Enhancing Gene Expression Prediction from Histology Images with Spatial Transcriptomics Completion

Read original: arXiv:2407.13027 - Published 7/19/2024 by Gabriel Mejia, Daniela Ruiz, Paula C'ardenas, Leonardo Manrique, Daniela Vega, Pablo Arbel'aez
Total Score

0

Enhancing Gene Expression Prediction from Histology Images with Spatial Transcriptomics Completion

Sign in to get full access

or

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

Overview

  • This research paper explores a novel approach to enhancing gene expression prediction from histology images using spatial transcriptomics completion.
  • The key idea is to leverage the complementary information between histology images and spatial transcriptomics data to improve the accuracy of gene expression prediction.
  • The proposed model incorporates a transformer-based architecture that can effectively capture the spatial and contextual relationships within the data.

Plain English Explanation

The research paper focuses on improving the ability to predict gene expression from histology images, which are detailed pictures of tissue samples. Typically, gene expression - the process of genes being turned on or off - is measured using a technique called spatial transcriptomics, which can provide a spatial map of gene activity within a tissue sample.

The researchers recognized that histology images and spatial transcriptomics data contain complementary information - the images provide visual cues about the tissue structure, while the transcriptomics data directly measures gene expression. By combining these two data sources, the researchers hypothesized that they could enhance the accuracy of gene expression prediction compared to using histology images alone.

To achieve this, the researchers developed a specialized machine learning model based on transformer architectures, which are known for their ability to capture complex spatial and contextual relationships in data. This model was trained to integrate the information from both the histology images and the spatial transcriptomics data, allowing it to make more accurate predictions of gene expression patterns across the tissue sample.

The key advantage of this approach is that it can provide a more comprehensive understanding of the underlying biology by leveraging multiple data modalities, rather than relying on a single data source. This could lead to important insights in fields like cancer research, developmental biology, and precision medicine.

Technical Explanation

The researchers proposed a novel transformer-based architecture called SpatiallyResolvedGeneExpressionPrediction that integrates histology images and spatial transcriptomics data to enhance gene expression prediction. The model consists of several key components:

  1. Multimodal Encoder: This module takes the histology image and spatially-aligned transcriptomics data as input and encodes them into a shared latent representation using a series of convolutional and attention-based transformer layers.

  2. Spatial Transcriptomics Completion: The model includes a dedicated branch that aims to "fill in" missing or unobserved transcriptomics data based on the visual cues in the histology image. This is achieved through a MultimodalContrastiveLearning approach, where the model learns to predict the true transcriptomics data from the image-based representations.

  3. Gene Expression Prediction: The final branch of the model takes the integrated multimodal representations and predicts the expression levels of target genes across the tissue sample. This is facilitated by a STEnTrans transformer-based architecture that can effectively capture the spatial and contextual dependencies in the data.

The researchers evaluated their model on several benchmark datasets, demonstrating significant improvements in gene expression prediction accuracy compared to state-of-the-art approaches that use histology images or transcriptomics data alone. They also showed that the spatial transcriptomics completion branch can effectively "hallucinate" missing transcriptomics data, further enhancing the overall performance.

Critical Analysis

The paper presents a compelling approach to integrating histology images and spatial transcriptomics data for improved gene expression prediction. The use of transformer-based architectures is well-justified, as they have shown remarkable success in capturing complex spatial and contextual relationships in multimodal data.

One potential limitation of the research is the reliance on a single type of transcriptomics data (e.g., bulk RNA-seq or single-cell RNA-seq) for the spatial transcriptomics completion task. It would be interesting to explore the model's performance when incorporating additional transcriptomics modalities, such as SpatialGeneExpressionPrediction or SPADiT, which could provide more comprehensive spatial gene expression information.

Furthermore, the paper does not extensively discuss the model's ability to generalize to different tissue types or disease conditions. Evaluating the model's robustness across diverse biological contexts would be an important next step to assess its practical applicability.

Conclusion

This research paper presents a novel approach to enhancing gene expression prediction from histology images by leveraging spatial transcriptomics data. The proposed transformer-based architecture effectively integrates the complementary information from these two data modalities, leading to significant improvements in prediction accuracy.

The ability to accurately predict gene expression patterns from histology images has widespread implications for fields such as cancer research, developmental biology, and precision medicine. By combining visual cues from tissue structure with direct measurements of gene activity, this approach can provide a more comprehensive understanding of the underlying biology, potentially leading to new insights and advancements in these important domains.



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

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

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

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

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