1
|
Feng L, Sun R, Zhang H, Zhang J, Peng Z, Li J, Gao Y, Xu Y, Cui J, Liu J, Yan J, Guo L, Yang L, Shen Y, Qi Z. Exploring the protective mechanisms of syringaresinol against myocardial infarction by experimental validation and network pharmacology. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167728. [PMID: 39985987 DOI: 10.1016/j.bbadis.2025.167728] [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: 09/25/2024] [Revised: 01/21/2025] [Accepted: 02/13/2025] [Indexed: 02/24/2025]
Abstract
Myocardial Infarction (MI) is a leading cause of mortality worldwide. Currently, effective treatments are still lacking. Increasing evidence supports the benefits of Syringaresinol (SYR) for the treatment of cardiovascular disease is accumulating. Nevertheless, whether SYR can alleviate MI is unknown. The study aims to investigate the protective effect of SYR against MI and elucidate its potential molecular mechanism. We found that SYR ameliorate MI-induced cardiac dysfunction, reduce infarct size, and alleviate myocardial hypertrophy, fibrosis, inflammation, as well as apoptosis. In addition, we collected targets related to SYR and MI through multiple databases, and obtained 281 potential therapeutic targets after intersection. GO and KEGG enrichment analysis found that these therapeutic targets were concentrated on inflammation, fibrosis, and apoptosis pathways. Based on the PPI network and combined with Centiscape2.2 and cytoHubba analysis, we obtained 10 hub proteins. The molecular docking results showed that SYR has strong bindings with 10 hub proteins. snRNA-seq data showed that CASP3 and NFKB1 were expressed in all cell types. In addition, the therapeutic targets of SYR are also mainly distributed in all cell types. Finally, we found that SYR could alleviate MI by partially reversing the expression of AKT1, EGFR, CASP3, SRC, NFKB1, HSP90AA1, HIF1A, MMP9 and ESR1 both in vivo and in vitro. Our findings suggested that SYR may protect against MI by reducing inflammatory, fibrotic and apoptotic effects via multiple targets and pathways, which provides a new theoretical foundation for the clinical therapy of MI.
Collapse
Affiliation(s)
- Lifeng Feng
- Department of Molecular Pharmacology, School of Medicine, Beichen Hospital, Nankai University, Tianjin 300071, China
| | - Runjia Sun
- Department of Molecular Pharmacology, School of Medicine, Beichen Hospital, Nankai University, Tianjin 300071, China
| | - Hanmo Zhang
- Department of Molecular Pharmacology, School of Medicine, Beichen Hospital, Nankai University, Tianjin 300071, China
| | - Junwei Zhang
- Department of Molecular Pharmacology, School of Medicine, Beichen Hospital, Nankai University, Tianjin 300071, China
| | - Zeyan Peng
- Department of Molecular Pharmacology, School of Medicine, Beichen Hospital, Nankai University, Tianjin 300071, China
| | - Jing Li
- Department of Molecular Pharmacology, School of Medicine, Beichen Hospital, Nankai University, Tianjin 300071, China
| | - Yang Gao
- Department of Molecular Pharmacology, School of Medicine, Beichen Hospital, Nankai University, Tianjin 300071, China
| | - Yang Xu
- Department of Molecular Pharmacology, School of Medicine, Beichen Hospital, Nankai University, Tianjin 300071, China
| | - Jianlin Cui
- Department of Molecular Pharmacology, School of Medicine, Beichen Hospital, Nankai University, Tianjin 300071, China
| | - Jie Liu
- Department of Molecular Pharmacology, School of Medicine, Beichen Hospital, Nankai University, Tianjin 300071, China
| | - Jie Yan
- Department of Molecular Pharmacology, School of Medicine, Beichen Hospital, Nankai University, Tianjin 300071, China
| | - Lihong Guo
- Institute of Digestive Disease, Shengli Oilfield Central Hospital, Dongying 257000, China.
| | - Liang Yang
- Department of Molecular Pharmacology, School of Medicine, Beichen Hospital, Nankai University, Tianjin 300071, China.
| | - Yanna Shen
- School of Medical Technology, Tianjin Medical University, Tianjin 300203, China.
| | - Zhi Qi
- Department of Molecular Pharmacology, School of Medicine, Beichen Hospital, Nankai University, Tianjin 300071, China; Institute of Digestive Disease, Shengli Oilfield Central Hospital, Dongying 257000, China; Tianjin Key Laboratory of General Surgery in Construction, Tianjin Union Medical Center, Tianjin 300122, China; The First Department of Critical Care Medicine, The First Affiliated Hospital of Shihezi University, Shihezi 832003, China.
| |
Collapse
|
2
|
Chen S, Wang W, Shen L, Liu H, Luo J, Ren Y, Cui S, Ye Y, Shi G, Cheng F, Su X, Dai L, Gou M, Deng H. A 3D-printed microdevice encapsulates vascularized islets composed of iPSC-derived β-like cells and microvascular fragments for type 1 diabetes treatment. Biomaterials 2025; 315:122947. [PMID: 39547136 DOI: 10.1016/j.biomaterials.2024.122947] [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: 05/23/2024] [Revised: 10/23/2024] [Accepted: 11/05/2024] [Indexed: 11/17/2024]
Abstract
Transplantation of insulin-secreting cells provides a promising method for re-establishing the autonomous blood glucose control ability of type 1 diabetes (T1D) patients, but the low survival of the transplanted cells hinder the therapeutic efficacy. In this study, we 3D-printed an encapsulation system containing β-like cells and microvascular fragments (MVF), to create a retrivable microdevice with vascularized islets in vivo for T1D therapy. The functional β-like cells were differentiated from the urine epithelial cell-derived induced pluripotent stem cells (UiPSCs). Single-cell RNA sequencing provided an integrative study and macroscopic developmental analyses of the entire process of differentiation, which revealed the developmental trajectory of differentiation in vitro follows the developmental pattern of embryonic pancreas in vivo. The MVF were isolated from the epididymal fat pad. The microdevice with a groove structure were rapidly fabricated by the digital light processing (DLP)-3D printing technology. The β-like cells and MVF were uniformly distributed in the device. After subcutaneous transplantation into C57BL/6 mice, the microdevice have less collagen accumulation and low immune cell infiltration. Moreover, the microdevice encapsulated vascularized islets reduced hyperglycemia in 33 % of the treated mice for up to 100 days without immunosuppressants, and the humanized C-peptide was also detected in the serum of the mice. In summary, we described the microdevice-protected vascularized islets for long-term treatment of T1D, with high safety and potential clinical transformative value, and may therefore provide a translatable solution to advance the research progress of β cell replacement therapy for T1D.
Collapse
Affiliation(s)
- Shuang Chen
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Wenshuang Wang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Lanlin Shen
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Haofan Liu
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Jing Luo
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Yushuang Ren
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Susu Cui
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Yixin Ye
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Gang Shi
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Fuyi Cheng
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Xiaolan Su
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Lei Dai
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Maling Gou
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China.
| | - Hongxin Deng
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
3
|
Miao X, Pan R, Chang F. Down-regulation of ATP8B2 in Foam Cells Inhibits Autophagic Flux and ox-LDL Degradation in Atherosclerosis. Cell Biochem Biophys 2025:10.1007/s12013-025-01728-z. [PMID: 40148707 DOI: 10.1007/s12013-025-01728-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/11/2025] [Indexed: 03/29/2025]
Abstract
Our study aims to screen and explore the potential molecular mechanisms of atherosclerosis using a comprehensive research approach combining bioinformatics analysis and molecular biology experiments. Bioinformatics analyses were conducted to screen for key genes with significantly differential expression in atherosclerosis. Subsequently, macrophages and foam cells induced from THP-1 cells were utilized to validate the function of these key genes through siRNA knockdown. Molecular biology experiments encompassed reverse transcription polymerase chain reaction (RT-PCR), Western Blotting, immunofluorescence staining, and JC-1 probe detection of mitochondrial membrane potential. ATP8B2, encoding a P4-ATPase, was significantly downregulated in both plaque tissues and circulating macrophages of atherosclerosis patients. This enzyme influences membrane fusion and other dynamic processes by affecting the asymmetric distribution of phospholipids within the bilayer. Knockdown of ATP8B2 expression significantly inhibited autophagic flux in macrophages, manifested by abnormal accumulation of LC3-II and p62 protein levels. Furthermore, downregulation of ATP8B2 expression significantly inhibited the degradation of oxidized low-density lipoprotein (ox-LDL) by macrophages. Simultaneously, reduced ATP8B2 expression led to decreased mitochondrial membrane potential and mitochondrial dysfunction. Our study unveils for the first time the crucial role of ATP8B2 in atherosclerosis, particularly in maintaining autophagic flux, promoting ox-LDL degradation, and sustaining mitochondrial homeostasis.
Collapse
Affiliation(s)
- Xiaodong Miao
- The second department of cardiology, The Fourth Hospital of Harbin, Harbin, China
| | - Rui Pan
- Department of Geriatrics, The Fourth Hospital of Harbin, Harbin, China
| | - Fei Chang
- The second department of cardiology, The Fourth Hospital of Harbin, Harbin, China.
| |
Collapse
|
4
|
Jiang Y, Long G, Huang X, Wang W, Cheng B, Pan W. Single-cell transcriptomic analysis reveals dynamic changes in the liver microenvironment during colorectal cancer metastatic progression. J Transl Med 2025; 23:336. [PMID: 40091048 PMCID: PMC11910851 DOI: 10.1186/s12967-025-06351-3] [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: 12/02/2024] [Accepted: 03/04/2025] [Indexed: 03/19/2025] Open
Abstract
BACKGROUND Metastasis is a leading cause of cancer-related deaths, with the liver being the most frequent site of metastasis in colorectal cancer. Previous studies have predominantly focused on the influence of the primary tumor itself on metastasis, with relatively limited research examining the changes within target organs. METHODS Using an orthotopic mouse model of colorectal cancer, single-cell sequencing was employed to profile the transcriptomic landscape of pre-metastatic and metastatic livers. The analysis focused on identifying cellular and molecular changes within the hepatic microenvironment, with particular emphasis on inflammatory pathways and immune cell populations. RESULTS A neutrophil subpopulation with high Prok2 expression was identified, showing elevated levels in the pre-metastatic and metastatic liver. Increased infiltration of Prok2⁺ neutrophils correlated with poor prognosis in liver metastatic colorectal cancer patients. In the liver metastatic niche (MN), these neutrophils showed high App and Cd274 (PD-L1) expression, suppressing macrophage phagocytosis and promoting T-cell exhaustion. CONCLUSION A Prok2⁺ neutrophil subpopulation infiltrated both pre-metastatic and macro-metastatic liver environments, potentially driving immunosuppression through macrophage inhibition and T-cell exhaustion. Targeting Prok2⁺ neutrophils could represent a novel therapeutic strategy for preventing liver metastasis in colorectal cancer patients.
Collapse
Affiliation(s)
- Yue Jiang
- Department of General Surgery (Department of Pancreatic Hepatobiliary Surgery), The Sixth Affiliated Hospital, Sun Yat-Sen University, No. 26, Yuancun Erheng Road, Tianhe District, Guangzhou, 510655, Guangdong, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Disease, The Sixth Affiliated Hospital, Sun Yat-Sen University, No. 26, Yuancun Erheng Road, Tianhe District, Guangzhou, 510655, Guangdong, China
- Guangdong Institute of Gastroenterology, Guangzhou, 510655, Guangdong, China
| | - Guojie Long
- Department of General Surgery (Department of Pancreatic Hepatobiliary Surgery), The Sixth Affiliated Hospital, Sun Yat-Sen University, No. 26, Yuancun Erheng Road, Tianhe District, Guangzhou, 510655, Guangdong, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Disease, The Sixth Affiliated Hospital, Sun Yat-Sen University, No. 26, Yuancun Erheng Road, Tianhe District, Guangzhou, 510655, Guangdong, China
- Guangdong Institute of Gastroenterology, Guangzhou, 510655, Guangdong, China
| | - Xiaoming Huang
- Department of General Surgery (Department of Pancreatic Hepatobiliary Surgery), The Sixth Affiliated Hospital, Sun Yat-Sen University, No. 26, Yuancun Erheng Road, Tianhe District, Guangzhou, 510655, Guangdong, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Disease, The Sixth Affiliated Hospital, Sun Yat-Sen University, No. 26, Yuancun Erheng Road, Tianhe District, Guangzhou, 510655, Guangdong, China
- Guangdong Institute of Gastroenterology, Guangzhou, 510655, Guangdong, China
| | - Wenyu Wang
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Disease, The Sixth Affiliated Hospital, Sun Yat-Sen University, No. 26, Yuancun Erheng Road, Tianhe District, Guangzhou, 510655, Guangdong, China.
- Guangdong Institute of Gastroenterology, Guangzhou, 510655, Guangdong, China.
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510655, China.
| | - Bing Cheng
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Disease, The Sixth Affiliated Hospital, Sun Yat-Sen University, No. 26, Yuancun Erheng Road, Tianhe District, Guangzhou, 510655, Guangdong, China.
- Guangdong Institute of Gastroenterology, Guangzhou, 510655, Guangdong, China.
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510655, China.
| | - Weidong Pan
- Department of General Surgery (Department of Pancreatic Hepatobiliary Surgery), The Sixth Affiliated Hospital, Sun Yat-Sen University, No. 26, Yuancun Erheng Road, Tianhe District, Guangzhou, 510655, Guangdong, China.
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Disease, The Sixth Affiliated Hospital, Sun Yat-Sen University, No. 26, Yuancun Erheng Road, Tianhe District, Guangzhou, 510655, Guangdong, China.
- Guangdong Institute of Gastroenterology, Guangzhou, 510655, Guangdong, China.
| |
Collapse
|
5
|
Brash JT, Diez-Pinel G, Rinaldi L, Castellan RFP, Fantin A, Ruhrberg C. Endothelial transcriptomic, epigenomic and proteomic data challenge the proposed role for TSAd in vascular permeability. Angiogenesis 2025; 28:21. [PMID: 40080216 PMCID: PMC11906500 DOI: 10.1007/s10456-025-09971-x] [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: 10/08/2024] [Accepted: 02/23/2025] [Indexed: 03/15/2025]
Abstract
The vascular endothelial growth factor VEGF drives excessive vascular permeability to cause tissue-damaging oedema in neovascular and inflammatory diseases across multiple organs. Several molecular pathways have been implicated in VEGF-induced hyperpermeability, including binding of the VEGF-activated tyrosine kinase receptor VEGFR2 by the T-cell specific adaptor (TSAd) to recruit a SRC family kinase to induce junction opening between vascular endothelial cells (ECs). Inconsistent with a universal role for TSAd in permeability signalling, immunostaining approaches previously reported TSAd only in dermal and kidney vasculature. To address this discrepancy, we have mined publicly available omics data for expression of TSAd and other permeability-relevant signal transducers in multiple organs affected by VEGF-induced vascular permeability. Unexpectedly, TSAd transcripts were largely absent from EC single cell RNAseq data, whereas transcripts for other permeability-relevant signal transducers were detected readily. TSAd transcripts were also lacking from half of the EC bulk RNAseq datasets examined, and in the remaining datasets appeared at low levels concordant with models of leaky transcription. Epigenomic EC data located the TSAd promoter to closed chromatin in ECs, and mass spectrometry-derived EC proteomes typically lacked TSAd. By suggesting that TSAd is not actively expressed in ECs, our findings imply that TSAd is likely not critical for linking VEGFR2 to downstream signal transducers for EC junction opening.
Collapse
Affiliation(s)
- James T Brash
- UCL Institute of Ophthalmology, University College London, 11-43 Bath Street, London, EC1V 9EL, UK
| | - Guillermo Diez-Pinel
- UCL Institute of Ophthalmology, University College London, 11-43 Bath Street, London, EC1V 9EL, UK
| | - Luca Rinaldi
- Department of Biosciences, University of Milan, Via G. Celoria 26, 20133, Milan, Italy
| | - Raphael F P Castellan
- UCL Institute of Ophthalmology, University College London, 11-43 Bath Street, London, EC1V 9EL, UK
| | - Alessandro Fantin
- Department of Biosciences, University of Milan, Via G. Celoria 26, 20133, Milan, Italy.
| | - Christiana Ruhrberg
- UCL Institute of Ophthalmology, University College London, 11-43 Bath Street, London, EC1V 9EL, UK.
| |
Collapse
|
6
|
Luo R, Liu J, Wang T, Zhao W, Wang Y, Wen J, Wang H, Zhou X. The Landscape of Malignant Transition: Unraveling Cancer Cell-of-Origin and Heterogeneous Tissue Microenvironment. Cancer Lett 2025:217591. [PMID: 40054660 DOI: 10.1016/j.canlet.2025.217591] [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: 01/20/2025] [Revised: 02/10/2025] [Accepted: 02/25/2025] [Indexed: 03/12/2025]
Abstract
Understanding disease progression and sophisticated tumor ecosystems is imperative for investigating tumorigenesis mechanisms and developing novel prevention strategies. Here, we dissected heterogeneous microenvironments during malignant transitions by leveraging data from 1396 samples spanning 13 major tissues. Within transitional stem-like subpopulations highly enriched in precancers and cancers, we identified 30 recurring cellular states strongly linked to malignancy, including hypoxia and epithelial senescence, revealing a high degree of plasticity in epithelial stem cells. By characterizing dynamics in stem-cell crosstalk with the microenvironment along the pseudotime axis, we found differential roles of ANXA1 at different stages of tumor development. In precancerous stages, reduced ANXA1 levels promoted monocyte differentiation toward M1 macrophages and inflammatory responses, whereas during malignant progression, upregulated ANXA1 fostered M2 macrophage polarization and cancer-associated fibroblast transformation by increasing TGF-β production. Our spatiotemporal analysis further provided insights into mechanisms responsible for immunosuppression and a potential target to control evolution of precancer and mitigate the risk for cancer development.
Collapse
Affiliation(s)
- Ruihan Luo
- Laboratory of Hepatic AI Translation and West China Biomedical Big Data Center, West China Hospital, Sichuan University, Chengdu 610041, China; Center for Computational Systems Medicine, McWilliams School of Biomedical Informatics, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA; Med-X Center for Informatics, Sichuan University, Chengdu 610041, China.
| | - Jiajia Liu
- Center for Computational Systems Medicine, McWilliams School of Biomedical Informatics, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Tiangang Wang
- Center for Computational Systems Medicine, McWilliams School of Biomedical Informatics, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Weiling Zhao
- Center for Computational Systems Medicine, McWilliams School of Biomedical Informatics, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Yanfei Wang
- Center for Computational Systems Medicine, McWilliams School of Biomedical Informatics, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Jianguo Wen
- Center for Computational Systems Medicine, McWilliams School of Biomedical Informatics, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Hongyu Wang
- Department of Diagnostic and Interventional Imaging, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA; Center for Nursing Research, Cizik School of Nursing, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Xiaobo Zhou
- Center for Computational Systems Medicine, McWilliams School of Biomedical Informatics, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA; McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| |
Collapse
|
7
|
Pietrogrande G, Shaker MR, Stednitz SJ, Soheilmoghaddam F, Aguado J, Morrison SD, Zambrano S, Tabassum T, Javed I, Cooper-White J, Davis TP, O'Brien TJ, Scott EK, Wolvetang EJ. Valproic acid-induced teratogenicity is driven by senescence and prevented by Rapamycin in human spinal cord and animal models. Mol Psychiatry 2025; 30:986-998. [PMID: 39227432 PMCID: PMC11835743 DOI: 10.1038/s41380-024-02732-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Revised: 08/21/2024] [Accepted: 08/27/2024] [Indexed: 09/05/2024]
Abstract
Valproic acid (VPA) is an effective and widely used anti-seizure medication but is teratogenic when used during pregnancy, affecting brain and spinal cord development for reasons that remain largely unclear. Here we designed a genetic recombinase-based SOX10 reporter system in human pluripotent stem cells that enables tracking and lineage tracing of Neural Crest cells (NCCs) in a human organoid model of the developing neural tube. We found that VPA induces extensive cellular senescence and promotes mesenchymal differentiation of human NCCs. We next show that the clinically approved drug Rapamycin inhibits senescence and restores aberrant NCC differentiation trajectory after VPA exposure in human organoids and in developing zebrafish, highlighting the therapeutic promise of this approach. Finally, we identify the pioneer factor AP1 as a key element of this process. Collectively our data reveal cellular senescence as a central driver of VPA-associated neurodevelopmental teratogenicity and identifies a new pharmacological strategy for prevention. These results exemplify the power of genetically modified human stem cell-derived organoid models for drug discovery.
Collapse
Affiliation(s)
- Giovanni Pietrogrande
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, St. Lucia, Brisbane, QLD, 4072, Australia.
| | - Mohammed R Shaker
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, St. Lucia, Brisbane, QLD, 4072, Australia
| | - Sarah J Stednitz
- Department of Anatomy & Physiology, University of Melbourne, Parkville, VIC, Australia
| | - Farhad Soheilmoghaddam
- School of Chemical Engineering, University of Queensland, St. Lucia, QLD, 4072, Australia
| | - Julio Aguado
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, St. Lucia, Brisbane, QLD, 4072, Australia
| | - Sean D Morrison
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, St. Lucia, Brisbane, QLD, 4072, Australia
| | - Samuel Zambrano
- School of Medicine, Vita-Salute San Raffaele University, Milan, 20132, Italy
- Division of Genetics and Cell Biology, San Raffaele Scientific Institute, Milan, 20132, Italy
| | - Tahmina Tabassum
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, St. Lucia, Brisbane, QLD, 4072, Australia
| | - Ibrahim Javed
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, St. Lucia, Brisbane, QLD, 4072, Australia
| | - Justin Cooper-White
- School of Chemical Engineering, University of Queensland, St. Lucia, QLD, 4072, Australia
| | - Thomas P Davis
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, St. Lucia, Brisbane, QLD, 4072, Australia
| | - Terence J O'Brien
- Department of Neuroscience, The Central Clinical School, Alfred Health, Monash University, Melbourne, VIC, Australia
- The Departments of Medicine and Neurology, The Royal Melbourne Hospital, The University of Melbourne, Parkville, VIC, Australia
| | - Ethan K Scott
- Department of Anatomy & Physiology, University of Melbourne, Parkville, VIC, Australia
- Queensland Brain Institute, The University of Queensland, St. Lucia, Brisbane, QLD, 4072, Australia
| | - Ernst J Wolvetang
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, St. Lucia, Brisbane, QLD, 4072, Australia
| |
Collapse
|
8
|
Wu J, Li Z, Zhou W, Hu Z, Gao K, Yang J, Zhang W. Identification and validation of a novel signature based on immune‑related genes from epithelial cells to predict prognosis and treatment response in patients with lung squamous cell cancer by integrated analysis of single‑cell and bulk RNA sequencing. Oncol Lett 2025; 29:158. [PMID: 39911151 PMCID: PMC11795163 DOI: 10.3892/ol.2025.14904] [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: 08/28/2024] [Accepted: 12/04/2024] [Indexed: 02/07/2025] Open
Abstract
Epithelial cells are associated with tumor immunity through interstitial transformation, yet the role of epithelial immune-related genes (EIGs) in this process remains unclear. Comprehending the mechanisms behind EIGs within lung squamous cell carcinoma (LUSC) may offer an explanation to these issues. The present study aimed to explore the biological role of EIGs in patients with LUSC. Based on data from the Gene Expression Omnibus and The Cancer Genome Atlas databases, a survival model and nomogram was established. This model and nomogram were used to study the mechanism of EIGs in LUSC and its medical significance by enrichment analysis, tumor microenvironment, immune cell infiltration and immune function correlation analysis. Finally, reverse transcription-quantitative PCR (RT-qPCR) and external dataset were used to assess the expression of the EIGs. The survival model was used to develop 4 EIGs as predictors for patient outcomes. Survival curves revealed that higher risk patients had more negative outcomes. This model and the nomogram developed based entirely on this model had an accurate prognosis predictive LUSC. The enrichment analysis indicated that pathways related to antigen processing and presentation, as well as Epstein-Barr virus infection, were prevalent in the high-risk populations. The research on immune infiltration demonstrated a notable rise in activated dendritic cells and neutrophils in the high-risk group. Furthermore, the results revealed that the high-risk populations are particularly susceptible to the effects of afureserpine, gefitinib and savolitinib. Finally, the outcomes of RT-qPCR were consistent with those of the bioinformatics analysis. In conclusion, the risk evaluation model and nomogram are effective in forecasting the prognosis and guiding drug selection for patients with LUSC. A worse prognosis in patients with high risk may be associated with certain viral infections and antigen processing and presentation.
Collapse
Affiliation(s)
- Jiajun Wu
- Department of Thoracic Surgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330000, P.R. China
| | - Zhifeng Li
- Department of Thoracic Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, P.R. China
| | - Weijun Zhou
- Department of Thoracic Surgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330000, P.R. China
| | - Zhuozheng Hu
- Department of Thoracic Surgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330000, P.R. China
| | - Kun Gao
- Department of Thoracic Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, P.R. China
| | - Jin Yang
- Department of Thoracic Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, P.R. China
| | - Wenxiong Zhang
- Department of Thoracic Surgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330000, P.R. China
| |
Collapse
|
9
|
Liu C, Wang K, Mei J, Zhao R, Shen J, Zhang W, Li L, Roy B, Fang X. Integrative single-cell and spatial transcriptome analysis reveals heterogeneity of human liver progenitor cells. Hepatol Commun 2025; 9:e0662. [PMID: 40008906 PMCID: PMC11868439 DOI: 10.1097/hc9.0000000000000662] [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: 09/06/2024] [Accepted: 01/02/2025] [Indexed: 02/27/2025] Open
Abstract
BACKGROUND Liver progenitor cells (LPCs) with bipotential differentiation capacities are essential for restoring liver homeostasis and hepatocyte population after damage. However, the low proportion and shared markers with epithelial cells make studying LPC heterogeneity difficult, especially in humans. To address this gap, we explored over 259,400 human liver single cells across 4 conditions (fetal, healthy, cirrhotic, and HCC-affected livers). METHODS Human liver tissue samples were analyzed using spatial transcriptomics sequencing technologies to describe the heterogeneity of LPCs. Liver tissue was characterized by LPC heterogeneity at single-cell resolution by employing cellular modules, differentiation trajectories, and gene co-expression patterns. RESULTS We annotated and identified 1 LPC cluster, 3 LPC subpopulations, and 4 distinct cellular modules, indicating the heterogeneity within LPC and the diversity between LPCs and epithelial cells. LPCs showed spatial colocalization with cholangiocytes and comprised a small proportion (2.95±1.91%) within the merged epithelial cells and LPC populations, exhibiting marked differences in marker expression patterns compared to those in mice. LPCs exhibited distinct cellular states in functional restoration, activation, proliferation, and cell transition. Additionally, the gene co-expression network of LPCs exhibited 3 unique modules, reflecting the distinct connectivity of genes encoding apolipoproteins and heat shock proteins in the gene co-expression network modules. CONCLUSIONS Our study provides valuable insights into the multifaceted heterogeneity of human LPCs. Future studies focusing on spatial gene expression dynamics will contribute to our understanding of the spatial arrangement of liver regeneration.
Collapse
Affiliation(s)
- Chuanjun Liu
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
- BGI Research, Shenzhen, China
| | - Kai Wang
- Department of Hepatobiliary and Pancreatic Surgery and Minimally Invasive Surgery, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
| | | | - Ruizhen Zhao
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
- BGI Research, Shenzhen, China
| | | | - Wei Zhang
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
| | | | - Bhaskar Roy
- Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, China
| | | |
Collapse
|
10
|
Lyu T, Wu K, Zhou Y, Kong T, Li L, Wang K, Fu P, Wei P, Chen M, Zheng J. Single-Cell RNA Sequencing Reveals the Tumor Heterogeneity and Immunosuppressive Microenvironment in Urothelial Carcinoma. Cancer Sci 2025; 116:710-723. [PMID: 39726326 PMCID: PMC11875766 DOI: 10.1111/cas.16436] [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: 07/12/2024] [Revised: 11/24/2024] [Accepted: 12/09/2024] [Indexed: 12/28/2024] Open
Abstract
Urothelial carcinoma (UC) can arise from either the lower urinary tract or the upper tract; they represent different disease entities and require different clinical treatment strategies. A full understanding of the cellular characteristics in UC may guide the development of novel therapies. Here, we performed single-cell transcriptome analysis from four patients with UC of the bladder (UCB), five patients with UC of the ureter (UCU), and four patients with UC of the renal pelvis (UCRP) to develop a comprehensive cell atlas of UC. We found the rare epithelial cell subtype EP9 with epithelial-to-mesenchymal transition (EMT) and cancer stem cell (CSC) features, and specifically expressed SOX6, which was associated with poor prognosis. We also found that ACKR1+ endothelial cells and inflammatory cancer-associated fibroblasts (iCAFs) were more enriched in UCU, which may promote pathogenesis. While ESM1+ endothelial cells may more actively participate in UCB and UCRP tumorigenesis by promoting angiogenesis. Additionally, CD8 + effector T cells were more enriched in UCU and UCRP patients, while Tregs were mainly enriched in UCB tumors. C1QC+ macrophages and LAMP3+ dendritic cells were more enriched in UCB, which is closely related to the formation of the heterogeneous immunosuppressive microenvironment. Furthermore, we found strong interactions between iCAFs, EP9, and Endo_ESM1, and different degrees of activation of the FGF-FGFR3 axis and immune checkpoint pathway were observed in different UC subtypes. Our study elucidated the cellular heterogeneity and the components of the microenvironment in UC arising from the upper and lower urinary tracts and provided novel therapeutic targets.
Collapse
Affiliation(s)
- Tianqi Lyu
- Cixi Institute of Biomedical Engineering, Chinese Academy of Science (CAS)Ningbo Institute of Materials Technology and Engineering, CAS NingboNingboChina
| | - Kerong Wu
- Department of Urology, Ningbo First HospitalSchool of Medicine Ningbo University, Zhejiang University Ningbo HospitalNingboChina
| | - Yincong Zhou
- Department of Bioinformatics, College of Life SciencesZhejiang UniversityHangzhouChina
| | - Tong Kong
- Cixi Institute of Biomedical Engineering, Chinese Academy of Science (CAS)Ningbo Institute of Materials Technology and Engineering, CAS NingboNingboChina
| | - Lin Li
- Cixi Institute of Biomedical Engineering, Chinese Academy of Science (CAS)Ningbo Institute of Materials Technology and Engineering, CAS NingboNingboChina
| | - Kaizhe Wang
- Cixi Institute of Biomedical Engineering, Chinese Academy of Science (CAS)Ningbo Institute of Materials Technology and Engineering, CAS NingboNingboChina
| | - Pan Fu
- Cixi Institute of Biomedical Engineering, Chinese Academy of Science (CAS)Ningbo Institute of Materials Technology and Engineering, CAS NingboNingboChina
| | - Pengyao Wei
- Cixi Institute of Biomedical Engineering, Chinese Academy of Science (CAS)Ningbo Institute of Materials Technology and Engineering, CAS NingboNingboChina
| | - Ming Chen
- Department of Bioinformatics, College of Life SciencesZhejiang UniversityHangzhouChina
| | - Jianping Zheng
- Cixi Institute of Biomedical Engineering, Chinese Academy of Science (CAS)Ningbo Institute of Materials Technology and Engineering, CAS NingboNingboChina
| |
Collapse
|
11
|
Huang Y, Zhu S, Yao S, Zhai H, Liu C, Han JDJ. Unraveling aging from transcriptomics. Trends Genet 2025; 41:218-235. [PMID: 39424502 DOI: 10.1016/j.tig.2024.09.006] [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: 07/31/2024] [Revised: 09/19/2024] [Accepted: 09/19/2024] [Indexed: 10/21/2024]
Abstract
Research into aging constitutes a pivotal endeavor aimed at elucidating the underlying biological mechanisms governing aging and age-associated diseases, as well as promoting healthy longevity. Recent advances in transcriptomic technologies, such as bulk RNA sequencing (RNA-seq), single-cell transcriptomics, and spatial transcriptomics, have revolutionized our ability to study aging at unprecedented resolution and scale. These technologies present novel opportunities for the discovery of biomarkers, elucidation of molecular pathways, and development of targeted therapeutic strategies for age-related disorders. This review surveys recent breakthroughs in different types of transcripts on aging, such as mRNA, long noncoding (lnc)RNA, tRNA, and miRNA, highlighting key findings and discussing their potential implications for future studies in this field.
Collapse
Affiliation(s)
- Yuanfang Huang
- Peking-Tsinghua Center for Life Sciences, Center for Quantitative Biology (CQB), Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, China
| | - Shouxuan Zhu
- Peking-Tsinghua Center for Life Sciences, Center for Quantitative Biology (CQB), Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, China
| | - Shuai Yao
- Peking-Tsinghua Center for Life Sciences, Center for Quantitative Biology (CQB), Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, China
| | - Haotian Zhai
- Peking-Tsinghua Center for Life Sciences, Center for Quantitative Biology (CQB), Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, China
| | - Chenyang Liu
- Peking-Tsinghua Center for Life Sciences, Center for Quantitative Biology (CQB), Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, China
| | - Jing-Dong J Han
- Peking-Tsinghua Center for Life Sciences, Center for Quantitative Biology (CQB), Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, China; Peking University Chengdu Academy for Advanced Interdisciplinary Biotechnologies, Chengdu, China.
| |
Collapse
|
12
|
Feng Y, Liu G, Li H, Cheng L. The landscape of cell lineage tracing. SCIENCE CHINA. LIFE SCIENCES 2025:10.1007/s11427-024-2751-6. [PMID: 40035969 DOI: 10.1007/s11427-024-2751-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Accepted: 09/30/2024] [Indexed: 03/06/2025]
Abstract
Cell fate changes play a crucial role in the processes of natural development, disease progression, and the efficacy of therapeutic interventions. The definition of the various types of cell fate changes, including cell expansion, differentiation, transdifferentiation, dedifferentiation, reprogramming, and state transitions, represents a complex and evolving field of research known as cell lineage tracing. This review will systematically introduce the research history and progress in this field, which can be broadly divided into two parts: prospective tracing and retrospective tracing. The initial section encompasses an array of methodologies pertaining to isotope labeling, transient fluorescent tracers, non-fluorescent transient tracers, non-fluorescent genetic markers, fluorescent protein, genetic marker delivery, genetic recombination, exogenous DNA barcodes, CRISPR-Cas9 mediated DNA barcodes, and base editor-mediated DNA barcodes. The second part of the review covers genetic mosaicism, genomic DNA alteration, TCR/BCR, DNA methylation, and mitochondrial DNA mutation. In the final section, we will address the principal challenges and prospective avenues of enquiry in the field of cell lineage tracing, with a particular focus on the sequencing techniques and mathematical models pertinent to single-cell genetic lineage tracing, and the value of pursuing a more comprehensive investigation at both the spatial and temporal levels in the study of cell lineage tracing.
Collapse
Affiliation(s)
- Ye Feng
- Shanghai YangZhi Rehabilitation Hospital (Shanghai Sunshine Rehabilitation Center), Tongji University School of Medicine, Shanghai, 201619, China.
| | - Guang Liu
- Department of Vascular Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200023, China.
| | - Haiqing Li
- Department of Cardiac Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| | - Lin Cheng
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| |
Collapse
|
13
|
Li C, Liao J, Chen B, Wang Q. Heterogeneity of the tumor immune cell microenvironment revealed by single-cell sequencing in head and neck cancer. Crit Rev Oncol Hematol 2025:104677. [PMID: 40023465 DOI: 10.1016/j.critrevonc.2025.104677] [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: 12/05/2024] [Revised: 02/16/2025] [Accepted: 02/26/2025] [Indexed: 03/04/2025] Open
Abstract
Head and neck cancer (HNC) is the sixth most common disease in the world. The recurrence rate of patients is relatively high, and the heterogeneity of tumor immune microenvironment (TIME) cells may be an important reason for this. Single-cell sequencing (SCS) is currently the most promising and mature application in cancer research. It can identify unique genes expressed in cells and study tumor heterogeneity. According to current research, the heterogeneity of immune cells has become an important factor affecting the occurrence and development of HNC. SCSs can provide effective therapeutic targets and prognostic factors for HNC patients through analyses of gene expression levels and cell heterogeneity. Therefore, this study analyzes the basic theory of HNC and the development of SCS technology, elaborating on the application of SCS technology in HNC and its potential value in identifying HNC therapeutic targets and biomarkers.
Collapse
Affiliation(s)
- Chunhong Li
- Department of Oncology, Suining Central Hospital, Suining, Sichuan 629000, China
| | - Jia Liao
- Department of Oncology, Suining Central Hospital, Suining, Sichuan 629000, China
| | - Bo Chen
- Department of Oncology, Suining Central Hospital, Suining, Sichuan 629000, China
| | - Qiang Wang
- Gastrointestinal Surgical Unit, Suining Central Hospital, Suining, Sichuan 629000, China.
| |
Collapse
|
14
|
Zhang R, Yang M, Schreiber J, O'Day DR, Turner JMA, Shendure J, Noble WS, Disteche CM, Deng X. Cross-species imputation and comparison of single-cell transcriptomic profiles. Genome Biol 2025; 26:40. [PMID: 40012008 PMCID: PMC11863430 DOI: 10.1186/s13059-025-03493-x] [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: 10/23/2023] [Accepted: 01/31/2025] [Indexed: 02/28/2025] Open
Abstract
Cross-species comparison and prediction of gene expression profiles are important to understand regulatory changes during evolution and to transfer knowledge learned from model organisms to humans. Single-cell RNA-seq (scRNA-seq) profiles enable us to capture gene expression profiles with respect to variations among individual cells; however, cross-species comparison of scRNA-seq profiles is challenging because of data sparsity, batch effects, and the lack of one-to-one cell matching across species. Moreover, single-cell profiles are challenging to obtain in certain biological contexts, limiting the scope of hypothesis generation. Here we developed Icebear, a neural network framework that decomposes single-cell measurements into factors representing cell identity, species, and batch factors. Icebear enables accurate prediction of single-cell gene expression profiles across species, thereby providing high-resolution cell type and disease profiles in under-characterized contexts. Icebear also facilitates direct cross-species comparison of single-cell expression profiles for conserved genes that are located on the X chromosome in eutherian mammals but on autosomes in chicken. This comparison, for the first time, revealed evolutionary and diverse adaptations of X-chromosome upregulation in mammals.
Collapse
Affiliation(s)
- Ran Zhang
- Department of Genome Sciences, University of Washington, Seattle, USA
- eScience Institute, University of Washington, Seattle, USA
| | - Mu Yang
- Department of Biomedical Informatics and Medical Education, University of Washington, Seattle, USA
| | | | - Diana R O'Day
- Brotman Baty Institute for Precision Medicine, University of Washington, Seattle, USA
| | - James M A Turner
- Sex Chromosome Biology Laboratory, The Francis Crick Institute, London, UK
| | - Jay Shendure
- Department of Genome Sciences, University of Washington, Seattle, USA
- Brotman Baty Institute for Precision Medicine, University of Washington, Seattle, USA
- Howard Hughes Medical Institute, Chevy Chase, USA
- Allen Center for Cell Lineage Tracing, Seattle, USA
| | - William Stafford Noble
- Department of Genome Sciences, University of Washington, Seattle, USA.
- Paul G. Allen School of Computer Science and Engineering, University of Washington, Seattle, USA.
| | - Christine M Disteche
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, USA.
- Department of Medicine, University of Washington, Seattle, USA.
| | - Xinxian Deng
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, USA.
| |
Collapse
|
15
|
Wong KK, Ab. Hamid SS. Multiomics in silico analysis identifies TM4SF4 as a cell surface target in hepatocellular carcinoma. PLoS One 2025; 20:e0307048. [PMID: 39999090 PMCID: PMC11856526 DOI: 10.1371/journal.pone.0307048] [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: 06/28/2024] [Accepted: 01/21/2025] [Indexed: 02/27/2025] Open
Abstract
The clinical application of cellular immunotherapy in hepatocellular carcinoma (HCC) is impeded by the lack of a cell surface target frequently expressed in HCC cells and with minimal presence in normal tissues to reduce on-target, off-tumor toxicity. To address this, an in silico multomics analysis was conducted to identify an optimal therapeutic target in HCC. A longlist of genes (n = 12,948) expressed in HCCs according to The Human Protein Atlas database were examined. Eight genes were shortlisted to identify one with the highest expression in HCCs, without being shed into circulation, and with restrictive expression profile in other normal human tissues. A total of eight genes were shortlisted and subsequently ranked according to the combination of their transcript and protein expression levels in HCC cases (n = 791) derived from four independent datasets. TM4SF4 was the top-ranked target with the highest expression in HCCs. TM4SF4 showed more favorable expression profile with significantly lower expression in normal human tissues but more highly expressed in HCC compared with seven other common HCC therapeutic targets. Furthermore, scRNA-seq and immunohistochemistry datasets showed that TM4SF4 was absent in immune cell populations but highly expressed in the bile duct canaliculi of hepatocytes, regions inaccessible to immune cells. In scRNA-seq dataset of HCCs, TM4SF4 expression was positively associated with mitochondrial components and oxidative phosphorylation Gene Ontologies in HCC cells (n = 15,787 cells), suggesting its potential roles in mitochondrial-mediated oncogenic effects in HCC. Taken together, TM4SF4 is proposed as a promising cell surface target in HCC due to its high expression in HCC cells with restricted expression profile in non-cancerous tissues, and association with HCC oncogenic pathways.
Collapse
Affiliation(s)
- Kah Keng Wong
- Department of Immunology, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian, Kelantan, Malaysia
| | - Suzina Sheikh Ab. Hamid
- Tissue Bank Unit, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian, Kelantan, Malaysia
- Department of Otorhinolaryngology-Head & Neck Surgery, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian, Kelantan, Malaysia
| |
Collapse
|
16
|
Sanborn MA, Wang X, Gao S, Dai Y, Rehman J. Unveiling the cell-type-specific landscape of cellular senescence through single-cell transcriptomics using SenePy. Nat Commun 2025; 16:1884. [PMID: 39987255 PMCID: PMC11846890 DOI: 10.1038/s41467-025-57047-7] [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: 04/29/2024] [Accepted: 02/06/2025] [Indexed: 02/24/2025] Open
Abstract
Senescent cells accumulate in most tissues with organismal aging, exposure to stressors, or disease progression. It is challenging to identify senescent cells because cellular senescence signatures and phenotypes vary widely across distinct cell types and tissues. Here we developed an analytical algorithm that defines cell-type-specific and universal signatures of cellular senescence across a wide range of cell types and tissues. We utilize 72 mouse and 64 human weighted single-cell transcriptomic signatures of cellular senescence to create the SenePy scoring platform. SenePy signatures better recapitulate in vivo cellular senescence than signatures derived from in vitro senescence studies. We use SenePy to map the kinetics of senescent cell accumulation in healthy aging as well as multiple disease contexts, including tumorigenesis, inflammation, and myocardial infarction. SenePy characterizes cell-type-specific in vivo cellular senescence and could lead to the identification of genes that serve as mediators of cellular senescence and disease progression.
Collapse
Grants
- R01-AG091545 U.S. Department of Health & Human Services | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- P01HL160469 U.S. Department of Health & Human Services | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- R01-HL152515 U.S. Department of Health & Human Services | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- R01 HL152515 NHLBI NIH HHS
- R01-HL163978 U.S. Department of Health & Human Services | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- P01 HL160469 NHLBI NIH HHS
- F31-AG090005 U.S. Department of Health & Human Services | NIH | National Institute on Aging (U.S. National Institute on Aging)
- T32- HL139439 U.S. Department of Health & Human Services | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- F31 AG090005 NIA NIH HHS
- R01 HL163978 NHLBI NIH HHS
- T32 HL139439 NHLBI NIH HHS
- R01 AG091545 NIA NIH HHS
- U.S. Department of Health & Human Services | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- U.S. Department of Health & Human Services | NIH | National Institute on Aging (U.S. National Institute on Aging)
Collapse
Affiliation(s)
- Mark A Sanborn
- Department of Biochemistry and Molecular Genetics, University of Illinois, College of Medicine, Chicago, Illinois, USA.
- Center for Bioinformatics and Quantitative Biology, University of Illinois Chicago, Chicago, Illinois, USA.
| | - Xinge Wang
- Department of Biochemistry and Molecular Genetics, University of Illinois, College of Medicine, Chicago, Illinois, USA
- Center for Bioinformatics and Quantitative Biology, University of Illinois Chicago, Chicago, Illinois, USA
- Department of Biomedical Engineering, University of Illinois Chicago, College of Engineering and College of Medicine, Chicago, Illinois, USA
| | - Shang Gao
- Department of Biochemistry and Molecular Genetics, University of Illinois, College of Medicine, Chicago, Illinois, USA
- Center for Bioinformatics and Quantitative Biology, University of Illinois Chicago, Chicago, Illinois, USA
- Department of Biomedical Engineering, University of Illinois Chicago, College of Engineering and College of Medicine, Chicago, Illinois, USA
| | - Yang Dai
- Center for Bioinformatics and Quantitative Biology, University of Illinois Chicago, Chicago, Illinois, USA
- Department of Biomedical Engineering, University of Illinois Chicago, College of Engineering and College of Medicine, Chicago, Illinois, USA
| | - Jalees Rehman
- Department of Biochemistry and Molecular Genetics, University of Illinois, College of Medicine, Chicago, Illinois, USA.
- Center for Bioinformatics and Quantitative Biology, University of Illinois Chicago, Chicago, Illinois, USA.
- Department of Biomedical Engineering, University of Illinois Chicago, College of Engineering and College of Medicine, Chicago, Illinois, USA.
- University of Illinois Cancer Center, Chicago, Illinois, USA.
| |
Collapse
|
17
|
Zhao B, Song K, Wei DQ, Xiong Y, Ding J. scCobra allows contrastive cell embedding learning with domain adaptation for single cell data integration and harmonization. Commun Biol 2025; 8:233. [PMID: 39948393 PMCID: PMC11825689 DOI: 10.1038/s42003-025-07692-x] [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: 07/12/2024] [Accepted: 02/06/2025] [Indexed: 02/16/2025] Open
Abstract
The rapid advancement of single-cell technologies has created an urgent need for effective methods to integrate and harmonize single-cell data. Technical and biological variations across studies complicate data integration, while conventional tools often struggle with reliance on gene expression distribution assumptions and over-correction. Here, we present scCobra, a deep generative neural network designed to overcome these challenges through contrastive learning with domain adaptation. scCobra effectively mitigates batch effects, minimizes over-correction, and ensures biologically meaningful data integration without assuming specific gene expression distributions. It enables online label transfer across datasets with batch effects, allowing continuous integration of new data without retraining. Additionally, scCobra supports batch effect simulation, advanced multi-omic integration, and scalable processing of large datasets. By integrating and harmonizing datasets from similar studies, scCobra expands the available data for investigating specific biological problems, improving cross-study comparability, and revealing insights that may be obscured in isolated datasets.
Collapse
Affiliation(s)
- Bowen Zhao
- State Key Laboratory of Microbial Metabolism, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
- Meakins-Christie Laboratories, Department of Medicine, McGill University Health Centre, Montreal, QC, Canada
- Division of Experimental Medicine, Department of Medicine, McGill University, Montreal, QC, Canada
| | - Kailu Song
- Meakins-Christie Laboratories, Department of Medicine, McGill University Health Centre, Montreal, QC, Canada
- Quantitative Life Sciences, McGill University, Montreal, QC, Canada
| | - Dong-Qing Wei
- State Key Laboratory of Microbial Metabolism, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Yi Xiong
- State Key Laboratory of Microbial Metabolism, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China.
| | - Jun Ding
- Meakins-Christie Laboratories, Department of Medicine, McGill University Health Centre, Montreal, QC, Canada.
- Division of Experimental Medicine, Department of Medicine, McGill University, Montreal, QC, Canada.
- Quantitative Life Sciences, McGill University, Montreal, QC, Canada.
- School of Computer Science, McGill University, Montreal, QC, Canada.
- Mila-Quebec AI Institute, Montreal, QC, Canada.
| |
Collapse
|
18
|
Zhang P, Wang X, Cen X, Zhang Q, Fu Y, Mei Y, Wang X, Wang R, Wang J, Ouyang H, Liang T, Xia H, Han X, Guo G. A deep learning framework for in silico screening of anticancer drugs at the single-cell level. Natl Sci Rev 2025; 12:nwae451. [PMID: 39872221 PMCID: PMC11771446 DOI: 10.1093/nsr/nwae451] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Revised: 11/28/2024] [Accepted: 11/28/2024] [Indexed: 01/30/2025] Open
Abstract
Tumor heterogeneity plays a pivotal role in tumor progression and resistance to clinical treatment. Single-cell RNA sequencing (scRNA-seq) enables us to explore heterogeneity within a cell population and identify rare cell types, thereby improving our design of targeted therapeutic strategies. Here, we use a pan-cancer and pan-tissue single-cell transcriptional landscape to reveal heterogeneous expression patterns within malignant cells, precancerous cells, as well as cancer-associated stromal and endothelial cells. We introduce a deep learning framework named Shennong for in silico screening of anticancer drugs for targeting each of the landscape cell clusters. Utilizing Shennong, we could predict individual cell responses to pharmacologic compounds, evaluate drug candidates' tissue damaging effects, and investigate their corresponding action mechanisms. Prioritized compounds in Shennong's prediction results include FDA-approved drugs currently undergoing clinical trials for new indications, as well as drug candidates reporting anti-tumor activity. Furthermore, the tissue damaging effect prediction aligns with documented injuries and terminated discovery events. This robust and explainable framework has the potential to accelerate the drug discovery process and enhance the accuracy and efficiency of drug screening.
Collapse
Affiliation(s)
- Peijing Zhang
- Bone Marrow Transplantation Center of the First Affiliated Hospital, and Center for Stem Cell and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou 310000, China
- Liangzhu Laboratory, Zhejiang University, Hangzhou 311121, China
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Regenerative Medicine, Zhejiang University, Hangzhou 310058, China
| | - Xueyi Wang
- Bone Marrow Transplantation Center of the First Affiliated Hospital, and Center for Stem Cell and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou 310000, China
| | - Xufeng Cen
- Liangzhu Laboratory, Zhejiang University, Hangzhou 311121, China
- Research Center of Clinical Pharmacy of the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Qi Zhang
- Department of Hepatobiliary and Pancreatic Surgery, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
- MOE Joint International Research Laboratory of Pancreatic Diseases, Hangzhou 310003, China
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
- Zhejiang University Cancer Center, Hangzhou 310058, China
| | - Yuting Fu
- Bone Marrow Transplantation Center of the First Affiliated Hospital, and Center for Stem Cell and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou 310000, China
| | - Yuqing Mei
- Bone Marrow Transplantation Center of the First Affiliated Hospital, and Center for Stem Cell and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou 310000, China
| | - Xinru Wang
- Bone Marrow Transplantation Center of the First Affiliated Hospital, and Center for Stem Cell and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou 310000, China
| | - Renying Wang
- Bone Marrow Transplantation Center of the First Affiliated Hospital, and Center for Stem Cell and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou 310000, China
| | - Jingjing Wang
- Bone Marrow Transplantation Center of the First Affiliated Hospital, and Center for Stem Cell and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou 310000, China
- Liangzhu Laboratory, Zhejiang University, Hangzhou 311121, China
| | - Hongwei Ouyang
- Liangzhu Laboratory, Zhejiang University, Hangzhou 311121, China
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Regenerative Medicine, Zhejiang University, Hangzhou 310058, China
| | - Tingbo Liang
- Department of Hepatobiliary and Pancreatic Surgery, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
- MOE Joint International Research Laboratory of Pancreatic Diseases, Hangzhou 310003, China
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
- Zhejiang University Cancer Center, Hangzhou 310058, China
| | - Hongguang Xia
- Liangzhu Laboratory, Zhejiang University, Hangzhou 311121, China
- Research Center of Clinical Pharmacy of the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
- Department of Biochemistry, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Xiaoping Han
- Bone Marrow Transplantation Center of the First Affiliated Hospital, and Center for Stem Cell and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou 310000, China
- Zhejiang Key Laboratory of Multi-omics Precision Diagnosis and Treatment of Liver Diseases, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310000, China
| | - Guoji Guo
- Bone Marrow Transplantation Center of the First Affiliated Hospital, and Center for Stem Cell and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou 310000, China
- Liangzhu Laboratory, Zhejiang University, Hangzhou 311121, China
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Regenerative Medicine, Zhejiang University, Hangzhou 310058, China
- Institute of Hematology, Zhejiang University, Hangzhou 310000, China
| |
Collapse
|
19
|
Yamazaki T, Cable EE, Schnabl B. Peroxisome proliferator-activated receptor delta and liver diseases. Hepatol Commun 2025; 9:e0646. [PMID: 39899669 DOI: 10.1097/hc9.0000000000000646] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Accepted: 12/16/2024] [Indexed: 02/05/2025] Open
Abstract
Peroxisome proliferator-activated receptors (PPARs) are nuclear receptors involved in transcriptional regulation and play an important role in many physiological and metabolic processes. Unlike PPAR-alpha and PPAR-gamma, PPAR-delta is ubiquitously expressed, and its activity is key to maintaining proper metabolic homeostasis within the liver. PPAR-delta not only regulates physiologic processes of lipid, glucose, and bile acid metabolism but also attenuates pathologic responses to alcohol metabolism, inflammation, fibrosis, and carcinogenesis, and is considered an important therapeutic target in liver diseases. Promising results have been reported in clinical trials for PPAR-delta agonists in liver disease, and the selective agonist seladelpar was recently conditionally approved in the United States as a new treatment option for primary biliary cholangitis. This review provides an overview of PPAR-delta's function and biology in the liver, examines its kinetics and therapeutic potential across different liver diseases, and discusses the current status of clinical trials involving its agonists.
Collapse
Affiliation(s)
- Tomoo Yamazaki
- Department of Medicine, University of California San Diego, La Jolla, California, USA
- Department of Medicine, Division of Gastroenterology and Hepatology, Shinshu University School of Medicine, Matsumoto, Japan
| | | | - Bernd Schnabl
- Department of Medicine, University of California San Diego, La Jolla, California, USA
- Department of Medicine, VA San Diego Healthcare System, San Diego, California, USA
| |
Collapse
|
20
|
Li X, Zhao H. Targeting secretory autophagy in solid cancers: mechanisms, immune regulation and clinical insights. Exp Hematol Oncol 2025; 14:12. [PMID: 39893499 PMCID: PMC11786567 DOI: 10.1186/s40164-025-00603-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2024] [Accepted: 01/25/2025] [Indexed: 02/04/2025] Open
Abstract
Secretory autophagy is a classical form of unconventional secretion that integrates autophagy with the secretory process, relying on highly conserved autophagy-related molecules and playing a critical role in tumor progression and treatment resistance. Traditional autophagy is responsible for degrading intracellular substances by fusing autophagosomes with lysosomes. However, secretory autophagy uses autophagy signaling to mediate the secretion of specific substances and regulate the tumor microenvironment (TME). Cytoplasmic substances are preferentially secreted rather than directed toward lysosomal degradation, involving various selective mechanisms. Moreover, substances released by secretory autophagy convey biological signals to the TME, inducing immune dysregulation and contributing to drug resistance. Therefore, elucidating the mechanisms underlying secretory autophagy is essential for improving clinical treatments. This review systematically summarizes current knowledge of secretory autophagy, from initiation to secretion, considering inter-tumor heterogeneity, explores its role across different tumor types. Furthermore, it proposes future research directions and highlights unresolved clinical challenges.
Collapse
Affiliation(s)
- Xinyu Li
- Department of General Surgery, Fourth Affiliated Hospital of China Medical University, Shenyang City, 110032, Liaoning Province, China
| | - Haiying Zhao
- Department of General Surgery, Fourth Affiliated Hospital of China Medical University, Shenyang City, 110032, Liaoning Province, China.
| |
Collapse
|
21
|
Li H, Côté P, Kuoch M, Ezike J, Frenis K, Afanassiev A, Greenstreet L, Tanaka-Yano M, Tarantino G, Zhang S, Whangbo J, Butty VL, Moiso E, Falchetti M, Lu K, Connelly GG, Morris V, Wang D, Chen AF, Bianchi G, Daley GQ, Garg S, Liu D, Chou ST, Regev A, Lummertz da Rocha E, Schiebinger G, Rowe RG. The dynamics of hematopoiesis over the human lifespan. Nat Methods 2025; 22:422-434. [PMID: 39639169 PMCID: PMC11908799 DOI: 10.1038/s41592-024-02495-0] [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: 11/29/2023] [Accepted: 09/19/2024] [Indexed: 12/07/2024]
Abstract
Over a lifetime, hematopoietic stem cells (HSCs) adjust their lineage output to support age-aligned physiology. In model organisms, stereotypic waves of hematopoiesis have been observed corresponding to defined age-biased HSC hallmarks. However, how the properties of hematopoietic stem and progenitor cells change over the human lifespan remains unclear. To address this gap, we profiled individual transcriptome states of human hematopoietic stem and progenitor cells spanning gestation, maturation and aging. Here we define the gene expression networks dictating age-specific differentiation of HSCs and the dynamics of fate decisions and lineage priming throughout life. We additionally identifiy and functionally validate a fetal-specific HSC state with robust engraftment and multilineage capacity. Furthermore, we observe that classification of acute myeloid leukemia against defined transcriptional age states demonstrates that utilization of early life transcriptional programs associates with poor prognosis. Overall, we provide a disease-relevant framework for heterochronic orientation of stem cell ontogeny along the real time axis of the human lifespan.
Collapse
Affiliation(s)
- Hojun Li
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA, USA.
- Division of Hematology/Oncology, Boston Children's Hospital, Boston, MA, USA.
- Harvard Medical School, Boston, MA, USA.
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Department of Pediatrics, University of California, San Diego, CA, USA.
- Division of Hematology/Oncology, Rady Children's Hospital, San Diego, CA, USA.
| | - Parker Côté
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Pediatrics, University of California, San Diego, CA, USA
| | - Michael Kuoch
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Jideofor Ezike
- Computational and Systems Biology Program, Massachusetts Institute of Technology, Cambridge, MA, USA
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Katie Frenis
- Division of Hematology/Oncology, Boston Children's Hospital, Boston, MA, USA
| | - Anton Afanassiev
- Department of Mathematics, University of British Columbia, Vancouver, British Columbia, Canada
| | - Laura Greenstreet
- Department of Mathematics, University of British Columbia, Vancouver, British Columbia, Canada
| | - Mayuri Tanaka-Yano
- Division of Hematology/Oncology, Boston Children's Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Giuseppe Tarantino
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Stephen Zhang
- Department of Mathematics, University of British Columbia, Vancouver, British Columbia, Canada
| | - Jennifer Whangbo
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Division of Hematology/Oncology, Boston Children's Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Vor Biopharma, Cambridge, MA, USA
| | - Vincent L Butty
- Barbara K. Ostrom Bioinformatics Facility, Integrated Genomics and Bioinformatics Core of the Koch Institute, Cambridge, MA, USA
| | - Enrico Moiso
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Marcelo Falchetti
- Departments of Microbiology, Immunology and Parasitology, Federal University of Santa Catarina, Florianopolis, Brazil
| | - Kate Lu
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Guinevere G Connelly
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Vivian Morris
- Division of Hematology/Oncology, Boston Children's Hospital, Boston, MA, USA
| | - Dahai Wang
- Division of Hematology/Oncology, Boston Children's Hospital, Boston, MA, USA
| | - Antonia F Chen
- Harvard Medical School, Boston, MA, USA
- Department of Orthopedic Surgery, Brigham and Women's Hospital, Boston, MA, USA
| | - Giada Bianchi
- Harvard Medical School, Boston, MA, USA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - George Q Daley
- Division of Hematology/Oncology, Boston Children's Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Salil Garg
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Pathology, Massachusetts General Hospital, Boston, MA, USA
| | - David Liu
- Harvard Medical School, Boston, MA, USA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Stella T Chou
- Division of Hematology, Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Pediatrics, University of Pennsylvania School of Medicine, Philadelphia, PA, USA
| | - Aviv Regev
- Division of Hematology/Oncology, Rady Children's Hospital, San Diego, CA, USA
- Genentech, South San Francisco, CA, USA
| | - Edroaldo Lummertz da Rocha
- Departments of Microbiology, Immunology and Parasitology, Federal University of Santa Catarina, Florianopolis, Brazil
| | - Geoffrey Schiebinger
- Department of Mathematics, University of British Columbia, Vancouver, British Columbia, Canada
| | - R Grant Rowe
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA, USA.
- Division of Hematology/Oncology, Boston Children's Hospital, Boston, MA, USA.
- Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
22
|
Sun Y, Huang Q, Sun J, Zhou H, Guo D, Peng L, Lin H, Li C, Shang H, Wang T, Chen Y, Huang Y, Hu C, Hu Z, Lu Y, Peng H. Mucosal-Associated Invariant T (MAIT) Cell-Mediated Immune Mechanisms of Peritoneal Dialysis-Induced Peritoneal Fibrosis and Therapeutic Targeting. J Am Soc Nephrol 2025:00001751-990000000-00539. [PMID: 39874111 DOI: 10.1681/asn.0000000627] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Accepted: 01/22/2025] [Indexed: 01/30/2025] Open
Abstract
Key Points
Peritoneal mucosal-associated invariant T (MAIT) cells were characterized by single-cell RNA sequencing, histological imaging, and flow cytometry.Activation of MAIT cells modulated glucose metabolism in mesothelial cells by TCRVα7.2-MHC class 1–related protein 1 signaling and triggered peritoneal fibrogenesis.Pharmacological inhibition of MAIT cell function by acetyl-6-formylpterin mitigated peritoneal fibrosis.
Background
Peritoneal fibrosis is a serious complication of long-term peritoneal dialysis (PD) and abdominal surgeries, yet effective treatments remain elusive. Given the known roles of mucosal-associated invariant T (MAIT) cells in immune responses and fibrotic diseases, we investigated their involvement in PD-induced peritoneal fibrosis to identify potential therapeutic targets.
Methods
We used single-cell RNA sequencing and flow cytometry to characterize the activation and function of peritoneal MAIT cells in patients undergoing long-term PD. Our investigation focused on the molecular pathways activated by these cells, particularly the MHC class 1–related protein 1 (MR1)-mediated interaction with mesothelial cells and subsequent activation of the mTOR complex 1 signaling pathway. We further assessed the effect of inhibiting MAIT cells on fibrogenesis using both in vitro models and Mr1 knockout mice.
Results
Our study revealed that long-term PD significantly enhanced the activation of MAIT cells, particularly the proinflammatory MAIT17 subtype. These activated cells contributed to peritoneal fibrogenesis by binding to the MR1 receptor on mesothelial cells, which triggered hyperglycolysis through the mTOR complex 1 pathway, ultimately leading to fibrogenesis. Notably, we demonstrated that blocking the MR1–MAIT interaction, either through genetic knockout or pharmacological inhibition with acetyl-6-formylpterin, effectively mitigated fibrosis.
Conclusions
This study identified MAIT cells as crucial drivers of PD-induced peritoneal fibrosis.
Collapse
Affiliation(s)
- Yuxiang Sun
- Nephrology Division, Department of Medicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- NHC Key Laboratory of Clinical Nephrology (Sun Yat-sen University) and Guangdong Provincial Key Laboratory of Nephrology, Guangzhou, China
| | - Qiang Huang
- Nephrology Division, Department of Medicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- NHC Key Laboratory of Clinical Nephrology (Sun Yat-sen University) and Guangdong Provincial Key Laboratory of Nephrology, Guangzhou, China
| | - Juan Sun
- Nephrology Division, Department of Medicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Hu Zhou
- Nephrology Division, Department of Medicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Dandan Guo
- Nephrology Division, Department of Medicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Long Peng
- Division of Cardiovascular Medicine, Department of Medicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Hongchun Lin
- Nephrology Division, Department of Medicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Canming Li
- Nephrology Division, Department of Medicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Hongli Shang
- Nephrology Division, Department of Medicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Tongtong Wang
- Department of Clinical Immunology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Yanxu Chen
- Organ Transplant Center, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Yong Huang
- Division of Gastrointestinal Surgery, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Cheng Hu
- Department of Urology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Zhaoyong Hu
- Nephrology Division, Department of Medicine, Baylor College of Medicine, Houston, Texas
| | - Yan Lu
- Department of Clinical Immunology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Hui Peng
- Nephrology Division, Department of Medicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- NHC Key Laboratory of Clinical Nephrology (Sun Yat-sen University) and Guangdong Provincial Key Laboratory of Nephrology, Guangzhou, China
| |
Collapse
|
23
|
Li Y, Zhu J, Yue C, Song S, Tian L, Wang Y. Recent advances in pancreatic α-cell transdifferentiation for diabetes therapy. Front Immunol 2025; 16:1551372. [PMID: 39911402 PMCID: PMC11794509 DOI: 10.3389/fimmu.2025.1551372] [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: 12/25/2024] [Accepted: 01/07/2025] [Indexed: 02/07/2025] Open
Abstract
As the global prevalence of diabetes mellitus rises, traditional treatments like insulin therapy and oral hypoglycemic agents often fail to achieve optimal glycemic control, leading to severe complications. Recent research has focused on replenishing pancreatic β-cells through the transdifferentiation of α-cells, offering a promising therapeutic avenue. This review explores the molecular mechanisms underlying α-cell to β-cell transdifferentiation, emphasizing key transcription factors such as Dnmt1, Arx, Pdx1, MafA, and Nkx6.1. The potential clinical applications, especially in type 1 and type 2 diabetes characterized by significant β-cell dysfunction, are addressed. Challenges, including low transdifferentiation efficiency, cell stability, and safety concerns, are also included. Future research directions include optimizing molecular pathways, enhancing transdifferentiation efficiency, and ensuring the long-term stability of β-cell identity. Overall, the ability to convert α-cells into β-cells represents a transformative strategy for diabetes treatment, offering hope for more effective and sustainable therapies for patients with severe β-cell loss.
Collapse
Affiliation(s)
- Yanjiao Li
- Department of Pharmacy, Qionglai Hospital of Traditional Chinese Medicine, Chengdu, China
- Clinical Immunology Translational Medicine Key Laboratory of Sichuan Province, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Jinyu Zhu
- Center for Geriatrics and Endocrinology, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Congyang Yue
- Center for Geriatrics and Endocrinology, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Siyuan Song
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, United States
| | - Limin Tian
- Center for Geriatrics and Endocrinology, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Yi Wang
- Clinical Immunology Translational Medicine Key Laboratory of Sichuan Province, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
- Center for Geriatrics and Endocrinology, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
- Center for Critical Care Medicine, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| |
Collapse
|
24
|
Pang Y, Qin Y, Du Z, Liu Q, Zhang J, Han K, Lu J, Yuan Z, Li J, Pan S, Dong X, Xu M, Wang D, Li S, Li Z, Chen Y, Zhao Z, Zhang Z, Chuan S, Song Y, Sun M, Jia X, Xia Z, Zhan L, Yue Z, Cui W, Wang J, Gu Y, Ni M, Yang H, Xu X, Liu X, Li Q, Fan G. Single-cell transcriptome atlas of lamprey exploring Natterin- induced white adipose tissue browning. Nat Commun 2025; 16:752. [PMID: 39820434 PMCID: PMC11739602 DOI: 10.1038/s41467-025-56153-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: 03/28/2024] [Accepted: 01/09/2025] [Indexed: 01/19/2025] Open
Abstract
Lampreys are early jawless vertebrates that are the key to understanding the evolution of vertebrates. However, the lack of cytomic studies on multiple lamprey organs has hindered progress in this field. Therefore, the present study constructed a comprehensive cell atlas comprising 604,460 cells/nuclei and 70 cell types from 14 lamprey tissue samples. Comparison of cellular evolution across species revealed that most lamprey cell types are homologous to those in jawed vertebrates. We discovered acinar- and islet-like cell populations despite the lack of parenchymal organs in lampreys, providing evidence of pancreatic function in vertebrates. Furthermore, we investigated the heterogeneity of lamprey immune cell populations. Natterin was highly expressed in granulocytes, and NATTERIN was localized to the lipid droplets. Moreover, we developed a transgenic mouse model expressing Natterin to elucidate the role of NATTERIN in lipid metabolism, whereas the browning of white adipose tissue was induced. These findings elucidate vertebrate cellular evolution and advance our understanding of adipose tissue plasticity and metabolic regulation in lampreys.
Collapse
Affiliation(s)
- Yue Pang
- College of Life Science, Liaoning Normal University, Dalian, 116081, China.
- Lamprey Research Center, Liaoning Normal University, Dalian, 116081, China.
| | - Yating Qin
- BGI Research, Qingdao, 266555, China
- Qingdao Key Laboratory of Marine Genomics, BGI Research, Qingdao, 266555, China
- Department of Biology, University of Copenhagen, Copenhagen, 2100, Denmark
- BGI Research, Hangzhou, 310030, China
| | - Zeyu Du
- College of Life Science, Liaoning Normal University, Dalian, 116081, China
- Lamprey Research Center, Liaoning Normal University, Dalian, 116081, China
| | - Qun Liu
- BGI Research, Qingdao, 266555, China
- Qingdao Key Laboratory of Marine Genomics, BGI Research, Qingdao, 266555, China
- Department of Biology, University of Copenhagen, Copenhagen, 2100, Denmark
| | - Jin Zhang
- College of Life Science, Liaoning Normal University, Dalian, 116081, China
- Lamprey Research Center, Liaoning Normal University, Dalian, 116081, China
| | - Kai Han
- BGI Research, Qingdao, 266555, China
- Qingdao Key Laboratory of Marine Genomics, BGI Research, Qingdao, 266555, China
- Department of Biology, University of Copenhagen, Copenhagen, 2100, Denmark
| | - Jiali Lu
- College of Life Science, Liaoning Normal University, Dalian, 116081, China
- Lamprey Research Center, Liaoning Normal University, Dalian, 116081, China
| | - Zengbao Yuan
- BGI Research, Qingdao, 266555, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Jun Li
- College of Life Science, Liaoning Normal University, Dalian, 116081, China
- Lamprey Research Center, Liaoning Normal University, Dalian, 116081, China
| | | | - Xinrui Dong
- College of Life Science, Liaoning Normal University, Dalian, 116081, China
- Lamprey Research Center, Liaoning Normal University, Dalian, 116081, China
| | - Mengyang Xu
- BGI Research, Qingdao, 266555, China
- BGI Research, Shenzhen, 518083, China
- Shenzhen Key Laboratory of marine biology genomics, BGI Research, Shenzhen, 518083, China
| | - Dantong Wang
- BGI Research, Qingdao, 266555, China
- BGI Research, Shenzhen, 518083, China
| | - Shuo Li
- State Key Laboratory of Mariculture Biobreeding and Sustainable Goods, Yellow Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Qingdao, Shandong, 266071, China
| | - Zhen Li
- BGI Research, Qingdao, 266555, China
| | | | - Zhisheng Zhao
- College of Life Science, Liaoning Normal University, Dalian, 116081, China
- Lamprey Research Center, Liaoning Normal University, Dalian, 116081, China
| | | | - Shunqin Chuan
- College of Life Science, Liaoning Normal University, Dalian, 116081, China
- Lamprey Research Center, Liaoning Normal University, Dalian, 116081, China
| | - Yue Song
- BGI Research, Qingdao, 266555, China
- Department of Biology, University of Copenhagen, Copenhagen, 2100, Denmark
| | - Mingjie Sun
- College of Life Science, Liaoning Normal University, Dalian, 116081, China
- Lamprey Research Center, Liaoning Normal University, Dalian, 116081, China
| | - Xiaodong Jia
- Joint Laboratory for Translational Medicine Research, Liaocheng People's Hospital, Liaocheng, Shandong, 252000, China
| | - Zhangyong Xia
- Department of Neurology, The Second People's Hospital of Liaocheng, Liaocheng, Shandong, 252000, China
| | | | - Zhen Yue
- BGI Research, Sanya, 572025, China
| | - Wei Cui
- BGI Research, Qingdao, 266555, China
| | - Jun Wang
- BGI Research, Qingdao, 266555, China
| | - Ying Gu
- BGI Research, Shenzhen, 518083, China
- BGI, Shenzhen, 518083, China
| | - Ming Ni
- MGI Tech, Shenzhen, 518083, China
| | - Huanming Yang
- BGI Research, Shenzhen, 518083, China
- BGI, Shenzhen, 518083, China
| | - Xun Xu
- BGI Research, Shenzhen, 518083, China
- BGI, Shenzhen, 518083, China
| | - Xin Liu
- BGI Research, Shenzhen, 518083, China.
- BGI, Shenzhen, 518083, China.
| | - Qingwei Li
- College of Life Science, Liaoning Normal University, Dalian, 116081, China.
- Lamprey Research Center, Liaoning Normal University, Dalian, 116081, China.
| | - Guangyi Fan
- BGI Research, Qingdao, 266555, China.
- Qingdao Key Laboratory of Marine Genomics, BGI Research, Qingdao, 266555, China.
- BGI Research, Hangzhou, 310030, China.
- BGI Research, Shenzhen, 518083, China.
- Shenzhen Key Laboratory of marine biology genomics, BGI Research, Shenzhen, 518083, China.
- BGI Research, Sanya, 572025, China.
| |
Collapse
|
25
|
Yu X, Zheng G, Xu L, Guo W, Chen G, Zhu Y, Li T, Rao M, Wang L, Cong R, Pei H. MobiChIP: a compatible library construction method of single-cell ChIP-seq based droplets. Mol Omics 2025; 21:32-37. [PMID: 39513632 DOI: 10.1039/d4mo00111g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2024]
Abstract
To illustrate epigenetic heterogeneity, versatile tools of single-cell ChIP-seq (scChIP-seq) are essential for both convenience and accuracy. We developed MobiChIP, a compatible ChIP-seq library construction method based on current sequencing platforms for single-cell applications. MobiChIP efficiently captures fragments from tagmented nuclei across various species and allows sample mixing from different tissues or species. This strategy offers robust nucleosome amplification and flexible sequencing without customized primers. MobiChIP reveals regulatory landscapes of chromatin with active (H3K27ac) and repressive (H3K27me3) histone modification in peripheral blood mononuclear cells (PBMCs) and accurately identifies epigenetic repression of the Hox gene cluster, outperforming ATAC-seq. Meanwhile, we also integrated scChIP-seq with scRNA-seq to further illustrate cellular genetic and epigenetic heterogeneity.
Collapse
Affiliation(s)
- Xianhong Yu
- The Key Laboratory for Biomedical Photonics of MOE at Wuhan National Laboratory for Optoelectronics - Hubei Bioinformatics and Molecular Imaging Key Laboratory, Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
- Shanghai MobiDrop Co., Ltd., Room 351, Building 1, Guoshoujing Road, Shanghai Free Trade Pilot Zone, Shanghai, 200000, China.
| | - Guantao Zheng
- Shanghai MobiDrop Co., Ltd., Room 351, Building 1, Guoshoujing Road, Shanghai Free Trade Pilot Zone, Shanghai, 200000, China.
| | - Liting Xu
- MobiDrop (Zhejiang) Co., Ltd., No. 1888 Longxiang Avenue, Tongxiang, Zhejiang Province, 314500, China.
| | - Weiyi Guo
- The Key Laboratory for Biomedical Photonics of MOE at Wuhan National Laboratory for Optoelectronics - Hubei Bioinformatics and Molecular Imaging Key Laboratory, Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
- MobiDrop (Zhejiang) Co., Ltd., No. 1888 Longxiang Avenue, Tongxiang, Zhejiang Province, 314500, China.
| | - Guodong Chen
- Shanghai MobiDrop Co., Ltd., Room 351, Building 1, Guoshoujing Road, Shanghai Free Trade Pilot Zone, Shanghai, 200000, China.
| | - Yiling Zhu
- MobiDrop (Zhejiang) Co., Ltd., No. 1888 Longxiang Avenue, Tongxiang, Zhejiang Province, 314500, China.
| | - Tingting Li
- Shanghai MobiDrop Co., Ltd., Room 351, Building 1, Guoshoujing Road, Shanghai Free Trade Pilot Zone, Shanghai, 200000, China.
| | - Mingming Rao
- Shanghai MobiDrop Co., Ltd., Room 351, Building 1, Guoshoujing Road, Shanghai Free Trade Pilot Zone, Shanghai, 200000, China.
| | - Linyan Wang
- Eye Center, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Zhejiang Provincial Key Laboratory of Ophthalmology, Zhejiang Provincial Clinical Research Center for Eye Diseases, Zhejiang Provincial Engineering Institute on Eye Diseases, Hangzhou, Zhejiang, China
| | - Rong Cong
- MobiDrop (Zhejiang) Co., Ltd., No. 1888 Longxiang Avenue, Tongxiang, Zhejiang Province, 314500, China.
| | - Hao Pei
- MobiDrop (Zhejiang) Co., Ltd., No. 1888 Longxiang Avenue, Tongxiang, Zhejiang Province, 314500, China.
| |
Collapse
|
26
|
Yang T, Zhang N, Yang N. Single-cell sequencing in diabetic retinopathy: progress and prospects. J Transl Med 2025; 23:49. [PMID: 39806376 PMCID: PMC11727737 DOI: 10.1186/s12967-024-06066-x] [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: 10/11/2024] [Accepted: 12/30/2024] [Indexed: 01/16/2025] Open
Abstract
Diabetic retinopathy is a major ocular complication of diabetes, characterized by progressive retinal microvascular damage and significant visual impairment in working-age adults. Traditional bulk RNA sequencing offers overall gene expression profiles but does not account for cellular heterogeneity. Single-cell RNA sequencing overcomes this limitation by providing transcriptomic data at the individual cell level and distinguishing novel cell subtypes, developmental trajectories, and intercellular communications. Researchers can use single-cell sequencing to draw retinal cell atlases and identify the transcriptomic features of retinal cells, enhancing our understanding of the pathogenesis and pathological changes in diabetic retinopathy. Additionally, single-cell sequencing is widely employed to analyze retinal organoids and single extracellular vesicles. Single-cell multi-omics sequencing integrates omics information, whereas stereo-sequencing analyzes gene expression and spatiotemporal data simultaneously. This review discusses the protocols of single-cell sequencing for obtaining single cells from retina and accurate sequencing data. It highlights the applications and advancements of single-cell sequencing in the study of normal retinas and the pathological changes associated with diabetic retinopathy. This underscores the potential of these technologies to deepen our understanding of the pathogenesis of diabetic retinopathy that may lead to the introduction of new therapeutic strategies.
Collapse
Affiliation(s)
- Tianshu Yang
- Department of Ophthalmology, Renmin Hospital of Wuhan University, Jiefang Road, Wuhan, Hubei, 430060, China
| | - Ningzhi Zhang
- Department of Ophthalmology, Renmin Hospital of Wuhan University, Jiefang Road, Wuhan, Hubei, 430060, China
| | - Ning Yang
- Department of Ophthalmology, Renmin Hospital of Wuhan University, Jiefang Road, Wuhan, Hubei, 430060, China.
| |
Collapse
|
27
|
Zhong H, Han W, Gomez-Cabrero D, Tegner J, Gao X, Cui G, Aranda M. Benchmarking cross-species single-cell RNA-seq data integration methods: towards a cell type tree of life. Nucleic Acids Res 2025; 53:gkae1316. [PMID: 39778870 PMCID: PMC11707536 DOI: 10.1093/nar/gkae1316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2024] [Revised: 11/23/2024] [Accepted: 12/27/2024] [Indexed: 01/11/2025] Open
Abstract
Cross-species single-cell RNA-seq data hold immense potential for unraveling cell type evolution and transferring knowledge between well-explored and less-studied species. However, challenges arise from interspecific genetic variation, batch effects stemming from experimental discrepancies and inherent individual biological differences. Here, we benchmarked nine data-integration methods across 20 species, encompassing 4.7 million cells, spanning eight phyla and the entire animal taxonomic hierarchy. Our evaluation reveals notable differences between the methods in removing batch effects and preserving biological variance across taxonomic distances. Methods that effectively leverage gene sequence information capture underlying biological variances, while generative model-based approaches excel in batch effect removal. SATURN demonstrates robust performance across diverse taxonomic levels, from cross-genus to cross-phylum, emphasizing its versatility. SAMap excels in integrating species beyond the cross-family level, especially for atlas-level cross-species integration, while scGen shines within or below the cross-class hierarchy. As a result, our analysis offers recommendations and guidelines for selecting suitable integration methods, enhancing cross-species single-cell RNA-seq analyses and advancing algorithm development.
Collapse
Affiliation(s)
- Huawen Zhong
- BioEngineering Program, Biological and Environmental Science and Engineering Division, King Abdullah University of Science and Technology (KAUST), Thuwal 23955-6900, Kingdom of Saudi Arabia
| | - Wenkai Han
- Computer Science Program, Computer, Electrical and Mathematical Sciences and Engineering Division, King Abdullah University of Science and Technology (KAUST), Thuwal 23955-6900, Kingdom of Saudi Arabia
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - David Gomez-Cabrero
- BioEngineering Program, Biological and Environmental Science and Engineering Division, King Abdullah University of Science and Technology (KAUST), Thuwal 23955-6900, Kingdom of Saudi Arabia
- Unit of Translational Bioinformatics, Navarrabiomed—Fundación Miguel Servet, Universidad Pública de Navarra (UPNA), IdiSNA, Pamplona, Spain
| | - Jesper Tegner
- BioEngineering Program, Biological and Environmental Science and Engineering Division, King Abdullah University of Science and Technology (KAUST), Thuwal 23955-6900, Kingdom of Saudi Arabia
- Computer Science Program, Computer, Electrical and Mathematical Sciences and Engineering Division, King Abdullah University of Science and Technology (KAUST), Thuwal 23955-6900, Kingdom of Saudi Arabia
- Unit of Computational Medicine, Department of Medicine, Center for Molecular Medicine, Karolinska Institutet, Karolinska University Hospital, L8:05, SE-171 76 Stockholm, Sweden
- Science for Life Laboratory, Tomtebodavagen 23A, SE-17165 Solna, Sweden
| | - Xin Gao
- Computer Science Program, Computer, Electrical and Mathematical Sciences and Engineering Division, King Abdullah University of Science and Technology (KAUST), Thuwal 23955-6900, Kingdom of Saudi Arabia
- Center of Excellence on Smart Health, King Abdullah University of Science and Technology (KAUST), Thuwal 23955-6900, Kingdom of Saudi Arabia
- Center of Excellence for Generative AI, King Abdullah University of Science and Technology (KAUST), Thuwal 23955-6900, Kingdom of Saudi Arabia
| | - Guoxin Cui
- BioEngineering Program, Biological and Environmental Science and Engineering Division, King Abdullah University of Science and Technology (KAUST), Thuwal 23955-6900, Kingdom of Saudi Arabia
- Marine Science Program, Biological and Environmental Science and Engineering Division, King Abdullah University of Science and Technology (KAUST), Thuwal 23955-6900, Kingdom of Saudi Arabia
| | - Manuel Aranda
- BioEngineering Program, Biological and Environmental Science and Engineering Division, King Abdullah University of Science and Technology (KAUST), Thuwal 23955-6900, Kingdom of Saudi Arabia
- Marine Science Program, Biological and Environmental Science and Engineering Division, King Abdullah University of Science and Technology (KAUST), Thuwal 23955-6900, Kingdom of Saudi Arabia
| |
Collapse
|
28
|
Utkina M, Shcherbakova A, Deviatiiarov R, Ryabova A, Loguinova M, Trofimov V, Kuznetsova A, Petropavlovskiy M, Salimkhanov R, Maksimov D, Albert E, Golubeva A, Asaad W, Urusova L, Bondarenko E, Lapshina A, Shutova A, Beltsevich D, Gusev O, Dzeranova L, Melnichenko G, Minniakhmetov I, Dedov I, Mokrysheva N, Popov S. Comparative evaluation of ACetic - MEthanol high salt dissociation approach for single-cell transcriptomics of frozen human tissues. Front Cell Dev Biol 2025; 12:1469955. [PMID: 39839668 PMCID: PMC11748064 DOI: 10.3389/fcell.2024.1469955] [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: 07/24/2024] [Accepted: 11/20/2024] [Indexed: 01/23/2025] Open
Abstract
Current dissociation methods for solid tissues in scRNA-seq studies do not guarantee intact single-cell isolation, especially for sensitive and complex human endocrine tissues. Most studies rely on enzymatic dissociation of fresh samples or nuclei isolation from frozen samples. Dissociating whole intact cells from fresh-frozen samples, commonly collected by biobanks, remains a challenge. Here, we utilized the acetic-methanol dissociation approach (ACME) to capture transcriptional profiles of individual cells from fresh-frozen tissue samples. This method combines acetic acid-based dissociation and methanol-based fixation. In our study, we optimized this approach for human endocrine tissue samples for the first time. We incorporated a high-salt washing buffer instead of the standard PBS to stabilize RNA and prevent RNases reactivation during rehydration. We have designated this optimized protocol as ACME HS (ACetic acid-MEthanol High Salt). This technique aims to preserve cell morphology and RNA integrity, minimizing transcriptome changes and providing a more accurate representation of mature mRNA. We compared the ability of enzymatic, ACME HS, and nuclei isolation methods to preserve major cell types, gene expression, and standard quality parameters across 41 tissue samples. Our results demonstrated that ACME HS effectively dissociates and fixes cells, preserving cell morphology and high RNA integrity. This makes ACME HS a valuable alternative for scRNA-seq protocols involving challenging tissues where obtaining a live cell suspension is difficult or disruptive.
Collapse
Affiliation(s)
- Marina Utkina
- Endocrinology Research Centre, Institute of Personalized Medicine, Moscow, Russia
| | | | - Ruslan Deviatiiarov
- Endocrinology Research Centre, Institute of Personalized Medicine, Moscow, Russia
- Graduate School of Medicine, Juntendo University, Bunkyo-ku, Japan
- Life Improvement by Future Technologies (LIFT) Center, Moscow, Russia
| | - Alina Ryabova
- Endocrinology Research Centre, Institute of Personalized Medicine, Moscow, Russia
| | - Marina Loguinova
- Endocrinology Research Centre, Institute of Personalized Medicine, Moscow, Russia
| | - Valentin Trofimov
- Endocrinology Research Centre, Institute of Personalized Medicine, Moscow, Russia
| | - Anna Kuznetsova
- Endocrinology Research Centre, Institute of Personalized Medicine, Moscow, Russia
| | | | - Rustam Salimkhanov
- Endocrinology Research Centre, Institute of Personalized Medicine, Moscow, Russia
| | - Denis Maksimov
- Faculty of Medicine, Lomonosov Moscow State University, Moscow, Russia
| | - Eugene Albert
- Faculty of Medicine, Lomonosov Moscow State University, Moscow, Russia
| | - Alexandra Golubeva
- Endocrinology Research Centre, Institute of Personalized Medicine, Moscow, Russia
| | - Walaa Asaad
- Endocrinology Research Centre, Institute of Personalized Medicine, Moscow, Russia
| | - Lilia Urusova
- Endocrinology Research Centre, Institute of Personalized Medicine, Moscow, Russia
| | - Ekaterina Bondarenko
- Endocrinology Research Centre, Institute of Personalized Medicine, Moscow, Russia
| | - Anastasia Lapshina
- Endocrinology Research Centre, Institute of Personalized Medicine, Moscow, Russia
| | - Alexandra Shutova
- Endocrinology Research Centre, Institute of Personalized Medicine, Moscow, Russia
| | - Dmitry Beltsevich
- Endocrinology Research Centre, Institute of Personalized Medicine, Moscow, Russia
| | - Oleg Gusev
- Endocrinology Research Centre, Institute of Personalized Medicine, Moscow, Russia
- Graduate School of Medicine, Juntendo University, Bunkyo-ku, Japan
- Life Improvement by Future Technologies (LIFT) Center, Moscow, Russia
- Regulatory Genomics Research Center, Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia
| | - Larisa Dzeranova
- Endocrinology Research Centre, Institute of Personalized Medicine, Moscow, Russia
| | - Galina Melnichenko
- Endocrinology Research Centre, Institute of Personalized Medicine, Moscow, Russia
| | - Ildar Minniakhmetov
- Endocrinology Research Centre, Institute of Personalized Medicine, Moscow, Russia
| | - Ivan Dedov
- Endocrinology Research Centre, Institute of Personalized Medicine, Moscow, Russia
| | - Natalya Mokrysheva
- Endocrinology Research Centre, Institute of Personalized Medicine, Moscow, Russia
| | - Sergey Popov
- Endocrinology Research Centre, Institute of Personalized Medicine, Moscow, Russia
| |
Collapse
|
29
|
Jia Y, Dong H, Li L, Wang F, Juan L, Wang Y, Guo H, Zhao T. xQTLatlas: a comprehensive resource for human cellular-resolution multi-omics genetic regulatory landscape. Nucleic Acids Res 2025; 53:D1270-D1277. [PMID: 39351883 PMCID: PMC11701524 DOI: 10.1093/nar/gkae837] [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: 07/22/2024] [Revised: 08/26/2024] [Accepted: 09/13/2024] [Indexed: 10/03/2024] Open
Abstract
Understanding how genetic variants influence molecular phenotypes in different cellular contexts is crucial for elucidating the molecular and cellular mechanisms behind complex traits, which in turn has spurred significant advances in research into molecular quantitative trait locus (xQTL) at the cellular level. With the rapid proliferation of data, there is a critical need for a comprehensive and accessible platform to integrate this information. To meet this need, we developed xQTLatlas (http://www.hitxqtl.org.cn/), a database that provides a multi-omics genetic regulatory landscape at cellular resolution. xQTLatlas compiles xQTL summary statistics from 151 cell types and 339 cell states across 55 human tissues. It organizes these data into 20 xQTL types, based on four distinct discovery strategies, and spans 13 molecular phenotypes. Each entry in xQTLatlas is meticulously annotated with comprehensive metadata, including the origin of the tissue, cell type, cell state and the QTL discovery strategies utilized. Additionally, xQTLatlas features multiscale data exploration tools and a suite of interactive visualizations, facilitating in-depth analysis of cell-level xQTL. xQTLatlas provides a valuable resource for deepening our understanding of the impact of functional variants on molecular phenotypes in different cellular environments, thereby facilitating extensive research efforts.
Collapse
Affiliation(s)
- Yuran Jia
- Faculty of Computing, Harbin Institute of Technology, Harbin 150001, China
| | - Hongchao Dong
- Faculty of Computing, Harbin Institute of Technology, Harbin 150001, China
| | - Linhao Li
- School of Medicine and Health, Harbin Institute of Technology, Harbin 150001, China
| | - Fang Wang
- Faculty of Computing, Harbin Institute of Technology, Harbin 150001, China
| | - Liran Juan
- School of Life Science and Technology, Harbin Institute of Technology, Harbin 150001, China
| | - Yadong Wang
- School of Medicine and Health, Harbin Institute of Technology, Harbin 150001, China
- Zhengzhou Research Institute, Harbin Institute of Technology, Harbin 450000, China
| | - Hongzhe Guo
- Faculty of Computing, Harbin Institute of Technology, Harbin 150001, China
- Zhengzhou Research Institute, Harbin Institute of Technology, Harbin 450000, China
| | - Tianyi Zhao
- Faculty of Computing, Harbin Institute of Technology, Harbin 150001, China
- School of Medicine and Health, Harbin Institute of Technology, Harbin 150001, China
- Zhengzhou Research Institute, Harbin Institute of Technology, Harbin 450000, China
| |
Collapse
|
30
|
Park CJ, Oh JE, Lin P, Zhou S, Bunnell M, Bikorimana E, Spinella MJ, Lim HJ, Ko CJ. A Dynamic Shift in Estrogen Receptor Expression During Granulosa Cell Differentiation in the Ovary. Endocrinology 2025; 166:bqaf006. [PMID: 39834231 DOI: 10.1210/endocr/bqaf006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2024] [Revised: 12/29/2024] [Accepted: 01/18/2025] [Indexed: 01/22/2025]
Abstract
This study uncovers a dynamic shift in estrogen receptor expression during granulosa cell (GC) differentiation in the ovary, highlighting a transition from estrogen receptor alpha (ESR1) to estrogen receptor beta (ESR2). Using a transgenic mouse model with Esr1-iCre-mediated Esr2 deletion, we demonstrate that ESR2 expression is absent in GCs derived from ESR1-expressing ovarian surface epithelium (OSE) cells. Single-cell analysis of the OSE-GC lineage reveals a developmental trajectory from Esr1-expressing OSE cells to Foxl2-expressing pre-GCs, culminating in GCs exclusively expressing Esr2. Transcriptome analyses identified vasculature-derived TGFβ1 ligands as key regulators of this transition. Supporting this, TGFβ1 treatment of cultured embryonic ovaries reduced Esr1 expression while promoting Esr2 expression. This study underscores the capability of GCs to switch from ESR1 to ESR2 expression as a fundamental aspect of normal differentiation.
Collapse
Affiliation(s)
- Chan Jin Park
- Department of Comparative Biosciences, College of Veterinary Medicine, University of Illinois at Urbana-Champaign, Urbana, IL 61802, USA
- Epivara, Inc., Research Park, Champaign, IL 61820, USA
| | - Ji-Eun Oh
- Department of Comparative Biosciences, College of Veterinary Medicine, University of Illinois at Urbana-Champaign, Urbana, IL 61802, USA
| | - PoChing Lin
- Department of Comparative Biosciences, College of Veterinary Medicine, University of Illinois at Urbana-Champaign, Urbana, IL 61802, USA
| | - Sherry Zhou
- Epivara, Inc., Research Park, Champaign, IL 61820, USA
| | - Mary Bunnell
- Department of Comparative Biosciences, College of Veterinary Medicine, University of Illinois at Urbana-Champaign, Urbana, IL 61802, USA
| | - Emmanuel Bikorimana
- Department of Comparative Biosciences, College of Veterinary Medicine, University of Illinois at Urbana-Champaign, Urbana, IL 61802, USA
| | - Michael J Spinella
- Department of Comparative Biosciences, College of Veterinary Medicine, University of Illinois at Urbana-Champaign, Urbana, IL 61802, USA
| | - Hyunjung Jade Lim
- Department of Veterinary Medicine, Konkuk University, Gwangjin-gu, Seoul 05029, Korea
| | - CheMyong J Ko
- Department of Comparative Biosciences, College of Veterinary Medicine, University of Illinois at Urbana-Champaign, Urbana, IL 61802, USA
| |
Collapse
|
31
|
Guo Q, Liu Q, He D, Xin M, Dai Y, Sun R, Li H, Zhang Y, Li J, Kong C, Gao Y, Zhi H, Li F, Ning S, Wang P. LnCeCell 2.0: an updated resource for lncRNA-associated ceRNA networks and web tools based on single-cell and spatial transcriptomics sequencing data. Nucleic Acids Res 2025; 53:D107-D115. [PMID: 39470723 PMCID: PMC11701739 DOI: 10.1093/nar/gkae947] [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: 09/10/2024] [Revised: 09/29/2024] [Accepted: 10/08/2024] [Indexed: 10/30/2024] Open
Abstract
We describe LnCeCell 2.0 (http://bio-bigdata.hrbmu.edu.cn/LnCeCell), an updated resource for lncRNA-associated competing endogenous RNA (ceRNA) networks and web tools based on single-cell and spatial transcriptomics sequencing (stRNA-seq) data. We have updated the LnCeCell 2.0 database with significantly expanded data and improved features, including (i) 257 single-cell RNA sequencing and stRNA-seq datasets across 86 diseases/phenotypes and 80 human normal tissues, (ii) 836 581 cell-specific and spatial spot-specific ceRNA interactions and functional networks for 1 002 988 cells and 367 971 spatial spots, (iii) 15 489 experimentally supported lncRNA biomarkers related to disease pathology, diagnosis and treatment, (iv) detailed annotation of cell type, cell state, subcellular and extracellular locations of ceRNAs through manual curation and (v) ceRNA expression profiles and follow-up clinical information of 20 326 cancer patients. Further, a panel of 24 flexible tools (including 8 comprehensive and 16 mini-analysis tools) was developed to investigate ceRNA-regulated mechanisms at single-cell/spot resolution. The CeCellTraject tool, for example, illustrates the detailed ceRNA distribution of different cell populations and explores the dynamic change of the ceRNA network along the developmental trajectory. LnCeCell 2.0 will facilitate the study of fine-tuned lncRNA-ceRNA networks with single-cell and spatial spot resolution, helping us to understand the regulatory mechanisms behind complex microbial ecosystems.
Collapse
Affiliation(s)
- Qiuyan Guo
- Department of Gynecology, the First Affiliated Hospital of Harbin Medical University, 23 Youzheng Road, Harbin 150081, China
| | - Qian Liu
- College of Bioinformatics Science and Technology, Harbin Medical University, 157 Baojian Road, Harbin 150081, China
| | - Danni He
- College of Bioinformatics Science and Technology, Harbin Medical University, 157 Baojian Road, Harbin 150081, China
| | - Mengyu Xin
- College of Bioinformatics Science and Technology, Harbin Medical University, 157 Baojian Road, Harbin 150081, China
| | - Yifan Dai
- College of Bioinformatics Science and Technology, Harbin Medical University, 157 Baojian Road, Harbin 150081, China
| | - Rui Sun
- College of Bioinformatics Science and Technology, Harbin Medical University, 157 Baojian Road, Harbin 150081, China
| | - Houxing Li
- College of Bioinformatics Science and Technology, Harbin Medical University, 157 Baojian Road, Harbin 150081, China
| | - Yujie Zhang
- College of Bioinformatics Science and Technology, Harbin Medical University, 157 Baojian Road, Harbin 150081, China
| | - Jiatong Li
- College of Bioinformatics Science and Technology, Harbin Medical University, 157 Baojian Road, Harbin 150081, China
| | - Congcong Kong
- Department of Gynecology, the First Affiliated Hospital of Harbin Medical University, 23 Youzheng Road, Harbin 150081, China
| | - Yue Gao
- College of Bioinformatics Science and Technology, Harbin Medical University, 157 Baojian Road, Harbin 150081, China
| | - Hui Zhi
- College of Bioinformatics Science and Technology, Harbin Medical University, 157 Baojian Road, Harbin 150081, China
| | - Feng Li
- College of Bioinformatics Science and Technology, Harbin Medical University, 157 Baojian Road, Harbin 150081, China
| | - Shangwei Ning
- College of Bioinformatics Science and Technology, Harbin Medical University, 157 Baojian Road, Harbin 150081, China
| | - Peng Wang
- College of Bioinformatics Science and Technology, Harbin Medical University, 157 Baojian Road, Harbin 150081, China
| |
Collapse
|
32
|
Sun F, Li H, Sun D, Fu S, Gu L, Shao X, Wang Q, Dong X, Duan B, Xing F, Wu J, Xiao M, Zhao F, Han JDJ, Liu Q, Fan X, Li C, Wang C, Shi T. Single-cell omics: experimental workflow, data analyses and applications. SCIENCE CHINA. LIFE SCIENCES 2025; 68:5-102. [PMID: 39060615 DOI: 10.1007/s11427-023-2561-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Accepted: 04/18/2024] [Indexed: 07/28/2024]
Abstract
Cells are the fundamental units of biological systems and exhibit unique development trajectories and molecular features. Our exploration of how the genomes orchestrate the formation and maintenance of each cell, and control the cellular phenotypes of various organismsis, is both captivating and intricate. Since the inception of the first single-cell RNA technology, technologies related to single-cell sequencing have experienced rapid advancements in recent years. These technologies have expanded horizontally to include single-cell genome, epigenome, proteome, and metabolome, while vertically, they have progressed to integrate multiple omics data and incorporate additional information such as spatial scRNA-seq and CRISPR screening. Single-cell omics represent a groundbreaking advancement in the biomedical field, offering profound insights into the understanding of complex diseases, including cancers. Here, we comprehensively summarize recent advances in single-cell omics technologies, with a specific focus on the methodology section. This overview aims to guide researchers in selecting appropriate methods for single-cell sequencing and related data analysis.
Collapse
Affiliation(s)
- Fengying Sun
- Department of Clinical Laboratory, the Affiliated Wuhu Hospital of East China Normal University (The Second People's Hospital of Wuhu City), Wuhu, 241000, China
| | - Haoyan Li
- Pharmaceutical Informatics Institute, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Dongqing Sun
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration (Tongji University), Ministry of Education, Orthopaedic Department, Tongji Hospital, Bioinformatics Department, School of Life Sciences and Technology, Tongji University, Shanghai, 200082, China
- Frontier Science Center for Stem Cells, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Shaliu Fu
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration (Tongji University), Ministry of Education, Orthopaedic Department, Tongji Hospital, Bioinformatics Department, School of Life Sciences and Technology, Tongji University, Shanghai, 200082, China
- Translational Medical Center for Stem Cell Therapy and Institute for Regenerative Medicine, Shanghai East Hospital, Bioinformatics Department, School of Life Sciences and Technology, Tongji University, Shanghai, 200082, China
- Research Institute of Intelligent Computing, Zhejiang Lab, Hangzhou, 311121, China
- Shanghai Research Institute for Intelligent Autonomous Systems, Shanghai, 201210, China
| | - Lei Gu
- Center for Single-cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Xin Shao
- Pharmaceutical Informatics Institute, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
- National Key Laboratory of Chinese Medicine Modernization, Innovation Center of Yangtze River Delta, Zhejiang University, Jiaxing, 314103, China
| | - Qinqin Wang
- Center for Single-cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Xin Dong
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration (Tongji University), Ministry of Education, Orthopaedic Department, Tongji Hospital, Bioinformatics Department, School of Life Sciences and Technology, Tongji University, Shanghai, 200082, China
- Frontier Science Center for Stem Cells, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Bin Duan
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration (Tongji University), Ministry of Education, Orthopaedic Department, Tongji Hospital, Bioinformatics Department, School of Life Sciences and Technology, Tongji University, Shanghai, 200082, China
- Translational Medical Center for Stem Cell Therapy and Institute for Regenerative Medicine, Shanghai East Hospital, Bioinformatics Department, School of Life Sciences and Technology, Tongji University, Shanghai, 200082, China
- Research Institute of Intelligent Computing, Zhejiang Lab, Hangzhou, 311121, China
- Shanghai Research Institute for Intelligent Autonomous Systems, Shanghai, 201210, China
| | - Feiyang Xing
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration (Tongji University), Ministry of Education, Orthopaedic Department, Tongji Hospital, Bioinformatics Department, School of Life Sciences and Technology, Tongji University, Shanghai, 200082, China
- Frontier Science Center for Stem Cells, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Jun Wu
- Center for Bioinformatics and Computational Biology, Shanghai Key Laboratory of Regulatory Biology, the Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Minmin Xiao
- Department of Clinical Laboratory, the Affiliated Wuhu Hospital of East China Normal University (The Second People's Hospital of Wuhu City), Wuhu, 241000, China.
| | - Fangqing Zhao
- Beijing Institutes of Life Science, Chinese Academy of Sciences, Beijing, 100101, China.
| | - Jing-Dong J Han
- Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Center for Quantitative Biology (CQB), Peking University, Beijing, 100871, China.
| | - Qi Liu
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration (Tongji University), Ministry of Education, Orthopaedic Department, Tongji Hospital, Bioinformatics Department, School of Life Sciences and Technology, Tongji University, Shanghai, 200082, China.
- Translational Medical Center for Stem Cell Therapy and Institute for Regenerative Medicine, Shanghai East Hospital, Bioinformatics Department, School of Life Sciences and Technology, Tongji University, Shanghai, 200082, China.
- Research Institute of Intelligent Computing, Zhejiang Lab, Hangzhou, 311121, China.
- Shanghai Research Institute for Intelligent Autonomous Systems, Shanghai, 201210, China.
| | - Xiaohui Fan
- Pharmaceutical Informatics Institute, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China.
- National Key Laboratory of Chinese Medicine Modernization, Innovation Center of Yangtze River Delta, Zhejiang University, Jiaxing, 314103, China.
- Zhejiang Key Laboratory of Precision Diagnosis and Therapy for Major Gynecological Diseases, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, China.
| | - Chen Li
- Center for Single-cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| | - Chenfei Wang
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration (Tongji University), Ministry of Education, Orthopaedic Department, Tongji Hospital, Bioinformatics Department, School of Life Sciences and Technology, Tongji University, Shanghai, 200082, China.
- Frontier Science Center for Stem Cells, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China.
| | - Tieliu Shi
- Department of Clinical Laboratory, the Affiliated Wuhu Hospital of East China Normal University (The Second People's Hospital of Wuhu City), Wuhu, 241000, China.
- Center for Bioinformatics and Computational Biology, Shanghai Key Laboratory of Regulatory Biology, the Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, 200241, China.
- Key Laboratory of Advanced Theory and Application in Statistics and Data Science-MOE, School of Statistics, East China Normal University, Shanghai, 200062, China.
| |
Collapse
|
33
|
He KJ, Gong G. Prognostic prediction and immune infiltration analysis based on lysosome and senescence state identifies MMP12 as a novel therapy target in gastric cancer. Int Immunopharmacol 2024; 143:113344. [PMID: 39401475 DOI: 10.1016/j.intimp.2024.113344] [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: 06/21/2024] [Revised: 09/21/2024] [Accepted: 10/04/2024] [Indexed: 10/30/2024]
Abstract
BACKGROUND AND AIMS As humans undergo the aging process, they become more vulnerable to various types of cancers, including gastric cancer (GC), which is frequently associated with aging. The senescent phenotype is closely linked to lysosomes, but research on the combined impact of senescence and lysosomes on GC prognosis is scarce. METHODS To construct and validate a prognostic model for gastric cancer (GC), we obtained gene expression and clinical data of GC patients from Cancer Genome Atlas (TCGA) databases. We employed Least Absolute Shrinkage and Selection Operator (LASSO) Cox regression for model construction and ConsensusClusterPlus R package for generating cluster heatmaps. The model's predictive ability was evaluated through Kaplan-Meier survival analysis and ROC curve analysis. Our analysis included an assessment of the senescence and lysosome state using expression profiles and immune infiltration analysis through CIBERSORT methods. Finally, we validated potential gene targets through cellular experiments. RESULTS "In this research, we discovered two subtypes of gastric cancer (GC), Cluster 1 and Cluster 2. These subtypes are characterized by the presence of lysosomes and senescence, and we have identified distinct molecular features unique to each subtype. We observed that Cluster 2 had a lower survival prognosis compared to Cluster 1. Additionally, we have developed a risk prediction model that takes into consideration the presence of lysosomes and senescence. Patients in the high-risk group, as predicted by our model, experienced shorter survival times. Further analysis included immune infiltration, immune checkpoint, and chemotherapy evaluation of GC patients. We have displayed the frequency of mutations and copy number variations (CNVs) in visual formats. Our cellular experiments demonstrated that the MMP12 gene serves as a protective factor in GC cells." CONCLUSIONS In conclusion, we have clarified the extensive relationship between lysosomes and senescence in GC and developed a risk signature to forecast the prognosis of GC patients. MMP12 could be a promising protective factor for GC patients and might present a novel concept for anticipating the efficacy of targeted therapies and immunotherapies in GC patients.
Collapse
Affiliation(s)
- Ke-Jie He
- The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Quzhou City, Zhejiang Province, China.
| | - Guoyu Gong
- School of Medicine, Xiamen University, Xiamen, China
| |
Collapse
|
34
|
Wang J, Ye F, Chai H, Jiang Y, Wang T, Ran X, Xia Q, Xu Z, Fu Y, Zhang G, Wu H, Guo G, Guo H, Ruan Y, Wang Y, Xing D, Xu X, Zhang Z. Advances and applications in single-cell and spatial genomics. SCIENCE CHINA. LIFE SCIENCES 2024:10.1007/s11427-024-2770-x. [PMID: 39792333 DOI: 10.1007/s11427-024-2770-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Accepted: 10/10/2024] [Indexed: 01/12/2025]
Abstract
The applications of single-cell and spatial technologies in recent times have revolutionized the present understanding of cellular states and the cellular heterogeneity inherent in complex biological systems. These advancements offer unprecedented resolution in the examination of the functional genomics of individual cells and their spatial context within tissues. In this review, we have comprehensively discussed the historical development and recent progress in the field of single-cell and spatial genomics. We have reviewed the breakthroughs in single-cell multi-omics technologies, spatial genomics methods, and the computational strategies employed toward the analyses of single-cell atlas data. Furthermore, we have highlighted the advances made in constructing cellular atlases and their clinical applications, particularly in the context of disease. Finally, we have discussed the emerging trends, challenges, and opportunities in this rapidly evolving field.
Collapse
Affiliation(s)
- Jingjing Wang
- Bone Marrow Transplantation Center of the First Affiliated Hospital & Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Fang Ye
- Bone Marrow Transplantation Center of the First Affiliated Hospital & Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Haoxi Chai
- Life Sciences Institute and The Second Affiliated Hospital, Zhejiang University, Hangzhou, 310058, China
| | - Yujia Jiang
- BGI Research, Shenzhen, 518083, China
- BGI Research, Hangzhou, 310030, China
| | - Teng Wang
- Biomedical Pioneering Innovation Center (BIOPIC) and School of Life Sciences, Peking University, Beijing, 100871, China
- Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, 100871, China
| | - Xia Ran
- Bone Marrow Transplantation Center of the First Affiliated Hospital & Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, 310058, China
- Institute of Hematology, Zhejiang University, Hangzhou, 310000, China
| | - Qimin Xia
- Biomedical Pioneering Innovation Center (BIOPIC) and School of Life Sciences, Peking University, Beijing, 100871, China
| | - Ziye Xu
- Department of Laboratory Medicine of The First Affiliated Hospital & Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Yuting Fu
- Bone Marrow Transplantation Center of the First Affiliated Hospital & Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, 310058, China
- Center for Stem Cell and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Guodong Zhang
- Bone Marrow Transplantation Center of the First Affiliated Hospital & Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, 310058, China
- Center for Stem Cell and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Hanyu Wu
- Bone Marrow Transplantation Center of the First Affiliated Hospital & Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, 310058, China
- Center for Stem Cell and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Guoji Guo
- Bone Marrow Transplantation Center of the First Affiliated Hospital & Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, 310058, China.
- Center for Stem Cell and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, 310058, China.
- Zhejiang Provincial Key Lab for Tissue Engineering and Regenerative Medicine, Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Regenerative Medicine, Hangzhou, 310058, China.
- Institute of Hematology, Zhejiang University, Hangzhou, 310000, China.
| | - Hongshan Guo
- Bone Marrow Transplantation Center of the First Affiliated Hospital & Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, 310058, China.
- Institute of Hematology, Zhejiang University, Hangzhou, 310000, China.
| | - Yijun Ruan
- Life Sciences Institute and The Second Affiliated Hospital, Zhejiang University, Hangzhou, 310058, China.
| | - Yongcheng Wang
- Department of Laboratory Medicine of The First Affiliated Hospital & Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, 310058, China.
| | - Dong Xing
- Biomedical Pioneering Innovation Center (BIOPIC) and School of Life Sciences, Peking University, Beijing, 100871, China.
- Beijing Advanced Innovation Center for Genomics (ICG), Peking University, Beijing, 100871, China.
| | - Xun Xu
- BGI Research, Shenzhen, 518083, China.
- BGI Research, Hangzhou, 310030, China.
- Guangdong Provincial Key Laboratory of Genome Read and Write, BGI Research, Shenzhen, 518083, China.
| | - Zemin Zhang
- Biomedical Pioneering Innovation Center (BIOPIC) and School of Life Sciences, Peking University, Beijing, 100871, China.
| |
Collapse
|
35
|
Otoničar J, Lazareva O, Mallm JP, Simovic-Lorenz M, Philippos G, Sant P, Parekh U, Hammann L, Li A, Yildiz U, Marttinen M, Zaugg J, Noh KM, Stegle O, Ernst A. HIPSD&R-seq enables scalable genomic copy number and transcriptome profiling. Genome Biol 2024; 25:316. [PMID: 39696535 DOI: 10.1186/s13059-024-03450-0] [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: 09/05/2023] [Accepted: 11/29/2024] [Indexed: 12/20/2024] Open
Abstract
Single-cell DNA sequencing (scDNA-seq) enables decoding somatic cancer variation. Existing methods are hampered by low throughput or cannot be combined with transcriptome sequencing in the same cell. We propose HIPSD&R-seq (HIgh-throughPut Single-cell Dna and Rna-seq), a scalable yet simple and accessible assay to profile low-coverage DNA and RNA in thousands of cells in parallel. Our approach builds on a modification of the 10X Genomics platform for scATAC and multiome profiling. In applications to human cell models and primary tissue, we demonstrate the feasibility to detect rare clones and we combine the assay with combinatorial indexing to profile over 17,000 cells.
Collapse
Affiliation(s)
- Jan Otoničar
- Group Genome Instability in Tumors, German Cancer Research Center (DKFZ), Heidelberg, Germany
- German Cancer Consortium (DKTK), DKFZ, Core Center, Heidelberg, Germany
- Faculty of Biosciences, Heidelberg University, Heidelberg, Germany
| | - Olga Lazareva
- Division of Computational Genomics and Systems Genetics, German Cancer Research Center, Heidelberg, Germany
- European Molecular Biology Laboratory, Genome Biology Unit, Heidelberg, Germany
- Junior Clinical Cooperation Unit, Multiparametric Methods for Early Detection of Prostate Cancer, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Jan-Philipp Mallm
- Single Cell Open Lab, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Center for Quantitative Analysis of Molecular and Cellular Biosystems (BioQuant), Heidelberg University, Heidelberg, Germany
| | - Milena Simovic-Lorenz
- Group Genome Instability in Tumors, German Cancer Research Center (DKFZ), Heidelberg, Germany
- German Cancer Consortium (DKTK), DKFZ, Core Center, Heidelberg, Germany
| | - George Philippos
- Group Genome Instability in Tumors, German Cancer Research Center (DKFZ), Heidelberg, Germany
- German Cancer Consortium (DKTK), DKFZ, Core Center, Heidelberg, Germany
- Faculty of Biosciences, Heidelberg University, Heidelberg, Germany
| | - Pooja Sant
- Single Cell Open Lab, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Urja Parekh
- Group Genome Instability in Tumors, German Cancer Research Center (DKFZ), Heidelberg, Germany
- German Cancer Consortium (DKTK), DKFZ, Core Center, Heidelberg, Germany
- Faculty of Biosciences, Heidelberg University, Heidelberg, Germany
| | - Linda Hammann
- Group Genome Instability in Tumors, German Cancer Research Center (DKFZ), Heidelberg, Germany
- German Cancer Consortium (DKTK), DKFZ, Core Center, Heidelberg, Germany
| | - Albert Li
- Group Genome Instability in Tumors, German Cancer Research Center (DKFZ), Heidelberg, Germany
- German Cancer Consortium (DKTK), DKFZ, Core Center, Heidelberg, Germany
| | - Umut Yildiz
- European Molecular Biology Laboratory, Genome Biology Unit, Heidelberg, Germany
| | - Mikael Marttinen
- European Molecular Biology Laboratory, Structural and Computational Biology Unit, Heidelberg, Germany
| | - Judith Zaugg
- European Molecular Biology Laboratory, Structural and Computational Biology Unit, Heidelberg, Germany
- Molecular Medicine Partnership Unit, University of Heidelberg, Heidelberg, Germany
| | - Kyung Min Noh
- European Molecular Biology Laboratory, Genome Biology Unit, Heidelberg, Germany
| | - Oliver Stegle
- Division of Computational Genomics and Systems Genetics, German Cancer Research Center, Heidelberg, Germany.
- European Molecular Biology Laboratory, Genome Biology Unit, Heidelberg, Germany.
- Wellcome Sanger Institute, Wellcome Genome Campus, Cambridge, UK.
| | - Aurélie Ernst
- Group Genome Instability in Tumors, German Cancer Research Center (DKFZ), Heidelberg, Germany.
- German Cancer Consortium (DKTK), DKFZ, Core Center, Heidelberg, Germany.
| |
Collapse
|
36
|
Liu T, Li K, Wang Y, Li H, Zhao H. Evaluating the Utilities of Foundation Models in Single-cell Data Analysis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.09.08.555192. [PMID: 38464157 PMCID: PMC10925156 DOI: 10.1101/2023.09.08.555192] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/12/2024]
Abstract
Foundation Models (FMs) have made significant strides in both industrial and scientific domains. In this paper, we evaluate the performance of FMs for single-cell sequencing data analysis through comprehensive experiments across eight downstream tasks pertinent to single-cell data. Overall, the top FMs include scGPT, Geneformer, and CellPLM by considering model performances and user accessibility among ten single-cell FMs. However, by comparing these FMs with task-specific methods, we found that single-cell FMs may not consistently excel than task-specific methods in all tasks, which challenges the necessity of developing foundation models for single-cell analysis. In addition, we evaluated the effects of hyper-parameters, initial settings, and stability for training single-cell FMs based on a proposed scEval framework, and provide guidelines for pre-training and fine-tuning, to enhance the performances of single-cell FMs. Our work summarizes the current state of single-cell FMs, points to their constraints and avenues for future development, and offers a freely available evaluation pipeline to benchmark new models and improve method development.
Collapse
|
37
|
Wu A, Li H, Gao M, Liang J, Huang J, Farrés J, Cao D, Li G. The pan-cancer landscape of aldo-keto reductase1B10 reveals that its expression is diminished in gastric cancer. Front Immunol 2024; 15:1488042. [PMID: 39712017 PMCID: PMC11659136 DOI: 10.3389/fimmu.2024.1488042] [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: 08/29/2024] [Accepted: 11/18/2024] [Indexed: 12/24/2024] Open
Abstract
Introduction Aldo-keto reductase 1B10 (AKR1B10) is a multifunctional enzyme, which is important in cancer development and progression, but the landscape of AKR1B10 in pan-cancers and in tumor microenvironment is unclear. Method This study integrated the sequencing data of 33 cancer types, including gastric cancer, from TCGA project to explored the expression pattern and genetic and epigenetic alterations of AKR1B10. The association of AKR1B10 expression with clinical progression of cancers was evaluated by Kaplan-Meier analysis; the potential role of AKR1B10 in tumor microenvironment (TME) and immune-related gene expression were analyzed by PURITY, ESTIMATE, TIMER and CIBERSORT algorithms. The expression of AKR1B10 and immune cell markers in gastric cancer were evaluated with multiplex immunofluorescence staining. Result Results indicated that AKR1B10 was highly expressed in the gastrointestinal tract in health donors, but the expression of AKR1B10 was significantly changed in most of cancer types, which may be ascribed to DNA methylation in its promoter. The AKR1B10 expression in cancers and its value in disease progression was bidirectional and functionally enriched in metabolism in pan-cancers. In tumor microenvironment, AKR1B10 was significantly correlated with immune cell infiltrations and immune gene expression. In the stomach, along with the diminishing of AKR1B10 expression, CD68+ macrophage increased and CD19+ B cell decreased in gastric cancer. Discussion These data indicates that AKR1B10 may be an important factor in the development and progression and a potential therapeutic target for multiple cancers, but plays as a protector in the gastric tissues.
Collapse
Affiliation(s)
- Anqi Wu
- Department of Clinical Research Center, The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, China
- Hunan Province Key Laboratory of Basic and Clinical Pharmacological Research on Gastrointestinal Tumors, The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, China
| | - Hao Li
- Department of Pathology, The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, China
| | - Mengnan Gao
- Department of Gastroenterology, The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, China
| | - Juan Liang
- Department of Gastroenterology, The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, China
| | - Jiaqi Huang
- Department of Gastroenterology, The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, China
| | - Jaume Farrés
- Department of Biochemistry and Molecular Biology, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Deliang Cao
- Hunan Province Key Laboratory of Cancer Cellular and Molecular Pathology, Hengyang Medical School, Cancer Research Institute, University of South China, Hengyang, China
| | - Guoqing Li
- Hunan Province Key Laboratory of Basic and Clinical Pharmacological Research on Gastrointestinal Tumors, The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, China
- Department of Gastroenterology, The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, China
| |
Collapse
|
38
|
Luo CH, Hu LH, Liu JY, Xia L, Zhou L, Sun RH, Lin CC, Qiu X, Jiang B, Yang MY, Zhang XH, Yang XB, Chen GQ, Lu Y. CDK9 recruits HUWE1 to degrade RARα and offers therapeutic opportunities for cutaneous T-cell lymphoma. Nat Commun 2024; 15:10594. [PMID: 39632829 PMCID: PMC11618697 DOI: 10.1038/s41467-024-54354-3] [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: 04/23/2024] [Accepted: 11/05/2024] [Indexed: 12/07/2024] Open
Abstract
Cutaneous T-cell lymphoma (CTCL) is a heterogeneous non-Hodgkin lymphoma originating in the skin and invading the systemic hematopoietic system. Current treatments, including chemotherapy and monoclonal antibodies yielded limited responses with high incidence of side effects, highlighting the need for targeted therapy. Screening with small inhibitors library, herein we identify cyclin dependent kinase 9 (CDK9) as a driver of CTCL growth. Single-cell RNA-seq analysis reveals a CDK9high malignant T cell cluster with a unique actively proliferating feature. Inhibition, depletion or proteolysis targeting chimera (PROTAC)-mediated degradation of CDK9 significantly reduces CTCL cell growth in vitro and in murine models. CDK9 also promotes degradation of retinoic acid receptor α (RARα) via recruiting the E3 ligase HUWE1. Co-administration of CDK9-PROTAC (GT-02897) with all-trans retinoic acid (ATRA) leads to synergistic attenuation of tumor growth in vitro and in xenograft models, providing a potential translational treatment for complete eradication of CTCL.
Collapse
MESH Headings
- Humans
- Animals
- Cyclin-Dependent Kinase 9/metabolism
- Cyclin-Dependent Kinase 9/antagonists & inhibitors
- Lymphoma, T-Cell, Cutaneous/metabolism
- Lymphoma, T-Cell, Cutaneous/drug therapy
- Lymphoma, T-Cell, Cutaneous/pathology
- Lymphoma, T-Cell, Cutaneous/genetics
- Mice
- Ubiquitin-Protein Ligases/metabolism
- Ubiquitin-Protein Ligases/genetics
- Cell Line, Tumor
- Tumor Suppressor Proteins/metabolism
- Tumor Suppressor Proteins/genetics
- Retinoic Acid Receptor alpha/metabolism
- Retinoic Acid Receptor alpha/genetics
- Tretinoin/metabolism
- Tretinoin/pharmacology
- Xenograft Model Antitumor Assays
- Cell Proliferation/drug effects
- Skin Neoplasms/drug therapy
- Skin Neoplasms/metabolism
- Skin Neoplasms/pathology
- Skin Neoplasms/genetics
- Proteolysis/drug effects
- Female
- Mice, Inbred NOD
Collapse
Affiliation(s)
- Chen-Hui Luo
- Department of Pathophysiology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Institute of Dermatology, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Li-Hong Hu
- Institute of Dermatology, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Jie-Yang Liu
- Institute of Dermatology, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Li Xia
- Department of Core Facility of Basic Medical Sciences, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Li Zhou
- Department of Core Facility of Basic Medical Sciences, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ren-Hong Sun
- Gluetacs Therapeutics (Shanghai) Co., Ltd., Shanghai, China
| | - Chen-Cen Lin
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, Shanghai, China
| | - Xing Qiu
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, Shanghai, China
| | - Biao Jiang
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, Shanghai, China
| | - Meng-Ying Yang
- Institute of Dermatology, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.
| | - Xue-Hong Zhang
- Center of Genome and Personalized Medicine, Institute of Cancer Stem Cell, Dalian Medical University, Dalian, China.
| | - Xiao-Bao Yang
- Gluetacs Therapeutics (Shanghai) Co., Ltd., Shanghai, China.
| | - Guo-Qiang Chen
- Department of Pathophysiology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
- Institute of Aging & Tissue Regeneration, State Key Laboratory of Systems Medicine for Cancer, Research Units of Stress and Tumor (2019RU043), Chinese Academy of Medical Sciences, Ren-Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
- School of Basic Medicine and Life Science, Hainan Academy of Medical Sciences, Hainan Medical University, Haikou, China.
| | - Ying Lu
- Institute of Dermatology, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.
| |
Collapse
|
39
|
Kim J, Eo EY, Kim B, Lee H, Kim J, Koo BK, Kim HJ, Cho S, Kim J, Cho YJ. Transcriptomic Analysis of Air-Liquid Interface Culture in Human Lung Organoids Reveals Regulators of Epithelial Differentiation. Cells 2024; 13:1991. [PMID: 39682739 DOI: 10.3390/cells13231991] [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: 10/07/2024] [Revised: 11/17/2024] [Accepted: 11/29/2024] [Indexed: 12/18/2024] Open
Abstract
To develop in vitro respiratory models, it is crucial to identify the factors involved in epithelial cell differentiation. In this study, we comprehensively analyzed the effects of air-liquid interface (ALI) culture on epithelial cell differentiation using single-cell RNA sequencing (scRNA-seq). ALI culture induced a pronounced shift in cell composition, marked by a fivefold increase in ciliated cells and a reduction of more than half in basal cells. Transcriptional signatures associated with epithelial cell differentiation, analyzed using iPathwayGuide software, revealed the downregulation of VEGFA and upregulation of CDKN1A as key signals for epithelial differentiation. Our findings highlight the efficacy of the ALI culture for replicating the human lung airway epithelium and provide valuable insights into the crucial factors that influence human ciliated cell differentiation.
Collapse
Affiliation(s)
- Jieun Kim
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Seoul National University College of Medicine, Seoul National University Bundang Hospital, Seongnam 13620, Republic of Korea
- Department of Biomedical Science, CHA University, Seongnam 13488, Republic of Korea
| | - Eun-Young Eo
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Seoul National University College of Medicine, Seoul National University Bundang Hospital, Seongnam 13620, Republic of Korea
| | - Bokyong Kim
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Seoul National University College of Medicine, Seoul National University Bundang Hospital, Seongnam 13620, Republic of Korea
| | - Heetak Lee
- Center for Genome Engineering, Institute for Basic Science, Daejeon 34126, Republic of Korea
| | - Jihoon Kim
- Center for Genome Engineering, Institute for Basic Science, Daejeon 34126, Republic of Korea
- Department of Medical and Biological Sciences, The Catholic University of Korea, Bucheon 14662, Republic of Korea
| | - Bon-Kyoung Koo
- Center for Genome Engineering, Institute for Basic Science, Daejeon 34126, Republic of Korea
| | - Hyung-Jun Kim
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Seoul National University College of Medicine, Seoul National University Bundang Hospital, Seongnam 13620, Republic of Korea
| | - Sukki Cho
- Department of Thoracic and Cardiovascular Surgery, Seoul National University Bundang Hospital, Seongnam 13620, Republic of Korea
| | - Jinho Kim
- Department of Genomic Medicine, Seoul National University Bundang Hospital, Seongnam 13620, Republic of Korea
- Precision Medicine Center, Future Innovation Research Division, Seoul National University Bundang Hospital, Seongnam 13620, Republic of Korea
- Department of Laboratory Medicine, Seoul National University College of Medicine, Seoul National University Bundang Hospital, Seongnam 13620, Republic of Korea
| | - Young-Jae Cho
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Seoul National University College of Medicine, Seoul National University Bundang Hospital, Seongnam 13620, Republic of Korea
| |
Collapse
|
40
|
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
|
41
|
Kuemmerle LB, Luecken MD, Firsova AB, Barros de Andrade E Sousa L, Straßer L, Mekki II, Campi F, Heumos L, Shulman M, Beliaeva V, Hediyeh-Zadeh S, Schaar AC, Mahbubani KT, Sountoulidis A, Balassa T, Kovacs F, Horvath P, Piraud M, Ertürk A, Samakovlis C, Theis FJ. Probe set selection for targeted spatial transcriptomics. Nat Methods 2024; 21:2260-2270. [PMID: 39558096 PMCID: PMC11621025 DOI: 10.1038/s41592-024-02496-z] [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: 07/19/2022] [Accepted: 09/30/2024] [Indexed: 11/20/2024]
Abstract
Targeted spatial transcriptomic methods capture the topology of cell types and states in tissues at single-cell and subcellular resolution by measuring the expression of a predefined set of genes. The selection of an optimal set of probed genes is crucial for capturing the spatial signals present in a tissue. This requires selecting the most informative, yet minimal, set of genes to profile (gene set selection) for which it is possible to build probes (probe design). However, current selections often rely on marker genes, precluding them from detecting continuous spatial signals or new states. We present Spapros, an end-to-end probe set selection pipeline that optimizes both gene set specificity for cell type identification and within-cell type expression variation to resolve spatially distinct populations while considering prior knowledge as well as probe design and expression constraints. We evaluated Spapros and show that it outperforms other selection approaches in both cell type recovery and recovering expression variation beyond cell types. Furthermore, we used Spapros to design a single-cell resolution in situ hybridization on tissues (SCRINSHOT) experiment of adult lung tissue to demonstrate how probes selected with Spapros identify cell types of interest and detect spatial variation even within cell types.
Collapse
Affiliation(s)
- Louis B Kuemmerle
- Institute of Computational Biology, Helmholtz Zentrum München, Neuherberg, Germany
- Institute for Tissue Engineering and Regenerative Medicine, Helmholtz Zentrum München, Neuherberg, Germany
- School of Life Sciences Weihenstephan, Technical University of Munich, Freising, Germany
| | - Malte D Luecken
- Institute of Computational Biology, Helmholtz Zentrum München, Neuherberg, Germany
- Institute of Lung Health & Immunity, Helmholtz Munich, Member of the German Center for Lung Research (DZL), Munich, Germany
- German Center for Lung Research (DZL), Gießen, Germany
| | - Alexandra B Firsova
- SciLifeLab and Department of Molecular Biosciences, Stockholm University, Stockholm, Sweden
| | | | - Lena Straßer
- Institute of Computational Biology, Helmholtz Zentrum München, Neuherberg, Germany
| | | | - Francesco Campi
- Helmholtz AI, Helmholtz Zentrum München, Neuherberg, Germany
| | - Lukas Heumos
- Institute of Computational Biology, Helmholtz Zentrum München, Neuherberg, Germany
- School of Life Sciences Weihenstephan, Technical University of Munich, Freising, Germany
- Institute of Lung Biology and Disease and Comprehensive Pneumology Center, Helmholtz Zentrum München, German Center for Lung Research (DZL), Munich, Germany
| | - Maiia Shulman
- Institute of Computational Biology, Helmholtz Zentrum München, Neuherberg, Germany
| | - Valentina Beliaeva
- Institute of Computational Biology, Helmholtz Zentrum München, Neuherberg, Germany
| | - Soroor Hediyeh-Zadeh
- Institute of Computational Biology, Helmholtz Zentrum München, Neuherberg, Germany
- School of Life Sciences Weihenstephan, Technical University of Munich, Freising, Germany
| | - Anna C Schaar
- Institute of Computational Biology, Helmholtz Zentrum München, Neuherberg, Germany
- TUM School of Computation, Information and Technology, Technical University of Munich, Munich, Germany
- Munich Center for Machine Learning, Technical University of Munich, Munich, Germany
| | - Krishnaa T Mahbubani
- Department of Surgery, University of Cambridge and Cambridge NIHR Biomedical Research Centre, Cambridge, UK
| | | | - Tamás Balassa
- Synthetic and Systems Biology Unit, Biological Research Centre, Eötvös Loránd Research Network, Szeged, Hungary
| | | | - Peter Horvath
- Synthetic and Systems Biology Unit, Biological Research Centre, Eötvös Loránd Research Network, Szeged, Hungary
- Institute of AI for Health, Helmholtz Zentrum München, Neuherberg, Germany
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki, Finland
| | - Marie Piraud
- Helmholtz AI, Helmholtz Zentrum München, Neuherberg, Germany
| | - Ali Ertürk
- Institute for Tissue Engineering and Regenerative Medicine, Helmholtz Zentrum München, Neuherberg, Germany
- Institute for Stroke and Dementia Research, Klinikum der Universität München, Ludwig-Maximilians University Munich, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
- School of Medicine, Koç University, İstanbul, Turkey
| | - Christos Samakovlis
- SciLifeLab and Department of Molecular Biosciences, Stockholm University, Stockholm, Sweden
- Cardiopulmonary Institute, Justus Liebig University, Giessen, Germany
| | - Fabian J Theis
- Institute of Computational Biology, Helmholtz Zentrum München, Neuherberg, Germany.
- School of Life Sciences Weihenstephan, Technical University of Munich, Freising, Germany.
- School of Computation, Information and Technology, Technical University of Munich, Munich, Germany.
| |
Collapse
|
42
|
Liu T, Long W, Cao Z, Wang Y, He CH, Zhang L, Strittmatter SM, Zhao H. CosGeneGate selects multi-functional and credible biomarkers for single-cell analysis. Brief Bioinform 2024; 26:bbae626. [PMID: 39592241 PMCID: PMC11596696 DOI: 10.1093/bib/bbae626] [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: 07/12/2024] [Revised: 10/07/2024] [Accepted: 11/14/2024] [Indexed: 11/28/2024] Open
Abstract
MOTIVATION Selecting representative genes or marker genes to distinguish cell types is an important task in single-cell sequencing analysis. Although many methods have been proposed to select marker genes, the genes selected may have redundancy and/or do not show cell-type-specific expression patterns to distinguish cell types. RESULTS Here, we present a novel model, named CosGeneGate, to select marker genes for more effective marker selections. CosGeneGate is inspired by combining the advantages of selecting marker genes based on both cell-type classification accuracy and marker gene specific expression patterns. We demonstrate the better performance of the marker genes selected by CosGeneGate for various downstream analyses than the existing methods with both public datasets and newly sequenced datasets. The non-redundant marker genes identified by CosGeneGate for major cell types and tissues in human can be found at the website as follows: https://github.com/VivLon/CosGeneGate/blob/main/marker gene list.xlsx.
Collapse
Affiliation(s)
- Tianyu Liu
- Department of Biostatistics, Yale University, New Haven, CT, 06520, United States
- Interdepartmental Program in Computational Biology & Bioinformatics, Yale University, New Haven, CT, 06520, United States
| | - Wenxin Long
- Department of Biostatistics, Yale University, New Haven, CT, 06520, United States
- Department of Statistics, The Pennsylvania State University, University Park, PA, 16820, United States
| | - Zhiyuan Cao
- Department of Biostatistics, Yale University, New Haven, CT, 06520, United States
- Interdepartmental Program in Computational Biology & Bioinformatics, Yale University, New Haven, CT, 06520, United States
- Program of Health Informatics, Yale University, New Haven, CT, 06520, United States
| | - Yuge Wang
- Department of Biostatistics, Yale University, New Haven, CT, 06520, United States
| | - Chuan Hua He
- Department of Neurology, Yale University School of Medicine, New Haven, CT, 06520, United States
| | - Le Zhang
- Department of Neurology, Yale University School of Medicine, New Haven, CT, 06520, United States
- Department of Neuroscience, Yale University School of Medicine, New Haven, CT, 06520, United States
| | - Stephen M Strittmatter
- Department of Neurology, Yale University School of Medicine, New Haven, CT, 06520, United States
- Department of Neuroscience, Yale University School of Medicine, New Haven, CT, 06520, United States
- Cellular Neuroscience, Neurodegeneration and Repair Program, Yale University School of Medicine, New Haven, CT, 06520, United States
| | - Hongyu Zhao
- Department of Biostatistics, Yale University, New Haven, CT, 06520, United States
- Interdepartmental Program in Computational Biology & Bioinformatics, Yale University, New Haven, CT, 06520, United States
| |
Collapse
|
43
|
Yuan L, Sun S, Jiang Y, Zhang Q, Ye L, Zheng CH, Huang DS. scRGCL: a cell type annotation method for single-cell RNA-seq data using residual graph convolutional neural network with contrastive learning. Brief Bioinform 2024; 26:bbae662. [PMID: 39708840 DOI: 10.1093/bib/bbae662] [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: 09/26/2024] [Revised: 11/13/2024] [Accepted: 12/04/2024] [Indexed: 12/23/2024] Open
Abstract
Cell type annotation is a critical step in analyzing single-cell RNA sequencing (scRNA-seq) data. A large number of deep learning (DL)-based methods have been proposed to annotate cell types of scRNA-seq data and have achieved impressive results. However, there are several limitations to these methods. First, they do not fully exploit cell-to-cell differential features. Second, they are developed based on shallow features and lack of flexibility in integrating high-order features in the data. Finally, the low-dimensional gene features may lead to overfitting in neural networks. To overcome those limitations, we propose a novel DL-based model, cell type annotation of single-cell RNA-seq data using residual graph convolutional neural network with contrastive learning (scRGCL), based on residual graph convolutional neural network and contrastive learning for cell type annotation of single-cell RNA-seq data. scRGCL mainly consists of a residual graph convolutional neural network, contrastive learning, and weight freezing. A residual graph convolutional neural network is utilized to extract complex high-order features from data. Contrastive learning can help the model learn meaningful cell-to-cell differential features. Weight freezing can avoid overfitting and help the model discover the impact of specific gene expression on cell type annotation. To verify the effectiveness of scRGCL, we compared its performance with six methods (three shallow learning algorithms and three state-of-the-art DL-based methods) on eight single-cell benchmark datasets from two species (seven in human and one in mouse). Experimental results not only show that scRGCL outperforms competing methods but also demonstrate the generalizability of scRGCL for cell type annotation. scRGCL is available at https://github.com/nathanyl/scRGCL.
Collapse
Affiliation(s)
- Lin Yuan
- Key Laboratory of Computing Power Network and Information Security, Ministry of Education, Shandong Computer Science Center, Qilu University of Technology (Shandong Academy of Sciences), 3501 Daxue Road, 250353, Shandong, China
- Shandong Engineering Research Center of Big Data Applied Technology, Faculty of Computer Science and Technology, Qilu University of Technology (Shandong Academy of Sciences), 3501 Daxue Road, 250353, Shandong, China
- Shandong Provincial Key Laboratory of Industrial Network and Information System Security, Shandong Fundamental Research Center for Computer Science, 3501 Daxue Road, 250353, Shandong, China
| | - Shengguo Sun
- Key Laboratory of Computing Power Network and Information Security, Ministry of Education, Shandong Computer Science Center, Qilu University of Technology (Shandong Academy of Sciences), 3501 Daxue Road, 250353, Shandong, China
- Shandong Engineering Research Center of Big Data Applied Technology, Faculty of Computer Science and Technology, Qilu University of Technology (Shandong Academy of Sciences), 3501 Daxue Road, 250353, Shandong, China
- Shandong Provincial Key Laboratory of Industrial Network and Information System Security, Shandong Fundamental Research Center for Computer Science, 3501 Daxue Road, 250353, Shandong, China
| | - Yufeng Jiang
- Key Laboratory of Computing Power Network and Information Security, Ministry of Education, Shandong Computer Science Center, Qilu University of Technology (Shandong Academy of Sciences), 3501 Daxue Road, 250353, Shandong, China
- Shandong Engineering Research Center of Big Data Applied Technology, Faculty of Computer Science and Technology, Qilu University of Technology (Shandong Academy of Sciences), 3501 Daxue Road, 250353, Shandong, China
- Shandong Provincial Key Laboratory of Industrial Network and Information System Security, Shandong Fundamental Research Center for Computer Science, 3501 Daxue Road, 250353, Shandong, China
| | - Qinhu Zhang
- Ningbo Institute of Digital Twin, Eastern Institute of Technology, 568 Tongxin Road, 315201, Zhejiang, China
| | - Lan Ye
- Cancer Center, The Second Hospital of Shandong University, 247 Beiyuan Street, 250033, Shandong, China
| | - Chun-Hou Zheng
- Key Lab of Intelligent Computing and Signal Processing of Ministry of Education, School of Artificial Intelligence, Anhui University, 111 Jiulong Road, 230601, Anhui, China
| | - De-Shuang Huang
- Ningbo Institute of Digital Twin, Eastern Institute of Technology, 568 Tongxin Road, 315201, Zhejiang, China
| |
Collapse
|
44
|
Zhang X, Liu L. Senescent T Cells: The Silent Culprit in Acute Myeloid Leukemia Progression? Int J Mol Sci 2024; 25:12550. [PMID: 39684260 DOI: 10.3390/ijms252312550] [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: 10/31/2024] [Revised: 11/17/2024] [Accepted: 11/19/2024] [Indexed: 12/18/2024] Open
Abstract
Malignant tumors can evade immune surveillance and elimination through multiple mechanisms, with the induction of immune cell dysfunction serving as a crucial strategy. Mounting evidence indicates that T cell senescence constitutes the primary mechanism underlying T cell dysfunction in acute myeloid leukemia (AML) and represents one of the potential causes of immunotherapy failure. AML usually progresses rapidly and is highly susceptible to drug resistance, thereby resulting in recurrence and patient mortality. Hence, disrupting the immune interface within the bone marrow microenvironment of AML has emerged as a critical objective for synergistically enhancing tumor immunotherapy. In this review, we summarize the general characteristics, distinctive phenotypes, and regulatory signaling networks of senescent T cells and highlight their potential clinical significance in the bone marrow microenvironment of AML. Additionally, we discuss potential therapeutic strategies for alleviating and reversing T cell senescence.
Collapse
Affiliation(s)
- Xiaolan Zhang
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Lingbo Liu
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| |
Collapse
|
45
|
Zhang H, Qian Y, Zhang Y, Zhou X, Shen S, Li J, Sun Z, Wang W. Multi-omics analysis deciphers intercellular communication regulating oxidative stress to promote oral squamous cell carcinoma progression. NPJ Precis Oncol 2024; 8:272. [PMID: 39572698 PMCID: PMC11582705 DOI: 10.1038/s41698-024-00764-x] [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: 06/29/2024] [Accepted: 11/11/2024] [Indexed: 11/24/2024] Open
Abstract
Oral squamous cell carcinoma (OSCC) is a common malignant tumor in the head and neck, associated with high recurrence and poor prognosis. We performed an integrated analysis of single-cell RNA and spatial transcriptomic data from cancerous and normal tissues to create a comprehensive atlas of epithelial cells and cancer-associated fibroblasts (CAFs). Our findings show that AKR1C3+ epithelial cells, located at the tumor's stromal front, exhibit significant copy number variation and poor prognostic indicators, suggesting a role in tumor invasion. We also identified a distinct group of early-stage CAFs (named OSCC_Normal, characterized by ADH1B+, MFAP4+, and PLA2G2A+) that interact with AKR1C3+ cells, where OSCC_Normal may inhibit the FOXO1 redox switch in these epithelial cells via the IGF1/IGF1R pathway, causing oxidative stress overload. Conversely, AKR1C3+ cells use ITGA6/ITGB4 receptor to counteract the effects of OSCC_Normal, promoting cancer invasion. This study unveils complex interactions within the OSCC tumor microenvironment.
Collapse
Affiliation(s)
- Hongrong Zhang
- Department of Oral and Maxillofacial Surgery, Affiliated Stomatology Hospital of Kunming Medical University, Kunming, China
- Yunnan Key Laboratory of Stomatology, Kunming, China
| | - Yemei Qian
- Department of Oral and Maxillofacial Surgery, Affiliated Stomatology Hospital of Kunming Medical University, Kunming, China
- Yunnan Key Laboratory of Stomatology, Kunming, China
| | - Yang Zhang
- Department of Vascular Surgery, Fuwai Yunnan Cardiovascular Hospital, Affiliated Cardiovascular Hospital of Kunming Medical University, Kunming, China
| | - Xue Zhou
- Department of Oral and Maxillofacial Surgery, Affiliated Stomatology Hospital of Kunming Medical University, Kunming, China
| | - Shiying Shen
- Department of Oral and Maxillofacial Surgery, Affiliated Stomatology Hospital of Kunming Medical University, Kunming, China
- Yunnan Key Laboratory of Stomatology, Kunming, China
| | - Jingyi Li
- Department of Oral and Maxillofacial Surgery, Affiliated Stomatology Hospital of Kunming Medical University, Kunming, China
| | - Zheyi Sun
- Yunnan Key Laboratory of Stomatology, Kunming, China.
- Department of Endodontics, Affiliated Stomatology Hospital of Kunming Medical University, Kunming, China.
| | - Weihong Wang
- Department of Oral and Maxillofacial Surgery, Affiliated Stomatology Hospital of Kunming Medical University, Kunming, China.
- Yunnan Key Laboratory of Stomatology, Kunming, China.
| |
Collapse
|
46
|
Yang D, Fang Y, Liu X, Ma J, Xu J, Dong H, Ding H, Wang D, Liu Q, Zhang F. Lensless On-Chip Chemiluminescence Imaging for High-Throughput Single-Cell Heterogeneity Analysis. NANO LETTERS 2024; 24:14875-14883. [PMID: 39512117 DOI: 10.1021/acs.nanolett.4c04487] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2024]
Abstract
High-throughput single-cell heterogeneity imaging and analysis is essential for understanding complex biological systems and for advancing personalized precision disease diagnosis and treatment. Here, we present a miniaturized lensless chemiluminescence chip for high-throughput single-cell functional imaging with subcellular resolution. With the sensitive chemiluminescence sensing and wide field of view of contact lensless imaging, we demonstrated the chemiluminescent imaging of over 1000 single cells, and their membrane glycoprotein and the high-throughput single-cell heterogeneity of membrane protein imaging were examined for precision analysis. Furthermore, the functional adhesion and heterogeneity of single live cells were imaged and explored. This miniaturized lensless on-chip CL-CMOS imaging platform enables high-throughput single-cell imaging and analysis with high sensitivity and subcellular resolution, providing new techniques for the cellular study of biological heterogeneity and has potential application in precision disease diagnosis and treatment at the point-of-care settings.
Collapse
Affiliation(s)
- Dehong Yang
- Biosensor National Special Laboratory, Department of Biomedical Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Ying Fang
- Biosensor National Special Laboratory, Department of Biomedical Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Xiaoyin Liu
- Biosensor National Special Laboratory, Department of Biomedical Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Jinbiao Ma
- Biosensor National Special Laboratory, Department of Biomedical Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Jiahao Xu
- Biosensor National Special Laboratory, Department of Biomedical Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Hao Dong
- Intelligent Perception Research Institute, Zhejiang Lab, Hangzhou, 311100, China
| | - Haiying Ding
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
- Department of Pharmacy, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, 310005, China
| | - Di Wang
- Biosensor National Special Laboratory, Department of Biomedical Engineering, Zhejiang University, Hangzhou, 310027, China
- Intelligent Perception Research Institute, Zhejiang Lab, Hangzhou, 311100, China
| | - Qingjun Liu
- Biosensor National Special Laboratory, Department of Biomedical Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Fenni Zhang
- Biosensor National Special Laboratory, Department of Biomedical Engineering, Zhejiang University, Hangzhou, 310027, China
| |
Collapse
|
47
|
Aubin RG, Montelongo J, Hu R, Gunther E, Nicodemus P, Camara PG. Clustering-independent estimation of cell abundances in bulk tissues using single-cell RNA-seq data. CELL REPORTS METHODS 2024; 4:100905. [PMID: 39561717 PMCID: PMC11705773 DOI: 10.1016/j.crmeth.2024.100905] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Revised: 06/03/2024] [Accepted: 10/22/2024] [Indexed: 11/21/2024]
Abstract
Single-cell RNA sequencing has transformed the study of biological tissues by enabling transcriptomic characterizations of their constituent cell states. Computational methods for gene expression deconvolution use this information to infer the cell composition of related tissues profiled at the bulk level. However, current deconvolution methods are restricted to discrete cell types and have limited power to make inferences about continuous cellular processes such as cell differentiation or immune cell activation. We present ConDecon, a clustering-independent method for inferring the likelihood for each cell in a single-cell dataset to be present in a bulk tissue. ConDecon represents an improvement in phenotypic resolution and functionality with respect to regression-based methods. Using ConDecon, we discover the implication of neurodegenerative microglia inflammatory pathways in the mesenchymal transformation of pediatric ependymoma and characterize their spatial trajectories of activation. The generality of this approach enables the deconvolution of other data modalities, such as bulk ATAC-seq data.
Collapse
Affiliation(s)
- Rachael G Aubin
- Department of Genetics and Institute for Biomedical Informatics, Perelman School of Medicine, University of Pennsylvania, 3700 Hamilton Walk, Philadelphia, PA 19104, USA
| | - Javier Montelongo
- Department of Genetics and Institute for Biomedical Informatics, Perelman School of Medicine, University of Pennsylvania, 3700 Hamilton Walk, Philadelphia, PA 19104, USA
| | - Robert Hu
- Department of Genetics and Institute for Biomedical Informatics, Perelman School of Medicine, University of Pennsylvania, 3700 Hamilton Walk, Philadelphia, PA 19104, USA
| | - Elijah Gunther
- Department of Genetics and Institute for Biomedical Informatics, Perelman School of Medicine, University of Pennsylvania, 3700 Hamilton Walk, Philadelphia, PA 19104, USA
| | - Patrick Nicodemus
- Department of Genetics and Institute for Biomedical Informatics, Perelman School of Medicine, University of Pennsylvania, 3700 Hamilton Walk, Philadelphia, PA 19104, USA
| | - Pablo G Camara
- Department of Genetics and Institute for Biomedical Informatics, Perelman School of Medicine, University of Pennsylvania, 3700 Hamilton Walk, Philadelphia, PA 19104, USA.
| |
Collapse
|
48
|
Zeng Y, Ma Q, Chen J, Kong X, Chen Z, Liu H, Liu L, Qian Y, Wang X, Lu S. Single-cell sequencing: Current applications in various tuberculosis specimen types. Cell Prolif 2024; 57:e13698. [PMID: 38956399 PMCID: PMC11533074 DOI: 10.1111/cpr.13698] [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: 01/24/2024] [Revised: 05/21/2024] [Accepted: 06/07/2024] [Indexed: 07/04/2024] Open
Abstract
Tuberculosis (TB) is a chronic disease caused by Mycobacterium tuberculosis (M.tb) and responsible for millions of deaths worldwide each year. It has a complex pathogenesis that primarily affects the lungs but can also impact systemic organs. In recent years, single-cell sequencing technology has been utilized to characterize the composition and proportion of immune cell subpopulations associated with the pathogenesis of TB disease since it has a high resolution that surpasses conventional techniques. This paper reviews the current use of single-cell sequencing technologies in TB research and their application in analysing specimens from various sources of TB, primarily peripheral blood and lung specimens. The focus is on how these technologies can reveal dynamic changes in immune cell subpopulations, genes and proteins during disease progression after M.tb infection. Based on the current findings, single-cell sequencing has significant potential clinical value in the field of TB research. Next, we will focus on the real-world applications of the potential targets identified through single-cell sequencing for diagnostics, therapeutics and the development of effective vaccines.
Collapse
Affiliation(s)
- Yuqin Zeng
- National Clinical Research Center for Infectious DiseaseShenzhen Third People's HospitalShenzhenGuangdong ProvinceChina
| | - Quan Ma
- National Clinical Research Center for Infectious DiseaseShenzhen Third People's HospitalShenzhenGuangdong ProvinceChina
| | - Jinyun Chen
- National Clinical Research Center for Infectious DiseaseShenzhen Third People's HospitalShenzhenGuangdong ProvinceChina
| | - Xingxing Kong
- National Clinical Research Center for Infectious DiseaseShenzhen Third People's HospitalShenzhenGuangdong ProvinceChina
| | - Zhanpeng Chen
- National Clinical Research Center for Infectious DiseaseShenzhen Third People's HospitalShenzhenGuangdong ProvinceChina
| | - Huazhen Liu
- National Clinical Research Center for Infectious DiseaseShenzhen Third People's HospitalShenzhenGuangdong ProvinceChina
| | - Lanlan Liu
- National Clinical Research Center for Infectious DiseaseShenzhen Third People's HospitalShenzhenGuangdong ProvinceChina
| | - Yan Qian
- National Clinical Research Center for Infectious DiseaseShenzhen Third People's HospitalShenzhenGuangdong ProvinceChina
| | - Xiaomin Wang
- National Clinical Research Center for Infectious DiseaseShenzhen Third People's HospitalShenzhenGuangdong ProvinceChina
| | - Shuihua Lu
- National Clinical Research Center for Infectious DiseaseShenzhen Third People's HospitalShenzhenGuangdong ProvinceChina
| |
Collapse
|
49
|
Pereira Vasconcelos A, Santos E Silva JC, Simizo A, Peña Avila J, Nassar Reich Goldstein G, Prado de Oliveira PH, Mogollón García H, de Carvalho Fraga CA, Nakaya HI. Sex-Based Differences in Thyroid Plasma B Cell Infiltration: Implications for Autoimmune Disease Susceptibility. Endocrinology 2024; 165:bqae148. [PMID: 39487734 DOI: 10.1210/endocr/bqae148] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Revised: 08/15/2024] [Accepted: 10/31/2024] [Indexed: 11/04/2024]
Abstract
Thyroid autoimmune diseases, such as Hashimoto thyroiditis and Graves disease, are significantly more prevalent in women than in men, suggesting underlying biological differences in immune system function and regulation between sexes. Plasma B cells are crucial in autoimmunity due to their role in producing antibodies targeting self-antigens, but their presence in the thyroids of women without clinical autoimmune diseases remains largely unexplored. This study investigates the infiltration of plasma B cells in female thyroids specifically excluding those with any clinical signs of autoimmune diseases. Using bulk RNA-seq analysis, we identified significant sex differences in gene expression profiles, particularly in genes associated with plasma B cells. Single-cell RNA-seq and spatial transcriptomic analyses further revealed that the CXCL13-CXCR5 signaling axis plays a pivotal role in recruiting and organizing plasma B cells within the thyroid tissue. These findings suggest that the inherent presence of plasma B cells in the female thyroid, driven by CXCL13, may contribute to the higher risk of developing autoimmune thyroid diseases in women. Our study provides new insights into the immune landscape of the thyroid and underscores the importance of understanding sex-specific differences in immune cell distribution and function.
Collapse
Affiliation(s)
- Amanda Pereira Vasconcelos
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of São Paulo, São Paulo 05508, Brazil
| | - Juan Carlo Santos E Silva
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of São Paulo, São Paulo 05508, Brazil
| | - Adriana Simizo
- Hospital Israelita Albert Einstein, São Paulo 05620, Brazil
| | - Jonathan Peña Avila
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of São Paulo, São Paulo 05508, Brazil
| | - Gabriel Nassar Reich Goldstein
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of São Paulo, São Paulo 05508, Brazil
| | | | - Henry Mogollón García
- Department of Genetic, Evolution, Microbiology and Immunology, Biology Institute, Campinas State University, Campinas, São Paulo 13083, Brazil
| | | | - Helder I Nakaya
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of São Paulo, São Paulo 05508, Brazil
- Hospital Israelita Albert Einstein, São Paulo 05620, Brazil
| |
Collapse
|
50
|
Luo Y, Liang H. Developmental-status-aware transcriptional decomposition establishes a cell state panorama of human cancers. Genome Med 2024; 16:124. [PMID: 39468667 PMCID: PMC11514945 DOI: 10.1186/s13073-024-01393-6] [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: 05/10/2023] [Accepted: 10/03/2024] [Indexed: 10/30/2024] Open
Abstract
BACKGROUND Cancer cells evolve under unique functional adaptations that unlock transcriptional programs embedded in adult stem and progenitor-like cells for progression, metastasis, and therapeutic resistance. However, it remains challenging to quantify the stemness-aware cell state of a tumor based on its gene expression profile. METHODS We develop a developmental-status-aware transcriptional decomposition strategy using single-cell RNA-sequencing-derived tissue-specific fetal and adult cell signatures as anchors. We apply our method to various biological contexts, including developing human organs, adult human tissues, experimentally induced differentiation cultures, and bulk human tumors, to benchmark its performance and to reveal novel biology of entangled developmental signaling in oncogenic processes. RESULTS Our strategy successfully captures complex dynamics in developmental tissue bulks, reveals remarkable cellular heterogeneity in adult tissues, and resolves the ambiguity of cell identities in in vitro transformations. Applying it to large patient cohorts of bulk RNA-seq, we identify clinically relevant cell-of-origin patterns and observe that decomposed fetal cell signals significantly increase in tumors versus normal tissues and metastases versus primary tumors. Across cancer types, the inferred fetal-state strength outperforms published stemness indices in predicting patient survival and confers substantially improved predictive power for therapeutic responses. CONCLUSIONS Our study not only provides a general approach to quantifying developmental-status-aware cell states of bulk samples but also constructs an information-rich, biologically interpretable, cell-state panorama of human cancers, enabling diverse translational applications.
Collapse
Affiliation(s)
- Yikai Luo
- Graduate Program in Quantitative and Computational Biosciences, Baylor College of Medicine, Houston, TX, 77030, USA
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
- Division of Rheumatology, Department of Medicine, University of California, San Francisco, San Francisco, CA, 94143, USA
| | - Han Liang
- Graduate Program in Quantitative and Computational Biosciences, Baylor College of Medicine, Houston, TX, 77030, USA.
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA.
- Department of Systems Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA.
- Institute for Data Science in Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA.
| |
Collapse
|