1
|
Fan G, Dai L, Xie T, Li L, Tang L, Han X, Shi Y. Spatial analyses revealed CXCL5 and SLC6A14 as the markers of microvascular invasion in intrahepatic cholangiocarcinoma. Hepatol Commun 2025; 9:e0597. [PMID: 39670859 PMCID: PMC11637745 DOI: 10.1097/hc9.0000000000000597] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2024] [Accepted: 10/09/2024] [Indexed: 12/14/2024] Open
Abstract
BACKGROUND Microvascular invasion (MVI) is a critical prognostic factor in intrahepatic cholangiocarcinoma (ICC), strongly associated with postoperative recurrence. However, the phenotypic features and spatial organization of MVI remain inadequately understood. METHODS We performed a spatial transcriptomic analysis on 29,632 spots from six ICC samples, manually delineating MVI clusters using the cloupe software. Key biomarkers were identified and validated in an independent cohort of 135 ICC patients. Functional and survival analyses were conducted to assess clinical relevance, and cell-cell communication pathways were investigated. RESULTS MVI regions exhibited heightened proliferation, angiogenesis, and epithelial-mesenchymal transition, driven by increased expression of transcription factors SOX10, ZEB1, and SNAI2. CXCL5 and SLC6A14 were identified as potential MVI biomarkers and showed high expression in tumor-invasive areas. Serum CXCL5 demonstrated strong predictive power for vascular invasion (AUC = 0.92) and intrahepatic metastasis (AUC = 0.96). High expression of both CXCL5 and SLC6A14 was associated with the worst survival outcomes. MVI regions were enriched with immunosuppressive MRC1+ macrophages and exhibited elevated immune checkpoint expression, including HAVCR2 and TIGHT, indicative of immune resistance. Cell-cell communication analysis revealed CXCL5-CXCR2 and LGALS9-HAVCR2 as key ligand-receptor pairs contributing to the immunosuppressive microenvironment. CONCLUSIONS This study identifies CXCL5 and SLC6A14 as key biomarkers of MVI, highlighting their roles in tumor proliferation, immune resistance, and poor clinical outcomes. These findings provide valuable insights into the spatial organization of MVI and its contribution to ICC progression, offering potential therapeutic targets.
Collapse
Affiliation(s)
- Guangyu Fan
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing Key Laboratory of Clinical Study on Anticancer Molecular Targeted Drugs, Chaoyang District, Beijing, China
| | - Liyuan Dai
- Department of Clinical Laboratory, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing Key Laboratory of Clinical Study on Anticancer Molecular Targeted Drugs, Chaoyang District, Beijing, China
| | - Tongji Xie
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing Key Laboratory of Clinical Study on Anticancer Molecular Targeted Drugs, Chaoyang District, Beijing, China
| | - Lin Li
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Le Tang
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing Key Laboratory of Clinical Study on Anticancer Molecular Targeted Drugs, Chaoyang District, Beijing, China
| | - Xiaohong Han
- Department of Clinical Pharmacology Research Center, Peking Union Medical College Hospital, State Key Laboratory of Complex Severe and Rare Diseases, NMPA Key Laboratory for Clinical Research and Evaluation of Drug, Beijing Key Laboratory of Clinical PK & PD Investigation for Innovative Drugs, Chinese Academy of Medical Sciences & Peking Union Medical College, Dongcheng District, Beijing, China
| | - Yuankai Shi
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing Key Laboratory of Clinical Study on Anticancer Molecular Targeted Drugs, Chaoyang District, Beijing, China
| |
Collapse
|
2
|
Zhang D, Yu N, Sun X, Li H, Zhang W, Qiao X, Zhang W, Gao R. Deciphering spatial domains from spatially resolved transcriptomics through spatially regularized deep graph networks. BMC Genomics 2024; 25:1160. [PMID: 39614161 DOI: 10.1186/s12864-024-11072-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Accepted: 11/21/2024] [Indexed: 12/01/2024] Open
Abstract
BACKGROUND Recent advancements in spatially resolved transcriptomics (SRT) have opened up unprecedented opportunities to explore gene expression patterns within spatial contexts. Deciphering spatial domains is a critical task in spatial transcriptomic data analysis, aiding in the elucidation of tissue structural heterogeneity and biological functions. However, existing spatial domain detection methods ignore the consistency of expression patterns and spatial arrangements between spots, as well as the severe gene dropout phenomenon present in SRT data, resulting in suboptimal performance in identifying tissue spatial heterogeneity. RESULTS In this paper, we introduce a novel framework, spatially regularized deep graph networks (SR-DGN), which integrates gene expression profiles with spatial information to learn spatially-consistent and informative spot representations. Specifically, SR-DGN employs graph attention networks (GAT) to adaptively aggregate gene expression information from neighboring spots, considering local expression patterns between spots. In addition, the spatial regularization constraint ensures the consistency of neighborhood relationships between physical and embedded spaces in an end-to-end manner. SR-DGN also employs cross-entropy (CE) loss to model gene expression states, effectively mitigating the impact of noisy gene dropouts. CONCLUSIONS Experimental results demonstrate that SR-DGN outperforms state-of-the-art methods in spatial domain identification across SRT data from different sequencing platforms. Moreover, SR-DGN is capable of recovering known microanatomical structures, yielding clearer low-dimensional visualizations and more accurate spatial trajectory inferences.
Collapse
Affiliation(s)
- Daoliang Zhang
- Center of Intelligent Medicine, School of Control Science and Engineering, Shandong University, Jinan, Shandong, 250061, China
| | - Na Yu
- Center of Intelligent Medicine, School of Control Science and Engineering, Shandong University, Jinan, Shandong, 250061, China
| | - Xue Sun
- Center of Intelligent Medicine, School of Control Science and Engineering, Shandong University, Jinan, Shandong, 250061, China
| | - Haoyang Li
- Center of Intelligent Medicine, School of Control Science and Engineering, Shandong University, Jinan, Shandong, 250061, China
| | - Wenjing Zhang
- Center of Intelligent Medicine, School of Control Science and Engineering, Shandong University, Jinan, Shandong, 250061, China
| | - Xu Qiao
- Center of Intelligent Medicine, School of Control Science and Engineering, Shandong University, Jinan, Shandong, 250061, China.
| | - Wei Zhang
- Center of Intelligent Medicine, School of Control Science and Engineering, Shandong University, Jinan, Shandong, 250061, China.
| | - Rui Gao
- Center of Intelligent Medicine, School of Control Science and Engineering, Shandong University, Jinan, Shandong, 250061, China.
| |
Collapse
|
3
|
Pan Y, Fei L, Wang S, Chen H, Jiang C, Li H, Wang C, Yang Y, Zhang Q, Chen Y. Integrated analysis of single-cell, spatial and bulk RNA-sequencing identifies a cell-death signature for predicting the outcomes of head and neck cancer. Front Immunol 2024; 15:1487966. [PMID: 39575251 PMCID: PMC11578999 DOI: 10.3389/fimmu.2024.1487966] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Accepted: 10/16/2024] [Indexed: 11/24/2024] Open
Abstract
Background Cell death plays an essential role in carcinogenesis, but its function in the recurrence and postoperative prognosis of head and neck cancer (HNC), which ranks as the 7th most common malignancy globally, remains unclear. Methods Data from five main subtypes of HNC related single-cell RNA sequencing (scRNA-seq) were recruited to establish a single-cell atlas, and the distribution of cell death models (CDMs) across different tissues as well as cell subtypes were analyzed. Bulk RNA-seq from the Cancer Genome Atlas Program (TCGA) dataset was subjected to a machine learning-based integrative procedure for constructing a consensus cell death-related signature risk score (CDRscore) model and validated by external data. The biofunctions including different expression analysis, immune cell infiltration, genomic mutations, enrichment analysis as well as cellchat analysis were compared between the high- and low- risk score groups categorized by this CDRscore model. Finally, samples from laryngeal squamous cell cancer (LSCC) were conducted by spatial transcriptomics (ST) to further validate the results of CDRscore model. Results T cells from HNC patients manifested the highest levels of cell death while HPV infection attenuates malignant cell death based on single-cell atlas. CDMs are positively correlated with the tumor-cell stemness, immune-related score and T cells are infiltrated. A CDRscore model was established based on the transcription of ten cell death prognostic genes (MRPL10, DDX19A, NDFIP1, PCMT1, HPRT1, SLC2A3, EFNB2, HK1, BTG3 and MAP2K7). It functions as an independent prognostic factor for overall survival in HNC and displays stable and powerful performance validated by GSE41613 and GSE65858 datasets. Patients in high CDRscore manifested worse overall survival, more active of epithelial mesenchymal transition, TGF-β-related pathways and hypoxia, higher transcription of T cell exhausted markers, and stronger TP53 mutation. ST from LSCC showed that spots with high-risk scores were colocalized with TGF-β and the proliferating malignant cells, additionally, the risk scores have a negative correlation with TCR signaling but positive association with LAG3 transcription. Conclusion The CDRscore model could be utilized as a powerful prognostic indicator for HNC.
Collapse
Affiliation(s)
- Yue Pan
- Institute of Immunology, People’s Liberation Army (PLA), Third Military Medical University, Chongqing, China
| | - Lei Fei
- Institute of Immunology, People’s Liberation Army (PLA), Third Military Medical University, Chongqing, China
| | - Shihua Wang
- Department of Pharmacy, The General Hospital of Western Theater Command, Chengdu, Sichuan, China
| | - Hua Chen
- Department of Pharmacy, The General Hospital of Western Theater Command, Chengdu, Sichuan, China
| | - Changqing Jiang
- Department of Pharmacy, The General Hospital of Western Theater Command, Chengdu, Sichuan, China
| | - Hong Li
- Chongqing Renpin Otolaryngology Head and Neck Surgery Hospital, Chongqing, China
| | - Changsong Wang
- Department of Pathology, People’s Liberation Army Joint Logistic Support Force 989 Hospital, Luoyang, Henan, China
| | - Yao Yang
- Department of Pharmacy, The General Hospital of Western Theater Command, Chengdu, Sichuan, China
| | - Qinggao Zhang
- Chronic Disease Research Center, Medical College, Dalian University, Dalian, Liaoning, China
| | - Yongwen Chen
- Institute of Immunology, People’s Liberation Army (PLA), Third Military Medical University, Chongqing, China
| |
Collapse
|
4
|
Zhou Y, He W, Hou W, Zhu Y. Pianno: a probabilistic framework automating semantic annotation for spatial transcriptomics. Nat Commun 2024; 15:2848. [PMID: 38565531 PMCID: PMC11271244 DOI: 10.1038/s41467-024-47152-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Accepted: 03/20/2024] [Indexed: 04/04/2024] Open
Abstract
Spatial transcriptomics has revolutionized the study of gene expression within tissues, while preserving spatial context. However, annotating spatial spots' biological identity remains a challenge. To tackle this, we introduce Pianno, a Bayesian framework automating structural semantics annotation based on marker genes. Comprehensive evaluations underscore Pianno's remarkable prowess in precisely annotating a wide array of spatial semantics, ranging from diverse anatomical structures to intricate tumor microenvironments, as well as in estimating cell type distributions, across data generated from various spatial transcriptomics platforms. Furthermore, Pianno, in conjunction with clustering approaches, uncovers a region- and species-specific excitatory neuron subtype in the deep layer 3 of the human neocortex, shedding light on cellular evolution in the human neocortex. Overall, Pianno equips researchers with a robust and efficient tool for annotating diverse biological structures, offering new perspectives on spatial transcriptomics data.
Collapse
Affiliation(s)
- Yuqiu Zhou
- State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Institutes of Brain Science and Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China
| | - Wei He
- State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Institutes of Brain Science and Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China
| | - Weizhen Hou
- State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Institutes of Brain Science and Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China
| | - Ying Zhu
- State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Institutes of Brain Science and Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China.
| |
Collapse
|
5
|
Ren L, Huang D, Liu H, Ning L, Cai P, Yu X, Zhang Y, Luo N, Lin H, Su J, Zhang Y. Applications of single‑cell omics and spatial transcriptomics technologies in gastric cancer (Review). Oncol Lett 2024; 27:152. [PMID: 38406595 PMCID: PMC10885005 DOI: 10.3892/ol.2024.14285] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Accepted: 01/19/2024] [Indexed: 02/27/2024] Open
Abstract
Gastric cancer (GC) is a prominent contributor to global cancer-related mortalities, and a deeper understanding of its molecular characteristics and tumor heterogeneity is required. Single-cell omics and spatial transcriptomics (ST) technologies have revolutionized cancer research by enabling the exploration of cellular heterogeneity and molecular landscapes at the single-cell level. In the present review, an overview of the advancements in single-cell omics and ST technologies and their applications in GC research is provided. Firstly, multiple single-cell omics and ST methods are discussed, highlighting their ability to offer unique insights into gene expression, genetic alterations, epigenomic modifications, protein expression patterns and cellular location in tissues. Furthermore, a summary is provided of key findings from previous research on single-cell omics and ST methods used in GC, which have provided valuable insights into genetic alterations, tumor diagnosis and prognosis, tumor microenvironment analysis, and treatment response. In summary, the application of single-cell omics and ST technologies has revealed the levels of cellular heterogeneity and the molecular characteristics of GC, and holds promise for improving diagnostics, personalized treatments and patient outcomes in GC.
Collapse
Affiliation(s)
- Liping Ren
- School of Healthcare Technology, Chengdu Neusoft University, Chengdu, Sichuan 611844, P.R. China
| | - Danni Huang
- Department of Radiology, Central South University Xiangya School of Medicine Affiliated Haikou People's Hospital, Haikou, Hainan 570208, P.R. China
| | - Hongjiang Liu
- School of Computer Science and Technology, Aba Teachers College, Aba, Sichuan 624099, P.R. China
| | - Lin Ning
- School of Healthcare Technology, Chengdu Neusoft University, Chengdu, Sichuan 611844, P.R. China
| | - Peiling Cai
- School of Basic Medical Sciences, Chengdu University, Chengdu, Sichuan 610106, P.R. China
| | - Xiaolong Yu
- Hainan Yazhou Bay Seed Laboratory, Sanya Nanfan Research Institute, Material Science and Engineering Institute of Hainan University, Sanya, Hainan 572025, P.R. China
| | - Yang Zhang
- Innovative Institute of Chinese Medicine and Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 611137, P.R. China
| | - Nanchao Luo
- School of Computer Science and Technology, Aba Teachers College, Aba, Sichuan 624099, P.R. China
| | - Hao Lin
- Center for Informational Biology, University of Electronic Science and Technology of China, Chengdu, Sichuan 611731, P.R. China
| | - Jinsong Su
- Research Institute of Integrated Traditional Chinese Medicine and Western Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 611137, P.R. China
| | - Yinghui Zhang
- School of Healthcare Technology, Chengdu Neusoft University, Chengdu, Sichuan 611844, P.R. China
| |
Collapse
|
6
|
Ma X, Zhao Q. Application of artificial intelligence in oncology. Semin Cancer Biol 2023; 97:68-69. [PMID: 37977345 DOI: 10.1016/j.semcancer.2023.11.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2023]
Affiliation(s)
- Xuelei Ma
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China.
| | - Qi Zhao
- Institute of Translational Medicine, Cancer Centre, Faculty of Health Sciences, University of Macau, Taipa, Macau Special Administrative region of China; MoE Frontiers Science Center for Precision Oncology, University of Macau, Taipa, Macau Special Administrative region of China.
| |
Collapse
|
7
|
Schneider F, Kaczorowski A, Jurcic C, Kirchner M, Schwab C, Schütz V, Görtz M, Zschäbitz S, Jäger D, Stenzinger A, Hohenfellner M, Duensing S, Duensing A. Digital Spatial Profiling Identifies the Tumor Periphery as a Highly Active Biological Niche in Clear Cell Renal Cell Carcinoma. Cancers (Basel) 2023; 15:5050. [PMID: 37894418 PMCID: PMC10605891 DOI: 10.3390/cancers15205050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Revised: 10/03/2023] [Accepted: 10/11/2023] [Indexed: 10/29/2023] Open
Abstract
Clear cell renal cell carcinoma (ccRCC) is characterized by a high degree of intratumoral heterogeneity (ITH). Besides genomic ITH, there is considerable functional ITH, which encompasses spatial niches with distinct proliferative and signaling activities. The full extent of functional spatial heterogeneity in ccRCC is incompletely understood. In the present study, a total of 17 ccRCC tissue specimens from different sites (primary tumor, n = 11; local recurrence, n = 1; distant metastasis, n = 5) were analyzed using digital spatial profiling (DSP) of protein expression. A total of 128 regions of interest from the tumor periphery and tumor center were analyzed for the expression of 46 proteins, comprising three major signaling pathways as well as immune cell markers. Results were correlated to clinico-pathological variables. The differential expression of granzyme B was validated using conventional immunohistochemistry and was correlated to the cancer-specific patient survival. We found that a total of 37 proteins were differentially expressed between the tumor periphery and tumor center. Thirty-five of the proteins were upregulated in the tumor periphery compared to the center. These included proteins involved in cell proliferation, MAPK and PI3K/AKT signaling, apoptosis regulation, epithelial-to-mesenchymal transition, as well as immune cell markers. Among the most significantly upregulated proteins in the tumor periphery was granzyme B. Granzyme B upregulation in the tumor periphery correlated with a significantly reduced cancer-specific patient survival. In conclusion, this study highlights the unique cellular contexture of the tumor periphery in ccRCC. The correlation between granzyme B upregulation in the tumor periphery and patient survival suggests local selection pressure for aggressive tumor growth and disease progression. Our results underscore the potential of spatial biology for biomarker discovery in ccRCC and cancer in general.
Collapse
Affiliation(s)
- Felix Schneider
- Molecular Urooncology, Department of Urology, University Hospital Heidelberg, Im Neuenheimer Feld 517, D-69120 Heidelberg, Germany
| | - Adam Kaczorowski
- Molecular Urooncology, Department of Urology, University Hospital Heidelberg, Im Neuenheimer Feld 517, D-69120 Heidelberg, Germany
| | - Christina Jurcic
- Molecular Urooncology, Department of Urology, University Hospital Heidelberg, Im Neuenheimer Feld 517, D-69120 Heidelberg, Germany
| | - Martina Kirchner
- Institute of Pathology, University Hospital Heidelberg, Im Neuenheimer Feld 224, D-69120 Heidelberg, Germany
| | - Constantin Schwab
- Institute of Pathology, University Hospital Heidelberg, Im Neuenheimer Feld 224, D-69120 Heidelberg, Germany
| | - Viktoria Schütz
- Department of Urology, University Hospital Heidelberg, and National Center for Tumor Diseases (NCT), Im Neuenheimer Feld 420, D-69120 Heidelberg, Germany
| | - Magdalena Görtz
- Department of Urology, University Hospital Heidelberg, and National Center for Tumor Diseases (NCT), Im Neuenheimer Feld 420, D-69120 Heidelberg, Germany
| | - Stefanie Zschäbitz
- Department of Medical Oncology, University Hospital Heidelberg, and National Center for Tumor Diseases (NCT), Im Neuenheimer Feld 460, D-69120 Heidelberg, Germany
| | - Dirk Jäger
- Department of Medical Oncology, University Hospital Heidelberg, and National Center for Tumor Diseases (NCT), Im Neuenheimer Feld 460, D-69120 Heidelberg, Germany
| | - Albrecht Stenzinger
- Institute of Pathology, University Hospital Heidelberg, Im Neuenheimer Feld 224, D-69120 Heidelberg, Germany
| | - Markus Hohenfellner
- Department of Urology, University Hospital Heidelberg, and National Center for Tumor Diseases (NCT), Im Neuenheimer Feld 420, D-69120 Heidelberg, Germany
| | - Stefan Duensing
- Molecular Urooncology, Department of Urology, University Hospital Heidelberg, Im Neuenheimer Feld 517, D-69120 Heidelberg, Germany
- Department of Urology, University Hospital Heidelberg, and National Center for Tumor Diseases (NCT), Im Neuenheimer Feld 420, D-69120 Heidelberg, Germany
| | - Anette Duensing
- Department of Urology, University Hospital Heidelberg, and National Center for Tumor Diseases (NCT), Im Neuenheimer Feld 420, D-69120 Heidelberg, Germany
- Cancer Therapeutics Program, UPMC Hillman Cancer Center, 5117 Centre Avenue, Pittsburgh, PA 15213, USA
- Department of Pathology, University of Pittsburgh School of Medicine, 200 Lothrop Street, Pittsburgh, PA 15213, USA
- Precision Oncology of Urological Malignancies, Department of Urology, University Hospital Heidelberg, Im Neuenheimer Feld 517, D-69120 Heidelberg, Germany
| |
Collapse
|