Appearance
This report is written by MaltSci based on the latest literature and research findings
How does spatial transcriptomics map gene expression?
Abstract
Spatial transcriptomics is a cutting-edge technology that integrates transcriptomic data with spatial context, revolutionizing our understanding of gene expression within tissues. This innovative approach enables high-resolution mapping of gene expression patterns, allowing researchers to investigate cellular microenvironments and complex biological interactions that are crucial for comprehending health and disease mechanisms. Traditional transcriptomic methods, which analyze gene expression in bulk samples, fall short in capturing the intricate spatial patterns necessary for a holistic understanding of biological processes. The significance of spatial transcriptomics is underscored by its applications in various fields, including cancer research, developmental biology, and regenerative medicine, where it has enhanced insights into cellular heterogeneity and tissue dynamics. Despite its transformative potential, spatial transcriptomics faces challenges such as technical hurdles, data analysis complexities, and biological variability, which must be addressed to facilitate broader adoption. This review provides an overview of the foundational principles, historical development, techniques, applications, challenges, and future directions of spatial transcriptomics, emphasizing its importance in advancing precision medicine and targeted therapies. As the field evolves, spatial transcriptomics is poised to become an essential tool in elucidating the molecular underpinnings of diseases and informing therapeutic strategies.
Outline
This report will discuss the following questions.
- 1 Introduction
- 2 Overview of Spatial Transcriptomics
- 2.1 Definition and Importance
- 2.2 Historical Development and Evolution
- 3 Techniques in Spatial Transcriptomics
- 3.1 In Situ Hybridization Methods
- 3.2 High-Throughput Sequencing Approaches
- 3.3 Integration with Imaging Techniques
- 4 Applications of Spatial Transcriptomics
- 4.1 Cancer Research
- 4.2 Developmental Biology
- 4.3 Neurobiology and Other Fields
- 5 Challenges and Limitations
- 5.1 Technical Limitations
- 5.2 Data Analysis Challenges
- 5.3 Biological Variability
- 6 Future Directions
- 6.1 Technological Advancements
- 6.2 Potential for Clinical Applications
- 6.3 Integration with Other Omics Technologies
- 7 Conclusion
1 Introduction
Spatial transcriptomics is an innovative technology that has revolutionized our understanding of gene expression by integrating transcriptomic data with spatial context within tissues. This approach allows researchers to map gene expression at unprecedented resolutions, providing insights into the cellular microenvironment and the complex interactions that occur within biological systems. As the field of molecular biology has evolved, traditional transcriptomic methods, which typically analyze gene expression in bulk tissue samples, have proven inadequate for capturing the intricate spatial patterns that are crucial for understanding biological processes in health and disease. The advent of spatial transcriptomics addresses this gap, enabling the analysis of gene expression while preserving the spatial architecture of tissues [1][2].
The significance of spatial transcriptomics lies in its potential applications across various domains, including developmental biology, cancer research, and regenerative medicine. By elucidating the spatial organization of gene expression, this technology can reveal insights into cellular heterogeneity, tissue dynamics, and the molecular underpinnings of diseases. For instance, spatial transcriptomics has been instrumental in identifying the spatial distribution of different cell types within tumors, thus enhancing our understanding of tumor microenvironments and informing therapeutic strategies [3][4]. Furthermore, the ability to investigate gene expression patterns in specific anatomical regions allows researchers to explore developmental processes and disease mechanisms with greater precision [5].
Despite its transformative potential, spatial transcriptomics faces several challenges and limitations. Technical hurdles, such as the need for specialized equipment and expertise, as well as data analysis complexities, can hinder the widespread adoption of this technology. Additionally, biological variability within tissues poses significant challenges in interpreting spatial transcriptomic data [6][7]. As the field continues to advance, addressing these challenges will be critical for the successful integration of spatial transcriptomics into routine research and clinical practice.
This review aims to provide a comprehensive overview of spatial transcriptomics, beginning with an exploration of its foundational principles and historical development. We will discuss various techniques employed in spatial transcriptomics, including in situ hybridization methods, high-throughput sequencing approaches, and the integration of imaging techniques [2][8]. Following this, we will delve into the applications of spatial transcriptomics across different fields, highlighting its contributions to cancer research, developmental biology, and neurobiology [1][3]. We will also examine the challenges and limitations associated with current methodologies, including technical constraints and data analysis challenges, and consider future directions for the field, focusing on technological advancements and the potential for clinical applications [5][9].
In conclusion, spatial transcriptomics represents a significant leap forward in our ability to study gene expression within its spatial context. By mapping gene expression patterns, this technology not only enhances our understanding of biological processes but also opens new avenues for research and therapeutic development. As we explore the intricacies of spatial transcriptomics, we will emphasize its importance in advancing our comprehension of cellular heterogeneity and tissue dynamics, ultimately contributing to the broader goals of precision medicine and targeted therapies.
2 Overview of Spatial Transcriptomics
2.1 Definition and Importance
Spatial transcriptomics is a cutting-edge technology that enables the mapping of gene expression within tissues while preserving the spatial context of cellular arrangements. This innovative approach integrates high-throughput transcriptomic analysis with spatial data, allowing researchers to generate high-resolution maps of gene expression patterns in intact cell and tissue samples. The significance of spatial transcriptomics lies in its ability to provide a comprehensive understanding of the spatial distribution of gene expression, which is crucial for elucidating the functional organization of tissues and understanding various biological processes and diseases.
The foundational concept of spatial transcriptomics is the combination of gene expression profiling with spatial coordinates. This technology emerged in the early 2010s and has rapidly evolved, incorporating methods such as in situ hybridization, in situ sequencing, and microdissection-based techniques, each offering advanced mapping resolution and precise assessments at the single-cell level [1]. By enabling researchers to know which genes are expressed by specific cells and in which locations within a tissue, spatial transcriptomics facilitates a deeper understanding of tissue architecture, cellular interactions, and the pathophysiology of diseases [3].
Spatial transcriptomics operates through several key methodologies. Techniques such as microdissection allow for the isolation of specific tissue regions, while in situ sequencing provides direct measurement of RNA molecules within their native context. These methods are complemented by advancements in high-throughput sequencing technologies, which have significantly enhanced the capability to analyze gene expression across various biological contexts, including cancer, developmental biology, and neurodegenerative diseases [2].
The practical applications of spatial transcriptomics are vast. For instance, it has been instrumental in characterizing the spatial organization of tissues in human skin research, revealing insights into dermatological diseases by mapping gene expression patterns related to skin function and dysfunction [3]. Furthermore, spatial transcriptomics has proven valuable in the field of autoimmune rheumatic diseases, where it has provided insights into immune cell distribution and interactions within affected tissues, thereby advancing personalized treatment strategies [1].
As the technology continues to evolve, the latest advancements include highly multiplexed methods and the ability to achieve single-cell resolution, which enhances the granularity of data obtained [2]. The integration of sophisticated data analysis techniques, such as statistical parametric mapping, enables the comparison of gene expression patterns across different samples, further enriching the insights gained from spatial transcriptomic studies [10].
In summary, spatial transcriptomics is a transformative approach that not only enhances our understanding of gene expression in a spatially resolved manner but also paves the way for significant breakthroughs in various fields of biomedical research. Its ability to correlate gene expression with tissue architecture and cellular interactions underscores its importance in advancing our knowledge of complex biological systems and disease mechanisms.
2.2 Historical Development and Evolution
Spatial transcriptomics is a transformative technology that integrates high-throughput transcriptomics with spatially resolved imaging to map gene expression patterns within intact tissues. This innovative approach allows researchers to generate high-resolution maps of gene expression while maintaining the spatial context of cells and tissues, thereby providing a more comprehensive understanding of biological processes compared to traditional bulk RNA sequencing methods.
Historically, the development of spatial transcriptomics has evolved significantly since its inception in the early 2010s. Initially, traditional techniques such as immunohistochemistry (IHC) and in situ hybridization (ISH) were employed to study gene expression, but these methods were limited by their low throughput and the inability to analyze multiple genes simultaneously. The advent of high-throughput RNA sequencing (RNA-seq) provided a breakthrough, enabling the unbiased measurement of gene expression at a genome-wide scale. However, this advancement came at the cost of losing spatial information about where those expressions occurred within the tissue.
The integration of spatial information into transcriptomics was made possible through various technological advancements. For instance, methods based on microdissection, in situ sequencing, and single-molecule fluorescence in situ hybridization (FISH) have been developed to allow precise mapping of gene expression patterns at the tissue section level [11]. These methods have enabled the exploration of complex biological phenomena such as tissue architecture, developmental biology, and disease mechanisms [2].
Recent innovations in spatial transcriptomics include highly multiplexed techniques and transcriptomic-wide analyses, achieving single-cell resolution [2]. These advancements have significantly broadened the applications of spatial transcriptomics, facilitating studies in various fields, including cancer research, neurodegenerative diseases, and infectious diseases [2].
Furthermore, the development of computational methods has enhanced the analysis of spatial transcriptomic data. Techniques such as SpatialCorr allow for the identification of gene sets with spatially varying correlation structures, revealing insights into coordinated gene expression within specific tissue regions [12]. Additionally, new frameworks such as GAAEST leverage deep learning to integrate spatial location information and gene expression data, facilitating the recognition of spatial domains and improving the accuracy of spatial transcriptomics [13].
The field continues to evolve with the introduction of methods that aim to increase accessibility and scalability. For example, imaging-free approaches that reconstruct spatial barcode locations through molecular diffusion and dimensionality reduction have been developed, allowing for the analysis of larger tissue samples without the need for specialized imaging equipment [14].
In summary, spatial transcriptomics represents a significant advancement in the mapping of gene expression, combining high-throughput sequencing with spatial data to enhance our understanding of biological systems. Its historical development reflects a continuous effort to overcome the limitations of traditional techniques, leading to the establishment of a powerful tool that is now integral to modern biomedical research. The ongoing evolution of this field promises to further elucidate the complexities of tissue organization and gene regulation in health and disease.
3 Techniques in Spatial Transcriptomics
3.1 In Situ Hybridization Methods
Spatial transcriptomics employs various in situ hybridization methods to map gene expression within tissues while preserving spatial information. This technology integrates high-plex RNA sequencing with spatial data, enabling the analysis of gene expression at the cellular level. The emergence of spatial transcriptomics in the early 2010s has been marked by significant advancements in techniques such as in situ hybridization, in situ sequencing, in situ spatial barcoding, and microdissection-based methods. Each of these techniques offers distinct advantages in terms of mapping resolution and spatial assessments at the single-cell level.
In situ hybridization methods are pivotal in spatial transcriptomics as they allow for the visualization of specific RNA molecules within the context of tissue architecture. These methods typically involve the use of labeled probes that bind to target mRNA transcripts, enabling their detection and localization. The application of in situ hybridization facilitates the identification of gene expression patterns in specific diseased foci, which is particularly beneficial in understanding complex diseases like autoimmune rheumatic diseases and neurological disorders. For instance, in the context of rheumatology, spatial transcriptomics provides insights into immune cell distribution and interactions within affected tissues, such as synovial tissue, kidneys, and salivary glands, thereby enhancing the understanding of disease processes and cellular interactions [1].
Moreover, the recent advancements in spatial transcriptomics, such as the development of reverse-padlock amplicon-encoding fluorescence in situ hybridization (RAEFISH), have improved the ability to achieve whole-genome coverage and single-molecule resolution in intact tissues. This method enables the spatial profiling of a vast number of genes, revealing not only transcript-specific subcellular localization but also the interactions among different cell types. Such detailed mapping is crucial for uncovering the gene programs that underlie cellular interactions and disease mechanisms [15].
Additionally, the integration of structured illumination microscopy (SIM) with in situ hybridization methods has shown promise in enhancing the detection performance of gene transcripts. By employing SIM, researchers can increase the detection efficiency of gene transcript spots, which is particularly advantageous when dealing with densely clustered RNA molecules. This improvement in data quality allows for more accurate mapping of gene expression patterns within tissues [16].
In summary, spatial transcriptomics utilizes in situ hybridization techniques to provide a comprehensive view of gene expression within the spatial context of tissues. The continuous evolution of these methods, coupled with innovations in imaging technologies, significantly enhances the resolution and accuracy of spatial transcriptomic analyses, facilitating a deeper understanding of cellular functions and interactions in various biological and pathological contexts [17][18].
3.2 High-Throughput Sequencing Approaches
Spatial transcriptomics employs advanced techniques to map gene expression within tissues, integrating spatial information with high-throughput sequencing methods. This innovative approach allows for the analysis of gene expression patterns at a cellular level while preserving the spatial context of the tissue, thereby providing insights into the complex organization of biological systems.
One of the key methodologies in spatial transcriptomics is the use of barcoded solid-phase RNA capture, which enables the profiling of gene expression in mammalian tissue sections. This technique combines traditional histological staining with spatially resolved RNA sequencing data, allowing for the simultaneous analysis of multiple genes across tissue sections. The process typically involves several steps: first, tissue sections are prepared and stained to provide morphological context; then, RNA is captured and sequenced, resulting in high-throughput gene expression data that retains spatial information [19].
The integration of high-plex RNA sequencing with spatial data has been pivotal in enhancing the resolution of gene expression mapping. Spatial transcriptomics can be applied to a variety of tissues and has seen significant advancements in methodologies such as in situ hybridization, in situ sequencing, and spatial barcoding. These techniques facilitate high-resolution assessments of gene expression at the single-cell level, enabling researchers to uncover cellular interactions and tissue architecture in detail [1].
Moreover, recent developments in deep learning algorithms and computational frameworks have further refined the mapping of gene expression. For instance, the introduction of models that leverage graph convolutional networks allows for the identification of spatially structured domains within transcriptomic data. These models can analyze transcriptomic data to find clusters of cells with similar expression patterns and their spatial distribution, enhancing the understanding of tissue organization and function [5].
The high-throughput nature of spatial transcriptomics is also supported by advancements in sequencing technologies, which enable the systematic measurement of gene expression levels across large tissue areas. This allows for the creation of comprehensive spatial maps that can reveal how gene activity orchestrates cellular arrangements in multicellular organisms [20].
In summary, spatial transcriptomics utilizes a combination of high-throughput sequencing techniques and advanced computational methods to map gene expression with spatial precision. This approach not only enhances the understanding of tissue architecture but also provides critical insights into the molecular mechanisms underlying various biological processes and diseases.
3.3 Integration with Imaging Techniques
Spatial transcriptomics is a revolutionary technology that allows for the mapping of gene expression within tissue samples while preserving the spatial context of those expressions. This integration of gene expression data with spatial coordinates enables researchers to obtain high-resolution maps of gene activity in their native tissue architecture, which is essential for understanding complex biological processes.
The core of spatial transcriptomics lies in its ability to combine high-plex RNA sequencing with spatial data. Various methods have been developed to achieve this, including in situ hybridization, in situ sequencing, and microdissection-based techniques, which offer advanced mapping resolution at the single-cell level [1]. These techniques facilitate the analysis of gene expression patterns directly within the tissue, providing insights into cellular interactions and the pathophysiology of diseases.
One significant advancement in spatial transcriptomics is the integration of histological imaging with transcriptomic data. For instance, a method that utilizes deep generative models can combine spatial gene expression data with histological images from the same tissue section, resulting in higher-resolution expression maps [21]. This approach allows for the prediction of spatial gene expression solely from histology images, thus enhancing the overall resolution and accuracy of gene expression mapping.
Moreover, the integration of imaging techniques with spatial transcriptomics also includes the use of AI to extract morphological features that describe the tissue [9]. This framework can either translate morphological data to predict gene expression or integrate morphological features into spatial transcriptomics, thereby enriching the dataset and improving the analysis of tissue architecture.
Furthermore, the application of machine learning models, such as the deep learning model for spatial gene clusters and expression (DeepSpaCE), has shown promise in predicting spatial gene expression patterns from histological images, which can significantly aid in the interpretation of complex tissues, such as those found in breast cancer [22]. This predictive capability enhances the utility of spatial transcriptomics, especially in clinical settings where high costs and specialized expertise may limit the accessibility of traditional methods.
Overall, the integration of spatial transcriptomics with advanced imaging techniques is paving the way for a more comprehensive understanding of gene expression dynamics within tissues, facilitating discoveries in developmental biology, cancer research, and various other fields. This synergy not only enhances the spatial resolution of gene expression studies but also allows for the exploration of tissue organization and cellular interactions at unprecedented levels of detail.
4 Applications of Spatial Transcriptomics
4.1 Cancer Research
Spatial transcriptomics (ST) is a transformative technology that enables the high-resolution mapping of gene expression within the context of intact tissues, providing crucial insights into the spatial organization of cells and their interactions. This approach integrates sequencing and imaging techniques to generate detailed maps of gene expression patterns, thereby allowing researchers to examine the intricate cellular architectures that characterize various biological states, particularly in cancer research.
The primary mechanism of spatial transcriptomics involves the capture of mRNA transcripts directly from tissue sections, followed by sequencing to determine the gene expression profiles associated with specific spatial coordinates. This process generates a spatially resolved transcriptome, which reveals not only the identity of expressed genes but also their precise locations within the tissue architecture. The ability to maintain spatial information while assessing gene expression marks a significant advancement over traditional bulk RNA sequencing methods, which often lose this critical spatial context.
In cancer research, spatial transcriptomics has emerged as an invaluable tool for exploring tumor heterogeneity, the tumor microenvironment, and the dynamics of cellular interactions. For instance, studies have utilized ST to delineate intratumor heterogeneity in various cancer types, including breast and liver cancers, by mapping distinct cellular niches and identifying unique gene expression profiles associated with different tumor regions. Such spatially informed analyses allow researchers to uncover the complex interplay between tumor cells and the surrounding stroma, immune cells, and vasculature, which are crucial for understanding tumor progression and response to therapies.
Recent advancements in spatial transcriptomics technologies have led to the development of highly multiplexed methods that enable transcriptomic-wide analysis at single-cell resolution. These methodologies facilitate the identification of specific cellular subpopulations within tumors and their functional states, thus providing insights into the mechanisms of drug resistance and potential therapeutic targets. For example, in liver cancer, spatial transcriptomics has revealed the role of lipid-associated macrophages in steatosis and the signaling pathways involved in liver regeneration and repair, thereby enhancing the understanding of disease pathogenesis [23].
Moreover, the integration of spatial transcriptomics with other modalities, such as single-cell RNA sequencing and immunohistochemistry, has been shown to enhance the interpretation of complex data sets, allowing for a more comprehensive understanding of tumor biology. This multi-omics approach not only aids in characterizing the tumor microenvironment but also supports the identification of biomarkers for personalized treatment strategies [24].
However, despite its potential, spatial transcriptomics faces several challenges, including technical limitations related to resolution and sequencing depth, as well as the need for high-quality samples and sophisticated data analysis tools. Future directions in this field emphasize the development of robust analytical frameworks and the integration of artificial intelligence and geospatial analytics to further leverage the capabilities of spatial transcriptomics [25].
In summary, spatial transcriptomics represents a significant leap forward in cancer research by enabling the detailed mapping of gene expression in its native spatial context. This technology not only enhances the understanding of tumor heterogeneity and microenvironmental interactions but also holds promise for advancing personalized cancer therapies. As the field continues to evolve, the integration of spatial transcriptomics with emerging technologies will likely yield new insights into cancer biology and treatment.
4.2 Developmental Biology
Spatial transcriptomics is a transformative technology that facilitates the mapping of gene expression within intact tissues while preserving the spatial context of cellular environments. This innovative approach integrates high-throughput transcriptomics with detailed spatial imaging, enabling researchers to produce high-resolution maps of gene expression patterns that reflect the natural organization of tissues.
In developmental biology, spatial transcriptomics plays a pivotal role by providing insights into the spatiotemporal dynamics of gene expression as organisms develop. The technology allows for the precise mapping of gene expression at the tissue section level, which is crucial for understanding the functional diversity driven by spatial heterogeneity in developing tissues. By overcoming the limitations of traditional transcriptomics, which often lacks spatial context, spatial transcriptomics provides a more comprehensive view of developmental processes.
Recent advancements in spatial transcriptomics have led to the development of highly multiplexed methods and single-cell resolution techniques. For instance, techniques such as in situ hybridization, in situ sequencing, and spatial barcoding have been instrumental in enhancing the resolution and accuracy of gene expression mapping. These methodologies allow researchers to analyze gene expression not only in bulk but also at the single-cell level, thereby revealing the interactions and developmental trajectories of individual cells within their native microenvironments[1][2][11].
Moreover, the integration of spatial transcriptomics with computational methods has significantly improved the ability to analyze complex datasets. Algorithms designed to detect spatial-temporal domains can trace developmental paths by identifying regions within tissues where cells share similar functions and developmental stages. This capability has been demonstrated in studies involving mouse embryos, where spatial-temporal domains were identified, revealing significant gene expression differences across various developmental contexts[26][26].
The applications of spatial transcriptomics in developmental biology extend to elucidating the mechanisms of tissue development and the cellular interactions that govern these processes. For example, spatial transcriptomics has been employed to construct comprehensive spatiotemporal cell atlases, as seen in murine cardiac development studies, where the spatial organization of cardiac cellular lineages and their interactions were mapped, providing insights into gene expression dynamics during early heart development[27].
Overall, spatial transcriptomics is a crucial tool for developmental biology, enabling researchers to visualize and understand the intricate relationships between gene expression, cellular architecture, and tissue function throughout the developmental process. Its continued advancement promises to further unravel the complexities of developmental biology and enhance our understanding of various biological systems.
4.3 Neurobiology and Other Fields
Spatial transcriptomics is a transformative technology that facilitates the high-resolution mapping of gene expression within intact tissues, preserving spatial context. This capability allows researchers to create detailed maps of gene expression patterns, which can provide insights into the organization and function of various biological systems.
The mapping process in spatial transcriptomics typically involves several advanced methodologies that integrate high-throughput RNA sequencing with spatially resolved data. Techniques such as microdissection, in situ sequencing, and single-molecule fluorescence in situ hybridization (FISH) are employed to capture gene expression at specific locations within the tissue. For instance, recent advancements have led to the development of highly multiplexed methods that enable transcriptomic-wide analysis at single-cell resolution, allowing researchers to discern not only which genes are expressed but also where within the tissue they are localized [2].
In neurobiology, spatial transcriptomics has emerged as a pivotal tool for understanding complex tissue architectures and gene expression patterns. By elucidating the spatial distribution of gene expression in the brain, researchers can gain insights into neural development, function, and pathology. For example, spatial transcriptomics has been applied to identify specific gene expression profiles associated with neurodegenerative diseases, enhancing our understanding of disease mechanisms and potential therapeutic targets [3].
Beyond neurobiology, spatial transcriptomics is making significant impacts in various fields including cancer research, developmental biology, and the study of infectious diseases. In cancer, it aids in the characterization of tumor microenvironments by mapping the spatial organization of different cell types and their interactions, which is crucial for understanding tumor biology and developing targeted therapies [1]. Additionally, in developmental biology, spatial transcriptomics allows for the investigation of gene expression dynamics during tissue development, providing insights into how spatial patterns of gene expression contribute to cellular differentiation and tissue formation [11].
The applications of spatial transcriptomics extend further into clinical settings, where it has the potential to inform personalized medicine. By analyzing gene expression patterns in specific diseased tissues, researchers can better understand the interactions between various cell populations in conditions such as autoimmune diseases and cancers, paving the way for more tailored therapeutic strategies [1].
In summary, spatial transcriptomics provides a powerful framework for mapping gene expression in a spatially resolved manner, with applications that span neurobiology, cancer research, developmental biology, and beyond. This technology not only enhances our understanding of tissue architecture and function but also holds promise for advancing clinical applications in precision medicine.
5 Challenges and Limitations
5.1 Technical Limitations
Spatial transcriptomics (ST) represents a transformative advancement in the field of gene expression analysis, allowing for the precise mapping of gene expression within the spatial context of tissues. This innovative approach integrates high-plex RNA sequencing with spatial data, enabling researchers to visualize the distribution of RNA molecules and gain insights into cellular organization and interactions in their native environments. However, despite its revolutionary capabilities, spatial transcriptomics faces several technical limitations that impact its effectiveness and applicability.
One of the primary challenges associated with spatial transcriptomics is the inherent technical noise and dropout events that can lead to incomplete data. For instance, single-cell transcriptomics, a component of ST, often suffers from these issues, resulting in a lack of reliable data on gene expression levels at the individual cell level (Lammi & Qu 2024) [28]. This can significantly affect the interpretation of results, particularly in complex tissues where cellular heterogeneity is pronounced.
Additionally, spatial transcriptomics technologies are often limited by their gene detection capacity and measurement accuracy. Imaging-based ST methods, while achieving single-cell spatial resolution, can struggle with the accurate quantification of gene expression profiles. This limitation can result in systematic biases toward the distribution of single-cell RNA sequencing data, which complicates the analysis and interpretation of spatial transcriptomics datasets (Zhao et al. 2025) [29].
Another significant technical limitation arises from the challenges of data integration and interpretation. The complexity of spatial transcriptomics data, which includes high-dimensional information, necessitates sophisticated computational tools for effective analysis. The integration of spatial data with other modalities, such as imaging or single-cell RNA sequencing, remains a challenge, as existing methods may not adequately account for the nuances of spatial context (Si et al. 2025) [30]. This complexity can hinder the development of scalable and universally applicable analysis methods.
Furthermore, the cost and time required for spatial transcriptomics analyses pose additional barriers to its widespread adoption. The advanced technologies and reagents necessary for these analyses can be prohibitively expensive, limiting access for many researchers and laboratories (Miyamoto et al. 2025) [1]. This is compounded by the need for specialized expertise to navigate the intricacies of the methodologies and data interpretation.
In summary, while spatial transcriptomics provides groundbreaking insights into gene expression patterns within tissues, it is not without its challenges. Technical limitations such as noise and dropout events, gene detection capacity, data integration complexities, and high costs continue to pose significant hurdles that researchers must address to fully leverage the potential of this powerful technology. Continued advancements in methodologies and computational tools are essential to overcome these challenges and enhance the reliability and applicability of spatial transcriptomics in biomedical research.
5.2 Data Analysis Challenges
Spatial transcriptomics (ST) represents a transformative approach in the field of genomics, allowing researchers to map gene expression within tissues while retaining the spatial context of the cellular environment. This methodology integrates high-plex RNA sequencing with spatial data, providing a detailed landscape of gene expression at the cellular level, which is crucial for understanding complex biological processes and disease mechanisms.
One of the primary challenges in spatial transcriptomics is the analysis of the resultant data. The high-dimensional nature of ST data poses significant computational challenges. Researchers must develop robust statistical methods to effectively interpret and visualize the spatial gene expression patterns. A critical initial step in this process is identifying genes that exhibit spatially varying expression, which is often complicated by the need to account for the spatial correlation structures inherent in the data. For instance, methods like SpatialCorr have been developed to identify gene sets with spatially varying correlation structures, allowing for a deeper understanding of the coordinated expression of groups of genes within specific tissue regions [12].
Moreover, the integration of spatial transcriptomic data with other modalities, such as histological images, further complicates the analysis. For example, the method SpatialSPM addresses the challenge of comparing spatial gene expression patterns across samples with different shapes and coordinates by reconstructing ST data into multi-dimensional image matrices [10]. This ensures comparability and enhances the analysis of gene expression across diverse biological conditions.
The complexity of the data also necessitates advanced computational techniques. As spatial transcriptomics continues to evolve, various deep learning approaches, such as DMGCN, have been introduced to facilitate domain identification and clustering of spatially structured domains [5]. These methods leverage graph convolutional networks to analyze transcriptomic data effectively, although they require substantial computational resources and expertise.
Additionally, researchers face challenges related to the high dimensionality of ST data, which can lead to the loss of biological insights during dimension reduction processes. Traditional clustering methods may overlook valuable information about gene co-expression patterns and spatial dependencies among cells [6]. Therefore, innovative approaches, such as joint Bayesian methods for estimating gene and spatial cell correlations, are essential for maintaining the integrity of the data during analysis [6].
In summary, while spatial transcriptomics provides unprecedented insights into gene expression patterns within tissues, the analysis of ST data is fraught with challenges. These include managing the high dimensionality of the data, ensuring accurate integration with other data types, and employing sophisticated statistical methods to reveal the biological significance of the findings. Addressing these challenges is critical for the successful application of spatial transcriptomics in research and clinical settings, ultimately enhancing our understanding of complex biological systems and diseases.
5.3 Biological Variability
Spatial transcriptomics (ST) is a transformative technology that maps gene expression patterns within their native spatial contexts in tissues, thereby offering unprecedented insights into the spatial organization of cellular functions. The fundamental principle of ST lies in its ability to retain spatial information while simultaneously capturing gene expression profiles, which traditional bulk or single-cell RNA sequencing methods cannot achieve. This capability is crucial for understanding the intricate cellular interactions and functional heterogeneity within complex biological systems, such as tumors or developing tissues [31].
Despite its potential, spatial transcriptomics faces several challenges and limitations, particularly regarding biological variability. One major challenge is the inherent spatial variability of gene expression across different regions of a tissue, which can complicate data interpretation. For instance, the expression of certain genes may vary significantly not only between different tissues but also within the same tissue type due to local microenvironmental factors [32]. Additionally, traditional methods often focus on identifying spatially variable genes, potentially overlooking the complex interactions among genes that contribute to biological pathways and cellular functions [32].
Another critical limitation is the modeling of cross-slice spatial variation, which is essential for accurately identifying genes with differential spatial expression patterns (DSEPs) across multiple conditions. This complexity arises from the need to disentangle inter-slice heterogeneity and ensure that the analysis can scale to large datasets [33]. Furthermore, while ST can elucidate spatially resolved gene expression, it often struggles with integrating data from different modalities, such as histological images or other omics data, which is necessary for a holistic understanding of tissue architecture [34].
Biological variability also manifests in the form of technical noise and dropout events, which can affect the accuracy and completeness of the data obtained from single-cell transcriptomics [28]. This noise can obscure the underlying biological signals and lead to misinterpretations of gene expression patterns. Moreover, the presence of highly abundant genes can cause transcript bleed into adjacent tissue regions, complicating the spatial resolution of gene expression data [35].
In summary, while spatial transcriptomics offers a powerful framework for mapping gene expression in a spatial context, the challenges posed by biological variability, technical noise, and the complexity of data integration necessitate ongoing methodological advancements and robust analytical frameworks to fully leverage the potential of this technology in biomedical research. Addressing these limitations will enhance our understanding of the intricate relationships between gene expression and tissue architecture, ultimately facilitating the translation of these insights into clinical applications [1].
6 Future Directions
6.1 Technological Advancements
Spatial transcriptomics is a revolutionary technique that enables the high-resolution mapping of gene expression within intact tissues, thereby preserving the spatial context of gene activity. This method integrates high-throughput transcriptomics with advanced imaging techniques to create detailed maps of gene expression patterns at the tissue section level. Recent advancements in spatial transcriptomics have led to the development of highly multiplexed methods and transcriptomic-wide analyses, allowing researchers to achieve single-cell resolution in their studies.
The technological advancements in spatial transcriptomics can be categorized into several key areas:
High-Resolution Mapping: Spatial transcriptomics allows for the creation of high-resolution maps of gene expression patterns, adding an additional layer of information to bulk sequencing data. This capability is particularly impactful in various fields such as tissue architecture, developmental biology, cancer research, and studies of neurodegenerative and infectious diseases (Robles-Remacho et al., 2023) [2].
Integration with Imaging Techniques: Recent developments have introduced imaging-free approaches that reconstruct spatial barcode locations through molecular diffusion and dimensionality reduction. This method enhances the accessibility and throughput of spatial transcriptomics, enabling large-scale studies without the need for specialized imaging equipment (Hu et al., 2025) [14].
Enhanced Analytical Methods: Advances in computational methods have improved the analysis of spatial transcriptomics data. For instance, methods like SpatialCorr enable the identification of gene sets with spatially varying correlation structures, revealing biological insights that were previously obscured (Bernstein et al., 2022) [12]. Additionally, the introduction of deep learning techniques, such as the Graph Attention-based Autoencoder for Spatial Transcriptomics (GAAEST), has significantly improved the recognition of spatial domains, enhancing the understanding of tissue organization (Wang et al., 2024) [13].
Scalability and Accessibility: The development of scalable methods for spatial transcriptomics, such as the joint Bayesian estimation approach, allows for simultaneous analysis of gene dependencies and spatial cell correlations. This scalability is crucial for studying complex tissues and understanding their functional organization (Chakrabarti et al., 2024) [6].
Future Directions: As spatial transcriptomics continues to evolve, future advancements are likely to focus on improving the resolution and accuracy of spatial gene expression mapping. This includes enhancing computational tools for data integration and analysis, which will facilitate the exploration of spatiotemporal dynamics in biological processes. The continued development of spatial transcriptomics technologies is expected to promote widespread adoption within the research community, enabling more comprehensive studies of cellular interactions and disease mechanisms (Deng et al., 2025) [11].
In summary, spatial transcriptomics is at the forefront of technological innovation in the biological sciences, with significant advancements paving the way for enhanced understanding of gene expression patterns in their native spatial contexts. These developments not only improve the precision of gene mapping but also open new avenues for research in various fields, including developmental biology, cancer, and autoimmune diseases.
6.2 Potential for Clinical Applications
Spatial transcriptomics is a revolutionary technology that enables the analysis of gene expression at the cellular level within tissues while retaining spatial location information. This capability is achieved through a combination of high-plex RNA sequencing and spatial data integration, allowing researchers to create detailed maps of gene expression patterns in their native spatial context. The evolution of spatial transcriptomics has been marked by the development of various techniques such as in situ hybridization, in situ sequencing, in situ spatial barcoding, and microdissection-based methods, each contributing to enhanced mapping resolution and precise spatial assessments at the single-cell level[1].
The potential clinical applications of spatial transcriptomics are vast and promising. In the context of autoimmune rheumatic diseases, for instance, this technology offers insights into the complex pathophysiology of conditions such as rheumatoid arthritis, systemic lupus erythematosus, and Sjögren's syndrome. By elucidating the interactions between different cell populations within disease foci—such as synovial tissue, kidneys, and salivary glands—spatial transcriptomics enhances the understanding of cellular interactions and disease processes, paving the way for personalized treatment strategies[1].
Moreover, the application of spatial transcriptomics in human cardiac tissues illustrates its utility in understanding cardiac development, electro-anatomy, immunology, and ischemic heart disease. Studies utilizing this technology can integrate spatial transcriptomics with other omics technologies to provide a more comprehensive view of human health and disease, highlighting its potential for translational research[36].
Looking ahead, the future directions for spatial transcriptomics involve addressing current challenges that limit its widespread adoption. This includes improving spatial resolution, enhancing data analysis methodologies, and developing standardized protocols for different tissue types and conditions. The integration of spatial transcriptomics with advanced computational tools and machine learning techniques, such as the proposed Graph Attention-based Autoencoder for Spatial Transcriptomics (GAAEST), exemplifies the ongoing efforts to refine data interpretation and enhance the identification of spatial domains with similar expression patterns[13].
In summary, spatial transcriptomics not only transforms our understanding of gene expression within its spatial context but also holds significant promise for clinical applications across various fields, including rheumatology and cardiology. The continuous advancements in this technology are expected to facilitate the translation of research findings into clinical practice, ultimately contributing to improved patient outcomes and personalized medicine approaches.
6.3 Integration with Other Omics Technologies
Spatial transcriptomics (ST) represents a significant advancement in the ability to map gene expression while preserving the spatial context within tissues. This technique allows researchers to visualize and analyze gene expression patterns in their native spatial arrangement, thereby providing insights into cellular organization, interactions, and functional states within complex biological systems.
The mapping process involves several key methodologies that facilitate the preservation of spatial information alongside transcriptomic data. Techniques such as in situ hybridization, in situ sequencing, and microdissection-based methods are employed to capture gene expression at high resolution. These approaches allow for the creation of detailed spatial maps that reveal how different cell types are distributed within tissues and how they interact with one another. For instance, the development of highly multiplexed methods has enabled transcriptomic-wide analysis at single-cell resolution, enhancing the understanding of tissue architecture in various contexts, including developmental biology and disease states[2][37].
In terms of future directions, the integration of spatial transcriptomics with other omics technologies is poised to enhance the depth and breadth of biological insights. The combination of spatial transcriptomics with proteomics, metabolomics, and genomic sequencing allows for a more comprehensive understanding of cellular functions and interactions. For example, integrating spatial transcriptomics data with proteomic profiles can elucidate how gene expression translates into protein localization and function within tissues[38][39]. This multimodal approach not only enriches the data but also provides a holistic view of the biological processes at play, thereby facilitating more accurate interpretations of complex biological phenomena.
Moreover, the development of computational tools to analyze and integrate these diverse datasets is crucial. Advanced machine learning and deep learning algorithms are being employed to decipher the spatial heterogeneity of tissues, identify functional regions, and understand cell-cell communication within the spatial transcriptomics framework[1][39]. As these computational methodologies continue to evolve, they will enable researchers to uncover novel biological insights and improve the predictive power of spatial transcriptomics in various research applications.
In summary, spatial transcriptomics not only provides a powerful platform for mapping gene expression in a spatially resolved manner but also holds great potential for integration with other omics technologies. This integration will likely lead to significant advancements in our understanding of biological systems, disease mechanisms, and therapeutic interventions[1][31].
7 Conclusion
Spatial transcriptomics has emerged as a transformative technology in the field of molecular biology, enabling researchers to map gene expression within tissues while preserving spatial context. The major findings highlight the significance of this approach in elucidating the intricate cellular microenvironments and interactions that govern biological processes in health and disease. Current research demonstrates its applications across various domains, including cancer research, developmental biology, and neurobiology, where it has provided valuable insights into cellular heterogeneity and tissue dynamics. However, challenges such as technical limitations, data analysis complexities, and biological variability remain significant hurdles that need to be addressed. Looking ahead, the future of spatial transcriptomics is promising, with ongoing advancements in technology and computational methods that will enhance its resolution and applicability in clinical settings. The integration of spatial transcriptomics with other omics technologies is also expected to deepen our understanding of complex biological systems and pave the way for personalized medicine, ultimately contributing to improved therapeutic strategies and patient outcomes.
References
- [1] Atsuko Tsujii Miyamoto;Hiroshi Shimagami;Atsushi Kumanogoh;Masayuki Nishide. Spatial transcriptomics in autoimmune rheumatic disease: potential clinical applications and perspectives.. Inflammation and regeneration(IF=6.6). 2025. PMID:39980019. DOI: 10.1186/s41232-025-00369-2.
- [2] Agustín Robles-Remacho;Rosario M Sanchez-Martin;Juan J Diaz-Mochon. Spatial Transcriptomics: Emerging Technologies in Tissue Gene Expression Profiling.. Analytical chemistry(IF=6.7). 2023. PMID:37814884. DOI: 10.1021/acs.analchem.3c02029.
- [3] Stavroula Tekkela;Georgios Theocharidis;John A McGrath;Alexandros Onoufriadis. Spatial transcriptomics in human skin research.. Experimental dermatology(IF=3.1). 2023. PMID:37150587. DOI: 10.1111/exd.14827.
- [4] Chuhan Wang;Adam S Chan;Xiaohang Fu;Shila Ghazanfar;Jinman Kim;Ellis Patrick;Jean Y H Yang. Benchmarking the translational potential of spatial gene expression prediction from histology.. Nature communications(IF=15.7). 2025. PMID:39934114. DOI: 10.1038/s41467-025-56618-y.
- [5] Xikeng Liang;Shutong Xiao;Lu Ba;Yuhui Feng;Zhicheng Ma;Fatima Adilova;Jing Qi;Shuilin Jin. Spatial domain identification method based on multi-view graph convolutional network and contrastive learning.. PLoS computational biology(IF=3.6). 2025. PMID:41105750. DOI: 10.1371/journal.pcbi.1013369.
- [6] Arhit Chakrabarti;Yang Ni;Bani K Mallick. Joint Bayesian estimation of cell dependence and gene associations in spatially resolved transcriptomic data.. Scientific reports(IF=3.9). 2024. PMID:38664448. DOI: 10.1038/s41598-024-60002-z.
- [7] Jessica Gillespie;Maciej Pietrzak;Min-Ae Song;Dongjun Chung. A Meta-Review of Spatial Transcriptomics Analysis Software.. Cells(IF=5.2). 2025. PMID:40710313. DOI: 10.3390/cells14141060.
- [8] Lambda Moses;Lior Pachter. Museum of spatial transcriptomics.. Nature methods(IF=32.1). 2022. PMID:35273392. DOI: 10.1038/s41592-022-01409-2.
- [9] Eduard Chelebian;Christophe Avenel;Carolina Wählby. Combining spatial transcriptomics with tissue morphology.. Nature communications(IF=15.7). 2025. PMID:40360467. DOI: 10.1038/s41467-025-58989-8.
- [10] Jungyoon Ohn;Mi-Kyoung Seo;Jeongbin Park;Daeseung Lee;Hongyoon Choi. SpatialSPM: statistical parametric mapping for the comparison of gene expression pattern images in multiple spatial transcriptomic datasets.. Nucleic acids research(IF=13.1). 2024. PMID:38676948. DOI: 10.1093/nar/gkae293.
- [11] Peilei Deng;Jiaruo Huang;Wencan He;Zhiyuan Li;Cun Guo;Guoxin Chen;Xiaoxu Li;Kejun Zhong;Wei Luo;Bo Kong. Opportunities and challenges in the application of spatiotemporal transcriptomics in plant research.. Frontiers in plant science(IF=4.8). 2025. PMID:41169728. DOI: 10.3389/fpls.2025.1684057.
- [12] Matthew N Bernstein;Zijian Ni;Aman Prasad;Jared Brown;Chitrasen Mohanty;Ron Stewart;Michael A Newton;Christina Kendziorski. SpatialCorr identifies gene sets with spatially varying correlation structure.. Cell reports methods(IF=4.5). 2022. PMID:36590683. DOI: 10.1016/j.crmeth.2022.100369.
- [13] Tianqi Wang;Huitong Zhu;Yunlan Zhou;Weihong Ding;Weichao Ding;Liangxiu Han;Xueqin Zhang. Graph attention automatic encoder based on contrastive learning for domain recognition of spatial transcriptomics.. Communications biology(IF=5.1). 2024. PMID:39424696. DOI: 10.1038/s42003-024-07037-0.
- [14] Chenlei Hu;Mehdi Borji;Giovanni J Marrero;Vipin Kumar;Jackson A Weir;Sachin V Kammula;Evan Z Macosko;Fei Chen. Scalable spatial transcriptomics through computational array reconstruction.. Nature biotechnology(IF=41.7). 2025. PMID:40181168. DOI: 10.1038/s41587-025-02612-0.
- [15] Yubao Cheng;Shengyuan Dang;Yuan Zhang;Yanbo Chen;Ruihuan Yu;Miao Liu;Shengyan Jin;Ailin Han;Samuel Katz;Siyuan Wang. Sequencing-free whole-genome spatial transcriptomics at single-molecule resolution.. Cell(IF=42.5). 2025. PMID:41038164. DOI: 10.1016/j.cell.2025.09.006.
- [16] Alejandro Linares;Carlo Brighi;Sergio Espinola;Francesco Bacchi;Álvaro H Crevenna. Structured Illumination Microscopy Improves Spot Detection Performance in Spatial Transcriptomics.. Cells(IF=5.2). 2023. PMID:37174710. DOI: 10.3390/cells12091310.
- [17] Dongshan Ya;Yingmei Zhang;Qi Cui;Yanlin Jiang;Jiaxin Yang;Ning Tian;Wenjing Xiang;Xiaohui Lin;Qinghua Li;Rujia Liao. Application of spatial transcriptome technologies to neurological diseases.. Frontiers in cell and developmental biology(IF=4.3). 2023. PMID:36936681. DOI: 10.3389/fcell.2023.1142923.
- [18] Katherine Benjamin;Aneesha Bhandari;Jessica D Kepple;Rui Qi;Zhouchun Shang;Yanan Xing;Yanru An;Nannan Zhang;Yong Hou;Tanya L Crockford;Oliver McCallion;Fadi Issa;Joanna Hester;Ulrike Tillmann;Heather A Harrington;Katherine R Bull. Multiscale topology classifies cells in subcellular spatial transcriptomics.. Nature(IF=48.5). 2024. PMID:38898271. DOI: 10.1038/s41586-024-07563-1.
- [19] Fredrik Salmén;Patrik L Ståhl;Annelie Mollbrink;José Fernández Navarro;Sanja Vickovic;Jonas Frisén;Joakim Lundeberg. Barcoded solid-phase RNA capture for Spatial Transcriptomics profiling in mammalian tissue sections.. Nature protocols(IF=16.0). 2018. PMID:30353172. DOI: 10.1038/s41596-018-0045-2.
- [20] Anjali Rao;Dalia Barkley;Gustavo S França;Itai Yanai. Exploring tissue architecture using spatial transcriptomics.. Nature(IF=48.5). 2021. PMID:34381231. DOI: 10.1038/s41586-021-03634-9.
- [21] Ludvig Bergenstråhle;Bryan He;Joseph Bergenstråhle;Xesús Abalo;Reza Mirzazadeh;Kim Thrane;Andrew L Ji;Alma Andersson;Ludvig Larsson;Nathalie Stakenborg;Guy Boeckxstaens;Paul Khavari;James Zou;Joakim Lundeberg;Jonas Maaskola. Super-resolved spatial transcriptomics by deep data fusion.. Nature biotechnology(IF=41.7). 2022. PMID:34845373. DOI: 10.1038/s41587-021-01075-3.
- [22] Taku Monjo;Masaru Koido;Satoi Nagasawa;Yutaka Suzuki;Yoichiro Kamatani. Efficient prediction of a spatial transcriptomics profile better characterizes breast cancer tissue sections without costly experimentation.. Scientific reports(IF=3.9). 2022. PMID:35260632. DOI: 10.1038/s41598-022-07685-4.
- [23] Naoto Fujiwara;Genki Kimura;Hayato Nakagawa. Emerging Roles of Spatial Transcriptomics in Liver Research.. Seminars in liver disease(IF=3.7). 2024. PMID:38574750. DOI: 10.1055/a-2299-7880.
- [24] Yang Jin;Yuanli Zuo;Gang Li;Wenrong Liu;Yitong Pan;Ting Fan;Xin Fu;Xiaojun Yao;Yong Peng. Advances in spatial transcriptomics and its applications in cancer research.. Molecular cancer(IF=33.9). 2024. PMID:38902727. DOI: 10.1186/s12943-024-02040-9.
- [25] Cenk Celik;Shi Pan;Eloise Withnell;Hou Wang Lam;Maria Secrier. Decrypting cancer's spatial code: from single cells to tissue niches.. Molecular oncology(IF=4.5). 2025. PMID:40711978. DOI: 10.1002/1878-0261.70100.
- [26] Zhangdi Song;Changyu Zheng;Jiaxing Chen. stTrace: detecting spatial-temporal domains from spatial transcriptome to trace developmental path.. Briefings in bioinformatics(IF=7.7). 2025. PMID:41241818. DOI: 10.1093/bib/bbaf606.
- [27] Jingmin Kang;Qingsong Li;Jie Liu;Lin Du;Peng Liu;Fuyan Liu;Yue Wang;Xunan Shen;Xujiao Luo;Ninghe Wang;Renhua Wu;Lei Song;Jizheng Wang;Xin Liu. Exploring the cellular and molecular basis of murine cardiac development through spatiotemporal transcriptome sequencing.. GigaScience(IF=3.9). 2025. PMID:39960664. DOI: 10.1093/gigascience/giaf012.
- [28] Mikko J Lammi;Chengjuan Qu. Spatial Transcriptomics, Proteomics, and Epigenomics as Tools in Tissue Engineering and Regenerative Medicine.. Bioengineering (Basel, Switzerland)(IF=3.7). 2024. PMID:39768053. DOI: 10.3390/bioengineering11121235.
- [29] Jinyue Zhao;Jiating Yu;Yuqing Cao;Fan Yuan;Ling-Yun Wu;Duanchen Sun. Enhancing Spatial Transcriptomics via Spatially Constrained Matrix Decomposition with EDGES.. Advanced science (Weinheim, Baden-Wurttemberg, Germany)(IF=14.1). 2025. PMID:40842021. DOI: 10.1002/advs.202508346.
- [30] Yichen Si;Joo Sang Lee;Goo Jun;Hyun Min Kang;Jun Hee Lee. Spatial omics enters the microscopic realm: opportunities and challenges.. Trends in genetics : TIG(IF=16.3). 2025. PMID:40461389. DOI: 10.1016/j.tig.2025.05.002.
- [31] Ran Zhou;Gaoxia Yang;Yan Zhang;Yuan Wang. Spatial transcriptomics in development and disease.. Molecular biomedicine(IF=10.1). 2023. PMID:37806992. DOI: 10.1186/s43556-023-00144-0.
- [32] Leqi Tian;Jiashun Xiao;Tianwei Yu. A robust statistical approach for finding informative spatially associated pathways.. Briefings in bioinformatics(IF=7.7). 2024. PMID:39451157. DOI: 10.1093/bib/bbae543.
- [33] Yan Cui;Zhiyuan Yuan. Prioritizing perturbation-responsive gene patterns using interpretable deep learning.. Nature communications(IF=15.7). 2025. PMID:40603296. DOI: 10.1038/s41467-025-61476-9.
- [34] Shuang Ge;Shuqing Sun;Huan Xu;Qiang Cheng;Zhixiang Ren. Deep learning in single-cell and spatial transcriptomics data analysis: advances and challenges from a data science perspective.. Briefings in bioinformatics(IF=7.7). 2025. PMID:40185158. DOI: 10.1093/bib/bbaf136.
- [35] Nick Howell;Zoe Weiss;Lori L Bonnycastle;Caleb M Grenko;Davide Randazzo;Christopher H Dampier;Neelam Sinha;Narisu Narisu;Amy J Swift;Michael R Erdos;Leslie G Biesecker;Francis S Collins;Catherine C Robertson;D Leland Taylor. A spatial transcriptomics dataset of pancreas sections in normal glucose tolerance and type 2 diabetic donors.. Scientific data(IF=6.9). 2025. PMID:40890166. DOI: 10.1038/s41597-025-05450-6.
- [36] Quynh Nguyen;Lin Wei Tung;Bruce Lin;Raam Sivakumar;Funda Sar;Gurpreet Singhera;Ying Wang;Jeremy Parker;Stephane Le Bihan;Amrit Singh;Fabio M V Rossi;Colin Collins;Jamil Bashir;Zachary Laksman. Spatial Transcriptomics in Human Cardiac Tissue.. International journal of molecular sciences(IF=4.9). 2025. PMID:39940764. DOI: 10.3390/ijms26030995.
- [37] Tsai-Ying Chen;Li You;Jose Angelito U Hardillo;Miao-Ping Chien. Spatial Transcriptomic Technologies.. Cells(IF=5.2). 2023. PMID:37626852. DOI: 10.3390/cells12162042.
- [38] Han-Eol Park;Song Hyun Jo;Rosalind H Lee;Christian P Macks;Taeyun Ku;Jihwan Park;Chung Whan Lee;Junho K Hur;Chang Ho Sohn. Spatial Transcriptomics: Technical Aspects of Recent Developments and Their Applications in Neuroscience and Cancer Research.. Advanced science (Weinheim, Baden-Wurttemberg, Germany)(IF=14.1). 2023. PMID:37026425. DOI: 10.1002/advs.202206939.
- [39] Chuanchao Zhang;Lequn Wang;Qianqian Shi. Computational modeling for deciphering tissue microenvironment heterogeneity from spatially resolved transcriptomics.. Computational and structural biotechnology journal(IF=4.1). 2024. PMID:38800634. DOI: 10.1016/j.csbj.2024.05.028.
MaltSci Intelligent Research Services
Search for more papers on MaltSci.com
spatial transcriptomics · gene expression · cellular microenvironment · cancer research · technical challenges
© 2025 MaltSci
