1
|
Wang W, Jiao Y, Du X, Ye Z. Immune-related glycosylation genes based classification predicts prognosis and therapy options of osteosarcoma. Gene 2025; 933:148985. [PMID: 39369757 DOI: 10.1016/j.gene.2024.148985] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2024] [Revised: 10/02/2024] [Accepted: 10/03/2024] [Indexed: 10/08/2024]
Abstract
Osteosarcoma is the most common primary bone malignancy, with a very poor prognosis. Aberrant glycosylation is close involvement in osteosarcoma. Accordingly, this study aimed at investigating the role of glycosylation genes in the prognosis and therapy options of osteosarcoma. The microenvironment of osteosarcoma was assessed using estimate algorithm. A total of 20 immune-related glycosylation genes (IRGGs) was identified using Pearson correlation analysis. Accordingly, osteosarcoma patients were divided into C1 and C2 type using consensus clustering. Multiple algorithms (Xcell, MCP-counter, ssGSEA, epic, quantiseq), cancer immune cycle analysis, and GSVA were applied to estimate the immune, molecule and metabolism characteristics of osteosarcoma, indicating that C1 type was featured with high immune infiltration, high glycosylation, enriched MEK signaling, and good prognosis, while C2 type was characterized by more metastasis, enriched immunotherapy-positive gene signatures, high tumor mutation burden, and poor prognosis. Results from TIDE algorithm and immunotherapy datasets suggested the C2 type's preference of immune checkpoint inhibitors (ICIs), while data of GDSC, CMap analysis and cell experiments indicated that C1 type was sensitivity to MEK inhibitor PD0325901. In addition, univariate Cox and Lasso analysis was combined to establish an IRGGs' risk score containing 6 genes (B3GNT8, FUT7, GAL3ST4, GALNT14, HS3ST2, and MFNG). The data of DCA and ROC indicated its well prediction of prognosis in osteosarcoma. Finally, cellular location analysis showed that the 6 genes not only distributed in tumor cells but also in immune cells. In summary, the classification and risk score based on IRGGs effectively predicted the prognosis and therapy options of osteosarcoma. Further studies on IRGGs may contribute to the understanding of cancer immunity in osteosarcoma.
Collapse
Affiliation(s)
- Wen Wang
- Zhejiang University, Hangzhou, Zhejiang 310058, China; Department of Orthopedics, Fenghua People's Hospital, 36 Gongyuan Road, Ningbo, Zhejiang 315502, China; Department of Orthopedics, Musculoskeletal Tumor Center, the Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, China
| | - Yunjia Jiao
- Clinical Laboratory, Minhang Hospital, Fudan University, No. 170, Xinsong Road, Shanghai 201199, China
| | - Xiaojing Du
- Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China.
| | - Zhaoming Ye
- Zhejiang University, Hangzhou, Zhejiang 310058, China; Department of Orthopedics, Musculoskeletal Tumor Center, the Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, China.
| |
Collapse
|
2
|
Xiong P, Huang Q, Mao Y, Qian H, Yang Y, Mou Z, Deng X, Wang G, He B, You Z. Identification of an immune-related gene panel for the diagnosis of pulmonary arterial hypertension using bioinformatics and machine learning. Int Immunopharmacol 2025; 144:113694. [PMID: 39616855 DOI: 10.1016/j.intimp.2024.113694] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2024] [Revised: 11/03/2024] [Accepted: 11/20/2024] [Indexed: 12/15/2024]
Abstract
OBJECTIVE This study aimed to screen an immune-related gene (IRG) panel and develop a novel approach for diagnosing pulmonary arterial hypertension (PAH) utilizing bioinformatics and machine learning (ML). METHODS Gene expression profiles were retrieved from the Gene Expression Omnibus (GEO) database to identify differentially expressed immune-related genes (IRG-DEGs). We employed five machine learning algorithms-LASSO, random forest (RF), boosted regression trees (BRT), XGBoost, and support vector machine recursive feature elimination (SVM-RFE) to identify biomarkers derived from IRG-DEGs associated with the diagnosis of PAH, incorporating them into the IRG-DEGs panel. Validation of these biomarker levels in lung tissue was conducted in a hypoxia-induced mouse model of PAH, investigating the correlation between AIMP1, IL-15, GLRX, SOD1, Fulton's index (RVHI), and the ratio of pulmonary artery medial thickness to external diameter (MT%). Subsequently, we developed a nomogram model based on the IRG-DEGs panel in lung tissue for diagnosing PAH. The expression, distribution, and pseudotime analysis of these biomarkers across various immune cell types were assessed using single-cell sequencing datasets. Finally, we evaluated the diagnostic utility of the nomogram model based on the IRG-DEGs panel in peripheral blood mononuclear cells (PBMCs) for diagnosing PAH. RESULTS A total of 36 upregulated and 17 downregulated IRG-DEGs were identified in lung tissue from patients with PAH. AIMP1, IL-15, GLRX, and SOD1 were subsequently selected as novel immune-related biomarkers for PAH through the aforementioned machine learning algorithms and incorporated into the IRG-DEGs panel. Experimental results from mice with PAH validated that the expression levels of AIMP1, IL-15, and GLRX in lung tissue were elevated, while SOD1 expression was significantly reduced. Additionally, GLRX and AIMP1 exhibited positive correlations with Fulton's index (RVHI). The expression levels of GLRX, IL-15, and AIMP1 showed positive correlations with MT%, whereas SOD1 exhibited negative correlations with MT%. Analysis of single-cell sequencing data further revealed that the levels of IRG-DEG panel members gradually increased during the pseudotime trajectory from PBMCs to macrophages, correlating with macrophage activation. The area under the curve (AUC) for diagnosing PAH using a nomogram model based on the IRG-DEGs panel derived from lung tissue samples and PBMCs was ≥0.969 and 0.900, respectively. CONCLUSIONS We developed an IRG-DEGs panel containing AIMP1, IL-15, GLRX, and SOD1, which may facilitate the diagnosis of pulmonary arterial hypertension (PAH). These findings provide novel insights that may enhance diagnostic and therapeutic approaches for PAH.
Collapse
Affiliation(s)
- Pan Xiong
- Department of General Practice, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing 400037, China
| | - Qiuhong Huang
- Department of General Practice, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing 400037, China
| | - Yang Mao
- Department of General Practice, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing 400037, China
| | - Hang Qian
- Institute of Respiratory Diseases, Department of Pulmonary and Critical Care Medicine, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing 400037, China
| | - Yi Yang
- Department of General Practice, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing 400037, China
| | - Ziye Mou
- Department of General Practice, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing 400037, China
| | - Xiaohui Deng
- Department of General Practice, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing 400037, China
| | - Guansong Wang
- Institute of Respiratory Diseases, Department of Pulmonary and Critical Care Medicine, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing 400037, China.
| | - Binfeng He
- Department of General Practice, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing 400037, China.
| | - Zaichun You
- Department of General Practice, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing 400037, China.
| |
Collapse
|
3
|
Liu T, Huang C, Sun L, Chen Z, Ge Y, Ji W, Chen S, Zhao Y, Wang M, Wang D, Zhu W. FAP + gastric cancer mesenchymal stromal cells via paracrining INHBA and remodeling ECM promote tumor progression. Int Immunopharmacol 2025; 144:113697. [PMID: 39615112 DOI: 10.1016/j.intimp.2024.113697] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Revised: 11/07/2024] [Accepted: 11/20/2024] [Indexed: 12/15/2024]
Abstract
Gastric cancer (GC) mesenchymal stromal cells (GCMSCs) are the predominant components of the tumor microenvironment (TME) and play a role in the occurrence, development, and metastasis of tumors. However, GCMSCs exhibit phenotypic and functional heterogeneity. The key population of GCMSCs which are vital to tumor progression remains elusive. The expression of fibroblast activation protein (FAP) in gastric cancer was analyzed and verified using clinical pathology data and single-cell RNA sequencing database of gastric cancer patients. FAP positive GCMSCs (FAP+ GCMSCs) were isolated via flow cytometry and characterized through transcriptomic sequencing. The impact of conditioned medium from FAP+ GCMSCs on gastric cancer cell lines was assessed using Enzyme-linked immunosorbent assay (ELISA) and Western blot analyses. Additionally, immunohistochemistry (IHC) and Masson's trichrome staining were employed to explore the association between FAP+ GCMSCs and extracellular matrix (ECM) deposition in gastric cancer tissues. Our study demonstrates that FAP is predominantly expressed in the mesenchymal stromal cells within the gastric cancer milieu. FAP+ GCMSCs exhibited enhanced proliferation, migration, contraction, and tumor-promoting capabilities compared to their FAP- counterparts. These cells significantly increased proliferation and migration of gastric cancer cells through the paracrine secretion of Inhibin Subunit Beta A (INHBA) and activation of the SMAD2/3 signaling pathway. Moreover, FAP+ GCMSCs also induced collagen deposition in ECM and then up-regulated invasion and stemness of GC cells. Mechanistically, this process was mediated by the interaction of collagen with Integrin Subunit Beta 1 (ITGB1), triggering the phosphorylation of Focal Adhesion Kinase (FAK) and Yes Associated Transcriptional Regulator (YAP). Our findings reveal that FAP+ GCSMCs enhanced the GC progression via releasing cytokine INHBA and remodeling ECM providing a theoretical basis for further exploration of tumor stromal-targeting therapy of gastric cancer.
Collapse
Affiliation(s)
- Ting Liu
- Department of Oncology, Digestive Disease Institute & Cancer Institute of Jiangsu University, Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, 212001, China; School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, 212013, China
| | - Chao Huang
- School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, 212013, China
| | - Li Sun
- Department of Clinical Laboratory, Affiliated Kunshan Hospital of Jiangsu University, Suzhou, Jiangsu Province, 215300, China
| | - Zhihong Chen
- Department of Gastrointestinal Surgery, Affiliated People's Hospital of Jiangsu University, Zhenjiang, Jiangsu, 212001, China
| | - Yan Ge
- Department of Gastrointestinal Surgery, Affiliated People's Hospital of Jiangsu University, Zhenjiang, Jiangsu, 212001, China
| | - Weimeng Ji
- School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, 212013, China
| | - Shihan Chen
- School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, 212013, China
| | - Yuanyuan Zhao
- School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, 212013, China
| | - Mei Wang
- School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, 212013, China
| | - Deqiang Wang
- Department of Oncology, Digestive Disease Institute & Cancer Institute of Jiangsu University, Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, 212001, China.
| | - Wei Zhu
- Department of Oncology, Digestive Disease Institute & Cancer Institute of Jiangsu University, Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, 212001, China; School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, 212013, China.
| |
Collapse
|
4
|
Guangzhao L, Xin W, Miaoqing W, Wenjuan M, Ranyi L, Zhizhong P, Rongxin Z, Gong C. IDO1 inhibitor enhances the effectiveness of PD-1 blockade in microsatellite stable colorectal cancer by promoting macrophage pro-inflammatory phenotype polarization. Cancer Immunol Immunother 2025; 74:71. [PMID: 39751692 PMCID: PMC11699167 DOI: 10.1007/s00262-024-03925-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: 09/13/2024] [Accepted: 12/13/2024] [Indexed: 01/04/2025]
Abstract
Microsatellite stable (MSS) colorectal cancer (CRC) is a subtype of CRC that generally exhibits resistance to immunotherapy, particularly immune checkpoint inhibitors such as PD-1 blockade. This study investigates the effects and underlying mechanisms of combining PD-1 blockade with IDO1 inhibition in MSS CRC. Bioinformatics analyses of TCGA-COAD and TCGA-READ cohorts revealed significantly elevated IDO1 expression in CRC tumors, correlating with tumor mutation burden across TCGA datasets. In vivo experiments demonstrated that the combination of IDO1 inhibition and PD-1 blockade significantly reduced tumor growth and increased immune cell infiltration, particularly pro-inflammatory macrophages and CD8+ T cells. IDO1 knockdown in CRC cell lines impaired tolerance to interferon-γ and increased apoptosis in vitro, which were rescued by the application of kynurenine, the end product of IDO1. IDO1 knockdown in MSS CRC enhanced the effectiveness of PD-1 blockade therapy in vivo. IDO1 knockdown cancer cells promoted pro-inflammatory macrophage polarization and enhanced phagocytic activity in vitro, associated with the upregulation of JAK2-STAT3-IL6 signaling pathway. These findings highlight the role of IDO1 in modulating the tumor immune microenvironment in MSS CRC and suggest that combining PD-1 blockade with IDO1 inhibition could enhance therapeutic efficacy by promoting macrophage pro-inflammatory polarization and infiltration through the JAK2-STAT3-IL6 pathway.
Collapse
Affiliation(s)
- Lv Guangzhao
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong, China
- Department of Colorectal Surgery, Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong, China
| | - Wang Xin
- Department of Radiotherapy, The Affiliated Qingyuan Hospital (Qingyuan People's Hospital), Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Wu Miaoqing
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong, China
- Department of Colorectal Surgery, Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong, China
- Digestive Diseases Center, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, China
| | - Ma Wenjuan
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong, China
- Department of Intensive Care Unit, Sun Yat-Sen University Cancer Center State Key Laboratory of Oncology in South China Guangdong Provincial Clinical Research Center for Cancer Guangzhou, Guangzhou, Guangdong, China
| | - Liu Ranyi
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong, China
| | - Pan Zhizhong
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong, China.
- Department of Colorectal Surgery, Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong, China.
| | - Zhang Rongxin
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong, China.
- Department of Colorectal Surgery, Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong, China.
| | - Chen Gong
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong, China.
- Department of Colorectal Surgery, Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong, China.
| |
Collapse
|
5
|
Jansma A, Yao Y, Wolfe J, Del Debbio L, Beentjes SV, Ponting CP, Khamseh A. High order expression dependencies finely resolve cryptic states and subtypes in single cell data. Mol Syst Biol 2025:10.1038/s44320-024-00074-1. [PMID: 39748128 DOI: 10.1038/s44320-024-00074-1] [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: 01/09/2024] [Revised: 10/24/2024] [Accepted: 10/31/2024] [Indexed: 01/04/2025] Open
Abstract
Single cells are typically typed by clustering into discrete locations in reduced dimensional transcriptome space. Here we introduce Stator, a data-driven method that identifies cell (sub)types and states without relying on cells' local proximity in transcriptome space. Stator labels the same single cell multiply, not just by type and subtype, but also by state such as activation, maturity or cell cycle sub-phase, through deriving higher-order gene expression dependencies from a sparse gene-by-cell expression matrix. Stator's finer resolution is clear from analyses of mouse embryonic brain, and human healthy or diseased liver. Rather than only coarse-scale labels of cell type, Stator further resolves cell types into subtypes, and these subtypes into stages of maturity and/or cell cycle phases, and yet further into portions of these phases. Among cryptically homogeneous embryonic cells, for example, Stator finds 34 distinct radial glia states whose gene expression forecasts their future GABAergic or glutamatergic neuronal fate. Further, Stator's fine resolution of liver cancer states reveals expression programmes that predict patient survival. We provide Stator as a Nextflow pipeline and Shiny App.
Collapse
Affiliation(s)
- Abel Jansma
- MRC Human Genetics Unit, Institute of Genetics & Cancer, University of Edinburgh, Edinburgh, EH4 2XU, UK
- Higgs Centre for Theoretical Physics, School of Physics & Astronomy, University of Edinburgh, Edinburgh, EH9 3FD, UK
| | - Yuelin Yao
- MRC Human Genetics Unit, Institute of Genetics & Cancer, University of Edinburgh, Edinburgh, EH4 2XU, UK
- School of Informatics, University of Edinburgh, Edinburgh, EH8 9AB, UK
| | - Jareth Wolfe
- MRC Human Genetics Unit, Institute of Genetics & Cancer, University of Edinburgh, Edinburgh, EH4 2XU, UK
| | - Luigi Del Debbio
- Higgs Centre for Theoretical Physics, School of Physics & Astronomy, University of Edinburgh, Edinburgh, EH9 3FD, UK
| | - Sjoerd V Beentjes
- MRC Human Genetics Unit, Institute of Genetics & Cancer, University of Edinburgh, Edinburgh, EH4 2XU, UK
- School of Mathematics, University of Edinburgh, Edinburgh, EH9 3FD, UK
| | - Chris P Ponting
- MRC Human Genetics Unit, Institute of Genetics & Cancer, University of Edinburgh, Edinburgh, EH4 2XU, UK.
| | - Ava Khamseh
- MRC Human Genetics Unit, Institute of Genetics & Cancer, University of Edinburgh, Edinburgh, EH4 2XU, UK.
- Higgs Centre for Theoretical Physics, School of Physics & Astronomy, University of Edinburgh, Edinburgh, EH9 3FD, UK.
- School of Informatics, University of Edinburgh, Edinburgh, EH8 9AB, UK.
| |
Collapse
|
6
|
Hu S, Qin J, Ding M, Gao R, Xiao Q, Lou J, Chen Y, Wang S, Pan Y. Bulk integrated single-cell-spatial transcriptomics reveals the impact of preoperative chemotherapy on cancer-associated fibroblasts and tumor cells in colorectal cancer, and construction of related predictive models using machine learning. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167535. [PMID: 39374811 DOI: 10.1016/j.bbadis.2024.167535] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Revised: 09/08/2024] [Accepted: 09/30/2024] [Indexed: 10/09/2024]
Abstract
BACKGROUND Preoperative chemotherapy (PC) is an important component of Colorectal cancer (CRC) treatment, but its effects on the biological functions of fibroblasts and epithelial cells in CRC are unclear. METHODS This study utilized bulk, single-cell, and spatial transcriptomic sequencing data from 22 independent cohorts of CRC. Through bioinformatics analysis and in vitro experiments, the research investigated the impact of PC on fibroblast and epithelial cells in CRC. Subpopulations associated with PC and CRC prognosis were identified, and a predictive model was constructed using machine learning. RESULTS PC significantly attenuated the pathways related to tumor progression in fibroblasts and epithelial cells. NOTCH3 + Fibroblast (NOTCH3 + Fib), TNNT1 + Epithelial (TNNT1 + Epi), and HSPA1A + Epithelial (HSPA1A + Epi) subpopulations were identified in the adjacent spatial region and were associated with poor prognosis in CRC. PC effectively diminished the presence of these subpopulations, concurrently inhibiting pathway activity and intercellular crosstalk. A risk signature model, named the Preoperative Chemotherapy Risk Signature Model (PCRSM), was constructed using machine learning. PCRSM emerged as an independent prognostic indicator for CRC, impacting both overall survival (OS) and recurrence-free survival (RFS), surpassing the performance of 89 previously published CRC risk signatures. Additionally, patients with a high PCRSM risk score showed sensitivity to fluorouracil-based adjuvant chemotherapy (FOLFOX) but resistance to single chemotherapy drugs (such as Bevacizumab and Oxaliplatin). Furthermore, this study predicted that patients with high PCRSM were resistant to anti-PD1therapy. CONCLUSION In conclusion, this study identified three cell subpopulations (NOTCH3 + Fib, TNNT1 + Epi, and HSPA1A + Epi) associated with PC, which can be targeted to improve the prognosis of CRC patients. The PCRSM model shows promise in enhancing the survival and treatment of CRC patients.
Collapse
Affiliation(s)
- Shangshang Hu
- School of Medicine, Southeast University, Nanjing 210009, Jiangsu, China
| | - Jian Qin
- School of Medicine, Southeast University, Nanjing 210009, Jiangsu, China
| | - Muzi Ding
- School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 211122, Jiangsu, China
| | - Rui Gao
- School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 211122, Jiangsu, China
| | - QianNi Xiao
- School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 211122, Jiangsu, China
| | - Jinwei Lou
- School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 211122, Jiangsu, China
| | - Yuhan Chen
- School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 211122, Jiangsu, China
| | - Shukui Wang
- School of Medicine, Southeast University, Nanjing 210009, Jiangsu, China; General Clinical Research Center, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, Jiangsu, China; Jiangsu Collaborative Innovation Center on Cancer Personalized Medicine, Nanjing Medical University, Nanjing 211100, Jiangsu, China.
| | - Yuqin Pan
- General Clinical Research Center, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, Jiangsu, China; Jiangsu Collaborative Innovation Center on Cancer Personalized Medicine, Nanjing Medical University, Nanjing 211100, Jiangsu, China.
| |
Collapse
|
7
|
Zhou XJ, Liu XF, Wang X, Cao XC. SITP: A single cell bioinformatics analysis flow captures proteasome markers in the development of breast cancer. Methods 2025; 233:1-10. [PMID: 39550019 DOI: 10.1016/j.ymeth.2024.11.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2024] [Revised: 11/06/2024] [Accepted: 11/13/2024] [Indexed: 11/18/2024] Open
Abstract
Single cell sequencing and related databases have been widely used in the exploration of cancer occurrence and development, but there is still no in-depth explanation of specific and complicated cellular protein modification processes. Ubiquitin-Proteasome System (UPS), as a specific and precise protein modification and degradation process, plays an important role in the biological functions of cancer cell proliferation and apoptosis. Proteasomes, vital multi-catalytic proteinases in eukaryotic cells, play a crucial role in protein degradation and contribute to tumor regulation. The 26S proteasome, part of the ubiquitin-proteasome system. In this study, we have enrolled a common SITP process including analysis of single cell sequencing to elucidate a flow that can capture typical proteasome markers in the oncogenesis and progression of breast cancer. PSMD11, a key component of the 26S proteasome regulatory particle, has been identified as a critical survival factor in cancer cells. Results suggest that PSMD11's rapid degradation is linked to acute apoptosis in cancer cells, making it a potential target for cancer treatment. Our study explored the potential mechanisms of PSMD11 in breast cancer development. The findings revealed the feasibility of disclosing ubiquitinating biomarkers from public database, as well as presented new evidence supporting PSMD11 as a potential therapeutic biomarker for breast cancer.
Collapse
Affiliation(s)
- Xue-Jie Zhou
- the First Department of Breast Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin 300060, PR China; Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin 300060, PR China; Key Laboratory of Cancer Prevention and Therapy, Tianjin 300060, PR China; Tianjin's Clinical Research Center for Cancer, Tianjin 300060, PR China
| | - Xiao-Feng Liu
- the First Department of Breast Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin 300060, PR China; Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin 300060, PR China; Key Laboratory of Cancer Prevention and Therapy, Tianjin 300060, PR China; Tianjin's Clinical Research Center for Cancer, Tianjin 300060, PR China
| | - Xin Wang
- the First Department of Breast Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin 300060, PR China; Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin 300060, PR China; Key Laboratory of Cancer Prevention and Therapy, Tianjin 300060, PR China; Tianjin's Clinical Research Center for Cancer, Tianjin 300060, PR China.
| | - Xu-Chen Cao
- the First Department of Breast Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin 300060, PR China; Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin 300060, PR China; Key Laboratory of Cancer Prevention and Therapy, Tianjin 300060, PR China; Tianjin's Clinical Research Center for Cancer, Tianjin 300060, PR China.
| |
Collapse
|
8
|
Li RF, Liu S, Gao Q, Fu M, Sun XY, Xiao M, Ge XY, Peng X. Inhibition of CDH11 Activates cGAS-STING by Stimulating Branched Chain Amino Acid Catabolism and Mitigates Lung Metastasis of Adenoid Cystic Carcinoma. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024:e2408751. [PMID: 39739317 DOI: 10.1002/advs.202408751] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/28/2024] [Revised: 10/10/2024] [Indexed: 01/02/2025]
Abstract
Salivary adenoid cystic carcinoma (SACC) is an intractable malignant tumor originates in the secretory glands and frequently metastasizes to the lungs. Hybrid epithelial-mesenchymal transition (EMT) cells within the tumors are correlated with augmented proliferative capacity and facilitation of lung metastasis. Single-cell RNA sequencing and spatial transcriptomic sequencing are employed to reveal the hybrid EMT subsets within the vascular fibroblast microenvironment. These hybrid EMT cells exhibit a pro-tumorigenic impact in vitro. Notably, cadherin 11 (CDH11), a specific marker for hybrid EMT cells, may exert its regulatory role in cellular function by interfering with branched-chain amino acids (BCAA) metabolism by inhibiting branched-chain ketoacid dehydrogenase to activate the mammalian target of the rapamycin pathway, thus making it a potential therapeutic target for SACC. Furthermore, celecoxib and its derivatives are specific CDH11 inhibitors that regulate BCAA metabolism, increase reactive oxygen species production, and subsequently activate the cyclic GMP-AMP synthase-stimulator of the interferongene pathway (cGAS-STING). They also inhibit lung metastasis in NOD-SCID mice in vivo. Overall, these findings suggest a promising treatment strategy that targets hybrid EMT cells to mitigate lung metastasis in SACC. Celecoxib may serve as a promising clinical intervention for the treatment of lung metastases in patients with SACC.
Collapse
Affiliation(s)
- Rui-Feng Li
- Department of Oral and Maxillofacial Surgery, Peking University School and Hospital of Stomatology, Beijing, 100081, P. R. China
- Central Laboratory, Peking University School and Hospital of Stomatology, Beijing, 100081, P. R. China
- National Center for Stomatology, Beijing, 100081, P. R. China
- National Clinical Research Center for Oral Diseases, Beijing, 100081, P. R. China
- National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, BeiJing, 100081, P. R. China
| | - Shuo Liu
- Department of Oral and Maxillofacial Surgery, Peking University School and Hospital of Stomatology, Beijing, 100081, P. R. China
- Central Laboratory, Peking University School and Hospital of Stomatology, Beijing, 100081, P. R. China
- National Center for Stomatology, Beijing, 100081, P. R. China
- National Clinical Research Center for Oral Diseases, Beijing, 100081, P. R. China
- National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, BeiJing, 100081, P. R. China
| | - Qian Gao
- Department of Oral and Maxillofacial Surgery, Peking University School and Hospital of Stomatology, Beijing, 100081, P. R. China
- Central Laboratory, Peking University School and Hospital of Stomatology, Beijing, 100081, P. R. China
- National Center for Stomatology, Beijing, 100081, P. R. China
- National Clinical Research Center for Oral Diseases, Beijing, 100081, P. R. China
- National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, BeiJing, 100081, P. R. China
| | - Min Fu
- Department of Oral and Maxillofacial Surgery, Peking University School and Hospital of Stomatology, Beijing, 100081, P. R. China
- Central Laboratory, Peking University School and Hospital of Stomatology, Beijing, 100081, P. R. China
- National Center for Stomatology, Beijing, 100081, P. R. China
- National Clinical Research Center for Oral Diseases, Beijing, 100081, P. R. China
- National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, BeiJing, 100081, P. R. China
| | - Xin-Yi Sun
- Department of Oral and Maxillofacial Surgery, Peking University School and Hospital of Stomatology, Beijing, 100081, P. R. China
- Central Laboratory, Peking University School and Hospital of Stomatology, Beijing, 100081, P. R. China
- National Center for Stomatology, Beijing, 100081, P. R. China
- National Clinical Research Center for Oral Diseases, Beijing, 100081, P. R. China
- National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, BeiJing, 100081, P. R. China
| | - Mian Xiao
- Department of Oral and Maxillofacial Surgery, Peking University School and Hospital of Stomatology, Beijing, 100081, P. R. China
- Central Laboratory, Peking University School and Hospital of Stomatology, Beijing, 100081, P. R. China
- National Center for Stomatology, Beijing, 100081, P. R. China
- National Clinical Research Center for Oral Diseases, Beijing, 100081, P. R. China
- National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, BeiJing, 100081, P. R. China
| | - Xi-Yuan Ge
- Department of Oral and Maxillofacial Surgery, Peking University School and Hospital of Stomatology, Beijing, 100081, P. R. China
- Central Laboratory, Peking University School and Hospital of Stomatology, Beijing, 100081, P. R. China
- National Center for Stomatology, Beijing, 100081, P. R. China
- National Clinical Research Center for Oral Diseases, Beijing, 100081, P. R. China
- National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, BeiJing, 100081, P. R. China
| | - Xin Peng
- Department of Oral and Maxillofacial Surgery, Peking University School and Hospital of Stomatology, Beijing, 100081, P. R. China
- Central Laboratory, Peking University School and Hospital of Stomatology, Beijing, 100081, P. R. China
- National Center for Stomatology, Beijing, 100081, P. R. China
- National Clinical Research Center for Oral Diseases, Beijing, 100081, P. R. China
- National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, BeiJing, 100081, P. R. China
| |
Collapse
|
9
|
Requena D, Medico JA, Soto-Ugaldi LF, Shirani M, Saltsman JA, Torbenson MS, Coffino P, Simon SM. Liver cancer multiomics reveals diverse protein kinase A disruptions convergently produce fibrolamellar hepatocellular carcinoma. Nat Commun 2024; 15:10887. [PMID: 39738196 DOI: 10.1038/s41467-024-55238-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Accepted: 12/03/2024] [Indexed: 01/01/2025] Open
Abstract
Fibrolamellar Hepatocellular Carcinoma (FLC) is a rare liver cancer characterized by a fusion oncokinase of the genes DNAJB1 and PRKACA, the catalytic subunit of protein kinase A (PKA). A few FLC-like tumors have been reported showing other alterations involving PKA. To better understand FLC pathogenesis and the relationships among FLC, FLC-like, and other liver tumors, we performed a massive multi-omics analysis. RNA-seq data of 1412 liver tumors from FLC, hepatocellular carcinoma, hepatoblastoma and intrahepatic cholangiocarcinoma are analyzed, obtaining transcriptomic signatures unrestricted by experimental processing methods. These signatures reveal which dysregulations are unique to specific tumors and which are common to all liver cancers. Moreover, the transcriptomic FLC signature identifies a unifying phenotype for all FLC tumors regardless of how PKA was activated. We study this signature at multi-omics and single-cell levels in the first spatial transcriptomic characterization of FLC, identifying the contribution of tumor, normal, stromal, and infiltrating immune cells. Additionally, we study FLC metastases, finding small differences from the primary tumors.
Collapse
Affiliation(s)
- David Requena
- Laboratory of Cellular Biophysics, The Rockefeller University, New York, NY, USA
| | - Jack A Medico
- Laboratory of Cellular Biophysics, The Rockefeller University, New York, NY, USA
| | - Luis F Soto-Ugaldi
- Laboratory of Cellular Biophysics, The Rockefeller University, New York, NY, USA
| | - Mahsa Shirani
- Laboratory of Cellular Biophysics, The Rockefeller University, New York, NY, USA
| | - James A Saltsman
- Laboratory of Cellular Biophysics, The Rockefeller University, New York, NY, USA
| | | | - Philip Coffino
- Laboratory of Cellular Biophysics, The Rockefeller University, New York, NY, USA
| | - Sanford M Simon
- Laboratory of Cellular Biophysics, The Rockefeller University, New York, NY, USA.
| |
Collapse
|
10
|
Liu Z, Wang S. A novel biomarker of COVI-19: MMP8 emerged by integrated bulk RNAseq and single-cell sequencing. Sci Rep 2024; 14:31086. [PMID: 39730651 DOI: 10.1038/s41598-024-82227-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Accepted: 12/03/2024] [Indexed: 12/29/2024] Open
Abstract
COVID-19 has been emerging as the most influential illness which has caused great costs to the heath of population and social economy. Sivelestat sodium (SS) is indicated as an effective cure for lung dysfunction, a characteristic symptom of COVID-19 infection, but its pharmacological target is still unclear. Therefore, a deep understanding of the pathological progression and molecular alteration is an urgent issue for settling the diagnosis and therapy problems of COVID-19. In this study, the bulk ribonucleic acid sequencing (RNA-seq) data of healthy donors and non-severe and severe COVID-19 patients were collected. Then, target differentially expressed genes (DEGs) were screened through integrating sequencing data and the pharmacological database. Besides, with the help of functional and molecular interaction analyses, the potential effect of target gene alteration on COVID-19 progression was investigated. Single-cell sequencing was performed to evaluate the cell distribution of target genes, and the possible interaction of gene-positive cells with other cells was explored by intercellular ligand-receptor pattern analysis. The results showed that matrix metalloproteinase 8 (MMP8) was upregulated in severe COVID-19 patients, which was also identified as a targeting site to SS. Additionally, MMP8 took a core part in the regulatory interaction network of the screened DEGs in COVID-19 and was dramatically correlated with the inflammatory signaling pathway. The further investigations indicated that MMP8 was mainly expressed in myelocytes with a high degree of heterogeneity. MMP8-positive myelocytes interacted with other cell types through RETN-TLR4 and RETN-CAP1 ligand-receptor patterns. These findings emphasize the important role of MMP8 in COVID-19 progression and provide a potential therapeutic target for COVID-19 patients.
Collapse
Affiliation(s)
- Zhenguo Liu
- Department of Intensive Care Unit, Shaanxi Provincial People's Hospital, Xi'an, 710068, Shaanxi, China
| | - Shunda Wang
- Department of Rehabilitative medicine, Shaanxi Provincial People's Hospital, No.256, Youyi West Road, Beilin District, Xi'an, 710068, Shaanxi, China.
| |
Collapse
|
11
|
Wu Y, Zhai Y, Ding Z, Xie T, Zhu W, Zhang C, Lu Y, Chen Y, Ren S, Hu Y, Li X, Zhong F, Liang Y, Wang S. Single-cell transcriptomics reveals tumor microenvironment changes and prognostic gene signatures in hepatocellular carcinoma. Int Immunopharmacol 2024; 143:113317. [PMID: 39447409 DOI: 10.1016/j.intimp.2024.113317] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Revised: 09/23/2024] [Accepted: 10/01/2024] [Indexed: 10/26/2024]
Abstract
BACKGROUND Hepatocellular Carcinoma (HCC) is the most common type of primary liver cancer, accounting for the majority of liver cancer cases. Hepatocellular Carcinoma not only exhibits high heterogeneity but also possesses an immune-suppressive tumor microenvironment that promotes tumor evasion, posing substantial difficulties for efficient therapy. Our aim is to utilize single-cell RNA transcriptome data to investigate the dynamic changes in the tumor microenvironment during the malignant progression of HCC, the communication among immune cells, and the marker genes associated with patient prognosis. METHODS We constructed expression matrices from open single-cell RNA transcriptome data (GSE149614) of HCC patients (representing stages I-IV), establishing single-cell RNA transcriptional atlases for different stages of HCC progression. For each stage, we conducted cell subgroup analysis to identify cell types at each stage. Horizontally, we explored the dynamic changes of the same cell type across different stages, performing trajectory analysis and prognosis analysis. Vertically, we investigated pairwise comparisons of different stages of HCC progression, probing the dynamic alterations in tumor microenvironment immune cell signaling pathways. Finally, potential drugs for the treatment of HCC were predicted based on relevant genes. FINDINGS As the HCC advances towards increased malignancy, there is a shift in the predominant composition of the tumor microenvironment, with a decline in the dominance of hepatic cells. Tumor-infiltrating immune cells migrate and accumulate within the tumor microenvironment, where T cells and myeloid cells display distinct patterns of change. Genes associated with cancer-associated fibroblasts (CAFs) and T cells are correlated with adverse patient outcomes. In the late stages of HCC, the tumor microenvironment is infiltrated by more myeloid-derived suppressor cells (MDSCs), and a prognostic model constructed based on genes related to myeloid cells can predict patient outcomes. Additionally, in the analysis of transcription factors, YY1 and MYC are found to be highly expressed. Cell communication analysis among tumor-infiltrating immune cells reveals significant differences in the main signaling pathways at different stages of HCC progression. Finally, drug sensitivity analysis based on key genes identifies Acetalax, Allopurinol, and Amonafide as potential candidates for HCC treatment.
Collapse
Affiliation(s)
- Yilin Wu
- School of Life Science and Food Engineering, Huaiyin Institute of Technology, Huai'an, China
| | - Yangyang Zhai
- Shanghai Key Laboratory of Biliary Tract Disease Research, Shanghai Research Center of Biliary Tract Disease, Department of General Surgery, Xinhua Hospital, Affiliated with Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhilong Ding
- Department of Hepatobiliary Surgery, The Affiliated Huaian Hospital of Xuzhou Medical University and Huai'an Second People's Hospital, Huai'an, Jiangsu, China
| | - Tong Xie
- School of Life Science and Food Engineering, Huaiyin Institute of Technology, Huai'an, China
| | - WeiJie Zhu
- School of Life Science and Food Engineering, Huaiyin Institute of Technology, Huai'an, China
| | - Cui Zhang
- School of Life Science and Food Engineering, Huaiyin Institute of Technology, Huai'an, China
| | - Ying Lu
- School of Life Science and Food Engineering, Huaiyin Institute of Technology, Huai'an, China
| | - Yunli Chen
- School of Life Science and Food Engineering, Huaiyin Institute of Technology, Huai'an, China
| | - Shiying Ren
- School of Life Science and Food Engineering, Huaiyin Institute of Technology, Huai'an, China
| | - Yihuai Hu
- School of Life Science and Food Engineering, Huaiyin Institute of Technology, Huai'an, China
| | - Xiangqian Li
- School of Life Science and Food Engineering, Huaiyin Institute of Technology, Huai'an, China
| | - Fei Zhong
- Department of Laboratory Medicine, The Affiliated Huaian Hospital of Xuzhou Medical University and Huai'an Second People's Hospital, Huai'an, Jiangsu, China.
| | - Yong Liang
- Department of Laboratory Medicine, The Affiliated Huaian Hospital of Xuzhou Medical University and Huai'an Second People's Hospital, Huai'an, Jiangsu, China.
| | - Shiyan Wang
- School of Life Science and Food Engineering, Huaiyin Institute of Technology, Huai'an, China.
| |
Collapse
|
12
|
Yu Y, Wang C, Wang Y, Shi H, Hu H, Du Y, Zhou Z. The conserved wobble uridine tRNA thiolase Ctu1 is required for angiogenesis and embryonic development. PLoS One 2024; 19:e0315854. [PMID: 39705244 DOI: 10.1371/journal.pone.0315854] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2024] [Accepted: 12/01/2024] [Indexed: 12/22/2024] Open
Abstract
Cytosolic thiouridylase is a conserved cytoplasmic tRNA thiolase composed of two different subunits, CTU1 and CTU2. CTU2 serves as a scaffold protein, while CTU1 catalyzes the 2-thiolation at the 34th wobble uridine of the anticodon loop. tRNAGlnUUG, tRNAGluUUC, and tRNALysUUU are the tRNA substrates that are modified with a thiol group at the C2 positions (s2) by CTU1, and also with a methoxycarbonylmethyl group at the C5 positions (mcm5) by Elongator and ALKBH8. mcm5s2U34 modification of the three tRNAs, and their modifying enzymes are involved in human disease and development. Elongator mutant animals exhibit severe phenotypes, while the biological function of Ctu1 in vertebrate animal models remains poorly characterized. Here, we applied antisense morpholino oligonucleotides targeting cytosolic thiouridylase subunit1 (ctu1) transcripts in a zebrafish model and small interfereing RNA against CTU1 transcript in human endothelial cells to define the phenotypes. We found that deficiency of ctu1 causes impaired angiogenesis and development in zebrafish embryos, and CTU1 is involved in proliferation, migration, and tube formation of human endothelial cells. We employed single-cell RNA sequencing to acquire the transcriptomic atlas from ctu1 and control morphant zebrafish. Comprehensive bioinformatics analysis, including pseudo-time, RNA velocity, cell-cell communication, and gene regulatory network inference revealed that ctu1 deficiency leads to the arrest of cell cycle, and the defects of nerve development and erythrocyte differentiation and the attenuation of several pro-angiogenic signaling pathways, e.g., angpt-tek and dll4-notch. Our findings show for the first time that CTU1 is essential for angiogenesis and embryonic development in vertebrates.
Collapse
Affiliation(s)
- Yangziwei Yu
- School of Medical Instrument and Food Engineering, University of Shanghai for Science and Technology, Shanghai, China
- Collaborative innovation Center for Biomedicine, Shanghai University of Medicine and Health Sciences, Shanghai, China
| | - Chuqiao Wang
- Collaborative innovation Center for Biomedicine, Shanghai University of Medicine and Health Sciences, Shanghai, China
- Department of Pharmaceutical Toxicology, School of Pharmacy, China Medical University, Shenyang, China
| | - Yan Wang
- Collaborative innovation Center for Biomedicine, Shanghai University of Medicine and Health Sciences, Shanghai, China
- Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Heng Shi
- Collaborative innovation Center for Biomedicine, Shanghai University of Medicine and Health Sciences, Shanghai, China
- Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Huiyuan Hu
- Department of Pharmaceutical Toxicology, School of Pharmacy, China Medical University, Shenyang, China
| | - Yibin Du
- Shanghai World Foreign Language Academy, Shanghai, China
| | - Zhaoli Zhou
- School of Medical Instrument and Food Engineering, University of Shanghai for Science and Technology, Shanghai, China
- Collaborative innovation Center for Biomedicine, Shanghai University of Medicine and Health Sciences, Shanghai, China
- Jiading District Central Hospital Affiliated Shanghai University of Medicine and Health Sciences, Shanghai University of Medicine and Health Sciences, Shanghai, China
| |
Collapse
|
13
|
Raffaele S, Clausen BH, Mannella FC, Wirenfeldt M, Marangon D, Tidgen SB, Corradini S, Madsen K, Lecca D, Abbracchio MP, Lambertsen KL, Fumagalli M. Characterisation of GPR17-expressing oligodendrocyte precursors in human ischaemic lesions and correlation with reactive glial responses. J Pathol 2024. [PMID: 39703181 DOI: 10.1002/path.6381] [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: 06/03/2024] [Revised: 10/14/2024] [Accepted: 11/20/2024] [Indexed: 12/21/2024]
Abstract
White matter damage and subsequent demyelination significantly contribute to long-term functional impairment after ischaemic stroke. Identifying novel pharmacological targets to restore myelin integrity by promoting the maturation of oligodendrocyte precursor cells (OPCs) into new myelinating oligodendrocytes may open new perspectives for ischaemic stroke treatment. In this respect, previous studies highlighted the role of the G protein-coupled membrane receptor 17 (GPR17) as a key regulator of OPC differentiation in experimental models of brain injury, including ischaemic stroke. To determine the translational value of GPR17 as a possible target in the context of human disease, we exploited immunohistochemistry to characterise the distribution of GPR17-expressing cells in brain tissue samples from ischaemic stroke cases and correlated it with the reactive state of neighbouring glial cells. The results showed that GPR17 specifically decorates a subpopulation of differentiation-committed OPCs, labelled by the peculiar marker breast carcinoma-amplified sequence 1 (BCAS1), that accumulates in the peri-infarct region in the later stages after the ischaemic event. Interestingly, the response of GPR17-expressing cells appears to be paralleled by the switch of reactive microglia/macrophages from a phagocytic to a dystrophic phenotype and by astrocytic scar formation. A negative correlation was found between GPR17-expressing OPCs and reactive microglia/macrophages and astrocytes surrounding chronic ischaemic lesions in female subjects, while the same relationship was less pronounced in males. These results were reinforced by bioinformatic analysis of a publicly available transcriptomic dataset, which implicated a possible role of inflammation and defective neuron-to-OPC communication in remyelination failure after ischaemic damage. Hence, these data strengthen the relevance of GPR17-based remyelinating therapies for the treatment of ischaemic stroke. © 2024 The Author(s). The Journal of Pathology published by John Wiley & Sons Ltd on behalf of The Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Stefano Raffaele
- Department of Pharmacological and Biomolecular Sciences 'Rodolfo Paoletti', Università degli Studi di Milano, Milan, Italy
| | - Bettina Hjelm Clausen
- Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
- Department of Clinical Research, Brain Research - Inter Disciplinary Guided Excellence (BRIDGE), University of Southern Denmark, Odense, Denmark
- Odense Patient data Explorative Network (OPEN), Department of Clinical Research, Odense University Hospital, University of Southern Denmark, Odense, Denmark
| | - Francesca Carolina Mannella
- Department of Pharmacological and Biomolecular Sciences 'Rodolfo Paoletti', Università degli Studi di Milano, Milan, Italy
| | - Martin Wirenfeldt
- Department of Clinical Research, Brain Research - Inter Disciplinary Guided Excellence (BRIDGE), University of Southern Denmark, Odense, Denmark
- Odense Patient data Explorative Network (OPEN), Department of Clinical Research, Odense University Hospital, University of Southern Denmark, Odense, Denmark
- Department of Pathology, South Denmark University Hospital, Odense, Denmark
| | - Davide Marangon
- Department of Pharmaceutical Sciences, Università degli Studi di Milano, Milan, Italy
| | - Sarah Boe Tidgen
- Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Silvia Corradini
- Department of Pharmacological and Biomolecular Sciences 'Rodolfo Paoletti', Università degli Studi di Milano, Milan, Italy
- Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Kirsten Madsen
- Department of Pathology, South Denmark University Hospital, Odense, Denmark
- Department of Cardiovascular and Renal Research, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Davide Lecca
- Department of Pharmaceutical Sciences, Università degli Studi di Milano, Milan, Italy
| | - Maria Pia Abbracchio
- Department of Pharmaceutical Sciences, Università degli Studi di Milano, Milan, Italy
| | - Kate Lykke Lambertsen
- Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
- Department of Clinical Research, Brain Research - Inter Disciplinary Guided Excellence (BRIDGE), University of Southern Denmark, Odense, Denmark
- Odense Patient data Explorative Network (OPEN), Department of Clinical Research, Odense University Hospital, University of Southern Denmark, Odense, Denmark
- Department of Neurology, Odense University Hospital, Odense, Denmark
| | - Marta Fumagalli
- Department of Pharmacological and Biomolecular Sciences 'Rodolfo Paoletti', Università degli Studi di Milano, Milan, Italy
| |
Collapse
|
14
|
Kim DW, Kim S, Han J, Belday K, Li E, Mahoney N, Blackshaw S, Rajaii F. Transcriptomic profiling of thyroid eye disease orbital fat demonstrates differences in adipogenicity and IGF-1R pathway. JCI Insight 2024; 9:e182352. [PMID: 39704170 DOI: 10.1172/jci.insight.182352] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Accepted: 11/05/2024] [Indexed: 12/21/2024] Open
Abstract
Despite recent advances in the treatment of thyroid eye disease thyroid-related eye disease (TED), marked gaps remain in our understanding of the underlying molecular mechanisms, particularly concerning the insulin-like growth factor-1 receptor (IGF-1R) pathway. To dissect the pathophysiology of TED, we used single-nucleus RNA-Seq to analyze orbital fat specimens from both patients with TED and matched individuals acting as controls. The analysis demonstrated a marked increase in the proportion of fibroblasts transitioning to adipogenesis in the orbital fat of patients with TED compared with that in control patients. This was associated with diverse alterations in immune cell composition. Significant alterations in the IGF-1R signaling pathway were noted between TED specimens and those from control patients, indicating a potential pathological mechanism driven by IGF-1R signaling abnormalities. Additionally, our data showed that linsitinib, a small-molecule inhibitor of IGF-1R, effectively reduced adipogenesis in TED orbital fibroblasts in vitro, suggesting its potential utility as a therapeutic agent. Our findings reveal that, beyond immune dysfunction, abnormal IGF-1R signaling leading to enhanced adipogenesis is a crucial pathogenic mechanism in TED.
Collapse
Affiliation(s)
- Dong Won Kim
- Danish Research Institute of Translational Neuroscience (DANDRITE), Nordic EMBL Partnership for Molecular Medicine, and
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Soohyun Kim
- Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Jeong Han
- Baylor College of Medicine, Houston, Texas, USA
| | - Karan Belday
- Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Emily Li
- Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Nicholas Mahoney
- Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Seth Blackshaw
- Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Department of Neurology
- Institute for Cell Engineering, and
- Kavli Neuroscience Discovery Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Fatemeh Rajaii
- Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
15
|
Li H, Jiao Y, Zhang Y, Liu J, Huang S. Exploring tumor microenvironment interactions and apoptosis pathways in NSCLC through spatial transcriptomics and machine learning. Cell Oncol (Dordr) 2024:10.1007/s13402-024-01025-6. [PMID: 39699801 DOI: 10.1007/s13402-024-01025-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/02/2024] [Indexed: 12/20/2024] Open
Abstract
BACKGROUND The most common type of lung cancer is non-small cell lung cancer (NSCLC), accounting for 85% of all cases. Programmed cell death (PCD), an important regulatory mechanism for cell survival and homeostasis, has become increasingly prominent in cancer research in recent years. As such, exploring the role of PCD in NSCLC may help uncover new mechanisms for therapeutic targets. METHODS We utilized the GEO database and TCGA NSCLC gene data to screen for co-expressed genes. To delve deeper, single-cell sequencing combined with spatial transcriptomics was employed to study the intrinsic mechanisms of programmed cell death in cells and their interaction with the tumor microenvironment. Furthermore, Mendelian randomization was applied to screen for causally related genes. Prognostic models were constructed using various machine learning algorithms, and multi-cohort multi-omics analyses were conducted to screen for genes. In vitro experiments were then carried out to reveal the biological functions of the genes and their relationship with apoptosis. RESULTS Cells with high programmed cell death activity primarily activate pathways related to apoptosis, cell migration, and hypoxia, while also exhibiting strong interactions with smooth muscle cells in the tumor microenvironment. Based on a set of programmed cell death genes, the prognostic model NSCLCPCD demonstrates strong predictive capabilities. Moreover, laboratory experiments confirm that SLC7A5 promotes the proliferation of NSCLC cells, and the knockout of SLC7A5 significantly increases tumor cell apoptosis. CONCLUSIONS Our data indicate that programmed cell death is predominantly associated with pathways related to apoptosis, tumor metastasis, and hypoxia. Additionally, it suggests that SLC7A5 is a significant risk indicator for the prognosis of non-small cell lung cancer (NSCLC) and may serve as an effective target for enhancing apoptosis in NSCLC tumor cells.
Collapse
Affiliation(s)
- Huimin Li
- Department of Internal Medicine Residency Training Base, Gongli Hospital of Shanghai Pudong New Area, Shanghai, 200135, China
| | - Yuheng Jiao
- Department of Heart Failure, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
| | - Yi Zhang
- Department of Otolaryngology, Gongli Hospital of Shanghai Pudong New Area, Shanghai, 200135, China
| | - Junzhi Liu
- Department of Otolaryngology, Gongli Hospital of Shanghai Pudong New Area, Shanghai, 200135, China.
| | - Shuixian Huang
- Department of Otolaryngology, Gongli Hospital of Shanghai Pudong New Area, Shanghai, 200135, China.
| |
Collapse
|
16
|
Rao J, Wang X, Wang Z. Integration of Microarray Data and Single-Cell Sequencing Analysis to Explore Key Genes Associated with Macrophage Infiltration in Heart Failure. J Inflamm Res 2024; 17:11257-11274. [PMID: 39717663 PMCID: PMC11665153 DOI: 10.2147/jir.s475633] [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: 04/26/2024] [Accepted: 12/14/2024] [Indexed: 12/25/2024] Open
Abstract
Background Cardiac macrophages are a heterogeneous population with high plasticity and adaptability, and their mechanisms in heart failure (HF) remain poorly elucidated. Methods We used single-cell and bulk RNA sequencing data to reveal the heterogeneity of non-cardiomyocytes and assess the immunoreactivity of each subpopulation. Additionally, we employed four integrated machine learning algorithms to identify macrophage-related genes with diagnostic value, and in vivo validation was performed. To assess the immune infiltration characteristics in HF, we utilized the CIBERSORT and single sample gene set enrichment analysis (ssGSEA). An unsupervised consensus clustering algorithm was applied to identify the macrophage-related HF subtypes. Furthermore, the scMetabolism was employed to explore the specific metabolic patterns of the macrophage subtypes. Finally, CellChat was used to investigate cell-cell interactions among the identified subtypes. Results The immunoreactivity score of macrophages in the HF was higher than that in the other cell types. GSEA of macrophage clusters indicated a significant enrichment of leukocyte-mediated immune processes, antigen processing, and presentation. The intersection of the results from machine learning revealed that SERPINA3, GPAT3, ANPEP, and FCER1G can serve as feature genes and form a diagnostic model with a good predictive capability. Unsupervised consensus clustering algorithms reveal the immune and metabolic subtypes of macrophages. The metabolic heterogeneity of macrophage subpopulations can lead to macrophage polarization into different types, which may be related to the metabolic reprogramming between glycolysis and mitochondrial oxidative phosphorylation. Cellular communication revealed that macrophages form a network of interactions with neutrophils to support each other's functions and maintenance. The complex efferent and afferent signals are closely associated with myocardial fibrosis. Conclusion SERPINA3, GPAT3, ANPEP, and FCER1G can potentially serve as immune therapeutic targets and central biomarkers. The immunological and metabolic heterogeneity of macrophages may offer a more precise direction to explore the mechanisms underlying HF and novel immunotherapies.
Collapse
Affiliation(s)
- Jin Rao
- Department of Cardiothoracic Surgery, Changzheng Hospital, Naval Medical University, Shanghai, People’s Republic of China
| | - Xuefu Wang
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai, People’s Republic of China
| | - Zhinong Wang
- Department of Cardiothoracic Surgery, Changzheng Hospital, Naval Medical University, Shanghai, People’s Republic of China
| |
Collapse
|
17
|
Chin WL, Cook AM, Chee J, Principe N, Hoang TS, Kidman J, Hmon KPW, Yeow Y, Jones ME, Hou R, Denisenko E, McDonnell AM, Hon CC, Moody J, Anderson D, Yip S, Cummins MM, Stockler MR, Kok PS, Brown C, John T, Kao SCH, Karikios DJ, O'Byrne KJ, Hughes BGM, Lake RA, Forrest ARR, Nowak AK, Lassmann T, Lesterhuis WJ. Coupling of response biomarkers between tumor and peripheral blood in patients undergoing chemoimmunotherapy. Cell Rep Med 2024:101882. [PMID: 39731918 DOI: 10.1016/j.xcrm.2024.101882] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2023] [Revised: 05/14/2024] [Accepted: 11/29/2024] [Indexed: 12/30/2024]
Abstract
Platinum-based chemotherapy in combination with anti-PD-L1 antibodies has shown promising results in mesothelioma. However, the immunological mechanisms underlying its efficacy are not well understood and there are no predictive biomarkers to guide treatment decisions. Here, we combine time course RNA sequencing (RNA-seq) of peripheral blood mononuclear cells with pre-treatment tumor transcriptome data from the single-arm, phase 2 DREAM trial (N = 54). Single-cell RNA-seq and T cell receptor sequencing (TCR-seq) reveal that CD8+ T effector memory (TEM) cells with stem-like properties are more abundant in peripheral blood of responders and that this population expands upon treatment. These peripheral blood changes are linked to the transcriptional state of the tumor microenvironment. Combining information from both compartments, rather than individually, is most predictive of response. Our study highlights complex interactions between the tumor and immune cells in peripheral blood during objective tumor responses to chemoimmunotherapy. This trial is registered with the Australian New Zealand Clinical Trials Registry, number ACTRN12616001170415.
Collapse
Affiliation(s)
- Wee Loong Chin
- National Centre for Asbestos Related Diseases, Institute for Respiratory Health, Nedlands, WA 6009, Australia; Medical School, University of Western Australia, Crawley, WA 6009, Australia; Department of Medical Oncology, Sir Charles Gairdner Hospital, Nedlands, WA 6009, Australia
| | - Alistair M Cook
- National Centre for Asbestos Related Diseases, Institute for Respiratory Health, Nedlands, WA 6009, Australia; School of Biomedical Sciences, University of Western Australia, Crawley, WA 6009, Australia
| | - Jonathan Chee
- National Centre for Asbestos Related Diseases, Institute for Respiratory Health, Nedlands, WA 6009, Australia; School of Biomedical Sciences, University of Western Australia, Crawley, WA 6009, Australia
| | - Nicola Principe
- National Centre for Asbestos Related Diseases, Institute for Respiratory Health, Nedlands, WA 6009, Australia; School of Biomedical Sciences, University of Western Australia, Crawley, WA 6009, Australia
| | - Tracy S Hoang
- National Centre for Asbestos Related Diseases, Institute for Respiratory Health, Nedlands, WA 6009, Australia; School of Biomedical Sciences, University of Western Australia, Crawley, WA 6009, Australia
| | - Joel Kidman
- National Centre for Asbestos Related Diseases, Institute for Respiratory Health, Nedlands, WA 6009, Australia; School of Biomedical Sciences, University of Western Australia, Crawley, WA 6009, Australia
| | - Khaing P W Hmon
- Harry Perkins Institute of Medical Research, QEII Medical Centre and Centre for Medical Research, The University of Western Australia, Nedlands, Perth, WA 6009, Australia
| | - Yen Yeow
- Harry Perkins Institute of Medical Research, QEII Medical Centre and Centre for Medical Research, The University of Western Australia, Nedlands, Perth, WA 6009, Australia
| | - Matthew E Jones
- Harry Perkins Institute of Medical Research, QEII Medical Centre and Centre for Medical Research, The University of Western Australia, Nedlands, Perth, WA 6009, Australia
| | - Rui Hou
- Harry Perkins Institute of Medical Research, QEII Medical Centre and Centre for Medical Research, The University of Western Australia, Nedlands, Perth, WA 6009, Australia
| | - Elena Denisenko
- Harry Perkins Institute of Medical Research, QEII Medical Centre and Centre for Medical Research, The University of Western Australia, Nedlands, Perth, WA 6009, Australia
| | - Alison M McDonnell
- National Centre for Asbestos Related Diseases, Institute for Respiratory Health, Nedlands, WA 6009, Australia; School of Biomedical Sciences, University of Western Australia, Crawley, WA 6009, Australia; The Kids Research Institute, University of Western Australia, Nedlands WA 6009, Australia
| | - Chung-Chau Hon
- RIKEN Center for Integrative Medical Sciences, Yokohama City, Kanagawa 230-0045, Japan
| | - Jonathan Moody
- RIKEN Center for Integrative Medical Sciences, Yokohama City, Kanagawa 230-0045, Japan
| | - Denise Anderson
- The Kids Research Institute, University of Western Australia, Nedlands WA 6009, Australia
| | - Sonia Yip
- National Health and Medical Research Council, Clinical Trials Centre, University of Sydney, Camperdown, NSW, Australia
| | - Michelle M Cummins
- National Health and Medical Research Council, Clinical Trials Centre, University of Sydney, Camperdown, NSW, Australia
| | - Martin R Stockler
- National Health and Medical Research Council, Clinical Trials Centre, University of Sydney, Camperdown, NSW, Australia
| | - Peey-Sei Kok
- National Health and Medical Research Council, Clinical Trials Centre, University of Sydney, Camperdown, NSW, Australia
| | - Chris Brown
- National Health and Medical Research Council, Clinical Trials Centre, University of Sydney, Camperdown, NSW, Australia
| | - Thomas John
- Department of Medical Oncology, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
| | - Steven C-H Kao
- Department of Medical Oncology, Chris O'Brien Lifehouse, Camperdown, NSW, Australia
| | - Deme J Karikios
- Department of Medical Oncology, Nepean Hospital, Kingswood, NSW, Australia
| | - Kenneth J O'Byrne
- Department of Medical Oncology, Princess Alexandra Hospital, Brisbane, QLD, Australia
| | - Brett G M Hughes
- Royal Brisbane and Women's Hospital, Brisbane, QLD, Australia; School of Medicine, University of Queensland, Brisbane, QLD, Australia
| | - Richard A Lake
- National Centre for Asbestos Related Diseases, Institute for Respiratory Health, Nedlands, WA 6009, Australia; Medical School, University of Western Australia, Crawley, WA 6009, Australia
| | - Alistair R R Forrest
- Harry Perkins Institute of Medical Research, QEII Medical Centre and Centre for Medical Research, The University of Western Australia, Nedlands, Perth, WA 6009, Australia.
| | - Anna K Nowak
- National Centre for Asbestos Related Diseases, Institute for Respiratory Health, Nedlands, WA 6009, Australia; Medical School, University of Western Australia, Crawley, WA 6009, Australia; Department of Medical Oncology, Sir Charles Gairdner Hospital, Nedlands, WA 6009, Australia.
| | - Timo Lassmann
- The Kids Research Institute, University of Western Australia, Nedlands WA 6009, Australia.
| | - W Joost Lesterhuis
- National Centre for Asbestos Related Diseases, Institute for Respiratory Health, Nedlands, WA 6009, Australia; School of Biomedical Sciences, University of Western Australia, Crawley, WA 6009, Australia; The Kids Research Institute, University of Western Australia, Nedlands WA 6009, Australia.
| |
Collapse
|
18
|
Chen X, Zhou B, Wang S, Jiang X, Ping Y, Xia J, Yu F, Li Y, Zhang M, Ding Y. Intestinal metaplasia key molecules and UPP1 activation via Helicobacter pylori /NF-kB: drivers of malignant progression in gastric cancer. Cancer Cell Int 2024; 24:399. [PMID: 39695769 DOI: 10.1186/s12935-024-03598-6] [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: 08/18/2024] [Accepted: 12/04/2024] [Indexed: 12/20/2024] Open
Abstract
Gastric cancer (GC) remains a significant global health challenge due to its high morbidity and mortality rates. The development of GC is a multi-hit process and the exploration of precancerous lesions is crucial. To elucidate the molecular and cellular dynamics underlying gastric carcinogenesis, we conducted an integrative single-cell RNA sequencing analysis of 26,028 high-quality cells from gastric antral mucosa biopsies across various stages, including non-atrophic gastritis, chronic atrophic gastritis, intestinal metaplasia, and early gastric cancer. By constructing a detailed single-cell atlas, we identified distinct epithelial cell subpopulations and their corresponding molecular signatures. We focused on the biological link between gastric epithelial cells and cancer cells. Notably, we observed that gland mucous cells acquired an intestinal-like stem cell phenotype during metaplasia, with MUC6, MUC2 and OLFM4 emerging as the specific markers for unique endocrine cells in early malignant lesions. Additionally, our analysis highlighted UPP1 as a key oncogene, with its expression progressively increasing from normal epithelial cells to malignant cells. UPP1 upregulation was shown to promote GC cell proliferation and migration, implicating it in the oncogenic process. Further, we explored the impact of Helicobacter pylori infection on gene expression, revealing that Helicobacter pylori infection upregulates UPP1 via the NF-κB pathway. Our cell-cell communication analysis underscored the significant role of the Macrophage migration inhibitory factor pathway in the tumor microenvironment, contributing to GC progression. Various key molecules involved in intestinal metaplasia, along with UPP1 and the Macrophage migration inhibitory factor pathway, collectively illustrate the multifaceted nature and complexity of gastric cancer evolution, highlighting the cumulative impacts that drive tumorigenesis.
Collapse
Affiliation(s)
- Xuyu Chen
- Department of Gastroenterology, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, China
| | - Bengang Zhou
- Dalian Medical University, Dalian, Liaoning, China
| | - Siying Wang
- Department of Oncology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Xin Jiang
- Department of Gastroenterology, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, China
| | - Yukun Ping
- Department of Neurology, Northern Jiangsu People's Hospital, Yangzhou, China
| | - Jianlei Xia
- Department of Gastroenterology, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, China
| | - Feiyu Yu
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, College of Veterinary Medicine, Yangzhou University, Yangzhou, China
| | - Yaoyao Li
- Department of Gastroenterology, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, China.
| | - Min Zhang
- Department of Gastroenterology, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, China.
| | - Yanbing Ding
- Department of Gastroenterology, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, China.
| |
Collapse
|
19
|
Park G, Foster CA, Malone-Perez M, Hasan A, Macias JJ, Frazer JK. Diverse Epithelial Lymphocytes in Zebrafish Revealed Using a Novel Scale Biopsy Method. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2024; 213:1902-1914. [PMID: 39503619 PMCID: PMC11626784 DOI: 10.4049/jimmunol.2300818] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Accepted: 10/09/2024] [Indexed: 11/08/2024]
Abstract
Zebrafish (Danio rerio) are a compelling model for studying lymphocytes because zebrafish and humans have similar adaptive immune systems, including their lymphocytes. Antibodies that recognize zebrafish proteins are sparse, so many investigators use transgenic, lymphocyte-specific fluorophore-labeled lines. Human and zebrafish lymphocyte types are conserved, but many aspects of zebrafish lymphocyte biology remain uninvestigated, including lymphocytes in peripheral tissues, like epidermis. This study is, to our knowledge, the first study to focus on zebrafish epidermal lymphocytes, using scales. Obtaining zebrafish blood via nonlethal methods is difficult; scales represent a source to longitudinally sample live fish. We developed a novel biopsy technique, collecting scales to analyze epithelial lymphocytes from several transgenic lines. We imaged scales via confocal microscopy and demonstrated multiple lymphocyte types in scales/epidermis, quantifying them flow cytometrically. We profiled gene expression of scale, thymic, and kidney-marrow (analogous to mammalian bone marrow) lymphocytes from the same animals, revealing B- and T-lineage signatures. Single-cell quantitative real-time PCR and RNA sequencing show not only canonical B and T cells but also novel lymphocyte populations not described previously. To validate longitudinal scale biopsies, we serially sampled scales from fish treated with dexamethasone, demonstrating epidermal lymphocyte responses. To analyze cells functionally, we employed a bead-ingestion assay, showing that thymic, marrow, and epidermal lymphocytes have phagocytic activity. In summary, we establish a novel, nonlethal technique to obtain zebrafish lymphocytes, providing the first quantification, expression profiling, and functional data from zebrafish epidermal lymphocytes.
Collapse
Affiliation(s)
- Gilseung Park
- Depts. of Cell Biology, University of Oklahoma Health Sciences Center, OK, USA
| | - Clay A. Foster
- Depts. of Pediatrics, Section of Pediatric Hematology-Oncology, University of Oklahoma Health Sciences Center, OK, USA
| | - Megan Malone-Perez
- Depts. of Pediatrics, Section of Pediatric Hematology-Oncology, University of Oklahoma Health Sciences Center, OK, USA
| | - Ameera Hasan
- Depts. of Microbiology & Immunology, University of Oklahoma Health Sciences Center, OK, USA
| | - Jose Juan Macias
- Depts. of Microbiology & Immunology, University of Oklahoma Health Sciences Center, OK, USA
| | - J. Kimble Frazer
- Depts. of Cell Biology, University of Oklahoma Health Sciences Center, OK, USA
- Depts. of Pediatrics, Section of Pediatric Hematology-Oncology, University of Oklahoma Health Sciences Center, OK, USA
- Depts. of Microbiology & Immunology, University of Oklahoma Health Sciences Center, OK, USA
| |
Collapse
|
20
|
Liu H, Da W, Mu J, He X, Li Z, Gong T, Wang J, Min L, Lu M, Tu C. Integration of single-cell and bulk analysis reveals TBXAS1 as a key platelet-related gene causing poor prognosis in osteosarcoma. Front Genet 2024; 15:1519529. [PMID: 39720182 PMCID: PMC11667113 DOI: 10.3389/fgene.2024.1519529] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Accepted: 11/26/2024] [Indexed: 12/26/2024] Open
Abstract
Background Platelets are associated with poor prognosis in most tumors, but their specific pathogenic mechanism in osteosarcoma is not yet clear. The objective of this study is to conduct an in-depth analysis of how genes closely related to platelet function impact the prognosis of osteosarcoma patients. We hope that through this research, we can uncover the potential mechanisms of these genes in the development and progression of osteosarcoma, thereby providing new therapeutic strategies and theoretical foundations for improving the prognosis of osteosarcoma patients. Method We collected the blood routine test data of patients who were initially diagnosed with osteosarcoma at the Department of Bone Tumors, West China Hospital, from January 2012 to January 2022. By applying the LASSO-COX regression analysis, a statistical method, we found that the platelet count is associated with the prognosis of osteosarcoma patients. To further explore this relationship, we obtained single-cell data and bulk RNA data of osteosarcoma patients from the TARGET database and GEO database, respectively. By analyzing these data, we revealed at the transcriptomic level how platelets contribute to the poor prognosis in osteosarcoma patients. Result Platelets are associated with the prognosis of osteosarcoma patients (HR = 3.9, 95% CI = 1.9-8.1, P < 0.001). Through the analysis of transcriptomic data from the TARGET database and GEO database, we found significant heterogeneity in tumor-specific pathways and immune infiltration under different platelet-related gene expression patterns. Among these, TBXAS1 was identified as a key gene that affects the prognosis of osteosarcoma patients. In addition, single-cell data analysis showed that the platelet-related gene TBXAS1 is mainly enriched in macrophages, and markers of macrophages are significantly associated with poor prognosis in osteosarcoma patients. Conclusion TBXAS1 is a key platelet-related gene that leads to poor prognosis in osteosarcoma, and this gene may affect the prognosis of osteosarcoma patients by interacting with macrophages.
Collapse
Affiliation(s)
- Han Liu
- Department of Orthopedics, Orthopaedic Research Institute, West China Hospital, Sichuan University, Chengdu, China
| | - Wacili Da
- Department of Orthopedics Surgery, Orthopeadic Research Institute, West China Hospital, West China Medical School, Sichuan University, Chengdu, Sichuan, China
| | - Jianhua Mu
- Department of Orthopedics, Orthopaedic Research Institute, West China Hospital, Sichuan University, Chengdu, China
| | - Xuanhong He
- Department of Orthopedics, Orthopaedic Research Institute, West China Hospital, Sichuan University, Chengdu, China
| | - Zhuangzhuang Li
- Department of Orthopedics, Orthopaedic Research Institute, West China Hospital, Sichuan University, Chengdu, China
| | - Taojun Gong
- Department of Orthopedics, Orthopaedic Research Institute, West China Hospital, Sichuan University, Chengdu, China
| | - Jingjing Wang
- Department of Endocrine, Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Li Min
- Department of Orthopedics, Orthopaedic Research Institute, West China Hospital, Sichuan University, Chengdu, China
| | - Minxun Lu
- Department of Orthopedics, Orthopaedic Research Institute, West China Hospital, Sichuan University, Chengdu, China
| | - Chongqi Tu
- Department of Orthopedics, Orthopaedic Research Institute, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
21
|
Jin X, Tian Y, Zhu H, Sun Y, Zhang Z. Computer-aided analysis reveals metallothionein-positive cancer-associated fibroblasts promote angiogenesis in gastric adenocarcinoma. Discov Oncol 2024; 15:751. [PMID: 39636347 PMCID: PMC11621267 DOI: 10.1007/s12672-024-01614-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Accepted: 11/21/2024] [Indexed: 12/07/2024] Open
Abstract
Gastric adenocarcinoma (GC), along with its tumor microenvironment (TME), poses great challenges for clinical treatment strategies. Single-cell sequencing has become an important tool for analyzing TME heterogeneity, cell subpopulation, and gene expression patterns. 56 GC single-cell sequencing samples were analyzed, focusing on TME by delineating cancer cells, cancer-associated fibroblasts (CAFs), and macrophages. The spatial transcriptome was used to clarify the distribution characteristics of each cellular component in the tissue slice. Despite the widespread genetic mutations observed in cancer cells, certain recurrent alterations were identified in specific chromosomal regions. The heterogeneity among GC cells is profound, four cancer cell subpopulations were identified through drug sensitivity profiling. Subtype 4, although only present in some samples, demonstrates the strongest stemness and metabolic activity, possibly indicative of an early-stage cancer subpopulation. Their drug sensitivity profiles may hold promise for guiding clinical intervention. In addition, robust spatial co-localization patterns were observed between CAFs, M2 macrophages, and endothelial cells. CAFs were further categorized into six subgroups, among which a novel subgroup termed metallothionein(mt)-positive CAF (mtCAF), characterized by elevated expression of metallothionein 1X (MT1X) and subsequent vascular endothelial growth factor A (VEGFA) secretion, was identified. Immunohistochemistry preliminary confirmed the presence of this unique CAF subgroup. Additionally, M2d macrophages, besides exhibiting high VEGFA expression, also demonstrated various growth factors such as Aamphiregulin (AREG). The M2d-mtCAF axis may play an important role in GC angiogenesis. This study not only enhances our understanding of the TME heterogeneity in GC but also sheds light on the interaction between CAFs and tumor-associated macrophages (TAMs) in tumor angiogenesis.
Collapse
Affiliation(s)
- Xiaolong Jin
- Department of Gastroenterology, Taizhou Central Hospital (Taizhou University Hospital), Taizhou, Zhejiang, China
| | - Yu Tian
- Health Science Center, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Haoran Zhu
- Health Science Center, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Yuewen Sun
- Health Science Center, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Zhenxing Zhang
- Department of Stomatology, Taizhou Central Hospital (Taizhou University Hospital), No. 999, Donghai Avenue, Taizhou, 318000, Zhejiang, People's Republic of China.
| |
Collapse
|
22
|
Wang X, Zhang Y. Multi-omics joint screening of biomarkers related to M2 macrophages in gastric cancer. Discov Oncol 2024; 15:738. [PMID: 39623254 PMCID: PMC11612128 DOI: 10.1007/s12672-024-01623-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Accepted: 11/25/2024] [Indexed: 12/06/2024] Open
Abstract
BACKGROUND Due to high mortality rate and limited treatments in gastric cancer (GC), call for deeper exploration of M2 macrophages as biomarkers is needed. METHODS The data for this study were obtained from the Gene Expression Omnibus (GEO) and Genomic Data Commons (GDC). The Seurat package was utilized for single-cell RNA sequencing (scRNA-seq) analysis. FindAllMarkers was used to identify genes highly expressed among different cell subsets. DESeq2 package was leveraged to screen differentially expressed genes (DEGs), while limma package was utilized for identifying differentially expressed proteins (DEPs). Enrichment analyses of the genes were conducted using KOBAS-i database. MultipleROC was applied to evaluate the diagnostic potential of biomarkers, and rms package was utilized to construct diagnostic models. hTFtarget database was utilized to predict potential transcription factors (TFs). Finally, cell-based assays were performed to validate the expression and potential biological functions of the screened key markers. RESULTS This study found that M2 macrophages were enriched in protein, endoplasmic reticulum, and virus-related pathways. A total of 4146 DEGs and 1946 DEPs were obtained through screening, with 254 common DEGs/DEPs. The results of gene function enrichment analysis suggested that it may affect the occurrence and development of GC through DNA replication and cell cycle. This study identified three biomarkers, HSPH1, HSPD1, and IFI30, and constructed a diagnostic model based on these three genes. The AUC value greater than 0.8 proved the reliability of the model. Through screening TFs, SPI1 and KLF5 were found to be the common TFs for the three biomarkers. The expression of the three genes IFI30, HSPD1 and HSPH1 was up-regulated in GC cells, and IFI30 may play a facilitating role in the migration and invasion of GC cells. CONCLUSION This study identified three biomarkers and constructed a diagnostic model, providing a new perspective for the research and treatment of GC.
Collapse
Affiliation(s)
- Xilong Wang
- Tumor Hematology Department, Liaoyang Central Hospital, Liaoyang, 111000, China
| | - Ying Zhang
- General Surgery Department, Liaoyang Central Hospital, Liaoyang, 111000, China.
| |
Collapse
|
23
|
Zhang S, Fang X, Chang M, Zheng M, Guo L, Xu Y, Shu J, Nie Q, Li Z. Cross-species single-cell analysis reveals divergence and conservation of peripheral blood mononuclear cells. BMC Genomics 2024; 25:1169. [PMID: 39623297 PMCID: PMC11613757 DOI: 10.1186/s12864-024-11030-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2024] [Accepted: 11/11/2024] [Indexed: 12/06/2024] Open
Abstract
BACKGROUND Single-cell transcriptome sequencing (scRNA-seq) has revolutionized the study of immune cells by overcoming the limitations of traditional antibody-based identification and isolation methods. This advancement allows us to obtain comprehensive gene expression profiles from a diverse array of vertebrate species, facilitating the identification of various cell types. Comparative immunology across vertebrates presents a promising approach to understanding the evolution of immune cell types. In this study, we conducted a comparative transcriptome analysis of peripheral blood mononuclear cells (PBMCs) at the single-cell level across 12 species. RESULTS Our findings shed light on the cellular compositional features of PBMCs, spanning from fish to mammals. Notably, we identified genes that exhibit vertebrate universality in characterizing immune cells. Moreover, our investigation revealed that monocytes have maintained a conserved transcriptional regulatory program throughout evolution, emphasizing their pivotal role in orchestrating immune cells to execute immune programs. CONCLUSIONS This comprehensive analysis provides valuable insights into the evolution of immune cells across vertebrates.
Collapse
Affiliation(s)
- Siyu Zhang
- State Key Laboratory of Swine and Poultry Breeding Industry, Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong, 510642, China
- Key Laboratory of Chicken Genetics, Breeding and Reproduction, Ministry of Agriculture and Rural Affair, South China Agricultural University, Guangzhou, Guangdong, 510642, China
| | - Xiang Fang
- Department of Infectious Diseases and Public Health, Jockey Club College of Veterinary Medicine and Life Sciences, City University of Hong Kong, Hong Kong, 999077, China
| | - Mengyang Chang
- Institute of Aquatic Biotechnology, College of Life Sciences, Qingdao University, Qingdao, Liaoning, 266071, China
| | - Ming Zheng
- State Key Laboratory of Swine and Poultry Breeding Industry, Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong, 510642, China
- Key Laboratory of Chicken Genetics, Breeding and Reproduction, Ministry of Agriculture and Rural Affair, South China Agricultural University, Guangzhou, Guangdong, 510642, China
| | - Lijin Guo
- State Key Laboratory of Swine and Poultry Breeding Industry, Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong, 510642, China
- Key Laboratory of Chicken Genetics, Breeding and Reproduction, Ministry of Agriculture and Rural Affair, South China Agricultural University, Guangzhou, Guangdong, 510642, China
| | - Yibin Xu
- State Key Laboratory of Swine and Poultry Breeding Industry, Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong, 510642, China
- Key Laboratory of Chicken Genetics, Breeding and Reproduction, Ministry of Agriculture and Rural Affair, South China Agricultural University, Guangzhou, Guangdong, 510642, China
| | - Jingting Shu
- Key Laboratory for Poultry Genetics and Breeding of Jiangsu Province, Jiangsu Institute of Poultry Science, Yangzhou, Jiangsu, 225125, China.
| | - Qinghua Nie
- State Key Laboratory of Swine and Poultry Breeding Industry, Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong, 510642, China.
- Key Laboratory of Chicken Genetics, Breeding and Reproduction, Ministry of Agriculture and Rural Affair, South China Agricultural University, Guangzhou, Guangdong, 510642, China.
| | - Zhenhui Li
- State Key Laboratory of Swine and Poultry Breeding Industry, Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong, 510642, China.
- Key Laboratory of Chicken Genetics, Breeding and Reproduction, Ministry of Agriculture and Rural Affair, South China Agricultural University, Guangzhou, Guangdong, 510642, China.
| |
Collapse
|
24
|
Yang CX, Sin DD, Ng RT. SMART: spatial transcriptomics deconvolution using marker-gene-assisted topic model. Genome Biol 2024; 25:304. [PMID: 39623485 PMCID: PMC11610197 DOI: 10.1186/s13059-024-03441-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Accepted: 11/20/2024] [Indexed: 12/06/2024] Open
Abstract
While spatial transcriptomics offer valuable insights into gene expression patterns within the spatial context of tissue, many technologies do not have a single-cell resolution. Here, we present SMART, a marker gene-assisted deconvolution method that simultaneously infers the cell type-specific gene expression profile and the cellular composition at each spot. Using multiple datasets, we show that SMART outperforms the existing methods in realistic settings. It also provides a two-stage approach to enhance its performance on cell subtypes. The covariate model of SMART enables the identification of cell type-specific differentially expressed genes across conditions, elucidating biological changes at a single-cell-type resolution.
Collapse
Affiliation(s)
- Chen Xi Yang
- Centre for Heart Lung Innovation, St. Paul's Hospital, University of British Columbia, Vancouver, BC, Canada.
- Department of Bioinformatics, Faculty of Science, University of British Columbia, Vancouver, BC, Canada.
| | - Don D Sin
- Centre for Heart Lung Innovation, St. Paul's Hospital, University of British Columbia, Vancouver, BC, Canada
- Division of Respiratory Medicine, Department of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Raymond T Ng
- Centre for Heart Lung Innovation, St. Paul's Hospital, University of British Columbia, Vancouver, BC, Canada
- Department of Bioinformatics, Faculty of Science, University of British Columbia, Vancouver, BC, Canada
- Department of Computer Science, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
25
|
Mondal S, Becskei A. Gene choice in cancer cells is exclusive in ion transport but concurrent in DNA replication. Comput Struct Biotechnol J 2024; 23:2534-2547. [PMID: 38974885 PMCID: PMC11226983 DOI: 10.1016/j.csbj.2024.06.004] [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: 02/29/2024] [Revised: 06/04/2024] [Accepted: 06/04/2024] [Indexed: 07/09/2024] Open
Abstract
Cancers share common cellular and physiological features. Little is known about whether distinctive gene expression patterns can be displayed at the single-cell level by gene families in cancer cells. The expression of gene homologs within a family can exhibit concurrence and exclusivity. Concurrence can promote all-or-none expression patterns of related genes and underlie alternative physiological states. Conversely, exclusive gene families express the same or similar number of homologs in each cell, allowing a broad repertoire of cell identities to be generated. We show that gene families involved in the cell-cycle and antigen presentation are expressed concurrently. Concurrence in the DNA replication complex MCM reflects the replicative status of cells, including cell lines and cancer-derived organoids. Exclusive expression requires precise regulatory mechanism, but cancer cells retain this form of control for ion homeostasis and extend it to gene families involved in cell migration. Thus, the cell adhesion-based identity of healthy cells is transformed to an identity based on migration in the population of cancer cells, reminiscent of epithelial-mesenchymal transition.
Collapse
Affiliation(s)
- Samuel Mondal
- Biozentrum, University of Basel, Spitalstrasse 41, Basel 4056, Switzerland
| | - Attila Becskei
- Biozentrum, University of Basel, Spitalstrasse 41, Basel 4056, Switzerland
| |
Collapse
|
26
|
Salihoglu R, Balkenhol J, Dandekar G, Liang C, Dandekar T, Bencurova E. Cat-E: A comprehensive web tool for exploring cancer targeting strategies. Comput Struct Biotechnol J 2024; 23:1376-1386. [PMID: 38596315 PMCID: PMC11001601 DOI: 10.1016/j.csbj.2024.03.024] [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: 01/27/2024] [Revised: 03/26/2024] [Accepted: 03/26/2024] [Indexed: 04/11/2024] Open
Abstract
Identifying potential cancer-associated genes and drug targets from omics data is challenging due to its diverse sources and analyses, requiring advanced skills and large amounts of time. To facilitate such analysis, we developed Cat-E (Cancer Target Explorer), a novel R/Shiny web tool designed for comprehensive analysis with evaluation according to cancer-related omics data. Cat-E is accessible at https://cat-e.bioinfo-wuerz.eu/. Cat-E compiles information on oncolytic viruses, cell lines, gene markers, and clinical studies by integrating molecular datasets from key databases such as OvirusTB, TCGA, DrugBANK, and PubChem. Users can use all datasets and upload their data to perform multiple analyses, such as differential gene expression analysis, metabolic pathway exploration, metabolic flux analysis, GO and KEGG enrichment analysis, survival analysis, immune signature analysis, single nucleotide variation analysis, dynamic analysis of gene expression changes and gene regulatory network changes, and protein structure prediction. Cancer target evaluation by Cat-E is demonstrated here on lung adenocarcinoma (LUAD) datasets. By offering a user-friendly interface and detailed user manual, Cat-E eliminates the need for advanced computational expertise, making it accessible to experimental biologists, undergraduate and graduate students, and oncology clinicians. It serves as a valuable tool for investigating genetic variations across diverse cancer types, facilitating the identification of novel diagnostic markers and potential therapeutic targets.
Collapse
Affiliation(s)
- Rana Salihoglu
- Department of Bioinformatics, University of Wurzburg, 97074 Wurzburg, Germany
| | - Johannes Balkenhol
- Department of Bioinformatics, University of Wurzburg, 97074 Wurzburg, Germany
- Rudolf Virchow Center for Integrative and Translational Bioimaging, University Hospital of Wurzburg, 97080 Wurzburg, Germany
| | - Gudrun Dandekar
- Chair of Tissue Engineering and Regenerative Medicine, University Hospital of Wurzburg, 97080 Wurzburg, Germany
| | - Chunguang Liang
- Department of Bioinformatics, University of Wurzburg, 97074 Wurzburg, Germany
- Institute of Immunology, Jena University Hospital, Friedrich-Schiller-University, 07743 Jena, Germany
| | - Thomas Dandekar
- Department of Bioinformatics, University of Wurzburg, 97074 Wurzburg, Germany
- Structural and Computational Biology Unit, European Molecular Biology Laboratory, 69117 Heidelberg, Germany
| | - Elena Bencurova
- Department of Bioinformatics, University of Wurzburg, 97074 Wurzburg, Germany
| |
Collapse
|
27
|
Zhong D, Liao Y, Chen W, Huang X, Liu J, Wang Z. TYROBP promotes the spread of pancreatic cancer by causing M2 TAM polarization. J Gastroenterol Hepatol 2024; 39:2926-2939. [PMID: 39496400 DOI: 10.1111/jgh.16783] [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: 08/07/2024] [Revised: 10/04/2024] [Accepted: 10/13/2024] [Indexed: 11/06/2024]
Abstract
BACKGROUND AND AIM M2-polarized tumor-associated macrophages (M2 TAMs) are known to promote cancer progression, and exosomes are crucial mediators of communication within the tumor microenvironment (TME). However, the specific role of exosomes derived from M2 TAMs in pancreatic cancer (PC) progression remains poorly understood. Tyrosine kinase binding protein (TYROBP, also known as DAP12 for DNAX activating protein-12) is a transmembrane signal transduction polypeptide that interacts with immune cell receptors, influencing cellular functions via signal transduction pathways. TYROBP is prominently found in M2 TAMs exosomes, facilitating its transfer to PC cells and suggesting a potential role in PC pathogenesis. METHODS This study initially confirmed the presence of TYROBP in M2 TAMs exosomes and its transfer to PC cells via exosomes. The impact of TYROBP on PC proliferation, apoptosis, migration, and invasion was investigated. Special attention was given to TYROBP's influence on PC metastasis and its underlying mechanisms, focusing particularly on the CD44/AKT/ERK signaling pathway. RESULTS TYROBP expression in PC cells did not significantly affect tumor cell proliferation or apoptosis but demonstrated a notable inhibitory effect on migration and invasion, which was mediated through the CD44/AKT/ERK pathway. Both in vivo and in vitro experiments consistently showed that TYROBP enhanced PC metastasis. CONCLUSIONS This study elucidates that TYROBP plays a direct role in promoting PC metastasis through its association with M2 TAMs polarization. Therefore, TYROBP represents a potential novel therapeutic target for interventions aimed at combatting PC progression.
Collapse
Affiliation(s)
- Dingwen Zhong
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
- Pancreas Treatment and Therapy Center of Xi'an Jiaotong University, Xi'an, China
- Department of Hepatobiliary and Pancreatic Surgery, Ganzhou People's Hospital, Ganzhou, Jiangxi, China
| | - Yonghui Liao
- Department of Hepatobiliary and Pancreatic Surgery, Ganzhou People's Hospital, Ganzhou, Jiangxi, China
| | - Wenhui Chen
- Department of Hepatobiliary and Pancreatic Surgery, Ganzhou People's Hospital, Ganzhou, Jiangxi, China
| | - Xianyu Huang
- Department of Hepatobiliary and Pancreatic Surgery, Ganzhou People's Hospital, Ganzhou, Jiangxi, China
| | - Jiaxin Liu
- Department of Hepatobiliary and Pancreatic Surgery, Ganzhou People's Hospital, Ganzhou, Jiangxi, China
| | - Zheng Wang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
- Pancreas Treatment and Therapy Center of Xi'an Jiaotong University, Xi'an, China
| |
Collapse
|
28
|
Yi L, Guo X, Liu Y, Jirimutu, Wang Z. Single-cell 5' RNA sequencing of camelid peripheral B cells provides insights into cellular basis of heavy-chain antibody production. Comput Struct Biotechnol J 2024; 23:1705-1714. [PMID: 38689719 PMCID: PMC11059136 DOI: 10.1016/j.csbj.2024.04.041] [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: 01/19/2024] [Revised: 04/15/2024] [Accepted: 04/15/2024] [Indexed: 05/02/2024] Open
Abstract
Camelids produce both conventional tetrameric antibodies (Abs) and dimeric heavy-chain antibodies (HCAbs). Although B cells that generate these two types of Abs exhibit distinct B cell receptors (BCRs), whether these two B cell populations differ in their phenotypes and developmental processes remains unclear. Here, we performed single-cell 5' RNA profiling of peripheral blood mononuclear cell samples from Bactrian camels before and after immunization. We characterized the functional subtypes and differentiation trajectories of circulating B cells in camels, and reconstructed single-cell BCR sequences. We found that in contrast to humans, the proportion of T-bet+ B cells was high among camelid peripheral B cells. Several marker genes of human B cell subtypes, including CD27 and IGHD, were expressed at low levels in the corresponding camel B cell subtypes. Camelid B cells expressing variable genes of HACbs (VHH) were widely present in various functional subtypes and showed highly overlapping differentiation trajectories with B cells expressing variable genes of conventional Abs (VH). After immunization, the transcriptional changes in VHH+ and VH+ B cells were largely consistent. Through structure modeling, we identified a variety of scaffold types among the reconstructed VHH sequences. Our study provides insights into the cellular context of HCAb production in camels and lays the foundation for developing single-B cell-based camelid single-domain Ab screening.
Collapse
Affiliation(s)
- Li Yi
- Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education, College of Food Science and Engineering, Inner Mongolia Agricultural University, Huhhot 010018, China
| | - Xin Guo
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Yuexing Liu
- Guangzhou Laboratory, Guangzhou 510005, China
| | - Jirimutu
- Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education, College of Food Science and Engineering, Inner Mongolia Agricultural University, Huhhot 010018, China
- Inner Mongolia China-Kazakhstan Camel Research Institute, Alxa 750306, China
| | - Zhen Wang
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| |
Collapse
|
29
|
Chi Y, Marini S, Wang GZ. BrainCellR: A precise cell type nomenclature pipeline for comparative analysis across brain single-cell datasets. Comput Struct Biotechnol J 2024; 23:4306-4314. [PMID: 39687760 PMCID: PMC11648093 DOI: 10.1016/j.csbj.2024.11.038] [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: 07/02/2024] [Revised: 11/24/2024] [Accepted: 11/25/2024] [Indexed: 12/18/2024] Open
Abstract
Single-cell studies in neuroscience require precise cell type classification and consistent nomenclature that allows for meaningful comparisons across diverse datasets. Current approaches often lack the ability to identify fine-grained cell types and establish standardized annotations at the cluster level, hindering comprehensive understanding of the brain's cellular composition. To facilitate data integration across multiple models and datasets, we designed BrainCellR. This pipeline provides researchers with a powerful and user-friendly tool for efficient cell type classification and nomination from single-cell transcriptomic data. While initially focused on brain studies, BrainCellR is applicable to other tissues with complex cellular compositions. BrainCellR goes beyond conventional classification approaches by incorporating a standardized nomenclature system for cell types at the cluster level. This feature enables consistent and comparable annotations across different studies, promoting data integration and providing deeper insights into the complex cellular landscape of the brain. All documents for BrainCellR, including source code, user manual and tutorials, are freely available at https://github.com/WangLab-SINH/BrainCellR.
Collapse
Affiliation(s)
- Yuhao Chi
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Simone Marini
- Department of Epidemiology, University of Florida, Gainesville, FL, USA
| | - Guang-Zhong Wang
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| |
Collapse
|
30
|
Zhao Y, Yu ZM, Cui T, Li LD, Li YY, Qian FC, Zhou LW, Li Y, Fang QL, Huang XM, Zhang QY, Cai FH, Dong FJ, Shang DS, Li CQ, Wang QY. scBlood: A comprehensive single-cell accessible chromatin database of blood cells. Comput Struct Biotechnol J 2024; 23:2746-2753. [PMID: 39050785 PMCID: PMC11266868 DOI: 10.1016/j.csbj.2024.06.015] [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: 04/16/2024] [Revised: 06/17/2024] [Accepted: 06/18/2024] [Indexed: 07/27/2024] Open
Abstract
The advent of single cell transposase-accessible chromatin sequencing (scATAC-seq) technology enables us to explore the genomic characteristics and chromatin accessibility of blood cells at the single-cell level. To fully make sense of the roles and regulatory complexities of blood cells, it is critical to collect and analyze these rapidly accumulating scATAC-seq datasets at a system level. Here, we present scBlood (https://bio.liclab.net/scBlood/), a comprehensive single-cell accessible chromatin database of blood cells. The current version of scBlood catalogs 770,907 blood cells and 452,247 non-blood cells from ∼400 high-quality scATAC-seq samples covering 30 tissues and 21 disease types. All data hosted on scBlood have undergone preprocessing from raw fastq files and multiple standards of quality control. Furthermore, we conducted comprehensive downstream analyses, including multi-sample integration analysis, cell clustering and annotation, differential chromatin accessibility analysis, functional enrichment analysis, co-accessibility analysis, gene activity score calculation, and transcription factor (TF) enrichment analysis. In summary, scBlood provides a user-friendly interface for searching, browsing, analyzing, visualizing, and downloading scATAC-seq data of interest. This platform facilitates insights into the functions and regulatory mechanisms of blood cells, as well as their involvement in blood-related diseases.
Collapse
Affiliation(s)
- Yu Zhao
- The First Affiliated Hospital & MOE Key Lab of Rare Pediatric Diseases, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
- School of Computer, University of South China, Hengyang, Hunan 421001, China
| | - Zheng-Min Yu
- School of Computer, University of South China, Hengyang, Hunan 421001, China
| | - Ting Cui
- The First Affiliated Hospital & MOE Key Lab of Rare Pediatric Diseases, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - Li-Dong Li
- School of Computer, University of South China, Hengyang, Hunan 421001, China
| | - Yan-Yu Li
- School of Medical Informatics, Daqing Campus, Harbin Medical University, Daqing 163319, China
| | - Feng-Cui Qian
- The First Affiliated Hospital, Cardiovascular Lab of Big Data and Imaging Artificial Intelligence, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - Li-Wei Zhou
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Ye Li
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - Qiao-Li Fang
- School of Computer, University of South China, Hengyang, Hunan 421001, China
| | - Xue-Mei Huang
- School of Computer, University of South China, Hengyang, Hunan 421001, China
| | - Qin-Yi Zhang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - Fu-Hong Cai
- School of Computer, University of South China, Hengyang, Hunan 421001, China
| | - Fu-Juan Dong
- School of Computer, University of South China, Hengyang, Hunan 421001, China
| | - De-Si Shang
- The First Affiliated Hospital, Cardiovascular Lab of Big Data and Imaging Artificial Intelligence, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - Chun-Quan Li
- The First Affiliated Hospital & MOE Key Lab of Rare Pediatric Diseases, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
- School of Computer, University of South China, Hengyang, Hunan 421001, China
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
- Hunan Provincial Key Laboratory of Multi-omics And Artificial Intelligence of Cardiovascular Diseases, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
- Hunan Provincial Maternal and Child Health Care Hospital, National Health Commission Key Laboratory of Birth Defect Research and Prevention, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
- The First Affiliated Hospital, Cardiovascular Lab of Big Data and Imaging Artificial Intelligence, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
- The First Affiliated Hospital, Institute of Cardiovascular Disease, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - Qiu-Yu Wang
- School of Computer, University of South China, Hengyang, Hunan 421001, China
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
- Hunan Provincial Key Laboratory of Multi-omics And Artificial Intelligence of Cardiovascular Diseases, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
- Hunan Provincial Maternal and Child Health Care Hospital, National Health Commission Key Laboratory of Birth Defect Research and Prevention, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| |
Collapse
|
31
|
Shao L, Chang Y, Liu J, Lin L, Chang L, Zhang J, Lan Z, Zhang H, Chen X. scRNA-Seq reveals age-dependent microglial evolution as a determinant of immune response following spinal cord injury. Brain Res Bull 2024; 219:111116. [PMID: 39515654 DOI: 10.1016/j.brainresbull.2024.111116] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Revised: 10/24/2024] [Accepted: 11/05/2024] [Indexed: 11/16/2024]
Abstract
Spinal cord injury (SCI) is a debilitating condition of the central nervous system (CNS) that leads to severe impairments in sensory and motor functions. Previous studies have pointed out that patient age is a critical factor influencing SCI prognosis. However, the role of microglia in age-related differences in SCI outcomes remains unclear. The current study aims to identify specific microglial subtypes and investigate their responses and functional differences in SCI recovery across different age groups. Single-cell RNA sequencing (scRNA-seq) data were obtained from the Gene Expression Omnibus (GEO) database, integrating multiple datasets to identify microglial subtypes. We performed pseudotime trajectory analysis and cell-cell communication analysis to understand microglial differentiation and interactions. Finally, immunofluorescence staining of mouse model samples was conducted to validate our bioinformatics findings. Microglia were classified into four subtypes: Homeostatic, Proliferating, Inflammatory A, and Inflammatory B. The Young SCI group exhibited a higher proportion of Homeostatic microglia and Inflammatory microglia A, whereas the old SCI group had more Inflammatory Microglia B but lacked Homeostatic Microglia. Gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analyses revealed that markers for homeostasis microglia were enriched in immune modulation pathways. While makers for Inflammatory Microglia were enriched in immune response pathways. Specifically, markers for Inflammatory microglia B were enriched in pathways associated with overactive immune response. Pseudotime analysis indicated that microglia in young mice predominantly differentiated into Inflammatory Microglia A and Homeostatic Microglia, whereas in old mice, they tended to only differentiate into Inflammatory Microglia B. CellChat analysis showed increased pro-inflammatory signaling generated by Inflammatory Microglia B, exclusively in the old group. Our study demonstrates significant differences in microglial subtypes and functions between different age groups following SCI. These findings provide novel insights into the development of age-related therapeutic strategies and microglia-targeted biological treatments for SCI.
Collapse
Affiliation(s)
- Lufei Shao
- Neurology Department, General Hospital of Ningxia Medical University, Yinchuan 750004, China; Ningxia nervous system disease Diagnosis and treatment Engineering Technology Research center, Yinchuan 750004, China
| | - Yueliang Chang
- Neurology Department, General Hospital of Ningxia Medical University, Yinchuan 750004, China
| | - Jinfang Liu
- Neurology Department, General Hospital of Ningxia Medical University, Yinchuan 750004, China
| | - Leilei Lin
- Orthopedics Department, General Hospital of Ningxia Medical University, Yinchuan 750004, China
| | - Long Chang
- Orthopedics Department, General Hospital of Ningxia Medical University, Yinchuan 750004, China
| | - Jialin Zhang
- Orthopedics Department, General Hospital of Ningxia Medical University, Yinchuan 750004, China
| | - Zhibin Lan
- Ningxia Key Laboratory of Clinical and Pathogenic Microbiology, General Hospital of Ningxia Medical University, Yinchuan 750004, China
| | - Honglai Zhang
- Ningxia Key Laboratory of Clinical and Pathogenic Microbiology, General Hospital of Ningxia Medical University, Yinchuan 750004, China
| | - Xiaolei Chen
- Orthopedics Department, General Hospital of Ningxia Medical University, Yinchuan 750004, China; Ningxia Key Laboratory of Clinical and Pathogenic Microbiology, General Hospital of Ningxia Medical University, Yinchuan 750004, China.
| |
Collapse
|
32
|
Zhai R, Chang L, Jiang J, Wang B, Zhu W. Cellular and Molecular Basis of Environment-Induced Color Change in a Tree Frog. Animals (Basel) 2024; 14:3472. [PMID: 39682437 DOI: 10.3390/ani14233472] [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: 10/22/2024] [Revised: 11/19/2024] [Accepted: 11/28/2024] [Indexed: 12/18/2024] Open
Abstract
Background color matching is essential for camouflage and thermoregulation in ectothermic vertebrates, yet several key cellular-level questions remain unresolved. For instance, it is unclear whether the number of chromatophores or the activity of individual chromatophores plays a more critical role in this process. Using single-cell RNA sequencing (scRNA-seq), we investigated the cellular and molecular mechanisms underlying color change in Rhacophorus dugritei, which adapted to its background by displaying light-green skin on white and black skin on black within two days. We identified two types of chromatophores in their skin, both responsible for the observed color differences. Our findings reveal that morphological color change (MCC) is the dominant process, with the number of chromatophores being more influential in driving color change than the transcriptional activity of melanogenesis in individual cells. Additionally, melanophores from darker individuals exhibited increased activity in energy metabolism pathways, while those from lighter individuals showed stronger immune-related gene expression, suggesting that background adaptation involves more than just morphological changes. Overall, this study successfully applied single-cell sequencing technology to investigate skin pigmentation in a non-model organism. Our results suggest that MCC driven by chromatophore proliferation is a key mechanism of background adaptation, offering new insights into amphibian color adaptation and environmental adaptation in other vertebrates.
Collapse
Affiliation(s)
- Runliang Zhai
- Chengdu Institute of Biology, Chinese Academy of Sciences, Chengdu 610213, China
- University of Chinese Academy of Sciences, Beijing 101408, China
| | - Liming Chang
- Chengdu Institute of Biology, Chinese Academy of Sciences, Chengdu 610213, China
| | - Jianping Jiang
- Chengdu Institute of Biology, Chinese Academy of Sciences, Chengdu 610213, China
- University of Chinese Academy of Sciences, Beijing 101408, China
| | - Bin Wang
- Chengdu Institute of Biology, Chinese Academy of Sciences, Chengdu 610213, China
- University of Chinese Academy of Sciences, Beijing 101408, China
| | - Wei Zhu
- Chengdu Institute of Biology, Chinese Academy of Sciences, Chengdu 610213, China
- University of Chinese Academy of Sciences, Beijing 101408, China
| |
Collapse
|
33
|
Liu J, Luo Q, Zhao H, Yang M, Yang J, Wang Y, Zhao M, Mao J, Chen J, Guo B, Zhang L. Comprehensive gene set enrichment and variation analyses identify SUV39H1 as a potential prognostic biomarker for glioblastoma immunorelevance. Comput Struct Biotechnol J 2024; 23:4161-4176. [PMID: 39640533 PMCID: PMC11617780 DOI: 10.1016/j.csbj.2024.11.016] [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: 07/10/2024] [Revised: 10/25/2024] [Accepted: 11/05/2024] [Indexed: 12/07/2024] Open
Abstract
Glioblastoma (GBM) is the most common intracranial malignancy. SUV39H1 encodes a histone H3 lysine 9 methyltransferase that acts as an oncogene in several cancers; however, its role in GBM remains unknown. We obtained GBM transcriptome and clinical data from The Cancer Genome Atlas (TCGA) database on the UCSC Xena platform to perform differential and enrichment analyses of genes in the SUV39H1 high- and low-expression groups to construct a prognostic risk model. Analysis of SUV39H1 related biological processes in GBM was performed by gene set enrichment analysis (GSEA) and gene set variation analysis (GSVA). High- and low-risk subgroup mutation signatures were analyzed using maftools. Immune infiltration was evaluated using IOBR and CIBERSORT algorithms. We analyzed the cell types and intercellular communication networks in glioma stem cells (GSCs) using scRNA-seq. The effects on GBM cells and GSCs after inhibition of SUV39H1 were investigated in vitro. SUV39H1 was significantly overexpressed in GBM and associated with poor prognosis. SUV39H1-related differentially expressed genes were enriched in immune and inflammation related pathways, and GSEA revealed that these genes were significantly enriched in signaling pathways such as IL-18, oxidative phosphorylation, and regulation of TP53 activity. Mutational analysis revealed frequent alterations in TP53 and PTEN expression. In addition, the infiltration abundances of the five immune cell types were significantly different between the high- and low-expression groups. Analysis of cellular communication networks by scRNA-seq revealed a strong interaction between CRYAB-GSC and PTPRZ1-GSC in GSCs. In vitro experiments verified that knockdown of SUV39H1 inhibited the viability and proliferation of U87 and U251 glioblastoma cells and downregulated the expression of stemness markers Nestin and SOX2 in CSC1589 and TS576 GSC lines. Increased SUV39H1 expression is associated with immune cell infiltration and poor prognosis in patients with GBM. Inhibition of SUV39H1 restrains GBM growth and reduces the stem cell properties of GSC. Thus, SUV39H1 might be a prognostic predictor and immunotherapeutic target in patients with GBM.
Collapse
Affiliation(s)
- Jixuan Liu
- Key Laboratory of Pathobiology, Ministry of Education, Department of Biomedical Science, College of Basic Medical Sciences, Jilin University, Changchun, China
| | - Qian Luo
- Key Laboratory of Pathobiology, Ministry of Education, Department of Biomedical Science, College of Basic Medical Sciences, Jilin University, Changchun, China
| | - Haoran Zhao
- Key Laboratory of Pathobiology, Ministry of Education, Department of Biomedical Science, College of Basic Medical Sciences, Jilin University, Changchun, China
| | - Mei Yang
- Key Laboratory of Pathobiology, Ministry of Education, Department of Biomedical Science, College of Basic Medical Sciences, Jilin University, Changchun, China
| | - Jiaying Yang
- Key Laboratory of Pathobiology, Ministry of Education, Department of Biomedical Science, College of Basic Medical Sciences, Jilin University, Changchun, China
| | - Yingtong Wang
- The Undergraduate Center of Hospital of Stomatology, Jilin University, Changchun 130021, China
| | - Mengxin Zhao
- Key Laboratory of Pathobiology, Ministry of Education, Department of Biomedical Science, College of Basic Medical Sciences, Jilin University, Changchun, China
| | - Juanjuan Mao
- Key Laboratory of Pathobiology, Ministry of Education, Department of Biomedical Science, College of Basic Medical Sciences, Jilin University, Changchun, China
| | - Jiasi Chen
- Key Laboratory of Pathobiology, Ministry of Education, Department of Biomedical Science, College of Basic Medical Sciences, Jilin University, Changchun, China
| | - Baofeng Guo
- Department of Plastic Surgery, China-Japan Union Hospital, Jilin University, Changchun 130033, China
| | - Ling Zhang
- Key Laboratory of Pathobiology, Ministry of Education, Department of Biomedical Science, College of Basic Medical Sciences, Jilin University, Changchun, China
| |
Collapse
|
34
|
Jie L, Zhang C, Liu Y, Huang Z, Xu B, Zhu Z, Li Y, Wang P, Shi X. Mechanistic study of the regulation of mitochondrial function by the GPNMB/Nrf2/NF-κB signaling pathway mediated by Quzhi Tang to alleviate chondrocyte senescence. JOURNAL OF ETHNOPHARMACOLOGY 2024; 340:119165. [PMID: 39617085 DOI: 10.1016/j.jep.2024.119165] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Revised: 11/03/2024] [Accepted: 11/25/2024] [Indexed: 12/07/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Quzhi Tang (QZT) is a compound formula consisting of six traditional Chinese medicinal herbs. It has achieved good clinical results in the treatment of knee osteoarthritis (KOA), and the potential drug mechanisms involved are worth exploring in depth. MATERIALS AND METHODS Using single-cell transcriptome analysis, this study identified the key target of senescence, GPNMB. Then, it investigated the mechanism by which QZT regulates the GPNMB/Nrf2/NF-κB signaling pathway to repair mitochondrial damage and ameliorate the process of chondrocyte senescence. RESULTS We collected cartilage tissues from mice and identified GPNMB as a key target of chondrocyte senescence by combining transcriptomics, histopathology, molecular biology, and immunology methods. The effects of QZT on the level of chondrocyte senescence in mice and its ameliorative effect on KOA were studied. In in vivo experiments, we explored the mechanism of GPNMB in the development of senescence in detail and revealed that, after siRNA-GPNMB interference, chondrocytes exhibited reduced impairment of mitochondrial function and senescence under equal amounts of stimuli, increasing Nrf2 expression and reducing NF-κB expression. In addition, the level of oxidative stress increased in chondrocytes overexpressing GPNMB after lentiviral infiltration, aggravating the impairment of mitochondrial function. After treatment with QZT, chondrocytes overexpressing GPNMB were able to increase Nrf2 expression, decrease NF-κB expression, repair mitochondrial damage, and improve the degree of chondrocyte aging. CONCLUSION We concluded that the GPNMB/Nrf2/NF-κB signaling pathway plays an important role in chondrocyte senescence and that QZT was able to reduce intracellular oxidative stress and restore impaired mitochondrial function by regulating the expression level of the GPNMB/Nrf2/NF-κB signaling pathway, reducing the level of chondrocyte senescence in the KOA process.
Collapse
Affiliation(s)
- Lishi Jie
- Department of Orthopaedics and Traumatology, Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Provincial Hospital of Traditional Chinese Medicine, Nanjing, 210023, China; Key Laboratory for Metabolic Diseases in Chinese Medicine, First College of Clinical Medicine, Nanjing University of Chinese Medicine, Jiangsu Provincial Hospital of Traditional Chinese Medicine, Nanjing, 210023, China
| | - Chaofeng Zhang
- Department of Orthopaedics and Traumatology, Suzhou Hospital of Traditional Chinese Medicine, Suzhou, China
| | - Yujiang Liu
- Department of Orthopaedics and Traumatology, Suzhou Hospital of Traditional Chinese Medicine, Suzhou, China
| | - Zeling Huang
- Department of Orthopaedics and Traumatology, Suzhou Hospital of Traditional Chinese Medicine, Suzhou, China; Key Laboratory for Metabolic Diseases in Chinese Medicine, First College of Clinical Medicine, Nanjing University of Chinese Medicine, Jiangsu Provincial Hospital of Traditional Chinese Medicine, Nanjing, 210023, China
| | - Bo Xu
- Department of Orthopaedics and Traumatology, Suzhou Hospital of Traditional Chinese Medicine, Suzhou, China
| | - Zaishi Zhu
- Department of Orthopaedics and Traumatology, Suzhou Hospital of Traditional Chinese Medicine, Suzhou, China; Key Laboratory for Metabolic Diseases in Chinese Medicine, First College of Clinical Medicine, Nanjing University of Chinese Medicine, Jiangsu Provincial Hospital of Traditional Chinese Medicine, Nanjing, 210023, China
| | - Yuwei Li
- Department of Orthopaedics and Traumatology, Suzhou Hospital of Traditional Chinese Medicine, Suzhou, China
| | - Peimin Wang
- Department of Orthopaedics and Traumatology, Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Provincial Hospital of Traditional Chinese Medicine, Nanjing, 210023, China; Jiangsu Provincial Engineering Research Center of TCM External Medication Development and Application, Nanjing, China.
| | - Xiaoqing Shi
- Department of Orthopaedics and Traumatology, Suzhou Hospital of Traditional Chinese Medicine, Suzhou, China.
| |
Collapse
|
35
|
Hu M, Alkhairy S, Lee I, Pillich RT, Fong D, Smith K, Bachelder R, Ideker T, Pratt D. Evaluation of large language models for discovery of gene set function. Nat Methods 2024:10.1038/s41592-024-02525-x. [PMID: 39609565 DOI: 10.1038/s41592-024-02525-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Accepted: 10/21/2024] [Indexed: 11/30/2024]
Abstract
Gene set enrichment is a mainstay of functional genomics, but it relies on gene function databases that are incomplete. Here we evaluate five large language models (LLMs) for their ability to discover the common functions represented by a gene set, supported by molecular rationale and a self-confidence assessment. For curated gene sets from Gene Ontology, GPT-4 suggests functions similar to the curated name in 73% of cases, with higher self-confidence predicting higher similarity. Conversely, random gene sets correctly yield zero confidence in 87% of cases. Other LLMs (GPT-3.5, Gemini Pro, Mixtral Instruct and Llama2 70b) vary in function recovery but are falsely confident for random sets. In gene clusters from omics data, GPT-4 identifies common functions for 45% of cases, fewer than functional enrichment but with higher specificity and gene coverage. Manual review of supporting rationale and citations finds these functions are largely verifiable. These results position LLMs as valuable omics assistants.
Collapse
Affiliation(s)
- Mengzhou Hu
- Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Sahar Alkhairy
- Department of Computer Science and Engineering, University of California San Diego, La Jolla, CA, USA
| | - Ingoo Lee
- Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Rudolf T Pillich
- Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Dylan Fong
- Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Kevin Smith
- Department of Physics, University of California San Diego, La Jolla, CA, USA
| | - Robin Bachelder
- Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Trey Ideker
- Department of Medicine, University of California San Diego, La Jolla, CA, USA.
- Department of Computer Science and Engineering, University of California San Diego, La Jolla, CA, USA.
| | - Dexter Pratt
- Department of Medicine, University of California San Diego, La Jolla, CA, USA.
| |
Collapse
|
36
|
Shen X, Wu J, Zhang F, Bi Q, Sun Z, Wang W. Deciphering the impact of senescence in kidney transplant rejection: An integrative machine learning and multi-omics analysis via bulk and single-cell RNA sequencing. PLoS One 2024; 19:e0312272. [PMID: 39602449 PMCID: PMC11602102 DOI: 10.1371/journal.pone.0312272] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2024] [Accepted: 09/30/2024] [Indexed: 11/29/2024] Open
Abstract
BACKGROUND The demographic shift towards an older population presents significant challenges for kidney transplantation (KTx), particularly due to the vulnerability of aged donor kidneys to ischemic damage, delayed graft function, and reduced graft survival. KTx rejection poses a significant threat to allograft function and longevity of the kidney graft. The relationship between senescence and rejection remains elusive and controversial. METHODS Gene Expression Omnibus (GEO) provided microarray and single-cell RNA sequencing datasets. After integrating Senescence-Related Genes (SRGs) from multiple established databases, differential expression analysis, weighted gene co-expression network analysis (WGCNA), and machine learning algorithms were applied to identify predictive SRGs (pSRGs). A cluster analysis of rejection samples was conducted using the consensus clustering algorithm. Subsequently, we utilized multiple machine learning methods (RF, SVM, XGB, GLM and LASSO) based on pSRGs to develop the optimal Acute Rejection (AR) diagnostic model and long-term graft survival predictive signatures. Finally, we validated the role of pSRGs and senescence in kidney rejection through the single-cell landscape. RESULTS Thirteen pSRGs were identified, correlating with rejection. Two rejection clusters were divided (Cluster C1 and C2). GSVA analysis of two clusters underscored a positive correlation between senescence, KTx rejection occurrence and worse graft survival. A non-invasive diagnostic model (AUC = 0.975) and a prognostic model (1- Year AUC = 0.881; 2- Year AUC = 0.880; 3- Year AUC = 0.883) for graft survival were developed, demonstrating significant predictive capabilities to early detect acute rejection and long-term graft outcomes. Single-cell sequencing analysis provided a detailed cellular-level landscape of rejection, supporting the conclusions drawn from above. CONCLUSION Our comprehensive analysis underscores the pivotal role of senescence in KTx rejection, highlighting the potential of SRGs as biomarkers for diagnosing rejection and predicting graft survival, which may enhance personalized treatment strategies and improve transplant outcomes.
Collapse
Affiliation(s)
- Xihao Shen
- Department of Urology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
- Institute of Urology, Capital Medical University, Beijing, China
| | - Jiyue Wu
- Department of Urology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
- Institute of Urology, Capital Medical University, Beijing, China
| | - Feilong Zhang
- Department of Urology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
- Institute of Urology, Capital Medical University, Beijing, China
| | - Qing Bi
- Department of Urology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
- Institute of Urology, Capital Medical University, Beijing, China
| | - Zejia Sun
- Department of Urology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
- Institute of Urology, Capital Medical University, Beijing, China
| | - Wei Wang
- Department of Urology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
- Institute of Urology, Capital Medical University, Beijing, China
| |
Collapse
|
37
|
Li X, Peng C, Liu H, Dong M, Li S, Liang W, Li X, Bai J. Constructing methylation-driven ceRNA networks unveil tumor heterogeneity and predict patient prognosis. Hum Mol Genet 2024:ddae176. [PMID: 39603659 DOI: 10.1093/hmg/ddae176] [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: 09/05/2024] [Revised: 10/23/2024] [Accepted: 11/22/2024] [Indexed: 11/29/2024] Open
Abstract
Cancer development involves a complex interplay between genetic and epigenetic factors, with emerging evidence highlighting the pivotal role of competitive endogenous RNA (ceRNA) networks in regulating gene expression. However, the influence of ceRNA networks by aberrant DNA methylation remains incompletely understood. In our study, we proposed DMceNet, a computational method to characterize the effects of DNA methylation on ceRNA regulatory mechanisms and apply it across eight prevalent cancers. By integrating methylation and transcriptomic data, we constructed methylation-driven ceRNA networks and identified a dominant role of lncRNAs within these networks in two key ways: (i) 17 cancer-shared differential methylation lncRNAs (DMlncs), including PVT1 and CASC2, form a Common Cancer Network (CCN) affecting key pathways such as the G2/M checkpoint, and (ii) 24 cancer-specific DMlncs construct unique ceRNA networks for each cancer type. For instance, in LUAD and STAD, hypomethylation drives DMlncs like PCAT6 and MINCR, disrupting the Wnt signaling pathway and apoptosis. We further investigated the characteristics of these methylation-driven ceRNA networks at the cellular level, revealing how methylation-driven dysregulation varies across distinct cell populations within the tumor microenvironment. Our findings also demonstrate the prognostic potential of cancer-specific ceRNA relationships, highlighting their relevance in predicting patient survival outcomes. This integrated transcriptomic and epigenomic analysis provides new insights into cancer biology and regulatory mechanisms.
Collapse
Affiliation(s)
- Xinyu Li
- College of Bioinformatics Science and Technology, Harbin Medical University, 194 Xuefu Road, Harbin, Heilongjiang 150081, China
| | - Chuo Peng
- College of Bioinformatics Science and Technology, Harbin Medical University, 194 Xuefu Road, Harbin, Heilongjiang 150081, China
| | - Hongyu Liu
- College of Bioinformatics Science and Technology, Harbin Medical University, 194 Xuefu Road, Harbin, Heilongjiang 150081, China
| | - Mingjie Dong
- College of Bioinformatics Science and Technology, Harbin Medical University, 194 Xuefu Road, Harbin, Heilongjiang 150081, China
| | - Shujuan Li
- College of Bioinformatics Science and Technology, Harbin Medical University, 194 Xuefu Road, Harbin, Heilongjiang 150081, China
| | - Weixin Liang
- College of Bioinformatics Science and Technology, Harbin Medical University, 194 Xuefu Road, Harbin, Heilongjiang 150081, China
| | - Xia Li
- College of Bioinformatics Science and Technology, Harbin Medical University, 194 Xuefu Road, Harbin, Heilongjiang 150081, China
- Key Laboratory of Reproductive Health Diseases Research and Translation, Ministry of Education, College of Biomedical Information and Engineering, Hainan Medical University, No. 3 Xueyuan Road, Haikou, Hainan 571199, China
| | - Jing Bai
- College of Bioinformatics Science and Technology, Harbin Medical University, 194 Xuefu Road, Harbin, Heilongjiang 150081, China
- Key Laboratory of Reproductive Health Diseases Research and Translation, Ministry of Education, College of Biomedical Information and Engineering, Hainan Medical University, No. 3 Xueyuan Road, Haikou, Hainan 571199, China
| |
Collapse
|
38
|
He E, Li Y, Zhao R, Kong Q, Shao Y, Wang C, Liu B, Jiang Y, Liu Q, Cui H. IL7 as a Risk Factor for Prostate Cancer: Implications for T Cell Apoptosis and Infiltration in the Tumor Microenvironment. Prostate 2024. [PMID: 39593187 DOI: 10.1002/pros.24830] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Revised: 10/23/2024] [Accepted: 11/11/2024] [Indexed: 11/28/2024]
Abstract
BACKGROUND Prostate cancer's complex interplay with the immune microenvironment prompted an investigation into immune-related pathogenic mechanisms and potential therapeutic targets. METHODS Within the GSE176031 data set, Seurat meticulously dissected single-cell profiles from radical prostatectomy patients. Leveraging CellMarker and SingleR cell identities were precisely annotated. Then, monocle traced pseudotime trajectories, illuminating cellular paths, complemented by CellChat's insights into intricate intercellular communications. Furthermore, mendelian randomization (MR) robustly substantiated causal associations within prostate cancer contexts. RESULTS Employing single-cell analysis on intraoperative tumor and normal tissue, we identified 15 distinct cell types, notably observing a significant T cell reduction in tumor samples. Intercellular communication analysis revealed multiple pathways between epithelial cells and T cells, highlighting interleukin (IL)-IL7R-IL2RG interactions. IL7R, crucial in T cell apoptosis, showed differential expression across T cell development stages. Patients with IL7 amplification had poorer outcomes (p < 0.05), supported by MR in two cohorts (ieu-b-4809 cohort: odds ratio [OR] = 1.005, p = 0.002, 95% confidence interval [CI] [1.002-1.008]; ebi-a-GCST90018905: OR = 1.063, p = 0.032, 95% CI [1.005-1.125]), confirming IL7 as a prostate cancer risk factor. CONCLUSIONS These findings suggest T cell depletion via IL7-IL7R signaling may drive prostate cancer progression, offering novel therapeutic insights.
Collapse
Affiliation(s)
- Enyang He
- Tianjin Medical University, Tianjin, China
- Graduate School of Tianjin Medical University, Tianjin, China
| | - Yaowen Li
- Tianjin Medical University, Tianjin, China
- The First Central Hospital of Tianjin, Tianjin, China
| | - Rui Zhao
- Tianjin Medical University, Tianjin, China
- General Hospital of Tianjin Medical University, Tianjin, China
| | - Qinyan Kong
- West China Hospital of Sichuan University, Chengdu, China
| | - Yi Shao
- Tianjin Medical University, Tianjin, China
- The Second Hospital of Tianjin Medical University, Tianjin, China
| | - Cong Wang
- Tianjin Medical University, Tianjin, China
- General Hospital of Tianjin Medical University, Tianjin, China
| | - Baoqun Liu
- Tianjin Medical University, Tianjin, China
- The First Central Hospital of Tianjin, Tianjin, China
| | - Yvhang Jiang
- Tianjin Medical University, Tianjin, China
- General Hospital of Tianjin Medical University, Tianjin, China
| | - Qian Liu
- Tianjin Medical University, Tianjin, China
- The First Central Hospital of Tianjin, Tianjin, China
| | - Hualei Cui
- Tianjin Medical University, Tianjin, China
- Graduate School of Tianjin Medical University, Tianjin, China
| |
Collapse
|
39
|
Ren YF, Ma Q, Zeng X, Huang CX, Ren JL, Li F, Tong JJ, He JW, Zhong Y, Tan SY, Jiang H, Zhang LF, Lai HZ, Xiao P, Zhuang X, Wu P, You LT, Shi W, Fu X, Zheng C, You FM. Single-cell RNA sequencing reveals immune microenvironment niche transitions during the invasive and metastatic processes of ground-glass nodules and part-solid nodules in lung adenocarcinoma. Mol Cancer 2024; 23:263. [PMID: 39580469 PMCID: PMC11585206 DOI: 10.1186/s12943-024-02177-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Accepted: 11/16/2024] [Indexed: 11/25/2024] Open
Abstract
BACKGROUND Radiographically, ground-glass nodules (GGN) and part-solid nodules (PSN) in lung adenocarcinoma (LUAD) have significant heterogeneity in their clinical manifestations, biological characteristics, and prognosis. This study aimed to explore the heterogeneity of LUAD in different radiological phenotypes and associated factors influencing tumor evolution. METHODS We performed single-cell RNA sequencing (scRNA-seq) on tumor tissues from eight and seven cases of GGN- and PSN-LUAD, respectively, at different disease stages, including minimally invasive adenocarcinoma (MIA), invasive adenocarcinoma (IAC), and metastatic lung cancer (MLC). Additionally, we analyzed adjacent normal tissues from four cases. Immunohistochemistry, multiplex immunofluorescence, and external scRNA-seq data were employed to confirm the expression of signature genes as well as the distribution patterns of CXCL9 + TAMs and TREM2 + TAMs. A LUAD mouse model was generated using gene editing, organoid culture, and orthotopic transplantation techniques, and comprehensive analyses such as histopathology, RNA sequencing, and Western blotting were performed to validate key pathways. RESULTS Diverse cellular compositions were observed in the tumor microenvironment (TME) during GGN- and PSN-LUAD invasion and metastasis. Notably, CXCL9 + and TREM2 + tumor-associated macrophages (TAMs) exhibited the most significant enrichment changes. It was found that GGN-LUAD exhibited a stronger immune response than PSN-LUAD, with increased interaction between CXCL9 + TAMs and CD8 + tissue-resident memory T cells during invasion stage (MIA-IAC). Conversely, greater interactions between TREM2 + TAMs and tumor cells were observed in PSN-LUAD during the MLC stage. Additionally, TREM2 + TAMs were found to differentiate into TREM2 + /SPP1 + and TREM2 + /SPP1- TAMs at different stages, which promotes tumor progression. This study also emphasizes that during the transdifferentiation process of GGN- and PSN-LUAD, IFN-γ activates the STAT1 signaling pathway to regulate the activation of CXCL9 + TAMs, and further recruiting CD8 + Trm cells and activating T cells through MHC class I antigen presentation. The role of the IFN-γ/STAT1 pathway in the occurrence and development of LUAD was further validated by animal experiments. CONCLUSIONS Our findings offer a potential therapeutic strategy to maintain a dynamic balance within the TME and improve the immunotherapy efficacy by modulating the relative proportions and functional states of CXCL9 + TAMs and TREM2 + TAMs.
Collapse
Affiliation(s)
- Yi-Feng Ren
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, Sichuan Province, China
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, Sichuan Province, China
| | - Qiong Ma
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, Sichuan Province, China
| | - Xiao Zeng
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, Sichuan Province, China
| | - Chun-Xia Huang
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, Sichuan Province, China
| | - Jia-Li Ren
- LC-Bio Technologies (Hangzhou) CO., LTD, Hangzhou, 310018, Zhejiang Province, China
| | - Fang Li
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, Sichuan Province, China
| | - Jia-Jing Tong
- LC-Bio Technologies (Hangzhou) CO., LTD, Hangzhou, 310018, Zhejiang Province, China
| | - Jia-Wei He
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, Sichuan Province, China
| | - Yang Zhong
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, Sichuan Province, China
| | - Shi-Yan Tan
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, Sichuan Province, China
| | - Hua Jiang
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, Sichuan Province, China
| | - Long-Fei Zhang
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, Sichuan Province, China
| | - Heng-Zhou Lai
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, Sichuan Province, China
| | - Ping Xiao
- Department of Thoracic Surgery, School of Medicine, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, University of Electronic Science and Technology of China, Chengdu, 610042, Sichuan Province, China
| | - Xiang Zhuang
- Department of Thoracic Surgery, School of Medicine, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, University of Electronic Science and Technology of China, Chengdu, 610042, Sichuan Province, China
| | - Peng Wu
- LC-Bio Technologies (Hangzhou) CO., LTD, Hangzhou, 310018, Zhejiang Province, China
| | - Li-Ting You
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, Sichuan Province, China
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan Province, China
| | - Wei Shi
- Department of Anesthesiology, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan Province, China
| | - Xi Fu
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, Sichuan Province, China
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, Sichuan Province, China
| | - Chuan Zheng
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, Sichuan Province, China.
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, Sichuan Province, China.
| | - Feng-Ming You
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, Sichuan Province, China.
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, Sichuan Province, China.
| |
Collapse
|
40
|
Wang W, Lu J, Pan N, Zhang H, Dai J, Li J, Chi C, Zhang L, Wang L, Zhang M. Identification of early Alzheimer's disease subclass and signature genes based on PANoptosis genes. Front Immunol 2024; 15:1462003. [PMID: 39650656 PMCID: PMC11621049 DOI: 10.3389/fimmu.2024.1462003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Accepted: 11/05/2024] [Indexed: 12/11/2024] Open
Abstract
Introduction Alzheimer's disease (AD) is one of the most prevalent forms of dementia globally and remains an incurable condition that often leads to death. PANoptosis represents an emerging paradigm in programmed cell death, integrating three critical processes: pyroptosis, apoptosis, and necroptosis. Studies have shown that apoptosis, necroptosis, and pyroptosis play important roles in AD development. Therefore, targeting PANoptosis genes might lead to novel therapeutic targets and clinically relevant therapeutic approaches. This study aims to identify different molecular subtypes of AD and potential drugs for treating AD based on PANoptosis. Methods Differentially expressed PANoptosis genes associated with AD were identified via Gene Expression Omnibus (GEO) dataset GSE48350, GSE5281, and GSE122063. Least Absolute Shrinkage and Selection Operator (LASSO) regression was employed to construct a risk model linked to these PANoptosis genes. Consensus clustering analysis was conducted to define AD subtypes based on these genes. We further performed gene set variation analysis (GSVA), functional enrichment analysis, and immune cell infiltration analysis to investigate differences between the identified AD subtypes. Additionally, a protein-protein interaction (PPI) network was established to identify hub genes, and the DGIdb database was consulted to identify potential therapeutic compounds targeting these hub genes. Single-cell RNA sequencing analysis was utilized to assess differences in gene expression at the cellular level across subtypes. Results A total of 24 differentially expressed PANoptosis genes (APANRGs) were identified in AD, leading to the classification of two distinct AD subgroups. The results indicate that these subgroups exhibit varying disease progression states, with the early subtype primarily linked to dysfunctional synaptic signaling. Furthermore, we identified hub genes from the differentially expressed genes (DEGs) between the two clusters and predicted 38 candidate drugs and compounds for early AD treatment based on these hub genes. Single-cell RNA sequencing analysis revealed that key genes associated with the early subtype are predominantly expressed in neuronal cells, while the differential genes for the metabolic subtype are primarily found in endothelial cells and astrocytes. Conclusion In summary, we identified two subtypes, including the AD early synaptic abnormality subtype as well as the immune-metabolic subtype. Additionally, ten hub genes, SLC17A7, SNAP25, GAD1, SLC17A6, SLC32A1, PVALB, SYP, GRIN2A, SLC12A5, and SYN2, were identified as marker genes for the early subtype. These findings may provide valuable insights for the early diagnosis of AD and contribute to the development of innovative therapeutic strategies.
Collapse
Affiliation(s)
- Wenxu Wang
- School of Medical Information and Engineering, Xuzhou Medical University, Xuzhou, Jiangsu, China
- College of Life Science, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Jincheng Lu
- School of Medical Information and Engineering, Xuzhou Medical University, Xuzhou, Jiangsu, China
- College of Life Science, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Ningyun Pan
- School of Medical Information and Engineering, Xuzhou Medical University, Xuzhou, Jiangsu, China
- College of Life Science, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Huiying Zhang
- School of Mathematics and System Science, Shandong University of Science and Technology, Qingdao, Shandong, China
| | - Jingcen Dai
- School of Medical Information and Engineering, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Jie Li
- School of Medical Information and Engineering, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Cheng Chi
- School of Medical Information and Engineering, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Liumei Zhang
- School of Medical Information and Engineering, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Liang Wang
- Laboratory Medicine, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, China
| | - Mengying Zhang
- School of Medical Information and Engineering, Xuzhou Medical University, Xuzhou, Jiangsu, China
- College of Life Science, Xuzhou Medical University, Xuzhou, Jiangsu, China
| |
Collapse
|
41
|
Ma X, Lin L, Zhao Q, Iqbal M. TriTan: an efficient triple nonnegative matrix factorization method for integrative analysis of single-cell multiomics data. Brief Bioinform 2024; 26:bbae615. [PMID: 39581871 PMCID: PMC11586128 DOI: 10.1093/bib/bbae615] [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: 06/12/2024] [Revised: 10/15/2024] [Accepted: 11/19/2024] [Indexed: 11/26/2024] Open
Abstract
Single-cell multiomics have opened up tremendous opportunities for understanding gene regulatory networks underlying cell states by simultaneously profiling transcriptomes, epigenomes, and proteomes of the same cell. However, existing computational methods for integrative analysis of these high-dimensional multiomics data are either computationally expensive or limited in interpretation. These limitations pose challenges in the implementation of these methods in large-scale studies and hinder a more in-depth understanding of the underlying regulatory mechanisms. Here, we propose TriTan (Triple inTegrative fast non-negative matrix factorization), an efficient joint factorization method for single-cell multiomics data. TriTan implements a highly efficient factorization algorithm, greatly improving its computational performance. Three matrix factorization produced by TriTan helps in clustering cells, identifying signature features for each cell type, and uncovering feature associations across omics, which facilitates the identification of domains of regulatory chromatin and the prediction of cell-type-specific regulatory networks. We applied TriTan to the single-cell multiomics data obtained from different technologies and benchmarked it against the state-of-the-art methods where it shows highly competitive performance. Furthermore, we showed a range of downstream analyses conducted utilizing TriTan outputs, highlighting its capacity to facilitate interpretation in biological discovery.
Collapse
Affiliation(s)
- Xin Ma
- Division of Informatics, Imaging and Data Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Oxford Rd, Manchester, M13 9PL, UK
| | - Lijing Lin
- Centre for Health Informatics, Division of Informatics, Imaging and Data Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Oxford Rd, Manchester, M13 9PL, UK
| | - Qian Zhao
- Division of Informatics, Imaging and Data Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Oxford Rd, Manchester, M13 9PL, UK
| | - Mudassar Iqbal
- Division of Informatics, Imaging and Data Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Oxford Rd, Manchester, M13 9PL, UK
| |
Collapse
|
42
|
Feng J, Liang Y, Yu T. ADM: adaptive graph diffusion for meta-dimension reduction. Brief Bioinform 2024; 26:bbae612. [PMID: 39584700 PMCID: PMC11586774 DOI: 10.1093/bib/bbae612] [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: 08/05/2024] [Revised: 10/18/2024] [Accepted: 11/12/2024] [Indexed: 11/26/2024] Open
Abstract
Dimension reduction is essential for analyzing high-dimensional data, with various techniques developed to address diverse data characteristics. However, individual methods often struggle to capture all intricate patterns and complex structures simultaneously. To overcome this limitation, we introduce ADM (Adaptive graph Diffusion for Meta-dimension reduction), a novel meta-dimension reduction method grounded in graph diffusion theory. ADM integrates results from multiple dimension reduction techniques, leveraging their individual strengths while mitigating their specific weaknesses.ADM utilizes dynamic Markov processes to transform Euclidean space results into an information space, revealing intrinsic nonlinear manifold structures that are hard to capture by conventional methods. A critical advancement in ADM is its adaptive diffusion mechanism, which dynamically selects optimal diffusion time scales for each sample, enabling effective representation of multi-scale structures. This approach generates robust, high-quality low-dimensional representations that capture both local and global data structures while reducing noise and technique-specific distortions. We demonstrate ADM's efficacy on simulated and real-world datasets, including various omics data types. Results show that ADM provides clearer separation between biological groups and reveals more meaningful patterns compared to existing methods, advancing the analysis and visualization of complex biological data.
Collapse
Affiliation(s)
- Junning Feng
- School of Data Science, the Chinese University of Hong Kong, Shenzhen (CUHK-Shenzhen), 518172 Guangdong, China
- Faculty of Innovation Engineering, Macau University of Science and Technology, 999078 MacaoSpecial Administrative Region of China
| | - Yong Liang
- Chinese Medicine Guangdong Laboratory, Hengqin 519031 Guangdong, China
| | - Tianwei Yu
- School of Data Science, the Chinese University of Hong Kong, Shenzhen (CUHK-Shenzhen), 518172 Guangdong, China
| |
Collapse
|
43
|
Wong KH, Rodriguez NA, Traylor-Knowles N. Exploring the Unknown: How Can We Improve Single-cell RNAseq Cell Type Annotations in Non-model Organisms? Integr Comp Biol 2024; 64:1291-1299. [PMID: 39013613 DOI: 10.1093/icb/icae112] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2024] [Revised: 07/05/2024] [Accepted: 07/08/2024] [Indexed: 07/18/2024] Open
Abstract
Single-cell RNA sequencing (scRNAseq) is a powerful tool to describe cell types in multicellular organisms across the animal kingdom. In standard scRNAseq analysis pipelines, clusters of cells with similar transcriptional signatures are given cell type labels based on marker genes that infer specialized known characteristics. Since these analyses are designed for model organisms, such as humans and mice, problems arise when attempting to label cell types of distantly related, non-model species that have unique or divergent cell types. Consequently, this leads to limited discovery of novel species-specific cell types and potential mis-annotation of cell types in non-model species while using scRNAseq. To address this problem, we discuss recently published approaches that help annotate scRNAseq clusters for any non-model organism. We first suggest that annotating with an evolutionary context of cell lineages will aid in the discovery of novel cell types and provide a marker-free approach to compare cell types across distantly related species. Secondly, machine learning has greatly improved bioinformatic analyses, so we highlight some open-source programs that use reference-free approaches to annotate cell clusters. Lastly, we propose the use of unannotated genes as potential cell markers for non-model organisms, as many do not have fully annotated genomes and these data are often disregarded. Improving single-cell annotations will aid the discovery of novel cell types and enhance our understanding of non-model organisms at a cellular level. By unifying approaches to annotate cell types in non-model organisms, we can increase the confidence of cell annotation label transfer and the flexibility to discover novel cell types.
Collapse
Affiliation(s)
- Kevin H Wong
- Department of Marine Biology and Ecology, Rosenstiel School of Marine, Atmospheric, and Earth Science, University of Miami, Miami, Florida, USA, 33149
| | - Natalia Andrade Rodriguez
- Department of Marine Biology and Ecology, Rosenstiel School of Marine, Atmospheric, and Earth Science, University of Miami, Miami, Florida, USA, 33149
| | - Nikki Traylor-Knowles
- Department of Marine Biology and Ecology, Rosenstiel School of Marine, Atmospheric, and Earth Science, University of Miami, Miami, Florida, USA, 33149
| |
Collapse
|
44
|
Yin M, Feng C, Yu Z, Zhang Y, Li Y, Wang X, Song C, Guo M, Li C. sc2GWAS: a comprehensive platform linking single cell and GWAS traits of human. Nucleic Acids Res 2024:gkae1008. [PMID: 39565208 DOI: 10.1093/nar/gkae1008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Revised: 10/01/2024] [Accepted: 10/23/2024] [Indexed: 11/21/2024] Open
Abstract
Identifying cell populations associated with risk variants is essential for uncovering cell-specific mechanisms that drive disease development and progression. Integrating genome-wide association studies (GWAS) with single-cell RNA sequencing (scRNA-seq) has become an effective strategy for detecting trait-cell relationships. The accumulation of trait-related single cell data has led to an urgent need for its comprehensively processing. To address this, we developed sc2GWAS (https://bio.liclab.net/sc2GWAS/), which aims to document large-scale GWAS trait-cell regulatory pairs at single-cell resolution and provide comprehensive annotations and enrichment analyses for these related pairs. The current version of sc2GWAS curates a total of 15 078 310 candidate trait-cell pairs from > 6 300 000 individual cells, offering a valuable resource for exploring complex regulatory relationships between traits and cells. We applied strict quality control measures on both scRNA-seq data and GWAS data, ensuring the reliability and accuracy of the datasets for the identification of trait-relevant cells and genes. In addition, sc2GWAS provides ranked lists of trait-relevant genes and extensive (epi) genetic annotations, making it a valuable resource for downstream analyses. We demonstrate the utility of the platform by investigating Alzheimer's disease, where we identified significant associations between the disease and microglial cells, with the APOE gene emerging as particularly significant. This platform facilitates detailed research into complex trait-cell and trait-gene interactions, we anticipate that sc2GWAS will become a comprehensive and valuable platform for exploring GWAS trait-cell regulatory mechanisms.
Collapse
Affiliation(s)
- Mingxue Yin
- The First Affiliated Hospital & National Health Commission Key Laboratory of Birth Defect Research and Prevention, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
- Insititute of Biochemistry and Molecular Biology, Hengyang Medical College, University of South China, Hengyang, Hunan 421001, China
- The First Affiliated Hospital, Cardiovascular Lab of Big Data and Imaging Artificial Intelligence, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
- Hunan Provincial Key Laboratory of Multi-omics And Artificial Intelligence of Cardiovascular Diseases, University of South China, Hengyang, Hunan 421001, China
| | - Chenchen Feng
- School of Computer, University of South China, Hengyang, Hunan 421001, China
| | - Zhengmin Yu
- The First Affiliated Hospital & National Health Commission Key Laboratory of Birth Defect Research and Prevention, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
- Insititute of Biochemistry and Molecular Biology, Hengyang Medical College, University of South China, Hengyang, Hunan 421001, China
- The First Affiliated Hospital, Cardiovascular Lab of Big Data and Imaging Artificial Intelligence, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
- Hunan Provincial Key Laboratory of Multi-omics And Artificial Intelligence of Cardiovascular Diseases, University of South China, Hengyang, Hunan 421001, China
- School of Computer, University of South China, Hengyang, Hunan 421001, China
| | - Yuexin Zhang
- The First Affiliated Hospital & National Health Commission Key Laboratory of Birth Defect Research and Prevention, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
- Insititute of Biochemistry and Molecular Biology, Hengyang Medical College, University of South China, Hengyang, Hunan 421001, China
- The First Affiliated Hospital, Cardiovascular Lab of Big Data and Imaging Artificial Intelligence, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
- Hunan Provincial Key Laboratory of Multi-omics And Artificial Intelligence of Cardiovascular Diseases, University of South China, Hengyang, Hunan 421001, China
| | - Ye Li
- The First Affiliated Hospital & National Health Commission Key Laboratory of Birth Defect Research and Prevention, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
- Insititute of Biochemistry and Molecular Biology, Hengyang Medical College, University of South China, Hengyang, Hunan 421001, China
- The First Affiliated Hospital, Cardiovascular Lab of Big Data and Imaging Artificial Intelligence, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
- Hunan Provincial Key Laboratory of Multi-omics And Artificial Intelligence of Cardiovascular Diseases, University of South China, Hengyang, Hunan 421001, China
| | - Xuan Wang
- The First Affiliated Hospital & National Health Commission Key Laboratory of Birth Defect Research and Prevention, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
- The First Affiliated Hospital, Cardiovascular Lab of Big Data and Imaging Artificial Intelligence, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
- Hunan Provincial Key Laboratory of Multi-omics And Artificial Intelligence of Cardiovascular Diseases, University of South China, Hengyang, Hunan 421001, China
| | - Chao Song
- The First Affiliated Hospital & National Health Commission Key Laboratory of Birth Defect Research and Prevention, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
- The First Affiliated Hospital, Cardiovascular Lab of Big Data and Imaging Artificial Intelligence, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
- Hunan Provincial Key Laboratory of Multi-omics And Artificial Intelligence of Cardiovascular Diseases, University of South China, Hengyang, Hunan 421001, China
| | - Maozu Guo
- School of Electrical and Information Engineering, Beijing University of Civil Engineering and Architecture, Beijing 100044, China
| | - Chunquan Li
- The First Affiliated Hospital & National Health Commission Key Laboratory of Birth Defect Research and Prevention, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
- Insititute of Biochemistry and Molecular Biology, Hengyang Medical College, University of South China, Hengyang, Hunan 421001, China
- The First Affiliated Hospital, Cardiovascular Lab of Big Data and Imaging Artificial Intelligence, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
- Hunan Provincial Key Laboratory of Multi-omics And Artificial Intelligence of Cardiovascular Diseases, University of South China, Hengyang, Hunan 421001, China
- School of Computer, University of South China, Hengyang, Hunan 421001, China
- The First Affiliated Hospital, Institute of Cardiovascular Disease, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
- The First Affiliated Hospital, Department of Cardiology, Hengyang Medical School, University of South China, Hengyang, China
- Department of Cell Biology and Genetics, School of Basic Medical Sciences, Hengyang Medical School, University of South China, Hengyang, Hunan421001, China
- Key Laboratory of Rare Pediatric Diseases, Ministry of Education, University of South China, Hengyang, Hunan 421001, China
| |
Collapse
|
45
|
Zhou L, Liu H, Chen Y, Hua L, Wu X, Gao X, Mao L. Unveiling Leydig cell heterogeneity and its role in male infertility: A single-cell transcriptomic study of human testicular tissue. Reprod Biol 2024; 25:100972. [PMID: 39566254 DOI: 10.1016/j.repbio.2024.100972] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Revised: 11/02/2024] [Accepted: 11/12/2024] [Indexed: 11/22/2024]
Abstract
Male infertility and impaired spermatogenesis are significant concerns in reproductive health, often linked to disruptions in the cellular and molecular processes within the testis. The cellular composition and transcriptional dynamics of human testicular tissue are crucial for understanding these issues. Previous studies have largely relied on bulk tissue analysis, which obscures the distinct roles and interactions of specific cell types. Here, through a comprehensive single-cell transcriptomic analysis of human testes across various developmental stages and pathological conditions, we reveal the intricate cellular heterogeneity and the molecular mechanisms underlying testicular function. Our study identifies significant disruptions in the differentiation trajectories of Germ cells in conditions such as Klinefelter syndrome (KS), AZFa deletion, and Sertoli-cell-only syndrome (SCOS). We further uncover key transcription factors and regulatory networks governing Leydig cell function, particularly those related to steroidogenesis and hormonal regulation. These findings highlight the organized yet complex cellular and molecular landscape of the testis and uncover critical pathways altered in male infertility. Collectively, our data suggest that targeted therapeutic strategies could be developed to address specific disruptions in testicular cell populations and their associated regulatory networks.
Collapse
Affiliation(s)
- Liwei Zhou
- Department of Urology, Xinghua People's Hospital Affiliated to Yangzhou University, Taizhou 225700, Jiangsu, China
| | - Hanchao Liu
- Department of Urology and Andrology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310016, Zhejiang, China
| | - Yuming Chen
- Department of Urology, Xinghua People's Hospital Affiliated to Yangzhou University, Taizhou 225700, Jiangsu, China
| | - Lin Hua
- Department of Urology and Andrology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310016, Zhejiang, China
| | - Xiaolong Wu
- Department of Urology and Andrology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310016, Zhejiang, China
| | - Xintao Gao
- Department of Urology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310016, Zhejiang, China.
| | - Le Mao
- Department of Vascular Surgery, Shanghai Geriatric Medical Center, Shanghai, China; Department of Vascular Surgery, Zhongshan Hospital, Fudan University, Shanghai, China; Institute of Vascular Surgery, Fudan University, Shanghai, China; National Clinical Research Center for Interventional Medicine, Shanghai, China.
| |
Collapse
|
46
|
Gao J, Liu Y, Tao L, Zeng P, Ye G, Zheng Y, Zhang N. Single-cell data revealed the regulatory mechanism of TNK cell heterogeneity in liver metastasis from gastric cancer. Discov Oncol 2024; 15:664. [PMID: 39549183 PMCID: PMC11569111 DOI: 10.1007/s12672-024-01528-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Accepted: 11/04/2024] [Indexed: 11/18/2024] Open
Abstract
AIM The present work set out to classify cell subpopulations related to liver metastasis from gastric cancer (GC) and the mechanisms of their interactions with other immune cell subpopulations. BACKGROUND GC is characterized by a high degree of heterogeneity and liver metastasis. Exploring the mechanism of liver metastasis of GC from the perspective of heterogeneity of the tumor microenvironment (TME) might help improve the efficacy of GC treatment. OBJECTIVE Based on the cellular subpopulation characteristics of GC with liver metastasis, the regulatory mechanisms contributing to GC progression were analyzed, with special focuses on the roles of signaling pathways, transcription factors (TFs) and ligand-receptor pairs. METHODS The GSE163558 dataset was downloaded from the Gene Expression Omnibus (GEO) database to collect single-cell transcriptomic data of GC patients and their metastasis groups for cell clustering and relevant analyses. Differentially expressed genes (DEGs) in the GC and GC liver metastasis groups were screened and subjected to Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analysis. SCENIC analysis was used to mine TFs that affected cellular subpopulations during liver metastasis from GC. The relative expression levels of TFs in GC were determined using qRT-PCR. Transwell and wound healing assays were utilized to verify the regulation of the TFs on the migration and invasion of GC cells. Interaction network between the cellular subpopulations was developed applying CellChat. RESULTS Single-cell clustering was performed to group six major cell subpopulations, namely, Myeloid cells, B cells, Mast cells, Epithelial cells, Fibroblasts, and TNK cells, among which the number of TNK cells was significantly increased in the GC liver metastasis group. Differentially enriched pathways of TNK cells between GC and GC liver metastasis groups mainly included IL-17 and Pi3k-Akt signaling pathways. TNK cell subsets could be further categorized into CD8 T cells, Exhausted T cells, NK cells, NKT cells, and Treg cells, with the GC liver metastasis group showing significantly more CD8 T cells and NKT cells. FOS and JUNB were the TFs of TNK cell marker genes that contributed to liver metastasis from GC and the invasion and migration of GC cell lines. Significant differences in immune cell communication ligand-receptor pairs existed between the GC and GC liver metastasis groups. CONCLUSION This study revealed the critical role of TNK cell subsets in GC with liver metastasis applying single-cell transcriptomics analysis. The findings provided an important theoretical basis for developing novel therapies to inhibit liver metastasis from GC.
Collapse
Affiliation(s)
- Jun Gao
- Department of Gastrointestinal Surgery, Jiangxi Province Hospital of Integrated Chinese and Western Medicine, Nanchang, 330003, China
| | - Yujuan Liu
- Department of Emergency, Jiangxi Province Hospital of Integrated Chinese and Western Medicine, Nanchang, 330003, China
| | - Lu Tao
- Department of Emergency, Jiangxi Province Hospital of Integrated Chinese and Western Medicine, Nanchang, 330003, China
| | - Peng Zeng
- Department of Emergency, Jiangxi Province Hospital of Integrated Chinese and Western Medicine, Nanchang, 330003, China
| | - Guiying Ye
- Department of Emergency, Jiangxi Province Hospital of Integrated Chinese and Western Medicine, Nanchang, 330003, China
| | - Ying Zheng
- Department of Emergency, Jiangxi Province Hospital of Integrated Chinese and Western Medicine, Nanchang, 330003, China
| | - Nai Zhang
- Department of Emergency, Jiangxi Province Hospital of Integrated Chinese and Western Medicine, Nanchang, 330003, China.
| |
Collapse
|
47
|
Wiśniewski J, Więcek K, Ali H, Pyrc K, Kula-Păcurar A, Wagner M, Chen HC. Distinguishable topology of the task-evoked functional genome networks in HIV-1 reservoirs. iScience 2024; 27:111222. [PMID: 39559761 PMCID: PMC11570469 DOI: 10.1016/j.isci.2024.111222] [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: 08/05/2024] [Revised: 10/07/2024] [Accepted: 10/18/2024] [Indexed: 11/20/2024] Open
Abstract
HIV-1 reservoirs display a heterogeneous nature, lodging both intact and defective proviruses. To deepen our understanding of such heterogeneous HIV-1 reservoirs and their functional implications, we integrated basic concepts of graph theory to characterize the composition of HIV-1 reservoirs. Our analysis revealed noticeable topological properties in networks, featuring immunologic signatures enriched by genes harboring intact and defective proviruses, when comparing antiretroviral therapy (ART)-treated HIV-1-infected individuals and elite controllers. The key variable, the rich factor, played a pivotal role in classifying distinct topological properties in networks. The host gene expression strengthened the accuracy of classification between elite controllers and ART-treated patients. Markov chain modeling for the simulation of different graph networks demonstrated the presence of an intrinsic barrier between elite controllers and non-elite controllers. Overall, our work provides a prime example of leveraging genomic approaches alongside mathematical tools to unravel the complexities of HIV-1 reservoirs.
Collapse
Affiliation(s)
- Janusz Wiśniewski
- Quantitative Virology Research Group, Population Diagnostics Center, Łukasiewicz Research Network – PORT Polish Center for Technology Development, Stabłowicka 147, 54-066 Wrocław, Poland
| | - Kamil Więcek
- Quantitative Virology Research Group, Population Diagnostics Center, Łukasiewicz Research Network – PORT Polish Center for Technology Development, Stabłowicka 147, 54-066 Wrocław, Poland
| | - Haider Ali
- Molecular Virology Group, Małopolska Centre of Biotechnology, Jagiellonian University, Gronostajowa 7A str, 30-387 Kraków, Poland
- Doctoral School of Exact and Natural Sciences, Jagiellonian University, Łojasiewicza 11, 30-348 Kraków, Poland
| | - Krzysztof Pyrc
- Virogenetics Laboratory of Virology, Małopolska Centre of Biotechnology, Jagiellonian University, Gronostajowa 7A str, 30-387 Kraków, Poland
| | - Anna Kula-Păcurar
- Molecular Virology Group, Małopolska Centre of Biotechnology, Jagiellonian University, Gronostajowa 7A str, 30-387 Kraków, Poland
| | - Marek Wagner
- Innate Immunity Research Group, Life Sciences and Biotechnology Center, Łukasiewicz Research Network – PORT Polish Center for Technology Development, Stabłowicka 147, 54-066 Wrocław, Poland
| | - Heng-Chang Chen
- Quantitative Virology Research Group, Population Diagnostics Center, Łukasiewicz Research Network – PORT Polish Center for Technology Development, Stabłowicka 147, 54-066 Wrocław, Poland
| |
Collapse
|
48
|
Qin G, Narsinh K, Wei Q, Roach JC, Joshi A, Goetz SL, Moxon ST, Brush MH, Xu C, Yao Y, Glen AK, Morris ED, Ralevski A, Roper R, Belhu B, Zhang Y, Shmulevich I, Hadlock J, Glusman G. Generating Biomedical Knowledge Graphs from Knowledge Bases, Registries, and Multiomic Data. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.14.623648. [PMID: 39605475 PMCID: PMC11601480 DOI: 10.1101/2024.11.14.623648] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
As large clinical and multiomics datasets and knowledge resources accumulate, they need to be transformed into computable and actionable information to support automated reasoning. These datasets range from laboratory experiment results to electronic health records (EHRs). Barriers to accessibility and sharing of such datasets include diversity of content, size and privacy. Effective transformation of data into information requires harmonization of stakeholder goals, implementation, enforcement of standards regarding quality and completeness, and availability of resources for maintenance and updates. Systems such as the Biomedical Data Translator leverage knowledge graphs (KGs), structured and machine learning readable knowledge representation, to encode knowledge extracted through inference. We focus here on the transformation of data from multiomics datasets and EHRs into compact knowledge, represented in a KG data structure. We demonstrate this data transformation in the context of the Translator ecosystem, including clinical trials, drug approvals, cancer, wellness, and EHR data. These transformations preserve individual privacy. We provide access to the five resulting KGs through the Translator framework. We show examples of biomedical research questions supported by our KGs, and discuss issues arising from extracting biomedical knowledge from multiomics data.
Collapse
Affiliation(s)
- Guangrong Qin
- Institute for Systems Biology, 401 Terry Ave N, Seattle, WA 98109, USA
| | - Kamileh Narsinh
- Institute for Systems Biology, 401 Terry Ave N, Seattle, WA 98109, USA
| | - Qi Wei
- Institute for Systems Biology, 401 Terry Ave N, Seattle, WA 98109, USA
| | - Jared C. Roach
- Institute for Systems Biology, 401 Terry Ave N, Seattle, WA 98109, USA
| | - Arpita Joshi
- The Scripps Research Institute, 10550 N Torrey Pines Rd, La Jolla, CA 92037, USA
| | - Skye L. Goetz
- Institute for Systems Biology, 401 Terry Ave N, Seattle, WA 98109, USA
| | - Sierra T. Moxon
- Lawrence Berkeley National Laboratory, 1 Cyclotron Road, Berkeley, CA 94720, USA
| | - Matthew H. Brush
- UNC Chapel Hill, Department of Genetics, 120 Mason Farm Rd, Chapel Hill, NC 27599, USA
| | - Colleen Xu
- The Scripps Research Institute, 10550 N Torrey Pines Rd, La Jolla, CA 92037, USA
| | - Yao Yao
- Oregon State University, 1500 SW Jefferson Way, Corvallis, OR 97331
| | - Amy K. Glen
- Oregon State University, 1500 SW Jefferson Way, Corvallis, OR 97331
| | - Evan D. Morris
- Renaissance Computing Institute, 100 Europa Dr, Ste 540, Chapel Hill, NC 27517, USA
| | | | - Ryan Roper
- Institute for Systems Biology, 401 Terry Ave N, Seattle, WA 98109, USA
| | - Basazin Belhu
- Institute for Systems Biology, 401 Terry Ave N, Seattle, WA 98109, USA
| | - Yue Zhang
- Institute for Systems Biology, 401 Terry Ave N, Seattle, WA 98109, USA
| | - Ilya Shmulevich
- Institute for Systems Biology, 401 Terry Ave N, Seattle, WA 98109, USA
| | - Jennifer Hadlock
- Institute for Systems Biology, 401 Terry Ave N, Seattle, WA 98109, USA
| | - Gwênlyn Glusman
- Institute for Systems Biology, 401 Terry Ave N, Seattle, WA 98109, USA
| |
Collapse
|
49
|
Sun S, Chen X, Ding N, Zhang M, Li X, Chen L, Sun K, Liu Y. Gamma-aminobutyric acid-mediated neuro-immune interactions in glioblastoma: Implications for prognosis and immunotherapy response. Life Sci 2024; 357:123067. [PMID: 39322177 DOI: 10.1016/j.lfs.2024.123067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Revised: 09/11/2024] [Accepted: 09/16/2024] [Indexed: 09/27/2024]
Abstract
AIMS This study aimed to investigate the role of gamma-aminobutyric acid (GABA) in the glioblastoma (GBM) tumor immune microenvironment (TIME) and its impact on prognosis and response to immunotherapy. MAIN METHODS This study employed single-cell RNA sequencing (scRNA-seq) to delineate the TIME of GBM, utilized non-negative matrix factorization (NMF) for GABA-associated cell clustering, and performed pseudotime analysis for cellular trajectories. Additionally, we integrated immunohistochemistry (IHC), immunofluorescence (IF), and protein-protein interaction (PPI) analysis to explore the regulatory mechanisms within the tumor microenvironment. KEY FINDINGS The study identified distinct GABA-associated immune cell subtypes, particularly macrophages and T-cells, with unique gene expression and developmental trajectories. The development of the GABA-associated scoring model (GABAAS), introduced novel prognostic indicators, enhancing our ability to predict patient outcomes. This study also suggests that GABA-related genes, including NDRG2 and TIMP1, play a crucial role in immune modulation, with potential implications for immunotherapy responsiveness. SIGNIFICANCE The findings underscore the potential of targeting GABA-related genes (NDRG2 and TIMP1) and M2 macrophage to reshape the glioblastoma immune landscape, offering a new frontier in personalized neuro-immunotherapy. This approach holds promise to counter individual tumor immunosuppressive mechanisms, enhancing patient outcomes.
Collapse
Affiliation(s)
- Shanyue Sun
- Department of Neurosurgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China.
| | - Xinyuan Chen
- 4+4 Medical Doctor Program, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Nannan Ding
- Department of Nephrology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Miao Zhang
- Department of Radiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Xiaoru Li
- Department of Neurosurgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Lin Chen
- Department of Neurosurgery, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Kai Sun
- Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China; College of Medical Information and Artificial Intelligence & Central Hospital Affiliated to Shandong First Medical University, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China.
| | - Yingchao Liu
- Department of Neurosurgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China.
| |
Collapse
|
50
|
Li F, Song X, Fan W, Pei L, Liu J, Zhao R, Zhang Y, Li M, Song K, Sun Y, Zhang C, Zhang Y, Xu Y. SPathDB: a comprehensive database of spatial pathway activity atlas. Nucleic Acids Res 2024:gkae1041. [PMID: 39546631 DOI: 10.1093/nar/gkae1041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Revised: 09/26/2024] [Accepted: 10/19/2024] [Indexed: 11/17/2024] Open
Abstract
Spatial transcriptomics sequencing technology deepens our understanding of the diversity of cell behaviors, fates and states within complex tissue, which is often determined by the fine-tuning of regulatory network functional activities. Therefore, characterizing the functional activity within tissue space is helpful for revealing the functional features that drive spatial heterogeneity, and understanding complex biological processes. Here, we describe a database, SPathDB (http://bio-bigdata.hrbmu.edu.cn/SPathDB/), which aims to dissect the pathway-mediated multidimensional spatial heterogeneity in the context of functional activity. We manually curated spatial transcriptomics datasets and biological pathways from public data resources. SPathDB consists of 1689 868 spatial spots of 695 slices from 84 spatial transcriptome datasets of human and mouse, which involves 36 tissues, and also diseases such as cancer, and provides interactive analysis and visualization of the functional activities of 114 998 pathways across these spatial spots. SPathDB provides five flexible interfaces to retrieve and analyze pathways with highly variable functional activity across spatial spots, the distribution of pathway functional activities along pseudo-space axis, pathway-mediated spatial intercellular communications and the associations between spatial pathway functional activity and the occurrence of cell types. SPathDB will serve as a foundational resource for identifying functional features and elucidating underlying mechanisms of spatial heterogeneity.
Collapse
Affiliation(s)
- Feng Li
- College of Bioinformatics Science and Technology, Harbin Medical University, No.157 Baojian Road, Harbin 150081, China
| | - Xinyu Song
- College of Bioinformatics Science and Technology, Harbin Medical University, No.157 Baojian Road, Harbin 150081, China
| | - Wenli Fan
- College of Bioinformatics Science and Technology, Harbin Medical University, No.157 Baojian Road, Harbin 150081, China
| | - Liying Pei
- College of Bioinformatics Science and Technology, Harbin Medical University, No.157 Baojian Road, Harbin 150081, China
| | - Jiaqi Liu
- College of Bioinformatics Science and Technology, Harbin Medical University, No.157 Baojian Road, Harbin 150081, China
| | - Rui Zhao
- College of Bioinformatics Science and Technology, Harbin Medical University, No.157 Baojian Road, Harbin 150081, China
| | - Yifang Zhang
- College of Bioinformatics Science and Technology, Harbin Medical University, No.157 Baojian Road, Harbin 150081, China
| | - Mengyue Li
- College of Bioinformatics Science and Technology, Harbin Medical University, No.157 Baojian Road, Harbin 150081, China
| | - Kaiyue Song
- College of Bioinformatics Science and Technology, Harbin Medical University, No.157 Baojian Road, Harbin 150081, China
| | - Yu Sun
- College of Bioinformatics Science and Technology, Harbin Medical University, No.157 Baojian Road, Harbin 150081, China
| | - Chunlong Zhang
- College of Bioinformatics Science and Technology, Harbin Medical University, No.157 Baojian Road, Harbin 150081, China
| | - Yunpeng Zhang
- College of Bioinformatics Science and Technology, Harbin Medical University, No.157 Baojian Road, Harbin 150081, China
| | - Yanjun Xu
- College of Bioinformatics Science and Technology, Harbin Medical University, No.157 Baojian Road, Harbin 150081, China
| |
Collapse
|