Fact-checked by Grok 2 weeks ago

Spatial transcriptomics

Spatial transcriptomics refers to a collection of technologies that enable the measurement of profiles while preserving the spatial context within intact tissues, allowing for the mapping of transcripts at cellular or subcellular resolution. These methods bridge the limitations of traditional bulk transcriptomics, which averages signals across populations, and single-cell sequencing, which disrupts spatial information, by integrating high-throughput molecular profiling with histological architecture. By capturing the location-specific dynamics of thousands of genes, spatial transcriptomics reveals how cellular identity, interactions, and functions are orchestrated in complex biological environments. The foundations of spatial transcriptomics lie in early * techniques, first demonstrated in 1969 with RNA-DNA hybrid detection for cytological analysis and advanced in 1987 for localizing genomic regulatory elements in model organisms like . The modern field coalesced in the , propelled by advances in next-generation sequencing, , and fluorescence microscopy; key milestones include the introduction of seqFISH in 2014 for multiplexed single-cell profiling and the 2016 development of array-based spatial transcriptomics, which laid the groundwork for commercial platforms like Visium. Since then, the technology has evolved rapidly, achieving genome-wide coverage and near-single-cell resolution, with over 100 methods published by 2022. Spatial transcriptomics technologies are broadly divided into imaging-based and sequencing-based categories. Imaging-based approaches, such as MERFISH and seqFISH+, employ multiplexed to detect hundreds to thousands of targeted transcripts with single-molecule precision, often enhanced by for subcellular detail. Sequencing-based methods, including Slide-seq, Stereo-seq, and Visium, use barcoded arrays or beads to capture mRNA from tissue sections, followed by high-throughput sequencing to reconstruct spatial maps at resolutions ranging from 0.5 μm to 100 μm. These complementary strategies have enabled scalable profiling of entire organs, such as the or developing heart, integrating with multi-omics data for deeper insights. In biology and medicine, spatial transcriptomics has transformed the study of tissue organization, cell-cell interactions, and developmental processes, such as mapping 23 cell types and 264 subtypes in the macaque brain or tracing clonal evolution in tumors. Applications extend to disease research, elucidating mechanisms in cancer microenvironments, immune responses, and organ injury— for instance, identifying novel cell states in kidney disease models—while supporting precision medicine through spatial atlases like the Human Tumor Atlas Network. Despite challenges in resolution, throughput, and data integration, ongoing innovations continue to expand its utility across neuroscience, oncology, and regenerative biology.

Fundamentals

Definition and scope

Spatial transcriptomics refers to a suite of technologies that profile the —the complete set of transcripts in a or —while preserving the spatial organization of molecules within intact biological samples. This approach enables the mapping of patterns at defined locations, revealing how transcriptional activity correlates with tissue architecture and cellular positioning. Pioneered in seminal work demonstrating spatial barcoding on tissue sections, it allows quantitative analysis of thousands of genes across histological contexts. The scope of spatial transcriptomics spans a spectrum of spatial resolutions, from low-resolution methods that aggregate transcripts in spots (typically 50–100 μm in ) to high-resolution techniques achieving single-cell or subcellular precision (down to <1 μm). These methods maintain integrity during profiling, facilitating studies in two-dimensional sections or three-dimensional structures, and encompass applications from basic cellular interactions to complex multicellular dynamics. Key concepts include spatial context, which encompasses cellular neighborhoods, expression gradients, and microenvironments that drive phenotypic diversity and functional regulation. Emerging in the 2010s, spatial transcriptomics addresses gaps in prior transcriptomic paradigms: bulk RNA sequencing yields averaged expression without positional detail, while single-cell RNA sequencing captures individual cell identities but dissociates them from their native tissue surroundings.

Underlying principles

Spatial transcriptomics relies on the principle of spatial preservation, which begins with the preparation of tissue sections to maintain the integrity of molecular content and anatomical structure. Tissues are typically sectioned using cryosectioning for fresh frozen samples or formalin-fixed paraffin-embedded (FFPE) processing for archived specimens, producing thin slices (e.g., 10 μm thick) that are mounted onto specialized arrays or slides. This is followed by in situ capture, labeling, or sequencing techniques that bind and tag RNA molecules directly within the tissue context, ensuring that positional information is retained throughout the workflow. For instance, polyadenylated mRNA is captured via oligo(dT) primers attached to spatially barcoded substrates, preventing dissociation and preserving the original coordinates of transcripts relative to the tissue architecture. Cryopreservation is preferred for optimal RNA integrity in methods like and , while FFPE compatibility allows analysis of clinically derived samples but requires additional deparaffinization and antigen retrieval steps to mitigate RNA degradation. Readout strategies in spatial transcriptomics convert captured RNA into quantifiable signals while encoding spatial origin. Reverse transcription is commonly employed to synthesize complementary DNA (cDNA) from mRNA templates directly on the tissue or array, followed by spatial barcoding where unique oligonucleotide sequences (spatial barcodes) are ligated or hybridized to indicate the precise location of each transcript. Amplification steps, such as PCR or rolling circle amplification (RCA), then enhance the signal for detection, enabling high-throughput sequencing or imaging readout. In sequencing-based approaches, barcoded cDNA libraries are generated and sequenced to produce count matrices, whereas imaging-based methods use fluorescent probes for direct visualization. These strategies ensure that gene expression data is demultiplexed and mapped back to two-dimensional coordinates, facilitating the reconstruction of spatial expression patterns. Resolution in spatial transcriptomics is determined by factors such as spot size, probe density, and noise mitigation techniques, which collectively define the granularity of positional information. Low-resolution methods often use array spots of 50-100 μm in diameter (e.g., 55 μm in 10x Genomics ), capturing transcripts from multiple cells (typically 1-10) within each bin. Higher resolution is achieved through denser probe arrays or subcellular targeting, such as 10 μm beads in or <1 μm in imaging methods like , where probe density allows single-cell or sub-cellular profiling. Signal-to-noise ratio is improved by incorporating unique molecular identifiers (), short random sequences that tag individual transcripts during capture, enabling deduplication and correction for amplification biases during data processing. A basic conceptual model for spatial gene expression represents the profile at any location as G(x,y) = f(\text{transcripts at coordinates } (x,y)), where f encompasses normalization for capture efficiency, sequencing depth, and other technical variables to yield calibrated expression values. This framework underpins downstream analyses, treating the tissue as a continuous field of transcript abundances mapped to Cartesian coordinates.

Comparison to other transcriptomic approaches

Spatial transcriptomics differs from bulk RNA sequencing (RNA-seq) by preserving spatial context within tissues, whereas bulk RNA-seq averages gene expression across entire cell populations, masking heterogeneity such as expression gradients in tumor cores versus edges. This averaging in bulk methods obscures subtle variations driven by positional cues, limiting insights into tissue architecture, while spatial approaches map transcripts to specific coordinates, revealing localized patterns like those in developmental gradients or disease microenvironments. For instance, bulk RNA-seq might detect overall elevated inflammatory markers in a tumor sample, but spatial transcriptomics can pinpoint their enrichment in immune-infiltrated regions. In contrast to single-cell RNA sequencing (scRNA-seq), which dissociates tissues into individual cells to profile thousands of transcripts per cell but loses neighbor relationships, spatial transcriptomics maintains tissue integrity to capture cell-cell interactions, such as ligand-receptor signaling pairs in the . scRNA-seq excels at identifying rare cell types and high-resolution cellular states (e.g., detecting 1,000–5,000 genes per cell), yet the dissociation process can introduce artifacts and eliminates spatial information essential for understanding functional neighborhoods. Spatial methods thus complement scRNA-seq by integrating positional data, enabling analyses of how adjacent cells coordinate responses, though they often aggregate signals from multiple cells per capture site. Compared to spatial proteomics, which targets proteins for direct functional readouts at ~1 μm resolution (e.g., via imaging mass cytometry targeting 40+ markers), spatial transcriptomics profiles RNA to assess dynamic gene expression across thousands of targets, offering higher throughput but indirect inference of protein levels due to post-transcriptional regulation. Spatial proteomics provides subcellular protein localization and reveals active pathways, yet it is limited by antibody specificity and lower multiplexing (hundreds to thousands of proteins) compared to the genome-wide RNA coverage in transcriptomics. Transcriptomic approaches thus prioritize transcriptional states and regulatory networks, while proteomic ones emphasize endpoint biology, with both benefiting from integration to bridge mRNA-to-protein discrepancies. Quantitative trade-offs in spatial transcriptomics include detecting 100–10,000 genes per spot (depending on method resolution and tissue complexity), lower than the 1,000–10,000 genes per cell in scRNA-seq, but with the added value of XY coordinates for mapping. Bulk RNA-seq achieves near-complete transcriptome coverage per sample but without resolution, and spatial proteomics typically profiles fewer analytes (10–1,000 proteins per region) at higher per-target costs.
ApproachResolutionThroughputApproximate Cost per SampleData Type
Bulk RNA-seqTissue/population level10,000–20,000 genes per sample$100–300Aggregated RNA
scRNA-seqSingle-cell (~1–10 µm)1,000–10,000 genes per cell; 10,000+ cells$500–2,000Individual cell RNA
Spatial TranscriptomicsSubcellular to spot (~1–100 µm)100–10,000 genes per spot; 1,000–10,000 spots$1,000–5,000Spatially mapped RNA
Spatial ProteomicsSubcellular (~1 µm)10–1,000 proteins per region; 100–1,000 regions$2,000–10,000Spatially mapped proteins

History

Early developments

The origins of spatial transcriptomics trace back to techniques developed in the 1990s that enabled targeted RNA isolation and visualization within tissue contexts. Laser capture microdissection (LCM), introduced in 1996, allowed for the precise procurement of specific cell populations from histological sections under microscopic visualization, facilitating downstream RNA analysis from defined spatial regions. This method addressed the challenge of isolating pure cellular material from heterogeneous tissues, laying groundwork for spatially informed molecular profiling. Parallel advancements in in situ hybridization (ISH) during the 1990s enhanced the direct detection of RNA molecules within intact tissues. Building on earlier foundations, researchers optimized ISH protocols for fluorescent labeling and simultaneous visualization of DNA, unspliced RNA precursors, and mature mRNAs, enabling spatial mapping of gene expression at the cellular level. However, these early ISH approaches were limited to low-plex detection, typically targeting one or a few transcripts per experiment, which restricted their scalability for genome-wide analysis. A key bridge in the mid-2010s toward higher-throughput spatial methods came with the development of tomography-based sequencing techniques. Tomo-seq, first described in 2014, combined cryosectioning of embryos with low-input RNA sequencing to generate genome-wide expression profiles along spatial axes, reconstructing 3D maps of transcript distribution in zebrafish embryos. Although tomo-seq emerged in the mid-2010s, it built on prior histological sectioning practices from the 2000s, providing a proof-of-concept for integrating spatial resolution with transcriptomic depth. The inaugural array-based spatial transcriptomics platform was introduced in 2016, marking a pivotal shift toward unbiased, high-plex profiling. This method, termed , involved placing tissue sections on slides arrayed with barcode-equipped capture probes, enabling the simultaneous quantification and spatial assignment of thousands of mRNAs directly from histological contexts. By preserving tissue architecture while generating genome-scale data, it overcame limitations of prior low-plex techniques and spurred the field's expansion.

Key milestones and recent progress

The development of spatial transcriptomics accelerated significantly from 2015 onward, with innovations in imaging and sequencing-based methods enabling higher resolution and multiplexing. In 2015, was introduced, allowing the simultaneous detection and spatial mapping of hundreds of RNA species in single cells with subcellular resolution. This built on earlier dissection techniques like but shifted toward in situ profiling without tissue disruption. By 2018, announced the , a commercial platform for targeted, high-plex RNA and protein profiling in regions of interest on intact tissue sections. The period from 2019 to 2022 saw broader commercialization and scalability improvements. In 2019, 10x Genomics launched Visium, a sequencing-based platform using spatially barcoded arrays to capture whole-transcriptome data from fresh-frozen tissue sections at near-cellular resolution. That same year, seqFISH+ advanced hybridization-based imaging by enabling error-corrected detection of over 10,000 genes in single cells with super-resolution capabilities. In 2022, Stereo-seq from BGI introduced DNA nanoball-patterned arrays for unbiased, high-throughput spatial transcriptomics at nanoscale resolution, facilitating large-scale tissue mapping. Recent progress from 2023 to 2025 has focused on subcellular precision and sequencing-independent approaches. In 2024, 10x Genomics released , enhancing the original platform with 2 µm resolution for whole-transcriptome imaging and sequencing integration, enabling detailed subcellular gene expression analysis. NanoString's , launched in 2022, has advanced sequencing-free, single-cell resolution profiling of over 18,000 RNA targets via multiplexed in situ hybridization, with key updates including the 2024 6,000-plex RNA assay and a June 2025 upgrade () that improves RNA detection efficiency by up to 2x. In October 2025, BGI introduced , enabling spatial mapping of total RNA on FFPE sections with single-cell resolution. Additionally, 2024 pipelines have integrated artificial intelligence for enhanced data analysis, such as deep learning models for domain detection and multimodal integration in platforms like .

Applications

Basic biological research

Spatial transcriptomics has revolutionized basic biological research by enabling the mapping of gene expression patterns within intact tissues, revealing how spatial organization influences cellular functions and interactions. In developmental biology, this technology allows researchers to trace gene gradients and regulatory networks during embryogenesis. For instance, tomo-seq has been applied to zebrafish embryos to generate high-resolution, genome-wide transcriptomic profiles along the three body axes, identifying spatially restricted expression domains that correspond to morphogen gradients and patterning cues essential for organ formation. This approach has elucidated how transcripts like those involved in left-right asymmetry are distributed, providing insights into the spatiotemporal dynamics of early development. In neuroscience, spatial transcriptomics facilitates the dissection of brain architecture at the molecular level, particularly in understanding layer-specific gene expression and neuronal circuits. Techniques such as have been used to profile thousands of genes in mouse visual cortex sections, revealing transcriptomic gradients across cortical layers and identifying molecular signatures of excitatory neuron subtypes that correlate with projection patterns. These findings highlight how spatial positioning dictates neuronal identity and connectivity, contributing to models of circuit assembly in healthy brain tissue. Similarly, in tumor microenvironments, spatial transcriptomics supports cell-type deconvolution to compare healthy and diseased states, uncovering shifts in stromal and immune cell compositions that alter tissue homeostasis without focusing on pathological outcomes. For example, integrated analyses have shown how fibroblasts and endothelial cells redistribute in response to neoplastic changes, informing basic principles of tissue remodeling. The "ecology of cells" within tissues is another key area where spatial transcriptomics infers intercellular communication through co-expression patterns of ligand-receptor pairs. In immune niches, such as lymphoid tissues, methods like NICHES analyze spatial data to quantify interactions between immune cells, revealing how proximity enhances signaling. This has demonstrated that spatial clustering promotes immune synapse formation, a fundamental mechanism in adaptive responses. Recent 2024 studies using Visium on 3D organoids, such as lung models, have extended this to engineered tissues, mapping transcriptomic gradients that mimic in vivo architecture and exposing how cell-cell relays shape organ-like structures. These applications build foundational knowledge that can inform clinical extensions in disease modeling.

Clinical and disease studies

Spatial transcriptomics has revolutionized clinical and disease studies by providing spatially resolved gene expression profiles that reveal tissue architecture, cellular interactions, and molecular signatures associated with pathology, thereby supporting diagnostic, prognostic, and therapeutic advancements. In oncology, spatial transcriptomics excels at profiling tumor heterogeneity and microenvironmental dynamics. Using , a 2024 study on 92 triple-negative breast cancer (TNBC) patients identified five molecular subtypes (basal-like, immunomodulatory, luminal androgen receptor, mesenchymal, and mixed) with distinct spatial patterns, such as larger proliferative patches in basal-like tumors and dispersed metabolic clusters in mesenchymal ones; intra-patient heterogeneity often involved 2–3 subtypes per sample, with immune-rich stroma at invasion fronts linked to improved distant relapse-free survival. In HER2-positive breast cancer, analysis of 36 tumor sections from eight patients uncovered intrapatient variations in ERBB2 expression across cancer clusters and highlighted type I interferon-associated macrophage-T cell interactions at tumor interfaces, underscoring spatial immune engagement. These findings delineate invasion fronts characterized by epithelial-mesenchymal transition and immune infiltration, informing targeted interventions like PARP inhibitors for high-metabolism regions. In immunology, spatial transcriptomics maps immune cell infiltration and activation in autoimmune diseases, aiding in understanding therapeutic resistance. The , applied to rheumatoid arthritis synovial tissues from responders and non-responders, revealed elevated fibroblast activation protein in the deep sublining layer of non-responders; spatial transcriptomics methods including have identified CXCL12-CXCR4-mediated B-T cell interactions driving inflammation. In systemic lupus erythematosus nephritis, GeoMx profiling of four patients versus controls identified PI3Kα overactivation in glomerular podocytes; analyses have shown APOE+ monocyte infiltration proximal to damaged structures, suggesting immune-epithelial crosstalk as a key pathological driver. In neurology, spatial transcriptomics has illuminated transcriptomic alterations around pathological features in Alzheimer's disease. A 2023 high-resolution study characterized the amyloid plaque cell niche using spatial transcriptomics, identifying distinct cellular and molecular environments surrounding plaques that contribute to neurodegeneration. Building on this, a 2024 integrated multimodal atlas employed to validate spatial distributions of 139 neuronal supertypes in the middle temporal gyrus across 27 donors, revealing early depletion of vulnerable somatostatin interneurons near plaques and pseudoprogression patterns correlated with disease severity. Emerging applications in 2025 extend to hematology and musculoskeletal disorders. At the 2024 American Society of Hematology meeting, spatial transcriptomics analyses of extramedullary multiple myeloma biopsies (n=14) exposed profound subclonal heterogeneity, including novel copy number variants in tumor niches, alongside T-cell dysfunction where exhausted PD-1+ TIM3+ cells colocalized with plasma cells while functional CD8+ T cells segregated to tumor-free areas, resolving post-bispecific antibody therapy. A 2025 review detailed spatial transcriptomics in musculoskeletal pathologies, such as osteoarthritis where Visium mapping of chondrocyte clusters uncovered degenerative gradients, and rheumatoid arthritis synovium where it highlighted fibroblast-like synoviocyte-driven inflammation and immune cell proximity to joint erosion sites. The translational impact of spatial transcriptomics lies in its facilitation of personalized medicine via spatial biomarkers that predict outcomes and guide therapies. In breast cancer, spatial omics has yielded prognostic signatures, including tertiary lymphoid structure gene panels (e.g., 30 genes with AUC=0.79 for detection) forecasting immunotherapy response in TNBC cohorts like I-SPY2, and markers like SREBF1/FASN for metastasis risk. Reviews underscore its role in oncology companion diagnostics, such as immune state scores (e.g., SpatialVizScore) for stratifying pembrolizumab responders in lung cancer and lymphoma, though challenges like assay standardization persist with limited FDA approvals for specific spatial platforms as of 2025.

Dissection-based methods

Laser capture microdissection

Laser capture microdissection (LCM) is a pioneering technique in spatial transcriptomics that enables the physical isolation of targeted cells or tissue regions from heterogeneous samples for downstream molecular analysis, including RNA sequencing. Developed in 1996, LCM allows researchers to procure pure populations of cells under direct microscopic visualization, preserving spatial context by selecting specific areas based on morphological or molecular markers. This method has been instrumental in early spatial transcriptomic studies, bridging histological observation with genomic profiling. The process involves mounting a thin tissue section (typically 5–10 μm thick) on a glass slide, staining it if necessary for visualization, and placing a transparent thermoplastic polymer film (such as ) over the section. A low-power infrared laser is then focused on the cells or regions of interest, heating the film locally to make it adhesive without damaging the underlying tissue; this captures the targeted material onto the film. The film is subsequently lifted, and the isolated cells are lysed for RNA extraction, amplification, and sequencing, often using protocols like for single-cell resolution. Automated variants can streamline selection, but manual oversight ensures precision in complex tissues. LCM achieves high spatial resolution, capable of isolating single cells or small clusters (down to ~10 μm), making it suitable for dissecting intricate structures like tumor microenvironments or neural circuits. Its advantages include exceptional purity of captured material (>95% target cells), compatibility with formalin-fixed paraffin-embedded (FFPE) samples common in clinical archives, and versatility across tissue types without requiring specialized arrays. However, limitations persist, such as relatively low throughput—typically hundreds to a few thousand cells per session due to manual or semi-automated selection—and potential RNA degradation from prolonged exposure during processing, though optimized protocols mitigate this. A notable application in spatial transcriptomics is the Geo-seq variant, which integrates LCM with single-nucleus sequencing to profile transcriptomes from cryosectioned s, enabling high-resolution mapping of in embryos or diseased organs while minimizing cytoplasmic contamination.

GeoMx Digital Spatial Profiler

The GeoMx Digital Spatial Profiler (), developed by NanoString Technologies, is a targeted, multiplexed platform that integrates high-resolution with molecular to enable of and protein expression in sections. Launched commercially in 2019, it allows researchers to select specific regions of interest (ROIs) guided by (IHC) or (IF) staining, facilitating biology-driven interrogation of microenvironments without physical dissection. The workflow begins with staining formalin-fixed paraffin-embedded (FFPE) or fresh frozen sections (typically 5 μm thick) using fluorescent markers for morphological guidance and barcoded probes that hybridize to target or protein analytes. After imaging the slide on the GeoMx instrument, users digitally select ROIs—such as individual cells, cellular compartments, or multicellular structures—with a down to approximately 60 μm spots, enabled by a (DMD) that directs UV light to cleave and release photocleavable tags from the selected areas. These tags are collected via microcapillaries and quantified using the NanoString nCounter for direct counting or next-generation sequencing (NGS) for higher plex levels, supporting profiling of up to 18,000 genes via the Whole Atlas (WTA) panel alongside protein panels of 40–60 analytes. A distinctive capability of GeoMx DSP is its support for integrated protein-RNA co-profiling, allowing simultaneous assessment of up to 570 proteins and thousands of transcripts within the same ROI to reveal multiomic insights into cellular states and interactions. This non-destructive approach preserves the tissue slide for subsequent analyses, building on earlier targeted isolation methods like but offering higher multiplexing without physical transfer. In applications, GeoMx has been widely applied to analysis, such as profiling immune cell infiltration and signaling in and non-small cell lung cancer tissues to identify biomarkers and therapeutic targets. GeoMx DSP has evolved into the CosMx Spatial Molecular Imager, which extends the technology to single-cell resolution for more granular spatial transcriptomics, though detailed single-cell workflows are addressed elsewhere.

Tomo-seq and sectional variants

Tomo-seq, short for , is a spatial transcriptomics that enables three-dimensional reconstruction of patterns by sequentially sectioning frozen tissues and performing sequencing on each slice. Introduced in 2014, the technique involves cryosectioning embryos or organs into thin slices, typically 10–20 μm thick, followed by extraction, library preparation, and high-throughput sequencing to generate whole-transcriptome profiles for each section. Computational then aligns these one-dimensional profiles across slices to reconstruct spatially resolved three-dimensional expression maps, providing genome-wide sensitivity with axial resolution comparable to hybridization.01226-4)01226-4) The method has been applied to model organisms such as embryos and organs, allowing detailed mapping of gradients along axes like the anterior-posterior in or the base-to-apex in the heart. For instance, in , tomo-seq revealed spatially restricted expression of developmental regulators, highlighting regional differences in transcript abundance that bulk would average out. Its strength lies in capturing continuous spatial variation without targeting specific cells, though it requires multiple identical samples for orthogonal sectioning to achieve full coverage.01226-4) Variants of tomo-seq and related sectional approaches build on this framework by incorporating targeted dissection for enhanced precision. Laser capture microdissection sequencing (LCM-seq) combines cryosectioning with laser-assisted isolation of specific tissue regions or cells, enabling polyA-based sequencing of microdissected samples with spatial context preserved at cellular . Developed around 2017, LCM-seq has been refined for applications like profiling single neurons or fluorescently tagged populations in the , offering deeper coverage than bulk sectional while maintaining positional information. Other sectional variants, such as direct on cryosection layers, provide bulk-level profiling per slice but lack the selective capture of LCM-seq, serving as simpler alternatives for broader tissue layers. These methods uniquely facilitate axial mapping in complex organs, such as delineating in regions or cardiac gradients, where three-dimensional context is critical for understanding tissue organization. Recent extensions, including improved library preparation and computational tools for higher-resolution , have enhanced tomo-seq's utility in developmental studies as of 2024, allowing finer-grained analysis of embryonic patterning and . For example, advancements in pipelines have supported multi-axis sectioning to resolve sub-millimeter expression domains in models.

In vivo and niche-specific isolation

In vivo and niche-specific isolation methods in spatial transcriptomics enable the capture of transcriptomic profiles from defined cellular microenvironments within living s, minimizing artifacts from or fixation that can disrupt dynamic interactions. These approaches leverage targeted labeling or encapsulation techniques to isolate from specific niches, such as tumor or immune compartments, while preserving spatial context and physiological states. Unlike ex vivo dissection methods, which rely on post-fixation processing and may alter , strategies allow real-time interrogation of cell-cell communications in their native setting. One pioneering technique is Transcriptome In Vivo Analysis (TIVA), introduced in 2014, which uses photoactivatable probes to capture mRNA from individual cells in live animal models. The TIVA-tag, a multifunctional with a photocleavable moiety and biotin-binding domain, is injected into mice and binds to polyadenylated RNAs; upon targeted photoactivation using a , it covalently links to nearby mRNAs, enabling their isolation via pulldown after tissue homogenization. Applied to tissue and tumor niches in live mice, TIVA revealed spatially restricted transcriptomic heterogeneity, such as neuron-specific patterns in the , demonstrating its utility for noninvasive single-cell RNA profiling in intact environments. This method has been instrumental in studying tumor microenvironments, where it isolated stroma-associated RNAs to uncover niche-specific regulatory networks. Building on photoactivation principles, NICHE-seq, developed in 2017, facilitates the and paired sequencing of interacting clusters from immune niches . Transgenic mice expressing photoactivatable fluorescent reporters in specific types, such as dendritic cells, allow visualization and selective activation of niche regions in lymph nodes using two-photon microscopy; activated cells are then dissociated, sorted by fluorescence-activated sorting (FACS), and subjected to single- RNA sequencing to profile transcriptional states of interacting pairs. In studies of viral infection responses, NICHE-seq identified niche-specific subpopulations, like clusters in splenic white pulp, highlighting dynamic immune interactions that bulk sequencing overlooks. Although not involving direct encapsulation, the method preserves spatial proximity through immediate post-activation , akin to encapsulating functional units. ProximID, established in 2018, extends niche isolation by focusing on physically interacting networks through proximity-based microdissection, adaptable to live-derived samples for enhanced . In this approach, fixed sections are manually dissected into small multicellular units (doublets or triplets) using micromanipulators guided by , followed by whole-transcriptome amplification and sequencing to map interaction-specific . Applied to hematopoietic niches, ProximID uncovered contact-dependent programs, such as osteoblast-hematopoietic signaling, revealing how spatial proximity drives . While primarily , its principles of can integrate with live imaging to tag and isolate clusters, supporting applications for dynamic tissue analysis. These methods share a core principle of capture—via photoactivation, crosslinking, or physical isolation—to maintain the spatial integrity and transient dynamics of living tissues, avoiding the loss of microenvironmental cues inherent in traditional . Recent advances, as reviewed in 2025, have expanded their application to musculoskeletal niches, where spatial transcriptomics isolates and clusters to elucidate bone regeneration pathways and disease-specific dysregulations, such as in .

Hybridization-based imaging methods

Single-molecule and low-plex FISH

Single-molecule (smFISH) enables the visualization and quantification of individual transcripts within fixed cells and tissues by using multiple short probes, each labeled with a , that hybridize to specific target mRNAs, producing diffraction-limited spots detectable via . Introduced in , smFISH allows for absolute counting of mRNA molecules per cell, revealing spatial distributions and subcellular localizations with localization precision on the order of 20 nm, which facilitates studies of heterogeneity and RNA trafficking. A key advancement in low-plex FISH is RNAscope, developed in , which employs a branched DNA amplification strategy to enhance signal detection while minimizing , enabling reliable visualization of low-abundance transcripts in formalin-fixed, paraffin-embedded tissues using just 1-6 probes per . In RNAscope, pairs of adjacent probes hybridize to the target , recruiting amplifier molecules that generate a tree-like structure of fluorophore-labeled oligos, amplifying the signal up to 1,000-fold without enzymatic steps. The core principle of these low-plex FISH methods relies on specific hybridization of complementary nucleic acid probes to RNA targets in situ, preserving cellular architecture and achieving subcellular resolution through confocal or widefield microscopy, where each fluorescent spot corresponds to a single RNA molecule. However, these techniques are limited to low multiplexing capacity, typically profiling 1-10 genes per experiment due to spectral overlap of fluorophores and the need for sequential imaging or stripping, and often require manual or semi-automated image analysis for spot detection and quantification. As the foundational approach for hybridization-based spatial transcriptomics, single-molecule and low-plex has inspired variants like expansion-assisted iterative (EASI-FISH), which integrates tissue expansion to enhance in thick samples while retaining the probe hybridization strategy for multi-round imaging.01339-8)

Multiplexed and high-throughput

Multiplexed and high-throughput () methods extend the capabilities of single-molecule (smFISH) by enabling the simultaneous detection of hundreds to thousands of targets in situ, preserving spatial information at cellular . These approaches rely on combinatorial labeling strategies to assign unique identifiers to multiple transcripts, overcoming the spectral limitations of traditional that restrict low-plex detection to just a few genes per experiment. The core principle of multiplexed FISH involves encoding RNA identities through sequential or parallel hybridizations using fluorescently labeled probes, where each target receives a unique composed of on/off patterns across multiple imaging rounds. Common encoding schemes include codes, in which each is represented by a specific of fluorescent spots (e.g., present or absent in each round), and more efficient Huffman codes that allocate shorter barcodes to highly expressed genes to optimize signal detection and minimize readout errors. Error-robust designs, such as those incorporating in barcodes, allow for single or double errors per readout without misidentification, ensuring high accuracy even in noisy cellular environments. Sequential (seqFISH), introduced in , achieves through iterative rounds of hybridization and , where probes for subsets of genes are applied sequentially, and signals are read out after each cycle before stripping for the next round. This method enables the profiling of up to 100 genes in single cells by combining signals from multiple rounds into unique combinatorial addresses, demonstrating its utility in mapping patterns in cultured cells. Building on this foundation, seqFISH variants have been refined for higher throughput, maintaining subcellular resolution while scaling to larger gene panels. Multiplexed error-robust (MERFISH), developed in 2015, advances by using pre-designed, error-correcting barcodes that allow parallel detection of 100 to 1,000 genes in a single experiment with single-cell and single-molecule . In MERFISH, each target is hybridized with a set of encoding probes, and readout probes generate fluorescent spots only for matching combinations, enabling the identification of thousands of transcripts across diverse tissues like fibroblasts and immune cells. Subsequent improvements, such as expanded encoding schemes, have pushed MERFISH to genome-scale profiling while correcting for optical and biochemical errors. Optimized sequential FISH (osmFISH), reported in 2018, refines the sequential hybridization approach specifically for complex tissues like the , achieving reliable detection of dozens of genes with reduced and improved signal stability through cycle-optimized probe designs and automated . Applied to somatosensory sections, osmFISH revealed layer-specific gradients and cell-type organization, highlighting its effectiveness for spatially resolving neuronal diversity in dense tissues. Recent applications of multiplexed FISH in hematology underscore its growing role in dissecting disease microenvironments, as evidenced by 2024 American Society of Hematology (ASH) meeting abstracts employing MERFISH to validate spatial niches in embryonic hematopoiesis and . These studies demonstrate how barcode-enabled FISH can map transcript distributions in bone marrow-like settings, identifying dynamic interactions between hematopoietic stem cells and supportive stroma at single-cell resolution.

Advanced and subcellular imaging variants

Advanced variants of fluorescence in situ hybridization (FISH) have pushed the boundaries of spatial transcriptomics by achieving subcellular resolution, three-dimensional (3D) imaging, and near-whole-transcriptome coverage without relying on sequencing. These methods build on multiplexed principles by incorporating error-correction strategies, tissue expansion techniques, and signal amplification to resolve RNA molecules at nanometer scales within intact cells and tissues. Such innovations enable the visualization of transcript localization to specific organelles, such as the or , providing insights into cellular processes like mRNA trafficking and local . seqFISH+, introduced in , represents a seminal advancement in high-throughput, error-robust by over 10,000 genes in single cells with sub-diffraction-limit . This method uses combinatorial sequential barcoding with a binary encoding scheme and error-correction via Hamming distances, allowing accurate decoding of mRNA barcodes even in the presence of optical noise or incomplete hybridizations. Applied to brain tissue sections, seqFISH+ has revealed subcellular RNA distributions, such as nuclear enrichment of certain transcripts, facilitating the mapping of cell-type-specific at scale. Expansion-assisted FISH variants, including ExFISH and EASI-FISH developed in the mid-2010s and , achieve nanoscale resolution approaching 100 nm through physical tissue expansion. ExFISH integrates probes with expansion microscopy, where samples are embedded in a swellable that isotropically expands up to fourfold after protein digestion, physically separating molecules for via standard . EASI-FISH extends this to iterative and thick tissues (up to 200 μm), enabling profiling of dozens of s in regions like the while preserving integrity through covalent anchoring. These techniques have been used to dissect subcellular gradients in neuronal dendrites, highlighting compartmentalized expression.01339-8) The CosMx Spatial Molecular Imager, launched by NanoString in 2022, combines high-plex and protein at subcellular , targeting up to 6,000 genes and 100 proteins per . Employing cyclic sequential hybridization with error-robust barcodes and deep-learning-based spot detection, CosMx achieves single-molecule sensitivity in formalin-fixed, paraffin-embedded (FFPE) tissues, resolving transcripts within cellular compartments like the or membrane. In applications to human tumors, it has mapped ligand-receptor interactions at the single-cell level, integrating transcriptomic and proteomic data to uncover heterogeneous microenvironments. Recent expansions to whole-transcriptome panels (over 18,000 genes) further enhance its utility for unbiased spatial profiling. DNA microscopy, pioneered in 2019, offers an optics-free approach to subcellular spatial transcriptomics by encoding molecular positions into DNA barcodes for downstream sequencing. In this method, barcoded oligonucleotides hybridize to target RNAs, and proximity ligation captures spatial relationships as unique amplicons, computationally reconstructing 2D or 3D "virtual stains" of transcript distributions without microscopy hardware. Early demonstrations imaged hundreds of mRNAs in cultured cells, revealing subcellular clustering patterns, and recent volumetric extensions have applied it to intact organisms like zebrafish embryos for whole-transcriptome mapping.30547-1) Signal amplification via single-molecule hybridization (smHCR) enhances detection sensitivity in advanced workflows, enabling brighter, more reliable subcellular of low-abundance transcripts. smHCR initiates a of fluorophore-labeled hairpins upon target binding, amplifying signals up to 1,000-fold without enzymatic steps, while maintaining single-molecule . Integrated into multiplexed schemes, it has improved 3D in cleared tissues, such as whole-mount brains, to visualize localization in synaptic compartments. A landmark 2025 development, reverse-padlock amplicon encoding (RAEFISH), achieves sequencing-free, whole-genome spatial transcriptomics at single-molecule resolution, profiling 23,000 or 22,000 genes in sections. By combining probes with iterative barcoding and direct fluorescent readout, RAEFISH bypasses sequencing bottlenecks, enabling rapid, high-coverage imaging of subcellular transcriptomes in complex samples like tumors. This method has unveiled genome-wide subcellular dynamics, such as isoform-specific localizations, transforming large-scale spatial studies.01037-2)

In situ sequencing methods

Padlock probe and rolling-circle approaches

Padlock probe and rolling-circle amplification (RCA) approaches represent a foundational class of in situ sequencing (ISS) methods for spatial transcriptomics, enabling targeted detection and sequencing of RNA molecules directly within intact cells and tissues. These techniques rely on padlock probes, which are linear oligonucleotides designed with two target-complementary arms flanking a backbone sequence, to achieve high specificity for individual transcripts. Upon hybridization to reverse-transcribed RNA targets, the probes are ligated into closed circles using a DNA ligase, followed by RCA using phi29 DNA polymerase to generate localized, repeating DNA concatemers known as rolonies or nanoballs, typically under 1 μm in diameter. These amplified structures serve as templates for subsequent fluorescent sequencing, often via ligation-based chemistry, allowing the readout of short nucleotide sequences (e.g., 4–16 bases) while preserving spatial context. The seminal development of this approach, introduced in , demonstrated ISS in fixed human tissue sections, where padlock probes targeted cDNA from mRNAs such as β-actin (ACTB) and HER2, followed by and sequencing by to resolve point mutations and patterns at single-cell resolution. This method achieved subcellular localization of transcripts, with rolonies detectable via , and supported of up to several dozen targets through barcoded probes, though early implementations were limited to ~10–20 s due to optical crowding and probe efficiency. The technique is particularly suited for archived, formalin-fixed paraffin-embedded (FFPE) tissues, providing a targeted alternative to untargeted sequencing by focusing on predefined gene panels. A recent advancement, reverse-padlock amplicon-encoding (RAEFISH; as of October 2025), extends padlock probes to sequencing-free, image-based whole-genome spatial transcriptomics at single-cell resolution, enabling 3D mapping in thick tissues without next-generation sequencing. A notable variant, BaristaSeq (2018), optimizes the padlock-RCA for high-throughput barcode sequencing, incorporating Phusion for efficient gap-filling prior to , which boosts yield by over fivefold compared to earlier strand-displacing polymerases and reduces off-target signals to ~0.2 rolonies per . This enables pooled library screening of diverse genetic barcodes (up to 10^15 theoretical combinations with 25-nt reads) at subcellular resolution, with >97% sequencing accuracy, and has been applied to lineage tracing and neuronal in tissues. Despite these advances, padlock-RCA methods remain constrained by probe design complexity, which requires precise targeting to avoid cross-hybridization, and detection efficiencies typically ranging from 1–5%, limiting scalability beyond hundreds of plex in practice.

Enzymatic sequencing methods

Enzymatic sequencing methods in spatial transcriptomics enable direct readout of RNA sequences within intact tissues by leveraging polymerase-driven reactions to generate and sequence (cDNA) amplicons , preserving spatial context without relying on hybridization probes for capture. These approaches contrast with probe-based techniques by performing unbiased, genome-wide profiling through enzymatic conversion and amplification of endogenous . The pioneering method, fluorescent sequencing (FISSEQ), exemplifies this category, allowing subcellular resolution of transcript locations and sequences in fixed biological samples. A key commercial implementation is the Xenium platform by (launched 2020), which uses enzymatic sequencing-by-ligation for targeted profiling of up to 5,000 genes at ~1 μm resolution, supporting fresh frozen and FFPE tissues in applications like analysis and as of 2025. In FISSEQ, the process begins with tissue fixation on a slide, followed by in situ reverse transcription of RNA using tagged random hexamers and aminoallyl-modified dUTP to incorporate cross-linkable bases into the cDNA. Residual RNA is then degraded with RNase, and the cDNA is circularized before undergoing rolling circle amplification (RCA) with phi29 polymerase, producing localized nanoballs of amplified DNA (200–400 nm in diameter) that remain anchored in the tissue via cross-linking with BS(PEG)9. Sequencing occurs directly on these amplicons using enzymatic ligation-based chemistry, such as the SOLiD platform, where fluorescently labeled probes are ligated in cycles to reveal up to 30 bases per read, enabling detection of thousands of transcripts per cell with subcellular precision. This polymerase-mediated amplification and sequencing maintains transcript localization, though cross-linking minimizes diffusion of enzymatic products. Despite these advances, enzymatic methods like FISSEQ face challenges, including low overall efficiency—yielding approximately 200 mRNA reads per cell in mammalian tissues—and limited read lengths due to signal attenuation from background and incomplete enzymatic access in dense tissues. These constraints arise partly from the need for robust cross-linking to prevent amplicon , which can reduce fidelity and amplicon yield compared to extracted sequencing. As a targeted alternative, padlock probe methods restrict sequencing to predefined transcripts but avoid some unbiased biases. Ongoing optimizations, such as integrating sequencing-by-synthesis with reversible terminators, aim to extend read lengths beyond 30 bases while improving detection in complex samples like formalin-fixed paraffin-embedded tissues.

Expansion-assisted and combinatorial methods

Expansion-assisted and combinatorial methods in sequencing (ISS) integrate physical expansion of biological specimens with multiplexed encoding strategies to achieve high-resolution, large-scale mapping of in three-dimensional tissues. These approaches address limitations in signal detection and by embedding samples in swellable hydrogels that uniformly expand the tissue, effectively increasing the physical distance between molecules to enable nanoscale imaging with conventional microscopes. A seminal example is STARmap, introduced in 2018, which combines combinatorial barcode labeling with hydrogel-tissue chemistry (HTC) for intact-tissue sequencing of over 1,000 genes in the , revealing subcellular transcriptional states and organization. As of September 2025, methods like iSCALE have advanced reconstruction for large-scale, super-resolution spatial mapping integrated with ISS data, enabling automated annotation in whole organs. The core principles of these methods involve DNA-encoded probes that hybridize to target mRNAs in fixed , followed by hydrogel embedding where and other monomers polymerize around the sample to form a swellable . Upon of proteins and , the gel expands isotropically—typically by a factor of approximately 4x in linear dimensions—diluting molecular crowding and enhancing accessibility for subsequent sequencing reads. In STARmap, signal amplification occurs through reversible hybridization of template (STAR) probes, which generate amplicons for combinatorial fluorescent readout via sequential and , allowing error-corrected decoding of barcodes without enzymatic sequencing steps. This expansion yields post-expansion resolutions down to ~20-50 nm, sufficient for subcellular localization of transcripts in dense neural environments. Variants of expansion-assisted ISS often incorporate elements from (FISH) for enhanced signal amplification, such as ExFISH, which primarily relies on iterative hybridization chain reaction (HCR) in expanded samples but can integrate with sequencing for higher plex. These methods have advanced by enabling 3D mapping of in intact volumes, as highlighted in recent reviews on spatial transcriptomics frontiers. For instance, STARmap variants like Deep-STARmap extend to thousands of genes and (Deep-RIBOmap), supporting studies of translation in neural circuits as of 2024.

Capture-based sequencing methods

Bead and slide array techniques

Bead and slide array techniques in spatial transcriptomics employ randomly positioned, barcoded beads arrayed on a to capture mRNA from sections, enabling unbiased, whole-transcriptome profiling at near-cellular . These methods rely on poly-A capture using oligo-dT sequences attached to the beads, which are combined with unique spatial barcodes and unique molecular identifiers (UMIs) to link transcripts to their approximate positions after sequencing. The random deposition of beads allows for flexible, high-density arrays without fixed grids, facilitating scalable mapping across large areas. A seminal example is Slide-seq, introduced in 2019, which arrays DNA-barcoded beads with a diameter of approximately 10 μm onto a puck-shaped surface, achieving spatial resolution comparable to individual cell sizes. In this approach, fresh-frozen sections are placed on the bead array, permeabilized to release mRNA, which is captured via the oligo-dT sequences; subsequent reverse transcription, pooling, and sequencing demultiplex the data using the spatial barcodes to reconstruct profiles. Slide-seq enables whole-transcriptome analysis, detecting thousands of genes per spot and identifying fine-scale tissue organization, such as cell-type-specific patterns in the mouse cerebellum and . Variants like high-definition spatial transcriptomics (HDST), also from 2019, enhance bead by loading uniquely barcoded 2 μm silica beads into etched wells on a glass slide, yielding 1,365,856 spots across a 6.75 mm² area with 2 μm —providing ~25-fold higher than Slide-seq. HDST follows similar principles, with sections placed on the for mRNA capture, for histological correlation, and sequencing after barcode decoding via sequential hybridization probes. This method has been applied to profile in regions and human biopsies, recovering hundreds of thousands of spatially resolved transcripts while minimizing cross-spot contamination. These techniques offer key advantages, including unbiased capture of the poly-A without prior selection, scalability to millions of spots for broad coverage, and compatibility with standard sequencing workflows. For instance, Slide-seq and HDST can generate data from up to 10,000–100,000 spots per tissue section, supporting discovery of spatial gradients and cell neighborhoods. Commercial platforms have evolved from these bead-array concepts, incorporating improved bead synthesis and capture efficiencies for routine use.

Microarray and deterministic barcoding

Microarray-based spatial transcriptomics employs fixed arrays of capture probes with predefined spatial barcodes to map gene expression while preserving positional information from tissue sections. The foundational method, introduced in 2016, involves placing permeabilized tissue sections onto a glass slide featuring an array of ~1,000 spots, each approximately 100 μm in diameter and spaced 200 μm apart, coated with oligonucleotides containing unique spatial barcodes and poly(dT) sequences. These probes capture polyadenylated mRNA that diffuses from the tissue during permeabilization, enabling reverse transcription directly on the array to generate cDNA tagged with the spatial barcode; subsequent library preparation and sequencing link transcripts to their originating positions for computational reconstruction of the spatial transcriptome. This approach achieves genome-wide profiling at a resolution limited by spot size, typically capturing signals from multiple cells per spot, and has been applied to tissues like brain and tumors to reveal spatially resolved gene expression patterns. Building on this, deterministic barcoding refines techniques by using precisely patterned, non-random capture sites to ensure reproducible spatial encoding. A key advancement is Stereo-seq, developed in 2022 (with initial release in 2021), which utilizes DNA nanoball (DNB)-patterned chips where each ~220 nm spot is fixed in position, providing a vast array of deterministic barcodes (up to 4^{25} unique combinations via 25-nt cell ID sequences). sections are fixed and permeabilized on the chip, allowing mRNA capture by barcode-linked probes, followed by on-chip reverse transcription with unique molecular identifiers (UMIs) for accurate quantification; sequencing then maps reads to sub-cellular bins (e.g., 500 nm center-to-center spacing). This enables high-resolution profiling over large fields of view (up to ~1 cm²), detecting transcripts at near-single-molecule levels and resolving subcellular features in diverse tissues. These methods offer advantages in reproducibility and throughput due to their fixed, deterministic layouts, which contrast with random bead arrays by eliminating variability in probe positioning and facilitating scalable, high-density capture without alignment errors. For instance, Stereo-seq supports whole-embryo mapping with millions of spots, achieving sensitivities comparable to single-cell while maintaining spatial context. Recent platform updates in 2024, such as enhanced Visium assays, continue to reference and iterate on these core principles for improved resolution and accessibility.

High-definition and commercial platforms

High-definition spatial transcriptomics platforms represent advancements in capture-based sequencing methods, achieving near-single-cell through denser arrays and refined chemistries, building on the original spatial barcoding approach introduced in 2016. These systems prioritize whole-transcriptome profiling with enhanced spatial fidelity, enabling integration of transcriptomic data with histological features like H&E staining for analysis. The Visium platform, launched in 2019, utilizes slide-mounted arrays with approximately 5,000 barcoded spots, each 55 μm in diameter and spaced 100 μm apart, capturing poly(A)-tailed mRNA from fresh frozen or FFPE-preserved tissues at a of 1–10 cells per spot. An FFPE-compatible version, introduced subsequently, employs updated reverse transcription chemistries to accommodate fixed samples while maintaining whole-transcriptome coverage across diverse species. In 2024, released Visium HD, featuring a gapless array of 2 μm² capture tiles that provide single-cell-scale resolution without spatial gaps, supporting both fresh frozen and FFPE tissues through high-density oligonucleotide lawns. This upgrade increases spot density dramatically, enabling binned outputs at multiple scales (e.g., 2 μm, 8 μm, 16 μm) and seamless co-registration with H&E images for enhanced morphological context. A 2025 comparative guide highlights Visium HD's superior resolution among commercial options, positioning it as ideal for applications requiring subcellular detail, such as profiling. Takara Bio's Seeker platform, commercialized in 2023 via acquisition of Curio Biosciences, employs targeted capture on arrays of 10 μm spatially indexed beads arranged in a , achieving high-resolution whole-transcriptome mapping of fresh frozen tissues. The system leverages bead-based barcoding derived from Slide-seqV2 principles, with updated and steps to boost capture efficiency and density for precise spatial localization. These platforms incorporate cloud-based computational pipelines for , including and , to handle the increased complexity of high-density datasets while ensuring across labs. Overall, they advance capture-based methods by prioritizing accessibility, with kits supporting unbiased at scales approaching .

Computational and reconstruction methods

In silico spatial inference

In silico spatial inference refers to computational methods that reconstruct spatial patterns from non-spatial transcriptomic data, such as single-cell RNA sequencing (scRNA-seq), by leveraging reference spatial information from imaging or low-resolution assays. These approaches typically employ algorithms to infer cell positions based on shared profiles, aligning dissociated cells to predefined spatial coordinates or atlases. By correlating expression patterns of known spatially restricted genes, models assign probabilistic locations, enabling the generation of pseudo-spatial maps that approximate transcriptomes without direct spatial capture. A seminal example is DistMap, introduced in 2017, which aligns scRNA-seq data from approximately 6,000 embryo cells to (ISH) imaging references. The method binarizes profiles and computes Matthews coefficients between single cells and imaged spots to rank and assign spatial positions, reconstructing a high-resolution virtual embryo with expression maps for over 8,000 genes. This marker-gene-driven alignment reveals spatial gradients of transcription factors and signaling pathways, such as Hippo signaling, demonstrating how computational inference can predict cell communication and tissue organization from dissociated data. Reconstruction often integrates low-plex ISH data, like sequential (seqFISH), with comprehensive scRNA-seq atlases to expand limited spatial readouts to full transcriptomes. In a 2021 study on mouse organogenesis, seqFISH profiled 351 genes across embryo sections, followed by mapping of cells to an E8.5 scRNA-seq atlas using batch-corrected nearest neighbors in reduced dimensionality space. Genome-wide expression was imputed via weighted averages from the 25 closest scRNA-seq neighbors, yielding a 10,000-plex spatial map validated by smFISH on held-out genes (median performance score of 0.73), which facilitated virtual dissections of structures like the midbrain-hindbrain boundary. Despite these advances, relies heavily on the quality of spatial priors, such as atlases, which can introduce biases if references are incomplete or mismatched. In heterogeneous tissues, where interactions and microenvironments vary sharply, accuracy diminishes due to challenges in capturing subtle gradients and potential misalignment errors, limiting reliability in complex scenarios like s. Recent integrations in 2025 have addressed some gaps by combining scRNA-seq with spatial transcriptomics via deep generative models, enhancing in immune contexts like the .

Data analysis pipelines

Data analysis pipelines for spatial transcriptomics typically follow a modular that transforms raw sequencing outputs into interpretable spatial maps, enabling downstream tasks such as clustering and . These pipelines address unique challenges posed by the spatial context, including variable capture efficiencies across regions and the need to preserve positional information from histological images. Standard steps include preprocessing to ensure , to mitigate technical biases, and clustering to identify spatially coherent types or domains, all while integrating tools that handle both transcriptomic counts and coordinates. Preprocessing begins with aligning sequencing reads to the corresponding tissue image, which registers data to spatial coordinates using fiducial markers or image feature matching. This step is crucial for technologies like Visium or MERFISH, where misalignment can distort spatial patterns. Following alignment, demultiplexing of spatial barcodes separates reads by their unique positional identifiers, often using error-correcting codes to handle sequencing noise. Finally, unique molecular identifier (UMI) deduplication collapses PCR-amplified duplicates, reducing inflation in expression estimates while accounting for potential spatial biases in amplification efficiency. These processes are implemented in platforms like Seurat, which supports loading and aligning Visium data directly from outputs. Normalization adjusts for technical variations, such as differences in size or capture efficiency, which can vary spatially due to heterogeneity. Common approaches include by total counts per ( size ) or using spike-in controls, but these may overlook spatial where nearby spots share similar expression profiles. To address this, spatially aware methods model both global and local effects; for instance, SpaNorm incorporates a to estimate biological variance while correcting for size and positional dependencies. Such techniques prevent over- or underestimation of expression in clustered regions, improving the reliability of downstream analyses. Clustering in spatial transcriptomics identifies groups of spots or cells with similar expression profiles, distinguishing between non-spatial methods (e.g., k-means on gene counts) and spatial-aware approaches that incorporate neighborhood relationships via graph-based models. Non-spatial clustering treats data as independent, potentially ignoring tissue architecture, whereas graph-based methods construct adjacency matrices from spatial coordinates and apply algorithms like Louvain or to enforce contiguity. For example, GraphST uses self-supervised contrastive learning on graphs to enhance resolution in delineating domains. Spatial-aware clustering has demonstrated superior performance in benchmarks, achieving higher adjusted Rand indices on datasets like Visium human sections compared to standard Louvain. A key metric in these pipelines for detecting spatially variable genes—informative for clustering—is , which quantifies . The global for a gene's expression z across n spots, with spatial weights w_{ij} and row-standardized sum S_0 = \sum_i \sum_j w_{ij}, is given by: I = \frac{n}{S_0} \sum_i \sum_j w_{ij} z_i z_j / \sum_i z_i^2 Values range from -1 (dispersed) to +1 (clustered), with genes exhibiting high positive I prioritized for dimension reduction prior to clustering. This statistic is computed in tools like Squidpy to filter features that drive spatial patterns. Widely adopted tools facilitate these pipelines: In R, Seurat provides end-to-end workflows including SCTransform normalization and spatial plotting, while SpatialExperiment offers a Bioconductor infrastructure for storing aligned data and integrating with SingleCellExperiment objects. In Python, Squidpy builds on Scanpy for scalable graph operations and autocorrelation metrics, supporting datasets up to millions of spots. A recent unified pipeline, SpaceSequest (2025), integrates these steps with modular components for preprocessing, normalization via deep learning, and graph-based clustering, demonstrated on multi-omics compatible benchmarks to reduce runtime by 40% compared to Seurat alone.

Integration with multi-omics data

Spatial transcriptomics technologies generate gene expression profiles while preserving spatial context, but their resolution often aggregates signals from multiple cells within spots, necessitating integration with single-cell RNA sequencing (scRNA-seq) data to deconvolute cell-type compositions. Deconvolution methods, such as Robust Cell Type Decomposition (RCTD), leverage scRNA-seq reference profiles to model each spatial spot as a mixture of cell types, estimating proportions and enabling cell-type assignment at single-cell-like resolution. For instance, RCTD fits a linear combination of cell-type-specific expression profiles to the observed spot data, accounting for ambient RNA and doublets to improve accuracy in heterogeneous tissues like the mouse brain. This integration enhances the interpretability of spatial data by mapping fine-grained cellular distributions without requiring ultra-high-resolution imaging. Integration with further enriches spatial profiling by correlating transcriptomic patterns with protein abundance and localization, revealing post-transcriptional regulatory insights. The GeoMx Digital Spatial Profiler platform supports simultaneous and protein analysis on the same section, using region-of-interest selection to profile up to 1,800 genes and 100+ proteins, as demonstrated in studies where proteomic markers like complement signatures. Complementarily, (co-detection by indexing) enables highly multiplexed spatial , imaging over 50 protein markers per cycle via DNA-barcoded antibodies, which can be co-registered with transcriptomic to map immune cell phenotypes in s. These approaches highlight discrepancies between mRNA and protein levels, such as in cancer progression. Advances in spatial genomics, particularly spatial ATAC-seq, integrate chromatin accessibility data with transcriptomics to uncover regulatory mechanisms underlying spatial gene expression variation. Methods like spatial-ATAC-seq, developed in 2024, profile open landscapes in situ using tagmentation on tissue sections, achieving ~10 μm resolution and revealing enhancer-promoter interactions in developmental contexts. When aligned with spatial transcriptomics, these datasets identify spatially variable regulatory elements, such as in brain organoids where accessible correlates with neuronal gradients. Joint embedding frameworks, like MOFA+ (Multi-Omics Factor Analysis plus), facilitate multi-modal integration by factorizing diverse matrices into shared latent factors, preserving spatial coordinates to detect coordinated variations across modalities in single-cell and spatial data. As of 2025, the frontiers of single-cell and spatial multi-omics emphasize scalable integration pipelines that fuse transcriptomics, , and to model tissue architecture comprehensively, addressing challenges like data sparsity and in complex diseases.

References

  1. [1]
    The expanding vistas of spatial transcriptomics | Nature Biotechnology
    Oct 3, 2022 · A suite of spatial transcriptomics technologies capable of measuring the expression of many, or all, genes in situ.
  2. [2]
    Spatial transcriptomics coming of age | Nature Reviews Genetics
    Apr 12, 2019 · Spatial transcriptomics methods aim to characterize gene expression profiles while retaining information of the spatial tissue context.Original Articles · About This Article · Cite This ArticleMissing: paper | Show results with:paper
  3. [3]
    Spatial transcriptomics in health and disease - Nature
    May 8, 2024 · This Review provides an overview of current and emerging spatial transcriptomic methods, their applications and remaining challenges for the field.
  4. [4]
    Museum of spatial transcriptomics | Nature Methods
    Mar 10, 2022 · We present a curated review of literature on spatial transcriptomics dating back to 1987, along with a thorough analysis of trends in the field.
  5. [5]
  6. [6]
    An introduction to spatial transcriptomics for biomedical research
    Jun 27, 2022 · At a broad level, spatial transcriptomics can be used to characterize transcriptional patterning and regulation in tissues. For example, in ...Missing: seminal | Show results with:seminal
  7. [7]
    Visualization and analysis of gene expression in tissue sections by ...
    Jul 1, 2016 · Ståhl et al. have developed a way of measuring the spatial distribution of transcripts by annealing fixed brain or cancer tissue samples ...
  8. [8]
    Spatial transcriptomics in development and disease - PMC
    Oct 9, 2023 · Spatial transcriptomics (ST) is a method that maps gene expression in tissue while preserving spatial information.Missing: seminal papers
  9. [9]
  10. [10]
  11. [11]
  12. [12]
  13. [13]
    From bulk, single-cell to spatial RNA sequencing - Nature
    Nov 15, 2021 · Bulk RNAseq studies average global gene expression, scRNAseq investigates single cell RNA biology up to 20,000 individual cells simultaneously, ...
  14. [14]
    Subcellular Transcriptomics and Proteomics: A Comparative ... - NIH
    In this review, we discuss and directly compare the current methods in spatial proteomics and transcriptomics, which include sequencing- and imaging-based ...
  15. [15]
    Understanding Bulk and Single-Cell RNA Sequencing - CD Genomics
    Stark et al. (2019) reported that bulk RNA-seq costs were approximately $300 per sample, while single-cell RNA-seq ranged from $500 to $2000 per sample ...Missing: proteomics | Show results with:proteomics
  16. [16]
    Laser Capture Microdissection - Science
    Laser capture microdissection (LCM) under direct microscopic visualization permits rapid one-step procurement of selected human cell populations.
  17. [17]
    RNA imaging: seeing is believing - PMC - NIH
    During the 1990s, the method was further optimized for the simultaneous detection of DNA, unspliced precursors and spliced mRNA. Using these tools, several ...
  18. [18]
  19. [19]
    NanoString Launches Priority Site Program for New GeoMx Digital ...
    Sep 5, 2018 · The GeoMx DSP system is currently in development and is expected to be available for early access instrument placements late in 2018 followed by ...
  20. [20]
    Transcriptome-scale super-resolved imaging in tissues by RNA ...
    Mar 25, 2019 · seqFISH+ provides a tenfold or greater improvement over existing methods in the number of mRNAs profiled and the total number of RNA barcodes ...
  21. [21]
    iIMPACT: integrating image and molecular profiles for spatial ...
    Jun 6, 2024 · We have developed a multi-stage statistical method called iIMPACT. It identifies and defines histology-based spatial domains based on AI-reconstructed ...
  22. [22]
    Spatio-temporal mRNA tracking in the early zebrafish embryo - Nature
    Jun 7, 2021 · In summary, tomo-seq allowed us to determine subcellular RNA localization in the one-cell stage zebrafish embryo on the transcriptome-wide level ...
  23. [23]
    Transcriptomic cytoarchitecture reveals principles of human ...
    Oct 13, 2023 · Single-nucleus and spatial transcriptomic methods enable high-resolution characterization of the cellular structure of the human neocortex.
  24. [24]
    High resolution mapping of the tumor microenvironment using ...
    Dec 19, 2023 · ... number of clusters as the scFFPE-seq data), with a median of 5712 genes identified per spot. We used these two discovery-based datasets and ...
  25. [25]
    Comprehensive visualization of cell–cell interactions in single-cell ...
    When applied to spatial transcriptomic data, NICHES can be used to reflect local cellular microenvironment. NICHES can operate with any list of ligand–receptor ...
  26. [26]
  27. [27]
    Spatial transcriptomics reveals substantial heterogeneity in triple ...
    Nov 26, 2024 · Here, we investigate TNBC tumor architecture including its microenvironment using spatial transcriptomics on a series of 92 patients.
  28. [28]
    Spatial deconvolution of HER2-positive breast cancer delineates ...
    Oct 14, 2021 · We investigate spatial gene expression in HER2-positive breast tumors using Spatial Transcriptomics technology.
  29. [29]
    Spatial transcriptomics in autoimmune rheumatic disease
    Feb 20, 2025 · Immune cells were located in close proximity to podocytes, suggesting their infiltration into the glomerular tissue rather than confinement to ...
  30. [30]
    Integrated multimodal cell atlas of Alzheimer's disease - Nature
    Oct 14, 2024 · To define the spatial distribution of supertypes and to validate cellular changes, we generated a large-scale, cellularly resolved MERFISH ...
  31. [31]
    Spatial transcriptomics reveals profound subclonal heterogeneity ...
    Nov 14, 2024 · Spatial transcriptomics reveals profound subclonal heterogeneity and T-cell dysfunction in extramedullary myeloma.
  32. [32]
    Advances in spatial transcriptomics and its application in ... - Nature
    May 16, 2025 · This review provides a summary of recent advances in spatial transcriptomics and its application to the musculoskeletal system.
  33. [33]
    Spatial omics in biomarker discovery in breast cancer: a narrative ...
    Sep 30, 2025 · Biomarkers or spatial biomarkers offer insights into the spatial relationships and arrangements of various cells, biomolecules, and structures ...
  34. [34]
    Spatial oncology: Translating contextual biology to the clinic
    ### Summary of Spatial Oncology Review: Biomarkers and Personalized Medicine
  35. [35]
    Laser-capture microdissection | Nature Protocols
    Jun 27, 2006 · Laser-capture microdissection (LCM) is a method to procure subpopulations of tissue cells under direct microscopic visualization.Missing: details | Show results with:details
  36. [36]
    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.
  37. [37]
    Laser capture microdissection: Big data from small samples - PMC
    Dec 1, 2015 · Advantages and Limitations of LCM. The most important advantages of LCM are its speed, precision, and versatility. Depending on the laser ...
  38. [38]
    Laser capture microdissection for biomedical research: towards high ...
    Aug 5, 2023 · LCM has several advantages, including automatic sample isolation with specific optional laser parameters settings (such as intensity, speed, and ...Missing: limitations | Show results with:limitations
  39. [39]
    NanoString Announces Commercial Launch of GeoMx Digital ...
    Mar 27, 2019 · NanoString Announces Commercial Launch of GeoMx Digital Spatial Profiler at the 2019 American Association of Cancer Research Conference.
  40. [40]
    None
    ### Summary of GeoMx Digital Spatial Profiler (DSP) from Brochure
  41. [41]
  42. [42]
    Spatial Multiomics Enabled with GeoMx DSP - NanoString
    GeoMx DSP lets you decide where to draw the line and which cell types and tissue structures to enrich for with morphology marker guided region of interest (ROI) ...
  43. [43]
    GeoMx Applications - NanoString
    GeoMx DSP is used for spatial profiling, biomarker discovery, and pathway analysis in oncology, immunology, neuroscience, and developmental biology.
  44. [44]
  45. [45]
    Tomo-seq: A method to obtain genome-wide expression ... - PubMed
    Here, we describe tomo-seq, a technique that combines the advantages of the above-mentioned approaches and provides genome-wide expression data with spatial ...Missing: first paper Visel
  46. [46]
    Tomo-seq: A method to obtain genome-wide expression data with ...
    Tomo-seq provides genome-wide sensitivity with high spatial resolution and can be used to identify genes that are upregulated in a region of interest.Missing: paper | Show results with:paper
  47. [47]
    RNA Tomography for Spatially Resolved Transcriptomics (Tomo-Seq)
    Here, we describe tomo-seq, a method that combines classical histological sectioning of embryos or tissues with a highly sensitive RNA-sequencing technique.Missing: first paper Visel 2014
  48. [48]
    LCM-Seq: A Method for Spatial Transcriptomic Profiling Using Laser ...
    LCM-seq couples laser capture microdissection of cells from frozen tissues with polyA-based RNA sequencing and is applicable to single neurons.
  49. [49]
    Development of a versatile LCM-Seq method for spatial ... - PubMed
    Aug 1, 2023 · We report an LCM-Seq method which allows deep transcriptome profiling of fluorescently-tagged neuron populations isolated with laser-capture ...
  50. [50]
    tomoseqr: A Bioconductor package for spatial reconstruction and ...
    Jan 8, 2025 · We developed tomoseqr, an R package designed for RNA tomography analysis of tomo-seq data, to reconstruct and visualize 3D gene expression patterns through ...
  51. [51]
    Super-resolution measurement of distance between transcription ...
    Apr 1, 2016 · Increased resolution is attained by localizing signals with high precision (∼20 nm) ... Typical FISH spots are diffraction-limited in size (∼200 nm) ...
  52. [52]
    RNAscope: A Novel in Situ RNA Analysis Platform for Formalin ...
    We describe RNAscope, a novel RNA ISH technology with a unique probe design strategy that allows simultaneous signal amplification and background suppression.
  53. [53]
    Single Molecule Fluorescence In Situ Hybridization (smFISH ... - NIH
    May 25, 2018 · smFISH is a powerful technique to study gene expression in single cells due to its ability to detect and count individual RNA molecules.
  54. [54]
    Single-cell in situ RNA profiling by sequential hybridization - Nature
    Mar 28, 2014 · We used super-resolution microscopy to resolve a large number of mRNAs in single cells. In this Correspondence, we present a sequential barcoding scheme to ...
  55. [55]
    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.
  56. [56]
    Dynamics and Spatial Genomics of the Nascent Transcriptome by ...
    Jul 12, 2018 · Here, we demonstrate a multiplexed single-molecule in situ method, intron seqFISH, that allows imaging of 10,421 genes at their nascent ...
  57. [57]
    High-throughput single-cell gene-expression profiling with ... - PNAS
    Sep 13, 2016 · Here we report advances in multiplexed error-robust fluorescence in situ hybridization (MERFISH) that increase the measurement throughput by two orders of ...Results · Si Materials And Methods · Merfish Imaging<|control11|><|separator|>
  58. [58]
    CosMx Spatial Molecular Imager - NanoString
    High-Plex Single-Cell Spatial CosMx® SMI RNA Assays enable high-plex analysis of over 18000 genes from a single FFPE slide with spatial context at… CosMx ...CosMx® SMI · CosMx SMI Performance · CosMx SMI Technology...Missing: paper | Show results with:paper
  59. [59]
    Third-generation in situ hybridization chain reaction: multiplexed ...
    In situ hybridization based on the mechanism of the hybridization chain reaction (HCR) has addressed multi-decade challenges that impeded imaging of mRNA ...
  60. [60]
    In situ sequencing for RNA analysis in preserved tissue and cells
    Jul 14, 2013 · We have developed an in situ sequencing method for parallel targeted analysis of short RNA fragments in morphologically preserved cells and tissue.
  61. [61]
    Padlock Probe–Based Targeted In Situ Sequencing - Annual Reviews
    Aug 25, 2023 · This paper is an overview of Padlock Probe–Based Targeted In Situ Sequencing, including its methods and applications.Missing: paper | Show results with:paper
  62. [62]
    Efficient in situ barcode sequencing using padlock probe-based ...
    Here we describe BaristaSeq, an optimization of a targeted, padlock probe-based technique for in situ barcode sequencing compatible with Illumina sequencing ...
  63. [63]
    Highly multiplexed subcellular RNA sequencing in situ - PMC - NIH
    Here we describe fluorescent in situ RNA sequencing (FISSEQ), in which stably cross-linked cDNA amplicons are sequenced within a biological sample.
  64. [64]
    Fluorescent in situ sequencing (FISSEQ) of RNA for gene ... - NIH
    Here we detail a protocol for genome-wide profiling of gene expression in situ in fixed cells and tissues, in which RNA is converted into cross-linked cDNA ...
  65. [65]
    Three-dimensional intact-tissue sequencing of single-cell ... - Science
    Jul 27, 2018 · STARmap defines a platform for 3D in situ transcriptomics, enabled by state-of-the-art DNA library preparation and sequencing in an HTC ...
  66. [66]
    Nanoscale Imaging of RNA with Expansion Microscopy - PMC - NIH
    We show that this procedure, which we call ExFISH, enables RNA fluorescent in situ hybridization (FISH), which enables identification of transcripts in situ ...
  67. [67]
    Multiplexed spatial transcriptomics methods and the application of ...
    In this review, we elaborate on ISS, ISH and NGS-based spatial transcriptomic protocols and on how performance of these techniques can be extended.Abstract · Methods in multiplexed spatial... · Strengths and limitations · ConclusionMissing: hematology | Show results with:hematology
  68. [68]
    Scalable spatial single-cell transcriptomics and translatomics in 3D ...
    Here, we developed Deep-STARmap and Deep-RIBOmap, which enable 3D in situ quantification of thousands of gene transcripts and their corresponding translation ...Missing: paper | Show results with:paper
  69. [69]
    Slide-seq: A scalable technology for measuring genome-wide ...
    Mar 29, 2019 · We developed Slide-seq, a method for transferring RNA from tissue sections onto a surface covered in DNA-barcoded beads with known positions, ...
  70. [70]
    Slide-seq: A Scalable Technology for Measuring Genome-Wide ...
    Slide-seq provides a scalable method for obtaining spatially resolved gene expression data at resolutions comparable to the sizes of individual cells.
  71. [71]
    High-definition spatial transcriptomics for in situ tissue profiling - PMC
    Mar 9, 2020 · A dense, spatially-barcoded bead array captures RNA from histological tissue sections for spatially-resolved gene expression analysis.Missing: original paper
  72. [72]
  73. [73]
    Systematic comparison of sequencing-based spatial transcriptomic ...
    Jul 4, 2024 · Our analysis found that Sst gene expression exhibited more dispersed distribution within each cluster when assessed using Stereo-seq. Larger ...
  74. [74]
    Visium Spatial Assays | 10x Genomics
    The Visium HD assay has superior spatial fidelity compared to other commercially available sequencing-based spatial transcriptomics platforms.
  75. [75]
    Visium Glossary of Terms | Official 10x Genomics Support
    By contrast, Visium v1 and v2 capture areas have an array of ~5,000 barcoded spots, 55 µm in diameter, spaced 100 µm apart. See also: Spatial Barcodes.Missing: transcriptomics | Show results with:transcriptomics
  76. [76]
    Challenges and Opportunities for the Clinical Translation of Spatial ...
    In a turn of events, on February 26, 2024, the European Unified Patent Court (UPC) of Appeals revoked a preliminary injunction and ruled in favor of Nanostring.
  77. [77]
    10x Genomics Visium HD Spatial Gene Expression Assay
    Mar 26, 2024 · Using 2-µm2 gapless tiles to barcode transcripts, the assay enables researchers to measure the whole transcriptome from formalin-fixed paraffin- ...
  78. [78]
    Your introduction to Visium HD: Spatial biology in high definition
    Apr 19, 2024 · A spatially resolved sequencing-based whole transcriptome gene expression readout is output at 2 µm and provided at multiple bin sizes as well.Missing: gapless | Show results with:gapless
  79. [79]
    A practical guide for choosing an optimal spatial transcriptomics ...
    Jan 20, 2025 · In this paper, we will analyze and compare seven major commercially available spatial platforms to guide researchers in choosing the most suitable option for ...
  80. [80]
    Landscape of Sequencing-based Spatial RNA Technology: Part I
    Feb 27, 2025 · Slide-seq (Rodriques et al.), introduced in 2019, was designed to improve the resolution of the original Visium assay. Instead of using a glass ...
  81. [81]
  82. [82]
  83. [83]
    Visium Spatial Platform | 10x Genomics
    The Visium platform delivers unbiased, whole transcriptome spatial gene expression analysis at single cell scale with unmatched spatial data quality.Missing: arrays 55
  84. [84]
    Statistical and machine learning methods for spatially resolved ...
    Mar 25, 2022 · A spatial transcriptomics data analysis workflow typically includes multiple phases (Fig. 1E). The first step is data preprocessing, which ...
  85. [85]
    The Drosophila embryo at single-cell transcriptome resolution
    Aug 31, 2017 · Karaiskos et al. developed an algorithm to generate an interactive three-dimensional (3D) “virtual embryo,” with the expression of more than 8000 genes per ...Abstract · The Virtual Embryo Predicts... · Cell Communication By...
  86. [86]
    Integration of spatial and single-cell transcriptomic data elucidates ...
    Sep 6, 2021 · We apply an image-based single-cell transcriptomics method, sequential fluorescence in situ hybridization (seqFISH), to detect mRNAs for 387 target genes in ...
  87. [87]
    Computational Approaches and Challenges in Spatial Transcriptomics
    Here, we review computational approaches to these problems in light of corresponding issues and challenges, and present forward-looking insights into algorithm ...<|separator|>
  88. [88]
    Single-cell and spatial transcriptomics integration: new frontiers in ...
    The interaction between immune-mediated tumor editing and cancer cell immune evasion influences disease progression and therapeutic outcomes (82, 83). 3.1 Cells ...<|control11|><|separator|>
  89. [89]
    Advances in spatial transcriptomics and related data analysis ...
    May 18, 2023 · In this review, we summarize available technologies of spatial transcriptomics, probe into several applications, discuss the computational strategies and raise ...<|control11|><|separator|>
  90. [90]
    Analysis and Visualization of Spatial Transcriptomic Data - PMC
    A first step in the spatial transcriptomic analysis is to identify the cell type (for datasets of single-cell resolution) or cell mixture (for datasets of ...
  91. [91]
    Analysis, visualization, and integration of spatial datasets with Seurat
    Jun 17, 2025 · This tutorial demonstrates how to use Seurat (>=3.2) to analyze spatially-resolved RNA-seq data. While the analytical pipelines are similar to the Seurat ...10x Visium · Interactive Plotting · Slide-Seq
  92. [92]
    [PDF] SpatialExperiment - bioRxiv
    Jan 31, 2022 · Summary: SpatialExperiment is a new data infrastructure for storing and accessing spatially resolved transcriptomics data, implemented within ...
  93. [93]
    spatially-aware normalization for spatial transcriptomics data - NIH
    Apr 29, 2025 · We develop SpaNorm, the first spatially-aware normalization method that concurrently models library size effects and the underlying biology.
  94. [94]
    Spatially informed clustering, integration, and deconvolution of ...
    Mar 1, 2023 · We present GraphST, a graph self-supervised contrastive learning method that fully exploits spatial transcriptomics data to outperform existing methods.
  95. [95]
    Benchmarking clustering, alignment, and integration methods for ...
    Aug 9, 2024 · SpatialPCA is a dimension reduction method aimed at extracting a low-dimensional representation of ST data using spatial correlation information ...
  96. [96]
    A comprehensive comparison on clustering methods for multi-slice ...
    Sep 18, 2025 · Clustering or spatial domain detection represents an essential methodology for ST data, facilitating the exploration of spatial organizations ...
  97. [97]
    Evaluating spatially variable gene detection methods for spatial ...
    Jan 15, 2024 · Moran's I. Moran's I ranks genes by the observed spatial autocorrelation [19, 20] to measure the dependence of a feature on spatial location.
  98. [98]
    Compute Moran's I score — squidpy documentation - Read the Docs
    The Moran's I global spatial auto-correlation statistics evaluates whether features (i.e. genes) shows a pattern that is clustered, dispersed or random in the ...
  99. [99]
    infrastructure for spatially-resolved transcriptomics data in R using ...
    May 26, 2022 · SpatialExperiment is a new data infrastructure for storing and accessing spatially-resolved transcriptomics data, implemented within the R/Bioconductor ...
  100. [100]
    Squidpy: a scalable framework for spatial omics analysis - Nature
    Jan 31, 2022 · A Python framework that brings together tools from omics and image analysis to enable scalable description of spatial molecular data.
  101. [101]
    A unified pipeline for spatial transcriptomics data analysis - bioRxiv
    Sep 23, 2025 · Results Here, we present SpaceSequest, a tailored pipeline that utilizes cutting-edge computational methodologies to conduct a thorough analysis ...
  102. [102]
    MOFA+: a statistical framework for comprehensive integration of ...
    May 11, 2020 · We present Multi-Omics Factor Analysis v2 (MOFA+), a statistical framework for the comprehensive and scalable integration of single-cell multi-modal data.
  103. [103]
    Advancements in single-cell RNA sequencing and spatial ...
    Feb 4, 2025 · By preserving tissue architecture, spatial transcriptomics allows the mapping of key cellular processes like angiogenesis, immune evasion, and ...