frontier-banner
Frontiers
Home>Frontiers>

Nature Genetics | Single-cell and spatial transcriptomics reveal a fibrosis-associated network in Crohn's disease

Nature Genetics | Single-cell and spatial transcriptomics reveal a fibrosis-associated network in Crohn's disease
--

This study comprehensively解析es the cellular composition, gene expression characteristics, and spatial organization of intestinal fibrosis in Crohn's disease using single-cell and spatial transcriptomic technologies. It reveals the interaction network between inflammatory-associated fibroblasts (IAF), immune cells, and endothelial cells, offering new insights into the mechanisms and therapeutic strategies for fibrotic diseases.

 

Literature Overview

This article, 'Single-cell and spatial transcriptomics of stricturing Crohn’s disease highlights a fibrosis-associated network', published in the journal Nature Genetics, reviews and summarizes the cellular and gene expression atlas of Crohn’s disease (CD)-related intestinal fibrosis. Based on 61 samples covering 21 CD patients and 10 non-IBD controls, the study utilized single-cell RNA sequencing (scRNA-seq) and spatial transcriptomic technology (Visium) to reveal dynamic changes in fibroblasts, immune cells, and the enteric nervous system in fibrotic regions. It also identifies tissue microenvironments highly correlated with Crohn's disease risk gene modules.

Background Knowledge

Crohn's disease is a chronic inflammatory bowel disease often accompanied by fibrotic complications following prolonged inflammation, leading to intestinal lumen stenosis and functional impairment. Currently, there are limited effective anti-fibrotic treatment options, and traditional mouse models fail to accurately replicate the complex pathology of human CD. In recent years, single-cell sequencing and spatial transcriptomic techniques have emerged as critical tools for analyzing disease microenvironments, enabling the identification of cell subtypes, their gene expression programs, and intercellular communication networks. This study combines both omics approaches to reveal the spatial organization of fibrotic Crohn's disease tissue at single-cell resolution and identifies multicellular networks associated with disease progression, offering novel insights into understanding intestinal fibrosis mechanisms.

 

 

Research Methods and Experiments

The study included 61 intestinal tissue samples from 21 CD patients and 10 non-IBD controls, including biopsies and surgically resected tissues. The samples were classified into four categories—non-IBD, non-stricturing, inflamed non-stricturing, and stricturing—based on macroscopic pathology and endoscopic scoring. scRNA-seq was performed to isolate and sequence epithelial and non-epithelial cells, while spatial transcriptomics (Visium) was used to map gene expression within tissue sections. Cellular composition changes and gene expression patterns were evaluated using Bray-Curtis dissimilarity analysis, Dirichlet regression, and Lasso regression modeling. Additionally, an inflammatory-associated fibroblast (IAF) scoring system was developed to identify core cellular programs associated with fibrosis.

Key Conclusions and Perspectives

  • IgG+ plasma cells, CCR7-hi CD4+ T cells, and inflammatory fibroblasts are significantly increased in stricturing tissues, whereas intestinal epithelial cells and M2 macrophages are decreased
  • Spatial transcriptomic analysis reveals enrichment of tertiary lymphoid structures (TLS) in stricturing tissues, showing co-localization of B cells, Tfh cells, and dendritic cells
  • Fibroblast subtypes exhibit distinct spatial distributions: IL11+ inflammatory fibroblasts accumulate in epithelial injury zones, while COL1A1+ collagen-high fibroblasts localize near the muscularis and serosa layers
  • IAF scores are significantly correlated with GZMB-hi pDCs, T cell proliferation, and Tfh cell frequency, indicating their role in local immune activation
  • Spatial analysis shows that CD risk genes exhibit specific enrichment in epithelial, lymphoid follicle, and inflammatory regions, with enhanced expression when co-localized with T cells or fibroblasts

Research Significance and Prospects

This study, for the first time at single-cell and spatial resolution, reveals the cellular composition and gene networks of intestinal fibrosis in Crohn’s disease, highlighting the central role of fibroblast-immune cell interactions in fibrosis. Future research may further investigate the dynamic regulatory mechanisms of IAF programs and validate subcellular RNA localization using high-resolution spatial techniques to identify potential therapeutic targets for anti-fibrotic treatment. Additionally, integrating multi-omics and longitudinal studies could track fibrosis progression and evaluate molecular signatures of therapeutic responses.

 

 

Conclusion

This study systematically characterizes cellular heterogeneity and gene expression programs in stricturing Crohn's disease tissues through integrated single-cell and spatial transcriptomic approaches. It identifies fibroblast subtypes and their spatial localization patterns that are highly associated with intestinal fibrosis. The study also reveals gene module enrichment in tertiary lymphoid structures and spatially specific expression of CD risk genes, providing key clues for fibrosis mechanism research and therapeutic interventions. These findings not only deepen our understanding of CD-associated fibrosis but also provide an analytical framework applicable to other fibrotic diseases such as pulmonary fibrosis and cirrhosis.

 

Reference:
Lingjia Kong, Sathish Subramanian, Åsa Segerstolpe, Christopher S Smillie, and Ramnik J Xavier. Single-cell and spatial transcriptomics of stricturing Crohn’s disease highlights a fibrosis-associated network. Nature genetics.
Humanization
Monoclonal antibody therapeutics typically originate from non-human sources (usually mice), which may trigger immune responses in humans. Antibody humanization aims to modify the variable region sequences of antibodies to obtain antibodies that do not elicit immune responses. We utilized nearly one billion antibody sequences from the OAS database to establish an antibody humanness evaluation AI model capable of distinguishing between human and non-human antibody variable region sequences. The scores output by the model are negatively correlated with the experimental immunogenicity (ADA) of existing FDA-approved antibody therapies. Following the approach of Marks and Hummer, we combined this model with a Beam Search algorithm to develop an antibody sequence humanization tool. This tool aims to maximize the level of humaness of antibodies while minimizing number of mutations and maintaining key characteristics such as affinity, thereby reducing their immunogenicity.