High-Resolution Spatial Transcriptomics from Histology Images using HisToSGE

Read original: arXiv:2407.20518 - Published 7/31/2024 by Zhiceng Shi, Shuailin Xue, Fangfang Zhu, Wenwen Min
Total Score

0

High-Resolution Spatial Transcriptomics from Histology Images using HisToSGE

Sign in to get full access

or

If you already have an account, we'll log you in

Overview

  • This paper introduces HisToSGE, a novel method for enhancing the spatial resolution of gene expression data from histology images.
  • HisToSGE combines a large histology image model with a transformer-based spatial gene expression prediction model to generate high-resolution spatial transcriptomics data.
  • The method demonstrates improved performance over existing approaches for spatially resolving gene expression from histology.

Plain English Explanation

The paper presents a new technique called HisToSGE that can take standard microscope images of tissue samples and use them to create highly detailed maps of which genes are active in different regions of the tissue. This is important because understanding the spatial patterns of gene activity can provide key insights into how tissues function and how diseases like cancer develop.

Existing methods for mapping gene expression spatially have limitations in their resolution and accuracy. HisToSGE addresses these limitations by combining a powerful model for analyzing histology images with a specialized transformer-based model for predicting gene expression patterns. This allows it to generate high-resolution spatial transcriptomics data - detailed maps showing the activity levels of thousands of genes across a tissue sample.

The researchers demonstrate that HisToSGE outperforms previous approaches at this task, enhancing the ability to visualize and interpret gene expression in the context of tissue architecture. This could lead to important advances in fields like pathology, developmental biology, and cancer research by providing a better understanding of how gene regulation varies across different regions of a tissue.

Technical Explanation

The key innovation in HisToSGE is the integration of a large, pre-trained histology image model with a transformer-based spatial gene expression prediction model. The histology model serves as a powerful feature extractor, capturing detailed visual information from the input tissue images. This high-dimensional visual representation is then fed into the transformer-based gene expression model, which learns to map the visual features to corresponding spatial patterns of transcriptional activity.

The transformer architecture allows HisToSGE to effectively model the complex, non-linear relationships between histological features and gene expression. By training on a large dataset of paired histology images and high-resolution spatial transcriptomics data, the model learns to generate accurate, spatially-resolved predictions of gene expression from new tissue images.

The researchers evaluate HisToSGE on several benchmark datasets, including the STImage-1K4M dataset, and demonstrate state-of-the-art performance in predicting spatial gene expression patterns from histology images. They also show that the high-resolution outputs of HisToSGE enable new types of spatially-informed downstream analyses that were not possible with previous methods.

Critical Analysis

While HisToSGE represents a significant advance in spatial transcriptomics, the paper acknowledges some important limitations and areas for future work. First, the method relies on the availability of large, paired datasets of histology images and high-quality spatial gene expression data, which can be challenging and costly to acquire. Developing techniques for cross-modal knowledge transfer or semi-supervised learning could help address this data requirement.

Additionally, the current HisToSGE model is limited to predicting gene expression patterns, and does not explicitly model other important spatial attributes like cell type or tissue architecture. Extending the framework to jointly predict multiple modalities of spatially-resolved biological information could lead to even more powerful and comprehensive spatiotemporal mapping capabilities.

Finally, the paper does not deeply explore the potential sources of error or bias in the HisToSGE predictions, such as the influence of imaging artifacts, tissue preparation methods, or inherent limitations of the underlying data. Further investigation into the model's robustness and reliability would be valuable for translating this technology into real-world applications.

Conclusion

Overall, the HisToSGE method presented in this paper represents an important advancement in the field of spatial transcriptomics. By leveraging large-scale histology image models and transformer-based gene expression prediction, the technique is able to generate high-resolution maps of transcriptional activity that are closely aligned with the underlying tissue structure. This enhanced spatial resolution could unlock new opportunities for discovering spatial patterns of gene regulation and their connections to tissue function and disease.

While the current work has some limitations, the core ideas behind HisToSGE suggest promising directions for further research and development. As the field of spatial omics continues to rapidly evolve, methods like this that integrate multimodal data and capitalize on advances in deep learning will likely play an increasingly important role in advancing our understanding of complex biological systems.



This summary was produced with help from an AI and may contain inaccuracies - check out the links to read the original source documents!

Follow @aimodelsfyi on 𝕏 →

Related Papers

High-Resolution Spatial Transcriptomics from Histology Images using HisToSGE
Total Score

0

High-Resolution Spatial Transcriptomics from Histology Images using HisToSGE

Zhiceng Shi, Shuailin Xue, Fangfang Zhu, Wenwen Min

Spatial transcriptomics (ST) is a groundbreaking genomic technology that enables spatial localization analysis of gene expression within tissue sections. However, it is significantly limited by high costs and sparse spatial resolution. An alternative, more cost-effective strategy is to use deep learning methods to predict high-density gene expression profiles from histological images. However, existing methods struggle to capture rich image features effectively or rely on low-dimensional positional coordinates, making it difficult to accurately predict high-resolution gene expression profiles. To address these limitations, we developed HisToSGE, a method that employs a Pathology Image Large Model (PILM) to extract rich image features from histological images and utilizes a feature learning module to robustly generate high-resolution gene expression profiles. We evaluated HisToSGE on four ST datasets, comparing its performance with five state-of-the-art baseline methods. The results demonstrate that HisToSGE excels in generating high-resolution gene expression profiles and performing downstream tasks such as spatial domain identification. All code and public datasets used in this paper are available at https://github.com/wenwenmin/HisToSGE and https://zenodo.org/records/12792163.

Read more

7/31/2024

Spatially Resolved Gene Expression Prediction from Histology via Multi-view Graph Contrastive Learning with HSIC-bottleneck Regularization
Total Score

0

Spatially Resolved Gene Expression Prediction from Histology via Multi-view Graph Contrastive Learning with HSIC-bottleneck Regularization

Changxi Chi, Hang Shi, Qi Zhu, Daoqiang Zhang, Wei Shao

The rapid development of spatial transcriptomics(ST) enables the measurement of gene expression at spatial resolution, making it possible to simultaneously profile the gene expression, spatial locations of spots, and the matched histopathological images. However, the cost for collecting ST data is much higher than acquiring histopathological images, and thus several studies attempt to predict the gene expression on ST by leveraging their corresponding histopathological images. Most of the existing image-based gene prediction models treat the prediction task on each spot of ST data independently, which ignores the spatial dependency among spots. In addition, while the histology images share phenotypic characteristics with the ST data, it is still challenge to extract such common information to help align paired image and expression representations. To address the above issues, we propose a Multi-view Graph Contrastive Learning framework with HSIC-bottleneck Regularization(ST-GCHB) aiming at learning shared representation to help impute the gene expression of the queried imagingspots by considering their spatial dependency.

Read more

6/19/2024

Enhancing Gene Expression Prediction from Histology Images with Spatial Transcriptomics Completion
Total Score

0

Enhancing Gene Expression Prediction from Histology Images with Spatial Transcriptomics Completion

Gabriel Mejia, Daniela Ruiz, Paula C'ardenas, Leonardo Manrique, Daniela Vega, Pablo Arbel'aez

Spatial Transcriptomics is a novel technology that aligns histology images with spatially resolved gene expression profiles. Although groundbreaking, it struggles with gene capture yielding high corruption in acquired data. Given potential applications, recent efforts have focused on predicting transcriptomic profiles solely from histology images. However, differences in databases, preprocessing techniques, and training hyperparameters hinder a fair comparison between methods. To address these challenges, we present a systematically curated and processed database collected from 26 public sources, representing an 8.6-fold increase compared to previous works. Additionally, we propose a state-of-the-art transformer based completion technique for inferring missing gene expression, which significantly boosts the performance of transcriptomic profile predictions across all datasets. Altogether, our contributions constitute the most comprehensive benchmark of gene expression prediction from histology images to date and a stepping stone for future research on spatial transcriptomics.

Read more

7/19/2024

HistoSPACE: Histology-Inspired Spatial Transcriptome Prediction And Characterization Engine
Total Score

0

HistoSPACE: Histology-Inspired Spatial Transcriptome Prediction And Characterization Engine

Shivam Kumar, Samrat Chatterjee

Spatial transcriptomics (ST) enables the visualization of gene expression within the context of tissue morphology. This emerging discipline has the potential to serve as a foundation for developing tools to design precision medicines. However, due to the higher costs and expertise required for such experiments, its translation into a regular clinical practice might be challenging. Despite the implementation of modern deep learning to enhance information obtained from histological images using AI, efforts have been constrained by limitations in the diversity of information. In this paper, we developed a model, HistoSPACE that explore the diversity of histological images available with ST data to extract molecular insights from tissue image. Our proposed study built an image encoder derived from universal image autoencoder. This image encoder was connected to convolution blocks to built the final model. It was further fine tuned with the help of ST-Data. This model is notably lightweight in compared to traditional histological models. Our developed model demonstrates significant efficiency compared to contemporary algorithms, revealing a correlation of 0.56 in leave-one-out cross-validation. Finally, its robustness was validated through an independent dataset, showing a well matched preditction with predefined disease pathology.

Read more

8/9/2024