Sign in to use this feature.

Years

Between: -

Subjects

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Journals

Article Types

Countries / Regions

Search Results (19)

Search Parameters:
Keywords = visium

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
19 pages, 3009 KiB  
Article
PD-1-Positive CD8+ T Cells and PD-1-Positive FoxP3+ Cells in Tumor Microenvironment Predict Response to Neoadjuvant Chemoimmunotherapy in Gastric Cancer Patients
by Liubov A. Tashireva, Anna Yu. Kalinchuk, Elena O. Shmakova, Elisaveta A. Tsarenkova, Dmitriy M. Loos, Pavel Iamschikov, Ivan A. Patskan, Alexandra V. Avgustinovich, Sergey V. Vtorushin, Irina V. Larionova and Evgeniya S. Grigorieva
Cancers 2025, 17(14), 2407; https://doi.org/10.3390/cancers17142407 - 21 Jul 2025
Viewed by 353
Abstract
Background/Objectives: In gastric cancer, only a subset of patients benefit clinically from neoadjuvant chemoimmunotherapy, underscoring the need for robust biomarkers that can predict treatment responses and guide personalized immunotherapy. This study aimed to characterize the immune microenvironment of gastric tumors and identify predictive [...] Read more.
Background/Objectives: In gastric cancer, only a subset of patients benefit clinically from neoadjuvant chemoimmunotherapy, underscoring the need for robust biomarkers that can predict treatment responses and guide personalized immunotherapy. This study aimed to characterize the immune microenvironment of gastric tumors and identify predictive markers associated with therapeutic efficacy. Methods: We prospectively enrolled 16 patients with histologically confirmed, PD-L1–positive (CPS ≥ 1) gastric adenocarcinoma (T2–4N0–1M0). All patients received eight cycles of FLOT chemotherapy combined with pembrolizumab. Treatment response was assessed by Mandard tumor regression grading. Spatial transcriptomic profiling (10x Genomics Visium) and multiplex immunofluorescence were used to evaluate tumor-infiltrating immune cell subsets and PD-1 expression at baseline and after treatment. Results: Transcriptomic analysis differentiated the immune landscapes of responders from non-responders. Responders exhibited elevated expression of IL1B, CXCL5, HMGB1, and IFNGR2, indicative of an inflamed tumor microenvironment and type I/II interferon signaling. In contrast, non-responders demonstrated upregulation of immunosuppressive genes such as LGALS3, IDO1, and CD55, along with enrichment in oxidative phosphorylation and antigen presentation pathways. Multiplex immunofluorescence confirmed a higher density of FoxP3+ regulatory T cells in non-responders (median 5.36% vs. 2.41%; p = 0.0032). Notably, PD-1+ CD8+ T cell and PD-1+ FoxP3+ Treg frequencies were significantly elevated in non-responders, suggesting that PD-1 expression within cytotoxic and regulatory compartments may contribute to immune evasion. No substantial differences were observed in PD-L1 CPS or PD-1+ B cells and PD-1+ macrophages. Conclusions: Our findings identify PD-1+ CD8+ T cells and PD-1+ FoxP3+ Tregs as potential biomarkers of resistance to neoadjuvant chemoimmunotherapy in gastric cancer. Transcriptional programs centered on IL1B/CXCL5 and LGALS3/IDO1 define distinct immune phenotypes that may guide future combination strategies targeting both effector and suppressive arms of the tumor immune response. Full article
Show Figures

Figure 1

30 pages, 1689 KiB  
Review
Applications of Spatial Transcriptomics in Veterinary Medicine: A Scoping Review of Research, Diagnostics, and Treatment Strategies
by Rachael M. Weiderman, Mahamudul Hasan and Laura C. Miller
Int. J. Mol. Sci. 2025, 26(13), 6163; https://doi.org/10.3390/ijms26136163 - 26 Jun 2025
Viewed by 839
Abstract
Spatial transcriptomics is an emerging technology that maps gene expression within tissue architecture. Its expanding use in medicine and veterinary science supports research, precision diagnostics, biomarker discovery, and development of targeted treatment strategies. While spatial transcriptomics applications in human health are well-documented with [...] Read more.
Spatial transcriptomics is an emerging technology that maps gene expression within tissue architecture. Its expanding use in medicine and veterinary science supports research, precision diagnostics, biomarker discovery, and development of targeted treatment strategies. While spatial transcriptomics applications in human health are well-documented with significant publication diversity and volume, published applications in veterinary medicine remain limited. A comprehensive search of PubMed was conducted, focusing on studies published from 2016 to early 2025 that employed spatial transcriptomics in the context of disease research, diagnosis, or treatment in human or animal health. The review followed the Arksey and O’Malley framework and adhered to Preferred Reporting Items for Systematic Reviews and Meta-Analyses Extension for Scoping Reviews (PRISMA-ScR) guidelines. A total of 1398 studies met the inclusion criteria. The studies highlighted emerging trends of comparative research with animal model use for human health research. Commonly used spatial transcriptomics platforms included 10× Visium, Slide-seq, Nanostring (GeoMx, CosMX), and multiplexed error-robust fluorescence in situ hybridization (MERFISH). Key gaps in publications include limited veterinary representation, interspecies comparisons, standardized methods, public data use, and therapeutic studies, alongside biases in disease, species, organ, and geography. This review presents the current landscape of spatial transcriptomics publications for human and animal research and medicine, providing comprehensive data and highlighting underrepresented research areas and gaps for future consideration. Full article
Show Figures

Figure 1

16 pages, 3056 KiB  
Article
Muscle Spatial Transcriptomic Reveals Heterogeneous Profiles in Juvenile Dermatomyositis and Persistence of Abnormal Signature After Remission
by Margot Tragin, Séverine A. Degrelle, Baptiste Periou, Brigitte Bader-Meunier, Christine Barnerias, Christine Bodemer, Isabelle Desguerre, Mathieu Paul Rodero, François Jérôme Authier and Cyril Gitiaux
Cells 2025, 14(12), 939; https://doi.org/10.3390/cells14120939 - 19 Jun 2025
Viewed by 571
Abstract
This study aimed to investigate the spatial heterogeneity of molecular signature in the muscle of juvenile dermatomyositis (JDM) patients before and after treatment. Unsupervised reference-free deconvolution of spatial transcriptomics and standardized morphometry were performed in two JDM muscle biopsies with different clinical severity [...] Read more.
This study aimed to investigate the spatial heterogeneity of molecular signature in the muscle of juvenile dermatomyositis (JDM) patients before and after treatment. Unsupervised reference-free deconvolution of spatial transcriptomics and standardized morphometry were performed in two JDM muscle biopsies with different clinical severity at disease onset and compared to healthy muscle. Identified signatures were scored in two additional JDM muscle biopsies from the same patient before and after remission. Disappearance of the normal muscle signature mostly corresponding to mitochondrial biology was observed in JDM. Three pathological transcriptomic signatures were isolated, related to “myofibrillar stress”, “muscle remodeling” and “interferon signaling” signatures. The “myofibrillar stress signature” was prominent in the most severe biopsy while the “muscle remodeling” signature was mostly present in the biopsy from the patient with good outcome. These signatures unveiled genes not previously associated with JDM including ANKRD1 and FSLT1 for “myofibrillar stress” and “muscle remodeling” signatures, respectively. Post-treatment analysis of muscle after two years remission showed a persistence of pathological signatures. This pilot study of JDM muscle identified spatially distributed pathological signatures that persist after remission. This work paves the way for a better understanding of the pathophysiology in affected muscle and the identification of biomarkers that predict relapse. Full article
Show Figures

Figure 1

12 pages, 3391 KiB  
Article
The Influence of Anteroposterior Head Inclination on the Perceived Consonance of the Smile Arc and Lower Lip Curvature on Photographs: A Cross-Sectional Study
by Amir Reza Khadem, Matteo Togninalli, Gregory S. Antonarakis and Cristina Vento
J. Clin. Med. 2025, 14(5), 1658; https://doi.org/10.3390/jcm14051658 - 28 Feb 2025
Viewed by 645
Abstract
Objectives: To determine the extent to which anteroposterior head inclination influences smile arc curvature assessment on frontal photographs. Materials and Methods: Sixty-three young adults participated in this study. Each had five standardized frontal-view photographs captured with posed smiles at five anteroposterior head inclinations [...] Read more.
Objectives: To determine the extent to which anteroposterior head inclination influences smile arc curvature assessment on frontal photographs. Materials and Methods: Sixty-three young adults participated in this study. Each had five standardized frontal-view photographs captured with posed smiles at five anteroposterior head inclinations (−20°, −10°, 0°, +10°, +20°) using a cervical range of motion device. Two curves were traced per photograph: one following the shape of the lower lip and the other the incisal edge of the maxillary anterior teeth from canine to canine (smile line). These curvatures were approximated by quadratic function and compared for concordance based on the maximum curvature of the obtained functions. A score was calculated, with 0 denoting a consonant smile (perfect concordance) and 2 a non-consonant smile. Results: Among the sixty-three participants, fifty-nine were included in the analysis after excluding those with insufficient tooth exposure in the photographs for the smile line assessment. The analysis revealed that the perceived smile line was more consonant (concordant with lower lip curvature) with a −20° head anteroposterior inclination (score: 0.146), and the least consonant with +20° anteroposterior inclination (score: 1.326), with statistically significant differences (p < 0.05). Conclusions: The smile arc curvature assessment on frontal photographs may be influenced by the anteroposterior inclination of the head on frontal photographs. However, due to the two-dimensional nature of this study, further investigations incorporating three-dimensional imaging are recommended. Full article
(This article belongs to the Special Issue Orthodontics: Current Advances and Future Options)
Show Figures

Figure 1

13 pages, 11901 KiB  
Article
Spatial Gene Expression of Human Coronary Arteries Revealed the Molecular Features of Diffuse Intimal Thickening in Explanted Hearts
by Boaz Li, Samuel Leung, Maria Elishaev, Wan Hei Cheng, Giuseppe Mocci, Johan L. M. Björkegren, Chi Lai, Amrit Singh and Ying Wang
Int. J. Mol. Sci. 2025, 26(5), 1949; https://doi.org/10.3390/ijms26051949 - 24 Feb 2025
Cited by 1 | Viewed by 2295
Abstract
Diffuse intimal thickening (DIT) is a pre-clinical stage of atherosclerosis characterized by thickened intima. The molecular basis of its susceptibility to atherogenesis is unknown, and mechanistic investigations cannot be performed in commonly used mouse models, in which DIT does not exist. Vascular smooth [...] Read more.
Diffuse intimal thickening (DIT) is a pre-clinical stage of atherosclerosis characterized by thickened intima. The molecular basis of its susceptibility to atherogenesis is unknown, and mechanistic investigations cannot be performed in commonly used mouse models, in which DIT does not exist. Vascular smooth muscle cells (SMCs) are the predominant cell type that occupies the intima and media of DIT. The molecular differences between these two layers may reveal the earliest phenotypic changes in SMCs to promote atherosclerosis. We benchmarked the RNA quality of human coronary arteries from autopsies (n = 7) and explanted hearts (n = 7) and performed Visium spatial gene expression on tissue sections with DIT. Although autopsy samples met the RNA quality standard for Visium (DV200 ≥ 30%), only arteries from explanted hearts exhibited reliable sequencing performance. Genes enriched in TGF-β-mediated remodeling of the extracellular matrix were overrepresented in the intima. SMCs enriched in the intima are dedifferentiated, but unlike those in the atherosclerotic lesions, they are not pro-inflammatory. Our findings indicate that autopsy samples are not ideal to distinguish subtle differences among cell phenotypes. SMCs in thickened intima may lead to lipid retention but not necessarily the onset of atherosclerosis. Full article
(This article belongs to the Special Issue Molecular Mechanism in Cardiovascular Pathology)
Show Figures

Figure 1

18 pages, 558 KiB  
Review
Spatial Transcriptomics in Human Cardiac Tissue
by 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 and Zachary Laksman
Int. J. Mol. Sci. 2025, 26(3), 995; https://doi.org/10.3390/ijms26030995 - 24 Jan 2025
Cited by 1 | Viewed by 3364
Abstract
Spatial transcriptomics has transformed our understanding of gene expression by preserving the spatial context within tissues. This review focuses on the application of spatial transcriptomics in human cardiac tissues, exploring current technologies with a focus on commercially available platforms. We also highlight key [...] Read more.
Spatial transcriptomics has transformed our understanding of gene expression by preserving the spatial context within tissues. This review focuses on the application of spatial transcriptomics in human cardiac tissues, exploring current technologies with a focus on commercially available platforms. We also highlight key studies utilizing spatial transcriptomics to investigate cardiac development, electro-anatomy, immunology, and ischemic heart disease. These studies demonstrate how spatial transcriptomics can be used in conjunction with other omics technologies to provide a more comprehensive picture of human health and disease. Despite its transformative potential, spatial transcriptomics comes with several challenges that limit its widespread adoption and broader application. By addressing these limitations and fostering interdisciplinary collaboration, spatial transcriptomics has the potential to become an essential tool in cardiovascular research. We hope this review serves as a practical guide for researchers interested in adopting spatial transcriptomics, particularly those with limited prior experience, by providing insights into current technologies, applications, and considerations for successful implementation. Full article
(This article belongs to the Special Issue New Insights in Translational Bioinformatics: Second Edition)
Show Figures

Figure 1

30 pages, 3753 KiB  
Article
Spatial Transcriptomic Profiling of Human Saphenous Vein Exposed to Ex Vivo Arterial Haemodynamics—Implications for Coronary Artery Bypass Graft Patency and Vein Graft Disease
by Liam W. McQueen, Shameem S. Ladak, Georgia R. Layton, Marcin Wozniak, Charles Solomon, Zein El-Dean, Gavin J. Murphy and Mustafa Zakkar
Int. J. Mol. Sci. 2024, 25(19), 10368; https://doi.org/10.3390/ijms251910368 - 26 Sep 2024
Viewed by 1880
Abstract
Vein graft disease is the process by which saphenous vein grafts, utilised for revascularisation during coronary artery bypass graft surgery, undergo an inflammation-driven intimal hyperplasia and accelerated atherosclerosis process in subsequent years after implantation. The role of the arterial circulation, particularly the haemodynamic [...] Read more.
Vein graft disease is the process by which saphenous vein grafts, utilised for revascularisation during coronary artery bypass graft surgery, undergo an inflammation-driven intimal hyperplasia and accelerated atherosclerosis process in subsequent years after implantation. The role of the arterial circulation, particularly the haemodynamic properties’ impact on graft patency, have been investigated but have not to date been explored in depth at the transcriptomic level. We have undertaken the first-in-man spatial transcriptomic analysis of the long saphenous vein in response to ex vivo acute arterial haemodynamic stimulation, utilising a combination of a custom 3D-printed perfusion bioreactor and the 10X Genomics Visium Spatial Gene Expression technology. We identify a total of 413 significant genes (372 upregulated and 41 downregulated) differentially expressed in response to arterial haemodynamic conditions. These genes were associated with pathways including NFkB, TNF, MAPK, and PI3K/Akt, among others. These are established pathways involved in the initiation of an early pro-inflammatory response, leukocyte activation and adhesion signalling, tissue remodelling, and cellular differentiation. Utilising unsupervised clustering analysis, we have been able to classify subsets of the expression based on cell type and with spatial resolution. These findings allow for further characterisation of the early saphenous vein graft transcriptional landscape during the earliest stage of implantation that contributes to vein graft disease, in particular validation of pathways and druggable targets that could contribute towards the therapeutic inhibition of processes underpinning vein graft disease. Full article
(This article belongs to the Section Molecular Pathology, Diagnostics, and Therapeutics)
Show Figures

Figure 1

26 pages, 2978 KiB  
Review
Scoping Review: Methods and Applications of Spatial Transcriptomics in Tumor Research
by Kacper Maciejewski and Patrycja Czerwinska
Cancers 2024, 16(17), 3100; https://doi.org/10.3390/cancers16173100 - 6 Sep 2024
Cited by 1 | Viewed by 5161
Abstract
Spatial transcriptomics (ST) examines gene expression within its spatial context on tissue, linking morphology and function. Advances in ST resolution and throughput have led to an increase in scientific interest, notably in cancer research. This scoping study reviews the challenges and practical applications [...] Read more.
Spatial transcriptomics (ST) examines gene expression within its spatial context on tissue, linking morphology and function. Advances in ST resolution and throughput have led to an increase in scientific interest, notably in cancer research. This scoping study reviews the challenges and practical applications of ST, summarizing current methods, trends, and data analysis techniques for ST in neoplasm research. We analyzed 41 articles published by the end of 2023 alongside public data repositories. The findings indicate cancer biology is an important focus of ST research, with a rising number of studies each year. Visium (10x Genomics, Pleasanton, CA, USA) is the leading ST platform, and SCTransform from Seurat R library is the preferred method for data normalization and integration. Many studies incorporate additional data types like single-cell sequencing and immunohistochemistry. Common ST applications include discovering the composition and function of tumor tissues in the context of their heterogeneity, characterizing the tumor microenvironment, or identifying interactions between cells, including spatial patterns of expression and co-occurrence. However, nearly half of the studies lacked comprehensive data processing protocols, hindering their reproducibility. By recommending greater transparency in sharing analysis methods and adapting single-cell analysis techniques with caution, this review aims to improve the reproducibility and reliability of future studies in cancer research. Full article
Show Figures

Figure 1

18 pages, 3022 KiB  
Article
Spatial Transcriptomics Identifies Cellular and Molecular Characteristics of Scleroderma Skin Lesions: Pilot Study in Juvenile Scleroderma
by Tianhao Liu, Deren Esencan, Claudia M. Salgado, Chongyue Zhao, Ying-Ju Lai, Theresa Hutchins, Anwesha Sanyal, Wei Chen and Kathryn S. Torok
Int. J. Mol. Sci. 2024, 25(17), 9182; https://doi.org/10.3390/ijms25179182 - 23 Aug 2024
Cited by 2 | Viewed by 3047
Abstract
Juvenile localized and systemic scleroderma are rare autoimmune diseases which cause significant disability and morbidity in children. The mechanisms driving juvenile scleroderma remain unclear, necessitating further cellular and molecular level studies. The Visium CytAssist spatial transcriptomics (ST) platform, which preserves the spatial location [...] Read more.
Juvenile localized and systemic scleroderma are rare autoimmune diseases which cause significant disability and morbidity in children. The mechanisms driving juvenile scleroderma remain unclear, necessitating further cellular and molecular level studies. The Visium CytAssist spatial transcriptomics (ST) platform, which preserves the spatial location of cells and simultaneously sequences the whole transcriptome, was employed to profile the histopathological slides from skin lesions of juvenile scleroderma patients. (1) Spatial domains were identified from ST data and exhibited strong concordance with the pathologist’s annotations of anatomical structures. (2) The integration of paired ST data and single-cell RNA sequencing (scRNA-seq) from the same patients validated the comparable accuracy of the two platforms and facilitated the estimation of cell type composition in ST data. (3) The pathologist-annotated immune infiltrates, such as perivascular immune infiltrates, were clearly delineated by the ST analysis, underscoring the biological relevance of the findings. This is the first study utilizing spatial transcriptomics to investigate skin lesions in juvenile scleroderma patients. The validity of the ST data was corroborated by gene expression analyses and the pathologist’s assessments. Integration with scRNA-seq data facilitated the cell type-level analysis and validation. Analyses of immune infiltrates through combined ST data and pathological review enhances our understanding of the pathogenesis of juvenile scleroderma. Full article
(This article belongs to the Special Issue Targeted Therapy for Immune Diseases)
Show Figures

Figure 1

20 pages, 34930 KiB  
Article
A Spatial Transcriptomics Browser for Discovering Gene Expression Landscapes across Microscopic Tissue Sections
by Maria Schmidt, Susanna Avagyan, Kristin Reiche, Hans Binder and Henry Loeffler-Wirth
Curr. Issues Mol. Biol. 2024, 46(5), 4701-4720; https://doi.org/10.3390/cimb46050284 - 13 May 2024
Cited by 1 | Viewed by 2424
Abstract
A crucial feature of life is its spatial organization and compartmentalization on the molecular, cellular, and tissue levels. Spatial transcriptomics (ST) technology has opened a new chapter of the sequencing revolution, emerging rapidly with transformative effects across biology. This technique produces extensive and [...] Read more.
A crucial feature of life is its spatial organization and compartmentalization on the molecular, cellular, and tissue levels. Spatial transcriptomics (ST) technology has opened a new chapter of the sequencing revolution, emerging rapidly with transformative effects across biology. This technique produces extensive and complex sequencing data, raising the need for computational methods for their comprehensive analysis and interpretation. We developed the ST browser web tool for the interactive discovery of ST images, focusing on different functional aspects such as single gene expression, the expression of functional gene sets, as well as the inspection of the spatial patterns of cell–cell interactions. As a unique feature, our tool applies self-organizing map (SOM) machine learning to the ST data. Our SOM data portrayal method generates individual gene expression landscapes for each spot in the ST image, enabling its downstream analysis with high resolution. The performance of the spatial browser is demonstrated by disentangling the intra-tumoral heterogeneity of melanoma and the microarchitecture of the mouse brain. The integration of machine-learning-based SOM portrayal into an interactive ST analysis environment opens novel perspectives for the comprehensive knowledge mining of the organization and interactions of cellular ecosystems. Full article
(This article belongs to the Section Bioinformatics and Systems Biology)
Show Figures

Figure 1

14 pages, 9810 KiB  
Article
Unveiling the Genomic Landscape of Intraductal Carcinoma of the Prostate Using Spatial Gene Expression Analysis
by Ryuta Watanabe, Noriyoshi Miura, Mie Kurata, Riko Kitazawa, Tadahiko Kikugawa and Takashi Saika
Int. J. Mol. Sci. 2024, 25(9), 4818; https://doi.org/10.3390/ijms25094818 - 28 Apr 2024
Cited by 6 | Viewed by 2225
Abstract
Intraductal carcinoma of the prostate (IDCP) has recently attracted increasing interest owing to its unfavorable prognoses. To effectively identify the IDCP-specific gene expression profile, we took a novel approach of characterizing a typical IDCP case using spatial gene expression analysis. A formalin-fixed, paraffin-embedded [...] Read more.
Intraductal carcinoma of the prostate (IDCP) has recently attracted increasing interest owing to its unfavorable prognoses. To effectively identify the IDCP-specific gene expression profile, we took a novel approach of characterizing a typical IDCP case using spatial gene expression analysis. A formalin-fixed, paraffin-embedded sample was subjected to Visium CytAssist Spatial Gene Expression analysis. IDCP within invasive prostate cancer sites was recognized as a distinct cluster separate from other invasive cancer clusters. Highly expressed genes defining the IDCP cluster, such as MUC6, MYO16, NPY, and KLK12, reflected the aggressive nature of high-grade prostate cancer. IDCP sites also showed increased hypoxia markers HIF1A, BNIP3L, PDK1, and POGLUT1; decreased fibroblast markers COL1A2, DCN, and LUM; and decreased immune cell markers CCR5 and FCGR3A. Overall, these findings indicate that the hypoxic tumor microenvironment and reduced recruitment of fibroblasts and immune cells, which reflect morphological features of IDCP, may influence the aggressiveness of high-grade prostate cancer. Full article
(This article belongs to the Special Issue Molecular Mechanisms Underlying the Progression of Prostate Cancer)
Show Figures

Figure 1

16 pages, 6170 KiB  
Article
Spatial Computational Hepatic Molecular Biomarker Reveals LSEC Role in Midlobular Liver Zonation Fibrosis in DILI and NASH Liver Injury
by Munish Puri
Int. J. Transl. Med. 2024, 4(2), 208-223; https://doi.org/10.3390/ijtm4020012 - 23 Mar 2024
Cited by 1 | Viewed by 3188
Abstract
The liver is structurally organized into zonation, where Liver Sinusoidal Endothelial Cells (LSECs) play a crucial role during chronic liver injury and the early stages of fibrosis. Fibrosis can be reversed if diagnosed early at the molecular level in zonation before progressing to [...] Read more.
The liver is structurally organized into zonation, where Liver Sinusoidal Endothelial Cells (LSECs) play a crucial role during chronic liver injury and the early stages of fibrosis. Fibrosis can be reversed if diagnosed early at the molecular level in zonation before progressing to advanced stages like bridging fibrosis. This study identified zonation marker genes using scRNA-seq and spatial transcriptomics molecular profiling technologies in a normal and diseased fibrotic human liver. DGE analysis was performed over LSECs, and we identified the top 20 expressed genes in the periportal, perivenous, and intermediate acinar zones. Multi-omics and scRNA-seq analysis over Visium images and ECs liver cells showed OIT3, DNASE1L3, CLEC4G, LYVE1, FCN2, and CRHBP as commonly expressed mid-lobular zonation-specific genes. Also, this study detected STAB2, F8, AQP1, TEK, TIMP3, TIE1, and CTSL genes as expressed in DILI and NASH EC populations. The connection between LSEC marker genes in zone 2 and liver fibrosis holds significant promise for advancing our understanding in developing new therapeutic strategies for fibrosis reversal and designing computational molecular biomarkers in NASH and DILI fibrotic liver diseases. Full article
Show Figures

Figure 1

22 pages, 20479 KiB  
Article
Unique Spatial Transcriptomic Profiling of the Murine Femoral Fracture Callus: A Preliminary Report
by Will Jiang, Dennis L. Caruana, Jungho Back and Francis Y. Lee
Cells 2024, 13(6), 522; https://doi.org/10.3390/cells13060522 - 16 Mar 2024
Cited by 7 | Viewed by 2792
Abstract
Fracture callus formation is a dynamic stage of bone activity and repair with precise, spatially localized gene expression. Metastatic breast cancer impairs fracture healing by disrupting bone homeostasis and imparting an altered genomic profile. Previous sequencing techniques such as single-cell RNA and in [...] Read more.
Fracture callus formation is a dynamic stage of bone activity and repair with precise, spatially localized gene expression. Metastatic breast cancer impairs fracture healing by disrupting bone homeostasis and imparting an altered genomic profile. Previous sequencing techniques such as single-cell RNA and in situ hybridization are limited by missing spatial context and low throughput, respectively. We present a preliminary approach using the Visium CytAssist spatial transcriptomics platform to provide the first spatially intact characterization of genetic expression changes within an orthopedic model of impaired fracture healing. Tissue slides prepared from BALB/c mice with or without MDA-MB-231 metastatic breast cancer cells were used. Both unsupervised clustering and histology-based annotations were performed to identify the hard callus, soft callus, and interzone for differential gene expression between the wild-type and pathological fracture model. The spatial transcriptomics platform successfully localized validated genes of the hard (Dmp1, Sost) and soft callus (Acan, Col2a1). The fibrous interzone was identified as a region of extensive genomic heterogeneity. MDA-MB-231 samples demonstrated downregulation of the critical bone matrix and structural regulators that may explain the weakened bone structure of pathological fractures. Spatial transcriptomics may represent a valuable tool in orthopedic research by providing temporal and spatial context. Full article
Show Figures

Figure 1

20 pages, 8243 KiB  
Article
Spatial Transcriptomics Reveals Signatures of Histopathological Changes in Muscular Sarcoidosis
by Hippolyte Lequain, Cyril Dégletagne, Nathalie Streichenberger, Julie Valantin, Thomas Simonet, Laurent Schaeffer, Pascal Sève and Pascal Leblanc
Cells 2023, 12(23), 2747; https://doi.org/10.3390/cells12232747 - 30 Nov 2023
Cited by 5 | Viewed by 3062
Abstract
Sarcoidosis is a multisystemic disease characterized by non-caseating granuloma infiltrating various organs. The form with symptomatic muscular involvement is called muscular sarcoidosis. The impact of immune cells composing the granuloma on the skeletal muscle is misunderstood. Here, we investigated the granuloma–skeletal muscle interactions [...] Read more.
Sarcoidosis is a multisystemic disease characterized by non-caseating granuloma infiltrating various organs. The form with symptomatic muscular involvement is called muscular sarcoidosis. The impact of immune cells composing the granuloma on the skeletal muscle is misunderstood. Here, we investigated the granuloma–skeletal muscle interactions through spatial transcriptomics on two patients affected by muscular sarcoidosis. Five major transcriptomic clusters corresponding to perigranuloma, granuloma, and three successive muscle tissue areas (proximal, intermediate, and distal) around the granuloma were identified. Analyses revealed upregulated pathways in the granuloma corresponding to the activation of T-lymphocytes and monocytes/macrophages cytokines, the upregulation of extracellular matrix signatures, and the induction of the TGF-β signaling in the perigranuloma. A comparison between the proximal and distal muscles to the granuloma revealed an inverse correlation between the distance to the granuloma and the upregulation of cellular response to interferon-γ/α, TNF-α, IL-1,4,6, fibroblast proliferation, epithelial to mesenchymal cell transition, and the downregulation of muscle gene expression. These data shed light on the intercommunications between granulomas and the muscle tissue and provide pathophysiological mechanisms by showing that granuloma immune cells have a direct impact on proximal muscle tissue by promoting its progressive replacement by fibrosis via the expression of pro-inflammatory and profibrosing signatures. These data could possibly explain the evolution towards a state of disability for some patients. Full article
(This article belongs to the Special Issue Advances in Muscle Research in Health and Disease)
Show Figures

Graphical abstract

14 pages, 4256 KiB  
Article
Identification of Niche-Specific Gene Signatures between Malignant Tumor Microenvironments by Integrating Single Cell and Spatial Transcriptomics Data
by Jahanzeb Saqib, Beomsu Park, Yunjung Jin, Junseo Seo, Jaewoo Mo and Junil Kim
Genes 2023, 14(11), 2033; https://doi.org/10.3390/genes14112033 - 31 Oct 2023
Cited by 5 | Viewed by 3857
Abstract
The tumor microenvironment significantly affects the transcriptomic states of tumor cells. Single-cell RNA sequencing (scRNA-seq) helps elucidate the transcriptomes of individual cancer cells and their neighboring cells. However, cell dissociation results in the loss of information on neighboring cells. To address this challenge [...] Read more.
The tumor microenvironment significantly affects the transcriptomic states of tumor cells. Single-cell RNA sequencing (scRNA-seq) helps elucidate the transcriptomes of individual cancer cells and their neighboring cells. However, cell dissociation results in the loss of information on neighboring cells. To address this challenge and comprehensively assess the gene activity in tissue samples, it is imperative to integrate scRNA-seq with spatial transcriptomics. In our previous study on physically interacting cell sequencing (PIC-seq), we demonstrated that gene expression in single cells is affected by neighboring cell information. In the present study, we proposed a strategy to identify niche-specific gene signatures by harmonizing scRNA-seq and spatial transcriptomic data. This approach was applied to the paired or matched scRNA-seq and Visium platform data of five cancer types: breast cancer, gastrointestinal stromal tumor, liver hepatocellular carcinoma, uterine corpus endometrial carcinoma, and ovarian cancer. We observed distinct gene signatures specific to cellular niches and their neighboring counterparts. Intriguingly, these niche-specific genes display considerable dissimilarity to cell type markers and exhibit unique functional attributes independent of the cancer types. Collectively, these results demonstrate the potential of this integrative approach for identifying novel marker genes and their spatial relationships. Full article
(This article belongs to the Special Issue Applications and Progress in Single-Cell Genomics)
Show Figures

Figure 1

Back to TopTop