Visium HD Analysis Tutorial: A Comprehensive Guide
This tutorial delves into Visium HD spatial transcriptomics, covering data acquisition, processing with Loupe Browser, and advanced analysis using LatchBio.
Explore gene expression mapping, tissue segmentation, and cell type deconvolution, alongside comparisons to other technologies like 10x Visium.

Visium HD represents a significant advancement in spatial transcriptomics, enabling researchers to analyze gene expression patterns within intact tissues at an unprecedented resolution – approaching single-cell fidelity. Unlike traditional RNA sequencing, which loses spatial context, Visium HD preserves the location of each transcript, providing a comprehensive view of cellular organization and interactions.
This technology utilizes a spatially resolved barcoding approach, capturing mRNA from tissue sections directly on a slide. The resulting data reveals transcriptome-wide gene expression alongside precise spatial coordinates. This allows for the investigation of how gene expression varies across different tissue compartments, identifying spatially distinct cell populations, and uncovering novel biological insights.
Visium HD builds upon the foundation of earlier Visium technology, offering dramatically improved resolution and sensitivity. It’s becoming increasingly vital for understanding complex biological processes in development, disease, and response to therapies. The ability to integrate this data with histological images further enhances the interpretability and impact of spatial transcriptomic studies, as demonstrated in recent research mapping gene expression in the epipharyngeal mucosa.
Understanding the Visium HD Workflow
The Visium HD workflow encompasses several key stages, beginning with fresh-frozen tissue sectioning and mounting onto a specialized slide. This slide contains spatially barcoded oligo probes designed to capture mRNA molecules released from the tissue. Following tissue preparation, the slide undergoes hybridization, where mRNA binds to the barcoded probes.
Next, the captured mRNA is converted into cDNA, amplified, and sequenced using Illumina platforms. The resulting sequencing data contains both gene expression information and spatial coordinates, encoded by the barcode associated with each transcript. Initial data processing involves demultiplexing, alignment, and quality control steps.

Crucially, the workflow integrates seamlessly with Loupe Browser for initial visualization and processing. Advanced analysis often leverages tools like LatchBio, providing infrastructure for large-scale spatial data management and analysis. The entire process, from tissue preparation to data interpretation, requires careful attention to detail to ensure accurate and reliable results, as highlighted by recent advancements from 10x Genomics.

Visium HD Data Acquisition and Quality Control
Visium HD data acquisition begins with sequencing on Illumina platforms, generating FASTQ files containing read data. Initial quality control (QC) is paramount, assessing read quality scores, library complexity, and potential contamination. Demultiplexing separates reads based on sample barcodes, preparing them for alignment.
Alignment to the reference genome is a critical step, utilizing tools to map reads to their genomic origin while preserving spatial information encoded in the barcodes. Post-alignment QC focuses on mapping rates, unique molecular identifier (UMI) counts, and the detection of potential batch effects. Low-quality reads or poorly mapped transcripts are filtered out.
Visium HD’s high resolution necessitates stringent QC to ensure data accuracy. Metrics like the number of genes detected per spot and the distribution of UMI counts are evaluated. Outlier spots exhibiting aberrant expression patterns are identified and potentially removed. Proper QC ensures reliable downstream analysis, as emphasized by advancements in spatial genomics reported in the Global Strategic Business Report.
Data Import and Initial Processing in Loupe Browser
Loupe Browser, 10x Genomics’ software, is central to Visium HD data processing. Importing the processed data – typically a CLoupe file generated from Cell Ranger – initiates the workflow. The software visualizes the spatial gene expression data, displaying spots overlaid on the tissue image.
Initial processing involves filtering, normalization, and visualization. Filtering removes low-quality spots based on metrics like UMI counts and gene detection. Normalization accounts for variations in sequencing depth across spots, ensuring comparable gene expression levels. Loupe Browser facilitates interactive exploration of the data, allowing users to inspect gene expression within individual spots and across the tissue section.
Key features include the ability to view gene expression heatmaps, identify highly variable genes, and perform basic spatial clustering. Users can also overlay histological data, such as H&E staining, to correlate gene expression patterns with tissue morphology. This initial exploration sets the stage for more advanced analysis, as demonstrated in applications mapping gene expression to cellular locations.
Image Alignment and Tissue Segmentation
Precise image alignment is crucial for accurate spatial transcriptomics analysis. Visium HD data requires aligning the gene expression spots to the corresponding tissue morphology, typically obtained from histology images like H&E staining. Loupe Browser provides tools for this alignment process, utilizing image registration algorithms to map spots to the tissue.

Following alignment, tissue segmentation delineates different anatomical regions or structures within the tissue. This can be performed manually, drawing regions of interest (ROIs) around specific areas, or semi-automatically using image segmentation algorithms. Accurate segmentation is vital for focused analysis, allowing researchers to compare gene expression patterns between distinct tissue compartments.
Effective segmentation enhances downstream analysis, such as differential gene expression, by isolating gene expression data to specific regions. The quality of alignment and segmentation directly impacts the reliability of spatial gene expression patterns identified, ensuring accurate correlation between gene activity and tissue architecture. Proper segmentation is key to understanding spatial biology.
Spatial Domain Identification and Clustering
After image alignment and tissue segmentation, identifying spatial domains – regions exhibiting similar gene expression profiles – is a key step. Loupe Browser and platforms like LatchBio employ clustering algorithms to group spots based on their transcriptomic similarity. These algorithms, often utilizing dimensionality reduction techniques, reveal spatially coherent patterns of gene expression.
Clustering methods categorize spots into distinct domains, representing potentially different cell types, tissue structures, or functional regions. Parameters like the number of clusters and the distance metric significantly influence the resulting domain organization. Visualizing these clusters overlaid on the tissue image reveals the spatial distribution of gene expression patterns.
Identifying these domains allows researchers to move beyond individual gene expression levels and understand how genes are co-regulated within specific spatial contexts. Further analysis can then focus on characterizing the biological functions associated with each identified spatial domain, providing insights into tissue organization and function.
Differential Gene Expression Analysis
Differential gene expression (DGE) analysis identifies genes with significantly altered expression levels between defined spatial domains or conditions. Utilizing tools within Loupe Browser or dedicated platforms like LatchBio, researchers can compare gene expression profiles across different regions of the tissue. This process is crucial for pinpointing genes driving observed spatial differences.
Statistical methods, such as negative binomial models, are commonly employed to account for the discrete nature of RNA sequencing data and identify genes exhibiting statistically significant expression changes. Adjustments for multiple testing are essential to control the false discovery rate and ensure reliable results. The analysis considers factors like spot count and library size normalization.
Results are typically presented as fold changes and adjusted p-values, highlighting genes that are upregulated or downregulated in specific spatial domains. Identifying these differentially expressed genes provides valuable clues about the biological processes occurring within those regions, furthering our understanding of tissue function.

Gene Ontology (GO) Enrichment Analysis
Gene Ontology (GO) enrichment analysis is a vital step following differential gene expression analysis. It helps interpret the biological meaning of identified gene lists by determining whether specific GO terms – representing biological processes, molecular functions, and cellular components – are overrepresented within the differentially expressed genes.
Tools like those integrated within LatchBio or standalone GO enrichment platforms are used to perform this analysis. The process compares the genes of interest against a comprehensive database of genes annotated with GO terms. Statistical tests, such as hypergeometric tests, assess the significance of the observed enrichment.
Significant GO terms suggest the biological pathways and functions that are actively altered in specific spatial domains. For example, enrichment of terms related to immune response might indicate inflammation in a particular tissue region. This provides a functional context to the spatial transcriptomic data, aiding in hypothesis generation and deeper biological insights.
Visualization of Spatial Gene Expression Patterns
Visualizing spatial gene expression is crucial for interpreting Visium HD data. The Loupe Browser provides initial visualization capabilities, displaying gene expression levels overlaid onto the tissue image. However, more sophisticated visualization techniques often enhance understanding.
Heatmaps are commonly used to represent gene expression across spatial domains, revealing patterns of upregulation or downregulation. Feature plots highlight the expression of specific genes, allowing researchers to pinpoint their localization within the tissue. UMAP or t-SNE plots, colored by gene expression, can reveal spatial clustering patterns.
Furthermore, tools like LatchBio offer advanced visualization options, including interactive spatial atlases and the ability to overlay histological data. These visualizations facilitate the identification of spatially distinct gene expression signatures, revealing how gene activity varies across the tissue landscape and contributing to a deeper understanding of tissue organization and function.
Integration with Histological Data
Integrating histological data with Visium HD spatial transcriptomics provides critical contextual information. Hematoxylin and eosin (H&E) staining, for example, reveals tissue morphology, enabling researchers to correlate gene expression patterns with anatomical structures.
This integration is typically achieved by aligning the histological image with the spatial transcriptomics data during the image alignment step in the Visium HD workflow. The Loupe Browser allows for direct overlay of gene expression data onto the histological image, facilitating visual comparison.
Advanced tools, such as those offered by LatchBio, further enhance this integration, enabling quantitative analysis of gene expression within specific histological features. This combined approach allows for a more comprehensive understanding of tissue biology, linking gene expression changes to specific cellular environments and pathological conditions. Ultimately, this synergy unlocks deeper insights than either modality could provide alone.
Advanced Analysis: Cell Type Deconvolution
Cell type deconvolution is a crucial advanced analysis technique for Visium HD data, addressing the limitation of spatial resolution. Since each Visium HD spot captures transcripts from multiple cells, deconvolution algorithms estimate the proportion of each cell type within each spot.
This process leverages reference datasets – typically single-cell RNA sequencing (scRNA-seq) data – containing gene expression signatures for different cell types. Algorithms then computationally dissect the bulk RNA-seq signal from each Visium HD spot, attributing it to constituent cell populations.
LatchBio provides tools specifically designed for cell type deconvolution, streamlining this complex analysis. Accurate deconvolution reveals spatially resolved cell type distributions, offering insights into tissue organization and cellular interactions. This is particularly valuable for understanding complex tissues and disease microenvironments, revealing how cell type composition varies across the tissue landscape.
Utilizing LatchBio for Visium HD Analysis
LatchBio emerges as a powerful platform for comprehensive Visium HD data analysis, offering a streamlined workflow from raw data to biological insights. It provides white-labeled data infrastructure, analysis tools, and customizable delivery portals, catering to both researchers and solution providers.
LatchBio facilitates efficient data import, quality control, and processing of Visium HD datasets. Its capabilities extend to advanced analyses like cell type deconvolution, spatial domain identification, and differential gene expression. Notably, LatchBio recently released a 25 million cell spatial atlas, demonstrating its capacity for large-scale spatial data integration.

The platform’s user-friendly interface and scalable infrastructure empower researchers to explore complex spatial transcriptomic data with ease. LatchBio’s tools accelerate discovery by automating key analytical steps and providing interactive visualizations, ultimately enhancing the interpretation of Visium HD results and accelerating spatial biology research.
Comparison with Other Spatial Transcriptomics Technologies (e.g., 10x Visium)
Visium HD represents a significant advancement in spatial transcriptomics, particularly when contrasted with its predecessor, the original 10x Visium technology. While both platforms map gene expression within tissues, Visium HD achieves single-cell resolution, a substantial improvement over the original’s spot-based approach.
Traditional Visium utilized spots to capture transcripts, averaging gene expression across multiple cells. Visium HD, however, enables transcriptome-wide spatial gene expression analysis at a much finer granularity. Other spatial transcriptomics methods, like in situ sequencing (ISS), offer alternative approaches, each with unique strengths and limitations regarding resolution, throughput, and cost.
Visium HD balances high resolution with relatively straightforward workflows, making it a compelling option for researchers seeking detailed spatial information. The choice of technology depends on specific research questions, budget constraints, and the desired level of cellular detail.
Troubleshooting Common Analysis Issues

Visium HD data analysis, while powerful, can present challenges. Common issues include low capture efficiency in certain tissue regions, leading to sparse data and impacting downstream analysis. Image alignment failures can occur, particularly with tissues exhibiting significant folds or distortions, requiring careful parameter adjustments within Loupe Browser.

Another frequent problem is batch effects arising from variations in tissue processing or sequencing runs. These necessitate normalization strategies during data processing. Unexpected clustering patterns might indicate issues with tissue segmentation or the presence of contaminating cells.
Furthermore, difficulties can arise during cell type deconvolution, especially with complex tissues. Thorough quality control, including assessment of mitochondrial gene expression and unique molecular identifier (UMI) counts, is crucial. Utilizing resources like LatchBio can aid in identifying and resolving these issues, ensuring robust and reliable results.
Best Practices for Visium HD Data Analysis
To maximize the insights from Visium HD data, adhere to several best practices. Begin with meticulous tissue handling and sectioning to preserve spatial integrity. Prioritize high-quality image acquisition for accurate alignment within Loupe Browser. Implement rigorous quality control measures, filtering out low-quality spots based on UMI counts and mitochondrial gene expression.

Employ appropriate normalization techniques to mitigate batch effects and ensure data comparability. When performing spatial domain identification, carefully consider the resolution and clustering parameters. Validate findings using histological data, such as H&E staining, to confirm biological relevance.
Leverage tools like LatchBio for advanced analysis and visualization. Thorough documentation of each step, from data acquisition to final analysis, is essential for reproducibility. Finally, remember that spatial transcriptomics complements, rather than replaces, traditional RNA sequencing, offering a unique perspective on gene expression.
Future Directions in Visium HD Analysis and Applications
The future of Visium HD analysis is bright, with ongoing developments promising even greater resolution and deeper biological understanding. Expect advancements in computational methods for cell type deconvolution, enabling more precise identification of cellular components within tissues. Integration with multi-omics data, such as proteomics and metabolomics, will provide a holistic view of spatial biology.
Further refinement of LatchBio and similar platforms will streamline workflows and enhance data accessibility. We anticipate expanded applications in disease research, particularly in oncology and neurobiology, to unravel complex spatial patterns driving disease progression.
The development of new staining techniques compatible with Visium HD will allow for simultaneous detection of gene expression and protein localization. Ultimately, these advancements will empower researchers to explore the intricate interplay between genes, cells, and their microenvironment with unprecedented detail, revolutionizing our understanding of tissue organization and function.