chemistry, Campus Universitário de Santiago, University of Aveiro, Aveiro, 3810-193, Portugal
| | - Sara Nadine
- CICECO-Aveiro Institute of Materials, Department of Chemistry, Campus Universitário de Santiago, University of Aveiro, Aveiro, 3810-193, Portugal
| | - João F Mano
- CICECO-Aveiro Institute of Materials, Department of Chemistry, Campus Universitário de Santiago, University of Aveiro, Aveiro, 3810-193, Portugal
| |
Collapse
|
14
|
Gatlin V, Gupta S, Romero S, Chapkin RS, Cai JJ. Exploring cell-to-cell variability and functional insights through differentially variable gene analysis. NPJ Syst Biol Appl 2025; 11:29. [PMID: 40113778 PMCID: PMC11926233 DOI: 10.1038/s41540-025-00507-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Accepted: 02/26/2025] [Indexed: 03/22/2025] Open
Abstract
Single-cell RNA sequencing (scRNA-seq) has revolutionized our understanding of cellular variability by capturing gene expression profiles of individual cells. The importance of cell-to-cell variability in determining and shaping cell function has been widely appreciated. Nevertheless, differential expression (DE) analysis remains a cornerstone method in analytical practice. Current computational analyses overlook the rich information encoded by variability within the single-cell gene expression data by focusing exclusively on mean expression. To offer a deeper understanding of cellular systems, there is a need for approaches to assess data variability rather than just the mean. Here we present spline-DV, a statistical framework for differential variability (DV) analysis using scRNA-seq data. The spline-DV method identifies genes exhibiting significantly increased or decreased expression variability among cells derived from two experimental conditions. Case studies show that DV genes identified using spline-DV are representative and functionally relevant to tested cellular conditions, including obesity, fibrosis, and cancer.
Collapse
Affiliation(s)
- Victoria Gatlin
- Department of Veterinary Integrative Biosciences, School of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX, 77843, USA
- CPRIT Single Cell Data Science Core, Texas A&M University, College Station, TX, 77843, USA
| | - Shreyan Gupta
- Department of Veterinary Integrative Biosciences, School of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX, 77843, USA
- CPRIT Single Cell Data Science Core, Texas A&M University, College Station, TX, 77843, USA
| | - Selim Romero
- Department of Veterinary Integrative Biosciences, School of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX, 77843, USA
- CPRIT Single Cell Data Science Core, Texas A&M University, College Station, TX, 77843, USA
- Department of Nutrition, Texas A&M University, College Station, TX, 77843, USA
| | - Robert S Chapkin
- CPRIT Single Cell Data Science Core, Texas A&M University, College Station, TX, 77843, USA
- Department of Nutrition, Texas A&M University, College Station, TX, 77843, USA
| | - James J Cai
- Department of Veterinary Integrative Biosciences, School of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX, 77843, USA.
- CPRIT Single Cell Data Science Core, Texas A&M University, College Station, TX, 77843, USA.
- Department of Electrical and Computer Engineering, Texas A&M University, College Station, TX, 77843, USA.
| |
Collapse
|
15
|
Jawwad T, Kamkaew M, Phongkitkarun K, Chusorn P, Jamnongsong S, Lam EWF, Sampattavanich S. Exploring the Single-Cell Dynamics of FOXM1 Under Cell Cycle Perturbations. Cell Prolif 2025:e70019. [PMID: 40091487 DOI: 10.1111/cpr.70019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Revised: 02/18/2025] [Accepted: 02/21/2025] [Indexed: 03/19/2025] Open
Abstract
The cell cycle is crucial for maintaining normal cellular functions and preventing replication errors. FOXM1, a key transcription factor, plays a pivotal role in regulating cell cycle progression and is implicated in various physiological and pathological processes, including cancers like liver, prostate, breast, lung and colon cancer. Despite previous research, our understanding of FOXM1 dynamics under different cell cycle perturbations and its connection to heterogeneous cell fate decisions remains limited. In this study, we investigated FOXM1 behaviour in individual cells exposed to various perturbagens. We found that different drugs induce diverse responses due to heterogeneous FOXM1 dynamics at the single-cell level. Single-cell analysis identified six distinct cellular phenotypes: on-time cytokinesis, cytokinesis delay, cell cycle delay, G1 arrest, G2 arrest and cell death, observed across different drug types and doses. Specifically, treatments with PLK1, CDK1, CDK1/2 and Aurora kinase inhibitors revealed varied FOXM1 dynamics leading to heterogeneous cellular outcomes. Our findings affirm that the dynamics of FOXM1 are essential in shaping cellular outcomes, influencing the signals that dictate responses to various stimuli. Our results gave insights into how FOXM1 dynamics contribute to cell cycle fate decisions, especially under different cell cycle perturbations.
Collapse
Affiliation(s)
- Tooba Jawwad
- Siriraj Center of Research Excellence for Systems Pharmacology, Department of Pharmacology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Maliwan Kamkaew
- Siriraj Center of Research Excellence for Systems Pharmacology, Department of Pharmacology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Kriengkrai Phongkitkarun
- Siriraj Center of Research Excellence for Systems Pharmacology, Department of Pharmacology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
- Department of Biomedical Engineering, Faculty of Engineering, Mahidol University, Nakhon Pathom, Thailand
| | - Porncheera Chusorn
- Siriraj Center of Research Excellence for Systems Pharmacology, Department of Pharmacology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
- Faculty of Liberal Arts and Science, Roi Et Rajabhat University, Roi Et, Thailand
| | - Supawan Jamnongsong
- Siriraj Center of Research Excellence for Systems Pharmacology, Department of Pharmacology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Eric W-F Lam
- Department of Surgery and Cancer, Imperial College London, London, UK
| | - Somponnat Sampattavanich
- Siriraj Center of Research Excellence for Systems Pharmacology, Department of Pharmacology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| |
Collapse
|
16
|
Hecker FA, Leggio B, Koenig T, Niehaus K, Geibel S. Cell Painting of insect gut cells for exploration of molecular responses of insect epithelia to insecticides. In Vitro Cell Dev Biol Anim 2025:10.1007/s11626-025-01028-z. [PMID: 40097748 DOI: 10.1007/s11626-025-01028-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Accepted: 02/16/2025] [Indexed: 03/19/2025]
Abstract
Cell Painting is a sophisticated high-content imaging technique that has been predominantly applied to mammalian cells. Recent advancements have extended its applicability to the first insect cell line, the ovarian cell line Sf9, revealing significant insights into similarities and differences in cellular responses between different taxonomic groups. This study explores the utility of Cell Painting in Helicoverpa zea gut-derived cells, specifically the RP-HzGUT-AW1 cell line, to assess the specifics of insect epithelial cells in response to chemical treatments. Upon adaptation of the analysis pipeline to accommodate their unique morphology and characteristics, our investigations revealed distinct responses of RP-HzGUT-AW1 cells compared to the ovarian insect cell line Sf9. Variations were obtained not only in the dose-response behavior to treatments but also in the overall detectability of specific modes of action. Specifically, processes that relate to osmoregulation and the formation of epithelial structures showed the most significant and distinct responses. This suggests that the specific morphological and physiological attributes of these gut-derived insect cells contribute to unique phenotypic profiles, which enables in-depth interpretation of drug efficacy and safety in these models.
Collapse
Affiliation(s)
| | - Bruno Leggio
- Bayer SAS, Crop Science Division, R&D Small Molecules, Disease Control, Lyon, France
| | - Tim Koenig
- Bayer AG, Pharma Division, R&D Chemical Biology, Imaging and Omics, Wuppertal, Germany
| | - Karsten Niehaus
- Bielefeld University, Proteome and Metabolome Research, Bielefeld, Germany
| | - Sven Geibel
- Bayer AG, Crop Science Division, R&D Small Molecules, Hit Key, Monheim, Germany.
| |
Collapse
|
17
|
Ge L, Jiang C, Ma C, Han CY, Gong Y, Zhu L, Liu Q, Liu FL. Ultrasensitive Determination of Amino Acids in Single Cells by Chemical Isotope Labeling with Liquid Chromatography Mass Spectrometry Analysis. Anal Chem 2025; 97:5171-5178. [PMID: 39999418 DOI: 10.1021/acs.analchem.4c06441] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/27/2025]
Abstract
Amino acids play multiple critical roles in the regulation of various metabolic pathways and physiological processes in living organisms. Mass spectrometry (MS) has become the most pioneering platform for amino acid analysis. However, the simultaneous and sensitive determination of amino acids is still challenging because of their structural similarity and broad ranges of concentrations. To this end, a pair of isotope labeling reagents, d0/d3-2-((diazomethyl)phenyl)(9-methyl-1,3,4,9-tetrahydro-2H-pyrido[3,4-b]indol-2-yl) methanone (DMPI/d3-DMPI), were applied to label amino acid metabolites. The diazo groups on the pair of isotopomers (DMPI/d3-DMPI) can specifically react with the carboxyl groups on the amino acids. The results showed that the retention on reversed-phase column were enhanced and the detection sensitivities of 19 amino acids were increased benefiting from DMPI labeling strategy that transfers the hydrophobic indole heterocycle group of DMPI to the hydrophilic compounds of amino acids. The obtained limits of detection (LODs) of amino acids were in the range of 0.002-0.082 fmol. With this established method, we achieved the sensitive detection of amino acids in a single HUVE cell. Meanwhile, we found that the contents of amino acids in the serum of premature neonates were higher compared to normal neonates. Overall, this developed method provides great support of detection tool for the clinical metabolomic study of amino acids and the investigation of dynamic changes of amino acid metabolism in single cells.
Collapse
Affiliation(s)
- Li Ge
- School of Pharmacy, Anhui Medical University, Hefei 230032, China
- Department of Pediatrics, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, China
| | - Chuanling Jiang
- Department of Pharmacy and Biomedical Engineering, Clinical College of Anhui Medical University, Hefei 230031, China
| | - Chengjie Ma
- Key Laboratory of Tobacco Chemistry, Zhengzhou Tobacco Research Institute of CNTC, Zhengzhou 450001, China
| | - Chun-Yue Han
- School of Pharmacy, Anhui Medical University, Hefei 230032, China
| | - Yi Gong
- School of Pharmacy, Anhui Medical University, Hefei 230032, China
| | - Lili Zhu
- School of Pharmacy, Anhui Medical University, Hefei 230032, China
| | - Qi Liu
- School of Pharmacy, Anhui Medical University, Hefei 230032, China
| | - Fei-Long Liu
- School of Pharmacy, Anhui Medical University, Hefei 230032, China
| |
Collapse
|
18
|
Anantha P, Raj P, Saracino E, Kim JH, Kim JH, Convertino A, Gu L, Barman I. Uncovering Astrocyte Morphological Dynamics Using Optical Diffraction Tomography and Shape-Based Trajectory Inference. Adv Healthc Mater 2025; 14:e2402960. [PMID: 39740118 DOI: 10.1002/adhm.202402960] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Revised: 12/19/2024] [Indexed: 01/02/2025]
Abstract
Astrocytes, integral components of the central nervous system, are increasingly recognized for their multifaceted roles beyond support cells. Despite their acknowledged importance, understanding the intricacies of astrocyte morphological dynamics remains limited. Our study marks the first exploration of astrocytes using optical diffraction tomography (ODT), establishing a label-free, quantitative method to observe morphological changes in astrocytes over a 7-day in-vitro period. ODT offers quantitative insights into cell volume, dry mass, and area through label-free, real-time measurements-capabilities that are challenging to achieve with conventional imaging techniques. Through comprehensive analysis of 3D refractive index maps and shape characterization techniques, we capture the developmental trajectory and dynamic morphological transformations of astrocytes. Specifically, our observations reveal increased area and a transition to larger, flattened shapes, with alterations in cell volume and density, indicating shifts in cellular composition. By employing unsupervised clustering and pseudotime trajectory analysis, we introduce a novel morphological trajectory inference for neural cells, tracking the morphological evolution of astrocytes from elongated to evenly spread shapes. This analysis marks the first use of trajectory inference based solely on morphology for neural cell types, laying a foundation for future studies employing ODT to examine astrocyte dynamics and neural cell interactions across diverse substrates.
Collapse
Affiliation(s)
- Pooja Anantha
- Department of Mechanical Engineering, Johns Hopkins University, Baltimore, MD, 21218, USA
| | - Piyush Raj
- Department of Mechanical Engineering, Johns Hopkins University, Baltimore, MD, 21218, USA
| | - Emanuela Saracino
- Institute for Organic Synthesis and Photoreactivity (ISOF), National Research Council of Italy (CNR), Via P. Gobetti 101, Bologna, I-40129, Italy
| | - Joo Ho Kim
- Department of Materials Science and Engineering, Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD, 21218, USA
| | - Jeong Hee Kim
- Department of Mechanical Engineering, Johns Hopkins University, Baltimore, MD, 21218, USA
| | - Annalisa Convertino
- Institute for Microelectronics and Microsystems, National Research Council, via Fosso del Cavaliere 100, Rome, 00133, Italy
| | - Luo Gu
- Department of Materials Science and Engineering, Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD, 21218, USA
| | - Ishan Barman
- Department of Mechanical Engineering, Johns Hopkins University, Baltimore, MD, 21218, USA
- Department of Oncology, Johns Hopkins University, Baltimore, MD, 21287, USA
- The Russell H. Morgan Department of Radiology and Radiological Science, Division of Cancer Imaging Research, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| |
Collapse
|
19
|
Driouchi A, Bretan M, Davis BJ, Heckert A, Seeger M, Silva MB, Forrest WSR, Hsiung J, Tan J, Yang H, McSwiggen DT, Song L, Sule A, Abaie B, Chen H, Chhun B, Conroy B, Elliott LA, Gonzalez E, Ilkov F, Isaacs J, Labaria G, Lagana M, Larsen DD, Margolin B, Nguyen MK, Park E, Rine J, Tang Y, Vana M, Wilkey A, Zhang Z, Basham S, Ho JJ, Johnson S, Klammer AA, Lin K, Darzacq X, Betzig E, Berman RT, Anderson DJ. Oblique line scan illumination enables expansive, accurate and sensitive single-protein measurements in solution and in living cells. Nat Methods 2025; 22:559-568. [PMID: 39966678 PMCID: PMC11903300 DOI: 10.1038/s41592-025-02594-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Accepted: 12/17/2024] [Indexed: 02/20/2025]
Abstract
An ideal tool for the study of cellular biology would enable the measure of molecular activity nondestructively within living cells. Single-molecule localization microscopy (SMLM) techniques, such as single-molecule tracking (SMT), enable in situ measurements in cells but have historically been limited by a necessary tradeoff between spatiotemporal resolution and throughput. Here we address these limitations using oblique line scan (OLS), a robust single-objective light-sheet-based illumination and detection modality that achieves nanoscale spatial resolution and sub-millisecond temporal resolution across a large field of view. We show that OLS can be used to capture protein motion up to 14 μm2 s-1 in living cells. We further extend the utility of OLS with in-solution SMT for single-molecule measurement of ligand-protein interactions and disruption of protein-protein interactions using purified proteins. We illustrate the versatility of OLS by showcasing two-color SMT, STORM and single-molecule fluorescence recovery after photobleaching. OLS paves the way for robust, high-throughput, single-molecule investigations of protein function required for basic research, drug screening and systems biology studies.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - Kevin Lin
- Eikon Therapeutics, Hayward, CA, USA
| | - Xavier Darzacq
- Department of Molecular & Cell Biology, University of California, Berkeley, Berkeley, CA, USA
| | - Eric Betzig
- Department of Molecular & Cell Biology, University of California, Berkeley, Berkeley, CA, USA
| | | | | |
Collapse
|
20
|
Moreno-Justicia R, Van der Stede T, Stocks B, Laitila J, Seaborne RA, Van de Loock A, Lievens E, Samodova D, Marín-Arraiza L, Dmytriyeva O, Browaeys R, Van Vossel K, Moesgaard L, Yigit N, Anckaert J, Weyns A, Van Thienen R, Sahl RE, Zanoteli E, Lawlor MW, Wierer M, Mestdagh P, Vandesompele J, Ochala J, Hostrup M, Derave W, Deshmukh AS. Human skeletal muscle fiber heterogeneity beyond myosin heavy chains. Nat Commun 2025; 16:1764. [PMID: 39971958 PMCID: PMC11839989 DOI: 10.1038/s41467-025-56896-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Accepted: 01/28/2025] [Indexed: 02/21/2025] Open
Abstract
Skeletal muscle is a heterogenous tissue comprised primarily of myofibers, commonly classified into three fiber types in humans: one "slow" (type 1) and two "fast" (type 2A and type 2X). However, heterogeneity between and within traditional fiber types remains underexplored. We applied transcriptomic and proteomic workflows to 1050 and 1038 single myofibers from human vastus lateralis, respectively. Proteomics was conducted in males, while transcriptomics included ten males and two females. We identify metabolic, ribosomal, and cell junction proteins, in addition to myosin heavy chain isoforms, as sources of multi-dimensional variation between myofibers. Furthermore, whilst slow and fast fiber clusters are identified, our data suggests that type 2X fibers are not phenotypically distinct to other fast fibers. Moreover, myosin heavy chain-based classifications do not adequately describe the phenotype of myofibers in nemaline myopathy. Overall, our data indicates that myofiber heterogeneity is multi-dimensional with sources of variation beyond myosin heavy chain isoforms.
Collapse
Affiliation(s)
- Roger Moreno-Justicia
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Thibaux Van der Stede
- Department of Movement and Sports Sciences, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
- The August Krogh Section for Human Physiology, Department of Nutrition, Exercise and Sports, University of Copenhagen, Copenhagen, Denmark
| | - Ben Stocks
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Molecular Medicine and Surgery, Integrative Physiology, Karolinska Institutet, Stockholm, Sweden
| | - Jenni Laitila
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Robert A Seaborne
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Centre for Human and Applied Physiological Sciences, King's College London, London, United Kingdom
| | - Alexia Van de Loock
- Department of Movement and Sports Sciences, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
| | - Eline Lievens
- Department of Movement and Sports Sciences, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
| | - Diana Samodova
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Leyre Marín-Arraiza
- Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Oksana Dmytriyeva
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Robin Browaeys
- Bioinformatics Expertise Unit, VIB Center for Inflammation Research, Ghent, Belgium
- Department of Applied Mathematics, Computer Science and Statistics, Ghent University, Ghent, Belgium
| | - Kim Van Vossel
- Department of Movement and Sports Sciences, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
| | - Lukas Moesgaard
- The August Krogh Section for Human Physiology, Department of Nutrition, Exercise and Sports, University of Copenhagen, Copenhagen, Denmark
| | - Nurten Yigit
- Department of Biomolecular Medicine, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
| | - Jasper Anckaert
- Department of Biomolecular Medicine, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
| | - Anneleen Weyns
- Department of Movement and Sports Sciences, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
| | - Ruud Van Thienen
- Department of Movement and Sports Sciences, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
| | - Ronni E Sahl
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Edmar Zanoteli
- Department of Neurology, Faculdade de Medicina (FMUSP), Universidade de São Paulo, São Paulo, Brazil
| | - Michael W Lawlor
- Department of Pathology, Medical College of Wisconsin, Milwaukee, WI, USA
- Diverge Translational Science Laboratory, Milwaukee, WI, USA
| | - Michael Wierer
- Proteomics Research Infrastructure, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Pieter Mestdagh
- Department of Biomolecular Medicine, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
| | - Jo Vandesompele
- Department of Biomolecular Medicine, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
| | - Julien Ochala
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Morten Hostrup
- The August Krogh Section for Human Physiology, Department of Nutrition, Exercise and Sports, University of Copenhagen, Copenhagen, Denmark
| | - Wim Derave
- Department of Movement and Sports Sciences, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium.
| | - Atul S Deshmukh
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
21
|
Ma L, Liu J, Sun W, Zhao C, Yu L. scMFG: a single-cell multi-omics integration method based on feature grouping. BMC Genomics 2025; 26:132. [PMID: 39934664 PMCID: PMC11817349 DOI: 10.1186/s12864-025-11319-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Accepted: 02/03/2025] [Indexed: 02/13/2025] Open
Abstract
BACKGROUND Recent advancements in methodologies and technologies have enabled the simultaneous measurement of multiple omics data, which provides a comprehensive understanding of cellular heterogeneity. However, existing methods have limitations in accurately identifying cell types while maintaining model interpretability, especially in the presence of noise. METHODS We propose a novel method called scMFG, which leverages feature grouping and group integration techniques for the integration of single-cell multi-omics data. By organizing features with similar characteristics within each omics layer through feature grouping. Furthermore, scMFG ensures a consistent feature grouping approach across different omics layers, promoting comparability of diverse data types. Additionally, scMFG incorporates a matrix factorization-based approach to enable the integrated results remain interpretable. RESULTS We comprehensively evaluated scMFG's performance on four complex real-world datasets generated using diverse sequencing technologies, highlighting its robustness in accurately identifying cell types. Notably, scMFG exhibited superior performance in deciphering cellular heterogeneity at a finer resolution compared to existing methods when applied to simulated datasets. Furthermore, our method proved highly effective in identifying rare cell types, showcasing its robust performance and suitability for detecting low-abundance cellular populations. The interpretability of scMFG was successfully validated through its specific association of outputs with specific cell types or states observed in the neonatal mouse cerebral cortices dataset. Moreover, we demonstrated that scMFG is capable of identifying cell developmental trajectories even in datasets with batch effects. CONCLUSIONS Our work presents a robust framework for the analysis of single-cell multi-omics data, advancing our understanding of cellular heterogeneity in a comprehensive and interpretable manner.
Collapse
Affiliation(s)
- Litian Ma
- School of Computer Science and Technology, Xidian University, Xi'an, Shaanxi, 710071, China
| | - Jingtao Liu
- School of Computer Science and Technology, Xidian University, Xi'an, Shaanxi, 710071, China
| | - Wei Sun
- Department of Rehabilitation Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Chenguang Zhao
- Department of Rehabilitation Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China.
| | - Liang Yu
- School of Computer Science and Technology, Xidian University, Xi'an, Shaanxi, 710071, China.
| |
Collapse
|
22
|
Tu JJ, Yan H, Zhang XF, Lin Z. Precise gene expression deconvolution in spatial transcriptomics with STged. Nucleic Acids Res 2025; 53:gkaf087. [PMID: 39970279 PMCID: PMC11838043 DOI: 10.1093/nar/gkaf087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Revised: 01/07/2025] [Accepted: 02/02/2025] [Indexed: 02/21/2025] Open
Abstract
Spatially resolved transcriptomics (SRT) has transformed tissue biology by linking gene expression profiles with spatial information. However, sequencing-based SRT methods aggregate signals from multiple cell types within capture locations ("spots"), masking cell-type-specific gene expression patterns. Traditional cell-type deconvolution methods estimate cell compositions within spots but fail to resolve cell-type-specific gene expression, limiting their ability to uncover critical biological processes such as cellular interactions and microenvironmental dynamics. Here, we present STged (spatial transcriptomic gene expression deconvolution), a novel computational framework that goes beyond traditional deconvolution by reconstructing cell-type-specific gene expression profiles from mixed spots. STged integrates graph-based spatial correlations and reference-derived gene signatures using a non-negative least-squares regression framework, achieving precise and biologically meaningful deconvolution. Comprehensive simulations show that STged consistently outperforms existing methods in accuracy and robustness. Applications to human pancreatic ductal adenocarcinoma and human squamous cell carcinoma datasets reveal its capacity to identify microenvironment-specific highly variable genes, reconstruct spatial cell-cell communication networks, and resolve tissue architecture at near-single-cell resolution. In mouse kidney tissues, STged uncovers dynamic spatial gene expression patterns and distinct gene programs, advancing our understanding of tissue heterogeneity and cellular dynamics.
Collapse
Affiliation(s)
- Jia-Juan Tu
- School of Science, Hubei University of Technology, Wuhan 430079, China
- Department of Statistics, The Chinese University of Hong Kong, Hong Kong 999077, China
| | - Hong Yan
- Centre for Intelligent Multidimensional Data Analysis, Hong Kong 999077, China
- Department of Electrical Engineering, City University of Hong Kong, Hong Kong, China
| | - Xiao-Fei Zhang
- School of Mathematics and Statistics, and Hubei Key Laboratory of Mathematical Sciences, Central China Normal University, Wuhan 430079, China
- Key Laboratory of Nonlinear Analysis & Applications (Ministry of Education), Central China Normal University, Wuhan 430079, China
| | - Zhixiang Lin
- Department of Statistics, The Chinese University of Hong Kong, Hong Kong 999077, China
| |
Collapse
|
23
|
Seal S, Trapotsi MA, Spjuth O, Singh S, Carreras-Puigvert J, Greene N, Bender A, Carpenter AE. Cell Painting: a decade of discovery and innovation in cellular imaging. Nat Methods 2025; 22:254-268. [PMID: 39639168 PMCID: PMC11810604 DOI: 10.1038/s41592-024-02528-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Accepted: 09/24/2024] [Indexed: 12/07/2024]
Abstract
Modern quantitative image analysis techniques have enabled high-throughput, high-content imaging experiments. Image-based profiling leverages the rich information in images to identify similarities or differences among biological samples, rather than measuring a few features, as in high-content screening. Here, we review a decade of advancements and applications of Cell Painting, a microscopy-based cell-labeling assay aiming to capture a cell's state, introduced in 2013 to optimize and standardize image-based profiling. Cell Painting's ability to capture cellular responses to various perturbations has expanded owing to improvements in the protocol, adaptations for different perturbations, and enhanced methodologies for feature extraction, quality control, and batch-effect correction. Cell Painting is a versatile tool that has been used in various applications, alone or with other -omics data, to decipher the mechanism of action of a compound, its toxicity profile, and other biological effects. Future advances will likely involve computational and experimental techniques, new publicly available datasets, and integration with other high-content data types.
Collapse
Affiliation(s)
- Srijit Seal
- Broad Institute of MIT and Harvard, Cambridge, MA, USA.
- Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge, UK.
| | - Maria-Anna Trapotsi
- Imaging and Data Analytics, Clinical Pharmacology & Safety Sciences, R&D, AstraZeneca, Cambridge, UK.
| | - Ola Spjuth
- Department of Pharmaceutical Biosciences and Science for Life Laboratory, Uppsala University, Uppsala, Sweden
- Phenaros Pharmaceuticals AB, Uppsala, Sweden
| | | | - Jordi Carreras-Puigvert
- Department of Pharmaceutical Biosciences and Science for Life Laboratory, Uppsala University, Uppsala, Sweden
- Phenaros Pharmaceuticals AB, Uppsala, Sweden
| | - Nigel Greene
- Imaging and Data Analytics, Clinical Pharmacology & Safety Sciences, R&D, AstraZeneca, Waltham, MA, USA
| | - Andreas Bender
- Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge, UK
- STAR-UBB Institute, Babeş-Bolyai University, Cluj-Napoca, Romania
| | | |
Collapse
|
24
|
Wang Y, Qu Y, Wang H, Xue Y, Liang P, Ge Y, Peng H, Wang Y, Song Z, Bao X, Xu J, Li B. Microwell-assembled aluminum substrates for enhanced single-cell analysis: A novel approach for cancer cell profiling by Raman spectroscopy. Talanta 2025; 283:127149. [PMID: 39515049 DOI: 10.1016/j.talanta.2024.127149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Revised: 10/30/2024] [Accepted: 11/01/2024] [Indexed: 11/16/2024]
Abstract
Single-cell analysis is critical for advancing personalized medicine, as it reveals cell population heterogeneity that influences disease outcomes. We present a microwell-assembled aluminum substrate platform that enhances single-cell Raman spectroscopy in liquid suspension by isolating individual cells and preventing stacking and movement, which significantly improves signal stability and the signal-to-noise ratio (SNR). We applied this novel platform to analyze PC-9 lung cancer cells and BEAS-2B normal bronchial epithelial cells, identifying distinct biochemical differences. Notably, cancer cells showed higher levels of adenine, cytochromes, DNA/RNA, and unsaturated lipids, along with an increased unsaturation ratio and protein content. These findings were further validated using machine learning models. An eXtreme Gradient Boosting (XGBoost) model achieved perfect classification accuracy of 100 %, underscoring the robustness of the spectral features identified by our platform. Our platform not only enhances single-cell Raman signal detection but also holds promise for biomedical applications, including early cancer detection, treatment monitoring, and drug development. The high-throughput capacity of this platform featuring over 120,000 wells, along with its compatibility with techniques such as Raman-activated cell sorting (RACS) further extends its potential for clinical diagnostics and personalized medicine.
Collapse
Affiliation(s)
- Yuntong Wang
- Changchun Institute of Optics, Fine Mechanics and Physics, Chinese Academy of Sciences, Changchun, 130033, PR China; University of Chinese Academy of Sciences, Beijing, 100049, PR China; State Key Laboratory of Applied Optics, Changchun, 130033, PR China; Key Laboratory of Advanced Manufacturing for Optical Systems, Chinese Academy of Sciences, Changchun, 130033, PR China
| | - Yue Qu
- Haining High-tech Research Institute, Jiaxing, Zhejiang, 314408, PR China
| | - Huan Wang
- Changchun Institute of Optics, Fine Mechanics and Physics, Chinese Academy of Sciences, Changchun, 130033, PR China; University of Chinese Academy of Sciences, Beijing, 100049, PR China
| | - Ying Xue
- Hooke Laboratory, Changchun, 130033, PR China
| | - Peng Liang
- Changchun Institute of Optics, Fine Mechanics and Physics, Chinese Academy of Sciences, Changchun, 130033, PR China; University of Chinese Academy of Sciences, Beijing, 100049, PR China
| | - Yan Ge
- Changchun Institute of Optics, Fine Mechanics and Physics, Chinese Academy of Sciences, Changchun, 130033, PR China; University of Chinese Academy of Sciences, Beijing, 100049, PR China; State Key Laboratory of Applied Optics, Changchun, 130033, PR China; Key Laboratory of Advanced Manufacturing for Optical Systems, Chinese Academy of Sciences, Changchun, 130033, PR China
| | - Hao Peng
- Changchun Institute of Optics, Fine Mechanics and Physics, Chinese Academy of Sciences, Changchun, 130033, PR China; University of Chinese Academy of Sciences, Beijing, 100049, PR China; State Key Laboratory of Applied Optics, Changchun, 130033, PR China; Key Laboratory of Advanced Manufacturing for Optical Systems, Chinese Academy of Sciences, Changchun, 130033, PR China
| | - Yu Wang
- Changchun Institute of Optics, Fine Mechanics and Physics, Chinese Academy of Sciences, Changchun, 130033, PR China; University of Chinese Academy of Sciences, Beijing, 100049, PR China; State Key Laboratory of Applied Optics, Changchun, 130033, PR China; Key Laboratory of Advanced Manufacturing for Optical Systems, Chinese Academy of Sciences, Changchun, 130033, PR China
| | - Zhixiong Song
- Haining High-tech Research Institute, Jiaxing, Zhejiang, 314408, PR China
| | - Xiaodong Bao
- Changchun Institute of Optics, Fine Mechanics and Physics, Chinese Academy of Sciences, Changchun, 130033, PR China; University of Chinese Academy of Sciences, Beijing, 100049, PR China; State Key Laboratory of Applied Optics, Changchun, 130033, PR China; Key Laboratory of Advanced Manufacturing for Optical Systems, Chinese Academy of Sciences, Changchun, 130033, PR China
| | - Jiabao Xu
- Division of Biomedical Engineering, James Watt School of Engineering, University of Glasgow, Glasgow, G12 8LT, UK.
| | - Bei Li
- Changchun Institute of Optics, Fine Mechanics and Physics, Chinese Academy of Sciences, Changchun, 130033, PR China; University of Chinese Academy of Sciences, Beijing, 100049, PR China; State Key Laboratory of Applied Optics, Changchun, 130033, PR China; Key Laboratory of Advanced Manufacturing for Optical Systems, Chinese Academy of Sciences, Changchun, 130033, PR China.
| |
Collapse
|
25
|
Golchin A, Shams F, Moradi F, Sadrabadi AE, Parviz S, Alipour S, Ranjbarvan P, Hemmati Y, Rahnama M, Rasmi Y, Aziz SGG. Single-cell Technology in Stem Cell Research. Curr Stem Cell Res Ther 2025; 20:9-32. [PMID: 38243989 DOI: 10.2174/011574888x265479231127065541] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Revised: 09/23/2023] [Accepted: 10/04/2023] [Indexed: 01/22/2024]
Abstract
Single-cell technology (SCT), which enables the examination of the fundamental units comprising biological organs, tissues, and cells, has emerged as a powerful tool, particularly in the field of biology, with a profound impact on stem cell research. This innovative technology opens new pathways for acquiring cell-specific data and gaining insights into the molecular pathways governing organ function and biology. SCT is not only frequently used to explore rare and diverse cell types, including stem cells, but it also unveils the intricacies of cellular diversity and dynamics. This perspective, crucial for advancing stem cell research, facilitates non-invasive analyses of molecular dynamics and cellular functions over time. Despite numerous investigations into potential stem cell therapies for genetic disorders, degenerative conditions, and severe injuries, the number of approved stem cell-based treatments remains limited. This limitation is attributed to the various heterogeneities present among stem cell sources, hindering their widespread clinical utilization. Furthermore, stem cell research is intimately connected with cutting-edge technologies, such as microfluidic organoids, CRISPR technology, and cell/tissue engineering. Each strategy developed to overcome the constraints of stem cell research has the potential to significantly impact advanced stem cell therapies. Drawing on the advantages and progress achieved through SCT-based approaches, this study aims to provide an overview of the advancements and concepts associated with the utilization of SCT in stem cell research and its related fields.
Collapse
Affiliation(s)
- Ali Golchin
- Cellular and Molecular Research Center, Cellular and Molecular Medicine Institute, Urmia University of Medical Sciences, Urmia, Iran
- Department of Clinical Biochemistry and Applied Cell Sciences, School of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | - Forough Shams
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Shahid, Beheshti University of Medical Sciences, Tehran, Iran
| | - Faezeh Moradi
- Department of Tissue Engineering, School of Medicine, Tarbiat Modares University, Tehran, Iran
| | - Amin Ebrahimi Sadrabadi
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR , Tehran, Iran
| | - Shima Parviz
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Medical Sciences and Technologies, Shiraz, University of Medical Sciences, Shiraz, Iran
| | - Shahriar Alipour
- Cellular and Molecular Research Center, Cellular and Molecular Medicine Institute, Urmia University of Medical Sciences, Urmia, Iran
- Department of Clinical Biochemistry and Applied Cell Sciences, School of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | - Parviz Ranjbarvan
- Cellular and Molecular Research Center, Cellular and Molecular Medicine Institute, Urmia University of Medical Sciences, Urmia, Iran
- Department of Clinical Biochemistry and Applied Cell Sciences, School of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | - Yaser Hemmati
- Department of Prosthodontics, Dental Faculty, Urmia University of Medical Science, Urmia, Iran
| | - Maryam Rahnama
- Department of Clinical Biochemistry and Applied Cell Sciences, School of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | - Yousef Rasmi
- Department of Clinical Biochemistry and Applied Cell Sciences, School of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | - Shiva Gholizadeh-Ghaleh Aziz
- Department of Clinical Biochemistry and Applied Cell Sciences, School of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| |
Collapse
|
26
|
Boché A, Landras A, Morel M, Kellouche S, Carreiras F, Lambert A. Phenomics Demonstrates Cytokines Additive Induction of Epithelial to Mesenchymal Transition. J Cell Physiol 2025; 240:e31491. [PMID: 39565461 PMCID: PMC11747948 DOI: 10.1002/jcp.31491] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 10/09/2024] [Accepted: 11/06/2024] [Indexed: 11/21/2024]
Abstract
Epithelial to mesenchymal transition (EMT) is highly plastic with a programme where cells lose adhesion and become more motile. EMT heterogeneity is one of the factors for disease progression and chemoresistance in cancer. Omics characterisations are costly and challenging to use. We developed single cell phenomics with easy to use wide-field fluorescence microscopy. We analyse over 70,000 cells and combined 53 features. Our simplistic pipeline allows efficient tracking of EMT plasticity, with a single statistical metric. We discriminate four high EMT plasticity cancer cell lines along the EMT spectrum. We test two cytokines, inducing EMT in all cell lines, alone or in combination. The single cell EMT metrics demonstrate the additive effect of cytokines combination on EMT independently of cell line EMT spectrum. The effects of cytokines are also observed at the front of migration during wound healing assay. Single cell phenomics is uniquely suited to characterise the cellular heterogeneity in response to complex microenvironment and show potential for drug testing assays.
Collapse
Affiliation(s)
- Alphonse Boché
- Equipe de Recherche sur les Relations Matrice Extracellulaire‐Cellules, ERRMECe, (EA1391), Groupe Matrice Extracellulaire et Physiopathologie (MECuP), Institut des Matériaux, I‐MAT (FD4122)CY Cergy Paris UniversitéNeuville sur OiseVal d'OiseFrance
| | - Alexandra Landras
- Equipe de Recherche sur les Relations Matrice Extracellulaire‐Cellules, ERRMECe, (EA1391), Groupe Matrice Extracellulaire et Physiopathologie (MECuP), Institut des Matériaux, I‐MAT (FD4122)CY Cergy Paris UniversitéNeuville sur OiseVal d'OiseFrance
| | - Mathieu Morel
- PASTEUR, Department of Chemistry, École Normale SupérieurePSL University, Sorbonne Université, CNRSParisFrance
| | - Sabrina Kellouche
- Equipe de Recherche sur les Relations Matrice Extracellulaire‐Cellules, ERRMECe, (EA1391), Groupe Matrice Extracellulaire et Physiopathologie (MECuP), Institut des Matériaux, I‐MAT (FD4122)CY Cergy Paris UniversitéNeuville sur OiseVal d'OiseFrance
| | - Franck Carreiras
- Equipe de Recherche sur les Relations Matrice Extracellulaire‐Cellules, ERRMECe, (EA1391), Groupe Matrice Extracellulaire et Physiopathologie (MECuP), Institut des Matériaux, I‐MAT (FD4122)CY Cergy Paris UniversitéNeuville sur OiseVal d'OiseFrance
| | - Ambroise Lambert
- Equipe de Recherche sur les Relations Matrice Extracellulaire‐Cellules, ERRMECe, (EA1391), Groupe Matrice Extracellulaire et Physiopathologie (MECuP), Institut des Matériaux, I‐MAT (FD4122)CY Cergy Paris UniversitéNeuville sur OiseVal d'OiseFrance
| |
Collapse
|
27
|
Wen O, Wolff SC, Stallaert W, Li D, Purvis JE, Zikry TM. Spherical Manifolds Capture Drug-Induced Changes in Tumor Cell Cycle Behavior. PACIFIC SYMPOSIUM ON BIOCOMPUTING. PACIFIC SYMPOSIUM ON BIOCOMPUTING 2025; 30:473-487. [PMID: 39670390 PMCID: PMC11687821 DOI: 10.1142/9789819807024_0034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 05/16/2025]
Abstract
CDK4/6 inhibitors such as palbociclib block cell cycle progression and improve outcomes for many ER+/HER2- breast cancer patients. Unfortunately, many patients are initially resistant to the drug or develop resistance over time in part due to heterogeneity among individual tumor cells. To better understand these mechanisms of resistance, we used multiplex, single-cell imaging to profile cell cycle proteins in ER+ breast tumor cells under increasing palbociclib concentrations. We then applied spherical principal component analysis (SPCA), a dimensionality reduction method that leverages the inherently cyclical nature of the high-dimensional imaging data, to look for changes in cell cycle behavior in resistant cells. SPCA characterizes data as a hypersphere and provides a framework for visualizing and quantifying differences in cell cycles across treatment-induced perturbations. The hypersphere representations revealed shifts in the mean cell state and population heterogeneity. SPCA validated expected trends of CDK4/6 inhibitor response such as decreased expression of proliferation markers (Ki67, pRB), but also revealed potential mechanisms of resistance including increased expression of cyclin D1 and CDK2. Understanding the molecular mechanisms that allow treated tumor cells to evade arrest is critical for identifying targets of future therapies. Ultimately, we seek to further SPCA as a tool of precision medicine, targeting treatments by individual tumors, and extending this computational framework to interpret other cyclical biological processes represented by high-dimensional data.
Collapse
Affiliation(s)
- Olivia Wen
- Department of Biology, University of North Carolina at Chapel Hill, NC, United States
| | - Samuel C Wolff
- Computational Medicine Program, University of North Carolina at Chapel Hill, NC, United States
| | - Wayne Stallaert
- Department of Computational and Systems Biology, University of Pittsburgh, PA, United States
| | - Didong Li
- Department of Biostatistics, University of North Carolina at Chapel Hill, NC, United States
| | - Jeremy E Purvis
- Computational Medicine Program, University of North Carolina at Chapel Hill, NC, United States,
| | - Tarek M Zikry
- Computational Medicine Program, University of North Carolina at Chapel Hill, NC, United States,
| |
Collapse
|
28
|
Wang R, Zhou L, Yang Y, Zhao F, Sun X, Liu X, Zou Z, Liang G. Spatially Quantitative Imaging of Enzyme Activity in a Living Cell. J Am Chem Soc 2024; 146:34870-34877. [PMID: 39655641 DOI: 10.1021/jacs.4c14190] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2024]
Abstract
Enzyme activity plays a key role in cell heterogeneity. Its spatially quantitative imaging in a living cell not only directly displays but also helps people to understand cell heterogeneity. Current methods are hard to achieve due to the short intracellular retention or lack of internal reference of the imaging probes. Herein, we rationally designed a self-referenced Raman probe Val-Cit-Cys(StBu)-Pra-Gly-CBT (Yne-CBT) which takes an intracellular cathepsin B (CTSB)-initiated CBT-Cys click reaction to yield a long-retained cyclic dimer in cell. In the meantime, Raman signal changes of its two chemical bonds (C≡C and C≡N) after the reaction are used for self-referencing and quantitative Raman imaging of CTSB activity. In vitro experiments demonstrated that, with shell-isolated nanoparticle-enhanced Raman spectroscopy technique, 20 μM Yne-CBT was able to quantitatively detect CTSB activity with a limit of detection of 61.4 U L-1. Under a homemade microfluidic channel, Yne-CBT was successfully applied for spatially quantitative imaging CTSB activity in a living cell. Our strategy provides people with a facile method to directly and quantitatively display cell heterogeneity.
Collapse
Affiliation(s)
- Rui Wang
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, Nanjing, Jiangsu 210096, China
| | - Lei Zhou
- School of Science, China Pharmaceutical University, Nanjing, Jiangsu 210009, China
| | - Yueyan Yang
- School of Science, China Pharmaceutical University, Nanjing, Jiangsu 210009, China
| | - Furong Zhao
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, Nanjing, Jiangsu 210096, China
| | - Xianbao Sun
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, Nanjing, Jiangsu 210096, China
| | - Xiaoyang Liu
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, Nanjing, Jiangsu 210096, China
| | - Zhen Zou
- Key Laboratory of Chemical Biology & Traditional Chinese Medicine Research, Ministry of Education, Institute of Interdisciplinary Studies, College of Chemistry and Chemical Engineering, Hunan Normal University, Changsha, Hunan 410081, China
| | - Gaolin Liang
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, Nanjing, Jiangsu 210096, China
| |
Collapse
|
29
|
Zhang J, Lin H, Xu J, Zhang M, Ge X, Zhang C, Huang WE, Cheng JX. High-throughput single-cell sorting by stimulated Raman-activated cell ejection. SCIENCE ADVANCES 2024; 10:eadn6373. [PMID: 39661682 PMCID: PMC11633747 DOI: 10.1126/sciadv.adn6373] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Accepted: 05/21/2024] [Indexed: 12/13/2024]
Abstract
Raman-activated cell sorting isolates single cells in a nondestructive and label-free manner, but its throughput is limited by small spontaneous Raman scattering cross section. Coherent Raman scattering integrated with microfluidics enables high-throughput cell analysis, but faces challenges with small cells (<3 μm) and tissue sections. Here, we report stimulated Raman-activated cell ejection (S-RACE) that enables high-throughput single-cell sorting by integrating stimulated Raman imaging, in situ image decomposition, and laser-induced cell ejection. S-RACE allows ejection of live bacteria or fungi guided by their Raman signatures. Furthermore, S-RACE successfully sorted lipid-rich Rhodotorula glutinis cells from a cell mixture with a throughput of ~13 cells per second, and the sorting results were confirmed by downstream quantitative polymerase chain reaction. Beyond single cells, S-RACE shows high compatibility with tissue sections. Incorporating a closed-loop feedback control circuit further enables real-time SRS imaging-identification-ejection. In summary, S-RACE opens exciting opportunities for diverse single-cell sorting applications.
Collapse
Affiliation(s)
- Jing Zhang
- Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA
- Photonics Center, Boston University, Boston, MA 02215, USA
| | - Haonan Lin
- Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA
- Photonics Center, Boston University, Boston, MA 02215, USA
| | - Jiabao Xu
- Division of Biomedical Engineering, James Watt School of Engineering, University of Glasgow, Glasgow G12 8LT, UK
| | - Meng Zhang
- Photonics Center, Boston University, Boston, MA 02215, USA
- Department of Electrical and Computer Engineering, Boston University, Boston, MA 02215, USA
| | - Xiaowei Ge
- Photonics Center, Boston University, Boston, MA 02215, USA
- Department of Electrical and Computer Engineering, Boston University, Boston, MA 02215, USA
| | - Chi Zhang
- Department of Chemistry, Purdue University, 560 Oval Dr., West Lafayette, IN 47907, USA
| | - Wei E. Huang
- Department of Engineering Science, University of Oxford, Oxford OX1 3PJ, UK
| | - Ji-Xin Cheng
- Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA
- Photonics Center, Boston University, Boston, MA 02215, USA
- Department of Electrical and Computer Engineering, Boston University, Boston, MA 02215, USA
| |
Collapse
|
30
|
Yan Y, Ding L, Ding J, Zhou P, Su B. Recent Advances in Electrochemiluminescence Visual Biosensing and Bioimaging. Chembiochem 2024; 25:e202400389. [PMID: 38899794 DOI: 10.1002/cbic.202400389] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 06/18/2024] [Accepted: 06/20/2024] [Indexed: 06/21/2024]
Abstract
Electrochemiluminescence (ECL) is one of the most powerful techniques that meet the needs of analysis and detection in a variety of scenarios, because of its highly analytical sensitivity and excellent spatiotemporal controllability. ECL combined with microscopy (ECLM) offers a promising approach for quantifying and mapping a wide range of analytes. To date, ECLM has been widely used to image biological entities and processes, such as cells, subcellular structures, proteins and membrane transport properties. In this review, we first introduced the mechanisms of several classic ECL systems, then highlighted the progress of visual biosensing and bioimaging by ECLM in the last decade. Finally, the characteristics of ECLM were summarized, as well as some of the current challenges. The future research interests and potential directions for the application of ECLM were also outlooked.
Collapse
Affiliation(s)
- Yajuan Yan
- Key Laboratory of Excited-State Materials of Zhejiang Province, Department of Chemistry, Zhejiang University, Hangzhou, 310058, China
| | - Lurong Ding
- Key Laboratory of Excited-State Materials of Zhejiang Province, Department of Chemistry, Zhejiang University, Hangzhou, 310058, China
| | - Jialian Ding
- Key Laboratory of Excited-State Materials of Zhejiang Province, Department of Chemistry, Zhejiang University, Hangzhou, 310058, China
| | - Ping Zhou
- Key Laboratory of Excited-State Materials of Zhejiang Province, Department of Chemistry, Zhejiang University, Hangzhou, 310058, China
| | - Bin Su
- Key Laboratory of Excited-State Materials of Zhejiang Province, Department of Chemistry, Zhejiang University, Hangzhou, 310058, China
| |
Collapse
|
31
|
Ghanegolmohammadi F, Eslami M, Ohya Y. Systematic data analysis pipeline for quantitative morphological cell phenotyping. Comput Struct Biotechnol J 2024; 23:2949-2962. [PMID: 39104709 PMCID: PMC11298594 DOI: 10.1016/j.csbj.2024.07.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Revised: 07/09/2024] [Accepted: 07/10/2024] [Indexed: 08/07/2024] Open
Abstract
Quantitative morphological phenotyping (QMP) is an image-based method used to capture morphological features at both the cellular and population level. Its interdisciplinary nature, spanning from data collection to result analysis and interpretation, can lead to uncertainties, particularly among those new to this actively growing field. High analytical specificity for a typical QMP is achieved through sophisticated approaches that can leverage subtle cellular morphological changes. Here, we outline a systematic workflow to refine the QMP methodology. For a practical review, we describe the main steps of a typical QMP; in each step, we discuss the available methods, their applications, advantages, and disadvantages, along with the R functions and packages for easy implementation. This review does not cover theoretical backgrounds, but provides several references for interested researchers. It aims to broaden the horizons for future phenome studies and demonstrate how to exploit years of endeavors to achieve more with less.
Collapse
Affiliation(s)
- Farzan Ghanegolmohammadi
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
- Department of Integrated Biosciences, Graduate School of Frontier Sciences, The University of Tokyo, Kashiwa, Chiba, Japan
| | - Mohammad Eslami
- Harvard Ophthalmology AI Lab, Schepen’s Eye Research Institute of Massachusetts Eye and Ear Infirmary, Harvard Medical School, Boston, USA
| | - Yoshikazu Ohya
- Department of Integrated Biosciences, Graduate School of Frontier Sciences, The University of Tokyo, Kashiwa, Chiba, Japan
| |
Collapse
|
32
|
Li G, Ma Y, Zhang S, Lin W, Yao X, Zhou Y, Zhao Y, Rao Q, Qu Y, Gao Y, Chen L, Zhang Y, Han F, Sun M, Zhao C. A mechanistic systems biology model of brain microvascular endothelial cell signaling reveals dynamic pathway-based therapeutic targets for brain ischemia. Redox Biol 2024; 78:103415. [PMID: 39520909 PMCID: PMC11584692 DOI: 10.1016/j.redox.2024.103415] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Revised: 10/31/2024] [Accepted: 10/31/2024] [Indexed: 11/16/2024] Open
Abstract
Ischemic stroke is a significant threat to human health. Currently, there is a lack of effective treatments for stroke, and progress in new neuron-centered drug target development is relatively slow. On the other hand, studies have demonstrated that brain microvascular endothelial cells (BMECs) are crucial components of the neurovascular unit and play pivotal roles in ischemic stroke progression. To better understand the complex multifaceted roles of BMECs in the regulation of ischemic stroke pathophysiology and facilitate BMEC-based drug target discovery, we utilized a transcriptomics-informed systems biology modeling approach and constructed a mechanism-based computational multipathway model to systematically investigate BMEC function and its modulatory potential. Extensive multilevel data regarding complex BMEC pathway signal transduction and biomarker expression under various pathophysiological conditions were used for quantitative model calibration and validation, and we generated dynamic BMEC phenotype maps in response to various stroke-related stimuli to identify potential determinants of BMEC fate under stress conditions. Through high-throughput model sensitivity analyses and virtual target perturbations in model-based single cells, our model predicted that targeting succinate could effectively reverse the detrimental cell phenotype of BMECs under oxygen and glucose deprivation/reoxygenation, a condition that mimics stroke pathogenesis, and we experimentally validated the utility of this new target in terms of regulating inflammatory factor production, free radical generation and tight junction protection in vitro and in vivo. Our work is the first that complementarily couples transcriptomic analysis with mechanistic systems-level pathway modeling in the study of BMEC function and endothelium-based therapeutic targets in ischemic stroke.
Collapse
Affiliation(s)
- Geli Li
- School of Pharmacy, Nanjing Medical University, 210000, Nanjing, China; Gusu School, Nanjing Medical University, 215000, Suzhou, China
| | - Yuchen Ma
- School of Pharmacy, Nanjing Medical University, 210000, Nanjing, China
| | - Sujie Zhang
- School of Pharmacy, Nanjing Medical University, 210000, Nanjing, China
| | - Wen Lin
- School of Pharmacy, Nanjing Medical University, 210000, Nanjing, China
| | - Xinyi Yao
- School of Pharmacy, Nanjing Medical University, 210000, Nanjing, China
| | - Yating Zhou
- The First Affiliated Hospital of Nanjing Medical University, 210000, Nanjing, China
| | - Yanyong Zhao
- School of Pharmacy, Nanjing Medical University, 210000, Nanjing, China
| | - Qi Rao
- School of Pharmacy, Nanjing Medical University, 210000, Nanjing, China
| | - Yuchen Qu
- School of Pharmacy, Nanjing Medical University, 210000, Nanjing, China
| | - Yuan Gao
- QSPMed Technologies, 210000, Nanjing, China
| | - Lianmin Chen
- The First Affiliated Hospital of Nanjing Medical University, 210000, Nanjing, China
| | - Yu Zhang
- Department of Biomedical Engineering, School of Medicine, Johns Hopkins University, 21205, Baltimore, USA
| | - Feng Han
- Key Laboratory of Cardiovascular & Cerebrovascular Medicine, Drug Target and Drug Discovery Center, School of Pharmacy, Nanjing Medical University, 210000, Nanjing, China.
| | - Meiling Sun
- School of Basic Medical Sciences, Nanjing Medical University, 210000, Nanjing, China.
| | - Chen Zhao
- School of Pharmacy, Nanjing Medical University, 210000, Nanjing, China; The First Affiliated Hospital of Nanjing Medical University, 210000, Nanjing, China.
| |
Collapse
|
33
|
Lyu J, Chen C. Transcriptome and Temporal Transcriptome Analyses in Single Cells. Int J Mol Sci 2024; 25:12845. [PMID: 39684556 DOI: 10.3390/ijms252312845] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Revised: 11/21/2024] [Accepted: 11/26/2024] [Indexed: 12/18/2024] Open
Abstract
Transcriptome analysis in single cells, enabled by single-cell RNA sequencing, has become a prevalent approach in biomedical research, ranging from investigations of gene regulation to the characterization of tissue organization. Over the past decade, advances in single-cell RNA sequencing technology, including its underlying chemistry, have significantly enhanced its performance, marking notable improvements in methodology. A recent development in the field, which integrates RNA metabolic labeling with single-cell RNA sequencing, has enabled the profiling of temporal transcriptomes in individual cells, offering new insights into dynamic biological processes involving RNA kinetics and cell fate determination. In this review, we explore the chemical principles and design improvements that have enhanced single-molecule capture efficiency, improved RNA quantification accuracy, and increased cellular throughput in single-cell transcriptome analysis. We also illustrate the concept of RNA metabolic labeling for detecting newly synthesized transcripts and summarize recent advancements that enable single-cell temporal transcriptome analysis. Additionally, we examine data analysis strategies for the precise quantification of newly synthesized transcripts and highlight key applications of transcriptome and temporal transcriptome analyses in single cells.
Collapse
Affiliation(s)
- Jun Lyu
- Laboratory of Biochemistry and Molecular Biology, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Chongyi Chen
- Laboratory of Biochemistry and Molecular Biology, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
34
|
Beres B, Kovacs KD, Kanyo N, Peter B, Szekacs I, Horvath R. Label-Free Single-Cell Cancer Classification from the Spatial Distribution of Adhesion Contact Kinetics. ACS Sens 2024; 9:5815-5827. [PMID: 39082162 PMCID: PMC11590093 DOI: 10.1021/acssensors.4c01139] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Revised: 07/20/2024] [Accepted: 07/23/2024] [Indexed: 08/03/2024]
Abstract
There is an increasing need for simple-to-use, noninvasive, and rapid tools to identify and separate various cell types or subtypes at the single-cell level with sufficient throughput. Often, the selection of cells based on their direct biological activity would be advantageous. These steps are critical in immune therapy, regenerative medicine, cancer diagnostics, and effective treatment. Today, live cell selection procedures incorporate some kind of biomolecular labeling or other invasive measures, which may impact cellular functionality or cause damage to the cells. In this study, we first introduce a highly accurate single-cell segmentation methodology by combining the high spatial resolution of a phase-contrast microscope with the adhesion kinetic recording capability of a resonant waveguide grating (RWG) biosensor. We present a classification workflow that incorporates the semiautomatic separation and classification of single cells from the measurement data captured by an RWG-based biosensor for adhesion kinetics data and a phase-contrast microscope for highly accurate spatial resolution. The methodology was tested with one healthy and six cancer cell types recorded with two functionalized coatings. The data set contains over 5000 single-cell samples for each surface and over 12,000 samples in total. We compare and evaluate the classification using these two types of surfaces (fibronectin and noncoated) with different segmentation strategies and measurement timespans applied to our classifiers. The overall classification performance reached nearly 95% with the best models showing that our proof-of-concept methodology could be adapted for real-life automatic diagnostics use cases. The label-free measurement technique has no impact on cellular functionality, directly measures cellular activity, and can be easily tuned to a specific application by varying the sensor coating. These features make it suitable for applications requiring further processing of selected cells.
Collapse
Affiliation(s)
- Balint Beres
- Nanobiosensorics
Laboratory, Institute of Technical Physics and Materials Science, HUN-REN Centre for Energy Research, Konkoly-Thege út 29-33, Budapest H-1121, Hungary
- Department
of Automation and Applied Informatics, Faculty of Electrical Engineering
and Informatics, Budapest University of
Technology and Economics, Műegyetem rkp. 3, Budapest H-1111, Hungary
| | - Kinga Dora Kovacs
- Nanobiosensorics
Laboratory, Institute of Technical Physics and Materials Science, HUN-REN Centre for Energy Research, Konkoly-Thege út 29-33, Budapest H-1121, Hungary
- Department
of Biological Physics, Eötvös
University, Pázmány Péter stny. 1/A, Budapest H-1117, Hungary
| | - Nicolett Kanyo
- Nanobiosensorics
Laboratory, Institute of Technical Physics and Materials Science, HUN-REN Centre for Energy Research, Konkoly-Thege út 29-33, Budapest H-1121, Hungary
| | - Beatrix Peter
- Nanobiosensorics
Laboratory, Institute of Technical Physics and Materials Science, HUN-REN Centre for Energy Research, Konkoly-Thege út 29-33, Budapest H-1121, Hungary
| | - Inna Szekacs
- Nanobiosensorics
Laboratory, Institute of Technical Physics and Materials Science, HUN-REN Centre for Energy Research, Konkoly-Thege út 29-33, Budapest H-1121, Hungary
| | - Robert Horvath
- Nanobiosensorics
Laboratory, Institute of Technical Physics and Materials Science, HUN-REN Centre for Energy Research, Konkoly-Thege út 29-33, Budapest H-1121, Hungary
| |
Collapse
|
35
|
Yang D, Fang Y, Liu X, Ma J, Xu J, Dong H, Ding H, Wang D, Liu Q, Zhang F. Lensless On-Chip Chemiluminescence Imaging for High-Throughput Single-Cell Heterogeneity Analysis. NANO LETTERS 2024; 24:14875-14883. [PMID: 39512117 DOI: 10.1021/acs.nanolett.4c04487] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2024]
Abstract
High-throughput single-cell heterogeneity imaging and analysis is essential for understanding complex biological systems and for advancing personalized precision disease diagnosis and treatment. Here, we present a miniaturized lensless chemiluminescence chip for high-throughput single-cell functional imaging with subcellular resolution. With the sensitive chemiluminescence sensing and wide field of view of contact lensless imaging, we demonstrated the chemiluminescent imaging of over 1000 single cells, and their membrane glycoprotein and the high-throughput single-cell heterogeneity of membrane protein imaging were examined for precision analysis. Furthermore, the functional adhesion and heterogeneity of single live cells were imaged and explored. This miniaturized lensless on-chip CL-CMOS imaging platform enables high-throughput single-cell imaging and analysis with high sensitivity and subcellular resolution, providing new techniques for the cellular study of biological heterogeneity and has potential application in precision disease diagnosis and treatment at the point-of-care settings.
Collapse
Affiliation(s)
- Dehong Yang
- Biosensor National Special Laboratory, Department of Biomedical Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Ying Fang
- Biosensor National Special Laboratory, Department of Biomedical Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Xiaoyin Liu
- Biosensor National Special Laboratory, Department of Biomedical Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Jinbiao Ma
- Biosensor National Special Laboratory, Department of Biomedical Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Jiahao Xu
- Biosensor National Special Laboratory, Department of Biomedical Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Hao Dong
- Intelligent Perception Research Institute, Zhejiang Lab, Hangzhou, 311100, China
| | - Haiying Ding
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
- Department of Pharmacy, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, 310005, China
| | - Di Wang
- Biosensor National Special Laboratory, Department of Biomedical Engineering, Zhejiang University, Hangzhou, 310027, China
- Intelligent Perception Research Institute, Zhejiang Lab, Hangzhou, 311100, China
| | - Qingjun Liu
- Biosensor National Special Laboratory, Department of Biomedical Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Fenni Zhang
- Biosensor National Special Laboratory, Department of Biomedical Engineering, Zhejiang University, Hangzhou, 310027, China
| |
Collapse
|
36
|
Li R, Chen Y, Pan R, Hu S, Zhao S, Tian J, Zhao J. Single-Cell Multiplexed Signal Amplification Strategy Based on Catalytic Hairpin Self-Assembly and CRISPR/Cas12a for Exploring the Relationship between lncRNA HOTAIR and miRNA-122 in Individual Hepatocytes. Anal Chem 2024; 96:18096-18103. [PMID: 39473038 DOI: 10.1021/acs.analchem.4c03974] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2024]
Abstract
The long noncoding RNA (lncRNA) HOTAIR has been shown to act as an oncogene in a variety of cancers, including hepatocellular carcinoma (HCC). MicroRNA-122 (miR-122) is a key liver-specific miRNA that is frequently inhibited in HCC and is associated with poor prognosis. However, a potential relationship between HOTAIR and miR-122 in individual hepatocytes has not been explored. To this end, we propose here an intracellular catalytic hairpin self-assembly-CRISPR/Cas12a tandem multiplexed signal amplification strategy for the simultaneous quantification of HOTAIR and miRNA-122 in a single hepatocyte. We applied this method to analyze both normal HL-7702 liver cells and HepG2 HCC cells, and found that HL-7702 cells contained large amounts of miRNA-122, while the content of miRNA-122 in HepG2 cells was low. However, the level of HOTAIR in HepG2 cells was much higher than that in HL-7702 cells, confirming the overexpression of HOTAIR in HCC cells. We achieved the simultaneous absolute quantification of HOTAIR and miRNA-122 in single cells, providing an important method to study the relationships between these two RNA molecules in individual cells.
Collapse
Affiliation(s)
- Ruiyan Li
- State Key Laboratory for the Chemistry and Molecular Engineering of Medicinal Resources, Key Laboratory of Ecology of Rare and Endangered Species and Environmental Protection, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin 541004, China
| | - Yuhai Chen
- State Key Laboratory for the Chemistry and Molecular Engineering of Medicinal Resources, Key Laboratory of Ecology of Rare and Endangered Species and Environmental Protection, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin 541004, China
| | - Rongxiang Pan
- State Key Laboratory for the Chemistry and Molecular Engineering of Medicinal Resources, Key Laboratory of Ecology of Rare and Endangered Species and Environmental Protection, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin 541004, China
| | - Shengqiang Hu
- State Key Laboratory for the Chemistry and Molecular Engineering of Medicinal Resources, Key Laboratory of Ecology of Rare and Endangered Species and Environmental Protection, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin 541004, China
| | - Shulin Zhao
- State Key Laboratory for the Chemistry and Molecular Engineering of Medicinal Resources, Key Laboratory of Ecology of Rare and Endangered Species and Environmental Protection, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin 541004, China
| | - Jianniao Tian
- State Key Laboratory for the Chemistry and Molecular Engineering of Medicinal Resources, Key Laboratory of Ecology of Rare and Endangered Species and Environmental Protection, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin 541004, China
| | - Jingjin Zhao
- State Key Laboratory for the Chemistry and Molecular Engineering of Medicinal Resources, Key Laboratory of Ecology of Rare and Endangered Species and Environmental Protection, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin 541004, China
| |
Collapse
|
37
|
Zhao YN, Zhang X, Bai JJ, Jia HY, Chen ML, Wang JH. Inertial and Deterministic Lateral Displacement Integrated Microfluidic Chips for Epithelial-Mesenchymal Transition Analysis. Anal Chem 2024; 96:18187-18194. [PMID: 39484816 DOI: 10.1021/acs.analchem.4c04366] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2024]
Abstract
With the aim of efficiently sorting rare circulating tumor cells (CTCs) from blood and minimizing damage to CTCs during isolation, we constructed an inertia-assisted single-cell focusing generator (I-SCF) and a water droplet deterministic lateral displacement cell sorting (D-DLD) microfluidic system (IDIC) based on different sizes, the device is initially sorted by a continuous fluid swing and Dean flow-assisted helical micromixers, then flows through a droplet shaped DLD region, enabling single-cell focused sequencing and precise separation, improving cell separation efficiency (>95%) and purity, while ensuring a high single cells survival rate of more than 98.6%. Subsequently, breast cancer cell lines were run through our chip, and then the downstream epithelial-mesenchymal transition (EMT) process induced by TGF-β was detected, and the levels of three proteins, EpCAM, PD-L1, and N-cadherin, were analyzed to establish the relationship between PD-L1 and the EMT process. Compared with other analytical techniques such as the filtration method, the enrichment method and immunoaffinity capture methods, this method not only ensures the separation efficiency and purity, but also ensures the cell activity, and avoids missing the different results caused by the heterogeneity of CTCs due to the isolation of high purity (84.01%). The device has a high throughput processing capacity (5 mL of diluted whole blood/∼2.8 h). By using the chip, we can more easily and conveniently predict tumor stage and carry out cancer prevention and treatment in advance, and it is expected to be further developed into a clinical liquid biopsy technology in the future.
Collapse
Affiliation(s)
- Ya-Nan Zhao
- Research Center for Analytical Sciences, Department of Chemistry, College of Sciences, Northeastern University, Shenyang 110819, China
| | - Xuan Zhang
- Research Center for Analytical Sciences, Department of Chemistry, College of Sciences, Northeastern University, Shenyang 110819, China
| | - Jun-Jie Bai
- Research Center for Analytical Sciences, Department of Chemistry, College of Sciences, Northeastern University, Shenyang 110819, China
| | - Hao-Yu Jia
- Research Center for Analytical Sciences, Department of Chemistry, College of Sciences, Northeastern University, Shenyang 110819, China
| | - Ming-Li Chen
- Research Center for Analytical Sciences, Department of Chemistry, College of Sciences, Northeastern University, Shenyang 110819, China
| | - Jian-Hua Wang
- Research Center for Analytical Sciences, Department of Chemistry, College of Sciences, Northeastern University, Shenyang 110819, China
| |
Collapse
|
38
|
van Dijk R, Arevalo J, Babadi M, Carpenter AE, Singh S. Capturing cell heterogeneity in representations of cell populations for image-based profiling using contrastive learning. PLoS Comput Biol 2024; 20:e1012547. [PMID: 39527652 DOI: 10.1371/journal.pcbi.1012547] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2024] [Accepted: 10/10/2024] [Indexed: 11/16/2024] Open
Abstract
Image-based cell profiling is a powerful tool that compares perturbed cell populations by measuring thousands of single-cell features and summarizing them into profiles. Typically a sample is represented by averaging across cells, but this fails to capture the heterogeneity within cell populations. We introduce CytoSummaryNet: a Deep Sets-based approach that improves mechanism of action prediction by 30-68% in mean average precision compared to average profiling on a public dataset. CytoSummaryNet uses self-supervised contrastive learning in a multiple-instance learning framework, providing an easier-to-apply method for aggregating single-cell feature data than previously published strategies. Interpretability analysis suggests that the model achieves this improvement by downweighting small mitotic cells or those with debris and prioritizing large uncrowded cells. The approach requires only perturbation labels for training, which are readily available in all cell profiling datasets. CytoSummaryNet offers a straightforward post-processing step for single-cell profiles that can significantly boost retrieval performance on image-based profiling datasets.
Collapse
Affiliation(s)
| | - John Arevalo
- Imaging Platform, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, United States of America
| | - Mehrtash Babadi
- Data Sciences Platform, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, United States of America
| | - Anne E Carpenter
- Imaging Platform, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, United States of America
| | - Shantanu Singh
- Imaging Platform, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, United States of America
| |
Collapse
|
39
|
Huang Y, Zhou Z, Liu T, Tang S, Xin X. Exploring heterogeneous cell population dynamics in different microenvironments by novel analytical strategy based on images. NPJ Syst Biol Appl 2024; 10:129. [PMID: 39505883 PMCID: PMC11542073 DOI: 10.1038/s41540-024-00459-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Accepted: 10/21/2024] [Indexed: 11/08/2024] Open
Abstract
Understanding the dynamic states and transitions of heterogeneous cell populations is crucial for addressing fundamental biological questions. High-content imaging provides rich datasets, but it remains increasingly difficult to integrate and annotate high-dimensional and time-resolved datasets to profile heterogeneous cell population dynamics in different microenvironments. Using hepatic stellate cells (HSCs) LX-2 as model, we proposed a novel analytical strategy for image-based integration and annotation to profile dynamics of heterogeneous cell populations in 2D/3D microenvironments. High-dimensional features were extracted from extensive image datasets, and cellular states were identified based on feature profiles. Time-series clustering revealed distinct temporal patterns of cell shape and actin cytoskeleton reorganization. We found LX-2 showed more complex membrane dynamics and contractile systems with an M-shaped actin compactness trend in 3D culture, while they displayed rapid spreading in early 2D culture. This image-based integration and annotation strategy enhances our understanding of HSCs heterogeneity and dynamics in complex extracellular microenvironments.
Collapse
Affiliation(s)
- Yihong Huang
- Laboratory of Biophysics, School of Medicine, South China University of Technology, Guangzhou, Guangdong, China
| | - Zidong Zhou
- Laboratory of Biophysics, School of Medicine, South China University of Technology, Guangzhou, Guangdong, China
| | - Tianqi Liu
- Laboratory of Biophysics, School of Medicine, South China University of Technology, Guangzhou, Guangdong, China
| | - Shengnan Tang
- Laboratory of Biophysics, School of Medicine, South China University of Technology, Guangzhou, Guangdong, China
| | - Xuegang Xin
- Laboratory of Biophysics, School of Medicine, South China University of Technology, Guangzhou, Guangdong, China.
| |
Collapse
|
40
|
Wang C, Choi HJ, Woodbury L, Lee K. Interpretable Fine-Grained Phenotypes of Subcellular Dynamics via Unsupervised Deep Learning. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2403547. [PMID: 39239705 PMCID: PMC11538677 DOI: 10.1002/advs.202403547] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Revised: 08/09/2024] [Indexed: 09/07/2024]
Abstract
Uncovering fine-grained phenotypes of live cell dynamics is pivotal for a comprehensive understanding of the heterogeneity in healthy and diseased biological processes. However, this endeavor poses significant technical challenges for unsupervised machine learning, requiring the extraction of features that not only faithfully preserve this heterogeneity but also effectively discriminate between established biological states, all while remaining interpretable. To tackle these challenges, a self-training deep learning framework designed for fine-grained and interpretable phenotyping is presented. This framework incorporates an unsupervised teacher model with interpretable features to facilitate feature learning in a student deep neural network (DNN). Significantly, an autoencoder-based regularizer is designed to encourage the student DNN to maximize the heterogeneity associated with molecular perturbations. This method enables the acquisition of features with enhanced discriminatory power, while simultaneously preserving the heterogeneity associated with molecular perturbations. This study successfully delineated fine-grained phenotypes within the heterogeneous protrusion dynamics of migrating epithelial cells, revealing specific responses to pharmacological perturbations. Remarkably, this framework adeptly captured a concise set of highly interpretable features uniquely linked to these fine-grained phenotypes, each corresponding to specific temporal intervals crucial for their manifestation. This unique capability establishes it as a valuable tool for investigating diverse cellular dynamics and their heterogeneity.
Collapse
Affiliation(s)
- Chuangqi Wang
- Department of Immunology and MicrobiologyUniversity of Colorado Anschutz Medical CampusAuroraCO80045USA
- Department of Biomedical EngineeringWorcester Polytechnic InstituteWorcesterMA01609USA
| | - Hee June Choi
- Department of Biomedical EngineeringWorcester Polytechnic InstituteWorcesterMA01609USA
- Vascular Biology Program and Department of SurgeryBoston Children's HospitalHarvard Medical SchoolBostonMA02115USA
| | - Lucy Woodbury
- Department of Biomedical EngineeringWorcester Polytechnic InstituteWorcesterMA01609USA
- Department of Biomedical EngineeringUniversity of ArkansasFayettevilleAR72701USA
| | - Kwonmoo Lee
- Department of Biomedical EngineeringWorcester Polytechnic InstituteWorcesterMA01609USA
- Vascular Biology Program and Department of SurgeryBoston Children's HospitalHarvard Medical SchoolBostonMA02115USA
| |
Collapse
|
41
|
Sigawi T, Israeli A, Ilan Y. Harnessing Variability Signatures and Biological Noise May Enhance Immunotherapies' Efficacy and Act as Novel Biomarkers for Diagnosing and Monitoring Immune-Associated Disorders. Immunotargets Ther 2024; 13:525-539. [PMID: 39431244 PMCID: PMC11488351 DOI: 10.2147/itt.s477841] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Accepted: 09/27/2024] [Indexed: 10/22/2024] Open
Abstract
Lack of response to immunotherapies poses a significant challenge in treating immune-mediated disorders and cancers. While the mechanisms associated with poor responsiveness are not well defined and change between and among subjects, the current methods for overcoming the loss of response are insufficient. The Constrained Disorder Principle (CDP) explains biological systems based on their inherent variability, bounded by dynamic boundaries that change in response to internal and external perturbations. Inter and intra-subject variability characterize the immune system, making it difficult to provide a single therapeutic regimen to all patients and even the same patients over time. The dynamicity of the immune variability is also a significant challenge for personalizing immunotherapies. The CDP-based second-generation artificial intelligence system is an outcome-based dynamic platform that incorporates personalized variability signatures into the therapeutic regimen and may provide methods for improving the response and overcoming the loss of response to treatments. The signatures of immune variability may also offer a method for identifying new biomarkers for early diagnosis, monitoring immune-related disorders, and evaluating the response to treatments.
Collapse
Affiliation(s)
- Tal Sigawi
- Faculty of Medicine, Hebrew University and Department of Medicine, Hadassah Medical Center, Jerusalem, Israel
| | - Adir Israeli
- Faculty of Medicine, Hebrew University and Department of Medicine, Hadassah Medical Center, Jerusalem, Israel
| | - Yaron Ilan
- Faculty of Medicine, Hebrew University and Department of Medicine, Hadassah Medical Center, Jerusalem, Israel
| |
Collapse
|
42
|
Hao K, Barrett M, Samadi Z, Zarezadeh A, McGrath Y, Askary A. Reconstructing signaling history of single cells with imaging-based molecular recording. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.11.617908. [PMID: 39416000 PMCID: PMC11482953 DOI: 10.1101/2024.10.11.617908] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/19/2024]
Abstract
The intensity and duration of biological signals encode information that allows a few pathways to regulate a wide array of cellular behaviors. Despite the central importance of signaling in biomedical research, our ability to quantify it in individual cells over time remains limited. Here, we introduce INSCRIBE, an approach for reconstructing signaling history in single cells using endpoint fluorescence images. By regulating a CRISPR base editor, INSCRIBE generates mutations in genomic target sequences, at a rate proportional to signaling activity. The number of edits is then recovered through a novel ratiometric readout strategy, from images of two fluorescence channels. We engineered human cell lines for recording WNT and BMP pathway activity, and demonstrated that INSCRIBE faithfully recovers both the intensity and duration of signaling. Further, we used INSCRIBE to study the variability of cellular response to WNT and BMP stimulation, and test whether the magnitude of response is a stable, heritable trait. We found a persistent memory in the BMP pathway. Progeny of cells with higher BMP response levels are likely to respond more strongly to a second BMP stimulation, up to 3 weeks later. Together, our results establish a scalable platform for genetic recording and in situ readout of signaling history in single cells, advancing quantitative analysis of cell-cell communication during development and disease.
Collapse
Affiliation(s)
- Kai Hao
- Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles, CA, USA
| | - Mykel Barrett
- Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles, CA, USA
| | - Zainalabedin Samadi
- Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles, CA, USA
| | - Amirhossein Zarezadeh
- Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles, CA, USA
| | - Yuka McGrath
- Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles, CA, USA
| | - Amjad Askary
- Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles, CA, USA
| |
Collapse
|
43
|
Hu Y, Zou Y, Qiao L, Lin L. Integrative proteomic and metabolomic elucidation of cardiomyopathy with in vivo and in vitro models and clinical samples. Mol Ther 2024; 32:3288-3312. [PMID: 39233439 PMCID: PMC11489546 DOI: 10.1016/j.ymthe.2024.08.030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 07/16/2024] [Accepted: 08/30/2024] [Indexed: 09/06/2024] Open
Abstract
Cardiomyopathy is a prevalent cardiovascular disease that affects individuals of all ages and can lead to life-threatening heart failure. Despite its variety in types, each with distinct characteristics and causes, our understanding of cardiomyopathy at a systematic biology level remains incomplete. Mass spectrometry-based techniques have emerged as powerful tools, providing a comprehensive view of the molecular landscape and aiding in the discovery of biomarkers and elucidation of mechanisms. This review highlights the significant potential of integrating proteomic and metabolomic approaches with specialized databases to identify biomarkers and therapeutic targets across different types of cardiomyopathies. In vivo and in vitro models, such as genetically modified mice, patient-derived or induced pluripotent stem cells, and organ chips, are invaluable in exploring the pathophysiological complexities of this disease. By integrating omics approaches with these sophisticated modeling systems, our comprehension of the molecular underpinnings of cardiomyopathy can be greatly enhanced, facilitating the development of diagnostic markers and therapeutic strategies. Among the promising therapeutic targets are those involved in extracellular matrix remodeling, sarcomere damage, and metabolic remodeling. These targets hold the potential to advance precision therapy in cardiomyopathy, offering hope for more effective treatments tailored to the specific molecular profiles of patients.
Collapse
Affiliation(s)
- Yiwei Hu
- Department of Chemistry, Zhongshan Hospital, and Minhang Hospital, Fudan University, Shanghai 200000, China
| | - Yunzeng Zou
- Department of Chemistry, Zhongshan Hospital, and Minhang Hospital, Fudan University, Shanghai 200000, China.
| | - Liang Qiao
- Department of Chemistry, Zhongshan Hospital, and Minhang Hospital, Fudan University, Shanghai 200000, China.
| | - Ling Lin
- Department of Chemistry, Zhongshan Hospital, and Minhang Hospital, Fudan University, Shanghai 200000, China.
| |
Collapse
|
44
|
Cong L, Guo X, Wang J, Meng F, Zhao J, Xu W, Shi W, Liang C, Shi Z, Xu S. In-droplet multiplex immunoassays for hypoxia-induced single-cell cytokines. Talanta 2024; 278:126548. [PMID: 39008932 DOI: 10.1016/j.talanta.2024.126548] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Revised: 06/21/2024] [Accepted: 07/10/2024] [Indexed: 07/17/2024]
Abstract
Cytokine expression is an important biomarker in understanding hypoxia microenvironments in tumor growth and metastasis. In-droplet-based immunoassays performed above the target cell membrane were employed to track the cytokines of single cells with the aid of three types of immuno-nanoprobes (one capture nanoprobe and two reporter nanoprobes). Single cells and nanoprobes were co-packaged in water-in-oil microdroplets (about 100 μm in diameter) using a cross-shaped microfluidic chip. In each droplet, capture nanoprobes would be first fixed to the cell surface by linking to membrane proteins that have been streptavidinized. Then, the capture nanoprobes can collect cell-secreted cytokines (VEGF and IL-8) by the antibodies, followed by two reporter nanoprobes that emit distinguishable fluorescence. Fluorescence imaging was utilized to record the signal outputs of two reporter probes, which reflect cytokine expressions secreted by a single tumor cell. The cytokine levels at different degrees of hypoxia induction were assessed. Multiple chemometric methods were adopted to distinguish differences in the secretion of two cytokines and the results demonstrated a positive correlation. This study developed an in-droplet, dual-target, simultaneous biosensing strategy for a single cell, which is helpful for understanding the impacts of hypoxia microenvironments on cell cytokines that are vital for assessing early cancer diagnosis and prognosis.
Collapse
Affiliation(s)
- Lili Cong
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun, 130012, PR China
| | - Xiaolei Guo
- State Key Laboratory of Inorganic Synthesis and Preparative Chemistry, College of Chemistry, Jilin University, Changchun, 130012, PR China
| | - Jiaqi Wang
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun, 130012, PR China
| | - Fanxiang Meng
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun, 130012, PR China
| | - Junyi Zhao
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun, 130012, PR China
| | - Weiqing Xu
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun, 130012, PR China; Institute of Theoretical Chemistry, College of Chemistry, Jilin University, Changchun, 130012, PR China
| | - Wei Shi
- Key Lab for Molecular Enzymology & Engineering of Ministry of Education, Jilin University, Changchun, 130012, PR China
| | - Chongyang Liang
- Institute of Frontier Medical Science, Jilin University, Changchun, 130021, PR China
| | - Zhan Shi
- State Key Laboratory of Inorganic Synthesis and Preparative Chemistry, College of Chemistry, Jilin University, Changchun, 130012, PR China
| | - Shuping Xu
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun, 130012, PR China; Institute of Theoretical Chemistry, College of Chemistry, Jilin University, Changchun, 130012, PR China; Center for Supramolecular Chemical Biology, College of Chemistry, Jilin University, Changchun, 130012, PR China.
| |
Collapse
|
45
|
Arora S, Singh S, Mittal A, Desai N, Khatri DK, Gugulothu D, Lather V, Pandita D, Vora LK. Spheroids in cancer research: Recent advances and opportunities. J Drug Deliv Sci Technol 2024; 100:106033. [DOI: 10.1016/j.jddst.2024.106033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/16/2024]
|
46
|
Frank SA, Yanai I. The origin of novel traits in cancer. Trends Cancer 2024; 10:880-892. [PMID: 39112299 DOI: 10.1016/j.trecan.2024.07.005] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Revised: 07/06/2024] [Accepted: 07/12/2024] [Indexed: 10/11/2024]
Abstract
The traditional view of cancer emphasizes a genes-first process. Novel cancer traits arise by genetic mutations that spread to drive phenotypic change. However, recent data support a phenotypes-first process in which nonheritable cellular variability creates novel traits that later become heritably stabilized by genetic and epigenetic changes. Single-cell measurements reinforce the idea that phenotypes lead genotypes, showing how cancer evolution follows normal developmental plasticity and creates novel traits by recombining parts of different cellular developmental programs. In parallel, studies in evolutionary biology also support a phenotypes-first process driven by developmental plasticity and developmental recombination. These advances in cancer research and evolutionary biology mutually reinforce a revolution in our understanding of how cells and organisms evolve novel traits in response to environmental challenges.
Collapse
Affiliation(s)
- Steven A Frank
- Department of Ecology and Evolutionary Biology, University of California, Irvine, CA 92697-2525, USA.
| | - Itai Yanai
- Perlmutter Cancer Center, New York University (NYU) Grossman School of Medicine, New York, NY 10016, USA; Institute for Systems Genetics, NYU Langone Health, New York, NY 10016, USA
| |
Collapse
|
47
|
Liu X, Zheng X. Microfluidic-Based Electrical Operation and Measurement Methods in Single-Cell Analysis. SENSORS (BASEL, SWITZERLAND) 2024; 24:6359. [PMID: 39409403 PMCID: PMC11478560 DOI: 10.3390/s24196359] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Revised: 09/21/2024] [Accepted: 09/28/2024] [Indexed: 10/20/2024]
Abstract
Cellular heterogeneity plays a significant role in understanding biological processes, such as cell cycle and disease progression. Microfluidics has emerged as a versatile tool for manipulating single cells and analyzing their heterogeneity with the merits of precise fluid control, small sample consumption, easy integration, and high throughput. Specifically, integrating microfluidics with electrical techniques provides a rapid, label-free, and non-invasive way to investigate cellular heterogeneity at the single-cell level. Here, we review the recent development of microfluidic-based electrical strategies for single-cell manipulation and analysis, including dielectrophoresis- and electroporation-based single-cell manipulation, impedance- and AC electrokinetic-based methods, and electrochemical-based single-cell detection methods. Finally, the challenges and future perspectives of the microfluidic-based electrical techniques for single-cell analysis are proposed.
Collapse
Affiliation(s)
| | - Xiaolin Zheng
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, Bioengineering College, Chongqing University, Chongqing 400044, China
| |
Collapse
|
48
|
Greig JC, Tipping WJ, Graham D, Faulds K, Gould GW. New insights into lipid and fatty acid metabolism from Raman spectroscopy. Analyst 2024. [PMID: 39258960 DOI: 10.1039/d4an00846d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/12/2024]
Abstract
One of the challenges facing biology is to understand metabolic events at a single cellular level. While approaches to examine dynamics of protein distribution or report on spatiotemporal location of signalling molecules are well-established, tools for the dissection of metabolism in single living cells are less common. Advances in Raman spectroscopy, such as stimulated Raman scattering (SRS), are beginning to offer new insights into metabolic events in a range of experimental systems, including model organisms and clinical samples, and across a range of disciplines. Despite the power of Raman imaging, it remains a relatively under-used technique to approach biological problems, in part because of the specialised nature of the analysis. To raise the profile of this method, here we consider some key studies which illustrate how Raman spectroscopy has revealed new insights into fatty acid and lipid metabolism across a range of cellular systems. The powerful and non-invasive nature of this approach offers a new suite of tools for biomolecular scientists to address how metabolic events within cells informs on or underpins biological function. We illustrate potential biological applications, discuss some recent advances, and offer a direction of travel for metabolic research in this area.
Collapse
Affiliation(s)
- Justin C Greig
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, 161 Cathedral Street, Glasgow, UK.
| | | | - Duncan Graham
- Pure and Applied Chemistry, University of Strathclyde, UK
| | - Karen Faulds
- Pure and Applied Chemistry, University of Strathclyde, UK
| | - Gwyn W Gould
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, 161 Cathedral Street, Glasgow, UK.
| |
Collapse
|
49
|
Khodr V, Clauzier L, Machillot P, Sales A, Migliorini E, Picart C. Development of an automated high-content immunofluorescence assay of pSmads quantification: Proof-of-concept with drugs inhibiting the BMP/TGF-β pathways. Biotechnol J 2024; 19:e2400007. [PMID: 39295554 DOI: 10.1002/biot.202400007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Revised: 07/19/2024] [Accepted: 07/22/2024] [Indexed: 09/21/2024]
Abstract
INTRODUCTION Bone morphogenetic proteins (BMPs) and transforming growth factors (TGF-β) are members of the TGF-β superfamily, known for their roles in several physiological and pathological processes. These factors are known to bind in vivo to BMP and TGF-β receptors, respectively, which induces the phosphorylation of Smad (pSmad) transcription factors. This pathway is generally studied with Western blot and luciferase bioluminescence assay, which presents some limitations. PURPOSE In this work, we developed and optimized a high-throughput assay to study pSmad pathways using immunofluorescence (IF) as an alternative to Western blot. We aimed to overcome the technical challenges usually faced in the classical IF assay in image acquisition, analysis, and quantification. METHODS We used C2C12 cells as a cellular model. The cells were stimulated with BMP-2 and TGF-β1 that were delivered either in solution (soluble) or via a biomaterial presenting the growth factor (GF), that is in a "matrix-bound" manner. Image acquisition parameters, analysis methods, and quantification of pSmads using IF were optimized for cells cultured on two types of supports: on bare glass and on a biomimetic coating made by self-assembly of the biopolymers hyaluronic acid and poly(l-lysine), which was crosslinked and then loaded with the GFs. RESULTS We performed high-content kinetic studies of pSmad expression for cells cultured in 96-well microplates in response to soluble and matrix-bound BMP-2 and TGF-β1. The detection limit of the IF-based assay was found to be similar to Western blot. Additionally, we provide a proof-of-concept for drug testing using inhibitors of BMP and TGF-β receptors, under conditions where specific signaling pathways are engaged via the ligand/receptor interactions. Altogether, our findings offer perspectives for future mechanistic studies on cell signaling and for studies at the single cell level using imaging methods.
Collapse
Affiliation(s)
- Valia Khodr
- Université Grenoble Alpes, INSERM, CEA, U1292 Biosanté, CNRS EMR BRM, Grenoble cedex, France
- CNRS, Grenoble Institute of Technology, LMGP, UMR, Grenoble, France
| | - Laura Clauzier
- Université Grenoble Alpes, INSERM, CEA, U1292 Biosanté, CNRS EMR BRM, Grenoble cedex, France
| | - Paul Machillot
- Université Grenoble Alpes, INSERM, CEA, U1292 Biosanté, CNRS EMR BRM, Grenoble cedex, France
| | - Adrià Sales
- Université Grenoble Alpes, INSERM, CEA, U1292 Biosanté, CNRS EMR BRM, Grenoble cedex, France
| | - Elisa Migliorini
- Université Grenoble Alpes, INSERM, CEA, U1292 Biosanté, CNRS EMR BRM, Grenoble cedex, France
| | - Catherine Picart
- Université Grenoble Alpes, INSERM, CEA, U1292 Biosanté, CNRS EMR BRM, Grenoble cedex, France
- CNRS, Grenoble Institute of Technology, LMGP, UMR, Grenoble, France
- Institut Universitaire de France, Paris, France
| |
Collapse
|
50
|
Manohar SM. Shedding Light on Intracellular Proteins using Flow Cytometry. Cell Biochem Biophys 2024; 82:1693-1707. [PMID: 38831173 DOI: 10.1007/s12013-024-01338-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/24/2024] [Indexed: 06/05/2024]
Abstract
Intracellular protein abundance is routinely measured in mammalian cells using population-based techniques such as western blotting which fail to capture single cell protein levels or using fluorescence microscopy which is although suitable for single cell protein detection but not for rapid analysis of large no. of cells. Flow cytometry offers rapid, high-throughput, multiparameter-based analysis of intracellular protein expression in statistically significant no. of cells at single cell resolution. In past few decades, customized assays have been developed for flow cytometric detection of specific intracellular proteins. This review discusses the scope of flow cytometry for intracellular protein detection in mammalian cells along with specific applications. Technological advancements to overcome the limitations of traditional flow cytometry for the same are also discussed.
Collapse
Affiliation(s)
- Sonal M Manohar
- Department of Biological Sciences, Sunandan Divatia School of Science, SVKM's NMIMS (Deemed-to-be) University, Vile Parle (West), Mumbai, 400056, India.
| |
Collapse
|