Open Access

Deciphering the spatiotemporal transcriptional landscape of intestinal diseases (Review)

  • Authors:
    • Yajing Guo
    • Chao Ren
    • Yuxi He
    • Yue Wu
    • Xiaojun Yang
  • View Affiliations

  • Published online on: July 4, 2024     https://doi.org/10.3892/mmr.2024.13281
  • Article Number: 157
  • Copyright: © Guo et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

The intestines are the largest barrier organ in the human body. The intestinal barrier plays a crucial role in maintaining the balance of the intestinal environment and protecting the intestines from harmful bacterial invasion. Single‑cell RNA sequencing technology allows the detection of the different cell types in the intestine in two dimensions and the exploration of cell types that have not been fully characterized. The intestinal mucosa is highly complex in structure, and its proper functioning is linked to multiple structures in the proximal‑distal intestinal and luminal‑mucosal axes. Spatial localization is at the core of the efforts to explore the interactions between the complex structures. Spatial transcriptomics (ST) is a method that allows for comprehensive tissue analysis and the acquisition of spatially separated genetic information from individual cells, while preserving their spatial location and interactions. This approach also prevents the loss of fragile cells during tissue disaggregation. The emergence of ST technology allows us to spatially dissect enzymatic processes and interactions between multiple cells, genes, proteins and signals in the intestine. This includes the exchange of oxygen and nutrients in the intestine, different gradients of microbial populations and the role of extracellular matrix proteins. This regionally precise approach to tissue studies is gaining more acceptance and is increasingly applied in the investigation of disease mechanisms related to the gastrointestinal tract. Therefore, this review summarized the application of ST in gastrointestinal diseases.

Introduction

The intestinal tract contains a high concentration of microorganisms and neurons, which have a profound influence on both the physical and mental well-being of humans. The intestines are also known as the ‘second brain’ of the human body, and while they perform their normal functions, various intestinal diseases are inevitably present (13). These diseases can be broadly categorized into two groups: Functional diseases such as irritable bowel syndrome (IBS), functional dyspepsia and functional constipation, and organic diseases such as enterocolitis, ulcerative colitis (UC), Crohn's disease (CD) and various intestinal tumors (46). With the change in dietary habits and lifestyles (such as irregular eating, oversatiety or overhunger) of individuals, the incidence of IBS is on the rise, affecting ~20% of the general population. This not only diminishes the quality of life of individuals with the condition, but also causes a substantial healthcare burden (7,8). Furthermore, the prevalence of inflammatory bowel disease (IBD) has been increasing worldwide over the last century. Due to the impact of family history and weakened immune systems (9), there has been a notable rise in very early onset IBD, which is identified before the age of 6 years (10,11). This results in increased healthcare expenses for patients and can also hinder their career aspirations due to the enduring social stigma they face as they grow up. Chronic inflammation of the colon and accelerated renewal of epithelial cells can increase the risk of highly dysplastic growth, leading to the further development of colorectal cancer (CRC) (12). A previous study revealed that individuals with UC or CD are at a higher risk of developing both gastrointestinal and extraintestinal cancer (13,14). The etiology, severity and advancement of this intricate set of gastrointestinal disorders remain to be evaluated. For example, IBD might initially be misdiagnosed as IBS, leading to a delay in early treatment. The manifestations of IBD are varied and non-specific, with a wide range of differential diagnoses. The possibility of rapid growth and early metastasis of CRC is high, making early detection particularly crucial (1519). Given the prevalence and challenges in diagnosing and treating various gastrointestinal diseases, contemporary medical treatment urgently needs more appropriate and optimized experimental methods to explore and uncover the developmental origins of diseases in order to effectively manage disease progression and provide improved patient care.

The human immune system is a protective system covering the whole body, which can maintain the immune homeostasis of the body, while excluding exogenous antigenic substances (20). The gastrointestinal tract, as the initial line of defense for the immune system, is closely associated with the occurrence of numerous diseases (21). The intestinal barrier has a heterogeneous structure, including mechanical, chemical, microbial and immune barriers (22,23). The intestinal mucosal barrier is the first line of defense against penetration of luminal contents and performs various biological functions (24), not only maintaining local intestinal function, but also facilitating communication with the nervous and endocrine systems via the bidirectional signaling pathways of the brain-gut axis (25). Additionally, the barrier is involved in immune, metabolic and emotional regulation, which is closely linked to the function of intestinal microorganisms (26). The human body, functioning as a superorganism, coexists in symbiosis with trillions of beneficial bacteria and eukaryotic cells. The advancement of the Human Microbiome Project and METAgenomics (27,28) of the human intestinal tract has offered novel technological resources for the research of these microorganisms. There are ~500-1,000 different types of bacteria in the human body, which can reproduce to about 100 trillion individual cells in an adult-roughly 10 times the total number of a person's body cells. These microorganisms benefit from the warm environment of the intestines and serve a crucial role in digestion, vitamin synthesis, immune system regulation, pathogen elimination, toxin removal and maintenance of the normal functioning of the intestines (2932).

Since being awarded ‘Method of the Year 2020’ (33), spatial transcriptomics (ST) has gained popularity as a tool for studying disease mechanisms. When frozen tissue slices are attached to spatial transcriptomics slides, barcode primers bind and capture nearby mRNA from the tissue. The captured mRNA undergoes reverse transcription, resulting in cDNA containing spatial barcodes. By analyzing the sequence of spatial barcodes, the sequence transcribed by each mRNA can be mapped back to the starting position on the tissue slice, providing details of gene expression and its spatial distribution on the tissue slice (34,35). ST is often used in combination with other molecular research techniques such as proteomics, single-cell RNA sequencing (scRNA-seq), intestinal microbiomics and metabolomics. This approach has given researchers novel insights into the complexity of the mucosal immune system, and has provided increased opportunities for understanding intestinal disorders and identifying potential therapeutic targets. ST is the next wave in single-cell analysis, as research on diseases typically begins with single cells and then progresses to the spatial level (33). ST encompasses a range of techniques, such as in situ hybridization and targeted in situ sequencing (ISS). These methods, through multiple fluorescence in situ hybridization (FISH) or ISS, can preserve the three-dimensional spatial structure at the cellular resolution and reveal its functionality (36,37). In addition, there is the spatially-resolved transcript amplicon readout mapping (STARmap) technology, which combines hydrogel histochemistry, targeted signal amplification and ISS (38). These techniques allow exploration of the mechanisms of diseases, not only locally in the gut, but also in the nervous system to help understand brain-gut axis interactions. Fig. 1 illustrates the uses of ST in intestinal research.

In summary, ST can help understand the biological functions of the gut from multiple perspectives. First, generating high-resolution maps of neurons or the gut using ST indicates the development of the gut, the information exchange in the brain-gut axis and pain signaling. Second, with the assistance of ST, the physiopathological manifestations and healing of colonic tissues can be observed dynamically, and accurate judgments can be made about the development of intestinal diseases. Finally, spatially specifying changes in the gut microbiota helps understand the geographic features of the gut that have been altered by pathogens.

Development and evolution of ST technology

Since the 1990s, with the rapid development of high-throughput sequencing technology, transcriptomics has gradually emerged as a discipline to study all transcribed RNAs in cells. Researchers are starting to fully uncover gene expression patterns and regulatory networks. However, conventional transcriptomics techniques are unable to determine the effects of different subcellular structures and environments on gene expression. To more fully understand the regulatory processes of gene expression, ST is rapidly emerging to reveal the spatial distribution, subcellular location and interactions of genes within the cell, providing novel perspectives for understanding the spatial regulatory mechanisms of gene expression (39).

The development of transcriptome technology has gone through three important stages: The first stage is transcriptome sequencing (bulk RNA) of a large number of mixed cells, which reveals the average expression level of a single gene in a large cell population, but fails to demonstrate the transcriptional expression level in a single cell (40). The second stage is scRNA-seq, which constructs the expression profile of each cell at the single-cell level, reflecting cell-to-cell heterogeneity (41). However, single-cell spatial location information is lost during the preparation of the suspension, resulting in changes in gene expression profiles of some single cells during enzymatic digestion or after leaving a specific microenvironment; this is one of the key features of organ function. Driven by this need, the Joakim Lundeberg research group proposed the concept of ST in 2016 and reported the first ST technique based on in situ capture of RNA (42). This provides important research tools in a number of fields, including tissue-cell function, microenvironment interaction, lineage tracing of developmental processes and disease pathology research. Subsequently, a series of techniques capable of high-throughput in situ RNA detection analysis have all been grouped into the category of ST techniques. Although the principles of these techniques vary, they have a common element with regard to recording information about the spatial position of the detected RNA molecules (43).

In the late 1990s, certain studies on ST started the development of ST using technologies such as laser capture microdissection (LCM), microarrays or RNA sequencing (RNA-seq) and single-molecule FISH (smFISH). Some techniques from the 1980s, although not called ‘spatial transcriptomics’, also acquired transcriptional information in space. Moses and Pachter (42) refer to this part of the development of ST technology as the ‘prequel era’ in their online ebook ‘Museum of Spacial Transcriptomics’.

The prequel era technology depicts the general technical characteristics of the spatial transcriptome: Imaging, localization and expression level. At the same time, one of the spatial transcriptomes that can see the ‘prequel era’ has been working on imaging and single cells (resolution), and the expression amount is often not obtained by high throughput. Although spatial transcriptomics at that time was limited by resolution, ease of operation, available software, rich databases and other factors, particularly the high-throughput expression data based on NGS not being available in the pre-transcriptomics era, causing numerous technologies of that era to decline. However, still, in the pre-transcriptomics era, numerous beneficial attempts were made, and it can even be said that it provided a reference for the spatial transcriptomics technology of the present era (39).

By contrast, Moses and Pachter (42) refer to the technology after the ‘prequel era’ as the ‘current era’ technology. The foundations of a number of the ‘current era’ technologies were established in the 1970s to the early 2000s. For example, microarray technology, first reported in 1995 (44), was initially used to quantify transcripts hybridizing to cDNA printed on slides, but was used to quantify the transcriptome of LCM samples in 1999 (45). Currently, popular technologies, including ST and 10× Genomics, rely on this microarray technology to capture transcripts from tissues mounted on microarray slides (4648).

ST has undergone rapid development, with marked improvements in resolution and flux. The spatial transcriptome was declared to be the annual technical method by Nature Methods in 2020 and one of the seven noteworthy annual technologies by Nature magazine in 2022. In 2023, space omics became one of the 10 technologies selected by the World Economic Forum to have the highest potential and to have a positive impact on the world.

Advantages and disadvantages of the different ST techniques

Currently, the commonly used spatial transcriptome technologies are slide-DNA-seq, a method for capturing spatially resolved DNA sequences from intact tissue sections (49), ST, sequential FISH, multiple resistance error correction FISH (MERFISH), LCM-seq, geographical position sequencing (Geo-seq) and Topographic Single Cell Sequencing (Tomo-seq), which can be generally divided into four different types of strategies, each with their own advantages and disadvantages.

Computational strategies based on spatial reconstruction

Seurat Technology (50,51) is a computational spatial reconstruction strategy for cellular location inference by combining scRNA-seq and FISH technology. Specifically, the gene expression of individual cells is analyzed by Switching mechanism at 5′ end of the RNA transcript (Smart-seq) (52), then the cells are clustered into different types and signature genes are identified for each cell type. Subsequently, FISH is used to determine the spatial distribution of signature genes, and finally, the localization of cells is integrated with sequencing information and the spatial distribution information of signature cells. Smart-seq is a single-cell transcriptome sequencing technology for mRNA, which was developed by American and Swedish scientists, and can be used to perform large-scale sequencing of gene subtypes and alleles, among others, of single-cell RNA (52). The Smart-seq, which can generate full-length cDNA from single cells, is a landmark for in-depth studies of allele-specific expression, single nucleotide variant detection or splice variants. However, due to the technical limitations at the time of its creation, Smart-seq technology also has certain limitations, such as poor amplification efficiency for some low-abundance transcripts and the fact that the design of the primers may also affect the subsequent purification and require more cells for preliminary RNA extraction (53,54). This strategy reconstructs spatial information of tissues by combining scRNA-seq, in situ hybridization and prior knowledge, and presents a spatial trend or an overall layout of cells in a particular tissue, but does not fully match the actual cell coordinates (51).

Strategies based on laser microcutting

LCM (55) can cut individual cells or regions of interest in tissue sections at high resolution, and may be combined with transcriptome analysis by RNA-seq analysis, which can provide single-cell whole gene analysis and accurate location information. This technique for analyzing samples is not limited to use in mammals and can also be applied in plants (56,57). However, the spatial analysis technique based on LCM has certain limitations, including a large workload, high cost and the analysis of only a few cells (58).

LCM-seq is a specific technique involving cutting of frozen tissue or paraffin-embedded tissue, or single-cell cutting, combined with subsequent chip or NGS to obtain the transcriptomic data of single cells (59). In 2017, Chinese scientists established Geo-seq combined with LCM and scRNA-seq technology, which overcomes the problem of insufficient amounts of mRNA from a small number of cells and can achieve cell-level accuracy; this can be used not only for 3D reconstruction of a transcription map, but also to study the transcriptomic information of a small number of tissues or cells with special structures (60). In 2018, American scientists developed the TSCS technology. They used lasers to capture single cells, amplified the whole genome of the single cells, and attached independent label sequences. This increased the experimental throughput. After mixed sequencing, they matched the cell positions to tissues using labels and computational methods (61).

Tomo-seq technology was inspired by clinical multi-dimensional imaging technology. A research team sequenced all zebrafish embryos in three different directions and reconstructed different sections of the same axis to form complete 3D embryos by computational methods. However, this method has high requirements for computational methods, and necessitates three identical samples, which is not applicable in clinical practice. The accuracy also needs to be improved (62).

Strategies based on fluorescence imaging

With in situ transcriptomics based on fluorescence imaging, cell detection flux is no longer a problem. Early on, in 2015, Chen et al (43) proposed the MERFISH technique. MERFISH technology has been improved in both cell flux and mRNA species, and may be used for >1 million cells (63) and 10,000 mRNAs. This technique largely increases detectable regions, but also has novel or small sequence variants that cannot be detected, with higher error rates. The method also has limitations such as the requirement for preselected genes (64).

SmFISH uses a single probe sequence to bind the same transcript, using the fluorescent color molecules on the probe to display the expression level of the target molecule in the original image. Although smFISH has high sensitivity and subcellular spatial resolution, this method is susceptible to background influence and low flux due to its weak light signal (65). In 2014, researchers improved the invention of seqFISH, which uses multiple rounds of hybridization to assign unique barcodes to individual transcripts within a single cell. By performing consecutive rounds of hybridization, imaging and probe stripping, a distinct barcode is generated for each mRNA molecule, allowing for spatial transcriptomic mapping through color recognition (66). However, increasing the number of genes requires increasing the number of hybridization rounds, which is expensive and time-consuming and requires super-resolution microscopy (67).

Strategies based on the spatial barcode of the oligonucleotides

The spatial barcode-based strategies can also enable high-throughput detection of cells and are not limited to known sequences. These strategies mainly consist of ST technology (34) and Slide-seq technology (68).

In 2016, Spatial transcriptomics (ST) printed 1,007 pre-defined points on a 6.2×6.6 mm array, each with a spatial barcode, including paste part, sequencing fragments, position tags, random unique molecular identifier and mRNA capture poly (T), and each barcode was captured with mRNA molecules for library construction and sequencing to obtain a transcriptome spatial map (34). ST Technologies pioneered a newera of combining in situ RNA amplification with spatial barcode arrays. In 2018, 10× Genomics(https://www.10×genomics.com/) acquired ST Technologies, and in 2019, they released 10× Visium, reducing the distance from point to point from 200 to 100 microns for higher precision. ST indicates genome-wide spatial expression at the micrometer scale, but does not achieve single-cell resolution.

In 2019, Professors Alice Y. Ting and Howard Y. Chang from Stanford University introduced a new RNA localization detection technique called APEX-seq (69). The use of peroxidase APEX2 can produce covalent biotinylation of RNA molecules and has different sensitivities in different organelle regions, which allows the realization of transcriptome studies on different organelles. However, as cells are required to express APEX2, which is an engineered soybean ascorbate peroxidase, it cannot be used in clinical practice.

In 2020, NanoString Technologies released the GeoMx Digital Spatial Profiler, which is conjugated to DNA Oligo on antibodies or RNA (70). Each DNA Oligo corresponds to one target, with multiple reactions achieved through multiple targets. When the antibody or RNA is bound to the target on the tissue, ultraviolet light is used to cut off the linker between DNA Oligo and the antibody or RNA, thus releasing DNA Oligo for the next quantification of the transcriptome and proteome.

High-definition ST (HDST) and Slide-seq can implement spatial barcodes on microbeads (68,71). Slide-seq links spatial barcodes on a batch of 10-µm microbeads, and mRNA molecules in tissue sections are captured by microbead binding, which are then processed using ‘sequencing by oligo ligation detection’ sequencing to map tissue space by spatial barcode. HDST is a microwell arranged with 2 micron, each containing a microbead, which links a spatial barcode on the bead, bound to the mRNA in the tissue, and then allows Illumina, Inc. Sequencing.

In 2021, space omics technology was a research focus. The group of Professor Rong Fan from Yale University invented DBiT-seq spatial transcriptomics, which is a method of adding barcodes in a different way (72). Tissues are first attached to slides, with a minimum inter-hole distance of 10 micrometers achieved using microfluidic technology. Two sets of liquid containing barcodes are then flowed through the tissue sequentially from two axes, binding to mRNA and protein molecules in the tissue to form a 2D spatial barcode. Reverse transcription is performed inside the cells, and cDNA is collected for subsequent sequencing (73). Although the method can achieve simultaneous capture of the transcriptome and proteome, there may be barcode contamination, and the pore spacing can only be 10 µm, limiting the accuracy of the technique (74).

Chinese scientists established Stereo-seq space omics technology on a BGI sequencing platform. The technique is based on the DNA Nanoballs (DNB) sequencing technology. It works by depositing DNB containing random barcode sequences on a lithoetched modified chip. The nanosphere spacing can be 500 or 715 nm, using a rolling ring to amplify the barcode pool, and obtain the coordinate code of the DNB after the first round of sequencing. Then, by hybridization, connecting the molecular coding and polyT sequence, the tissue was loaded onto the chip and the barcode was captured with mRNA for a second round of sequencing to finally obtain the transcriptome map (75).

American scientists have released the Seq-Scope spatial group on the Illumina platform. This method involves two rounds of sequencing. In the first round, sequencing is done on the Illumina sequencing platform to amplify clusters on a chip with fixed labels. The distance between clusters can be achieved at 0.6–0.8 micrometers. After amplification, a restriction endonuclease is used to expose the Oligo-dT sequence. Following the capture of mRNA molecules in the tissue, a second round of sequencing is conducted to obtain the transcriptome map (76).

The evolution of the development of spatial transcriptomics is summarized in Fig. 2. In 1996, Emmert-Buck et al (55) proposed the LCM technique. In 1998, Femino et al (65) combined digital imaging microscopy and FISH technology. In 2002, Levsky et al (77) used a combined marker approach in order to increase the number of identifiable transcripts. In 2012, Lubeck and Cai (78) reported on the use of super-resolution microscopy and combinatorial markers combined. Subsequently, Ke et al (79) proposed the ISS method. In 2015, Lee et al (80) proposed and developed the fluorescence ISS technology. In 2014, Lubeck et al (67) developed the sequential hybridization technology. Subsequently, Chen et al (43) developed a method called MERFISH. In 2018, Wang et al (38) developed the STARmap method. Subsequently, Vickovic et al (70) proposed the HDST sequencing method and at the same time, Rodriques et al (68) invented a similar Slide-Seq method. In 2019, 10 Genomics introduced the Visium spatial transcriptome technology.

Figure 2.

Evolution of the development of spatial transcriptomics. In 1996, Emmert-Buck et al (55) proposed the LCM technique. In 1998, Femino et al (65) combined digital imaging microscopy and FISH technology. In 2002, Levsky et al (77) used a combined marker approach in order to increase the number of identifiable transcripts. In 2012, Lubeck and Cai (78) combined super-resolution microscopy and combinatorial markers. In 2013, Ke et al (79) proposed an ISS method. In 2014, Lubeck et al (67) developed the sequential hybridization technology. In 2015, Lee et al (80) proposed and developed fluorescence ISS technology. In 2015, Chen et al (43) developed a method known as MERFISH. In 2018, Wang et al (38) developed the STAR map method. In 2019, Vickovic et al (70) proposed the HDST sequencing method, while contemporaneously, Rodriques et al (68) invented a similar Slide-Seq method. In 2019, 10× Genomics introduced the Visium spatial transcriptome technology. APEX-seq, ascorbate peroxidase sequencing; BaristaSeq, barcode in situ targeting sequencing; BOLORAMIS, barcoded oligonucleotides ligated on RNA amplified for multiplexed and parallel in situ analyses; DBiT-seq, deterministic Barcoding in Tissue for spatial omics sequencing; DSP, digital Signal Processing; FISH, fluorescence in situ hybridization; FISSEQ, fluorescent in situ sequencing; Geo-seq, geographical position sequencing; HDST, high-definition ST; ISS, in situ sequencing; LCM, laser capture microdissection; MERFISH, multiple resistance error correction FISH; NGS, next-generation sequencing; NICHE-seq, combines photoactivatable fluorescent reporters, two-photon microscopy and single-cell RNA sequencing (scRNA-seq) to infer the cellular and molecular composition of niches.; osmFISH, ouroboros smFISH; ProximID, cell structures that interact with each other are manually dissociated into single cells and analyzed individually; RCA, rolling circle amplification; RNAscope, in situ hybridization technique for detecting target RNA within intact cells; ROI, region of interest; S1000, BMKMANU S1000; sci-Space, single-cell gene expression differences with spatial context; Seq-SCOPE, sub-micron resolution sequencing; seqFISH, sequential fluorescence in situ hybridization; seqFISH+, upgraded seqFISH; Slide-seq, slide sequencing; Slide-seq V2; smFISH, single-molecule FISH; ST, spatial transcriptomics; STAR map, spatially resolved transcript amplicon readout mapping; Stereo-seq, spatial enhanced resolution omics-sequencing; TIVA, transcriptome in vivo analysis; Tomo-seq, topographic single cell sequencing.

A spatial view of human intestinal development

The gut is the site of immune cell initiation at birth (81) and is closely associated with individual developmental abnormalities and illnesses. The gut is subsequently symbiotic with the gut microbiota, and these work together to supply nutrients to the body and maintain immune function (82,83). Marked advancements have been made in scRNA-seq, which have helped enhance the knowledge of the various types of cells in the gut and their biological characteristics (84). However, to the best of our knowledge, the molecular mechanisms underlying the formation of the unique intestinal morphology through the interplay of multiple cell types remain unclear. During the formation of the proto-gut embryo, three primary germ layers, the ectoderm, mesoderm and endoderm, are generated (8588). Once the primary germ layers of cells are formed, the endoderm undergoes intricate changes and extensive folding to create the embryonic gut tube (89). The intestinal loop is shaped around Carnegie stage (CS) 14 and then moves into the space outside the body of the embryo at CS 16, returning to the peritoneal cavity at post-conceptual week (PCW) 11. Between PCWs 8 and 12, the pseudostratified columnar epithelial cells give rise to chorionic villi and crypt structures, establishing a self-renewal circuit maintained by ESCs (90,91). Whereas various animal models have identified distinct intestinal cells and regional tissues driving the formation of the aforementioned structures, suggesting differential features across species (92,93), the mechanisms driving this process in humans are not fully understood. To gain a more precise understanding of human tissues, Fawkner-Corbett et al (94) used high-throughput scRNA-seq combined with ST technology to map high-resolution organ development by acquiring whole-layer intestinal tissues, which revealed previously unknown disease-related phenotypes in the adult intestinal tract and localized transcriptional signatures spatially to different feature regions. This offers a valuable tool for further exploration of the molecular origins and heritability of intestinal diseases (94).

Numerous research studies have used scRNA-seq techniques to identify the diversity of intestinal epithelial, mesenchymal and immune cell types in the adult intestines (9597). This has led to the exploration of several important questions using this information. Initially, the process of crypt-villus axis formation was elucidated. By scoring the activity of all modules (in the field of transcriptomics, modules can be understood as collections of genes with similar expression patterns), it was found that modules derived from endothelial cells, fibroblasts and pericytes are located deep in the tissue; epithelial-specific modules expressing Hedgehog pathway genes (such as the convolution donor receptor LDL receptor related protein 5) (94), are located in the vicinity of the canaliculus, and genes expressed by modules containing myofibroblasts (such as WNT2B) emerge later and are abundant at PCW 12, indicating that the ISC-myofibroblast signaling circuit is established only after crypt formation. These findings suggest that ST may partially restore the disrupted morphological gradients and increased physical distances during intestinal development (93). Furthermore, it can determine the hierarchical structure and differentiation functions of fibroblasts and myofibroblast subtypes. After classifying fibroblasts (98), intestinal fibroblasts were divided into different functional zones according to the characteristics of different phenotypes along the crypt structure to the villus axis (99). According to such anatomical zoning, as the main determinant of intestinal fibroblast heterogeneity, one of the original studies identified human colonic fibroblasts with numerous fibroblast subcharacteristics, Wnt differential expression and bone morphogenic protein signaling genes, which reflect the specific location along the anatomical axis of crypt villi (100). Antibodies in colon mesenchymal cells of UC patients were further investigated using scRNA-Seq and flow sorting to enrich CD90 cells. This study identified four populations of colonic stromal fibroblasts (S1-S4). After classifying fibroblasts, the analysis of various morphogens with different cells and genes expression revealed that the submucosal structural cells were mainly composed of S1 cells. S2 fibroblasts mainly contribute to maintaining the epithelial crypt ecological niche and promoting epithelial formation (101,102), whereas S3 fibroblasts are more commonly found in large vessels, emphasizing their role in forming the intestinal vascular support ecotone (93). S4 fibroblasts are closely associated with immune follicular cells in the adult colon and exhibit characteristics of follicular reticulocytes, which are crucial for the formation of lymphoid structures and are closely linked to the pathogenesis of UC (98). The development of blood vessels and nerves in the intestines, the formation of Peyer's patches (PP), gut-associated lymphoid tissue (GALT), and the immune system processes have been established (94). PPs are secondary lymphoid organs that interact with the external environment via the intestinal lumen. Spatial transcriptome analysis showed that fetal type 3 lymphocytes (innate lymphoid cells) express IL7RA and inhibitor of DNA binding 2, crucial genes for PP formation in the mouse uterus (103). Additionally, deficiencies in GDNF family receptor α3 also result in developmental disorders of PP. A study confirmed that GALT formation occurs in the small intestine prenatally and in the colon after birth (104), and lymphoid structures can be visualized by mapping factors enriched for B-cell-related genes onto colonic tissue. Enrichment of the NF-κB and TNF-α pathways has been detected in regions of immune and inflammatory activity, closely matching the spatial distribution of lymphoid clusters (105).

Landscape of molecular regionalization of the colon under physiological and pathological conditions

The intestinal epithelium maintains the normal physiology of the intestinal tract through continuous regeneration, and disruption of the regeneration process of intestinal epithelium can lead to pathogen translocation, which in turn mediates various chronic intestinal diseases (106). scRNA-seq technology has broadened the understanding of the cell types, cell subpopulations and cell states present in physiological and pathological states (107). scRNA-seq has also enabled the detection of subpopulations of cells abnormally driven (activation of KRAS in lung cancer is a poor prognostic indicator) by disease conditions, which is of considerable importance in the study of disease mechanisms (108,109). One study revealed that even though mice given dextran sodium sulfate (DSS) showed improvement in symptoms and body weight after a period of DSS discontinuation, the inflammatory changes in the colon did not fully return to normal levels (110). Therefore, it is crucial to analyze the transcriptomic landscape of the mucosal healing process in detail. Parigi et al (111) used ST to reveal previously unrecognized molecular regionalization in the colon of healthy mice. The study revealed a spatially organized transcriptional program for regionalized mucosal healing, demonstrating that factors in the proximal and distal colon have distinct functions and are involved in different regulatory processes, which was determined through dataset integration and associated pathway analyses. At the same time, the integration of longitudinal and ST data to observe the expression status of different modules at different stages can reveal the dynamic timing of colon tissue healing (110,111). Studies have revealed that clusterin accumulated during the repair phase following intestinal injury. Clusterin is seldom present in a healthy state, and further verification is needed to determine its association with infection. Multimodal spatial analysis will help identify important events related to tissue damage repair (111,112).

Macrophages are resident immune cells that exist in two primary states following activation: M1 macrophages and M2 macrophages. The functions and appearances of these macrophages vary depending on the microenvironment and location, and there is ongoing debate with regard to their roles and characteristics (113). What has been demonstrated is a higher expression of the M1 population in IBD samples, and that the phenotype and function of intestinal macrophages are related to their spatial distribution in the gut (114). ST has revealed the transcriptional status of macrophages in healthy and inflammatory states, defined specific markers for M2 macrophages, and clarified cell types and differences in the spatial distribution of different cells during intestinal inflammation (99). For example, ST showed that M1 macrophages and neutrophils were located near the mucosal surface in areas with intestinal ulcers. Spatial analysis has verified the diversity of macrophage populations and highlighted their interaction with inflammatory fibroblasts (115). This interaction enhances colony stimulating factor 2 expression and stimulates macrophage activation, leading to the release of IL-6 and TNF (115), which serve a role in the inflammatory process, and this spatial analysis technique is essential for comprehending the interactions between several specific cells.

Enhanced comprehension of the spatial dynamics of the gut microbiota

The gut microbiota is an integral part of the human body, and although a portion is highly conserved, the dynamic microbiota changes in response to age, and physiological and pathological states, and participates in processes such as neuromodulation, immunity and metabolism (116,117). The human gut consists mainly of Bacteroidota and Bacillota bacterial phyla, and an imbalance between these two phyla is considered to be a sign of microbial dysbiosis, which is closely linked to the pathogenesis of IBD (118). Dysfunction of the microbiota, such as an increase in facultative anaerobes and a decrease in obligate anaerobes, occurs during periods of active intestinal inflammation (119). Currently, the study of gut microbes is dominated by the use of genomics, which provides a comprehensive understanding of microbial categories and certain functions (120,121), but it does not offer spatial information about the microbes. The spatial organization of gut microbes can affect various properties such as colonization, metabolism and stability of the internal environment (122,123). It has been found that the distribution and abundance of different phyla is heterogeneous in different regions of the gut and that the microbial composition differs considerably between species (124).

The stomach, being the most acidic part of the digestive system, has fewer bacterial species. In the transition to the duodenum and ileum, the pH markedly increases and the oxygen pressure gradient decreases, creating a more favorable environment for bacterial growth. Although study results have indicated that the bacterial diversity in this area is either lower or equal to that of the stomach, there is a notable increase in the abundance of specific microbiota (125,126). This low diversity environment protects the intestinal tract to a certain extent, while bacterial overgrowth can potentially contribute to the onset of conditions such as IBS or functional dyspepsia (127). By the time it reaches the cecum and proximal colon, the intestinal lumen becomes intensely anaerobic and transit time is slowed due to the decreased pH, as a result of fermentation of fibers and complex polysaccharides. Dietary fibers provide ample nutrients for the growth of microbes, leading to a marked increase in both the quantity and variety of microbial species (128,129). In the human colon, there are higher concentrations of Bacteroidota, Pseudomonadota and Bacillota, and processes that shape the gut microbiota are thought to be mostly niche-driven (122). The intestinal epithelium has folds, villi and invaginations on its surface, known as crypts, which offer specialized protection to the intestine, but the crypts are heterogeneous in their response to injury. Paneth cells colonize the crypts of the small intestine and secrete antimicrobial peptides to achieve multi-level management of the intestinal flora and to maintain a stable intestinal flora structure. Since the large intestine lacks Paneth cells, it is guarded at the entrance of the colonic crypts by cup cells, which secrete mucin and form a mucus barrier to protect the epithelial cells (130). In addition, it has been previously reported that chemokines with antimicrobial activity against Escherichia coli and Salmonella enterica are found in colonic crypt compartments, which serve an important role in coordinating the normal influx of immune cells and inflammation in the intestine (131). Various microorganisms inhabit crypts, and some also extend their colonization beyond these areas, including into the PP, a specific region where bacteria directly interact with host tissues. This site serves as a sampling location for the microbiota of the immune system. Here, specialized cells access microbial and environmental antigens, passing them to antigen-presenting dendritic cells, participating in the immune process (132).

The degree of bacterial spatial organization in the gut microbiota varies, and the peristaltic contractions of the intestines can promote the fusion of bacteria in the gut. Therefore, even in the colon where the content flow rate is low, there is a high degree of mixing in the gut microbiota (133,134). A sample block sampling technique used to characterize the spatial organization of microbial communities at the 10–30-µm scale found well-mixed sites scattered with micron-scale clusters of specific taxa (126), which may be related to the heterogeneity of food and plant particles within the lumen (131,135). Bifidobacterium pseudolongum has been proved to colonize starch granules and plant granules, and Bacteroides colonizes undigested plant granules and is spatially segregated into multiple small colonies, suggesting that mucosal communities based on different intestinal regions could be useful in determining the role of mucosal geography in driving colonization of epithelial surfaces (136,137). Alterations in the microbiota are strongly associated with various human illnesses, such as IBD and CRC, as well as skin and mental health conditions (138,139). Clarifying the geographical distribution of gut microbiota affected by pathogens will provide novel insights for the understanding of host health and disease (140).

Identifying molecular features of human injury receptors and enhancing the understanding of brain-gut-related hypothalamic functions

IBD and IBS are commonly viewed as gastrointestinal conditions resulting from heightened sensitivity in the colon, along with dysfunction in other visceral and somatic organ (141,142). The interaction of sensory signals between multiple organs, known as ‘cross-organ sensitization’, is caused by the dorsal root ganglia transmitting sensory data for integrated processing (143). Injury receptors are located in the dorsal root ganglion and trigeminal ganglion, where they receive sensory signals related to bodily injury. These receptors are the first neurons in the pain pathway and express a range of receptors, allowing them to respond to various stimuli (144). In cases of cross-organ sensitization linked to intestinal dysfunction, the sensitization of colonic afferent neurons can lead to the cross-activation of reflex pathways in other uninjured organs, with the release of substance P, calcitonin gene-related peptide and excitatory amino acids to the peripheral organs, which activate the injury receptors and lower their thresholds (143). The excitability of these neurons increases in both acute and chronic pain states, and their excited phenotype changes are directly associated with the chronic pain state, so these neurons are key targets for the treatment of pain (144). ST enables researchers to draw high-resolution maps of human sensory neurons in the dorsal root ganglion, which can help identify more effective drug targets, therefore opening up novel avenues for pain management (145).

The mental, nervous, endocrine and immune regulation via the brain-gut axis serves an important role in the onset of certain intestinal diseases (146). The hypothalamus serves a leading role in regulating the basic social behaviors of the body and homeostatic functions, and is involved in the regulation of anxiety, depression, immune inflammation and gut microbiota (147,148). Studies have found that the volume of the hypothalamus in patients with IBD was larger than that of patients in the control group, and that it is also more strongly connected to other brain functional areas (149,150). This excessive connectivity can lead to the activation of abnormal anxiety circuits, mediating emotional disorders (151). However, the molecular characteristics of the development of the human hypothalamus are still unclear, and there is limited knowledge about the structure, spatial organization and function of the hypothalamic nucleus. The imaging-based single-cell transcriptomics method combines gene expression profiles with activity markers to directly image and analyze multiple RNAs in their natural cellular environment, thereby mapping out their spatial structure (152). By uniformly slicing the preoptic area, researchers have created a cellular map of the mouse hypothalamic preoptic area, identifying rare cell types and describing the spatial organization of specific neuronal cell types. This revealed their functions in different behaviors, providing potential insight into how different cell types communicate in physiological and pathological states (63). At the same time, ST can reveal the spatiotemporal transcriptional profile and cell type characteristics of human hypothalamic development. Mapping the spatiotemporal transcriptome of human hypothalamic development has demonstrated the asynchrony of spatial development of different neurons and neuroglial cells, identified key regulatory genes that neural progenitor cells and neural epithelial cells, and provided a deeper understanding of cellular network organization, circuit formation and the mechanisms of hypothalamic dysfunction (63,153). Analyzing the spatial transcription of key areas of the brain and gaining a comprehensive understanding of the connectivity patterns of the nervous system of the brain can provide valuable insights for studying the brain-gut axis, potentially offering novel therapeutic targets for the treatment of brain-gut-related diseases (153,154).

Applications in the study of CRC

During the progression of tumors, the interactions between cancer cells and other cell populations promote the heterogeneity of tumors. Experimental findings have demonstrated that the tumor microenvironment (TME) serves an important role in the development of cancer, and its spatial distribution and specific host-microbe cell interactions to some extent influence the progression of tumors (155,156). Single-cell sequencing of the genome, transcriptome and epigenome has contributed to the understanding of the internal structure of tumors, and more advanced ST is expected to decipher the complex principles and mechanisms of gene activity in three dimensions, which will have profound implications for life science research (157).

CRC is the third most common malignant tumor globally, and due to its high mortality rate due to metastasis, nearly 900,000 individuals die from CRC each year (158). Since CRC only presents with noticeable symptoms in the late stages, it is particularly important to identify relevant oncogenes and improve the early detection rate and early treatment of CRC (159,160). The combined application of multi-omics is important in studying the biological characteristics of the early local spread and distant metastasis of CRC (161). Cancer-associated fibroblasts (CAFs) are a major component of the stromal cells in numerous malignant tumors (109,162). In order to determine the interaction between the TME and CAFs in the pathogenesis of CRC, researchers have combined ST and bulk RNA-seq. Two types of CAFs were identified: Myo-CAFs and inflammatory CAFs. The latter not only promoted the progression and metastasis of tumors, but were also associated with the poor prognosis of the tumor (163). Certain studies have also found that spatial analysis can reveal the molecular and immunological characteristics of early-stage CRC, identify biomarkers associated with disease progression and prognosis, and reveal genes related to CRC invasion, providing a deeper understanding of the biological processes associated with tumor malignancy (164,165). The ST and GeoMx digital spatial profiling technologies of 10× Visium can identify the identity and location of the internal microbial community within tumors. The technologies can also detect areas with low vascularization, high levels of immune suppression and association with malignant tumors. Research has shown that the degree of vascularization in bacteria-colonized regions is lower than in bacteria-negative regions, with decreased expression of smooth muscle actin, decreased proliferation levels and downregulation of Ki-67 and p90 RSK. The bacteria-colonized microniches significantly increases the phosphorylation levels of JNK, ERK1, ERK2 and P38 in CRC tumors, revealing signal pathways that are activated in response to bacteria. The results indicated that the infected areas of CRC tumor tissue have lower proliferative potential compared to uninfected areas (166168).

The liver metastasis of CRC poses a significant challenge for clinicians, with 80% of CRC cases experiencing tumor metastasis to the liver before the primary tumor can be detected clinically, which is closely associated with low survival rates (169,170). There is controversy regarding whether or not aging cancer cells serve a positive role in the progression and metastasis of tumors. Researchers have used ST and multi-database integration to comprehensively map the whole transcriptome cell atlas of CRC and related liver metastasis. They analyzed the molecular specificity within the TME and the transcriptional heterogeneity of senescent cancer cells, which could identify the aging-dependent cancer ecosystem in CRC liver metastasis and potentially provide therapeutic targets for CRC-related cancer types (171,172).

Applications in intestinal diseases

As one of the eight regions of the digestive system, the gastrointestinal tract serves an important role in absorption, digestion, excretion, endocrine, nutritional metabolism, immune balance and other functions. Due to the diversity of intestinal cells, dissecting the characteristics, functions and internal operation of the cells in the intestinal mucosa has been a technical challenge (173,174). The advent of ST provides a higher-resolution research tool for developmental and cancer-related studies of the gut, adding new dimensions to the understanding of multiple gastrointestinal diseases (94). Table I shows that different ST technologies are applied to different intestinal diseases, and the corresponding conclusions are drawn from different materials, indicating that they are widely used in intestinal diseases (111,175181).

Table I.

Applications of spatial transcriptomics in intestinal diseases.

Table I.

Applications of spatial transcriptomics in intestinal diseases.

First author, yearTechnologyDisease or tissueConclusion(Refs.)
Pelka et al, 2021Slide-based sequencing and scRNA-seqCRCRevealed spatially organized multicellular immune centers in human CRC.(175)
Parigi et al, 2022LCM-seq and scRNA-seqIBDVisualized the gene expression landscape of the gut throughout injury and post-injury recovery, and described the spatial expression profile of IBD risk genes in the colon.(111)
Frede et al, 2022LCM-seq and scRNA-seqUCThe expansion of B cells hindered the healing of IBD. Assessment of the influence of B-cell subsets on epithelial-stromal cell interactions.(176)
Zhang et al, 2022 Immuno-LCM-RNA-seqSmall intestine lacteal cellsShowed partitioning of mouse small intestine lacteal cells.(177)
Ghaddar and De, 2022Neighbor-seqSmall intestinal epithelium, terminal respiratory tract and splenic white pulpAnalysis of cellular duplexes/multiplexes captured during standard scRNA-seq to infer cell-to-cell interactions. Proof of concept data in the small intestine.(178)
Zhu et al, 2023Slide-based sequencing and scRNA-seqGut microbiotaIntestinal-derived Akkermansia muciniphila may contribute to suppressing tumorigenesis by affecting the tumor commensal microbiome and reprogramming tumor metabolism.(179)
Sun et al, 2023LCM-seq and scRNA-seqGastric cancerIdentification of a ‘tumor-normal tissue interface’ region dominated by immune cells.(180)
Tun et al, 2024Seq-scopeIBDEnterorome dysregulation driven by Caudovirales expansion affected intestinal immunity and dysfunction.(181)

[i] LCM-seq, laser capture microdissection sequencing; scRNA-seq, single-cell RNA sequencing; Immuno-LCM-RNA-seq, laser capture microdissection sequencing combining with immunohistochemistry; Neighbor-seq, a method to identify and annotate the architecture of direct cell-cell interactions and relevant ligand-receptor signaling from the undissociated cell fractions in massively parallel single cell sequencing data; Seq-scope, a spatial barcoding technology with a resolution comparable to an optical microscope; CRC, colorectal cancer; UC, ulcerative colitis; IBD, inflammatory bowel disease.

Conclusions and prospects

The present review discusses the applications of ST technologies related to the gut. The rapid development of ST and its combined application with multi-omics has provided powerful technical support for studying the mechanisms involved in gut development and physiological pathology.

Due to the complex structure of the intestines, accurately mapping their spatial organization and molecular characteristics is crucial for understanding the progression of diseases. Various sequencing technologies have been continuously evolving, from scRNA-Seq to ST based on scRNA-Seq, enabling the visualization of the spatial positioning of tissues and cells. Given that low-resolution ST cannot meet higher imaging requirements, researchers have developed high-definition spatial genomics. HDST has been combined with powerful imaging modularization techniques through the modification of microbead arrays and the development of polynomial Bayesian classifiers, resulting in a marked increase in the resolution of the technique to 1,400 times higher than that of normal ST. Together with the highly specific nature of HDST signals, which can be interpreted through computational integration with morphological features and single-cell profiles, this has opened up novel avenues for high-resolution spatial analyses of cells and tissues (70,182). With the continuous development of sequencing technology, phenotypic information at the protein level on the cell surface has been gradually and well presented by novel technologies, and the use of DNA-barcoded antibodies allows for simultaneous analysis of the single-cell transcriptome and proteome, whereas NGS-based ST allows for the analysis of spatial multi-omics data. The single-cell multi-omics approach permits a more comprehensive understanding of intestinal biology and disease mechanisms, which facilitates the prediction of drug targets and clinical responses (183,184). More in-depth studies are already underway, including the use of ST to study epigenomic, proteomic or chromosomal structures (185), which can define neuroscience- and cancer-related cell types, provide spatial analyses that can present epigenetic traits at the single-cell level, and provide insight into the spatial state of protein molecules in tissues (186188).

Numerous discoveries in the life sciences are closely related to the biological function of tissue and cell interactions, where spatial relationships between cells determine cell fate and diseases emerge as abnormalities in spatial structures within tissues (189). ST reconstructs spatial organization based on single-cell sequencing and combines it with other methods, such as different sequencing techniques, computer programming and visualization technology to present biological processes more intuitively. These technologies have facilitated new discoveries in various fields ranging from intestinal development to various diseases of the intestines.

Efficient research methods serving research should also serve the clinic, and the clinical application of ST is one of the important issues in translational medicine (190). Complete and efficient sampling and testing protocols, as well as the analysis and interpretation of data, are key to applying ST in clinical settings. A number of transcriptome-based commercial platforms are progressively being developed for applications that utilize artificial intelligence (AI) to provide open datasets and accurate data processing methods (echnologies such as SOMDE, SEDR and STAGATE), leveraging the potentials of AI, which will contribute to the clinical application of ST (68,191193).

Acknowledgements

Not applicable.

Funding

The present study was supported by Chongqing Natural Science Foundation (grant no. cstc2021jcyj-msxmX0858).

Availability of data and materials

Not applicable.

Authors' contributions

YH and YG discussed the premise of the study and conceived the format. YG drafted the initial manuscript based on this discussion. CR completed the revisions of the manuscript. YH and YW generated the figures. YW revised the content of the manuscript. XY proposed the main idea of the article and coordinated the work to ensure the accuracy or completeness of the manuscript and address any issues that arise during the writing process. All authors have read and approved the final version of the manuscript. Data authentication is not applicable.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Caruso R, Lo BC and Núñez G: Host-microbiota interactions in inflammatory bowel disease. Nat Rev Immunol. 20:411–426. 2020. View Article : Google Scholar : PubMed/NCBI

2 

Round JL and Mazmanian SK: The gut microbiota shapes intestinal immune responses during health and disease. Nat Rev Immunol. 9:313–323. 2009. View Article : Google Scholar : PubMed/NCBI

3 

Sardinha-Silva A, Alves-Ferreira EVC and Grigg ME: Intestinal immune responses to commensal and pathogenic protozoa. Front Immunol. 13:9637232022. View Article : Google Scholar : PubMed/NCBI

4 

Longstreth GF, Thompson WG, Chey WD, Houghton LA, Mearin F and Spiller RC: Functional bowel disorders. Gastroenterology. 130:1480–1491. 2006. View Article : Google Scholar : PubMed/NCBI

5 

Mayer EA, Ryu HJ and Bhatt RR: The neurobiology of irritable bowel syndrome. Mol Psychiatry. 28:1451–1465. 2023. View Article : Google Scholar : PubMed/NCBI

6 

Flynn S and Eisenstein S: Inflammatory bowel disease presentation and diagnosis. Surg Clin North Am. 99:1051–1062. 2019. View Article : Google Scholar : PubMed/NCBI

7 

Bonetto S, Fagoonee S, Battaglia E, Grassini M, Saracco GM and Pellicano R: Recent advances in the treatment of irritable bowel syndrome. Pol Arch Intern Med. 131:709–715. 2021. View Article : Google Scholar : PubMed/NCBI

8 

Saha L: Irritable bowel syndrome: Pathogenesis, diagnosis, treatment, and evidence-based medicine. World J Gastroenterol. 20:6759–6773. 2014. View Article : Google Scholar : PubMed/NCBI

9 

Singh N and Bernstein CN: Environmental risk factors for inflammatory bowel disease. United European Gastroenterol J. 10:1047–1053. 2022. View Article : Google Scholar : PubMed/NCBI

10 

Rosen MJ, Dhawan A and Saeed SA: Inflammatory bowel disease in children and adolescents. JAMA Pediatr. 169:1053–1060. 2015. View Article : Google Scholar : PubMed/NCBI

11 

Zhang YZ and Li YY: Inflammatory bowel disease: Pathogenesis. World J Gastroenterol. 20:91–99. 2014. View Article : Google Scholar : PubMed/NCBI

12 

Neurath MF: IL-23 in inflammatory bowel diseases and colon cancer. Cytokine Growth Factor Rev. 45:1–8. 2019. View Article : Google Scholar : PubMed/NCBI

13 

Nadeem MS, Kumar V, Al-Abbasi FA, Kamal MA and Anwar F: Risk of colorectal cancer in inflammatory bowel diseases. Semin Cancer Biol. 64:51–60. 2020. View Article : Google Scholar : PubMed/NCBI

14 

Brackmann S, Andersen SN, Aamodt G, Langmark F, Clausen OPF, Aadland E, Fausa O, Rydning A and Vatn MH: Relationship between clinical parameters and the colitis-colorectal cancer interval in a cohort of patients with colorectal cancer in inflammatory bowel disease. Scand J Gastroenterol. 44:46–55. 2009. View Article : Google Scholar : PubMed/NCBI

15 

Borowitz SM: The epidemiology of inflammatory bowel disease: Clues to pathogenesis? Front Pediatr. 10:11037132023. View Article : Google Scholar : PubMed/NCBI

16 

Kaplan GG: The global burden of IBD: From 2015 to 2025. Nat Rev Gastroenterol Hepatol. 12:720–727. 2015. View Article : Google Scholar : PubMed/NCBI

17 

Zheng HB, de la Morena MT and Suskind DL: The growing need to understand very early onset inflammatory bowel disease. Front Immunol. 12:6751862021. View Article : Google Scholar : PubMed/NCBI

18 

Taylor S and Lobo AJ: Diagnosis and treatment of inflammatory bowel disease. Practitioner. 260:19–23. 2016.PubMed/NCBI

19 

Chachu KA and Osterman MT: How to diagnose and treat IBD mimics in the refractory IBD patient who does not have IBD. Inflamm Bowel Dis. 22:1262–1274. 2016. View Article : Google Scholar : PubMed/NCBI

20 

Parkin J and Cohen B: An overview of the immune system. Lancet. 357:1777–1789. 2001. View Article : Google Scholar : PubMed/NCBI

21 

Kayama H, Okumura R and Takeda K: Interaction between the microbiota, epithelia, and immune cells in the intestine. Annu Rev Immunol. 38:23–48. 2020. View Article : Google Scholar : PubMed/NCBI

22 

Groschwitz KR and Hogan SP: Intestinal barrier function: Molecular regulation and disease pathogenesis. J Allergy Clin Immunol. 124:3–22. 2009. View Article : Google Scholar : PubMed/NCBI

23 

Wu S, Yang L, Fu Y, Liao Z, Cai D and Liu Z: Intestinal barrier function and neurodegenerative disease. CNS Neurol Disord Drug Targets. Nov 24–2023.(Epub ahead of print).

24 

Wu Y, Tang L, Wang B, Sun Q, Zhao P and Li W: The role of autophagy in maintaining intestinal mucosal barrier. J Cell Physiol. 234:19406–19419. 2019. View Article : Google Scholar : PubMed/NCBI

25 

Ding JH, Jin Z, Yang XX, Lou J, Shan WX, Hu YX, Du Q, Liao QS, Xie R and Xu JY: Role of gut microbiota via the gut-liver-brain axis in digestive diseases. World J Gastroenterol. 26:6141–6162. 2020. View Article : Google Scholar : PubMed/NCBI

26 

Rutsch A, Kantsjö JB and Ronchi F: The gut-brain axis: How microbiota and host inflammasome influence brain physiology and pathology. Front Immunol. 11:6041792020. View Article : Google Scholar : PubMed/NCBI

27 

Integrative HMP (iHMP) Research Network Consortium, . The integrative human microbiome project. Nature. 569:641–648. 2019. View Article : Google Scholar : PubMed/NCBI

28 

Gueimonde M and Collado MC: Metagenomics and probiotics. Clin Microbiol Infect. 18 (Suppl 4):S32–S34. 2012. View Article : Google Scholar

29 

Adak A and Khan MR: An insight into gut microbiota and its functionalities. Cell Mol Life Sci. 76:473–493. 2019. View Article : Google Scholar : PubMed/NCBI

30 

Patterson E, Ryan PM, Cryan JF, Dinan TG, Ross RP, Fitzgerald GF and Stanton C: Gut microbiota, obesity and diabetes. Postgrad Med J. 92:286–300. 2016. View Article : Google Scholar : PubMed/NCBI

31 

Liu X, Chen Y, Zhang S and Dong L: Gut microbiota-mediated immunomodulation in tumor. J Exp Clin Cancer Res. 40:2212021. View Article : Google Scholar : PubMed/NCBI

32 

Qin J, Li R, Raes J, Arumugam M, Burgdorf KS, Manichanh C, Nielsen T, Pons N, Levenez F, Yamada T, et al: A human gut microbial gene catalogue established by metagenomic sequencing. Nature. 464:59–65. 2010. View Article : Google Scholar : PubMed/NCBI

33 

Marx V: Method of the year: Spatially resolved transcriptomics. Nat Methods. 18:9–14. 2021. View Article : Google Scholar : PubMed/NCBI

34 

Ståhl PL, Salmén F, Vickovic S, Lundmark A, Navarro JF, Magnusson J, Giacomello S, Asp M, Westholm JO, Huss M, et al: Visualization and analysis of gene expression in tissue sections by spatial transcriptomics. Science. 353:78–82. 2016. View Article : Google Scholar : PubMed/NCBI

35 

Wang Y, Liu B, Zhao G, Lee Y, Buzdin A, Mu X, Zhao J, Chen H and Li X: Spatial transcriptomics: Technologies, applications and experimental considerations. Genomics. 115:1106712023. View Article : Google Scholar : PubMed/NCBI

36 

Shah S, Lubeck E, Zhou W and Cai L: In situ transcription profiling of single cells reveals spatial organization of cells in the mouse hippocampus. Neuron. 92:342–357. 2016. View Article : Google Scholar : PubMed/NCBI

37 

Lee JH, Daugharthy ER, Scheiman J, Kalhor R, Yang JL, Ferrante TC, Terry R, Jeanty SS, Li C, Amamoto R, et al: Highly multiplexed subcellular RNA sequencing in situ. Science. 343:1360–1363. 2014. View Article : Google Scholar : PubMed/NCBI

38 

Wang X, Allen WE, Wright MA, Sylwestrak EL, Samusik N, Vesuna S, Evans K, Liu C, Ramakrishnan C, Liu J, et al: Three-dimensional intact-tissue sequencing of single-cell transcriptional states. Science. 361:eaat56912018. View Article : Google Scholar : PubMed/NCBI

39 

Asp M, Bergenstråhle J and Lundeberg J: Spatially Resolved transcriptomes-Next generation tools for tissue exploration. Bioessays. 42:e19002212020. View Article : Google Scholar : PubMed/NCBI

40 

Shi Y, Wu X, Zhou J, Cui W, Wang J, Hu Q, Zhang S, Han L, Zhou M, Luo J, et al: Single-nucleus RNA sequencing reveals that decorin expression in the amygdala regulates perineuronal nets expression and fear conditioning response after traumatic brain injury. Adv Sci (Weinh). 9:e21041122022. View Article : Google Scholar : PubMed/NCBI

41 

Li J, Wu C, Hu H, Qin G, Wu X, Bai F, Zhang J, Cai Y, Huang Y, Wang C, et al: Remodeling of the immune and stromal cell compartment by PD-1 blockade in mismatch repair-deficient colorectal cancer. Cancer Cell. 41:1152–1169.e7. 2023. View Article : Google Scholar : PubMed/NCBI

42 

Moses L and Pachter L: Museum of spatial transcriptomics. Nat Methods. 19:534–546. 2022. View Article : Google Scholar : PubMed/NCBI

43 

Chen KH, Boettiger AN, Moffitt JR, Wang S and Zhuang X: RNA imaging. Spatially resolved, highly multiplexed RNA profiling in single cells. Science. 348:aaa60902015. View Article : Google Scholar : PubMed/NCBI

44 

Schena M, Shalon D, Davis RW and Brown PO: Quantitative monitoring of gene expression patterns with a complementary DNA microarray. Science. 270:467–470. 1995. View Article : Google Scholar : PubMed/NCBI

45 

Luo L, Salunga RC, Guo H, Bittner A, Joy KC, Galindo JE, Xiao H, Rogers KE, Wan JS, Jackson MR and Erlander MG: Gene expression profiles of laser-captured adjacent neuronal subtypes. Nat Med. 5:117–122. 1999. View Article : Google Scholar : PubMed/NCBI

46 

Bhatia HS, Brunner AD, Öztürk F, Kapoor S, Rong Z, Mai H, Thielert M, Ali M, Al-Maskari R, Paetzold JC, et al: Spatial proteomics in three-dimensional intact specimens. Cell. 185:5040–5058.e19. 2022. View Article : Google Scholar : PubMed/NCBI

47 

Alfieri CM, Mattinzoli D, Ikehata M, Cresseri D, Moroni G, Vaira V, Ferri G, Ferrero S and Messa P: Laser capture microdissection on formalin-fixed and paraffin-embedded renal transplanted biopsies: Technical perspectives for clinical practice application. Exp Mol Pathol. 116:1045162020. View Article : Google Scholar : PubMed/NCBI

48 

Achanta S, Gorky J, Leung C, Moss A, Robbins S, Eisenman L, Chen J, Tappan S, Heal M, Farahani N, et al: A comprehensive integrated anatomical and molecular atlas of rat intrinsic cardiac nervous system. iScience. 23:1011402020. View Article : Google Scholar : PubMed/NCBI

49 

Zhao T, Chiang ZD, Morriss JW, LaFave LM, Murray EM, Del Priore I, Meli K, Lareau CA, Nadaf NM, Li J, et al: Spatial genomics enables multi-modal study of clonal heterogeneity in tissues. Nature. 601:85–91. 2022. View Article : Google Scholar : PubMed/NCBI

50 

Liao J, Lu X, Shao X, Zhu L and Fan X: Uncovering an organ's molecular architecture at single-cell resolution by spatially resolved transcriptomics. Trends Biotechnol. 39:43–58. 2021. View Article : Google Scholar : PubMed/NCBI

51 

Satija R, Farrell JA, Gennert D, Schier AF and Regev A: Spatial reconstruction of single-cell gene expression data. Nat Biotechnol. 33:495–502. 2015. View Article : Google Scholar : PubMed/NCBI

52 

Sun YM and Chen YQ: Principles and innovative technologies for decrypting noncoding RNAs: From discovery and functional prediction to clinical application. J Hematol Oncol. 13:1092020. View Article : Google Scholar : PubMed/NCBI

53 

Zhang L, Yu X, Zheng L, Zhang Y, Li Y, Fang Q, Gao R, Kang B, Zhang Q, Huang JY, et al: Lineage tracking reveals dynamic relationships of T cells in colorectal cancer. Nature. 564:268–272. 2018. View Article : Google Scholar : PubMed/NCBI

54 

Zhang Q, He Y, Luo N, Patel SJ, Han Y, Gao R, Modak M, Carotta S, Haslinger C, Kind D, et al: Landscape and dynamics of single immune cells in hepatocellular carcinoma. Cell. 179:829–845.e20. 2019. View Article : Google Scholar : PubMed/NCBI

55 

Emmert-Buck MR, Bonner RF, Smith PD, Chuaqui RF, Zhuang Z, Goldstein SR, Weiss RA and Liotta LA: Laser capture microdissection. Science. 274:998–1001. 1996. View Article : Google Scholar : PubMed/NCBI

56 

Liew LC, Narsai R, Wang Y, Berkowitz O, Whelan J and Lewsey MG: Temporal tissue-specific regulation of transcriptomes during barley (Hordeum vulgare) seed germination. Plant J. 101:700–715. 2020. View Article : Google Scholar : PubMed/NCBI

57 

Shaw R, Tian X and Xu J: Single-cell transcriptome analysis in plants: Advances and challenges. Mol Plant. 14:115–126. 2021. View Article : Google Scholar : PubMed/NCBI

58 

Guo W, Hu Y, Qian J, Zhu L, Cheng J, Liao J and Fan X: Laser capture microdissection for biomedical research: Towards high-throughput, multi-omics, and single-cell resolution. J Genet Genomics. 50:641–651. 2023. View Article : Google Scholar : PubMed/NCBI

59 

Nichterwitz S, Chen G, Aguila Benitez J, Yilmaz M, Storvall H, Cao M, Sandberg R, Deng Q and Hedlund E: Laser capture microscopy coupled with Smart-seq2 for precise spatial transcriptomic profiling. Nat Commun. 7:121392016. View Article : Google Scholar : PubMed/NCBI

60 

Chen J, Suo S, Tam PP, Han JDJ, Peng G and Jing N: Spatial transcriptomic analysis of cryosectioned tissue samples with Geo-seq. Nat Protoc. 12:566–580. 2017. View Article : Google Scholar : PubMed/NCBI

61 

Casasent AK, Schalck A, Gao R, Sei E, Long A, Pangburn W, Casasent T, Meric-Bernstam F, Edgerton ME and Navin NE: Multiclonal invasion in breast tumors identified by topographic single cell sequencing. Cell. 172:205–217.e12. 2018. View Article : Google Scholar : PubMed/NCBI

62 

Junker JP, Noël ES, Guryev V, Peterson KA, Shah G, Huisken J, McMahon AP, Berezikov E, Bakkers J and van Oudenaarden A: Genome-wide RNA Tomography in the zebrafish embryo. Cell. 159:662–675. 2014. View Article : Google Scholar : PubMed/NCBI

63 

Moffitt JR, Bambah-Mukku D, Eichhorn SW, Vaughn E, Shekhar K, Perez JD, Rubinstein ND, Hao J, Regev A, Dulac C and Zhuang X: Molecular, spatial, and functional single-cell profiling of the hypothalamic preoptic region. Science. 362:eaau53242018. View Article : Google Scholar : PubMed/NCBI

64 

Xia C, Fan J, Emanuel G, Hao J and Zhuang X: Spatial transcriptome profiling by MERFISH reveals subcellular RNA compartmentalization and cell cycle-dependent gene expression. Proc Natl Acad Sci USA. 116:19490–19499. 2019. View Article : Google Scholar : PubMed/NCBI

65 

Femino AM, Fay FS, Fogarty K and Singer RH: Visualization of single RNA transcripts in situ. Science. 280:585–590. 1998. View Article : Google Scholar : PubMed/NCBI

66 

Wang F, Flanagan J, Su N, Wang LC, Bui S, Nielson A, Wu X, Vo HT, Ma XJ and Luo Y: RNAscope: A novel in situ RNA analysis platform for formalin-fixed, paraffin-embedded tissues. J Mol Diagn. 14:22–29. 2012. View Article : Google Scholar : PubMed/NCBI

67 

Lubeck E, Coskun AF, Zhiyentayev T, Ahmad M and Cai L: Single-cell in situ RNA profiling by sequential hybridization. Nat Methods. 11:360–361. 2014. View Article : Google Scholar : PubMed/NCBI

68 

Rodriques SG, Stickels RR, Goeva A, Martin CA, Murray E, Vanderburg CR, Welch J, Chen LM, Chen F and Macosko EZ: Slide-seq: A scalable technology for measuring genome-wide expression at high spatial resolution. Science. 363:1463–1467. 2019. View Article : Google Scholar : PubMed/NCBI

69 

Fazal FM, Han S, Parker KR, Kaewsapsak P, Xu J, Boettiger AN, Chang HY and Ting AY: Atlas of subcellular RNA localization revealed by APEX-seq. Cell. 178:473–490.e26. 2019. View Article : Google Scholar : PubMed/NCBI

70 

Vickovic S, Eraslan G, Salmén F, Klughammer J, Stenbeck L, Schapiro D, Äijö T, Bonneau R, Bergenstråhle L, Navarro JF, et al: High-definition spatial transcriptomics for in situ tissue profiling. Nat Methods. 16:987–990. 2019. View Article : Google Scholar : PubMed/NCBI

71 

Srivatsan SR, Regier MC, Barkan E, Franks JM, Packer JS, Grosjean P, Duran M, Saxton S, Ladd JJ, Spielmann M, et al: Embryo-scale, single-cell spatial transcriptomics. Science. 373:111–117. 2021. View Article : Google Scholar : PubMed/NCBI

72 

Liu Y, Yang M, Deng Y, Su G, Enninful A, Guo CC, Tebaldi T, Zhang D, Kim D, Bai Z, et al: High-spatial-resolution multi-omics sequencing via deterministic barcoding in tissue. Cell. 183:1665–1681.e18. 2020. View Article : Google Scholar : PubMed/NCBI

73 

Su G, Qin X, Enninful A, Bai Z, Deng Y, Liu Y and Fan R: Spatial multi-omics sequencing for fixed tissue via DBiT-seq. STAR Protoc. 2:1005322021. View Article : Google Scholar : PubMed/NCBI

74 

Dixon EE, Wu H, Sulvarán-Guel E, Guo J and Humphreys BD: Spatially resolved transcriptomics and the kidney: Many opportunities. Kidney Int. 102:482–491. 2022. View Article : Google Scholar : PubMed/NCBI

75 

Chen A, Liao S, Cheng M, Ma K, Wu L, Lai Y, Qiu X, Yang J, Xu J, Hao S, et al: Spatiotemporal transcriptomic atlas of mouse organogenesis using DNA nanoball-patterned arrays. Cell. 185:1777–1792.e21. 2022. View Article : Google Scholar : PubMed/NCBI

76 

Cho CS, Xi J, Si Y, Park SR, Hsu JE, Kim M, Jun G, Kang HM and Lee JH: Microscopic examination of spatial transcriptome using Seq-Scope. Cell. 184:3559–3572.e22. 2021. View Article : Google Scholar : PubMed/NCBI

77 

Levsky JM, Shenoy SM, Pezo RC and Singer RH: Single-cell gene expression profiling. Science. 297:836–840. 2002. View Article : Google Scholar : PubMed/NCBI

78 

Lubeck E and Cai L: Single-cell systems biology by super- resolution imaging and combinatorial labeling. Nat Methods. 9:743–748. 2012. View Article : Google Scholar : PubMed/NCBI

79 

Ke R, Mignardi M, Pacureanu A, Svedlund J, Botling J, Wählby C and Nilsson M: In situ sequencing for RNA analysis in preserved tissue and cells. Nat Methods. 10:857–860. 2013. View Article : Google Scholar : PubMed/NCBI

80 

Lee JH, Daugharthy ER, Scheiman J, Kalhor R, Ferrante TC, Terry R, Turczyk BM, Yang JL, Lee HS, Aach J, et al: Fluorescent in situ sequencing (FISSEQ) of RNA for gene expression profiling in intact cells and tissues. Nat Protoc. 10:442–458. 2015. View Article : Google Scholar : PubMed/NCBI

81 

Olin A, Henckel E, Chen Y, Lakshmikanth T, Pou C, Mikes J, Gustafsson A, Bernhardsson AK, Zhang C, Bohlin K and Brodin P: Stereotypic immune system development in newborn children. Cell. 174:1277–1292.e14. 2018. View Article : Google Scholar : PubMed/NCBI

82 

Soderholm AT and Pedicord VA: Intestinal epithelial cells: At the interface of the microbiota and mucosal immunity. Immunology. 158:267–280. 2019. View Article : Google Scholar : PubMed/NCBI

83 

Schreurs RRCE, Baumdick ME, Sagebiel AF, Kaufmann M, Mokry M, Klarenbeek PL, Schaltenberg N, Steinert FL, van Rijn JM, Drewniak A, et al: Human fetal TNF-α-cytokine-producing CD4+ effector memory T cells promote intestinal development and mediate inflammation early in life. Immunity. 50:462–476.e8. 2019. View Article : Google Scholar : PubMed/NCBI

84 

Olsen TK and Baryawno N: Introduction to single-cell RNA sequencing. Curr Protoc Mol Biol. 122:e572018. View Article : Google Scholar : PubMed/NCBI

85 

Rodaway A and Patient R: Mesendoderm. An ancient germ layer? Cell. 105:169–172. 2001.PubMed/NCBI

86 

Zorn AM and Wells JM: Molecular basis of vertebrate endoderm development. Int Rev Cytol. 259:49–111. 2007. View Article : Google Scholar : PubMed/NCBI

87 

Zorn AM and Wells JM: Vertebrate endoderm development and organ formation. Annu Rev Cell Dev Biol. 25:221–251. 2009. View Article : Google Scholar : PubMed/NCBI

88 

Kimelman D and Griffin KJ: Vertebrate mesendoderm induction and patterning. Curr Opin Genet Dev. 10:350–356. 2000. View Article : Google Scholar : PubMed/NCBI

89 

Spence JR, Lauf R and Shroyer NF: Vertebrate intestinal endoderm development. Dev Dyn. 240:501–520. 2011. View Article : Google Scholar : PubMed/NCBI

90 

Que J, Okubo T, Goldenring JR, Nam KT, Kurotani R, Morrisey EE, Taranova O, Pevny LH and Hogan BL: Multiple dose-dependent roles for Sox2 in the patterning and differentiation of anterior foregut endoderm. Development. 134:2521–2531. 2007. View Article : Google Scholar : PubMed/NCBI

91 

Sherwood RI, Chen TYA and Melton DA: Transcriptional dynamics of endodermal organ formation. Dev Dyn. 238:29–42. 2009. View Article : Google Scholar : PubMed/NCBI

92 

Walton KD, Whidden M, Kolterud Å, Shoffner SK, Czerwinski MJ, Kushwaha J, Parmar N, Chandhrasekhar D, Freddo AM, Schnell S and Gumucio DL: Villification in the mouse: Bmp signals control intestinal villus patterning. Development. 143:427–436. 2016.PubMed/NCBI

93 

Grey RD: Morphogenesis of intestinal villi. I. Scanning electron microscopy of the duodenal epithelium of the developing chick embryo. J Morphol. 137:193–213. 1972. View Article : Google Scholar : PubMed/NCBI

94 

Fawkner-Corbett D, Antanaviciute A, Parikh K, Jagielowicz M, Gerós AS, Gupta T, Ashley N, Khamis D, Fowler D, Morrissey E, et al: Spatiotemporal analysis of human intestinal development at single-cell resolution. Cell. 184:810–826.e23. 2021. View Article : Google Scholar : PubMed/NCBI

95 

Martin JC, Chang C, Boschetti G, Ungaro R, Giri M, Grout JA, Gettler K, Chuang LS, Nayar S, Greenstein AJ, et al: Single-cell analysis of Crohn's disease lesions identifies a pathogenic cellular module associated with resistance to anti-TNF therapy. Cell. 178:1493–1508.e20. 2019. View Article : Google Scholar : PubMed/NCBI

96 

Parikh K, Antanaviciute A, Fawkner-Corbett D, Jagielowicz M, Aulicino A, Lagerholm C, Davis S, Kinchen J, Chen HH, Alham NK, et al: Colonic epithelial cell diversity in health and inflammatory bowel disease. Nature. 567:49–55. 2019. View Article : Google Scholar : PubMed/NCBI

97 

Smillie CS, Biton M, Ordovas-Montanes J, Sullivan KM, Burgin G, Graham DB, Herbst RH, Rogel N, Slyper M, Waldman J, et al: Intra- and inter-cellular rewiring of the human colon during ulcerative colitis. Cell. 178:714–730.e22. 2019. View Article : Google Scholar : PubMed/NCBI

98 

Fenderico N, van Scherpenzeel RC, Goldflam M, Proverbio D, Jordens I, Kralj T, Stryeck S, Bass TZ, Hermans G, Ullman C, et al: Anti-LRP5/6 VHHs promote differentiation of Wnt-hypersensitive intestinal stem cells. Nat Commun. 10:3652019. View Article : Google Scholar : PubMed/NCBI

99 

Kinchen J, Chen HH, Parikh K, Antanaviciute A, Jagielowicz M, Fawkner-Corbett D, Ashley N, Cubitt L, Mellado-Gomez E, Attar M, et al: Structural remodeling of the human colonic mesenchyme in inflammatory bowel disease. Cell. 175:372–386.e17. 2018. View Article : Google Scholar : PubMed/NCBI

100 

Powell DW, Pinchuk IV, Saada JI, Chen X and Mifflin RC: Mesenchymal cells of the intestinal lamina propria. Annu Rev Physiol. 73:213–237. 2011. View Article : Google Scholar : PubMed/NCBI

101 

McCarthy N, Manieri E, Storm EE, Saadatpour A, Luoma AM, Kapoor VN, Madha S, Gaynor LT, Cox C, Keerthivasan S, et al: Distinct mesenchymal cell populations generate the essential intestinal BMP signaling gradient. Cell Stem Cell. 26:391–402.e5. 2020. View Article : Google Scholar : PubMed/NCBI

102 

Degirmenci B, Valenta T, Dimitrieva S, Hausmann G and Basler K: GLI1-expressing mesenchymal cells form the essential Wnt-secreting niche for colon stem cells. Nature. 558:449–453. 2018. View Article : Google Scholar : PubMed/NCBI

103 

van Es JH, Sato T, van de Wetering M, Lyubimova A, Yee Nee AN, Gregorieff A, Sasaki N, Zeinstra L, van den Born M, Korving J, et al: Dll1+ secretory progenitor cells revert to stem cells upon crypt damage. Nat Cell Biol. 14:1099–1104. 2012. View Article : Google Scholar : PubMed/NCBI

104 

van de Pavert SA and Mebius RE: New insights into the development of lymphoid tissues. Nat Rev Immunol. 10:664–674. 2010. View Article : Google Scholar : PubMed/NCBI

105 

Scheich S, Chen J, Liu J, Schnütgen F, Enssle JC, Ceribelli M, Thomas CJ, Choi J, Morris V, Hsiao T, et al: Targeting N-linked glycosylation for the therapy of aggressive lymphomas. Cancer Discov. 13:1862–1883. 2023. View Article : Google Scholar : PubMed/NCBI

106 

Guan Q: A comprehensive review and update on the pathogenesis of inflammatory bowel disease. J Immunol Res. 2019:72472382019. View Article : Google Scholar : PubMed/NCBI

107 

Tang F, Barbacioru C, Wang Y, Nordman E, Lee C, Xu N, Wang X, Bodeau J, Tuch BB, Siddiqui A, et al: mRNA-Seq whole-transcriptome analysis of a single cell. Nat Methods. 6:377–382. 2009. View Article : Google Scholar : PubMed/NCBI

108 

Yu X, Abbas-Aghababazadeh F, Chen YA and Fridley BL: Statistical and bioinformatics analysis of data from bulk and single-cell RNA sequencing experiments. Methods Mol Biol. 2194:143–175. 2021. View Article : Google Scholar : PubMed/NCBI

109 

Nath A and Bild AH: Leveraging single-cell approaches in cancer precision medicine. Trends Cancer. 7:359–372. 2021. View Article : Google Scholar : PubMed/NCBI

110 

Czarnewski P, Parigi SM, Sorini C, Diaz OE, Das S, Gagliani N and Villablanca EJ: Conserved transcriptomic profile between mouse and human colitis allows unsupervised patient stratification. Nat Commun. 10:28922019. View Article : Google Scholar : PubMed/NCBI

111 

Parigi SM, Larsson L, Das S, Ramirez Flores RO, Frede A, Tripathi KP, Diaz OE, Selin K, Morales RA, Luo X, et al: The spatial transcriptomic landscape of the healing mouse intestine following damage. Nat Commun. 13:8282022. View Article : Google Scholar : PubMed/NCBI

112 

Ayyaz A, Kumar S, Sangiorgi B, Ghoshal B, Gosio J, Ouladan S, Fink M, Barutcu S, Trcka D, Shen J, et al: Single-cell transcriptomes of the regenerating intestine reveal a revival stem cell. Nature. 569:121–125. 2019. View Article : Google Scholar : PubMed/NCBI

113 

Smythies LE, Sellers M, Clements RH, Mosteller-Barnum M, Meng G, Benjamin WH, Orenstein JM and Smith PD: Human intestinal macrophages display profound inflammatory anergy despite avid phagocytic and bacteriocidal activity. J Clin Invest. 115:66–75. 2005. View Article : Google Scholar : PubMed/NCBI

114 

Martinez FO and Gordon S: The M1 and M2 paradigm of macrophage activation: Time for reassessment. F1000Prime Rep. 6:132014. View Article : Google Scholar : PubMed/NCBI

115 

Garrido-Trigo A, Corraliza AM, Veny M, Dotti I, Melón-Ardanaz E, Rill A, Crowell HL, Corbí Á, Gudiño V, Esteller M, et al: Macrophage and neutrophil heterogeneity at single-cell spatial resolution in human inflammatory bowel disease. Nat Commun. 14:45062023. View Article : Google Scholar : PubMed/NCBI

116 

Schoeler M and Caesar R: Dietary lipids, gut microbiota and lipid metabolism. Rev Endocr Metab Disord. 20:461–472. 2019. View Article : Google Scholar : PubMed/NCBI

117 

Milani C, Duranti S, Bottacini F, Casey E, Turroni F, Mahony J, Belzer C, Delgado Palacio S, Arboleya Montes S, Mancabelli L, et al: The first microbial colonizers of the human gut: Composition, activities, and health implications of the infant gut microbiota. Microbiol Mol Biol Rev. 81:e00036–17. 2017. View Article : Google Scholar : PubMed/NCBI

118 

Human Microbiome Project Consortium, . Structure, function and diversity of the healthy human microbiome. Nature. 486:207–214. 2012. View Article : Google Scholar : PubMed/NCBI

119 

Lloyd-Price J, Arze C, Ananthakrishnan AN, Schirmer M, Avila-Pacheco J, Poon TW, Andrews E, Ajami NJ, Bonham KS, Brislawn CJ, et al: Multi-omics of the gut microbial ecosystem in inflammatory bowel diseases. Nature. 569:655–662. 2019. View Article : Google Scholar : PubMed/NCBI

120 

Huang H, Ren Z, Gao X, Hu X, Zhou Y, Jiang J, Lu H, Yin S, Ji J, Zhou L and Zheng S: Integrated analysis of microbiome and host transcriptome reveals correlations between gut microbiota and clinical outcomes in HBV-related hepatocellular carcinoma. Genome Med. 12:1022020. View Article : Google Scholar : PubMed/NCBI

121 

Zhang SL, Cheng LS, Zhang ZY, Sun HT and Li JJ: Untangling determinants of gut microbiota and tumor immunologic status through a multi-omics approach in colorectal cancer. Pharmacol Res. 188:1066332023. View Article : Google Scholar : PubMed/NCBI

122 

Pereira FC and Berry D: Microbial nutrient niches in the gut. Environ Microbiol. 19:1366–1378. 2017. View Article : Google Scholar : PubMed/NCBI

123 

Kim HJ, Boedicker JQ, Choi JW and Ismagilov RF: Defined spatial structure stabilizes a synthetic multispecies bacterial community. Proc Natl Acad Sci USA. 105:18188–18193. 2008. View Article : Google Scholar : PubMed/NCBI

124 

Rausch P, Basic M, Batra A, Bischoff SC, Blaut M, Clavel T, Gläsner J, Gopalakrishnan S, Grassl GA, Günther C, et al: Analysis of factors contributing to variation in the C57BL/6J fecal microbiota across German animal facilities. Int J Med Microbiol. 306:343–355. 2016. View Article : Google Scholar : PubMed/NCBI

125 

Seekatz AM, Schnizlein MK, Koenigsknecht MJ, Baker JR, Hasler WL, Bleske BE, Young VB and Sun D: Spatial and temporal analysis of the stomach and small-intestinal microbiota in fasted healthy humans. mSphere. 4:e00126–19. 2019. View Article : Google Scholar : PubMed/NCBI

126 

Sheth RU, Li M, Jiang W, Sims PA, Leong KW and Wang HH: Spatial metagenomic characterization of microbial biogeography in the gut. Nat Biotechnol. 37:877–883. 2019. View Article : Google Scholar : PubMed/NCBI

127 

Choi CH and Chang SK: Role of small intestinal bacterial overgrowth in functional gastrointestinal disorders. J Neurogastroenterol Motil. 22:3–5. 2016. View Article : Google Scholar : PubMed/NCBI

128 

Evans DF, Pye G, Bramley R, Clark AG, Dyson TJ and Hardcastle JD: Measurement of gastrointestinal pH profiles in normal ambulant human subjects. Gut. 29:1035–1041. 1988. View Article : Google Scholar : PubMed/NCBI

129 

Macfarlane GT, Gibson GR and Cummings JH: Comparison of fermentation reactions in different regions of the human colon. J Appl Bacteriol. 72:57–64. 1992. View Article : Google Scholar : PubMed/NCBI

130 

Birchenough GM, Johansson ME, Gustafsson JK, Bergström JH and Hansson GC: New developments in goblet cell mucus secretion and function. Mucosal Immunol. 8:712–719. 2015. View Article : Google Scholar : PubMed/NCBI

131 

Kotarsky K, Sitnik KM, Stenstad H, Kotarsky H, Schmidtchen A, Koslowski M, Wehkamp J and Agace WW: A novel role for constitutively expressed epithelial-derived chemokines as antibacterial peptides in the intestinal mucosa. Mucosal Immunol. 3:40–48. 2010. View Article : Google Scholar : PubMed/NCBI

132 

McCallum G and Tropini C: The gut microbiota and its biogeography. Nat Rev Microbiol. 22:105–118. 2024. View Article : Google Scholar : PubMed/NCBI

133 

Cremer J, Arnoldini M and Hwa T: Effect of water flow and chemical environment on microbiota growth and composition in the human colon. Proc Natl Acad Sci USA. 114:6438–6443. 2017. View Article : Google Scholar : PubMed/NCBI

134 

Cremer J, Segota I, Yang CY, Arnoldini M, Sauls JT, Zhang Z, Gutierrez E, Groisman A and Hwa T: Effect of flow and peristaltic mixing on bacterial growth in a gut-like channel. Proc Natl Acad Sci USA. 113:11414–11419. 2016. View Article : Google Scholar : PubMed/NCBI

135 

Nagara Y, Takada T, Nagata Y, Kado S and Kushiro A: Microscale spatial analysis provides evidence for adhesive monopolization of dietary nutrients by specific intestinal bacteria. PLoS One. 12:e01754972017. View Article : Google Scholar : PubMed/NCBI

136 

Walker AW, Duncan SH, Harmsen HJ, Holtrop G, Welling GW and Flint HJ: The species composition of the human intestinal microbiota differs between particle-associated and liquid phase communities. Environ Microbiol. 10:3275–3283. 2008. View Article : Google Scholar : PubMed/NCBI

137 

Leitch ECMW, Walker AW, Duncan SH, Holtrop G and Flint HJ: Selective colonization of insoluble substrates by human faecal bacteria. Environ Microbiol. 9:667–679. 2007. View Article : Google Scholar : PubMed/NCBI

138 

Sobhani I, Bergsten E, Couffin S, Amiot A, Nebbad B, Barau C, de'Angelis N, Rabot S, Canoui-Poitrine F, Mestivier D, et al: Colorectal cancer-associated microbiota contributes to oncogenic epigenetic signatures. Proc Natl Acad Sci USA. 116:24285–24295. 2019. View Article : Google Scholar : PubMed/NCBI

139 

David LA, Maurice CF, Carmody RN, Gootenberg DB, Button JE, Wolfe BE, Ling AV, Devlin AS, Varma Y, Fischbach MA, et al: Diet rapidly and reproducibly alters the human gut microbiome. Nature. 505:559–563. 2014. View Article : Google Scholar : PubMed/NCBI

140 

Nguyen J, Pepin DM and Tropini C: Cause or effect? The spatial organization of pathogens and the gut microbiota in disease. Microbes Infect. 23:1048152021. View Article : Google Scholar : PubMed/NCBI

141 

Borghini R, Donato G, Alvaro D and Picarelli A: New insights in IBS-like disorders: Pandora's box has been opened; a review. Gastroenterol Hepatol Bed Bench. 10:79–89. 2017.PubMed/NCBI

142 

Ng QX, Soh AYS, Loke W, Lim DY and Yeo WS: The role of inflammation in irritable bowel syndrome (IBS). J Inflamm Res. 11:345–349. 2018. View Article : Google Scholar : PubMed/NCBI

143 

Qiao LY and Tiwari N: Spinal neuron-glia-immune interaction in cross-organ sensitization. Am J Physiol Gastrointest Liver Physiol. 319:G748–G760. 2020. View Article : Google Scholar : PubMed/NCBI

144 

North RY, Li Y, Ray P, Rhines LD, Tatsui CE, Rao G, Johansson CA, Zhang H, Kim YH, Zhang B, et al: Electrophysiological and transcriptomic correlates of neuropathic pain in human dorsal root ganglion neurons. Brain. 142:1215–1226. 2019. View Article : Google Scholar : PubMed/NCBI

145 

Tavares-Ferreira D, Shiers S, Ray PR, Wangzhou A, Jeevakumar V, Sankaranarayanan I, Cervantes AM, Reese JC, Chamessian A, Copits BA, et al: Spatial transcriptomics of dorsal root ganglia identifies molecular signatures of human nociceptors. Sci Transl Med. 14:eabj81862022. View Article : Google Scholar : PubMed/NCBI

146 

Bonaz BL and Bernstein CN: Brain-gut interactions in inflammatory bowel disease. Gastroenterology. 144:36–49. 2013. View Article : Google Scholar : PubMed/NCBI

147 

Tsigos C and Chrousos GP: Hypothalamic-pituitary-adrenal axis, neuroendocrine factors and stress. J Psychosom Res. 53:865–871. 2002. View Article : Google Scholar : PubMed/NCBI

148 

Kitaoka S: Inflammation in the brain and periphery found in animal models of depression and its behavioral relevance. J Pharmacol Sci. 148:262–266. 2022. View Article : Google Scholar : PubMed/NCBI

149 

De Bellis MD, Casey BJ, Dahl RE, Birmaher B, Williamson DE, Thomas KM, Axelson DA, Frustaci K, Boring AM, Hall J and Ryan ND: A pilot study of amygdala volumes in pediatric generalized anxiety disorder. Biol Psychiatry. 48:51–57. 2000. View Article : Google Scholar : PubMed/NCBI

150 

Schienle A, Ebner F and Schäfer A: Localized gray matter volume abnormalities in generalized anxiety disorder. Eur Arch Psychiatry Clin Neurosci. 261:303–307. 2011. View Article : Google Scholar : PubMed/NCBI

151 

Qiao J, Li A, Cao C, Wang Z, Sun J and Xu G: Aberrant functional network connectivity as a biomarker of generalized anxiety disorder. Front Hum Neurosci. 11:6262017. View Article : Google Scholar : PubMed/NCBI

152 

Moffitt JR, Hao J, Bambah-Mukku D, Lu T, Dulac C and Zhuang X: High-performance multiplexed fluorescence in situ hybridization in culture and tissue with matrix imprinting and clearing. Proc Natl Acad Sci USA. 113:14456–14461. 2016. View Article : Google Scholar : PubMed/NCBI

153 

Zhou X, Lu Y, Zhao F, Dong J, Ma W, Zhong S, Wang M, Wang B, Zhao Y, Shi Y, et al: Deciphering the spatial-temporal transcriptional landscape of human hypothalamus development. Cell Stem Cell. 29:328–343.e5. 2022. View Article : Google Scholar : PubMed/NCBI

154 

Oh SW, Harris JA, Ng L, Winslow B, Cain N, Mihalas S, Wang Q, Lau C, Kuan L, Henry AM, et al: A mesoscale connectome of the mouse brain. Nature. 508:207–214. 2014. View Article : Google Scholar : PubMed/NCBI

155 

Qian J, Olbrecht S, Boeckx B, Vos H, Laoui D, Etlioglu E, Wauters E, Pomella V, Verbandt S, Busschaert P, et al: A pan-cancer blueprint of the heterogeneous tumor microenvironment revealed by single-cell profiling. Cell Res. 30:745–762. 2020. View Article : Google Scholar : PubMed/NCBI

156 

Galeano Niño JL, Wu H, LaCourse KD, Kempchinsky AG, Baryiames A, Barber B, Futran N, Houlton J, Sather C, Sicinska E, et al: Effect of the intratumoral microbiota on spatial and cellular heterogeneity in cancer. Nature. 611:810–817. 2022. View Article : Google Scholar : PubMed/NCBI

157 

Ozato Y, Kojima Y, Kobayashi Y, Hisamatsu Y, Toshima T, Yonemura Y, Masuda T, Kagawa K, Goto Y, Utou M, et al: Spatial and single-cell transcriptomics decipher the cellular environment containing HLA-G+ cancer cells and SPP1+ macrophages in colorectal cancer. Cell Rep. 42:1119292023. View Article : Google Scholar : PubMed/NCBI

158 

Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A and Bray F: Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 71:209–249. 2021. View Article : Google Scholar : PubMed/NCBI

159 

Dekker E, Tanis PJ, Vleugels JLA, Kasi PM and Wallace MB: Colorectal cancer. Lancet. 394:1467–1480. 2019. View Article : Google Scholar : PubMed/NCBI

160 

Zhao W, Dai S, Yue L, Xu F, Gu J, Dai X and Qian X: Emerging mechanisms progress of colorectal cancer liver metastasis. Front Endocrinol (Lausanne). 13:10815852022. View Article : Google Scholar : PubMed/NCBI

161 

Brouwer NP, Webbink L, Haddad TS, Rutgers N, van Vliet S, Wood CS, Jansen PW, Lafarge MW; imCMS Consortium; de Wilt JH, ; et al: Transcriptomics and proteomics reveal distinct biology for lymph node metastases and tumour deposits in colorectal cancer. J Pathol. 261:401–412. 2023. View Article : Google Scholar : PubMed/NCBI

162 

Kobayashi H, Gieniec KA, Lannagan TRM, Wang T, Asai N, Mizutani Y, Iida T, Ando R, Thomas EM, Sakai A, et al: The origin and contribution of cancer-associated fibroblasts in colorectal carcinogenesis. Gastroenterology. 162:890–906. 2022. View Article : Google Scholar : PubMed/NCBI

163 

Peng Z, Ye M, Ding H, Feng Z and Hu K: Spatial transcriptomics atlas reveals the crosstalk between cancer-associated fibroblasts and tumor microenvironment components in colorectal cancer. J Transl Med. 20:3022022. View Article : Google Scholar : PubMed/NCBI

164 

Qi J, Sun H, Zhang Y, Wang Z, Xun Z, Li Z, Ding X, Bao R, Hong L, Jia W, et al: Single-cell and spatial analysis reveal interaction of FAP+ fibroblasts and SPP1+ macrophages in colorectal cancer. Nat Commun. 13:17422022. View Article : Google Scholar : PubMed/NCBI

165 

Valdeolivas A, Amberg B, Giroud N, Richardson M, Gálvez EJC, Badillo S, Julien-Laferrière A, Túrós D, Voith von Voithenberg L, Wells I, et al: Profiling the heterogeneity of colorectal cancer consensus molecular subtypes using spatial transcriptomics. NPJ Precis Oncol. 8:102024. View Article : Google Scholar : PubMed/NCBI

166 

Roelands J, van der Ploeg M, Ijsselsteijn ME, Dang H, Boonstra JJ, Hardwick JCH, Hawinkels LJAC, Morreau H and de Miranda NFCC: Transcriptomic and immunophenotypic profiling reveals molecular and immunological hallmarks of colorectal cancer tumourigenesis. Gut. 72:1326–1339. 2023. View Article : Google Scholar : PubMed/NCBI

167 

Zwing N, Failmezger H, Ooi CH, Hibar DP, Cañamero M, Gomes B, Gaire F and Korski K: Analysis of spatial organization of suppressive myeloid cells and effector T cells in colorectal cancer-A potential tool for discovering prognostic biomarkers in clinical research. Front Immunol. 11:5502502020. View Article : Google Scholar : PubMed/NCBI

168 

Liu HT, Chen SY, Peng LL, Zhong L, Zhou L, Liao SQ, Chen ZJ, Wang QL, He S and Zhou ZH: Spatially resolved transcriptomics revealed local invasion-related genes in colorectal cancer. Front Oncol. 13:10890902023. View Article : Google Scholar : PubMed/NCBI

169 

Manfredi S, Lepage C, Hatem C, Coatmeur O, Faivre J and Bouvier AM: Epidemiology and management of liver metastases from colorectal cancer. Ann Surg. 244:254–259. 2006. View Article : Google Scholar : PubMed/NCBI

170 

Hu Z, Ding J, Ma Z, Sun R, Seoane JA, Scott Shaffer J, Suarez CJ, Berghoff AS, Cremolini C, Falcone A, et al: Quantitative evidence for early metastatic seeding in colorectal cancer. Nat Genet. 51:1113–1122. 2019. View Article : Google Scholar : PubMed/NCBI

171 

Wang F, Long J, Li L, Wu ZX, Da TT, Wang XQ, Huang C, Jiang YH, Yao XQ, Ma HQ, et al: Single-cell and spatial transcriptome analysis reveals the cellular heterogeneity of liver metastatic colorectal cancer. Sci Adv. 9:eadf54642023. View Article : Google Scholar : PubMed/NCBI

172 

Garbarino O, Lambroia L, Basso G, Marrella V, Franceschini B, Soldani C, Pasqualini F, Giuliano D, Costa G, Peano C, et al: Spatial resolution of cellular senescence dynamics in human colorectal liver metastasis. Aging Cell. 22:e138532023. View Article : Google Scholar : PubMed/NCBI

173 

Cheng LK, O'Grady G, Du P, Egbuji JU, Windsor JA and Pullan AJ: Gastrointestinal system. Wiley Interdiscip Rev Syst Biol Med. 2:65–79. 2010. View Article : Google Scholar : PubMed/NCBI

174 

Peterson LW and Artis D: Intestinal epithelial cells: Regulators of barrier function and immune homeostasis. Nat Rev Immunol. 14:141–153. 2014. View Article : Google Scholar : PubMed/NCBI

175 

Pelka K, Hofree M, Chen JH, Sarkizova S, Pirl JD, Jorgji V, Bejnood A, Dionne D, Ge WH, Xu KH, et al: Spatially organized multicellular immune hubs in human colorectal cancer. Cell. 184:4734–4752.e20. 2021. View Article : Google Scholar : PubMed/NCBI

176 

Frede A, Czarnewski P, Monasterio G, Tripathi KP, Bejarano DA, Ramirez Flores RO, Sorini C, Larsson L, Luo X, Geerlings L, et al: B cell expansion hinders the stroma-epithelium regenerative cross talk during mucosal healing. Immunity. 55:2336–2351.e12. 2022. View Article : Google Scholar : PubMed/NCBI

177 

Zhang X, Hu C, Huang C, Wei Y, Li X, Hu M, Li H, Wu J, Czajkowsky DM, Guo Y and Shao Z: Robust Acquisition of spatial transcriptional programs in tissues with immunofluorescence-guided laser capture microdissection. Front Cell Dev Biol. 10:8531882022. View Article : Google Scholar : PubMed/NCBI

178 

Ghaddar B and De S: Reconstructing physical cell interaction networks from single-cell data using Neighbor-seq. Nucleic Acids Res. 50:e822022. View Article : Google Scholar : PubMed/NCBI

179 

Zhu Z, Cai J, Hou W, Xu K, Wu X, Song Y, Bai C, Mo YY and Zhang Z: Microbiome and spatially resolved metabolomics analysis reveal the anticancer role of gut Akkermansia muciniphila by crosstalk with intratumoral microbiota and reprogramming tumoral metabolism in mice. Gut Microbes. 15:21667002023. View Article : Google Scholar : PubMed/NCBI

180 

Sun C, Wang A, Zhou Y, Chen P, Wang X, Huang J, Gao J, Wang X, Shu L, Lu J, et al: Spatially resolved multi-omics highlights cell-specific metabolic remodeling and interactions in gastric cancer. Nat Commun. 14:26922023. View Article : Google Scholar : PubMed/NCBI

181 

Tun HM, Peng Y, Massimino L, Sin ZY, Parigi TL, Facoetti A, Rahman S, Danese S and Ungaro F: Gut virome in inflammatory bowel disease and beyond. Gut. 73:350–360. 2024. View Article : Google Scholar : PubMed/NCBI

182 

Zhao E, Stone MR, Ren X, Guenthoer J, Smythe KS, Pulliam T, Williams SR, Uytingco CR, Taylor SEB, Nghiem P, et al: Spatial transcriptomics at subspot resolution with BayesSpace. Nat Biotechnol. 39:1375–1384. 2021. View Article : Google Scholar : PubMed/NCBI

183 

Saviano A, Henderson NC and Baumert TF: Single-cell genomics and spatial transcriptomics: Discovery of novel cell states and cellular interactions in liver physiology and disease biology. J Hepatol. 73:1219–1230. 2020. View Article : Google Scholar : PubMed/NCBI

184 

Peterson VM, Zhang KX, Kumar N, Wong J, Li L, Wilson DC, Moore R, McClanahan TK, Sadekova S and Klappenbach JA: Multiplexed quantification of proteins and transcripts in single cells. Nat Biotechnol. 35:936–939. 2017. View Article : Google Scholar : PubMed/NCBI

185 

Thornton CA, Mulqueen RM, Torkenczy KA, Nishida A, Lowenstein EG, Fields AJ, Steemers FJ, Zhang W, McConnell HL, Woltjer RL, et al: Spatially mapped single-cell chromatin accessibility. Nat Commun. 12:12742021. View Article : Google Scholar : PubMed/NCBI

186 

Stoeckius M, Hafemeister C, Stephenson W, Houck-Loomis B, Chattopadhyay PK, Swerdlow H, Satija R and Smibert P: Simultaneous epitope and transcriptome measurement in single cells. Nat Methods. 14:865–868. 2017. View Article : Google Scholar : PubMed/NCBI

187 

Vickovic S, Lötstedt B, Klughammer J, Mages S, Segerstolpe Å, Rozenblatt-Rosen O and Regev A: SM-omics is an automated platform for high-throughput spatial multi-omics. Nat Commun. 13:7952022. View Article : Google Scholar : PubMed/NCBI

188 

Park HE, Jo SH, Lee RH, Macks CP, Ku T, Park J, Lee CW, Hur JK and Sohn CH: Spatial transcriptomics: Technical aspects of recent developments and their applications in neuroscience and cancer research. Adv Sci (Weinh). 10:e22069392023. View Article : Google Scholar : PubMed/NCBI

189 

Rao A, Barkley D, França GS and Yanai I: Exploring tissue architecture using spatial transcriptomics. Nature. 596:211–220. 2021. View Article : Google Scholar : PubMed/NCBI

190 

Zhang L, Chen D, Song D, Liu X, Zhang Y, Xu X and Wang X: Clinical and translational values of spatial transcriptomics. Signal Transduct Target Ther. 7:1112022. View Article : Google Scholar : PubMed/NCBI

191 

Li Y, Stanojevic S and Garmire LX: Emerging artificial intelligence applications in spatial transcriptomics analysis. Comput Struct Biotechnol J. 20:2895–2908. 2022. View Article : Google Scholar : PubMed/NCBI

192 

Hao M, Hua K and Zhang X: SOMDE: A scalable method for identifying spatially variable genes with self-organizing map. Bioinformatics. 37:4392–4398. 2021. View Article : Google Scholar : PubMed/NCBI

193 

Dries R, Zhu Q, Dong R, Eng CL, Li H, Liu K, Fu Y, Zhao T, Sarkar A, Bao F, et al: Giotto: A toolbox for integrative analysis and visualization of spatial expression data. Genome Biol. 22:782021. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

September-2024
Volume 30 Issue 3

Print ISSN: 1791-2997
Online ISSN:1791-3004

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Guo Y, Ren C, He Y, Wu Y and Yang X: Deciphering the spatiotemporal transcriptional landscape of intestinal diseases (Review). Mol Med Rep 30: 157, 2024
APA
Guo, Y., Ren, C., He, Y., Wu, Y., & Yang, X. (2024). Deciphering the spatiotemporal transcriptional landscape of intestinal diseases (Review). Molecular Medicine Reports, 30, 157. https://doi.org/10.3892/mmr.2024.13281
MLA
Guo, Y., Ren, C., He, Y., Wu, Y., Yang, X."Deciphering the spatiotemporal transcriptional landscape of intestinal diseases (Review)". Molecular Medicine Reports 30.3 (2024): 157.
Chicago
Guo, Y., Ren, C., He, Y., Wu, Y., Yang, X."Deciphering the spatiotemporal transcriptional landscape of intestinal diseases (Review)". Molecular Medicine Reports 30, no. 3 (2024): 157. https://doi.org/10.3892/mmr.2024.13281