Fact-checked by Grok 2 weeks ago

Single-cell analysis

Single-cell analysis encompasses a suite of technologies and methods designed to profile the molecular, genetic, and functional characteristics of individual within heterogeneous populations, thereby uncovering cell-to-cell variations that are obscured in traditional bulk analyses of tissues or cell mixtures. This approach has revolutionized by enabling the dissection of cellular diversity, rare cell types, and dynamic processes such as and response to stimuli at unprecedented . The field traces its roots to early techniques in the for observing individual cells, but modern single-cell analysis emerged in the late with innovations like in the 1970s for sorting and analyzing cells based on surface markers. Significant advances accelerated in the 2000s, driven by high-throughput sequencing and ; for instance, the first single-cell RNA sequencing (scRNA-seq) experiments were reported in 2009, allowing genome-wide profiling of individual cells. Subsequent developments include multimodal techniques that integrate data from multiple "" layers, such as combining transcriptomics with or , to provide a holistic view of cellular states. Key techniques in single-cell analysis span various modalities, including (e.g., single-cell DNA sequencing for detecting ), transcriptomics (e.g., scRNA-seq for ), (e.g., single-cell for accessibility), (e.g., or for protein levels), and spatial methods that preserve tissue context (e.g., ). Isolation methods range from manual micromanipulation and capture to automated approaches like and fluorescence-activated cell sorting, enabling high-throughput processing of thousands to millions of cells. Computational tools are integral for data analysis, addressing challenges like noise, sparsity, and dimensionality through clustering, trajectory inference, and integration algorithms. Applications of single-cell analysis are broad and transformative across biomedical fields, including identifying rare cancer cells and tumor heterogeneity to guide precision , tracing lineages in , profiling immune cell responses in infectious diseases like , and elucidating neuronal diversity in . In clinical contexts, it supports discovery, drug response prediction, and understanding disease mechanisms, such as in or autoimmune disorders, ultimately advancing . As the technology matures, ongoing challenges include scaling to whole organisms, improving sensitivity for low-abundance molecules, and standardizing best practices to ensure reproducibility.

Fundamentals

Definition and principles

Single-cell analysis refers to a suite of technologies and methods designed to profile the molecular features of individual cells, including DNA, RNA, proteins, and metabolites, at unprecedented resolution. This approach enables the dissection of complex biological systems by examining the unique characteristics of each cell within a population, rather than relying on aggregated data from ensembles of cells. By isolating and analyzing single cells, researchers can uncover variations in gene expression, mutations, and other molecular states that define cellular identity and function. At its core, single-cell analysis is grounded in the principle of cellular heterogeneity, which posits that seemingly uniform cell populations exhibit significant diversity in their molecular profiles due to factors such as developmental stage, environmental influences, and intrinsic variability. This heterogeneity is often driven by , where random fluctuations in transcription and other processes lead to cell-to-cell differences, even among genetically identical s. To access this level of detail, cell populations must typically be dissociated into single-cell suspensions, allowing for targeted molecular that reveals these subtle variations. Such principles underscore the necessity of single-cell to capture the full of biological dynamics that are averaged out in traditional methods. In contrast to bulk analysis, which measures average signals from thousands or millions of cells and thereby masks rare subpopulations, low-abundance transcripts, and transitional states, single-cell analysis illuminates these hidden elements. Bulk methods, such as conventional sequencing, provide a composite view that can obscure critical insights into mechanisms or developmental processes, whereas single-cell approaches detect subpopulations, cellular transitions, and quantify dynamic changes over time. This distinction is particularly vital in contexts like tumor microenvironments, where rare malignant or immune cells drive but are undetectable in bulk samples. Key concepts in single-cell analysis include the notion of cell state, which encapsulates a cell's current molecular configuration—often represented by its —as a snapshot of its functional identity and potential . Lineage tracing, facilitated by molecular barcodes or computational from single-cell data, allows reconstruction of cellular pedigrees and paths, providing insights into evolutionary relationships within tissues. These concepts enable a deeper understanding of how individual cells contribute to emergent properties in multicellular organisms.

Historical development

The roots of single-cell analysis trace back to the , when enabled the study of individual cells in diseased tissues. In 1858, advanced cellular pathology by demonstrating through microscopic examination that diseases arise at the cellular level, establishing the principle that "every cell comes from a cell" (omnis cellula e cellula) and emphasizing the importance of analyzing cells individually rather than tissues as a whole. The mid-20th century saw the development of flow cytometry, which allowed for the quantitative analysis and sorting of individual cells based on optical properties. Pioneered in the 1960s, this technique evolved with the invention of the fluorescence-activated cell sorter (FACS) in 1972 by Leonard Herzenberg and colleagues at Stanford University, enabling high-throughput separation of cells labeled with fluorescent antibodies. In the 1970s, patch-clamp electrophysiology revolutionized the functional analysis of single cells by permitting precise measurement of ion channel currents. Developed by Erwin Neher and Bert Sakmann in 1976, this method used a glass micropipette to form a tight seal on the cell membrane, allowing recordings from individual ion channels; their work earned the 1991 Nobel Prize in Physiology or Medicine. The 1980s introduced molecular tools for single-cell , with the advent of (PCR) enabling amplification of genetic material from minimal samples. The first application of reverse transcription PCR (RT-PCR) to single cells occurred in 1989, when Rappolee et al. developed a method to quantify mRNA phenotypes, such as expression, in individual cells or as few as 100 cRNA molecules. Technological drivers in the 1990s included , which miniaturized fluid handling for precise cell manipulation. In 1990, Andreas Manz and H. Michael Widmer proposed the concept of micro total analysis systems (μTAS), laying the groundwork for integrated microfluidic devices that facilitated single-cell isolation and analysis in the following decades. The early 2000s marked the emergence of next-generation sequencing (NGS), dramatically reducing costs and increasing throughput for genomic studies. Commercial NGS platforms, such as 454 and Illumina, launched in 2005, provided the sensitivity needed to sequence low-input material, setting the stage for single-cell applications by enabling whole-transcriptome profiling from tiny samples. Single-cell RNA sequencing (scRNA-seq) emerged in 2009 with et al.'s protocol for mRNA-Seq analysis of a single mouse blastomere, capturing over 75% of the transcriptome and revealing gene expression differences in embryos. The 2010s saw scale-up through commercial platforms like , founded in 2012 and launching its system in 2016, which used gel bead emulsions to profile thousands of cells simultaneously. Integration of CRISPR-Cas9 with single-cell analysis accelerated in the , enabling at resolution. Following 's discovery in 2012, methods combining CRISPR screens with scRNA-seq appeared by 2016, allowing perturbation of genes in individual cells and readout of transcriptomic effects to uncover regulatory networks. In the , single-cell analysis transitioned from low-throughput techniques like quantitative PCR (qPCR) on individual cells to high-throughput , exemplified by droplet-based methods developed around 2013. These encapsulate single cells in picoliter droplets for parallel processing, achieving millions of cells per run and democratizing large-scale heterogeneity studies. This trend continued into the with further scaling and integration of modalities. In the , single-cell analysis expanded with enhanced multi-omics integrations and computational advancements, notably applied to global health challenges like the , and the emergence of long-read sequencing for improved transcript isoform resolution as of 2025.

Sample Isolation and Preparation

Isolation techniques

Single-cell isolation techniques are essential for separating individual cells from heterogeneous tissues or cell cultures, enabling high-resolution downstream analyses such as transcriptomics and . These methods must balance cell viability, recovery efficiency, and throughput while minimizing stress that could alter cellular states. Common approaches include mechanical dissociation, fluorescence-activated cell sorting (FACS), microfluidic encapsulation, and emerging spatial techniques, each suited to different sample types and experimental scales. Mechanical methods involve physical and enzymatic disruption to liberate cells from tissues. Manual dissection uses fine tools like or scalpels to excise specific regions, preserving spatial context but limited to low throughput and manual labor intensity. Grinding or homogenization mechanically tissues into suspensions, often combined with to remove debris, though it risks low viability due to shear forces. Enzymatic , the most widely adopted mechanical variant, employs proteases such as or collagenase to degrade extracellular matrices; for instance, collagenase protocols at 37°C for 30-60 minutes yield high cell yields from solid tumors, achieving viability rates exceeding 80% when optimized with short incubation times to reduce stress-induced changes. -based , effective for adherent cell lines, maintains over 90% viability in isolations but requires neutralization to prevent over-. Recovery efficiency in these methods typically ranges from 50-80%, influenced by tissue type and concentration. Flow cytometry with fluorescence-activated cell sorting (FACS) enables label-based isolation by interrogating cells in a fluid stream using lasers and fluorescent markers. Cells are stained with antibodies targeting surface proteins, then sorted into collection tubes based on scatter and fluorescence parameters at rates up to 10,000-20,000 cells per second. This deterministic approach achieves high purity (>95%) for specific subpopulations, with live-cell viability averaging 84% ± 5% despite shear stresses from high flow rates through small nozzles. Recovery efficiency is around 32-35% for live cells, impacted by gating to exclude aggregates, making FACS ideal for rare cell enrichment from complex mixtures like blood or dissociated tissues. Microfluidic devices facilitate scalable isolation through miniaturization, often integrating encapsulation for high-throughput processing. Droplet-based systems like Drop-seq encapsulate individual in nanoliter oil droplets with barcoded beads, enabling parallel analysis of thousands of ; the method generates over 100,000 droplets per minute, with single-cell purity of 90-99% and transcript recovery efficiency of ~13%, though viability is not directly quantified and depends on gentle handling to avoid doublets (0.4-11%). Microwell platforms, such as BD Rhapsody, use arrays of nanoliter wells to trap probabilistically, supporting inputs from 100 to 800,000 per cartridge with up to 80% capture efficiency and throughput exceeding 640,000 per run, preserving viability through low-shear microwell partitioning. These systems excel in scalability for applications, with recovery rates enhanced by visual . Emerging techniques address limitations in precision and spatial fidelity. (LCM) uses or lasers to procure targeted cells from tissue sections under , achieving 62% success for single neurons with high quality (detecting >7,000 genes per cell) and minimal contamination, though throughput is low (tens of cells per hour) and viability is preserved via snap-freezing rather than live sorting. Inertial focusing in curved microfluidic channels leverages fluid inertia to align particles without labels, enabling high-resolution separation (down to 80 nm) at flow rates up to 32 µL/min, suitable for size-based single-cell isolation with potential for high viability in label-free scenarios, though primarily validated on particles rather than live cells. These methods expand options for spatially resolved or unbiased isolations, with ongoing improvements in automation boosting efficiency.

Preparation and preservation methods

Following , single-cell preparations require careful and preservation to maintain molecular integrity, particularly for sensitive analytes like and proteins, ensuring accurate downstream profiling. disrupts the to release contents while minimizing damage to . Chemical is the most widely adopted method in single-cell workflows, employing hypotonic buffers containing non-ionic detergents such as 0.2% combined with RNase inhibitors to gently permeabilize cells without excessive shearing. Mechanical , such as or bead beating, applies physical force to break membranes but is less common in single-cell applications due to risks of uneven and biomolecule fragmentation; it is occasionally used in microfluidic devices for high-throughput . Thermal , involving freeze-thaw cycles or controlled heating, denatures membrane proteins but can introduce variability in yield and is typically reserved for robust cell types or nuclei . Preservation strategies are essential to halt during or , with methods tailored to the layer. For transcriptomics, methanol fixation rapidly permeabilizes s and stabilizes by dehydrating the sample, preserving transcript profiles comparable to fresh cells in droplet-based assays, though it may reduce capture efficiency for low-abundance transcripts. fixation, often at 1-4% , cross-links proteins and is preferred for to maintain spatial and functional proteome states, enabling high recovery in single-cell without substantial loss of analytical depth. using 10% (DMSO) in freezing media protects live cells by preventing formation, yielding viable cells post-thaw with sequencing performance equivalent to non-frozen samples; this method outperforms fixation for overall cell recovery in multi-omics studies. Live-cell handling prioritizes immediate processing at 4°C in RNase-free buffers to avoid stress-induced artifacts, often incorporating viability enhancers like . Quality control assesses preparation efficacy to ensure high-fidelity data. Cell viability is routinely evaluated using exclusion, where live cells repel the dye while dead ones stain blue, targeting >80-90% viability to minimize low-quality profiles in sequencing libraries. RNA integrity is quantified via the (RIN), with scores >7 indicating sufficient preservation for single-cell RNA sequencing, as lower values correlate with fragmented transcripts and reduced gene detection. Contamination prevention is paramount, given the sensitivity of single-cell assays to nucleases and ambient . Protocols mandate RNase-free environments, including certified workspaces, diethyl pyrocarbonate-treated water, and single-use plastics to eliminate exogenous RNases that could degrade samples during handling. A key challenge is the inherent instability of mRNA, which has a cytoplasmic averaging several hours in mammalian cells (e.g., ~7 h) but degrades rapidly (within minutes) upon without inhibitors due to ubiquitous RNases, underscoring the need for swift stabilization to capture true cellular states.

Genomics

Techniques

Single-cell genomics techniques focus on sequencing the DNA of individual cells to identify genetic variations, including single nucleotide variants (SNVs), copy number variations (CNVs), and structural variants, which reveal cellular heterogeneity obscured in bulk analyses. Due to the picogram-level DNA content in a single cell, whole-genome amplification (WGA) is a prerequisite step to generate sufficient material for library preparation and sequencing. Common WGA methods include multiple displacement amplification (MDA), which uses phi29 DNA polymerase for isothermal amplification, achieving high accuracy (up to 95% genome coverage at sufficient depth) but susceptible to chimeric artifacts and coverage biases; degenerate oligonucleotide-primed PCR (DOP-PCR), a PCR-based approach with degenerate primers for more uniform amplification, though with lower fidelity; and multiple annealing and looping-based amplification cycles (MALBAC), which employs quasi-linear pre-amplification to minimize bias. Advanced methods like LIANTI (linear amplification via transposon insertion) and duplex sequencing-based approaches (e.g., SISSOR) further improve uniformity and error correction, enabling detection of low-frequency variants with coverages exceeding 90%. Following , single-cell whole-genome sequencing (scWGS) provides comprehensive profiling of the entire , typically requiring 50-100× depth for reliable variant calling, while single-cell whole-exome sequencing (scWES) targets protein-coding regions for efficient detection in genes of interest. High-throughput platforms, such as (e.g., Chromium) or microwell arrays, facilitate processing of thousands of cells, integrating barcoding for multiplexing. For CNV analysis, methods like Read-depth-based inference or segmentation algorithms are applied post-sequencing. Seminal studies include scWGS on cells using MDA, revealing punctuated clonal with CNV profiles across 66 cells at ~0.1× average depth. Targeted panels, such as Tapestri for single-cell , focus on specific loci (e.g., 200-500 genes) in applications like (AML), detecting mutations in pathways like /MAPK. Challenges persist, including allelic dropout (up to 50% in some methods), biases affecting GC-rich regions, and the need for ultra-deep sequencing (often >1 Tb per ) to overcome low input, limiting to millions of cells.

Applications

Single-cell genomics has transformed by dissecting intratumor heterogeneity and tracing clonal evolution. In , scWGS has identified subclonal CNVs and mutations driving , with studies showing branched evolutionary patterns in primary tumors and distant sites from over 1,000 cells. Similarly, in , it revealed EGFR variant subclones contributing to therapeutic resistance, informing precision oncology strategies. In , single-cell enables lineage tracing through somatic mutations accumulated over cell divisions. For example, SNV profiling in 136 human cortical neurons delineated four major clades, mapping early and mosaicism. Applications extend to recombination studies, where scWGS of individual cells has quantified crossover events and structures. In immunology, it profiles hematopoietic stem cell (HSC) diversity and clonal dynamics, such as analyzing 16,000 CD34+ cells to uncover variable contributions to blood production and aging-related biases. In infectious disease contexts, though less common, it aids in studying pathogen integration, like HIV proviral DNA in infected T cells. Beyond biomedicine, microbial ecology benefits from scWGS, sequencing over 12,000 ocean microbes to discover novel clades and gene content variations. Overall, these applications highlight single-cell genomics' role in resolving genetic mosaicism, with ongoing advances in multi-omics integration enhancing its utility as of 2024.

Transcriptomics

Techniques

Single-cell transcriptomics primarily focuses on single-cell RNA sequencing (scRNA-seq), which profiles the of individual cells to reveal heterogeneity. Techniques involve cell isolation, RNA capture, reverse transcription to (cDNA), library preparation, and sequencing. Isolation methods include fluorescence-activated cell sorting (FACS), , and plate-based approaches like limiting dilution or micromanipulation. Library preparation strategies vary by capture efficiency and coverage. Full-length methods, such as Smart-seq2, amplify the entire transcriptome using oligo-dT primers and template-switching, enabling detection of splice variants and low-abundance transcripts but with higher amplification bias. In contrast, 3'-end tagging methods like Drop-seq or use barcoded beads in droplets to capture mRNA termini, allowing high-throughput profiling of thousands of cells with unique molecular identifiers (UMIs) to mitigate duplicates, though they provide less isoform resolution. Single-nucleus RNA sequencing (snRNA-seq) adapts these for frozen or hard-to-dissociate tissues by isolating nuclei, reducing dissociation artifacts but capturing primarily nuclear . Major commercial platforms include , which supports scalable droplet encapsulation for up to 10,000+ cells per run, and Fluidigm's C1 system for lower-throughput microfluidic isolation. Recent advancements as of 2024 incorporate long-read sequencing (e.g., Oxford Nanopore) for full-length isoform detection and integration, such as Slide-seq or Visium, to retain tissue context. Limitations include technical noise from low input (e.g., ~10 pg per cell), dropout events for lowly expressed genes, and costs exceeding $1,000 per sample for high-depth sequencing.

Applications

In , scRNA-seq maps cell lineages and trajectories, such as pseudotime inference in differentiation, revealing regulatory networks driving . For example, profiling mouse has identified transient cell states and signaling pathways like Wnt and FGF. utilizes scRNA-seq to dissect tumor heterogeneity, identifying rare subpopulations, such as therapy-resistant clones in , and the , including immune infiltration patterns in lung adenocarcinoma. This informs precision by correlating expression profiles with drug responses. In and infectious diseases, applications include profiling immune cell dynamics during , where scRNA-seq revealed exhausted T-cell states and cytokine responses in severe cases, aiding design. It also tracks B-cell maturation and antigen-specific responses in autoimmune disorders like . Broader medical uses encompass biomarker discovery in , where β-cell heterogeneity informs , and , mapping neuronal diversity in the atlas. As of 2025, integrations with multi-omics enhance holistic insights, though challenges like batch effects and standardization persist.

Epigenomics

Techniques

Single-cell epigenomics techniques enable the profiling of epigenetic features, such as , accessibility, and modifications, at the resolution of individual cells to reveal regulatory heterogeneity. These methods address challenges like limited input material and high noise through adaptations of assays, including conversion, tagmentation, and antibody-based capture. For , single-cell (scBS-seq) provides base-resolution mapping by treating DNA with to convert unmethylated cytosines, followed by sequencing. Post-bisulfite adaptor tagging (PBAT) variants reduce DNA loss from degradation, achieving coverage of approximately 50% of CpG sites per cell. More recent enzymatic methods, such as those using and TET2 enzymes in sciEM, improve efficiency and enable better mapping of non-CpG methylation without damage. Limitations include high costs and incomplete coverage due to input constraints. Chromatin accessibility is commonly assessed using single-cell assay for transposase-accessible chromatin with sequencing (scATAC-seq), which employs Tn5 transposase to insert sequencing adapters into open chromatin regions. This method generates sparse but genome-wide profiles, with typical yields of 1,000–10,000 unique fragments per cell, allowing inference of regulatory elements and transcription factors. Advancements like droplet-based dsciATAC-seq enhance throughput to thousands of cells by combinatorial indexing, though data sparsity and low signal-to-noise ratios remain challenges. Histone modifications are profiled via single-cell chromatin immunoprecipitation sequencing (scChIP-seq) or cleavage under targets and tagmentation (scCUT&Tag), which uses antibodies to capture modified histones followed by Tn5 tagmentation for library preparation. scCUT&Tag reduces background noise compared to scChIP-seq, enabling detection of marks like H3K27ac or H3K4me3 in hundreds of cells. Recent multi-mark approaches, such as scChIX-seq, allow simultaneous profiling of multiple modifications using machine learning for demultiplexing, but require substantial computational resources and validation. Coverage is often limited to 10,000–100,000 reads per cell, focusing on promoter and enhancer regions. Three-dimensional genome organization is explored with single-cell (scHi-C), which captures interactions via proximity ligation in intact . Combinatorial indexing in sciHi-C improves to ~100 and scalability to thousands of cells, revealing cell-type-specific loops and compartments. However, it demands high sequencing depth (millions of reads per cell) and is sensitive to nucleus isolation quality.

Applications

Single-cell has advanced understanding of tumor heterogeneity in cancer by identifying epigenetic states driving progression and . For instance, scATAC-seq in has revealed subpopulations with distinct chromatin accessibility patterns associated with , such as enhanced accessibility at loci in lymph node-invading cells. In , integrated multi-omics analyses have mapped epigenetic evolution during , highlighting rare clones with resistant profiles. These insights support precision by pinpointing epigenetic biomarkers for targeted therapies. In , single-cell traces lineage commitment through dynamic epigenetic landscapes. scBS-seq in early mouse embryos has shown methylation waves priming cell fates, while scMNase-seq in lymphoid progenitors identified motifs like TCF-1 that epigenetically bias toward T-cell lineages. Such studies elucidate regulatory mechanisms in embryogenesis and , aiding models of congenital disorders. Applications in include profiling epigenetic regulation of immune responses. scATAC-seq in tumor-infiltrating + T cells from gliomas has uncovered accessibility changes linked to exhaustion and suppression, revealing potential reversal strategies. In broader contexts, these methods dissect heterogeneity in immune cell activation during infections, informing vaccine design and .

Antibody-based methods

Antibody-based methods in single-cell analysis rely on the high specificity of antibodies to detect and quantify proteins through affinity binding, enabling targeted of cellular proteomes without the need for ionization or fragmentation. These techniques are particularly valuable for studying known protein markers in heterogeneous cell populations, offering multiplexing capabilities that range from a few to over 40 targets per cell. They typically involve labeling antibodies with fluorophores, metal isotopes, or DNA barcodes, followed by detection via optical or mass-based readout systems. While these methods excel in specificity and established workflows, they are limited to predefined targets and can suffer from antibody or batch variability. Flow cytometry represents a cornerstone of antibody-based single-cell proteomics, where suspended cells are interrogated by lasers as they flow through a detection chamber, allowing simultaneous of surface and intracellular proteins. Antibodies conjugated to fluorophores bind specific epitopes, and emitted light is captured to generate multi-dimensional profiles. Conventional systems support up to 18-20 markers, but flow cytometry expands this to over 50 by resolving full emission spectra, enabling deep phenotyping of immune cells or tumor heterogeneity. Throughput reaches thousands of cells per second, making it suitable for large-scale sorting and analysis. Detection sensitivity is approximately 500 molecules per cell, sufficient for most abundant surface markers but challenging for low-expression proteins. Mass cytometry, or CyTOF (cytometry by time-of-flight), extends flow cytometry principles by replacing fluorophores with stable metal isotope tags on antibodies, eliminating spectral overlap and enabling multiplexing of 40 or more parameters per cell. Cells are labeled, nebulized into droplets, and ionized in a plasma torch, with ions separated by mass-to-charge ratio for detection. This approach provides quantitative, single-cell resolution data comparable to traditional flow but with higher dimensionality, ideal for dissecting complex signaling networks in immune or stem cell populations. Like flow cytometry, it processes up to 1,000 cells per second and achieves similar sensitivity thresholds of around 100-500 antibody molecules per cell. Seminal work demonstrated its utility in mapping over 30 markers in human bone marrow cells. Immunofluorescence microscopy offers spatial context for antibody-based detection, where fixed cells or tissues are stained with fluorescently tagged antibodies and imaged to visualize protein localization at subcellular . This method is inherently low-throughput, typically analyzing dozens to hundreds of cells per , but provides invaluable insights into protein distribution within cellular or tissue microenvironments. is limited to 3-5 markers without advanced techniques, due to channel constraints and . Sensitivity reaches 10-100 molecules per cell, enabling detection of low-abundance transcription factors or signaling molecules. It is often combined with sample fixation protocols to preserve , though it requires careful optimization to minimize non-specific . Microfluidic ELISA variants, including nanowell arrays, facilitate the capture and quantification of secreted proteins from individual cells by confining them in picoliter-scale compartments coated with capture antibodies. These platforms isolate single cells via gravity settling or encapsulation, allowing diffusion-limited accumulation of analytes for enzymatic amplification and fluorescent readout. They are particularly suited for studying or secretion in immune cells, with examples detecting up to 42 effectors from hybridomas or T cells. Throughput scales to thousands of cells per array, though parallel processing is lower than flow methods. Sensitivity is enhanced by the confined volume, achieving detection limits of ~100-1,000 molecules per cell over minutes to hours of secretion. A foundational nanowell system enabled high-efficiency screening of antigen-specific B cells. Barcoding strategies, such as (CO-Detection by indEXing), integrate DNA-conjugated antibodies with iterative hybridization and imaging to achieve high-plex spatial . In , antibodies are tagged with unique barcodes; during readout, complementary primers hybridize to barcodes, enabling sequential addition of fluorophores via polymerization, followed by stripping for the next cycle. This allows visualization of 40-60+ markers in intact tissues at single-cell resolution, with signal-to-noise ratios exceeding 80:1 and minimal cross-talk. It combines elements of and for deep phenotyping of tissue architecture, such as immune cell niches in or tumors. Detection sensitivity is around 100 molecules per cell, supporting analysis of both and intracellular targets. The method was pioneered for multiplexed imaging of over 30 antigens in murine sections.

Mass spectrometry-based methods

Mass spectrometry (MS)-based methods enable unbiased, discovery-oriented quantification of the at the single-cell level by directly analyzing peptides or intact proteins without relying on predefined targets, contrasting with antibody-dependent approaches. These techniques involve cell isolation, , enzymatic digestion (for ), and ionization followed by detection, allowing identification of thousands of proteins per cell while capturing post-translational modifications and sequence variants. Key challenges include low protein abundance (~10^5 to 10^8 molecules per cell), requiring ultra-sensitive instrumentation and minimized sample losses during processing. One prominent platform is nanodroplet processing in one pot for trace samples (NanoPOTS), introduced in 2018, which uses a microfluidic chip to confine sample preparation within nanowells (~200 nL volume) for efficient cell lysis, protein digestion, and peptide cleanup. This method achieves label-free detection of approximately 1,000-3,000 proteins per single mammalian cell, with coverage of ~20-50% of the expressed proteome depending on cell type, by minimizing adsorption losses and integrating with nano-liquid chromatography (nanoLC)-electrospray ionization (ESI)-MS. Subsequent automation via nested NanoPOTS (N2) chips has enhanced robustness and protein recovery for isobaric labeling workflows. Orbitrap-based mass spectrometers provide high-resolution (up to 480,000 FWHM) and high-mass-accuracy (~1-5 ppm) analysis of peptides generated post-digestion, crucial for distinguishing isobaric species in complex single-cell digests. Coupled with (DIA) or parallel reaction monitoring, instruments like the Eclipse or enable deeper coverage (~1,500-3,000 proteins per cell) through fast scanning and ion trapping in the analyzer after quadrupole selection and higher-energy collisional dissociation (HCD). Recent advancements, such as the 2023 , support with improved sensitivity for low-input samples. Emerging single-cell MS workflows achieve throughputs of 10-100 cells per hour, balancing speed with depth via automated sample handling and multiplexed analysis, though this lags behind transcriptomics platforms. Proteome coverage typically spans 20-50% of detectable proteins, influenced by cell size and preprocessing efficiency. Ionization strategies differ by application: matrix-assisted laser desorption/ionization (MALDI) suits spatial profiling by co-crystallizing analytes with a matrix (e.g., sinapinic acid) on tissue slides for laser-induced desorption, preserving cellular architecture but introducing matrix interference. In contrast, ESI facilitates flow-based analysis in nanoLC-MS setups, generating multiply charged ions from liquid droplets for sequential peptide separation and detection, ideal for high-throughput single-cell suspensions but requiring desalting. Hybrid approaches, like MALDI-2 with post-ionization, enhance sensitivity for both modalities.

Applications

Single-cell proteomics has advanced understanding of cellular heterogeneity in cancer, enabling proteogenomic analysis to identify molecular subtypes and therapeutic targets. For instance, in , it reveals protein-level variations driving tumor progression and response to , supporting precision . In , these methods dissect immune cell states within the , quantifying protein markers to uncover novel subsets and their interactions with cancer cells. Applications include profiling T cell exhaustion in solid tumors, informing efficacy as of 2024. For , single-cell traces protein dynamics during , highlighting heterogeneity in populations. Recent studies (2023) have mapped changes in neuronal progenitors, linking post-translational modifications to lineage commitment. Clinically, it aids biomarker discovery for diseases like Alzheimer's, identifying early protein signatures in cerebrospinal fluid-derived cells, and in infectious diseases such as , classifying infection severity via plasma profiling. As of 2025, spatial integrates these insights for tissue-level analysis in and other conditions.

Metabolomics

Techniques

Single-cell metabolomics techniques primarily rely on mass spectrometry (MS)-based approaches to detect and quantify small-molecule metabolites, enabling the of metabolic states at the individual level. These methods address the challenges of low metabolite abundance and small sample volumes by integrating high- and separation techniques. Key strategies include direct analysis without prior for spatial and extraction-based methods for targeted , with overall reaching attomole to femtomole ( to fmol) levels per . However, current coverage typically spans only 10-30% of the estimated cellular due to limitations in efficiency and spectral complexity. Capillary electrophoresis-MS (CE-MS) is particularly suited for analyzing polar metabolites, such as amino acids and nucleotides, in single cells. This technique separates charged species based on electrophoretic mobility in a narrow capillary, coupled online to MS for identification and quantification. Seminal work demonstrated detection of more than 100 metabolites from a single metacerebral neuron of Aplysia californica, including central carbon pathway intermediates, at concentrations down to the attomole range. Later advancements, such as sheathless nano-CE-MS, have enabled profiling of over 80 unique polar metabolites per cell in embryonic models like Xenopus laevis, highlighting metabolic heterogeneity during development. CE-MS excels in minimal sample dilution and rapid analysis (under 10 minutes per cell), though it requires precise control of capillary coatings to prevent analyte adsorption. Matrix-assisted laser desorption/ionization-MS (MALDI-MS) imaging provides spatial metabolomics without the need for physical isolation, allowing metabolite mapping directly from sections or arrays at subcellular . By applying a to the sample and ablating it with a , MALDI-MS generates images of hundreds of metabolites, including and small polar compounds, across thousands of s. For instance, high-resolution MALDI-MS (down to 1 μm) has visualized distributions in individual neurons, revealing compartmentalized metabolic gradients within single s. This approach supports high-throughput analysis, such as generating over 150,000 profiles from , and is valuable for studying -level heterogeneity while preserving spatial context. Limitations include interference for low-mass s (<200 Da) and the need for cryogenic sample preparation to maintain metabolite integrity. Microfluidic extraction coupled with nanoscale liquid chromatography-MS (nanoLC-MS) facilitates targeted analysis of non-polar metabolites like lipids and polar ones such as amino acids from isolated single cells. Microfluidic devices enable gentle cell positioning, lysis, and on-chip extraction into nanoliter volumes, minimizing loss and contamination before injection into nanoLC-MS for separation and detection. This method has profiled dozens of lipids from individual algal cells like Chlamydomonas reinhardtii, achieving femtomole (fmol) sensitivity for species like phosphatidylcholines. In algal models, similar setups identified over 50 lipid species per Chlamydomonas reinhardtii cell, underscoring its utility for membrane-related metabolism. The integration of automation allows throughput of tens to hundreds of cells per run, though optimization of extraction solvents is critical for broad coverage. Recent advances as of 2025 include high-throughput spatial methods like HT SpaceM, which detect >100 metabolites from >1,000 individual cells per hour using laser-ablation , enhancing scalability for complex tissues. Isotope labeling enhances analysis in single-cell metabolomics by tracing dynamic metabolic pathways, often using stable like ¹³C to monitor substrate incorporation over time. For example, ¹³C-glucose tracing via reveals glycolytic and TCA cycle by quantifying distributions in downstream metabolites. Early applications used multi-isotope imaging to track ¹³C-oleate uptake and metabolism in adipocytes, demonstrating intracellular at the single-cell level with sub-femtomole . More recent extensions to glucose labeling in tumor cells have mapped heterogeneous rates, integrating with CE- or nanoLC- for multiplexed analysis of labeled polar metabolites. This approach complements steady-state profiling by providing kinetic insights, though it requires controlled incubation times (minutes to hours) and advanced modeling. Cell lysis is commonly employed upstream for efficient isotope-labeled metabolite extraction in these workflows.

Applications

Single-cell metabolomics has revealed significant metabolic heterogeneity within populations, particularly in variations of that underpin the Warburg effect. In tumor subclones, individual cells exhibit differential reliance on aerobic , with some maintaining high production despite oxygen availability, enabling rapid and to hypoxic microenvironments. For instance, single-cell analysis of cells has identified subpopulations with elevated glycolytic flux, correlating with enhanced metastatic potential and resistance to therapies targeting . In drug metabolism studies, single-cell metabolomics has elucidated variations in cytochrome P450 (CYP450) activity among hepatocytes, highlighting functional heterogeneity in xenobiotic processing. Primary human hepatocytes exposed to drug challenges display distinct metabolic subgroups, where certain cells upregulate CYP450-mediated oxidation of substrates like acetaminophen, leading to differential toxicity responses and fatty acid accumulation profiles. This approach has identified resilient hepatocyte clusters with sustained phase I metabolism, informing personalized pharmacotherapy by revealing how inter-cellular differences influence drug clearance and adverse effects. Applications extend to microbial communities, where single-cell metabolomics uncovers metabolite exchange dynamics in consortia, fostering cooperative survival strategies. In intestinal bacterial populations, individual cells show heterogeneous production and uptake of and , enabling cross-feeding that stabilizes community structure under nutrient limitation. For example, analysis of consortia has demonstrated how species export succinate to support Firmicutes growth, illustrating metabolic interdependencies that drive ecosystem resilience and influence host health. A notable 2023 study applied single-cell to map metabolite gradients in neuronal progenitor cells, revealing spatially resolved variations in lipid and energy metabolites along cellular processes. This work identified dysregulated gradients in cells harboring CLN6 mutations, linking altered ceramide distribution to neuronal dysfunction in models and providing insights into progressive neurodegeneration. Furthermore, single-cell metabolomics has quantified NADPH/NADP+ ratios to probe states during , exposing cellular vulnerabilities in stress responses. In immune cells under challenge, individual macrophages exhibit fluctuating NADPH levels, with low-ratio subpopulations showing depleted pools and heightened ROS damage, while high-ratio cells maintain reductive capacity for survival. This granularity has advanced understanding of homeostasis in , guiding interventions.

Multi-Omics Integration

Integration strategies

Integration strategies in single-cell analysis encompass both computational algorithms for fusing disparate datasets and experimental protocols for simultaneously capturing multiple molecular layers from the same cells, enabling a more holistic view of cellular states. These approaches address the challenge of linking features across modalities, such as transcriptomes, proteomes, and epigenomes, to uncover coordinated regulatory mechanisms that single-modality might miss. Computationally, joint embedding methods project multi-omics data into a shared low-dimensional space to identify common variation. Seurat, through its weighted nearest neighbor (WNN) framework, integrates modalities like and ATAC by computing modality-specific embeddings and combining them based on feature importance, as demonstrated in analyses of paired single-cell datasets where cell types align across layers with minimal distortion. Similarly, Multi-Omics Factor Analysis (MOFA) decomposes multi-omics matrices into latent factors that capture shared and modality-specific sources of variation, applied successfully to single-cell and data to reveal differentiation trajectories with factors explaining up to 80% of total variance. (CCA) underpins many of these methods by maximizing correlations between paired datasets; for instance, bi-order CCA extends traditional CCA to align cells and features simultaneously in multimodal single-cell data, improving clustering concordance across and protein profiles. Experimental strategies facilitate direct multi-omics capture to minimize alignment errors inherent in computational fusion. enables simultaneous measurement of and surface protein levels using oligonucleotide-tagged antibodies, allowing quantification of up to 100 epitopes alongside in thousands of cells, as shown in immune cell profiling where protein markers refined transcriptomic cell-type assignments. For accessibility and , SHARE-seq profiles and from the same single nuclei via split-pool barcoding, capturing open regions and in parallel to link enhancers with target transcripts in embryonic development studies. More advanced multi-modal assays, such as TEA-seq, integrate transcripts, epitopes, and ATAC profiles in a single workflow using droplet-based partitioning, profiling over 10,000 cells per run to dissect T-cell heterogeneity with correlated multimodal features. Recent advances as of 2025 include methods like scMFG, which uses feature grouping for improved multi-omics integration, and comprehensive frameworks to guide method selection for diverse datasets. To handle discrepancies across modalities or batches, alignment techniques like the algorithm correct for technical variations while preserving biological signals; in multi-omics contexts, Harmony projects s into a corrected space, reducing batch effects in RNA-ATAC datasets and enhancing cross-modality matching. Successful integrations often achieve high concordance, with linked features (e.g., RNA-protein pairs) showing scores exceeding 0.7, as evaluated in benchmarks of methods like Seurat and MOFA on paired datasets.

Combined profiling approaches

Combined profiling approaches encompass experimental platforms that enable the simultaneous measurement of multiple layers—such as , epigenome, , or genetic perturbations—from individual cells, allowing direct linkage of molecular states without relying on post-acquisition computational alignment. These methods leverage advances in , barcoding, and sequencing to scale multi-omics , revealing coordinated regulatory mechanisms in heterogeneous populations like immune or tumor cells. By capturing modalities in a unified , they provide higher fidelity correlations compared to sequential assays, though challenges remain in balancing throughput, sensitivity, and cost. A foundational platform is Perturb-Seq, which integrates -Cas9-mediated genetic perturbations with single-cell RNA sequencing (scRNA-seq) to map gene functions through transcriptional readouts. In this approach, cells are transduced with a pooled library of CRISPR guide RNAs targeting specific genes, followed by scRNA-seq to quantify perturbation effects on the ; guide RNA barcodes are captured alongside transcripts for perturbation tracing. Originally demonstrated in 2016 on bone marrow-derived dendritic cells and K562 cell lines, Perturb-Seq profiled over 200,000 cells to uncover regulatory circuits, including targets and epistatic interactions during immune activation by . ECCITE-seq extends this paradigm by combining expanded perturbations, scRNA-seq, and surface protein profiling via oligonucleotide-tagged antibodies, enabling multimodal immune cell characterization. The method incorporates additional feature barcodes for clonotype detection and increased coverage, allowing simultaneous assessment of genetic effects on and protein levels in pooled screens. Introduced in 2019, ECCITE-seq was applied to primary T cells and natural killer cells, revealing molecular regulators of inhibitory checkpoints and enhancing resolution of functional heterogeneity in adaptive immunity. The Chromium Single Cell Multiome represents a widely adopted commercial platform for joint epigenomic and transcriptomic profiling, capturing accessibility via and 3' from the same nuclei in a droplet-based . Launched in 2020 and refined through 2021 updates, it processes up to 10,000 nuclei per sample across 8 parallel channels, facilitating discovery of gene-regulatory interactions in diverse tissues like and tumors. This has been instrumental in studies of and disease states, providing paired multimodal data for thousands of cells per run. Nanobiosensors offer an emerging avenue for direct co-detection of and proteins within live cells, utilizing nanoscale probes like nanowires or plasmonic nanostructures for intracellular, monitoring without sequencing. These sensors exploit optical or electrical signals to quantify dynamics, such as mRNA into proteins, at subcellular resolution. Representative implementations, including field-effect transistor-based arrays, have demonstrated sensitivity for multiplexed analyte detection in cellular microenvironments, supporting functional studies of signaling pathways. These platforms routinely achieve throughputs exceeding 10^3 cells with three or more modalities, as seen in trimodal assays combining , ATAC, and protein data, thereby scaling to dissect complex cellular responses.

Spatial Single-Cell Analysis

Spatial transcriptomics methods

methods enable the measurement of profiles while retaining the two-dimensional organization of cells within intact tissue sections, bridging the gap between dissociated single-cell RNA sequencing and traditional . These approaches are essential for understanding cellular heterogeneity in context, such as in developmental processes or microenvironments, and are divided into sequencing-based techniques that rely on barcoded capture arrays followed by next-generation sequencing, and imaging-based techniques that use fluorescent probes for direct visualization. Sequencing-based methods typically offer higher molecular throughput but coarser (10–55 μm), suitable for large tissue areas with 10^4–10^6 capture spots, while imaging-based methods provide subcellular resolution (~0.1–1 μm) but are limited to targeted panels of hundreds to thousands due to imaging constraints. Slide-seq, introduced in 2019, represents a sequencing-based approach that transfers from cryopreserved tissue sections onto a slide covered with randomly distributed DNA-barcoded beads, each with a unique spatial position decoded post-sequencing. This method achieves near-single-cell resolution of approximately 10 μm, enabling genome-wide profiling of thousands of genes across large areas and facilitating the identification of cell types and spatial patterns, as demonstrated in cerebellum and tissues. An improved version, Slide-seqV2, enhances transcript capture efficiency to 30–50% while maintaining scalability for high-throughput applications. Visium, commercialized by in 2019 and building on the foundational platform, uses glass slides with arrays of 55 μm-diameter spots, each containing spatially barcoded that capture polyadenylated mRNA from permeabilized sections. Following reverse transcription and library preparation, sequencing yields whole-transcriptome data per spot, often deconvoluted to infer single-cell contributions, across capture areas up to 6.5 mm × 6.5 mm with about 5,000 spots per slide. This untargeted method has been widely adopted for profiling complex s like tumors, providing positional maps at moderate resolution. MERFISH (multiplexed error-robust ), pioneered in , is an imaging-based technique that employs combinatorial encoding with multiple fluorophores and sequential hybridization rounds to assign unique, error-correcting barcodes to targeted species, allowing direct counting and localization at single-molecule . It profiles 100–1,000 genes across hundreds of s in ~1 mm² tissue areas, with subcellular precision (~200 nm), and has revealed subcellular distributions and cell-type-specific expression in diverse samples, including cell lines and sections. Extensions like expansion-MERFISH further improve and for broader transcriptomic coverage. SeqFISH (sequential ), originally developed in 2014 and refined in later iterations, uses successive rounds of probe hybridization and imaging with a small set of fluorophores to generate binary barcodes for identification, enabling high-plex profiling without sequencing. It achieves subcellular (~250 ) for up to 249 genes in over 10,000 cells across slices, capturing transcripts with copy numbers from 0 to over 100 per cell and uncovering robust in neural tissues. Variants such as seqFISH+ scale to thousands of genes by optimizing readouts and reducing optical crowding, supporting applications in fixed tissues up to ~1 mm².

Spatial multi-omics methods

Spatial multi-omics methods enable the simultaneous profiling of multiple molecular layers, such as proteins, , metabolites, and elements, while preserving their spatial organization within s. These approaches build on by integrating additional modalities to provide a more holistic view of cellular heterogeneity and interactions . Unlike single-modality techniques, spatial multi-omics addresses the limitations of dissociated single-cell analysis by maintaining , allowing researchers to correlate molecular profiles with positional context. One prominent method is spatial CITE-seq, which combines cellular indexing of transcriptomes and epitopes () with spatial profiling on slides. This technique uses oligonucleotide-tagged antibodies to detect up to 300 surface proteins alongside whole-transcriptome sequencing, achieving cellular resolution through digital spatial profiling. Developed in 2023, spatial CITE-seq has been applied to map immune cell states in tumors, revealing protein-RNA correlations that highlight functional diversity within spatial niches. Hydrogel embedding techniques, such as those involving tissue expansion and on-slide protein digestion, facilitate spatial proteomics integrated with metabolomics. In these methods, tissues are embedded in swellable hydrogels to enhance resolution, followed by mass spectrometry-based extraction of proteins and metabolites from defined regions. For instance, filter-aided expansion proteomics enables single-cell and organelle-level profiling of proteomes in formalin-fixed paraffin-embedded (FFPE) samples, while complementary metabolomics captures small molecules, providing insights into metabolic-protein linkages in diseased tissues. These approaches, advanced in 2024 and recognized as Nature Methods' Method of the Year, overcome diffusion barriers in traditional imaging by physically expanding samples up to fourfold, improving spatial precision for multi-layer analysis; for example, as of November 2025, spatial multi-omics has revealed links between pro-inflammatory chemokines and aggressive prostate cancer. NanoSIMS (nanoscale ) offers high-resolution isotope imaging for spatial multi-omics, particularly targeting metabolites and elemental composition at the single-cell level. By samples with a cesium and detecting secondary ions, NanoSIMS achieves sub-50 resolution to map stable isotope-labeled metabolites (e.g., 13C or 15N incorporation) alongside elements like carbon, , and within organelles. This method has been instrumental in quantifying metabolic fluxes in microbial communities and mammalian cells, revealing nanoscale heterogeneity in nutrient uptake and biosynthesis. In 2025, advances in AI-enhanced spatial multi-omics have pushed resolutions beyond optical limits, enabling sub- imaging of multiple layers. Techniques like Seq-Scope-eXpanded integrate tissue expansion with computational super-resolution algorithms to achieve near-nanometer transcriptomic and proteomic mapping, while AI platforms such as Thor deconvolute low-resolution data into cell-level multi-omics profiles with minimal artifacts. These innovations, including nano-array capture chips, have facilitated the integration of epigenomic, transcriptomic, and proteomic data in complex s, enhancing throughput and accuracy for large-scale studies. Despite these progresses, spatial multi-omics methods face challenges, including cross-talk between molecular layers due to during and signal overlap in multiplexed detection. Minimizing such requires optimized protocols, such as sequential or computational correction, to ensure layer-specific below detectable thresholds in high-plex assays. Additionally, integrating diverse data types demands robust strategies to avoid spatial distortions.

Computational Analysis

Data preprocessing and quality control

and are essential initial steps in single-cell analysis to ensure the reliability and accuracy of downstream computational analyses by removing artifacts and technical noise from raw datasets. These processes typically begin with assessing key quality metrics for individual s, such as library size (total molecular identifiers or UMIs per ), the number of detected genes per , and the percentage of mitochondrial , which can indicate viability. For instance, s with fewer than 500 UMIs or 200 genes are often filtered out as low-quality, while those exceeding 20% mitochondrial content are typically discarded to exclude dying or stressed s. Filtering extends to identifying and removing doublets or multiplets, which arise from the co-encapsulation of multiple cells during droplet-based single-cell sequencing (scRNA-seq) and can confound identification. The addresses this by simulating artificial doublets from the observed distribution and using k-nearest neighbors to score and remove suspected multiplets, achieving high sensitivity in datasets with doublet rates up to 10%. After filtering, corrects for technical variations like sequencing depth and batch effects; common approaches include log-transformation of counts after adding a pseudocount, or more advanced methods like scTransform, which models count distributions using regularized negative to stabilize variance across cells and batches. Single-cell datasets are prone to high dropout rates, where is undetected despite transcription, often exceeding 80% for lowly expressed genes due to technical limitations in capture efficiency. Imputation methods, such as (Markov Affinity-based Graph Imputation of Cells), mitigate this by diffusing information across similar cells in a manifold representation, recovering continuous patterns and reducing noise without over-smoothing biological variability. Popular open-source toolkits like Scanpy in and Seurat in provide integrated pipelines for these steps, enabling scalable processing of large datasets through functions for metric calculation, visualization of distributions (e.g., violin plots for library sizes), and automated filtering thresholds.

Dimensionality reduction and clustering

Dimensionality reduction is a fundamental step in single-cell analysis that compresses high-dimensional data into lower-dimensional representations, facilitating and pattern identification while preserving biological variance. In single-cell RNA sequencing (scRNA-seq), datasets often comprise thousands of cells and tens of thousands of genes, making direct analysis computationally intensive and visually intractable. Techniques like (PCA) provide an initial linear projection, capturing the largest sources of variance, typically retaining the top 50 principal components (PCs) for downstream tasks such as clustering. Non-linear methods further refine these projections for two-dimensional (2D) visualization, enabling intuitive exploration of cellular heterogeneity. (t-SNE) maps cells into 2D space by minimizing differences in probability distributions of pairwise similarities, commonly using a parameter of 30 to balance local and global structure preservation. Uniform manifold approximation and projection (UMAP) offers a faster alternative, preserving both local neighborhoods and global topology through graph-based optimization, often outperforming t-SNE in scalability and structure retention for datasets exceeding 10,000 cells. These methods, applied post-PCA, reveal clusters corresponding to cell types or states, though t-SNE can distort global distances and UMAP may require tuning of the minimum distance parameter (default 0.1) for optimal separation. Recent developments as of 2025 include advanced clustering frameworks like scMSCF, which enhances accuracy and stability for scRNA-seq data through multi-scale approaches. Clustering partitions reduced-dimensional data into groups of similar cells, typically using graph-based or partitioning on the or UMAP embeddings. The , a modularity optimization adapted for scRNA-seq, iteratively merges communities to maximize intra-cluster , with resolution parameters ranging from 0.4 to 1.0 to control cluster granularity—lower values yield broader groups, while higher values detect finer subpopulations. , in contrast, partitions data into a predefined number of centroids by minimizing within-cluster variance, suitable for spherical distributions but less effective for complex manifolds in single-cell data. Cluster quality is validated using metrics like the adjusted Rand index (), which measures agreement with ground-truth labels (ideal ARI > 0.8), highlighting Louvain's robustness in benchmarks across diverse datasets. Batch integration addresses technical variations across datasets, enabling joint analysis of multi-sample scRNA-seq experiments. aligns batches by identifying shared subspaces of variation, as implemented in Seurat, where it projects data into a corrected low-dimensional space before clustering. Mutual nearest neighbors (MNN) corrects batches by matching cells with shared biological states across high-dimensional space, preserving cell-type distinctions without assuming identical compositions, and scales to tens of thousands of cells with minimal computational overhead. For large-scale analyses involving millions of cells, and clustering require optimized implementations to manage memory and runtime. Methods like accelerated variants handle datasets of 10^6 cells by leveraging operations, achieving runtimes under 10 minutes on standard hardware. GPU-accelerated frameworks, such as those in cuML, further enhance scalability for UMAP and Louvain on datasets exceeding 10^6 cells, reducing computation time by 10-100 fold compared to CPU-based alternatives while maintaining ARI scores above 0.7.

Trajectory inference

Trajectory inference reconstructs the temporal progression of cell differentiation or response processes from static single-cell RNA sequencing snapshots by ordering cells along pseudotime axes that represent continuous state transitions. This approach assumes that cells sampled from an asynchronous population reflect underlying dynamics, enabling the modeling of processes like or perturbation responses without time-series data. Key inputs include gene expression gradients, which indicate progressive changes in transcript levels across cells, and bifurcation points, where trajectories split to represent divergences. Monocle 2 employs reversed with the Discriminative Dimensionality Reduction via Tree (DDRTree) algorithm to infer branching trajectories in an manner. DDRTree learns a low-dimensional by optimizing a manifold learning objective that minimizes reconstruction error while enforcing topology, allowing it to capture multiple fate decisions from noisy single-cell data. This method partitions the data into a principal where branches represent diverging cell fates, facilitating downstream analyses like pseudotime assignment and expression along paths. Slingshot infers cell by first constructing a (MST) on pre-clustered cells in reduced dimensions, then fitting simultaneous principal curves to define smooth, elastic paths along inferred . Starting from user-specified or automatically detected root clusters, it extends curves to endpoints while minimizing orthogonal distances to data points, providing pseudotimes for each . This geometry-based fitting ensures robustness to varying complexities, such as linear or multi-branching structures. Introduced in 2019, Partition-based Graph Abstraction (PAGA) reconciles clustering and by building a topology-preserving where nodes represent clusters and edges connectivity based on shared nearest neighbors. This abstraction captures coarse-grained manifold structure, from which fine-grained trajectories can be embedded using force-directed layouts, handling complex topologies like cycles or multi-fork branches more effectively than tree-based methods. PAGA's weights reflect confidence in transitions, aiding and validation of inferred . Recent advances as of 2025 include tools like scEGOT, a for with high interpretability, and Genes2Genes for aligning trajectories across datasets. Benchmarks evaluating these methods against simulated ground truth datasets show strong performance, with correlations between inferred and true pseudotimes often exceeding 0.8 for linear and simple branching trajectories, though accuracy varies with data complexity and noise levels. typically operates on clustered cells derived from to initialize lineage structures.

Cell-cell interaction modeling

Cell-cell interaction modeling in single-cell analysis involves computational inference of communication networks between cells, primarily through the detection of ligand-receptor (LR) pairs expressed in sender and receiver cells, respectively. These methods leverage single-cell sequencing (scRNA-seq) data to quantify co-expression patterns of known LR interactions, enabling the of signaling pathways within heterogeneous tissues. Central to these approaches are curated databases of LR pairs, such as those integrated into tools like CellPhoneDB, which compile over 2,000 directional interactions derived from protein-protein interaction resources including . One widely adopted method is CellPhoneDB, which infers cell-type-specific interactions by testing the of LR pair expression across cell clusters. It employs testing to generate a of expression levels, identifying significant pairs with a threshold of less than 0.05 after multiple-testing correction, while accounting for multi-subunit complexes in ligands and receptors. This approach has been instrumental in revealing context-specific communication, such as immune cell crosstalk in tumors. NicheNet extends LR inference by incorporating prior biological knowledge to predict downstream effects on target gene expression in receiver cells. It prioritizes ligands based on their ability to explain observed gene regulatory changes, using a network of signaling pathways and transcription factor regulons to score potential influences, thus providing mechanistic insights beyond mere pair detection. This method highlights how sender cells modulate receiver phenotypes, for instance, in developmental signaling cascades. To incorporate spatial context, methods adapt LR inference by weighting interactions according to cellular proximity in data, such as from 10x Visium platforms, where spot-level resolution approximates cell distances. Tools like Spacia model these spatial constraints using Bayesian frameworks to prioritize nearby cell pairs, enhancing the accuracy of communication networks in tissue microenvironments. This spatial awareness refines predictions from non-spatial scRNA-seq expression data. Recent progress as of 2025 includes enhanced tools like NICHES for single-cell resolution of niche signaling. Inferred networks are often visualized using Circos plots, which depict cell-type interactions as circular chord diagrams, with link thickness representing interaction strength or significance to facilitate intuitive exploration of complex communication landscapes.

Applications in Biology and Medicine

Developmental biology

Single-cell analysis has revolutionized the study of by enabling detailed mapping of embryogenesis, , and cell fate decisions at unprecedented resolution. In embryos, single-cell RNA sequencing (scRNA-seq) of over 100,000 cells from embryonic day 6.5 to 8.5 revealed the dynamic emergence of cell types during and early , identifying 25 distinct cell types including , neuromesodermal, and visceral lineages. This atlas highlighted key transcriptional programs driving formation, with primitive endoderm and epiblast cells transitioning into and progenitors. A pivotal finding from this work is the role of WNT signaling in specification, where WNT pathway activation in the posterior epiblast promotes formation and mesendoderm differentiation, as evidenced by enriched WNT target genes in nascent mesodermal cells. In differentiation, single-cell approaches have elucidated trajectories from induced pluripotent stem cells (iPSCs) to specialized lineages, such as s. For instance, scRNA-seq of differentiation from human iPSCs profiled thousands of cells across 100 days, reconstructing pseudotemporal trajectories that capture progressive maturation stages from neural progenitors to functional s, revealing regulatory networks involving FOXA2 and LMX1A transcription factors. These analyses demonstrate how single-cell resolution uncovers heterogeneity in differentiation efficiency and identifies bottlenecks, such as variable expression of neuronal markers like and , informing optimized protocols for . Organoids, three-dimensional cultures derived from cells, serve as models for , where single-cell analysis exposes cellular heterogeneity. In brain organoids generated from iPSCs, scRNA-seq integrated datasets from multiple protocols over cell types mimicking fetal brain development, including radial glia, intermediate progenitors, and neurons, but also revealed off-target populations like retinal cells and non-neural lineages contributing to variability across organoids. This heterogeneity underscores challenges in recapitulating organogenesis and highlights the need for refined culture conditions to enhance fidelity. Comparative single-cell studies across s provide evolutionary insights into conserved and divergent developmental mechanisms. A scRNA-seq atlas of , encompassing 91,232 cells from embryonic days 11.5 to 15, identified conserved gene modules for epiblast progression and allocation similar to and , yet revealed pig-specific expansions in extraembryonic , suggesting adaptive divergences in and body axis formation. Such cross-species analyses, often leveraging tools, illuminate how core pathways like WNT and are repurposed evolutionarily to shape embryogenesis.

Cancer research

Single-cell analysis has profoundly advanced by elucidating tumor heterogeneity at unprecedented resolution, enabling the dissection of cellular states within tumors that drive , progression, and therapeutic responses. Techniques such as single-cell RNA sequencing (scRNA-seq) and single-nucleus sequencing have revealed diverse subpopulations of cancer cells and their interactions within the tumor , shifting the from bulk tumor analyses to individualized cellular portraits. This approach has been instrumental in identifying mechanisms of tumor evolution, immune evasion, and , informing precision oncology strategies across various cancer types. In tumor microenvironments, single-cell analysis has illuminated the composition and dynamics of immune infiltration, particularly in immunogenic cancers like . For instance, scRNA-seq applied to metastatic samples from 19 patients profiled over 4,600 cells, identifying distinct malignant cell states associated with resistance to targeted therapies and revealing co-occurrence of immune cells such as T cells and macrophages that modulate the suppressive milieu. This work demonstrated how cells adopt a program mimicking resistant neural crest-like states, influencing immune cell recruitment and function within the tumor niche. Such insights have guided designs by highlighting actionable immune checkpoints in specific microenvironmental contexts. Clonal in cancers, driven by genetic alterations like copy number variations (CNVs), has been tracked using single-nucleus sequencing to overcome challenges posed by formalin-fixed tissues. In , analysis of 66 tumor cells from a primary pleomorphic lobular revealed early aneuploid rearrangements that stabilized during clonal expansion, with CNV profiles showing branched patterns where subclones acquired distinct genomic scars. This achieved high coverage (91% breadth) and quantified intratumor heterogeneity, showing that major clones dominated the tumor mass while minor subclones harbored potential drivers of . These findings underscore how single-cell genomic profiling reconstructs phylogenetic trees of tumor progression, aiding in the identification of evolutionarily stable targets for intervention. Therapy resistance, a major barrier in , has been probed through single-cell approaches that uncover rare drug-tolerant persister (DTP) cells responsible for . In non-small cell lung cancer with EGFR mutations, scRNA-seq of patient-derived models under treatment identified DTP subpopulations with activated pathways like YAP/TAZ and partial epithelial-mesenchymal transition signatures, persisting in a slow-cycling state post-therapy. These cells, comprising less than 1% of the population pre-treatment, exhibited transcriptomic plasticity influenced by the microenvironment, providing molecular markers for eradicating residual disease. Liquid biopsies leveraging single-cell analysis of circulating tumor cells (CTCs) offer non-invasive windows into systemic tumor dissemination and evolution. High-dimensional profiling of CTCs from patients demonstrated heterogeneous expression of epithelial and mesenchymal markers across individual cells, correlating with metastatic potential and differing from profiles. This approach has enabled real-time monitoring of clonal dynamics, such as emergence of therapy-resistant variants during , by isolating and sequencing viable CTCs from samples. By capturing rare events like epithelial-to-mesenchymal transitions, CTC single-cell analysis supports personalized tracking of tumor spread and response in advanced . The integration of single-cell data with large-scale resources like (TCGA) has culminated in pan-cancer atlases that delineate shared and cancer-specific cellular architectures. Efforts such as the 2018 TCGA immune landscape analysis across 33 cancer types revealed conserved immune exhaustion programs and identified pan-cancer regulatory networks involving myeloid cells and T lymphocytes. This atlas highlighted subtype-specific vulnerabilities, such as heightened in hypoxic niches common to several epithelial cancers, facilitating cross-tumor therapeutic hypotheses. By benchmarking single-cell heterogeneity against TCGA-derived genomic landscapes, these analyses have accelerated the discovery of universal biomarkers for responsiveness.

Immunology and infectious diseases

Single-cell analysis has revolutionized the study of immune responses in and infectious diseases by enabling the dissection of cellular heterogeneity, clonal dynamics, and functional states at unprecedented resolution. Techniques such as single-cell sequencing (scRNA-seq) and immune repertoire sequencing have revealed the diversity of T cell receptors (TCRs) and B cell receptors (BCRs), shedding light on adaptive immunity during infections and vaccinations. In infectious diseases, these methods have illuminated pathogen-specific responses, including hyperinflammatory states, while in , they highlight aberrant clonal expansions that drive pathology. Repertoire sequencing has been instrumental in characterizing TCR and BCR diversity following vaccination, providing insights into antigen-specific immune memory. For instance, single-cell profiling of T and B cell repertoires after mRNA vaccination demonstrated enrichment of spike protein-specific B cells and polyfunctional + T cells, with expanded clones showing high-affinity binding and . This approach has informed vaccine efficacy by quantifying repertoire convergence, where repeated immunizations lead to focused against conserved epitopes, as observed in studies of pertussis vaccines where predictable BCR heavy chain motifs emerged post-booster. Such analyses underscore how single-cell repertoire data can guide the design of next-generation vaccines targeting broad immune coverage. In infection dynamics, scRNA-seq has mapped the temporal evolution of immune responses, particularly during viral outbreaks like in 2020. A landmark study using scRNA-seq and scTCR-seq on fluid from patients distinguished mild from critical cases, revealing hyperinflammation driven by dysregulated monocyte-derived macrophages and exhausted T cells in severe disease, with over 10-fold higher pro-inflammatory signatures in critical patients. These findings highlighted clonal T cell exhaustion linked to persistent exposure, contributing to cytokine storms and informing therapeutic strategies to mitigate . In , single-cell analysis has uncovered clonal expansions underlying diseases like systemic lupus erythematosus (SLE). scRNA-seq of peripheral blood mononuclear cells from SLE patients identified expanded cytotoxic GZMH+ + T cell clones with heightened signatures, correlating with disease flares and tissue damage. Further, integrated single-cell multi-omics revealed that neoself-antigens trigger autoreactive TCR clonal bursts in SLE, distinguishing pathogenic from protective clones based on epigenetic accessibility and transcriptional profiles. This clonal focus has pinpointed therapeutic windows, such as depleting expanded autoreactive subsets to restore . For vaccine design, single-cell has traced B cell maturation pathways, optimizing immunogen strategies. Analysis of human tonsillar B cells post-vaccination-like stimulation showed that germinal center entry depends on early metabolic reprogramming and IRF4 upregulation, with trajectories predicting antibody affinity maturation rates up to 100-fold improvement in high-responders. These insights have advanced rational vaccine development, such as HIV immunogens that mimic natural B cell lineage trees to overcome maturation roadblocks. A key advancement is the delineation of anergic states in exhausted T cells, where single-cell profiling distinguishes hyporesponsiveness from full exhaustion. scRNA-seq identified transitional anergic + T cells in chronic settings, characterized by low IL-2 production and reversible Egr2 expression, contrasting with irreversible PD-1+ exhausted states. This nuance has implications for , as targeting anergic progenitors can reinvigorate responses in persistent infections without broad . As of 2025, single-cell analysis in has incorporated models to predict variant-specific T cell responses in emerging infectious diseases, enhancing vaccine strategies against evolving pathogens like variants.

References

  1. [1]
    Single-cell analysis: Advances and future perspectives - PMC - NIH
    Single-cell analysis allows the study of cell-to-cell variation within a cell population (organ, tissue, and cell culture).
  2. [2]
    Best practices for single-cell analysis across modalities - Nature
    Mar 31, 2023 · We summarize independent benchmarking studies of unimodal and multimodal single-cell analysis across modalities to suggest comprehensive best-practice ...
  3. [3]
    Single-cell technologies: From research to application - PMC
    Oct 18, 2022 · The earliest single-cell isolation was based on manual pipetting to analyze only a handful of cells.
  4. [4]
    Review The Technology and Biology of Single-Cell RNA Sequencing
    May 21, 2015 · The first transcriptomes generated via single-cell RNA-sequencing (scRNA-seq) were published in 2009 (Tang et al., 2009), only 2 years after the ...
  5. [5]
    Single-cell sequencing techniques from individual to multiomics ...
    Sep 15, 2020 · We review single-cell sequencing techniques for individual and multiomics profiling in single cells. We mainly describe single-cell genomic, epigenomic, and ...Missing: modalities | Show results with:modalities
  6. [6]
    Best practices for single-cell analysis across modalities - PMC - NIH
    Mar 31, 2023 · Recent advances in single-cell technologies have enabled high-throughput molecular profiling of cells across modalities and locations.
  7. [7]
  8. [8]
  9. [9]
    The life and work of Rudolf Virchow 1821–1902: “Cell theory ... - NIH
    Aug 19, 2016 · Virchow was the first to correctly link the origin of cancers from otherwise normal cells, believing that cancer is caused by severe irritation in the tissues.Missing: single history
  10. [10]
    Flow cytometry: retrospective, fundamentals and recent ... - NIH
    In 1972, Bonner et al. developed a new system named fluorescence activatedcell sorting (FACS) which purified sub-populations (Bonner et al. 1972) and was ...
  11. [11]
    Speed read: Recording channels - NobelPrize.org
    Sep 16, 2009 · Erwin Neher and Bert Sakmann's patch-clamp technique is based on an exquisitely simple idea. An extremely fine glass pipette with a very ...
  12. [12]
    Novel method for studying mRNA phenotypes in single or ... - PubMed
    With this method we have detected actin mRNA from a single cell, or less than 100 cRNA molecules, and have quantified differences in RNA concentrations of less ...Missing: first | Show results with:first
  13. [13]
    Microfluidic single-cell analysis of intracellular compounds - NIH
    Microfluidic devices have been an intensive field of research in the analytical and bioanalytical community since the 1990s and biologically relevant ...
  14. [14]
    Single-cell technologies: From research to application - ScienceDirect
    Nov 8, 2022 · A key technological milestone was the release of several massively parallel DNA sequencing (next-generation sequencing) platforms in 2005 and ...
  15. [15]
    mRNA-Seq whole-transcriptome analysis of a single cell - PubMed
    mRNA-Seq whole-transcriptome analysis of a single cell. Nat Methods. 2009 May;6(5):377-82. doi: 10.1038/nmeth.1315. Epub 2009 Apr 6. Authors. Fuchou Tang ...
  16. [16]
    New Commercial Developments in Single-cell Sequencing
    Sep 12, 2023 · We will briefly look back over the historical development of single-cell sequencing before summarising the latest commercial developments.
  17. [17]
    CRISPR screening in hematology research: from bulk to single-cell ...
    Oct 24, 2023 · In this review, we discuss the use of bulk CRISPR screening in hematology research, as well as the emergence of single-cell CRISPR screening and its added ...Crispr Screens · Single-Cell Crispr Screening · Grna Capture Approaches<|separator|>
  18. [18]
    Single-cell analysis and sorting using droplet-based microfluidics
    We present a droplet-based microfluidics protocol for high-throughput analysis and sorting of single cells. Compartmentalization of single cells in droplets ...
  19. [19]
    Dissociation of solid tumor tissues with cold active protease for ...
    Oct 17, 2019 · We demonstrate that both digestion time and collagenase contribute to the transcriptomic stress response in single cancer cells. Therefore ...
  20. [20]
    Tumor dissociation of highly viable cell suspensions for single-cell ...
    Dec 17, 2021 · This protocol describes a technique for digestion and single-cell isolation from mouse mammary tumors to achieve ∼90% of viable cells.Step-By-Step Method Details · Tissue Harvesting · Tissue Digestion<|separator|>
  21. [21]
    Considerations for high-yield, high-throughput cell enrichment - Nature
    Jan 18, 2019 · In FACS, the high fluid flow rates coupled with small nozzle diameters have the potential to reduce cellular viability due to high shear ...
  22. [22]
    Flow Cytometry vs Cell Sorting | Biocompare
    Jun 11, 2019 · “To maintain viability, rates in a single stream are typically limited to 10–20,000 cells/second,” he says. “So to scale up sort rates, it is ...Missing: per | Show results with:per
  23. [23]
    BD Rhapsody™ HT Xpress System - BD Biosciences
    Expanded Throughput · Million-cell studies now possible · Capable of capturing more than 640,000 cells per cartridge · Up to 80% cell capture rate (for certain ...
  24. [24]
    Laser capture microscopy coupled with Smart-seq2 for precise ...
    Jul 8, 2016 · Here we report a robust and highly efficient strategy for LCM coupled with full-length mRNA-sequencing (LCM-seq) developed for single-cell transcriptomics.
  25. [25]
    High-resolution particle separation by inertial focusing in ... - Nature
    Jul 6, 2021 · Within microfluidics, inertial focusing provides the means for a precise manipulation of particles, allowing for tasks such as focusing, ...
  26. [26]
    Preparation of Single-Cell RNA-Seq Libraries for Next Generation ...
    Start with high-quality RNA; use RNA with RNA integrity number >8; begin with a single RNA sample and work quickly; work in an RNase-free environment. PCR ...
  27. [27]
    Review of Microfluidic Methods for Cellular Lysis - MDPI
    In the following, the five most commonly used methods (chemical, mechanical, thermal, electrical methods, and laser lysis) are described using various ...
  28. [28]
    Methanol fixation is the method of choice for droplet-based single ...
    May 15, 2023 · Taken together, our results show that methanol fixation is the method of choice for performing droplet-based single-cell transcriptomics ...
  29. [29]
    Formaldehyde Fixation Helps Preserve the Proteome State during ...
    Feb 3, 2025 · We demonstrate that short-term FA fixation does not substantially affect protein recovery, even without heating and strong detergents, and maintains analytical ...
  30. [30]
    DMSO cryopreservation is the method of choice to preserve cells for ...
    Jul 23, 2019 · In this study, we compared DMSO cryopreservation, methanol fixation as well as CellCover reagent to preserve cells for scRNA-seq analysis using ...
  31. [31]
    7 Things to Consider When Preparing a Cell Suspension for scRNA ...
    Jun 29, 2023 · Enzymatic dissociation involves using enzymes to break down specific chemical bonds. Common enzymes are TrypLE, collagenase, dispase, and ...
  32. [32]
    Can the H&E staining process have a negative impact on RNA ...
    Nov 3, 2025 · In order to ensure the best possible results, we recommend using tissue with a RIN (RNA Integrity Number) score of 7 or greater. To evaluate ...
  33. [33]
    Non-invasive measurement of mRNA decay reveals translation ...
    Sep 7, 2018 · Whereas individual steps of mRNA degradation have been determined ... half-life parameters from the decay measurements. The overall ...
  34. [34]
  35. [35]
  36. [36]
    Single Cell Metabolomics Using Mass Spectrometry - NIH
    Mass spectrometry (MS) based techniques are gaining popularity for metabolomics research due to their high sensitivity, wide detection range, and capability ...Missing: per | Show results with:per
  37. [37]
    Single-Cell Metabolomics-Based Strategy for Studying the ...
    Mar 1, 2023 · (5) Tumor cells undergo a series of metabolic reprogramming to meet their need for rapid proliferation (e.g., the Warburg effect, which depends ...
  38. [38]
    The Development of Single-Cell Metabolism and Its Role ... - Frontiers
    Single-cell metabolomics identifies the metabolites of single cells in different states by mass spectrometry, and captures the molecular biological information ...
  39. [39]
    Single-cell metabolic profiling reveals subgroups of primary human ...
    Oct 17, 2023 · We identify four functional subgroups of hepatocytes responding differently to drug challenge and fatty acid accumulation.Missing: metabolomics | Show results with:metabolomics
  40. [40]
    Single-cell metabolomics reveals the metabolic heterogeneity ...
    Nov 8, 2021 · The study developed RespectM to detect over 700 metabolites in single microbial cells, revealing metabolic grouping based on genetic and ...
  41. [41]
    Neuronal progenitor cells-based metabolomics study reveals ...
    Oct 29, 2023 · Neuronal progenitor cells-based metabolomics study reveals dysregulated lipid metabolism and identifies putative biomarkers for CLN6 disease.Missing: single- | Show results with:single-
  42. [42]
    Metabolic profiling of single cells by exploiting NADH and FAD ...
    It was found that immune cells activated with specific stimuli increase NAD(P)H and that different purified immune cells exhibit different redox ratios [23].Missing: metabolomics NADP
  43. [43]
    Single cell metabolism: current and future trends - PubMed Central
    Single cell metabolomics is an emerging and rapidly developing field that complements developments in single cell analysis by genomics and proteomics.Missing: pmol | Show results with:pmol
  44. [44]
    High-throughput single-cell quantification of hundreds of proteins ...
    Sep 22, 2021 · In this article, we demonstrate the capabilities of Infinity Flow, a low-cost methodology and computational toolset for high-dimensional single- ...Missing: seminal | Show results with:seminal<|separator|>
  45. [45]
  46. [46]
  47. [47]
  48. [48]
  49. [49]
  50. [50]
    Trends in Mass Spectrometry-Based Single-Cell Proteomics
    Mass spectrometry (MS)-based single-cell proteomics (SCP) enables the study of protein expression at the individual cell level across thousands of proteins, ...Cell Isolation · Sample Preparation · Separation · MS Data Acquisition<|separator|>
  51. [51]
    High-throughput and high-efficiency sample preparation for single ...
    Oct 29, 2021 · We describe a nested nanoPOTS (N2) chip to improve protein recovery, operation robustness, and processing throughput for isobaric-labeling-based scProteomics ...Results · Cell Typing With... · Methods
  52. [52]
    Lipid imaging with time-of-flight secondary ion mass spectrometry ...
    Fundamental advances in secondary ion mass spectrometry (SIMS) now allow for the examination and characterization of lipids directly from biological ...
  53. [53]
    High-Resolution Secondary Ion Mass Spectrometry Analysis of Cell ...
    Dec 19, 2019 · This Feature describes the use a Cameca NanoSIMS instrument for directly imaging specific lipid and protein species in the plasma membranes of mammalian cells
  54. [54]
    Ultra-fast label-free quantification & proteome coverage
    Feb 1, 2024 · This dataset demonstrates that the Orbitrap Astral MS instrument is particularly well suited for single-cell proteomics research. To ...
  55. [55]
    Quantitative Label-Free Single-Cell Proteomics on Orbitrap Astral MS
    We investigate the capacity of the enhanced sensitivity of the Orbitrap Astral mass spectrometer to facilitate deeper proteome profiles from low-input to ...
  56. [56]
    [PDF] Making Single-cell Proteomics Mainstream - Thermo Fisher Scientific
    Single-cell proteomics seeks to complement transcriptome and genome data by generating comparative protein expression profiles of individual cells. The field ...Missing: seminal | Show results with:seminal
  57. [57]
    Unbiased spatial proteomics with single-cell resolution in tissues
    Jun 16, 2022 · Furthermore, MALDI is generally less quantitative and has less dynamic range than electrospray, which has led to its displacement in proteomics ...<|control11|><|separator|>
  58. [58]
    Comparing DESI-MSI and MALDI-MSI Mediated Spatial ...
    Jul 18, 2022 · MALDI is one of the most commonly used ionization methods linked to MSI as it provides a favorable balance between sample preparation, chemical ...
  59. [59]
    Computational strategies for single-cell multi-omics integration
    In this review, we first introduce recent developments in single-cell multi-omics in general and then focus on the available data integration strategies.Computational Strategies For... · 3. Single-Cell Multi-Omics... · 4. Computational Tools For...
  60. [60]
    Bi-order multimodal integration of single-cell data | Genome Biology
    May 9, 2022 · We present a novel mathematical solution named bi-order canonical correlation analysis (bi-CCA), which extends the widely used CCA approach.
  61. [61]
    Simultaneous trimodal single-cell measurement of transcripts ... - eLife
    Apr 9, 2021 · A trimodal assay that simultaneously measures transcriptomics (scRNA-seq), epitopes, and chromatin accessibility (scATAC-seq) from thousands of single cells, ...Missing: 2024 | Show results with:2024
  62. [62]
    Benchmarking multi-omics integration algorithms across single-cell ...
    In this paper, we benchmarked 12 multi-omics integration methods on three integration tasks via qualitative visualization and quantitative metrics.
  63. [63]
  64. [64]
    Page not found | Nature
    - **Status**: Insufficient relevant content
  65. [65]
    The technological landscape and applications of single-cell multi ...
    Jun 6, 2023 · Single-cell multi-omics technologies and methods characterize cell states and activities by simultaneously integrating various single-modality omics methods.
  66. [66]
    An introduction to spatial transcriptomics for biomedical research
    Jun 27, 2022 · Its commercialization by 10X Genomics, termed Visium, improved this to hexagonal 55 μm resolution, with plans for 2 μm capture areas in 2022.
  67. [67]
    Spatially resolved transcriptomics: a comprehensive review of their ...
    In this review, we introduce recent progresses and challenges on SRT from the perspectives of technologies and bioinformatic tools, as well as the ...
  68. [68]
    Slide-seq: A scalable technology for measuring genome-wide ...
    Mar 29, 2019 · Slide-seq provides a scalable method for obtaining spatially resolved gene expression data at resolutions comparable to the sizes of individual cells.
  69. [69]
    Spatially resolved, highly multiplexed RNA profiling in single cells
    We report multiplexed error-robust FISH (MERFISH), a single-molecule imaging method that allows thousands of RNA species to be imaged in single cells.
  70. [70]
  71. [71]
    Deciphering spatial domains from spatial multi-omics with SpatialGlue
    Jun 21, 2024 · Sequencing-based techniques include DBiT-seq1, spatial-CITE-seq2, spatial assay for transposase-accessible chromatin and RNA using sequencing ( ...
  72. [72]
    High-plex protein and whole transcriptome co-mapping at cellular ...
    Feb 23, 2023 · The scCITE-seq dataset was obtained from a published study. It was first cleaned by removing cells with fewer than ten total ADT UMIs and ...
  73. [73]
    Tissue characterization at an enhanced resolution across spatial ...
    Aug 2, 2024 · Pioneering technologies include spatial-CITE-seq (proteins and genes) and spatial ATAC-RNA-seq (chromatin accessibility and genes). The ...
  74. [74]
    Spatial proteomics of single cells and organelles on tissue ... - PubMed
    Oct 30, 2024 · Hydrogel-based tissue expansion combined with mass spectrometry (MS) offers an emerging spatial proteomics approach.Missing: XeGel metabolomics
  75. [75]
    Spatial proteomics of single cells and organelles on tissue slides ...
    Oct 30, 2024 · Hydrogel-based tissue expansion combined with mass spectrometry (MS) offers an emerging spatial proteomics approach.Missing: XeGel | Show results with:XeGel
  76. [76]
    Metabolic imaging at the nanoscale with Multi-Isotope Imaging Mass ...
    In this Current Protocol we introduce metabolic imaging by NanoSIMS, by focusing on two specific applications: quantifying protein turnover in single organelles ...
  77. [77]
    Biological explorations with nanoscale secondary ion mass ...
    With a spatial resolution of less than 50nm, the NanoSIMS instrument can easily image individual cells and resolve many subcellular structures. The NanoSIMS can ...
  78. [78]
    Thor: a platform for cell-level investigation of spatial transcriptomics ...
    Aug 5, 2025 · Thor and iStar enhanced spatial resolution to (near) cell resolution, iStar at times introduced artifacts, including excessive fusion, for ...<|separator|>
  79. [79]
    Spatiotemporal omics for biology and medicine - Cell Press
    Aug 22, 2024 · A significant advancement for resolution enhancement is the use of nano-array sequencing chips as capture carriers, which can achieve nanometer- ...
  80. [80]
    Spatial multi-omics: deciphering technological landscape of ...
    Aug 24, 2024 · The emergence of spatial multi-omics has helped address the limitations of single-cell sequencing, which often leads to the loss of spatial ...
  81. [81]
    SCANPY: large-scale single-cell gene expression data analysis
    Feb 6, 2018 · Scanpy is a scalable toolkit for analyzing single-cell gene expression data. It includes methods for preprocessing, visualization, clustering, pseudotime and ...
  82. [82]
    Common Considerations for Quality Control Filters for Single Cell ...
    Oct 27, 2022 · Here, we discuss the most common metrics and methods used for QC and filtering of cell barcodes in single cell RNA-seq data for gene expression analysis.
  83. [83]
    Systematic determination of the mitochondrial proportion in human ...
    An important QC metric is the mitochondrial proportion (mtDNA%), which is used as a threshold to filter out low-quality cells.
  84. [84]
    Normalization and variance stabilization of single-cell RNA-seq data ...
    Dec 23, 2019 · We present a modeling framework for the normalization and variance stabilization of molecular count data from scRNA-seq experiments.
  85. [85]
    Embracing the dropouts in single-cell RNA-seq analysis - Nature
    Mar 3, 2020 · The droplet-based 10X dataset contained count data of 70,118 cells for the same 23,433 genes, with an overall dropout rate of 93%. The Tabula ...
  86. [86]
    Accuracy, robustness and scalability of dimensionality reduction ...
    Dec 10, 2019 · Dimensionality reduction is an indispensable analytic component for many areas of single-cell RNA sequencing (scRNA-seq) data analysis.
  87. [87]
    A Comparison for Dimensionality Reduction Methods of Single-Cell ...
    We developed a strategy to evaluate the stability, accuracy, and computing cost of 10 dimensionality reduction methods using 30 simulation datasets and five ...Introduction · Materials and Methods · Results · Discussion
  88. [88]
    Dimensionality reduction for visualizing single-cell data using UMAP
    Dec 3, 2018 · A benchmarking analysis on single-cell RNA-seq and mass cytometry data reveals the best-performing technique for dimensionality reduction.
  89. [89]
    Benchmarking clustering algorithms on estimating the number of cell ...
    Feb 8, 2022 · Review of single-cell RNA-seq data clustering ... A systematic performance evaluation of clustering methods for single-cell RNA-seq data.
  90. [90]
  91. [91]
  92. [92]
    Accelerated dimensionality reduction of single-cell RNA sequencing ...
    Principal component analysis (PCA) is a widely used dimensionality reduction technique, but it has been criticized as being poorly suited to the sparse count ...Missing: review | Show results with:review
  93. [93]
    Driving Toward Billion-Cell Analysis and Biological Breakthroughs ...
    Jun 12, 2025 · Automatic spilling of data to host memory, enabling large-scale single-cell analysis across multiple GPU architectures. Dimensionality reduction ...Missing: scalability | Show results with:scalability
  94. [94]
    Page not found | Nature
    - **Insufficient relevant content**: The requested URL (https://www.nature.com/articles/s41592-018-0152-7) returns a "Page not found" error, indicating the page is unavailable or does not exist.
  95. [95]
    Slingshot: cell lineage and pseudotime inference for single-cell ...
    Jun 19, 2018 · We introduce Slingshot, a novel method for inferring cell lineages and pseudotimes from single-cell gene expression data.
  96. [96]
    PAGA: graph abstraction reconciles clustering with trajectory ...
    Finally, we show how PAGA abstracts transition graphs, for instance, from RNA velocity and compare to previous trajectory-inference algorithms.
  97. [97]
    [PDF] CellPhoneDB v5: inferring cell-cell communication from single-cell ...
    A total of 2,508 interactions have been classified as directional. (ligand-receptor), 242 as adhesion interaction and the remaining 161 as either ligand-ligand,.
  98. [98]
    Deciphering cell–cell interactions and communication from gene ...
    Nov 9, 2020 · One extensively used database contains ~2,400 human ligand–receptor pairs, obtained from multiple databases and literature curation.
  99. [99]
    CellPhoneDB: inferring cell-cell communication from ... - PubMed
    Here, we outline the structure and content of our repository, provide procedures for inferring cell-cell communication networks from single-cell RNA sequencing ...
  100. [100]
    NicheNet: modeling intercellular communication by linking ligands ...
    A method that predicts ligand-target links between interacting cells by combining their expression data with prior knowledge on signaling and gene regulatory ...
  101. [101]
    Evaluation of cell-cell interaction methods by integrating single-cell ...
    Oct 17, 2022 · Our study presents a comprehensive evaluation of cell-cell interaction tools for scRNA-seq. CellChat, CellPhoneDB, NicheNet, and ICELLNET show overall better ...
  102. [102]
    A single-cell atlas of pig gastrulation as a resource for comparative ...
    Jun 18, 2024 · Here we present a single-cell transcriptomic atlas of pig gastrulation, revealing cell-fate emergence dynamics, as well as conserved and divergent gene ...Missing: evolution | Show results with:evolution
  103. [103]
    Single‐cell proteomics by mass spectrometry: Advances and ...
    May 10, 2024 · This review aims to summarize application of SCP in cancer research, while revealing current development progress of SCP technology.2 Importance Of Scp In... · 3.1. 2 Cell Lysis And... · 5 Challenges And Future...
  104. [104]
    Single Cell Profiling of Circulating Tumor Cells
    Here we demonstrate feasibility of performing high dimensional single CTC profiling, providing early insight into CTC heterogeneity and allowing comparisons to ...Missing: seminal | Show results with:seminal