1
|
Tang H, Cai Y, Yang M, Tang S, Huang Q, Li H, Liu S, Teng H, Xie T, He M, Liu Y, Liao S. Single-cell and spatial transcriptomics reveals the key role of MCAM + tip-like endothelial cells in osteosarcoma metastasis. NPJ Precis Oncol 2025; 9:104. [PMID: 40221534 PMCID: PMC11993737 DOI: 10.1038/s41698-025-00896-8] [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: 05/07/2024] [Accepted: 03/24/2025] [Indexed: 04/14/2025] Open
Abstract
Osteosarcoma, the most common primary malignant bone tumor in children and adolescents, is highly aggressive and prone to metastasis. Endothelial cells (ECs) are involved in angiogenesis and play a key role in promoting the metastasis of tumor. However, research on tip-like ECs within osteosarcoma was extremely rare. In this study, a single-cell atlas of ECs was constructed using single-cell transcriptomic data. It was found that tip-like ECs were abundant in the primary tumors and metastatic foci. Gene sets score analysis indicated their enrichment in pathways associated with angiogenesis and metastasis. What's more, MCAM was highly expressed in tip-like ECs and was likely to promote the metastasis of osteosarcoma. MCAM was also found to be highly expressed in the ECs of metastatic lymph nodes when compared to normal lymph node samples. Meanwhile, spatial transcriptomics data confirmed the presence of MCAM-positive ECs in metastatic lymph node, closely localized to osteoblasts. In vitro assays, including qRT-PCR, tube formation, and immunofluorescence, validated the role of the MCAM gene in promoting angiogenesis. In conclusion, tip-like ECs may promote tumor metastasis by enhancing angiogenesis. MCAM was a functional gene for tip-like ECs and could serve as a target for the treatment of osteosarcoma.
Collapse
Affiliation(s)
- Haijun Tang
- Department of Spine and Osteopathic Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Yangjie Cai
- Department of Traumatic Orthopedic and Hand Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Mingxiu Yang
- Department of Spine and Osteopathic Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Shengping Tang
- Department of Traumatic Orthopedic and Hand Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Qian Huang
- Department of Traumatic Orthopedic and Hand Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Hening Li
- Department of Oncology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Shangyu Liu
- Department of Spine and Osteopathic Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Hongcai Teng
- Department of Spine and Osteopathic Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Tianyu Xie
- Department of Traumatic Orthopedic and Hand Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Maolin He
- Department of Spine and Osteopathic Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China.
| | - Yun Liu
- Department of Spine and Osteopathic Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China.
| | - Shijie Liao
- Department of Traumatic Orthopedic and Hand Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China.
| |
Collapse
|
2
|
Song H, Xie G, Li Y, Hu X, Yang Z, Zhao Y, Shi Q, Li H, Liu Z, Yin Z, Wang Z, Tong Z, Xu W. A single-cell atlas of bladder cancer unveils dynamic cellular composition and endothelial functional shifts during progression. Discov Oncol 2025; 16:500. [PMID: 40205274 PMCID: PMC11982012 DOI: 10.1007/s12672-025-02297-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2025] [Accepted: 04/02/2025] [Indexed: 04/11/2025] Open
Abstract
PURPOSE Bladder cancer (BC) is characterized by high heterogeneity, with non-muscle-invasive (NMIBC) and muscle-invasive (MIBC) stages differing significantly in clinical behavior and outcomes. The transition from NMIBC to MIBC involves extensive tumor microenvironment (TME) remodeling, particularly in endothelial cells (ECs), which drive angiogenesis and modulate immune and extracellular matrix (ECM) interactions. However, the precise roles of ECs in this progression remain poorly defined. METHODS Public single-cell RNA sequencing (scRNA-seq) datasets from 47 BC patients were analyzed to characterize endothelial cell heterogeneity and functional states across NMIBC and MIBC. Computational tools such as CellChat were applied to reconstruct cell-cell communication networks, focusing on pathways related to angiogenesis, immune crosstalk, and ECM remodeling. RESULTS Twelve major cell types were identified, with endothelial cells exhibiting distinct transcriptional profiles between NMIBC and MIBC. NMIBC-associated ECs promoted adhesion and migration through HMGB1 and CXCL12 signaling. In contrast, MIBC was enriched in an ADAM10+ endothelial subset associated with vascular remodeling and activation of Wnt signaling via CTNNB1. Key ligand-receptor interactions highlighted the dynamic roles of ECs in TME modulation during BC progression. CONCLUSIONS This study reveals stage-specific endothelial cell phenotypes and signaling networks in BC. The identification of an MIBC-specific ADAM10+ endothelial subset underscores its potential role in driving tumor progression and highlights opportunities for stage-adapted vascular-targeted therapies. These findings advance our understanding of BC pathogenesis and provide the foundation for novel therapeutic strategies.
Collapse
Affiliation(s)
- Hongjian Song
- NHC Key Laboratory of Molecular Probe and Targeted Theranostics, The Second Affiliated Hospital of Harbin Medical University, Harbin Medical University Cancer Hospital, Harbin Medical University, Harbin, 150001, China
- Department of Urology, Harbin Medical University Cancer Hospital, Harbin, 150001, China
- Heilongjiang Provincial Key Laboratory of Basic Medical Sciences in Urology Cancer, Harbin Medical University Cancer Hospital, Harbin, 150001, China
| | | | - Yaowei Li
- NHC Key Laboratory of Molecular Probe and Targeted Theranostics, The Second Affiliated Hospital of Harbin Medical University, Harbin Medical University Cancer Hospital, Harbin Medical University, Harbin, 150001, China
- Department of Urology, Harbin Medical University Cancer Hospital, Harbin, 150001, China
- Heilongjiang Provincial Key Laboratory of Basic Medical Sciences in Urology Cancer, Harbin Medical University Cancer Hospital, Harbin, 150001, China
| | - Xiaowei Hu
- Department of Urogenital Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, 150001, China
| | - Zongzheng Yang
- NHC Key Laboratory of Molecular Probe and Targeted Theranostics, The Second Affiliated Hospital of Harbin Medical University, Harbin Medical University Cancer Hospital, Harbin Medical University, Harbin, 150001, China
- Department of Urology, Harbin Medical University Cancer Hospital, Harbin, 150001, China
- BGI Research, Beijing, 102601, China
- Heilongjiang Provincial Key Laboratory of Basic Medical Sciences in Urology Cancer, Harbin Medical University Cancer Hospital, Harbin, 150001, China
| | - Yubo Zhao
- NHC Key Laboratory of Molecular Probe and Targeted Theranostics, The Second Affiliated Hospital of Harbin Medical University, Harbin Medical University Cancer Hospital, Harbin Medical University, Harbin, 150001, China
- Department of Urology, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150001, China
- Heilongjiang Provincial Key Laboratory of Basic Medical Sciences in Urology Cancer, Harbin Medical University Cancer Hospital, Harbin, 150001, China
- Center for Biomedical Materials and Engineering, Harbin Engineering University, Harbin, 150001, China
| | - Qing Shi
- NHC Key Laboratory of Molecular Probe and Targeted Theranostics, The Second Affiliated Hospital of Harbin Medical University, Harbin Medical University Cancer Hospital, Harbin Medical University, Harbin, 150001, China
- Department of Urology, Harbin Medical University Cancer Hospital, Harbin, 150001, China
- Heilongjiang Provincial Key Laboratory of Basic Medical Sciences in Urology Cancer, Harbin Medical University Cancer Hospital, Harbin, 150001, China
| | - Haonan Li
- NHC Key Laboratory of Molecular Probe and Targeted Theranostics, The Second Affiliated Hospital of Harbin Medical University, Harbin Medical University Cancer Hospital, Harbin Medical University, Harbin, 150001, China
- Department of Urology, Harbin Medical University Cancer Hospital, Harbin, 150001, China
- Heilongjiang Provincial Key Laboratory of Basic Medical Sciences in Urology Cancer, Harbin Medical University Cancer Hospital, Harbin, 150001, China
| | - Ziyi Liu
- NHC Key Laboratory of Molecular Probe and Targeted Theranostics, The Second Affiliated Hospital of Harbin Medical University, Harbin Medical University Cancer Hospital, Harbin Medical University, Harbin, 150001, China
- Department of Urology, Harbin Medical University Cancer Hospital, Harbin, 150001, China
- Heilongjiang Provincial Key Laboratory of Basic Medical Sciences in Urology Cancer, Harbin Medical University Cancer Hospital, Harbin, 150001, China
| | - Zhihao Yin
- NHC Key Laboratory of Molecular Probe and Targeted Theranostics, The Second Affiliated Hospital of Harbin Medical University, Harbin Medical University Cancer Hospital, Harbin Medical University, Harbin, 150001, China
- Department of Urology, Harbin Medical University Cancer Hospital, Harbin, 150001, China
- Heilongjiang Provincial Key Laboratory of Basic Medical Sciences in Urology Cancer, Harbin Medical University Cancer Hospital, Harbin, 150001, China
| | - Ziqi Wang
- NHC Key Laboratory of Molecular Probe and Targeted Theranostics, The Second Affiliated Hospital of Harbin Medical University, Harbin Medical University Cancer Hospital, Harbin Medical University, Harbin, 150001, China.
- Department of Urology, Harbin Medical University Cancer Hospital, Harbin, 150001, China.
- Heilongjiang Provincial Key Laboratory of Basic Medical Sciences in Urology Cancer, Harbin Medical University Cancer Hospital, Harbin, 150001, China.
- Department of Cystoscope Center, Harbin Medical University Cancer Hospital, Harbin, 150001, China.
| | - Zhichao Tong
- NHC Key Laboratory of Molecular Probe and Targeted Theranostics, The Second Affiliated Hospital of Harbin Medical University, Harbin Medical University Cancer Hospital, Harbin Medical University, Harbin, 150001, China.
- Department of Urogenital Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, 150001, China.
- Biobank, Harbin Medical University Cancer Hospital, Harbin, 150001, China.
- Heilongjiang Provincial Key Laboratory of Basic Medical Sciences in Urology Cancer, Harbin Medical University Cancer Hospital, Harbin, 150001, China.
| | - Wanhai Xu
- NHC Key Laboratory of Molecular Probe and Targeted Theranostics, The Second Affiliated Hospital of Harbin Medical University, Harbin Medical University Cancer Hospital, Harbin Medical University, Harbin, 150001, China.
- Department of Urology, Harbin Medical University Cancer Hospital, Harbin, 150001, China.
- Department of Urology, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150001, China.
- Heilongjiang Provincial Key Laboratory of Basic Medical Sciences in Urology Cancer, Harbin Medical University Cancer Hospital, Harbin, 150001, China.
| |
Collapse
|
3
|
Saddala MS, Chittineni MS, Hariharan N, Rias AL, Nagaraju GP. Mitigating ambient RNA and doublets effects on single cell transcriptomics analysis in cancer research. Cancer Lett 2025; 620:217693. [PMID: 40185305 DOI: 10.1016/j.canlet.2025.217693] [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: 01/19/2025] [Revised: 03/21/2025] [Accepted: 04/02/2025] [Indexed: 04/07/2025]
Abstract
In cancer biology, where understanding the tumor microenvironment at high resolution is vital, ambient RNA contamination becomes a considerable problem. This hinders accurate delineation of intratumoral heterogeneity, complicates the identification of potential biomarkers, and decelerates advancements in precision oncology. To solve this problem, several computational approaches are created to determine the ambient RNA contribution from scRNA-seq datasets. Techniques like SoupX and DecontX assist in assessing and eliminating ambient RNA contamination from primary gene expression profiles. Practical solutions like CellBender employ deep learning techniques to concurrently address ambient RNA contamination and background noise, offering a contemporary end-to-end strategy for data preparation. This high-quality, reliable data enables clinicians and researchers to make effective decisions that will help ensure interventions are rooted in reproducible evidence, giving hope for developing more effective targeted therapies.
Collapse
Affiliation(s)
| | - Midhuna Sree Chittineni
- Department of Bioinformatics, Northeastern University College of Science, Boston, MA-021 15, USA
| | - Niharitha Hariharan
- School of Medicine, Division of Hematology and Oncology, University of Alabama at Birmingham, Birmingham, AL 352 33, USA
| | - Anijah L Rias
- School of Medicine, Division of Hematology and Oncology, University of Alabama at Birmingham, Birmingham, AL 352 33, USA
| | - Ganji Purnachandra Nagaraju
- School of Medicine, Division of Hematology and Oncology, University of Alabama at Birmingham, Birmingham, AL 352 33, USA.
| |
Collapse
|
4
|
Wu Z, Ouyang S, Gao J, Hu J, Guo Q, Liu D, Ren K. Role of Radiomics-based Multiomics Panel in the Microenvironment and Prognosis of Hepatocellular Carcinoma. Acad Radiol 2025; 32:1961-1970. [PMID: 39765431 DOI: 10.1016/j.acra.2024.12.039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Revised: 11/20/2024] [Accepted: 12/18/2024] [Indexed: 04/11/2025]
Abstract
Hepatocellular carcinoma (HCC) is the most prevalent form of liver tumor, characterized by restricted therapeutic options and typically low long-term survival rates. Recently, immunotherapy has revolutionized HCC treatment, making the tumor microenvironment (TME) a research focus. Radiomics is increasingly crucial in HCC clinical decisions, offering advanced tools for TME characterization and prognosis assessment. Meanwhile, advancements in pathomics provide new insights into HCC's comprehensive traits and details. Advancements in genomics and transcriptomics enable the integration of radiomics and pathomics with genetic data to better understand HCC heterogeneity and its microenvironment, aiding prognostic assessments. This review provides a comprehensive overview of pivotal radiomics studies focused on TME prediction, underscoring the synergistic effects of integrating multiomics approaches for TME analysis and HCC outcome prediction. It critically examines the challenges and opportunities inherent in multiomics research, emphasizing its substantial significance in both research and clinical contexts.
Collapse
Affiliation(s)
- Ziqian Wu
- Department of Radiology, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen radiological Control Center, Xiamen 361102, Fujian, China (Z.W., J.G., Q.G., K.R.)
| | - Siyu Ouyang
- Department of Radiology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510630, Guangdong, China (S.O.)
| | - Jidong Gao
- Department of Radiology, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen radiological Control Center, Xiamen 361102, Fujian, China (Z.W., J.G., Q.G., K.R.)
| | - Jingyi Hu
- Department of Radiology, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Fujian, China (J.H.)
| | - Qiu Guo
- Department of Radiology, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen radiological Control Center, Xiamen 361102, Fujian, China (Z.W., J.G., Q.G., K.R.)
| | - Danyang Liu
- Department of Radiology, The Second Affiliated Hospital of Dalian Medical University, Dalian 116027, Liaoning, China (D.L.)
| | - Ke Ren
- Department of Radiology, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen radiological Control Center, Xiamen 361102, Fujian, China (Z.W., J.G., Q.G., K.R.).
| |
Collapse
|
5
|
Long X, Kwong TT, Cheng ASL, Chan SL. Targeting tumour endothelial cells in liver cancer: The end of beginning. J Hepatol 2025; 82:553-555. [PMID: 39725355 DOI: 10.1016/j.jhep.2024.12.032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Accepted: 12/12/2024] [Indexed: 12/28/2024]
Affiliation(s)
- Xiaohang Long
- School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Tsz Tung Kwong
- State Key Laboratory of Translational Oncology, Department of Clinical Oncology, Sir YK Pao Centre for Cancer, Hong Kong Cancer Institute, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China
| | - Alfred Sze-Lok Cheng
- School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Stephen Lam Chan
- State Key Laboratory of Translational Oncology, Department of Clinical Oncology, Sir YK Pao Centre for Cancer, Hong Kong Cancer Institute, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China.
| |
Collapse
|
6
|
Huijbers EJM, van Beijnum JR, van Loon K, Griffioen CJ, Volckmann R, Bassez A, Lambrechts D, Nunes Monteiro M, Jimenez CR, Hogendoorn PCW, Koster J, Griffioen AW. Embryonic reprogramming of the tumor vasculature reveals targets for cancer therapy. Proc Natl Acad Sci U S A 2025; 122:e2424730122. [PMID: 40096611 PMCID: PMC11962416 DOI: 10.1073/pnas.2424730122] [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: 11/28/2024] [Accepted: 01/27/2025] [Indexed: 03/19/2025] Open
Abstract
A sustained blood supply is critical for tumor growth, as it delivers the nutrients and oxygen required for development. Targeting of blood vessel formation via immunotherapies is an area of great importance. Knowing that certain embryonic genes, such as carcinoembryonic antigens (CEA) and oncofetal fibronectin, become reexpressed in malignant transformation, we hypothesized that a similar phenomenon holds true for tumor endothelial cells (TECs) as well. An approach for identification of highly selective tumor endothelial markers was conducted to develop targeted antiangiogenic immunotherapies. We first queried the transcriptome that is present during embryo development. We then performed a systematic search for genes selectively expressed in the mouse embryo at days E11 and E18, as compared to the transcriptome of the adult mouse. Subsequently, we queried for expression of these embryonic genes in sorted murine TECs. This approach identified among others the tumor endothelial antigens fibrillin-2 (Fbn2), elastin microfibril interface-located protein 2 (Emilin2) as well as the tumor endothelial antigens lysyl oxidase (Lox) and serine/cysteine protease inhibitor, clade E, member 1 (Serpine1; Pai-1). For these selected genes, functional involvement in angiogenesis was confirmed in in vitro bioassays. We subsequently used iBoost conjugate vaccine technology to develop vaccines against the selected targets. For all four targets, vaccination readily induced target-specific antibody responses in mice, resulting in inhibition of tumor growth. Access to highly specific tumor endothelial markers provides opportunities for direct targeting of the tumor vasculature with high specificity, without affecting healthy vasculature.
Collapse
Affiliation(s)
- Elisabeth J. M. Huijbers
- Department of Medical Oncology, Angiogenesis Laboratory, Amsterdam University Medical Center, Cancer Center Amsterdam, Amsterdam1081 HV, The Netherlands
- CimCure Besloten Vennootschap, Amsterdam1066 CX, The Netherlands
| | - Judy R. van Beijnum
- Department of Medical Oncology, Angiogenesis Laboratory, Amsterdam University Medical Center, Cancer Center Amsterdam, Amsterdam1081 HV, The Netherlands
| | - Karlijn van Loon
- Department of Medical Oncology, Angiogenesis Laboratory, Amsterdam University Medical Center, Cancer Center Amsterdam, Amsterdam1081 HV, The Netherlands
| | - Christian J. Griffioen
- Laboratory of Experimental Oncology and Radiobiology, Amsterdam University Medical Center, Cancer Center Amsterdam, Amsterdam1105 AZ, The Netherlands
| | - Richard Volckmann
- Laboratory of Experimental Oncology and Radiobiology, Amsterdam University Medical Center, Cancer Center Amsterdam, Amsterdam1105 AZ, The Netherlands
| | - Ayse Bassez
- Vlaams Instituut voor Biotechnologie-Katolieke Universiteit Leuven Center for Cancer Biology, Leuven3000, Belgium
| | - Diether Lambrechts
- Vlaams Instituut voor Biotechnologie-Katolieke Universiteit Leuven Center for Cancer Biology, Leuven3000, Belgium
| | - Madalena Nunes Monteiro
- Department of Medical Oncology, Oncoproteomics Laboratory, Amsterdam University Medical Center, Cancer Center Amsterdam, Amsterdam1081 HV, The Netherlands
| | - Connie R. Jimenez
- Department of Medical Oncology, Oncoproteomics Laboratory, Amsterdam University Medical Center, Cancer Center Amsterdam, Amsterdam1081 HV, The Netherlands
| | | | - Jan Koster
- Laboratory of Experimental Oncology and Radiobiology, Amsterdam University Medical Center, Cancer Center Amsterdam, Amsterdam1105 AZ, The Netherlands
| | - Arjan W. Griffioen
- Department of Medical Oncology, Angiogenesis Laboratory, Amsterdam University Medical Center, Cancer Center Amsterdam, Amsterdam1081 HV, The Netherlands
- CimCure Besloten Vennootschap, Amsterdam1066 CX, The Netherlands
| |
Collapse
|
7
|
Zhong J, Gao RR, Zhang X, Yang JX, Liu Y, Ma J, Chen Q. Dissecting endothelial cell heterogeneity with new tools. CELL REGENERATION (LONDON, ENGLAND) 2025; 14:10. [PMID: 40121354 PMCID: PMC11929667 DOI: 10.1186/s13619-025-00223-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Revised: 02/20/2025] [Accepted: 02/22/2025] [Indexed: 03/25/2025]
Abstract
The formation of a blood vessel network is crucial for organ development and regeneration. Over the past three decades, the central molecular mechanisms governing blood vessel growth have been extensively studied. Recent evidence indicates that vascular endothelial cells-the specialized cells lining the inner surface of blood vessels-exhibit significant heterogeneity to meet the specific needs of different organs. This review focuses on the current understanding of endothelial cell heterogeneity, which includes both intra-organ and inter-organ heterogeneity. Intra-organ heterogeneity encompasses arterio-venous and tip-stalk endothelial cell specialization, while inter-organ heterogeneity refers to organ-specific transcriptomic profiles and functions. Advances in single-cell RNA sequencing (scRNA-seq) have enabled the identification of new endothelial subpopulations and the comparison of gene expression patterns across different subsets of endothelial cells. Integrating scRNA-seq with other high-throughput sequencing technologies promises to deepen our understanding of endothelial cell heterogeneity at the epigenetic level and in a spatially resolved context. To further explore human endothelial cell heterogeneity, vascular organoids offer powerful tools for studying gene function in three-dimensional culture systems and for investigating endothelial-tissue interactions using human cells. Developing organ-specific vascular organoids presents unique opportunities to unravel inter-organ endothelial cell heterogeneity and its implications for human disease. Emerging technologies, such as scRNA-seq and vascular organoids, are poised to transform our understanding of endothelial cell heterogeneity and pave the way for innovative therapeutic strategies to address human vascular diseases.
Collapse
Affiliation(s)
- Jing Zhong
- Center for Cell Lineage Atlas, CAS Key Laboratory of Regenerative Biology, Joint School of Life Sciences, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou Medical University, Guangzhou, 510530, China
- University of Chinese Academy of Sciences, Beijing, China
- China-New Zealand Belt and Road Joint Laboratory on Biomedicine and Health, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangdong-Hong Kong Joint Laboratory for Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China
- Center for Cell Lineage Atlas, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China
| | - Rong-Rong Gao
- Biomedical Sciences College & Shandong Medicinal Biotechnology Centre, Shandong First Medical University & Shandong Academy of Medical Sciences, NHC Key Laboratory of Biotechnology Drugs (Shandong Academy of Medical Sciences); Key Lab for Rare & Uncommon Diseases of Shandong Province, Ji'nan 250117, Shandong, China
| | - Xin Zhang
- Center for Cell Lineage Atlas, CAS Key Laboratory of Regenerative Biology, Joint School of Life Sciences, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou Medical University, Guangzhou, 510530, China
- University of Chinese Academy of Sciences, Beijing, China
- China-New Zealand Belt and Road Joint Laboratory on Biomedicine and Health, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangdong-Hong Kong Joint Laboratory for Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China
- Center for Cell Lineage Atlas, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China
| | - Jia-Xin Yang
- The Innovation Centre of Ministry of Education for Development and Diseases, School of Medicine, South China University of Technology, Guangzhou, 510006, China
| | - Yang Liu
- The Innovation Centre of Ministry of Education for Development and Diseases, School of Medicine, South China University of Technology, Guangzhou, 510006, China.
| | - Jinjin Ma
- The Innovation Centre of Ministry of Education for Development and Diseases, School of Medicine, South China University of Technology, Guangzhou, 510006, China.
- The Institute of Future Health, South China of Technology, Guangzhou International Campus, Guangzhou, 511442, China.
| | - Qi Chen
- Center for Cell Lineage Atlas, CAS Key Laboratory of Regenerative Biology, Joint School of Life Sciences, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou Medical University, Guangzhou, 510530, China.
- China-New Zealand Belt and Road Joint Laboratory on Biomedicine and Health, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangdong-Hong Kong Joint Laboratory for Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China.
- Center for Cell Lineage Atlas, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China.
- Biomedical Sciences College & Shandong Medicinal Biotechnology Centre, Shandong First Medical University & Shandong Academy of Medical Sciences, NHC Key Laboratory of Biotechnology Drugs (Shandong Academy of Medical Sciences); Key Lab for Rare & Uncommon Diseases of Shandong Province, Ji'nan 250117, Shandong, China.
| |
Collapse
|
8
|
Sun X, Cai W, Li H, Gao C, Ma X, Guo Y, Fu D, Xiao D, Zhang Z, Wang Y, Yang S, Feng Y, Zhao T, Hao J. Endothelial-like cancer-associated fibroblasts facilitate pancreatic cancer metastasis via vasculogenic mimicry and paracrine signalling. Gut 2025:gutjnl-2024-333638. [PMID: 40122596 DOI: 10.1136/gutjnl-2024-333638] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Accepted: 03/08/2025] [Indexed: 03/25/2025]
Abstract
BACKGROUND Cancer-associated fibroblasts (CAFs) are highly heterogeneous in the progression of pancreatic ductal adenocarcinoma (PDAC) and vasculogenic mimicry (VM) refers to a phenomenon in which cancer cells adopt endothelial-like characteristics. OBJECTIVE To identify a novel protumoural CAF subtype undertaking VM. DESIGN We used single-cell RNA sequencing and mIHC to identify FAPα+CD144+ endothelial-like CAFs (endoCAFs) and combined prospective and retrospective analyses to assess its clinical outcomes. Tube formation, proliferation and invasion assay were conducted on cell lines, organoids, the orthotopic tumour model and LSL-KrasG12D/+, LSL-Trp53R172H/+ and Pdx1-Cre (KPC) mouse model. Mechanically, we performed cytokine array assays, RNA-sequencing, IP-mass spectrometry, ChIP and luciferase analyses. Importantly, an siRNA delivery nanosystem was developed to precisely target FAPα+CD144+endoCAFs in vivo. RESULTS FAPα+CD144+endoCAFs were present in the tumour microenvironment of PDAC, and patients with a higher CD144+CAFs proportion displayed poor prognosis of PDAC. FAPα+CD144+endoCAFs not only acquired a VM phenotype to provide metastatic conduits but also promoted the proliferation and invasion of tumour cells in situ through paracrine signalling, thereby actively facilitating the metastasis of tumour cells. The CD144-β-catenin-STAT3 signalling axis was activated, and CD144 and downstream secreted cytokines were transcriptionally upregulated to maintain the dual roles of endoCAFs. A CAF-targeting siRNA delivery nanosystem, via loading FAPα and siCD144, was administered to precisely target FAPα+CD144+ endoCAFs, which substantially inhibited their protumoural roles in vivo. CONCLUSION FAPα+CD144+endoCAFs can promote metastasis of PDAC via undertaking VM and paracrine through activation of the CD144-β-catenin-STAT3 signalling axis. CAF-targeting siRNA delivery nanosystem can inhibit tumour progression by precisely targeting FAPα+CD144+endoCAFs.
Collapse
Affiliation(s)
- Xugang Sun
- Pancreas Center, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin Key Laboratory of Digestive Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin, People's Republic of China
| | - Wenrun Cai
- The First Department of Breast Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, People's Republic of China
| | - Haorui Li
- Pancreas Center, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin Key Laboratory of Digestive Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin, People's Republic of China
| | - Chuntao Gao
- Pancreas Center, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin Key Laboratory of Digestive Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin, People's Republic of China
| | - Xi Ma
- Pancreas Center, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin Key Laboratory of Digestive Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin, People's Republic of China
| | - Yu Guo
- Pancreas Center, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin Key Laboratory of Digestive Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin, People's Republic of China
| | - Danqi Fu
- Pancreas Center, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin Key Laboratory of Digestive Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin, People's Republic of China
| | - Di Xiao
- Pancreas Center, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin Key Laboratory of Digestive Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin, People's Republic of China
| | - Zhaoyu Zhang
- Pancreas Center, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin Key Laboratory of Digestive Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin, People's Republic of China
| | - Yifei Wang
- Pancreas Center, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin Key Laboratory of Digestive Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin, People's Republic of China
| | - Shengyu Yang
- Cellular and Molecular Physiology, Penn State College of Medicine, Hershey, Pennsylvania, USA
| | - Yukuan Feng
- Pancreas Center, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin Key Laboratory of Digestive Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin, People's Republic of China
| | - Tiansuo Zhao
- Pancreas Center, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin Key Laboratory of Digestive Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin, People's Republic of China
| | - Jihui Hao
- Pancreas Center, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin Key Laboratory of Digestive Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin, People's Republic of China
| |
Collapse
|
9
|
Liu YT, Liu HM, Ren JG, Zhang W, Wang XX, Yu ZL, Fu QY, Xiong XP, Jia J, Liu B, Chen G. Immune-featured stromal niches associate with response to neoadjuvant immunotherapy in oral squamous cell carcinoma. Cell Rep Med 2025; 6:102024. [PMID: 40107247 PMCID: PMC11970382 DOI: 10.1016/j.xcrm.2025.102024] [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: 05/05/2024] [Revised: 11/19/2024] [Accepted: 02/19/2025] [Indexed: 03/22/2025]
Abstract
Tumor stromal cells (TSCs) play a crucial yet underexplored role in the tumor microenvironment (TME). This study uses single-cell sequencing and spatial transcriptomics on paired tumor specimens from 22 patients with oral squamous cell carcinoma (OSCC) enrolled in a randomized two-arm phase 2 trial, receiving neoadjuvant anti-PD-1 mono-immunotherapy or anti-PD-1 plus docetaxel-cisplatin-5-fluorouracil (TPF) immunochemotherapy. Single-cell analysis reveals increased TSCs within the TME of responders in immunochemotherapy. Notably, significant post-treatment upregulation of SELP+ high endothelial venules (HEVs) and APOD+ myofibroblastic cancer-associated fibroblasts (myCAFs), alongside a decline in STMN1+ capillary endothelial cells (cECs), is specific to the immunochemotherapy cohort. In contrast, MYF5+ muscle satellite cells (MSCs) are upregulated in non-responders to mono-immunotherapy. SELP+ HEVs and APOD+ myCAFs foster favorable immunomodulatory stromal niches for improved outcomes, while STMN1+ cECs and MYF5+ MSCs form immunosuppressive niches in tumor invasion regions, highlighting therapeutic targets. The trial was registered at ClinicalTrials.gov, and the registration number is NCT04649476.
Collapse
Affiliation(s)
- Yu-Tong Liu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Hai-Ming Liu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Jian-Gang Ren
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China; Department of Oral and Maxillofacial Surgery, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Wei Zhang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China; Department of Oral and Maxillofacial Surgery, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Xin-Xin Wang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Zi-Li Yu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China; Department of Oral and Maxillofacial Surgery, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Qiu-Yun Fu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China; Department of Oral and Maxillofacial Surgery, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Xue-Peng Xiong
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China; Department of Oral and Maxillofacial Surgery, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Jun Jia
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China; Department of Oral and Maxillofacial Surgery, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Bing Liu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China; Department of Oral and Maxillofacial Surgery, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Gang Chen
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China; Department of Oral and Maxillofacial Surgery, School and Hospital of Stomatology, Wuhan University, Wuhan, China; TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan, China; Frontier Science Center for Immunology and Metabolism, Wuhan University, Wuhan, China.
| |
Collapse
|
10
|
Turlej E, Domaradzka A, Radzka J, Drulis-Fajdasz D, Kulbacka J, Gizak A. Cross-Talk Between Cancer and Its Cellular Environment-A Role in Cancer Progression. Cells 2025; 14:403. [PMID: 40136652 PMCID: PMC11940884 DOI: 10.3390/cells14060403] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2025] [Revised: 02/27/2025] [Accepted: 03/06/2025] [Indexed: 03/27/2025] Open
Abstract
The tumor microenvironment is a dynamic and complex three-dimensional network comprising the extracellular matrix and diverse non-cancerous cells, including fibroblasts, adipocytes, endothelial cells and various immune cells (lymphocytes T and B, NK cells, dendritic cells, monocytes/macrophages, myeloid-derived suppressor cells, and innate lymphoid cells). A constantly and rapidly growing number of studies highlight the critical role of these cells in shaping cancer survival, metastatic potential and therapy resistance. This review provides a synthesis of current knowledge on the modulating role of the cellular microenvironment in cancer progression and response to treatment.
Collapse
Affiliation(s)
- Eliza Turlej
- Departament of Molecular Physiology and Neurobiology, University of Wrocław, ul. Sienkiewicza 21, 50-335 Wrocław, Poland; (E.T.); (A.D.); (J.R.)
| | - Aleksandra Domaradzka
- Departament of Molecular Physiology and Neurobiology, University of Wrocław, ul. Sienkiewicza 21, 50-335 Wrocław, Poland; (E.T.); (A.D.); (J.R.)
| | - Justyna Radzka
- Departament of Molecular Physiology and Neurobiology, University of Wrocław, ul. Sienkiewicza 21, 50-335 Wrocław, Poland; (E.T.); (A.D.); (J.R.)
| | - Dominika Drulis-Fajdasz
- Departament of Molecular Physiology and Neurobiology, University of Wrocław, ul. Sienkiewicza 21, 50-335 Wrocław, Poland; (E.T.); (A.D.); (J.R.)
| | - Julita Kulbacka
- Departament of Molecular and Cellular Biology, Faculty of Pharmacy, Wrocław Medical University, Borowska 211A, 50-556 Wrocław, Poland;
- Department of Immunology and Bioelectrochemistry, State Research Institute Centre for Innovative Medicine, LT-08406 Vilnius, Lithuania
| | - Agnieszka Gizak
- Departament of Molecular Physiology and Neurobiology, University of Wrocław, ul. Sienkiewicza 21, 50-335 Wrocław, Poland; (E.T.); (A.D.); (J.R.)
| |
Collapse
|
11
|
Ge S, Sun S, Xu H, Cheng Q, Ren Z. Deep learning in single-cell and spatial transcriptomics data analysis: advances and challenges from a data science perspective. Brief Bioinform 2025; 26:bbaf136. [PMID: 40185158 PMCID: PMC11970898 DOI: 10.1093/bib/bbaf136] [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: 12/16/2024] [Revised: 02/17/2025] [Accepted: 03/05/2025] [Indexed: 04/07/2025] Open
Abstract
The development of single-cell and spatial transcriptomics has revolutionized our capacity to investigate cellular properties, functions, and interactions in both cellular and spatial contexts. Despite this progress, the analysis of single-cell and spatial omics data remains challenging. First, single-cell sequencing data are high-dimensional and sparse, and are often contaminated by noise and uncertainty, obscuring the underlying biological signal. Second, these data often encompass multiple modalities, including gene expression, epigenetic modifications, metabolite levels, and spatial locations. Integrating these diverse data modalities is crucial for enhancing prediction accuracy and biological interpretability. Third, while the scale of single-cell sequencing has expanded to millions of cells, high-quality annotated datasets are still limited. Fourth, the complex correlations of biological tissues make it difficult to accurately reconstruct cellular states and spatial contexts. Traditional feature engineering approaches struggle with the complexity of biological networks, while deep learning, with its ability to handle high-dimensional data and automatically identify meaningful patterns, has shown great promise in overcoming these challenges. Besides systematically reviewing the strengths and weaknesses of advanced deep learning methods, we have curated 21 datasets from nine benchmarks to evaluate the performance of 58 computational methods. Our analysis reveals that model performance can vary significantly across different benchmark datasets and evaluation metrics, providing a useful perspective for selecting the most appropriate approach based on a specific application scenario. We highlight three key areas for future development, offering valuable insights into how deep learning can be effectively applied to transcriptomic data analysis in biological, medical, and clinical settings.
Collapse
Affiliation(s)
- Shuang Ge
- Shenzhen International Graduate School, Tsinghua University, 2279 Lishui Road, Nanshan District, Shenzhen 518055, Guangdong, China
- Pengcheng Laboratory, 6001 Shahe West Road, Nanshan District, Shenzhen 518055, Guangdong, China
| | - Shuqing Sun
- Shenzhen International Graduate School, Tsinghua University, 2279 Lishui Road, Nanshan District, Shenzhen 518055, Guangdong, China
| | - Huan Xu
- School of Public Health, Anhui University of Science and Technology, 15 Fengxia Road, Changfeng County, Hefei 231131, Anhui, China
| | - Qiang Cheng
- Department of Computer Science, University of Kentucky, 329 Rose Street, Lexington 40506, Kentucky, USA
- Institute for Biomedical Informatics, University of Kentucky, 800 Rose Street, Lexington 40506, Kentucky, USA
| | - Zhixiang Ren
- Pengcheng Laboratory, 6001 Shahe West Road, Nanshan District, Shenzhen 518055, Guangdong, China
| |
Collapse
|
12
|
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
|
13
|
Wang Y, Xue Y, Wang H, Qu Y, Zhang K, Shang L, Liang P, Chen F, Tang X, Luo W, Chin LK, Feng S, Li B. Automated Laser-Assisted Single-Cell Sorting for Cell Functional and RNA Sequencing. ACS Sens 2025; 10:846-856. [PMID: 39843241 DOI: 10.1021/acssensors.4c02417] [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] [Indexed: 01/24/2025]
Abstract
Accurate and efficient sorting of single target cells is crucial for downstream single-cell analysis, such as RNA sequencing, to uncover cellular heterogeneity and functional characteristics. However, conventional single-cell sorting techniques, such as manual micromanipulation or fluorescence-activated cell sorting, do not match current demands and are limited by low throughput, low sorting efficiency and precision, or limited cell viability. Here, we report an automated, highly efficient single-cell sorter, integrating laser-induced forward transfer (LIFT) with a high-throughput picoliter micropore array. The micropore array was surface-functionalized to manipulate liquid surface tension, facilitating the formation of single-cell picoliter droplets in the micropores to realize automated and highly efficient (>80%) single-cell isolation. Using an in-house built microscopic system, rare target cells were identified and automatically retrieved by LIFT with precise sorting efficiency (about 100%) for downstream single-cell analysis while maintaining high cell viability (about 80%). As a case demonstration, we demonstrated the accurate sorting of rare transfected PC-9 cells and post-transfection cell culture, minimizing cell loss and the risk of contamination. Furthermore, we performed single-cell RNA sequencing and showed that high-quality single-cell transcriptome information was efficiently and reliably obtained during cell sorting, preventing additional costs due to low sorting accuracy. The single-cell sorter will become invaluable for single-cell analysis, laying the foundation for multiomics analysis and precision medicine research.
Collapse
Affiliation(s)
- Yuntong Wang
- Changchun Institute of Optics, Fine Mechanics and Physics, Chinese Academy of Sciences, Changchun 130033, P. R. China
- University of Chinese Academy of Sciences, Beijing 100049, P. R. China
- State Key Laboratory of Applied Optics, Changchun 130033, P. R. China
- Key Laboratory of Advanced Manufacturing for Optical Systems, Chinese Academy of Sciences, Changchun 130033, P. R. China
| | - Ying Xue
- Hooke Laboratory, Changchun 130033, P. R. China
| | - Huan Wang
- Changchun Institute of Optics, Fine Mechanics and Physics, Chinese Academy of Sciences, Changchun 130033, P. R. China
- University of Chinese Academy of Sciences, Beijing 100049, P. R. China
| | - Yue Qu
- Hooke Laboratory, Changchun 130033, P. R. China
- Haining High-tech Research Institute, Jiaxing, Zhejiang 314408, P. R. China
| | | | - Lindong Shang
- Changchun Institute of Optics, Fine Mechanics and Physics, Chinese Academy of Sciences, Changchun 130033, P. R. China
- University of Chinese Academy of Sciences, Beijing 100049, P. R. China
- State Key Laboratory of Applied Optics, Changchun 130033, P. R. China
- Key Laboratory of Advanced Manufacturing for Optical Systems, Chinese Academy of Sciences, Changchun 130033, P. R. China
| | - Peng Liang
- Changchun Institute of Optics, Fine Mechanics and Physics, Chinese Academy of Sciences, Changchun 130033, P. R. China
- University of Chinese Academy of Sciences, Beijing 100049, P. R. China
- State Key Laboratory of Applied Optics, Changchun 130033, P. R. China
- Key Laboratory of Advanced Manufacturing for Optical Systems, Chinese Academy of Sciences, Changchun 130033, P. R. China
| | - Fuyuan Chen
- Changchun Institute of Optics, Fine Mechanics and Physics, Chinese Academy of Sciences, Changchun 130033, P. R. China
- University of Chinese Academy of Sciences, Beijing 100049, P. R. China
- State Key Laboratory of Applied Optics, Changchun 130033, P. R. China
- Key Laboratory of Advanced Manufacturing for Optical Systems, Chinese Academy of Sciences, Changchun 130033, P. R. China
| | - Xusheng Tang
- Changchun Institute of Optics, Fine Mechanics and Physics, Chinese Academy of Sciences, Changchun 130033, P. R. China
- University of Chinese Academy of Sciences, Beijing 100049, P. R. China
- State Key Laboratory of Applied Optics, Changchun 130033, P. R. China
- Key Laboratory of Advanced Manufacturing for Optical Systems, Chinese Academy of Sciences, Changchun 130033, P. R. China
| | - Wei Luo
- Department of Electrical and Electronic Engineering, The Hong Kong Polytechnic University, Kowloon, Hong Kong SAR 999077, China
| | - Lip Ket Chin
- Department of Electrical Engineering, City University of Hong Kong, Kowloon, Hong Kong SAR 999077, China
| | - Shilun Feng
- State Key Laboratory of Transducer Technology, Shanghai Institute of Microsystem and Information Technology, Chinese Academy of Sciences, Shanghai 200050, China
| | - Bei Li
- Changchun Institute of Optics, Fine Mechanics and Physics, Chinese Academy of Sciences, Changchun 130033, P. R. China
- University of Chinese Academy of Sciences, Beijing 100049, P. R. China
- State Key Laboratory of Applied Optics, Changchun 130033, P. R. China
- Key Laboratory of Advanced Manufacturing for Optical Systems, Chinese Academy of Sciences, Changchun 130033, P. R. China
| |
Collapse
|
14
|
Holm A, Graus MS, Wylie-Sears J, Tan JWH, Alvarez-Harmon M, Borgelt L, Nasim S, Chung L, Jain A, Sun M, Sun L, Brouillard P, Lekwuttikarn R, Qi Y, Teng J, Vikkula M, Kozakewich H, Mulliken JB, Francois M, Bischoff J. An endothelial SOX18-mevalonate pathway axis enables repurposing of statins for infantile hemangioma. J Clin Invest 2025; 135:e179782. [PMID: 39998898 PMCID: PMC11957709 DOI: 10.1172/jci179782] [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: 01/30/2024] [Accepted: 02/07/2025] [Indexed: 02/27/2025] Open
Abstract
Infantile hemangioma (IH) is the most common tumor in children and a paradigm for pathological vasculogenesis, angiogenesis, and regression. Propranolol, the mainstay of treatment, inhibits IH vessel formation via a β-adrenergic receptor-independent off-target effect of its R(+) enantiomer on endothelial SOX18 - a member of the SOX (SRY-related HMG-box) family of transcription factors. Transcriptomic profiling of patient-derived hemangioma stem cells uncovered the mevalonate pathway (MVP) as a target of R(+) propranolol. Loss and gain of function of SOX18 confirmed it is both necessary and sufficient for R(+) propranolol suppression of the MVP, including regulation of sterol regulatory element-binding protein 2 (SREBP2) and the rate-limiting enzyme HMG-CoA reductase (HMGCR). A biological relevance of the endothelial SOX18-MVP axis in IH patient tissue was demonstrated by nuclear colocalization of SOX18 and SREBP2. Functional validation in a preclinical IH xenograft model revealed that statins - competitive inhibitors of HMGCR - efficiently suppress IH vessel formation. We propose an endothelial SOX18-MVP axis as a central regulator of IH pathogenesis and suggest statin repurposing to treat IH. The pleiotropic effects of R(+) propranolol and statins along the SOX18-MVP axis to disable an endothelial cell-specific program may have therapeutic implications for other vascular disease entities involving pathological vasculogenesis and angiogenesis.
Collapse
Affiliation(s)
- Annegret Holm
- Vascular Biology Program, Department of Surgery, Boston Children’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Matthew S. Graus
- The David Richmond Laboratory for Cardiovascular Development: Gene Regulation and Editing, Centenary Institute, University of Sydney, Camperdown, New South Wales, Australia
| | - Jill Wylie-Sears
- Vascular Biology Program, Department of Surgery, Boston Children’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Jerry Wei Heng Tan
- Vascular Biology Program, Department of Surgery, Boston Children’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Maya Alvarez-Harmon
- Vascular Biology Program, Department of Surgery, Boston Children’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Luke Borgelt
- Vascular Biology Program, Department of Surgery, Boston Children’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Sana Nasim
- Vascular Biology Program, Department of Surgery, Boston Children’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Long Chung
- The David Richmond Laboratory for Cardiovascular Development: Gene Regulation and Editing, Centenary Institute, University of Sydney, Camperdown, New South Wales, Australia
| | - Ashish Jain
- Research Computing, Information Technology, Boston Children’s Hospital, Boston, Massachusetts, USA
| | - Mingwei Sun
- Research Computing, Information Technology, Boston Children’s Hospital, Boston, Massachusetts, USA
| | - Liang Sun
- Research Computing, Information Technology, Boston Children’s Hospital, Boston, Massachusetts, USA
| | - Pascal Brouillard
- Human Molecular Genetics, de Duve Institute, University of Louvain, VASCERN-VASCA European Reference Center, Brussels, Belgium
| | - Ramrada Lekwuttikarn
- Department of Dermatology, Lucile Packard Children’s Hospital at the Stanford University School of Medicine, Palo Alto, California, USA
| | - Yanfei Qi
- The David Richmond Laboratory for Cardiovascular Development: Gene Regulation and Editing, Centenary Institute, University of Sydney, Camperdown, New South Wales, Australia
| | - Joyce Teng
- Department of Dermatology, Lucile Packard Children’s Hospital at the Stanford University School of Medicine, Palo Alto, California, USA
| | - Miikka Vikkula
- Human Molecular Genetics, de Duve Institute, University of Louvain, VASCERN-VASCA European Reference Center, Brussels, Belgium
- WELBIO Department, WEL Research Institute, Wavre, Belgium
| | - Harry Kozakewich
- Department of Pathology, Boston Children’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - John B. Mulliken
- Department of Plastic and Oral Surgery, Boston Children’s Hospital; Department of Surgery, Harvard Medical School; Boston, Massachusetts, USA
| | - Mathias Francois
- The David Richmond Laboratory for Cardiovascular Development: Gene Regulation and Editing, Centenary Institute, University of Sydney, Camperdown, New South Wales, Australia
- School of Biomedical Sciences, University of Sydney, Camperdown, New South Wales, Australia
| | - Joyce Bischoff
- Vascular Biology Program, Department of Surgery, Boston Children’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
15
|
Huang Q, Yang W, Wang F, Huang R, Wang Q, Li X, Lei T, Yue S, Zou W, An Q, Yue J, Hu Q, Liu C. SELP + TEC:CD8 + T cell crosstalk associates with improved radiotherapy efficacy in cervical cancer. Mol Cancer 2025; 24:41. [PMID: 39901166 PMCID: PMC11789365 DOI: 10.1186/s12943-025-02244-7] [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: 08/28/2024] [Accepted: 01/20/2025] [Indexed: 02/05/2025] Open
Abstract
P-selectin (SELP) expression in tumor cells has been implicated in promoting tumor progression and treatment resistance across various cancers. However, our prior study identified SELP expression in a specific subpopulation of endothelial cells within cervical cancer (CC) and potentially linked to anti-cancer immune response. The precise mechanisms by which SELP influences anti-cancer immunity and its involvement in radiotherapy response in CC, however, remain elusive. To address these gaps, this study analyzed tumor tissue samples from 205 CC patients undergoing radiotherapy, scRNA-seq data from 42,159 cells of eight patients, and bulk RNA-sequencing data from 187 radiotherapy-treated patients. The results revealed that elevated SELP expression in tumor endothelial cells (TECs) was significantly correlated with improved survival outcomes in patients treated with radiotherapy. The SELPhigh group exhibited a prominent enrichment of immune-related pathways, coupled with a diminished enrichment in epithelial cell proliferation and angiogenesis pathways. Notably, this group demonstrated increased infiltration of CD8+ T cells and enhanced expression of chemokine receptors, including ACKR1. Furthermore, our data suggest that SELP+ TECs engage in crosstalk with CD8+ T cells via the ACKR1-CCL5 axis, which is associated with improved radiotherapy efficacy. In conclusion, these findings underscore the pivotal role of SELP+ TEC:CD8+ T cell interactions through the ACKR1-CCL5 pathway in enhancing radiotherapy response in CC. Targeting this crosstalk may offer novel therapeutic strategies to mitigate treatment resistance and improve patient survival.
Collapse
Affiliation(s)
- Qingyu Huang
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, 250117, China
| | - Wenhui Yang
- Department of Oncology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Fuhao Wang
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, 250117, China
| | - Rui Huang
- Department of Oncology, Chongqing University Fuling Hospital, Chongqing, China
| | - Qian Wang
- Department of Oncology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Xiaohui Li
- Department of Medical Oncology, Peking University First Hospital, Beijing, China
| | - Tianyu Lei
- Department of Oncology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Shengqin Yue
- Department of Oncology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Wenxue Zou
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, 250117, China
- Department of Oncology, Linyi Center Hospital, Linyi, Shandong, China
| | - Qi An
- Department of Oncology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Jinbo Yue
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, 250117, China.
| | - Qinyong Hu
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, 250117, China.
- Department of Oncology, Renmin Hospital of Wuhan University, Wuhan, 430060, China.
| | - Chao Liu
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, 250117, China.
| |
Collapse
|
16
|
Vojoudi H, Soroush M. Isolation of Biomolecules Using MXenes. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025; 37:e2415160. [PMID: 39663732 DOI: 10.1002/adma.202415160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/06/2024] [Revised: 11/14/2024] [Indexed: 12/13/2024]
Abstract
Biomolecule isolation is a crucial process in diverse biomedical and biochemical applications, including diagnostics, therapeutics, research, and manufacturing. Recently, MXenes, a novel class of two-dimensional nanomaterials, have emerged as promising adsorbents for this purpose due to their unique physicochemical properties. These biocompatible and antibacterial nanomaterials feature a high aspect ratio, excellent conductivity, and versatile surface chemistry. This timely review explores the potential of MXenes for isolating a wide range of biomolecules, such as proteins, nucleic acids, and small molecules, while highlighting key future research trends and innovative applications poised to transform the field. This review provides an in-depth discussion of various synthesis methods and functionalization techniques that enhance the specificity and efficiency of MXenes in biomolecule isolation. In addition, the mechanisms by which MXenes interact with biomolecules are elucidated, offering insights into their selective adsorption and customized separation capabilities. This review also addresses recent advancements, identifies existing challenges, and examines emerging trends that may drive the next wave of innovation in this rapidly evolving area.
Collapse
Affiliation(s)
- Hossein Vojoudi
- Department of Chemical and Biological Engineering, Drexel University, Philadelphia, PA, 19104, USA
| | - Masoud Soroush
- Department of Chemical and Biological Engineering, Drexel University, Philadelphia, PA, 19104, USA
- Department of Materials Science and Engineering, Drexel University, Philadelphia, PA, 19104, USA
| |
Collapse
|
17
|
Wei S, Han C, Mo S, Huang H, Luo X. Advancements in programmed cell death research in antitumor therapy: a comprehensive overview. Apoptosis 2025; 30:401-421. [PMID: 39487314 DOI: 10.1007/s10495-024-02038-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/24/2024] [Indexed: 11/04/2024]
Abstract
Cell death is a normal physiological process within cells that involves multiple pathways, such as normal DNA damage, cell cycle arrest, and programmed cell death (PCD). Cell death has been a hot spot of research in tumor-related fields, especially programmed cell death, which is a key form of cell death and is classified into different types according to the mechanism of occurrence, such as apoptosis, autophagy, necroptosis, pyroptosis, ferroptosis, and disulfidptosis. Given the important role of PCD in maintaining tissue homeostasis and inhibiting tumorigenesis and development, more and more basic and clinical studies are devoted to revealing its potential application in anti-tumor strategies. The purpose of this review is to systematically review the regulatory mechanisms of PCD and to summarize the latest research progress of anti-tumor treatment strategies based on PCD.
Collapse
Affiliation(s)
- Shuxin Wei
- School of Basic Medical Sciences, Guangxi Medical University, Nanning, 530021, China
- Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor, Ministry of Education, Guangxi Medical University, Nanning, 530021, China
| | - Chuangye Han
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, China
- Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor, Ministry of Education, Guangxi Medical University, Nanning, 530021, China
| | - Shutian Mo
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, China
| | - Hailian Huang
- School of Basic Medical Sciences, Guangxi Medical University, Nanning, 530021, China
- Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor, Ministry of Education, Guangxi Medical University, Nanning, 530021, China
| | - Xiaoling Luo
- School of Basic Medical Sciences, Guangxi Medical University, Nanning, 530021, China.
- Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor, Ministry of Education, Guangxi Medical University, Nanning, 530021, China.
- Department of Experimental Research, Guangxi Medical University Cancer Hospital, Nanning, 530021, China.
| |
Collapse
|
18
|
Guo W, Zhou B, Dou L, Guo L, Li Y, Qin J, Wang Z, Huai Q, Xue X, Li Y, Ying J, Xue Q, Gao S, He J. Single-cell RNA sequencing and spatial transcriptomics of esophageal squamous cell carcinoma with lymph node metastases. Exp Mol Med 2025; 57:59-71. [PMID: 39741182 PMCID: PMC11799171 DOI: 10.1038/s12276-024-01369-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: 01/13/2024] [Revised: 09/25/2024] [Accepted: 09/26/2024] [Indexed: 01/02/2025] Open
Abstract
Esophageal squamous cell carcinoma (ESCC) patients often face a grim prognosis due to lymph node metastasis. However, a comprehensive understanding of the cellular and molecular characteristics of metastatic lymph nodes in ESCC remains elusive. In this study involving 12 metastatic ESCC patients, we employed single-cell sequencing, spatial transcriptomics (ST), and multiplex immunohistochemistry (mIHC) to explore the spatial and molecular attributes of primary tumor samples, adjacent tissues, metastatic and non-metastatic lymph nodes. The analysis of 161,333 cells revealed specific subclusters of epithelial cells that were significantly enriched in metastatic lymph nodes, suggesting pro-metastatic characteristics. Furthermore, stromal cells in the tumor microenvironment, including MMP3+IL24+ fibroblasts, APLN+ endothelial cells, and CXCL12+ pericytes, were implicated in ESCC metastasis through angiogenesis, collagen production, and inflammatory responses. Exhausted CD8+ T cells in a cycling status were notably prevalent in metastatic lymph nodes, indicating their potential role in facilitating metastasis. We identified distinct cell-cell communication networks and specific ligand-receptor pathways. Our findings were validated through a spatial transcriptome map and mIHC. This study enhances our comprehension of the cellular and molecular aspects of metastatic lymph nodes in ESCC patients, offering potential insights into novel therapeutic strategies for these individuals.
Collapse
Affiliation(s)
- Wei Guo
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Key Laboratory of Minimally Invasive Therapy Research for Lung Cancer, Chinese Academy of Medical Sciences, Beijing, China
| | - Bolun Zhou
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Lizhou Dou
- Department of Endoscopy, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Lei Guo
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yong Li
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jianjun Qin
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zhen Wang
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Qilin Huai
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xuemin Xue
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yin Li
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jianming Ying
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Qi Xue
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Key Laboratory of Minimally Invasive Therapy Research for Lung Cancer, Chinese Academy of Medical Sciences, Beijing, China
| | - Shugeng Gao
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
- Key Laboratory of Minimally Invasive Therapy Research for Lung Cancer, Chinese Academy of Medical Sciences, Beijing, China.
| | - Jie He
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| |
Collapse
|
19
|
Guo S, Liu X, Cheng X, Jiang Y, Ji S, Liang Q, Koval A, Li Y, Owen LA, Kim IK, Aparicio A, Lee S, Sood AK, Kopetz S, Shen JP, Weinstein JN, DeAngelis MM, Chen R, Wang W. A deconvolution framework that uses single-cell sequencing plus a small benchmark data set for accurate analysis of cell type ratios in complex tissue samples. Genome Res 2025; 35:147-161. [PMID: 39586714 PMCID: PMC11789644 DOI: 10.1101/gr.278822.123] [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: 12/05/2023] [Accepted: 11/19/2024] [Indexed: 11/27/2024]
Abstract
Bulk deconvolution with single-cell/nucleus RNA-seq data is critical for understanding heterogeneity in complex biological samples, yet the technological discrepancy across sequencing platforms limits deconvolution accuracy. To address this, we utilize an experimental design to match inter-platform biological signals, hence revealing the technological discrepancy, and then develop a deconvolution framework called DeMixSC using this well-matched, that is, benchmark, data. Built upon a novel weighted nonnegative least-squares framework, DeMixSC identifies and adjusts genes with high technological discrepancy and aligns the benchmark data with large patient cohorts of matched-tissue-type for large-scale deconvolution. Our results using two benchmark data sets of healthy retinas and ovarian cancer tissues suggest much-improved deconvolution accuracy. Leveraging tissue-specific benchmark data sets, we applied DeMixSC to a large cohort of 453 age-related macular degeneration patients and a cohort of 30 ovarian cancer patients with various responses to neoadjuvant chemotherapy. Only DeMixSC successfully unveiled biologically meaningful differences across patient groups, demonstrating its broad applicability in diverse real-world clinical scenarios. Our findings reveal the impact of technological discrepancy on deconvolution performance and underscore the importance of a well-matched data set to resolve this challenge. The developed DeMixSC framework is generally applicable for accurately deconvolving large cohorts of disease tissues, including cancers, when a well-matched benchmark data set is available.
Collapse
Affiliation(s)
- Shuai Guo
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
| | - Xiaoqian Liu
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
| | - Xuesen Cheng
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas 77030, USA
| | - Yujie Jiang
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
- Department of Statistics, Rice University, Houston, Texas 77005, USA
| | - Shuangxi Ji
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
| | - Qingnan Liang
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas 77030, USA
| | - Andrew Koval
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
- Department of Statistics, Rice University, Houston, Texas 77005, USA
| | - Yumei Li
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas 77030, USA
| | - Leah A Owen
- Department of Ophthalmology, Jacobs School of Medicine and Biomedical Engineering, SUNY University at Buffalo, Buffalo, New York 14209, USA
- Department of Population Health Sciences, University of Utah School of Medicine, Salt Lake City, Utah 84108, USA
- Department of Ophthalmology and Visual Sciences, University of Utah School of Medicine, Salt Lake City, Utah 84132, USA
| | - Ivana K Kim
- USA Retina Service, Harvard Medical School, Massachusetts Eye and Ear, Boston, Massachusetts 02114, USA
| | - Ana Aparicio
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas 77230, USA
| | - Sanghoon Lee
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas 77230, USA
| | - Anil K Sood
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas 77230, USA
| | - Scott Kopetz
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
| | - John Paul Shen
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
| | - John N Weinstein
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
- Department of Systems Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
| | - Margaret M DeAngelis
- Department of Ophthalmology, Jacobs School of Medicine and Biomedical Engineering, SUNY University at Buffalo, Buffalo, New York 14209, USA
- Department of Population Health Sciences, University of Utah School of Medicine, Salt Lake City, Utah 84108, USA
- Department of Ophthalmology and Visual Sciences, University of Utah School of Medicine, Salt Lake City, Utah 84132, USA
- VA Western New York Healthcare System, Buffalo, New York 14215, USA
| | - Rui Chen
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas 77030, USA
| | - Wenyi Wang
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA;
| |
Collapse
|
20
|
Mo Y, Chen L, Zhang L, Zhao Q. Weakly Supervised Nuclei Segmentation with Point-Guided Attention and Self-Supervised Pseudo-Labeling. Bioengineering (Basel) 2025; 12:85. [PMID: 39851359 PMCID: PMC11761557 DOI: 10.3390/bioengineering12010085] [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: 12/20/2024] [Revised: 01/12/2025] [Accepted: 01/15/2025] [Indexed: 01/26/2025] Open
Abstract
Due to the labor-intensive manual annotations for nuclei segmentation, point-supervised segmentation based on nuclei coordinate supervision has gained recognition in recent years. Despite great progress, two challenges hinder the performance of weakly supervised nuclei segmentation methods: (1) The stable and effective segmentation of adjacent cell nuclei remains an unresolved challenge. (2) Existing approaches rely solely on initial pseudo-labels generated from point annotations for training, and inaccurate labels may lead the model to assimilate a considerable amount of noise information, thereby diminishing performance. To address these issues, we propose a method based on center-point prediction and pseudo-label updating for precise nuclei segmentation. First, we devise a Gaussian kernel mechanism that employs multi-scale Gaussian masks for multi-branch center-point prediction. The generated center points are utilized by the segmentation module to facilitate the effective separation of adjacent nuclei. Next, we introduce a point-guided attention mechanism that concentrates the segmentation module's attention around authentic point labels, reducing the noise impact caused by pseudo-labels. Finally, a label updating mechanism based on the exponential moving average (EMA) and k-means clustering is introduced to enhance the quality of pseudo-labels. The experimental results on three public datasets demonstrate that our approach has achieved state-of-the-art performance across multiple metrics. This method can significantly reduce annotation costs and reliance on clinical experts, facilitating large-scale dataset training and promoting the adoption of automated analysis in clinical applications.
Collapse
Affiliation(s)
| | | | | | - Qi Zhao
- Institute of Electronic Information Engineering, Beihang University, 37 Xueyuan Road, Haidian District, Beijing 100191, China; (Y.M.); (L.C.); (L.Z.)
| |
Collapse
|
21
|
Liang L, Chai C, Liu A, Nadukkandy AS, Kalaiselvan S, Brandt CB, Zhao W, Li H, Lin L, Wu J, Luo Y. Single-cell transcriptome analysis reveals reciprocal epithelial and endothelial cell evolution in ovarian cancer. iScience 2024; 27:111417. [PMID: 39717089 PMCID: PMC11665315 DOI: 10.1016/j.isci.2024.111417] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Revised: 05/26/2024] [Accepted: 11/14/2024] [Indexed: 12/25/2024] Open
Abstract
Tumor neovascularization mediated by endothelial cells (ECs) is essential for ovarian cancer (OC) progression, but interactions between epithelial cells and ECs are not well understood. Here, we analyze single-cell transcriptome of 87,847 epithelial cells and 11,696 ECs from fallopian tubes, primary and metastatic ovarian tumors. Cell differentiation trajectory analysis reveals that fallopian tube cells exhibit a potential development trend toward primary OC epithelial cells. We identify a sub-population of fallopian tube epithelial cells (FTSEC3), which highly express tumor cell markers and are enriched in vascular endothelial growth factor production. Two neovascularization-related EC phenotypes (MKI67+ proliferating ECs and ESM1+ tip cells) are specially found in ovarium tumors, which exhibit strong interactions with FTSEC3. We validate that genetic disruption of LAMININ and TGF-β with CRISPR in ECs inhibits sprouting angiogenesis. In summary, this study reveals a reciprocal evolution and interaction between epithelial and ECs in OC development and progression.
Collapse
Affiliation(s)
- Langchao Liang
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
- Lars Bolund Institute of Regenerative Medicine, Qingdao-Europe Advanced Institute for Life Science, BGI-Research, Qingdao 266555, China
| | - Chaochao Chai
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
- Lars Bolund Institute of Regenerative Medicine, Qingdao-Europe Advanced Institute for Life Science, BGI-Research, Qingdao 266555, China
| | - Anmin Liu
- HIM-BGI Omics Center, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, China
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou, China
| | | | | | | | - Wandong Zhao
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
- Lars Bolund Institute of Regenerative Medicine, Qingdao-Europe Advanced Institute for Life Science, BGI-Research, Qingdao 266555, China
| | - Hanbo Li
- Lars Bolund Institute of Regenerative Medicine, Qingdao-Europe Advanced Institute for Life Science, BGI-Research, Qingdao 266555, China
| | - Lin Lin
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
- Steno Diabetes Center Aarhus, Aarhus University Hospital, Aarhus, Denmark
| | - Jianmin Wu
- HIM-BGI Omics Center, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, China
- Institute of Genomic Medicine, Wenzhou Medical University, Wenzhou, China
| | - Yonglun Luo
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
- Steno Diabetes Center Aarhus, Aarhus University Hospital, Aarhus, Denmark
| |
Collapse
|
22
|
An J, Lu Y, Chen Y, Chen Y, Zhou Z, Chen J, Peng C, Huang R, Peng F. Spatial transcriptomics in breast cancer: providing insight into tumor heterogeneity and promoting individualized therapy. Front Immunol 2024; 15:1499301. [PMID: 39749323 PMCID: PMC11693744 DOI: 10.3389/fimmu.2024.1499301] [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: 09/20/2024] [Accepted: 12/05/2024] [Indexed: 01/04/2025] Open
Abstract
A comprehensive understanding of tumor heterogeneity, tumor microenvironment and the mechanisms of drug resistance is fundamental to advancing breast cancer research. While single-cell RNA sequencing has resolved the issue of "temporal dynamic expression" of genes at the single-cell level, the lack of spatial information still prevents us from gaining a comprehensive understanding of breast cancer. The introduction and application of spatial transcriptomics addresses this limitation. As the annual technical method of 2020, spatial transcriptomics preserves the spatial location of tissues and resolves RNA-seq data to help localize and differentiate the active expression of functional genes within a specific tissue region, enabling the study of spatial location attributes of gene locations and cellular tissue environments. In the context of breast cancer, spatial transcriptomics can assist in the identification of novel breast cancer subtypes and spatially discriminative features that show promise for individualized precise treatment. This article summarized the key technical approaches, recent advances in spatial transcriptomics and its applications in breast cancer, and discusses the limitations of current spatial transcriptomics methods and the prospects for future development, with a view to advancing the application of this technology in clinical practice.
Collapse
Affiliation(s)
- Junsha An
- West China School of Pharmacy, Sichuan University, Chengdu, China
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- Cardiovascular Department, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yajie Lu
- West China School of Pharmacy, Sichuan University, Chengdu, China
| | - Yuxi Chen
- West China School of Pharmacy, Sichuan University, Chengdu, China
| | - Yuling Chen
- West China School of Pharmacy, Sichuan University, Chengdu, China
| | - Zhaokai Zhou
- Department of Clinical Medicine, Zhengzhou University, Zhengzhou, China
| | - Jianping Chen
- School of Chinese Medicine, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Cheng Peng
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Ruizhen Huang
- Cardiovascular Department, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Fu Peng
- West China School of Pharmacy, Sichuan University, Chengdu, China
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, Sichuan University, Chengdu, China
| |
Collapse
|
23
|
Zhu XY, Liu WT, Hou XJ, Zong C, Yu W, Shen ZM, Qu SP, Tao M, Xue MM, Zhou DY, Bai HR, Gao L, Jiang JH, Zhao QD, Wei LX, Yang X, Han ZP, Zhang L. CD34 +CLDN5 + tumor associated senescent endothelial cells through IGF2-IGF2R signaling increased cholangiocellular phenotype in hepatocellular carcinoma. J Adv Res 2024:S2090-1232(24)00564-2. [PMID: 39674501 DOI: 10.1016/j.jare.2024.12.008] [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: 06/27/2024] [Revised: 11/02/2024] [Accepted: 12/05/2024] [Indexed: 12/16/2024] Open
Abstract
INTRODUCTION The heterogeneity of hepatocellular carcinoma (HCC) is linked to tumor malignancy and poor prognosis. Nevertheless, the precise mechanisms underlying the development of the cholangiocellular phenotype (CCA) within HCC remain unclear. Emerging studies support that the cross-talk among the host cells within tumor microenvironment (TME) sustains the cancer cell plasticity. OBJECTIVES This study sought to identify the specific cell types involved in the formation of CCA and to elucidate their functional roles in the progression of HCC. METHODS Single-cell RNA sequencing was employed to identify the specific cell types involved in the formation of CCA. Both in vitro and vivo analyses were used to identify the tumor-associated senescent ECs and investigate the function in TME. The diethylnitrosamine-induced model was utilized to investigate the interaction between senescent ECs and MSCs, aiming to elucidate their synergistic contributions to the progression of CCA. RESULTS Using single-cell RNA sequencing, we identified a distinct senescent-associated subset of endothelial cells (ECs), namely CD34+CLDN5+ ECs, which mainly enriched in tumor tissue. Further, the senescent ECs were observed to secrete IGF2, which recruited mesenchymal stem cells (MSCs) into the TME through IGF2R/MAPK signaling. In primary liver cancer model, MSCs exhibited a strong tumor-promoting effect, increasing the CCA and tumor malignancy after HCC formation. Interestingly, knockdown of IGF2R expression in MSCs inhibited the increase of CCA caused by MSCs in HCC. Meanwhile, it was revealed that MSCs released multiple inflammatory and trophic-related cytokines to enhance the cancer stem cell-like characteristics in HCC cells. Finally, we demonstrated that CEBPβ up-regulated IGF2 expression in tumor senescent ECs by combining with Igf2-promtor-sequence. CONCLUSIONS Together, our findings illustrated that tumor associated senescent ECs in HCC recruited the MSCs into TME, enhancing cancer stem cell (CSC)-like features of HCC cells and contributing to the CCA formation.
Collapse
Affiliation(s)
- Xin-Yu Zhu
- Changhai Clinical Research Unit, Changhai Hospital of Naval Medical University, Shanghai, China; Tumor Immunology and Metabolism Center, National Center for Liver Cancer, Naval Medical University, Shanghai, China
| | - Wen-Ting Liu
- Tumor Immunology and Metabolism Center, National Center for Liver Cancer, Naval Medical University, Shanghai, China; Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Xiao-Juan Hou
- Tumor Immunology and Metabolism Center, National Center for Liver Cancer, Naval Medical University, Shanghai, China
| | - Chen Zong
- Tumor Immunology and Metabolism Center, National Center for Liver Cancer, Naval Medical University, Shanghai, China
| | - Wei Yu
- Changhai Clinical Research Unit, Changhai Hospital of Naval Medical University, Shanghai, China; Tumor Immunology and Metabolism Center, National Center for Liver Cancer, Naval Medical University, Shanghai, China
| | - Zhe-Min Shen
- Tumor Immunology and Metabolism Center, National Center for Liver Cancer, Naval Medical University, Shanghai, China
| | - Shu-Ping Qu
- Department of Hepatic Surgery, Third Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Min Tao
- Changhai Clinical Research Unit, Changhai Hospital of Naval Medical University, Shanghai, China; Department of Clinical Pharmacology, The Second Hospital of Anhui Medical University, Hefei, China
| | - Meng-Meng Xue
- Changhai Clinical Research Unit, Changhai Hospital of Naval Medical University, Shanghai, China; Department of Clinical Pharmacology, The Second Hospital of Anhui Medical University, Hefei, China
| | - Dao-Yu Zhou
- Changhai Clinical Research Unit, Changhai Hospital of Naval Medical University, Shanghai, China; Tumor Immunology and Metabolism Center, National Center for Liver Cancer, Naval Medical University, Shanghai, China
| | - Hao-Ran Bai
- Department of Oncology, Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Lu Gao
- Tumor Immunology and Metabolism Center, National Center for Liver Cancer, Naval Medical University, Shanghai, China
| | - Jing-Hua Jiang
- Tumor Immunology and Metabolism Center, National Center for Liver Cancer, Naval Medical University, Shanghai, China
| | - Qiu-Dong Zhao
- Tumor Immunology and Metabolism Center, National Center for Liver Cancer, Naval Medical University, Shanghai, China
| | - Li-Xin Wei
- Tumor Immunology and Metabolism Center, National Center for Liver Cancer, Naval Medical University, Shanghai, China
| | - Xue Yang
- Tumor Immunology and Metabolism Center, National Center for Liver Cancer, Naval Medical University, Shanghai, China.
| | - Zhi-Peng Han
- Tumor Immunology and Metabolism Center, National Center for Liver Cancer, Naval Medical University, Shanghai, China; Department of Oncology, Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| | - Li Zhang
- Changhai Clinical Research Unit, Changhai Hospital of Naval Medical University, Shanghai, China.
| |
Collapse
|
24
|
Shang Y, Wang Z, Xi L, Wang Y, Liu M, Feng Y, Wang J, Wu Q, Xiang X, Chen M, Ding Y. Droplet-based single-cell sequencing: Strategies and applications. Biotechnol Adv 2024; 77:108454. [PMID: 39271031 DOI: 10.1016/j.biotechadv.2024.108454] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Revised: 08/22/2024] [Accepted: 09/10/2024] [Indexed: 09/15/2024]
Abstract
Notable advancements in single-cell omics technologies have not only addressed longstanding challenges but also enabled unprecedented studies of cellular heterogeneity with unprecedented resolution and scale. These strides have led to groundbreaking insights into complex biological systems, paving the way for a more profound comprehension of human biology and diseases. The droplet microfluidic technology has become a crucial component in many single-cell sequencing workflows in terms of throughput, cost-effectiveness, and automation. Utilizing a microfluidic chip to encapsulate and profile individual cells within droplets has significantly improved single-cell research. Therefore, this review aims to comprehensively elaborate the droplet microfluidics-assisted omics methods from a single-cell perspective. The strategies for using droplet microfluidics in the realms of genomics, epigenomics, transcriptomics, and proteomics analyses are first introduced. On this basis, the focus then turns to the latest applications of this technology in different sequencing patterns, including mono- and multi-omics. Finally, the challenges and further perspectives of droplet-based single-cell sequencing in both foundational research and commercial applications are discussed.
Collapse
Affiliation(s)
- Yuting Shang
- Department of Food Science & Engineering, College of Life Science and Technology, Jinan University, Guangzhou 510632, China
| | - Zhengzheng Wang
- Department of Food Science & Engineering, College of Life Science and Technology, Jinan University, Guangzhou 510632, China
| | - Liqing Xi
- Department of Food Science & Engineering, College of Life Science and Technology, Jinan University, Guangzhou 510632, China
| | - Yantao Wang
- Department of Food Science & Engineering, College of Life Science and Technology, Jinan University, Guangzhou 510632, China
| | - Meijing Liu
- Department of Food Science & Engineering, College of Life Science and Technology, Jinan University, Guangzhou 510632, China
| | - Ying Feng
- Department of Food Science & Engineering, College of Life Science and Technology, Jinan University, Guangzhou 510632, China
| | - Juan Wang
- College of Food Science, South China Agricultural University, Guangzhou 510432, China
| | - Qingping Wu
- National Health Commission Science and Technology Innovation Platform for Nutrition and Safety of Microbial Food, Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou 510070, China
| | - Xinran Xiang
- Jiangsu Key Laboratory of Huaiyang Food Safety and Nutrition Function Evaluation, Jiangsu Collaborative Innovation Center of Regional Modern Agriculture & Environmental Protection, Jiangsu Key Laboratory for Eco-Agricultural Biotechnology Around Hongze Lake, School of Life Science, Huaiyin Normal University, Huai'an 223300, China; Fujian Key Laboratory of Aptamers Technology, Fuzhou General Clinical Medical School (the 900th Hospital), Fujian Medical University, Fuzhou 350001, China.
| | - Moutong Chen
- National Health Commission Science and Technology Innovation Platform for Nutrition and Safety of Microbial Food, Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou 510070, China.
| | - Yu Ding
- Department of Food Science & Engineering, College of Life Science and Technology, Jinan University, Guangzhou 510632, China.
| |
Collapse
|
25
|
Hu R, Yang W, Li J, Jiang L, Li M, Zhang M, Kang Y, Cheng X, Zhu S, Zhao L, He W, Guo M, Ding S, Wu H, Cheng W. Multiple RNA Rapid In Situ Imaging Based on Cas9 Code Key System. SMALL METHODS 2024; 8:e2400195. [PMID: 38699929 DOI: 10.1002/smtd.202400195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Revised: 03/29/2024] [Indexed: 05/05/2024]
Abstract
Existing RNA in situ imaging strategies mostly utilize parallel repetitive nucleic acid self-assembly to achieve multiple analysis, with limitations of complicated systems and cumbersome steps. Here, a Cas9 code key system with key probe (KP) encoder and CRISPR/Cas9 signal exporter is developed. This system triggers T-protospacer adjacent motif (T-PAM structural transitions of multiple KP encoders to form coding products with uniform single-guide RNA (sgRNA) target sequences as tandem nodes. Only single sgRNA/Cas9 complex is required to cleave multiple coding products, enabling efficient "many-to-one" tandem signaling, and non-collateral cleavage activity-dependent automatic signaling output through active introduction of mismatched bases. Compared with conventional parallel multiple signaling analysis model, the proposed system greatly simplifies reaction process and enhances detection efficiency. Further, a rapid multiple RNA in situ imaging system is developed by combining the Cas9 code key system with a T-strand displacement amplification (T-SDA) signal amplifier. The constructed system is applied to tumor cells and clinicopathology slices, generating clear multi-mRNA imaging profiles in less than an hour with just one step. Therefore, this work provides reliable technical support for clinical tumor typing and molecular mechanism investigation.
Collapse
Affiliation(s)
- Ruiwei Hu
- The Center for Clinical Molecular Medical Detection, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400042, China
- Key Laboratory of Clinical Laboratory Diagnostics (Ministry of Education), College of Laboratory Medicine, Chongqing Medical University, Chongqing, 400016, China
| | - Wei Yang
- Laboratory Medicine Center, Department of Clinical Laboratory, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, 310014, China
| | - Jia Li
- The Center for Clinical Molecular Medical Detection, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400042, China
| | - Lanxin Jiang
- The Center for Clinical Molecular Medical Detection, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400042, China
| | - Menghan Li
- Key Laboratory of Clinical Laboratory Diagnostics (Ministry of Education), College of Laboratory Medicine, Chongqing Medical University, Chongqing, 400016, China
| | - Mengxuan Zhang
- Key Laboratory of Clinical Laboratory Diagnostics (Ministry of Education), College of Laboratory Medicine, Chongqing Medical University, Chongqing, 400016, China
| | - Yuexi Kang
- The Center for Clinical Molecular Medical Detection, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400042, China
| | - Xiaoxue Cheng
- The Center for Clinical Molecular Medical Detection, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400042, China
| | - Shasha Zhu
- The Center for Clinical Molecular Medical Detection, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400042, China
| | - Lina Zhao
- Key Laboratory of Clinical Laboratory Diagnostics (Ministry of Education), College of Laboratory Medicine, Chongqing Medical University, Chongqing, 400016, China
| | - Wen He
- Key Laboratory of Clinical Laboratory Diagnostics (Ministry of Education), College of Laboratory Medicine, Chongqing Medical University, Chongqing, 400016, China
| | - Minghui Guo
- Key Laboratory of Clinical Laboratory Diagnostics (Ministry of Education), College of Laboratory Medicine, Chongqing Medical University, Chongqing, 400016, China
| | - Shijia Ding
- Key Laboratory of Clinical Laboratory Diagnostics (Ministry of Education), College of Laboratory Medicine, Chongqing Medical University, Chongqing, 400016, China
| | - Haiping Wu
- Key Laboratory of Clinical Laboratory Diagnostics (Ministry of Education), College of Laboratory Medicine, Chongqing Medical University, Chongqing, 400016, China
| | - Wei Cheng
- The Center for Clinical Molecular Medical Detection, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400042, China
- Key Laboratory of Clinical Laboratory Diagnostics (Ministry of Education), College of Laboratory Medicine, Chongqing Medical University, Chongqing, 400016, China
| |
Collapse
|
26
|
Su W, Ling Y, Yang X, Wu Y, Xing C. Tumor microenvironment remodeling after neoadjuvant chemoradiotherapy in local advanced rectal cancer revealed by single-cell RNA sequencing. J Transl Med 2024; 22:1037. [PMID: 39558398 PMCID: PMC11575152 DOI: 10.1186/s12967-024-05747-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: 04/10/2024] [Accepted: 10/08/2024] [Indexed: 11/20/2024] Open
Abstract
BACKGROUND The use of neoadjuvant chemoradiotherapy (neoCRT) followed by surgery has markedly enhanced the quality of survival in patients suffering from local advanced rectal cancer (LARC). Enhancing this treatment requires a deep understanding of its underlying mechanism. The heterogeneous nature of the tumor microenvironment (TME) significantly impacts therapeutic responses, presenting complex therapeutic challenges. METHODS In this comprehensive study, we explored the intricate cellular and molecular shifts within the TME of LARC after neoCRT administration. Using single-cell transcriptomic analysis, we meticulously examined 32,417 cells sourced from six samples, each representing different tumor regression grades (TRG: 0 versus 2). This detailed analysis enabled us to characterize the various cell subpopulations, encompassing epithelial cells, lymphocytes, myeloid cells, endothelial cells, and fibroblasts. Additionally, we identified their marker genes for deconvolution calculation in the READ cohort of the TCGA project. And we obtain their marker genes for deconvolution calculation in the READ cohort of the TCGA project. RESULTS Through cluster analysis and pathway comparisons of malignant tumor cells, we discerned that samples with poor tumor regression exhibit enhanced metabolic versatility and adaptability, enabling them to counteract the impacts of both radiotherapy and chemotherapy. Interestingly, within the TRG2 cohort, we observed a predominant immunosuppressive state in the TME, characterized by the activation of CD4 + regulatory T cells, maintained CD8 + T cell functionality, and a heightened M1 to M2 macrophage ratio. Moreover, the differing outcomes of neoCRT were reflected in the varying interaction dynamics between macrophages (M1 and M2) and CD4+/CD8 + T cells. Furthermore, our data reveal that neoCRT intricately modulates fibroblasts and endothelial cells, primarily through the extracellular matrix remodeling pathway, which orchestrates tumor angiogenesis. All changes were validated through immunofluorescence staining on intraoperative samples before and after treatment. To summarize, our investigation presents a comprehensive exploration of the cellular and molecular metamorphoses within the TME post-neoCRT. CONCLUSIONS By unveiling the sophisticated interaction between the multifaceted cells within the TME and their respective reactions to neoCRT, we establish a robust platform for ensuing future investigations. This study paves the way for novel therapeutic strategies that leverage these insights to bolster the efficacy of neoCRT in managing LARC.
Collapse
Affiliation(s)
- Wenzhao Su
- Department of Gastroenterology, the Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, Sanxiang Road 1055, Suzhou, CN, 215000, China
| | - Yuhang Ling
- Huzhou Key Laboratory of Translational Medicine, First People's Hospital of Huzhou, Huzhou, Zhejiang Province, CN, 313000, China
| | - Xiaodong Yang
- Department of Gastroenterology, the Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, Sanxiang Road 1055, Suzhou, CN, 215000, China
| | - Yong Wu
- Department of Gastroenterology, the Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, Sanxiang Road 1055, Suzhou, CN, 215000, China.
| | - Chungen Xing
- Department of Gastroenterology, the Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, Sanxiang Road 1055, Suzhou, CN, 215000, China.
| |
Collapse
|
27
|
Ma Y, Yi C, Cai N, Chen J. Integration of single-cell and spatial transcriptome sequencing identifies CDKN2A as a senescent biomarker in endothelial cells implicating hepatocellular carcinoma malignancy. J Cancer Res Clin Oncol 2024; 150:487. [PMID: 39503880 PMCID: PMC11541268 DOI: 10.1007/s00432-024-06017-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2024] [Accepted: 10/25/2024] [Indexed: 11/09/2024]
Abstract
PURPOSE Highly complex tumor microenvironment makes hepatocellular carcinoma (HCC) as one of the most malignant tumors worldwide. The role of cellular senescence in HCC has been gradually recognized. The present study aimed to comprehensively elucidate the senescence-related features of HCC in single-cell and spatial dimension. METHODS Single-cell RNA sequencing (scRNA-Seq) data was used to clarify the heterogeneity of senescence-related genes (SRGs) among multiple cell types within HCC. Spatial transcriptome RNA sequencing (stRNA-Seq) data was used for depicting SRGs features in spatial dimension. A prognostic model based on SRGs was constructed by using of bulk sequencing (bulk-Seq) data of HCC. The cell-cell interaction of senescent endothelial cells (ECs) in tumor microenvironment was analyzed. Then, the role of senescent ECs was verified through in vitro and in vivo experiments. RESULTS The level of senescence demonstrated substantial heterogeneity among different cell types within tumor microenvironment of HCC, where ECs exhibited the most prominent senescent phenotype. Senescent ECs activated specific regulatory pathways through communicating with other cell types, with a potential impact on tumor progression. Spatial analysis revealed senescent ECs mainly located in the core region of HCC. The interaction of senescent ECs and immune cells implicated their role in tumor progression and immunotherapeutic response. In addition, CDKN2A was identified as an independent risk factor for HCC prognosis by constructing a prognostic model. Patients with high risk displayed an even worse outcome. The experimental verification indicated senescence of ECs determined by CDKN2A exhibited a secretory phenotype. Furthermore, senescent ECs with CDKN2A overexpression promote the proliferation and migration of HCC. CONCLUSION The present study recognizes the critical effect of senescent ECs defined by CDKN2A in the promotion of tumor progression, which sheds new light on the investigation of ECs senescence in HCC.
Collapse
Affiliation(s)
- Yue Ma
- Hepatobiliary Surgery, Department of General Surgery, Huashan Hospital & Cancer Metastasis Institute, Fudan University, Shanghai, 200040, P.R. China
| | - Chenhe Yi
- Hepatobiliary Surgery, Department of General Surgery, Huashan Hospital & Cancer Metastasis Institute, Fudan University, Shanghai, 200040, P.R. China
| | - Ning Cai
- Hepatic Surgery Centre, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, P.R. China
| | - Jinhong Chen
- Hepatobiliary Surgery, Department of General Surgery, Huashan Hospital & Cancer Metastasis Institute, Fudan University, Shanghai, 200040, P.R. China.
| |
Collapse
|
28
|
Skandha Gopalan K, Bergers G. The Pan-Tumor Vasculature under the Transcriptomic Magnifying Glass. Cancer Res 2024; 84:3502-3504. [PMID: 39240688 DOI: 10.1158/0008-5472.can-24-3219] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Accepted: 09/05/2024] [Indexed: 09/08/2024]
Abstract
In the first pan-cancer analysis of the tumor vasculature, Pan and colleagues profile nearly 200,000 endothelial cells (EC) and mural cells, identifying novel subclusters and cell states using consensus trajectory inference. They identify differentiation trajectories in vascular and lymphatic ECs and subtype the pericyte (PC) population. During sprouting angiogenesis, venous cells dedifferentiate and transition to capillary and, finally, arterial ECs. Capillary ECs transition via "three angiogenic stages" (SI-SIII), during which APLN+ TipS1 cells were identified as potential modulators of tumor-induced neovascularization and antiangiogenic therapy response. In lymphatic ECs, differentiation was inversely correlated between the lymphangiogenic (T1) and antigen-presenting (T2) trajectories, with T2 associated with a better prognosis. Although several PC clusters were identified, BASP1+ matrix-associated PCs were associated with APLN+ TipS1 cells and had a worse prognosis. These findings present transcriptional validation of previous experimental findings and serve as a resource to examine the tumor vascular microenvironment in detail.
Collapse
Affiliation(s)
- Krish Skandha Gopalan
- Department of Oncology, VIB-KU Leuven Center for Cancer Biology, and KU Leuven, Leuven, Belgium
| | - Gabriele Bergers
- Department of Oncology, VIB-KU Leuven Center for Cancer Biology, and KU Leuven, Leuven, Belgium
| |
Collapse
|
29
|
Tosato G, Wang Y. Celebrating the 1945 JNCI pioneering contribution to antiangiogenic therapy for cancer. J Natl Cancer Inst 2024; 116:1715-1720. [PMID: 39178374 DOI: 10.1093/jnci/djae181] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Revised: 07/25/2024] [Accepted: 07/29/2024] [Indexed: 08/25/2024] Open
Affiliation(s)
- Giovanna Tosato
- Laboratory of Cellular Oncology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Yuyi Wang
- Laboratory of Cellular Oncology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
30
|
Alnaqbi H, Becker LM, Mousa M, Alshamsi F, Azzam SK, Emini Veseli B, Hymel LA, Alhosani K, Alhusain M, Mazzone M, Alsafar H, Carmeliet P. Immunomodulation by endothelial cells: prospects for cancer therapy. Trends Cancer 2024; 10:1072-1091. [PMID: 39289084 DOI: 10.1016/j.trecan.2024.08.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Revised: 07/26/2024] [Accepted: 08/09/2024] [Indexed: 09/19/2024]
Abstract
Growing evidence highlights the importance of tumor endothelial cells (TECs) in the tumor microenvironment (TME) for promoting tumor growth and evading immune responses. Immunomodulatory endothelial cells (IMECs) represent a distinct plastic phenotype of ECs that exerts the ability to modulate immunity in health and disease. This review discusses our current understanding of IMECs in cancer biology, scrutinizing insights from single-cell reports to compare their characteristics and function dynamics across diverse tumor types, conditions, and species. We investigate possible implications of exploiting IMECs in the context of cancer treatment, particularly examining their influence on the efficacy of existing therapies and the potential to leverage them as targets in optimizing immunotherapeutic strategies.
Collapse
Affiliation(s)
- Halima Alnaqbi
- Center for Biotechnology, Khalifa University, Abu Dhabi, United Arab Emirates; Department of Biomedical Engineering and Biotechnology, Khalifa University, Abu Dhabi, United Arab Emirates
| | - Lisa M Becker
- Laboratory of Angiogenesis and Vascular Metabolism, Center for Cancer Biology, VIB, Department of Oncology, University of Leuven, Leuven, Belgium
| | - Mira Mousa
- Center for Biotechnology, Khalifa University, Abu Dhabi, United Arab Emirates; Department of Public Health and Epidemiology, Khalifa University, Abu Dhabi, United Arab Emirates
| | - Fatima Alshamsi
- Center for Biotechnology, Khalifa University, Abu Dhabi, United Arab Emirates; Laboratory of Angiogenesis and Vascular Metabolism, Center for Cancer Biology, VIB, Department of Oncology, University of Leuven, Leuven, Belgium
| | - Sarah K Azzam
- Center for Biotechnology, Khalifa University, Abu Dhabi, United Arab Emirates
| | - Besa Emini Veseli
- Laboratory of Angiogenesis and Vascular Metabolism, Center for Cancer Biology, VIB, Department of Oncology, University of Leuven, Leuven, Belgium
| | - Lauren A Hymel
- Laboratory of Angiogenesis and Vascular Metabolism, Center for Cancer Biology, VIB, Department of Oncology, University of Leuven, Leuven, Belgium
| | - Khalood Alhosani
- Department of Biological Sciences, College of Medicine and Health Sciences, Khalifa University, Abu Dhabi, United Arab Emirates
| | - Marwa Alhusain
- Department of Biological Sciences, College of Medicine and Health Sciences, Khalifa University, Abu Dhabi, United Arab Emirates
| | - Massimiliano Mazzone
- Laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer Biology, VIB, Leuven, Belgium; Laboratory of Tumor Inflammation and Angiogenesis, Department of Oncology, KU Leuven, Leuven, Belgium.
| | - Habiba Alsafar
- Center for Biotechnology, Khalifa University, Abu Dhabi, United Arab Emirates; Department of Biomedical Engineering and Biotechnology, Khalifa University, Abu Dhabi, United Arab Emirates
| | - Peter Carmeliet
- Center for Biotechnology, Khalifa University, Abu Dhabi, United Arab Emirates; Laboratory of Angiogenesis and Vascular Metabolism, Center for Cancer Biology, VIB, Department of Oncology, University of Leuven, Leuven, Belgium.
| |
Collapse
|
31
|
Wu X, Li B, Wang Y, Xue J, Zhao H, Huang Z, Zheng Z, Liang N, Wei Z. Microfluidic Chip-Based Automatic System for Sequencing Patient-Derived Organoids at the Single-Cell Level. Anal Chem 2024; 96:17027-17036. [PMID: 39399894 DOI: 10.1021/acs.analchem.4c05111] [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: 10/15/2024]
Abstract
Genetically sequencing patient-derived organoids (PDOs) at the single-cell level has emerged as a promising method to infer cell-level heterogeneity of original organs and improve cancer precision medicine. Unfortunately, because of the limited starting quantity and uncontrolled establishing process of PDOs, the existing single-cell sequencing technologies, either manual-operation-based or microfluid-based, are inefficient in processing PDOs originating from clinical tissue samples. To address such issues, this study presents a microfluidic chip-based automatic system for sequencing organoids at the single-cell level, named as MASSO. By performing all required procedures, including PDO establishment/culturing/digesting and single-cell isolation/lysis/whole-genome amplification, in a single microfluidic chip, the possible loss of precious PDO is avoided, and the high quality of on-chip whole-genome amplification of a single PDO cell is ensured. By automating the entire operation process, possible human error is eliminated, and the data repeatability is improved, therefore bridging the technical gap between laboratorial proof-of-concept studies and clinical practices. After characterizing the organoid single-cell whole-genome amplification chip (named as OSA-Chip) and the MASSO, the first successful attempt, to the best of our knowledge, on whole-genome sequencing lung cancer PDO at the single-cell level was performed by MASSO. The results reveal that the MASSO is capable of not only identifying common cancer-related mutations but also discovering specific mutations that affect drug responses, therefore laying the technical foundation for efficiently understanding the cell-level heterogeneities of PDOs and corresponding original organs.
Collapse
Affiliation(s)
- Xin Wu
- Department of Biomedical Engineering, School of Medical Technology, Beijing Institute of Technology, Beijing 100081, China
| | - Bowen Li
- Department of Thoracic Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
- Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| | - Yadong Wang
- Department of Thoracic Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
- Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| | - Jianchao Xue
- Department of Thoracic Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
- Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| | - Huiting Zhao
- Department of Biomedical Engineering, School of Medical Technology, Beijing Institute of Technology, Beijing 100081, China
| | - Zhicheng Huang
- Department of Thoracic Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
- Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| | - Zhibo Zheng
- Department of Thoracic Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| | - Naixin Liang
- Department of Thoracic Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| | - Zewen Wei
- Department of Biomedical Engineering, School of Medical Technology, Beijing Institute of Technology, Beijing 100081, China
| |
Collapse
|
32
|
Jiang H, Gao B, Meng Z, Wang Y, Jiao T, Li J, Li X, Cao Y, Zhang X, Li C, Lu S. Integrative multi-omics analysis reveals the role of tumor-associated endothelial cells and their signature in prognosis of intrahepatic cholangiocarcinoma. J Transl Med 2024; 22:948. [PMID: 39427165 PMCID: PMC11490089 DOI: 10.1186/s12967-024-05750-2] [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: 07/11/2024] [Accepted: 10/08/2024] [Indexed: 10/21/2024] Open
Abstract
This study aims to investigate the interplay between tumor-associated endothelial cells (TECs) and immune cells within the tumor microenvironment (TME) and its impact on tumor prognosis. We conducted single-cell RNA sequencing (scRNA-seq) of tumor, normal, and lymph node tissues obtained from intrahepatic cholangiocarcinoma (ICC) patients to reveal the role of TECs in tumor angiogenesis and their significant heterogeneity. Meanwhile, we identified genes highly expressed in TECs and constructed TEC signatures (TEC.Sig). Next, we calculated TEC scores of samples based on TEC.Sig. Patients with higher TEC scores exhibited a higher frequency of KRAS mutations, which was associated with increased infiltration of neutrophils and immature dendritic cells (iDCs), and decreased numbers of natural killer (NK), CD4 + T, and CD8 + T effector memory (Tem) cells, indicating an inflammation-dominated immunosuppressive phenotype. In contrast, BAP1 mutations and CXCL12 overexpression showed a contrasting trend. Spatial transcriptomics analysis and histological experiments further confirmed that TECs interacted with various tumor-killing immune cells through the CXCL12/CXCR4 axis. Multiple tumor immunotherapy datasets confirmed that the TEC.Sig could predict patient responses to immunotherapy. The TEC score is a promising and reliable biomarker for predicting genetic mutations and prognosis in ICC patients. Enhancing the regulation of the CXCL12/CXCR4 signaling pathway may represent a potential novel therapeutic target for ICC treatment.
Collapse
Affiliation(s)
- Hao Jiang
- Medical School of Chinese People's Liberation Army (PLA), Beijing, China
- Faculty of Hepato-Pancreato-Biliary Surgery, The First Medical Center of Chinese PLA General Hospital, Beijing, China
- Key Laboratory of Digital Hepatobiliary Surgery of Chinese PLA, Beijing, China
| | - Biao Gao
- Medical School of Chinese People's Liberation Army (PLA), Beijing, China
- Faculty of Hepato-Pancreato-Biliary Surgery, The First Medical Center of Chinese PLA General Hospital, Beijing, China
- Key Laboratory of Digital Hepatobiliary Surgery of Chinese PLA, Beijing, China
- Nankai University School of Medicine, Nankai University, Tianjin, China
| | - Zihe Meng
- Key Laboratory of Digital Hepatobiliary Surgery of Chinese PLA, Beijing, China
- College of Basic Medical Science, Inner Mongolia Medical University, Hohhot, Inner Mongolia Autonomous Region, China
| | - Yafei Wang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Henan University of Science and Technology, Luoyang, China
| | - Tianyu Jiao
- Medical School of Chinese People's Liberation Army (PLA), Beijing, China
- Faculty of Hepato-Pancreato-Biliary Surgery, The First Medical Center of Chinese PLA General Hospital, Beijing, China
- Key Laboratory of Digital Hepatobiliary Surgery of Chinese PLA, Beijing, China
| | - Junfeng Li
- Medical School of Chinese People's Liberation Army (PLA), Beijing, China
- Faculty of Hepato-Pancreato-Biliary Surgery, The First Medical Center of Chinese PLA General Hospital, Beijing, China
- Key Laboratory of Digital Hepatobiliary Surgery of Chinese PLA, Beijing, China
| | - Xuerui Li
- Medical School of Chinese People's Liberation Army (PLA), Beijing, China
- Faculty of Hepato-Pancreato-Biliary Surgery, The First Medical Center of Chinese PLA General Hospital, Beijing, China
- Key Laboratory of Digital Hepatobiliary Surgery of Chinese PLA, Beijing, China
- Nankai University School of Medicine, Nankai University, Tianjin, China
| | - Yinbiao Cao
- Medical School of Chinese People's Liberation Army (PLA), Beijing, China
- Faculty of Hepato-Pancreato-Biliary Surgery, The First Medical Center of Chinese PLA General Hospital, Beijing, China
- Key Laboratory of Digital Hepatobiliary Surgery of Chinese PLA, Beijing, China
| | - Xianzhou Zhang
- Department of Hepatic Biliary Pancreatic Surgery, Cancer Hospital Affiliated to Zhengzhou University, Zhengzhou, 450000, Henan, China.
| | - Chonghui Li
- Medical School of Chinese People's Liberation Army (PLA), Beijing, China.
- Faculty of Hepato-Pancreato-Biliary Surgery, The First Medical Center of Chinese PLA General Hospital, Beijing, China.
- Key Laboratory of Digital Hepatobiliary Surgery of Chinese PLA, Beijing, China.
| | - Shichun Lu
- Medical School of Chinese People's Liberation Army (PLA), Beijing, China.
- Faculty of Hepato-Pancreato-Biliary Surgery, The First Medical Center of Chinese PLA General Hospital, Beijing, China.
- Key Laboratory of Digital Hepatobiliary Surgery of Chinese PLA, Beijing, China.
| |
Collapse
|
33
|
Guelfi S, Hodivala-Dilke K, Bergers G. Targeting the tumour vasculature: from vessel destruction to promotion. Nat Rev Cancer 2024; 24:655-675. [PMID: 39210063 DOI: 10.1038/s41568-024-00736-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/25/2024] [Indexed: 09/04/2024]
Abstract
As angiogenesis was recognized as a core hallmark of cancer growth and survival, several strategies have been implemented to target the tumour vasculature. Yet to date, attempts have rarely been so diverse, ranging from vessel growth inhibition and destruction to vessel normalization, reprogramming and vessel growth promotion. Some of these strategies, combined with standard of care, have translated into improved cancer therapies, but their successes are constrained to certain cancer types. This Review provides an overview of these vascular targeting approaches and puts them into context based on our subsequent improved understanding of the tumour vasculature as an integral part of the tumour microenvironment with which it is functionally interlinked. This new knowledge has already led to dual targeting of the vascular and immune cell compartments and sets the scene for future investigations of possible alternative approaches that consider the vascular link with other tumour microenvironment components for improved cancer therapy.
Collapse
Affiliation(s)
- Sophie Guelfi
- Department of Oncology, VIB-KU Leuven Center for Cancer Biology and KU Leuven, Leuven, Belgium
| | - Kairbaan Hodivala-Dilke
- Barts Cancer Institute, Queen Mary University of London, John Vane Science Centre, London, UK.
| | - Gabriele Bergers
- Department of Oncology, VIB-KU Leuven Center for Cancer Biology and KU Leuven, Leuven, Belgium.
| |
Collapse
|
34
|
Cheng X, Cao Y, Liu X, Li Y, Li Q, Gao D, Yu Q. Single-cell and spatial omics unravel the spatiotemporal biology of tumour border invasion and haematogenous metastasis. Clin Transl Med 2024; 14:e70036. [PMID: 39350478 PMCID: PMC11442492 DOI: 10.1002/ctm2.70036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2024] [Revised: 08/14/2024] [Accepted: 09/16/2024] [Indexed: 10/04/2024] Open
Abstract
Solid tumours exhibit a well-defined architecture, comprising a differentiated core and a dynamic border that interfaces with the surrounding tissue. This border, characterised by distinct cellular morphology and molecular composition, serves as a critical determinant of the tumour's invasive behaviour. Notably, the invasive border of the primary tumour represents the principal site for intravasation of metastatic cells. These cells, known as circulating tumour cells (CTCs), function as 'seeds' for distant dissemination and display remarkable heterogeneity. Advancements in spatial sequencing technology are progressively unveiling the spatial biological features of tumours. However, systematic investigations specifically targeting the characteristics of the tumour border remain scarce. In this comprehensive review, we illuminate key biological insights along the tumour body-border-haematogenous metastasis axis over the past five years. We delineate the distinctive landscape of tumour invasion boundaries and delve into the intricate heterogeneity and phenotype of CTCs, which orchestrate haematogenous metastasis. These insights have the potential to explain the basis of tumour invasion and distant metastasis, offering new perspectives for the development of more complex and precise clinical interventions and treatments.
Collapse
Affiliation(s)
- Xifu Cheng
- Department of Gastroenterology and Hepatologythe Second Affiliated HospitalJiangxi Medical CollegeNanchang UniversityNanchangChina
- Department of Pathogen Biology and ImmunologySchool of Basic Medical SciencesJiangxi Medical CollegeNanchang UniversityNanchangChina
| | - Yuke Cao
- Department of Gastroenterology and Hepatologythe Second Affiliated HospitalJiangxi Medical CollegeNanchang UniversityNanchangChina
| | - Xiangyi Liu
- Queen Mary SchoolJiangxi Medical CollegeNanchang UniversityNanchangChina
| | - Yuanheng Li
- Queen Mary SchoolJiangxi Medical CollegeNanchang UniversityNanchangChina
| | - Qing Li
- Department of Oncologythe Second Affiliated HospitalJiangxi Medical CollegeNanchang UniversityNanchangChina
| | - Dian Gao
- Department of Gastroenterology and Hepatologythe Second Affiliated HospitalJiangxi Medical CollegeNanchang UniversityNanchangChina
- Department of Pathogen Biology and ImmunologySchool of Basic Medical SciencesJiangxi Medical CollegeNanchang UniversityNanchangChina
| | - Qiongfang Yu
- Department of Gastroenterology and Hepatologythe Second Affiliated HospitalJiangxi Medical CollegeNanchang UniversityNanchangChina
| |
Collapse
|
35
|
Jin X, Zhang R, Fu Y, Zhu Q, Hong L, Wu A, Wang H. Unveiling aging dynamics in the hematopoietic system insights from single-cell technologies. Brief Funct Genomics 2024; 23:639-650. [PMID: 38688725 DOI: 10.1093/bfgp/elae019] [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: 02/10/2024] [Revised: 04/10/2024] [Accepted: 04/15/2024] [Indexed: 05/02/2024] Open
Abstract
As the demographic structure shifts towards an aging society, strategies aimed at slowing down or reversing the aging process become increasingly essential. Aging is a major predisposing factor for many chronic diseases in humans. The hematopoietic system, comprising blood cells and their associated bone marrow microenvironment, intricately participates in hematopoiesis, coagulation, immune regulation and other physiological phenomena. The aging process triggers various alterations within the hematopoietic system, serving as a spectrum of risk factors for hematopoietic disorders, including clonal hematopoiesis, immune senescence, myeloproliferative neoplasms and leukemia. The emerging single-cell technologies provide novel insights into age-related changes in the hematopoietic system. In this review, we summarize recent studies dissecting hematopoietic system aging using single-cell technologies. We discuss cellular changes occurring during aging in the hematopoietic system at the levels of the genomics, transcriptomics, epigenomics, proteomics, metabolomics and spatial multi-omics. Finally, we contemplate the future prospects of single-cell technologies, emphasizing the impact they may bring to the field of hematopoietic system aging research.
Collapse
Affiliation(s)
- Xinrong Jin
- Zhejiang Key Laboratory of Medical Epigenetics, School of Basic Medical Sciences, The Third People's Hospital of Deqing, Deqing Hospital of Hangzhou Normal University, Hangzhou Normal University, Hangzhou 311121, China
| | - Ruohan Zhang
- Zhejiang Key Laboratory of Medical Epigenetics, School of Basic Medical Sciences, The Third People's Hospital of Deqing, Deqing Hospital of Hangzhou Normal University, Hangzhou Normal University, Hangzhou 311121, China
| | - Yunqi Fu
- Zhejiang Key Laboratory of Medical Epigenetics, School of Basic Medical Sciences, The Third People's Hospital of Deqing, Deqing Hospital of Hangzhou Normal University, Hangzhou Normal University, Hangzhou 311121, China
| | - Qiunan Zhu
- Zhejiang Key Laboratory of Medical Epigenetics, School of Basic Medical Sciences, The Third People's Hospital of Deqing, Deqing Hospital of Hangzhou Normal University, Hangzhou Normal University, Hangzhou 311121, China
| | - Liquan Hong
- Zhejiang Key Laboratory of Medical Epigenetics, School of Basic Medical Sciences, The Third People's Hospital of Deqing, Deqing Hospital of Hangzhou Normal University, Hangzhou Normal University, Hangzhou 311121, China
| | - Aiwei Wu
- Zhejiang Key Laboratory of Medical Epigenetics, School of Basic Medical Sciences, The Third People's Hospital of Deqing, Deqing Hospital of Hangzhou Normal University, Hangzhou Normal University, Hangzhou 311121, China
| | - Hu Wang
- Zhejiang Key Laboratory of Medical Epigenetics, School of Basic Medical Sciences, The Third People's Hospital of Deqing, Deqing Hospital of Hangzhou Normal University, Hangzhou Normal University, Hangzhou 311121, China
| |
Collapse
|
36
|
Xiong X, Wang X, Liu CC, Shao ZM, Yu KD. Deciphering breast cancer dynamics: insights from single-cell and spatial profiling in the multi-omics era. Biomark Res 2024; 12:107. [PMID: 39294728 PMCID: PMC11411917 DOI: 10.1186/s40364-024-00654-1] [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: 06/28/2024] [Accepted: 09/10/2024] [Indexed: 09/21/2024] Open
Abstract
As one of the most common tumors in women, the pathogenesis and tumor heterogeneity of breast cancer have long been the focal point of research, with the emergence of tumor metastasis and drug resistance posing persistent clinical challenges. The emergence of single-cell sequencing (SCS) technology has introduced novel approaches for gaining comprehensive insights into the biological behavior of malignant tumors. SCS is a high-throughput technology that has rapidly developed in the past decade, providing high-throughput molecular insights at the individual cell level. Furthermore, the advent of multitemporal point sampling and spatial omics also greatly enhances our understanding of cellular dynamics at both temporal and spatial levels. The paper provides a comprehensive overview of the historical development of SCS, and highlights the most recent advancements in utilizing SCS and spatial omics for breast cancer research. The findings from these studies will serve as valuable references for future advancements in basic research, clinical diagnosis, and treatment of breast cancer.
Collapse
Affiliation(s)
- Xin Xiong
- Department of Breast Surgery, Key Laboratory of Breast Cancer in Shanghai, Cancer Institute, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Xin Wang
- Department of Anesthesiology, Fudan University Shanghai Cancer Center, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Cui-Cui Liu
- Department of Breast Surgery, Key Laboratory of Breast Cancer in Shanghai, Cancer Institute, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Zhi-Ming Shao
- Department of Breast Surgery, Key Laboratory of Breast Cancer in Shanghai, Cancer Institute, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Ke-Da Yu
- Department of Breast Surgery, Key Laboratory of Breast Cancer in Shanghai, Cancer Institute, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
37
|
Liang W, Zhu Z, Xu D, Wang P, Guo F, Xiao H, Hou C, Xue J, Zhi X, Ran R. The burgeoning spatial multi-omics in human gastrointestinal cancers. PeerJ 2024; 12:e17860. [PMID: 39285924 PMCID: PMC11404479 DOI: 10.7717/peerj.17860] [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: 03/27/2024] [Accepted: 07/14/2024] [Indexed: 09/19/2024] Open
Abstract
The development and progression of diseases in multicellular organisms unfold within the intricate three-dimensional body environment. Thus, to comprehensively understand the molecular mechanisms governing individual development and disease progression, precise acquisition of biological data, including genome, transcriptome, proteome, metabolome, and epigenome, with single-cell resolution and spatial information within the body's three-dimensional context, is essential. This foundational information serves as the basis for deciphering cellular and molecular mechanisms. Although single-cell multi-omics technology can provide biological information such as genome, transcriptome, proteome, metabolome, and epigenome with single-cell resolution, the sample preparation process leads to the loss of spatial information. Spatial multi-omics technology, however, facilitates the characterization of biological data, such as genome, transcriptome, proteome, metabolome, and epigenome in tissue samples, while retaining their spatial context. Consequently, these techniques significantly enhance our understanding of individual development and disease pathology. Currently, spatial multi-omics technology has played a vital role in elucidating various processes in tumor biology, including tumor occurrence, development, and metastasis, particularly in the realms of tumor immunity and the heterogeneity of the tumor microenvironment. Therefore, this article provides a comprehensive overview of spatial transcriptomics, spatial proteomics, and spatial metabolomics-related technologies and their application in research concerning esophageal cancer, gastric cancer, and colorectal cancer. The objective is to foster the research and implementation of spatial multi-omics technology in digestive tumor diseases. This review will provide new technical insights for molecular biology researchers.
Collapse
Affiliation(s)
- Weizheng Liang
- Central Laboratory, The First Affiliated Hospital of Hebei North University, Zhangjiakou, Hebei province, China
| | - Zhenpeng Zhu
- Department of Surgery, The First Affiliated Hospital of Hebei North University, Zhangjiakou, Hebei Province, China
- Hebei North University, Zhangjiakou, Hebei Province, China
| | - Dandan Xu
- Central Laboratory, The First Affiliated Hospital of Hebei North University, Zhangjiakou, Hebei province, China
| | - Peng Wang
- Department of Surgery, The First Affiliated Hospital of Hebei North University, Zhangjiakou, Hebei Province, China
- Hebei North University, Zhangjiakou, Hebei Province, China
| | - Fei Guo
- Department of Surgery, The First Affiliated Hospital of Hebei North University, Zhangjiakou, Hebei Province, China
| | - Haoshan Xiao
- Department of Surgery, The First Affiliated Hospital of Hebei North University, Zhangjiakou, Hebei Province, China
- Hebei North University, Zhangjiakou, Hebei Province, China
| | - Chenyang Hou
- Department of Surgery, The First Affiliated Hospital of Hebei North University, Zhangjiakou, Hebei Province, China
- Hebei North University, Zhangjiakou, Hebei Province, China
| | - Jun Xue
- Department of Surgery, The First Affiliated Hospital of Hebei North University, Zhangjiakou, Hebei Province, China
| | - Xuejun Zhi
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Hebei North University, Zhangjiakou, Hebei province, China
| | - Rensen Ran
- Central Laboratory, The First Affiliated Hospital of Hebei North University, Zhangjiakou, Hebei province, China
- Department of Chemical Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, China
| |
Collapse
|
38
|
Augustin HG, Koh GY. A systems view of the vascular endothelium in health and disease. Cell 2024; 187:4833-4858. [PMID: 39241746 DOI: 10.1016/j.cell.2024.07.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Revised: 06/26/2024] [Accepted: 07/05/2024] [Indexed: 09/09/2024]
Abstract
The dysfunction of blood-vessel-lining endothelial cells is a major cause of mortality. Although endothelial cells, being present in all organs as a single-cell layer, are often conceived as a rather inert cell population, the vascular endothelium as a whole should be considered a highly dynamic and interactive systemically disseminated organ. We present here a holistic view of the field of vascular research and review the diverse functions of blood-vessel-lining endothelial cells during the life cycle of the vasculature, namely responsive and relaying functions of the vascular endothelium and the responsive roles as instructive gatekeepers of organ function. Emerging translational perspectives in regenerative medicine, preventive medicine, and aging research are developed. Collectively, this review is aimed at promoting disciplinary coherence in the field of angioscience for a broader appreciation of the importance of the vasculature for organ function, systemic health, and healthy aging.
Collapse
Affiliation(s)
- Hellmut G Augustin
- European Center for Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany; Division of Vascular Oncology and Metastasis, German Cancer Research Center Heidelberg (DKFZ), 69120 Heidelberg, Germany.
| | - Gou Young Koh
- Center for Vascular Research, Institute for Basic Science (IBS), Daejeon 34141, Republic of Korea; Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Republic of Korea.
| |
Collapse
|
39
|
Chen Y, Deng X, Li Y, Han Y, Peng Y, Wu W, Wang X, Ma J, Hu E, Zhou X, Shen E, Zeng S, Cai C, Qin Y, Shen H. Comprehensive molecular classification predicted microenvironment profiles and therapy response for HCC. Hepatology 2024; 80:536-551. [PMID: 38537130 PMCID: PMC11332383 DOI: 10.1097/hep.0000000000000869] [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: 09/30/2023] [Accepted: 02/07/2024] [Indexed: 06/06/2024]
Abstract
BACKGROUND AND AIMS Tumor microenvironment (TME) heterogeneity leads to a discrepancy in survival prognosis and clinical treatment response for patients with HCC. The clinical applications of documented molecular subtypes are constrained by several issues. APPROACH AND RESULTS We integrated 3 single-cell data sets to describe the TME landscape and identified 6 prognosis-related cell subclusters. Unsupervised clustering of subcluster-specific markers was performed to generate transcriptomic subtypes. The predictive value of these molecular subtypes for prognosis and treatment response was explored in multiple external HCC cohorts and the Xiangya HCC cohort. TME features were estimated using single-cell immune repertoire sequencing, mass cytometry, and multiplex immunofluorescence. The prognosis-related score was constructed based on a machine-learning algorithm. Comprehensive single-cell analysis described TME heterogeneity in HCC. The 5 transcriptomic subtypes possessed different clinical prognoses, stemness characteristics, immune landscapes, and therapeutic responses. Class 1 exhibited an inflamed phenotype with better clinical outcomes, while classes 2 and 4 were characterized by a lack of T-cell infiltration. Classes 5 and 3 indicated an inhibitory tumor immune microenvironment. Analysis of multiple therapeutic cohorts suggested that classes 5 and 3 were sensitive to immune checkpoint blockade and targeted therapy, whereas classes 1 and 2 were more responsive to transcatheter arterial chemoembolization treatment. Class 4 displayed resistance to all conventional HCC therapies. Four potential therapeutic agents and 4 targets were further identified for high prognosis-related score patients with HCC. CONCLUSIONS Our study generated a clinically valid molecular classification to guide precision medicine in patients with HCC.
Collapse
Affiliation(s)
- Yihong Chen
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Xiangying Deng
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yin Li
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Ying Han
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yinghui Peng
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Wantao Wu
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Xinwen Wang
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Jiayao Ma
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Erya Hu
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Xin Zhou
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Edward Shen
- Department of Life Science, McMaster University, Hamilton, Ontario, Canada
| | - Shan Zeng
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Changjing Cai
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yiming Qin
- Department of Spine Surgery and Orthopaedics, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Hong Shen
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| |
Collapse
|
40
|
Li J, Wang D, Tang F, Ling X, Zhang W, Zhang Z. Pan-cancer integrative analyses dissect the remodeling of endothelial cells in human cancers. Natl Sci Rev 2024; 11:nwae231. [PMID: 39345334 PMCID: PMC11429526 DOI: 10.1093/nsr/nwae231] [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: 04/03/2024] [Revised: 06/04/2024] [Accepted: 06/23/2024] [Indexed: 10/01/2024] Open
Abstract
Therapeutics targeting tumor endothelial cells (TECs) have been explored for decades, with only suboptimal efficacy achieved, partly due to an insufficient understanding of the TEC heterogeneity across cancer patients. We integrated single-cell RNA-seq data of 575 cancer patients from 19 solid tumor types, comprehensively charting the TEC phenotypic diversities. Our analyses uncovered underappreciated compositional and functional heterogeneity in TECs from a pan-cancer perspective. Two subsets, CXCR4 + tip cells and SELE + veins, represented the prominent angiogenic and proinflammatory phenotypes of TECs, respectively. They exhibited distinct spatial organization patterns, and compared to adjacent non-tumor tissues, tumor tissue showed an increased prevalence of CXCR4 + tip cells, yet with SELE + veins depleted. Such functional and spatial characteristics underlie their differential associations with the response of anti-angiogenic therapies and immunotherapies. Our integrative resources and findings open new avenues to understand and clinically intervene in the tumor vasculature.
Collapse
Affiliation(s)
- Jinhu Li
- Biomedical Pioneering Innovation Center (BIOPIC), School of Life Sciences, Peking University, Beijing 100871, China
| | - Dongfang Wang
- Biomedical Pioneering Innovation Center (BIOPIC), School of Life Sciences, Peking University, Beijing 100871, China
| | - Fei Tang
- Biomedical Pioneering Innovation Center (BIOPIC), School of Life Sciences, Peking University, Beijing 100871, China
- Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, China
| | - Xinnan Ling
- Biomedical Pioneering Innovation Center (BIOPIC), School of Life Sciences, Peking University, Beijing 100871, China
| | - Wenjie Zhang
- Biomedical Pioneering Innovation Center (BIOPIC), School of Life Sciences, Peking University, Beijing 100871, China
| | - Zemin Zhang
- Biomedical Pioneering Innovation Center (BIOPIC), School of Life Sciences, Peking University, Beijing 100871, China
- Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, China
| |
Collapse
|
41
|
Lei JX, Wang R, Hu C, Lou X, Lv MY, Li C, Gai B, Wu XJ, Dou R, Cai D, Gao F. Deciphering tertiary lymphoid structure heterogeneity reveals prognostic signature and therapeutic potentials for colorectal cancer: a multicenter retrospective cohort study. Int J Surg 2024; 110:5627-5640. [PMID: 38833363 PMCID: PMC11392219 DOI: 10.1097/js9.0000000000001684] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Accepted: 05/10/2024] [Indexed: 06/06/2024]
Abstract
BACKGROUND Tertiary lymphoid structures (TLSs) exert a crucial role in the tumor microenvironment (TME), impacting tumor development, immune escape, and drug resistance. Nonetheless, the heterogeneity of TLSs in colorectal cancer (CRC) and their impact on prognosis and treatment response remain unclear. METHODS The authors collected genome, transcriptome, clinicopathological information, and digital pathology images from multiple sources. An unsupervised clustering algorithm was implemented to determine diverse TLS patterns in CRC based on the expression levels of 39 TLS signature genes (TSGs). Comprehensive explorations of heterogeneity encompassing mutation landscape, TME, biological characteristics, response to immunotherapy, and drug resistance were conducted using multiomics data. TLSscore was then developed to quantitatively assess TLS patterns of individuals for further clinical applicability. RESULTS Three distinct TLS patterns were identified in CRC. Cluster 1 exhibited upregulation of proliferation-related pathways, high metabolic activity, and intermediate prognosis, while Cluster 2 displayed activation of stromal and carcinogenic pathways and a worse prognosis. Both Cluster 1 and Cluster 2 may potentially benefit from adjuvant chemotherapy. Cluster 3, characterized by the activation of immune regulation and activation pathways, demonstrated a favorable prognosis and enhanced responsiveness to immunotherapy. The authors subsequently employed a regularization algorithm to construct the TLSscore based on nine core genes. Patients with lower TLSscore trended to prolonged prognosis and a more prominent presence of TLSs, which may benefit from immunotherapy. Conversely, those with higher TLSscore exhibited increased benefits from adjuvant chemotherapy. CONCLUSIONS The authors identified distinct TLS patterns in CRC and characterized their heterogeneity through multiomics analyses. The TLSscore held promise for guiding clinical decision-making and further advancing the field of personalized medicine in CRC.
Collapse
Affiliation(s)
- Jia-Xin Lei
- Department of General Surgery (Colorectal Surgery), The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou Province
- School of Medicine, Shenzhen Campus of Sun Yat-Sen University, ShenzhenGuangdong Province
| | - Runxian Wang
- Department of Gastrointestinal Surgery, The Fifth Affiliated Hospital of Sun Yat-sen University, ZhuhaiGuangdong Province
| | - Chuling Hu
- Department of General Surgery (Colorectal Surgery), The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou Province
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University Guangzhou, Guangdong Province
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University Guangzhou
| | - Xiaoying Lou
- Department of Pathology, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou
| | - Min-Yi Lv
- Department of General Surgery (Colorectal Surgery), The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou Province
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University Guangzhou, Guangdong Province
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University Guangzhou
| | - Chenghang Li
- Artificial Intelligence Thrust, The Hong Kong University of Science and Technology, Guangzhou, People's Republic of China
| | - Baowen Gai
- Department of General Surgery (Colorectal Surgery), The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou Province
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University Guangzhou, Guangdong Province
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University Guangzhou
| | - Xiao-Jian Wu
- Department of General Surgery (Colorectal Surgery), The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou Province
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University Guangzhou, Guangdong Province
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University Guangzhou
| | - Ruoxu Dou
- Department of Gastrointestinal Surgery, The Fifth Affiliated Hospital of Sun Yat-sen University, ZhuhaiGuangdong Province
| | - Du Cai
- Department of General Surgery (Colorectal Surgery), The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou Province
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University Guangzhou, Guangdong Province
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University Guangzhou
| | - Feng Gao
- Department of General Surgery (Colorectal Surgery), The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou Province
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University Guangzhou, Guangdong Province
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University Guangzhou
| |
Collapse
|
42
|
Declercq M, Treps L, Geldhof V, Conchinha NV, de Rooij LPMH, Subramanian A, Feyeux M, Cotinat M, Boeckx B, Vinckier S, Dupont L, Vermeulen F, Boon M, Proesmans M, Libbrecht L, Pirenne J, Monbaliu D, Jochmans I, Dewerchin M, Eelen G, Roskams T, Verleden S, Lambrechts D, Carmeliet P, Witters P. Single-cell RNA sequencing of cystic fibrosis liver disease explants reveals endothelial complement activation. Liver Int 2024; 44:2382-2395. [PMID: 38847551 DOI: 10.1111/liv.15963] [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: 11/27/2023] [Revised: 04/04/2024] [Accepted: 04/21/2024] [Indexed: 08/30/2024]
Abstract
BACKGROUND & AIMS Cystic fibrosis (CF) is considered a multisystemic disorder in which CF-associated liver disease (CFLD) is the third most common cause of mortality. Currently, no effective treatment is available for CFLD because its pathophysiology is still unclear. Interestingly, CFLD exhibits identical vascular characteristics as non-cirrhotic portal hypertension, recently classified as porto-sinusoidal vascular disorders (PSVD). METHODS Since endothelial cells (ECs) are an important component in PSVD, we performed single-cell RNA sequencing (scRNA-seq) on four explant livers from CFLD patients to identify differential endothelial characteristics which could contribute to the disease. We comprehensively characterized the endothelial compartment and compared it with publicly available scRNA-seq datasets from cirrhotic and healthy livers. Key gene signatures were validated ex vivo on patient tissues. RESULTS We found that ECs from CF liver explants are more closely related to healthy than cirrhotic patients. In CF patients we also discovered a distinct population of liver sinusoidal ECs-coined CF LSECs-upregulating genes involved in the complement cascade and coagulation. Finally, our immunostainings further validated the predominant periportal location of CF LSECs. CONCLUSIONS Our work showed novel aspects of human liver ECs at the single-cell level thereby supporting endothelial involvement in CFLD, and reinforcing the hypothesis that ECs could be a driver of PSVD. Therefore, considering the vascular compartment in CF and CFLD may help developing new therapeutic approaches for these diseases.
Collapse
Affiliation(s)
- Mathias Declercq
- Department of Development and Regeneration, Woman and Child Unit, KU Leuven, Leuven, Belgium
- Laboratory of Angiogenesis and Vascular Metabolism, Department of Oncology and Leuven Cancer Institute (LKI), KU Leuven, VIB Center for Cancer Biology, VIB, Leuven, Belgium
| | - Lucas Treps
- Laboratory of Angiogenesis and Vascular Metabolism, Department of Oncology and Leuven Cancer Institute (LKI), KU Leuven, VIB Center for Cancer Biology, VIB, Leuven, Belgium
- Nantes Université, INSERM UMR 1307, CNRS UMR 6075, Université d'Angers, CRCI2NA, Nantes, France
| | - Vincent Geldhof
- Laboratory of Angiogenesis and Vascular Metabolism, Department of Oncology and Leuven Cancer Institute (LKI), KU Leuven, VIB Center for Cancer Biology, VIB, Leuven, Belgium
| | - Nadine V Conchinha
- Laboratory of Angiogenesis and Vascular Metabolism, Department of Oncology and Leuven Cancer Institute (LKI), KU Leuven, VIB Center for Cancer Biology, VIB, Leuven, Belgium
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - Laura P M H de Rooij
- Laboratory of Angiogenesis and Vascular Metabolism, Department of Oncology and Leuven Cancer Institute (LKI), KU Leuven, VIB Center for Cancer Biology, VIB, Leuven, Belgium
- The CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Abhishek Subramanian
- Laboratory of Angiogenesis and Vascular Metabolism, Department of Oncology and Leuven Cancer Institute (LKI), KU Leuven, VIB Center for Cancer Biology, VIB, Leuven, Belgium
- Department of Biotechnology, Indian Institute of Technology, Hyderabad, Telangana, India
| | - Magalie Feyeux
- Nantes Université, CHU Nantes, CNRS, Inserm, BioCore, US16, SFR Bonamy, Nantes, France
| | - Marine Cotinat
- Nantes Université, INSERM UMR 1307, CNRS UMR 6075, Université d'Angers, CRCI2NA, Nantes, France
| | - Bram Boeckx
- Laboratory for Translational Genetics, Center for Cancer Biology, VIB, Leuven, Belgium
- Laboratory for Translational Genetics, Department of Human Genetics, KU Leuven, Leuven, Belgium
| | - Stefan Vinckier
- Laboratory of Angiogenesis and Vascular Metabolism, Department of Oncology and Leuven Cancer Institute (LKI), KU Leuven, VIB Center for Cancer Biology, VIB, Leuven, Belgium
| | - Lieven Dupont
- Department of Pneumology, UZ Leuven, Leuven, Belgium
- Department of Chronic Diseases and Metabolism, Respiratory Diseases and Thoracic Surgery, KU Leuven, Leuven, Belgium
| | - Francois Vermeulen
- Department of Development and Regeneration, Woman and Child Unit, KU Leuven, Leuven, Belgium
- Department of Pediatrics, Pediatric Pulmonology, University Hospital of Leuven, Leuven, Flanders, Belgium
| | - Mieke Boon
- Department of Development and Regeneration, Woman and Child Unit, KU Leuven, Leuven, Belgium
- Department of Pediatrics, Pediatric Pulmonology, University Hospital of Leuven, Leuven, Flanders, Belgium
| | - Marijke Proesmans
- Department of Development and Regeneration, Woman and Child Unit, KU Leuven, Leuven, Belgium
- Department of Pediatrics, Pediatric Pulmonology, University Hospital of Leuven, Leuven, Flanders, Belgium
| | - Louis Libbrecht
- Department of Pathology, Cliniques Universitaires Saint-Luc, Brussels, Belgium
- Department of Pathology, AZ Groeninge, Kortrijk, Belgium
- Laboratory of Hepatology, KU Leuven, Leuven, Belgium
| | - Jacques Pirenne
- Transplantation Research Group, Department of Immunology, Microbiology and Transplantation, KU Leuven, Leuven, Belgium
- Department of Abdominal Transplant Surgery, University Hospitals Leuven, Leuven, Belgium
| | - Diethard Monbaliu
- Transplantation Research Group, Department of Immunology, Microbiology and Transplantation, KU Leuven, Leuven, Belgium
- Department of Abdominal Transplant Surgery, University Hospitals Leuven, Leuven, Belgium
| | - Ina Jochmans
- Transplantation Research Group, Department of Immunology, Microbiology and Transplantation, KU Leuven, Leuven, Belgium
- Department of Abdominal Transplant Surgery, University Hospitals Leuven, Leuven, Belgium
| | - Mieke Dewerchin
- Laboratory of Angiogenesis and Vascular Metabolism, Department of Oncology and Leuven Cancer Institute (LKI), KU Leuven, VIB Center for Cancer Biology, VIB, Leuven, Belgium
| | - Guy Eelen
- Laboratory of Angiogenesis and Vascular Metabolism, Department of Oncology and Leuven Cancer Institute (LKI), KU Leuven, VIB Center for Cancer Biology, VIB, Leuven, Belgium
| | - Tania Roskams
- Department of Imaging and Pathology, Translational Cell and Tissue Research, KU Leuven and University Hospitals Leuven, Leuven, Belgium
| | - Stijn Verleden
- Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), Department CHROMETA, KU Leuven, Leuven, Belgium
- Department of ASTARC, University of Antwerp, Wilrijk, Belgium
| | - Diether Lambrechts
- Laboratory for Translational Genetics, Center for Cancer Biology, VIB, Leuven, Belgium
- Laboratory for Translational Genetics, Department of Human Genetics, KU Leuven, Leuven, Belgium
| | - Peter Carmeliet
- Laboratory of Angiogenesis and Vascular Metabolism, Department of Oncology and Leuven Cancer Institute (LKI), KU Leuven, VIB Center for Cancer Biology, VIB, Leuven, Belgium
- Center for Biotechnology, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates
| | - Peter Witters
- Department of Development and Regeneration, Woman and Child Unit, KU Leuven, Leuven, Belgium
- Department of Paediatrics and Metabolic Center, University Hospitals Leuven, Leuven, Belgium
| |
Collapse
|
43
|
Fu Z, Zhang L, Chen R, Zhan J, Zhong J, Zheng W, Zou J, Wang P, Deng X, Lin AY, Wang DD, Lin PP, He R. Biphasic co-detection of melanoma aneuploid tumor cells and tumor endothelial cells in guidance of specifying the field cancerized surgical excision margin and administering immunotherapy. Cancer Lett 2024; 598:217099. [PMID: 38971491 DOI: 10.1016/j.canlet.2024.217099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2024] [Revised: 06/18/2024] [Accepted: 06/29/2024] [Indexed: 07/08/2024]
Abstract
An optimum safety excision margin (EM) delineated by precise demarcation of field cancerization along with reliable biomarkers that enable predicting and timely evaluating patients' response to immunotherapy significantly impact effective management of melanoma. In this study, optimized biphasic "immunofluorescence staining integrated with fluorescence insitu hybridization" (iFISH) was conducted along the diagnosis-metastasis-treatment-cellular MRD axis to longitudinally co-detect a full spectrum of intact CD31- aneuploid tumor cells (TCs), CD31+ aneuploid tumor endothelial cells (TECs), viable and necrotic circulating TCs (CTCs) and circulating TECs (CTECs) expressing PD-L1, Ki67, p16 and Vimentin in unsliced specimens of the resected primary tumor, EM, dissected sentinel lymph nodes (SLNs) and peripheral blood in an early-stage melanoma patient. Numerous PD-L1+ aneuploid TCs and TECs were detected at the conventional safety EM (2 cm), quantitatively indicating the existence of a field cancerized EM for the first time. Contrary to highly heterogeneous PD-L1 expression and degrees of Chr8 aneuploidy in TCs and TECs in the primary lesions as well as CTCs and CTECs in peripheral blood, almost all TCs and TECs in SLNs and EM were homogeneously PD-L1+ haploid cells. Dynamic monitoring and cellular MRD assessment revealed that, in contrast to PD-L1+ CTCs being responsive to the immune checkpoint inhibitor (ICI-anti-PD-1), multiploid (≥pentasomy 8) PD-L1+ and Ki67+ CTECs were respectively resistant to ICI-sensitized T cells. In therapeutically stressed lymphatic and hematogenous metastatic cascades, stratified phenotypic and karyotypic profiling of iFISH tissue and liquid biopsied TCs, TECs, CTCs and CTECs in future large-cohort studies will enable appropriate re-specification of the optimal safety EM and distribution mapping of in-depth characterized, subcategorized target cells to help illustrate their metastatic relevance, ultimately improving risk stratification and clinical intervention of tumor progression, metastases, therapy resistance and cancer relapse.
Collapse
Affiliation(s)
- Zhengzheng Fu
- Department of Dermatologic Surgery and Dermatologic Oncology, Dermatology Hospital of Southern Medical University, Guangdong Provincial Dermatology Hospital, Guangzhou, China
| | - Lina Zhang
- Department of Cellular and Molecular Biology, Beijing Chest Hospital, Capital Medical University, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, China
| | - Rongyi Chen
- Division of Cutaneous Oncology, Department of Dermatology, Dermatology Hospital of Southern Medical University, Guangdong Provincial Dermatology Hospital, Guangzhou, China
| | - Jipang Zhan
- Department of Dermatologic Surgery and Dermatologic Oncology, Dermatology Hospital of Southern Medical University, Guangdong Provincial Dermatology Hospital, Guangzhou, China
| | - Jing Zhong
- Department of Dermatologic Surgery and Dermatologic Oncology, Dermatology Hospital of Southern Medical University, Guangdong Provincial Dermatology Hospital, Guangzhou, China
| | - Wen Zheng
- Department of Dermatologic Surgery and Dermatologic Oncology, Dermatology Hospital of Southern Medical University, Guangdong Provincial Dermatology Hospital, Guangzhou, China
| | - Jingwen Zou
- Department of Dermatologic Surgery and Dermatologic Oncology, Dermatology Hospital of Southern Medical University, Guangdong Provincial Dermatology Hospital, Guangzhou, China
| | - Peng Wang
- Department of Pathology, Dermatology Hospital of Southern Medical University, Guangdong Provincial Dermatology Hospital, Guangzhou, China
| | - Xiaohua Deng
- Department of Pathology, Dermatology Hospital of Southern Medical University, Guangdong Provincial Dermatology Hospital, Guangzhou, China
| | | | | | | | - Renliang He
- Department of Dermatologic Surgery and Dermatologic Oncology, Dermatology Hospital of Southern Medical University, Guangdong Provincial Dermatology Hospital, Guangzhou, China.
| |
Collapse
|
44
|
Jin Y, Zhang J, Xing J, Li Y, Yang H, Ouyang L, Fang Z, Sun L, Jin B, Huang P, Yang H, Du S, Sang X, Mao Y. Multicellular 3D bioprinted human gallbladder carcinoma for in vitromimicry of tumor microenvironment and intratumoral heterogeneity. Biofabrication 2024; 16:045028. [PMID: 39121870 DOI: 10.1088/1758-5090/ad6d8c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Accepted: 08/09/2024] [Indexed: 08/12/2024]
Abstract
Gallbladder carcinoma (GBC) is a malignant hepatobiliary cancer characterized by an intricate tumor microenvironments (TME) and heterogeneity. The traditional GBC 2D culture models cannot faithfully recapitulate the characteristics of the TME. Three-dimensional (3D) bioprinting enables the establishment of high-throughput and high-fidelity multicellular GBC models. In this study, we designed a concentric cylindrical tetra-culture model to reconstitute the spatial distribution of cells in tumor tissue, with the inner portion containing GBC cells, and the outer ring containing a mixture of endothelial cells, fibroblasts, and macrophages. We confirmed the survival, proliferation, biomarker expression and gene expression profiles of GBC 3D tetra-culture models. Hematoxylin-eosin (HE) and immunofluorescence staining verified the morphology and robust expression of GBC/endothelial/fibroblast/macrophage biomarkers in GBC 3D tetra-culture models. Single-cell RNA sequencing revealed two distinct subtypes of GBC cells within the model, glandular epithelial and squamous epithelial cells, suggesting the mimicry of intratumoral heterogeneity. Comparative transcriptome profile analysis among variousin vitromodels revealed that cellular interactions and the TME in 3D tetra-culture models reshaped the biological processes of tumor cells to a more aggressive phenotype. GBC 3D tetra-culture models restored the characteristics of the TME as well as intratumoral heterogeneity. Therefore, this model is expected to have future applications in tumor biology research and antitumor drug development.
Collapse
Affiliation(s)
- Yukai Jin
- Department of Liver Surgery, Peking Union Medical College (PUMC) Hospital, PUMC & Chinese Academy of Medical Sciences (CAMS), Beijing, People's Republic of China
- Department of Gastric Surgery, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, People's Republic of China
| | - Jiangang Zhang
- Department of Liver Surgery, Peking Union Medical College (PUMC) Hospital, PUMC & Chinese Academy of Medical Sciences (CAMS), Beijing, People's Republic of China
- Eight-Year Medical Doctor Program, Peking Union Medical College (PUMC) Hospital, PUMC & Chinese Academy of Medical Sciences (CAMS), Beijing, People's Republic of China
| | - Jiali Xing
- Department of Liver Surgery, Peking Union Medical College (PUMC) Hospital, PUMC & Chinese Academy of Medical Sciences (CAMS), Beijing, People's Republic of China
| | - Yiran Li
- Department of Liver Surgery, Peking Union Medical College (PUMC) Hospital, PUMC & Chinese Academy of Medical Sciences (CAMS), Beijing, People's Republic of China
| | - Huiyu Yang
- Department of Liver Surgery, Peking Union Medical College (PUMC) Hospital, PUMC & Chinese Academy of Medical Sciences (CAMS), Beijing, People's Republic of China
- Eight-Year Medical Doctor Program, Peking Union Medical College (PUMC) Hospital, PUMC & Chinese Academy of Medical Sciences (CAMS), Beijing, People's Republic of China
| | - Liujian Ouyang
- Department of Endocrinology, Children's Hospital of Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, Zhejiang, People's Republic of China
| | - Zhiyuan Fang
- Department of Liver Surgery, Peking Union Medical College (PUMC) Hospital, PUMC & Chinese Academy of Medical Sciences (CAMS), Beijing, People's Republic of China
- Eight-Year Medical Doctor Program, Peking Union Medical College (PUMC) Hospital, PUMC & Chinese Academy of Medical Sciences (CAMS), Beijing, People's Republic of China
| | - Lejia Sun
- Department of Liver Surgery, Peking Union Medical College (PUMC) Hospital, PUMC & Chinese Academy of Medical Sciences (CAMS), Beijing, People's Republic of China
- Colorectal Surgery Division, Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, People's Republic of China
| | - Bao Jin
- Department of Liver Surgery, Peking Union Medical College (PUMC) Hospital, PUMC & Chinese Academy of Medical Sciences (CAMS), Beijing, People's Republic of China
| | - Pengyu Huang
- Institute of Biomedical Engineering, PUMC & CAMS, Tianjin, People's Republic of China
| | - Huayu Yang
- Department of Liver Surgery, Peking Union Medical College (PUMC) Hospital, PUMC & Chinese Academy of Medical Sciences (CAMS), Beijing, People's Republic of China
| | - Shunda Du
- Department of Liver Surgery, Peking Union Medical College (PUMC) Hospital, PUMC & Chinese Academy of Medical Sciences (CAMS), Beijing, People's Republic of China
| | - Xinting Sang
- Department of Liver Surgery, Peking Union Medical College (PUMC) Hospital, PUMC & Chinese Academy of Medical Sciences (CAMS), Beijing, People's Republic of China
| | - Yilei Mao
- Department of Liver Surgery, Peking Union Medical College (PUMC) Hospital, PUMC & Chinese Academy of Medical Sciences (CAMS), Beijing, People's Republic of China
| |
Collapse
|
45
|
Ren F, Wang L, Wang Y, Wang J, Wang Y, Song X, Zhang G, Nie F, Lin S. Single-cell transcriptome profiles the heterogeneity of tumor cells and microenvironments for different pathological endometrial cancer and identifies specific sensitive drugs. Cell Death Dis 2024; 15:571. [PMID: 39112478 PMCID: PMC11306564 DOI: 10.1038/s41419-024-06960-8] [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: 04/16/2024] [Revised: 07/23/2024] [Accepted: 07/29/2024] [Indexed: 08/10/2024]
Abstract
Endometrial cancer (EC) is a highly heterogeneous malignancy characterized by varied pathology and prognoses, and the heterogeneity of its cancer cells and the tumor microenvironment (TME) remains poorly understood. We conducted single-cell RNA sequencing (scRNA-seq) on 18 EC samples, encompassing various pathological types to delineate their specific unique transcriptional landscapes. Cancer cells from diverse pathological sources displayed distinct hallmarks labeled as immune-modulating, proliferation-modulating, and metabolism-modulating cancer cells in uterine clear cell carcinomas (UCCC), well-differentiated endometrioid endometrial carcinomas (EEC-I), and uterine serous carcinomas (USC), respectively. Cancer cells from the UCCC exhibited the greatest heterogeneity. We also identified potential effective drugs and confirmed their effectiveness using patient-derived EC organoids for each pathological group. Regarding the TME, we observed that prognostically favorable CD8+ Tcyto and NK cells were prominent in normal endometrium, whereas CD4+ Treg, CD4+ Tex, and CD8+ Tex cells dominated the tumors. CXCL3+ macrophages associated with M2 signature and angiogenesis were exclusively found in tumors. Prognostically relevant epithelium-specific cancer-associated fibroblasts (eCAFs) and SOD2+ inflammatory CAFs (iCAFs) predominated in EEC-I and UCCC groups, respectively. We also validated the oncogenic effects of SOD2+ iCAFs in vitro. Our comprehensive study has yielded deeper insights into the pathogenesis of EC, potentially facilitating personalized treatments for its varied pathological types.
Collapse
Affiliation(s)
- Fang Ren
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China.
| | - Lingfang Wang
- Zhejiang Provincial Key Laboratory of Precision Diagnosis and Therapy for Major Gynecological Diseases, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yuyouye Wang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Jiaxuan Wang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Yuanpei Wang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Xiaole Song
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Gong Zhang
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Fangfang Nie
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Shitong Lin
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China.
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, 430022, Wuhan, Hubei, PR China.
| |
Collapse
|
46
|
Li J, Zhang Y, Liang C, Yan X, Hui X, Liu Q. Advancing precision medicine in gliomas through single-cell sequencing: unveiling the complex tumor microenvironment. Front Cell Dev Biol 2024; 12:1396836. [PMID: 39156969 PMCID: PMC11327033 DOI: 10.3389/fcell.2024.1396836] [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: 03/06/2024] [Accepted: 07/22/2024] [Indexed: 08/20/2024] Open
Abstract
Glioblastoma (GBM) displays an infiltrative growth characteristic that recruits neighboring normal cells to facilitate tumor growth, maintenance, and invasion into the brain. While the blood-brain barrier serves as a critical natural defense mechanism for the central nervous system, GBM disrupts this barrier, resulting in the infiltration of macrophages from the peripheral bone marrow and the activation of resident microglia. Recent advancements in single-cell transcriptomics and spatial transcriptomics have refined the categorization of cells within the tumor microenvironment for precise identification. The intricate interactions and influences on cell growth within the tumor microenvironment under multi-omics conditions are succinctly outlined. The factors and mechanisms involving microglia, macrophages, endothelial cells, and T cells that impact the growth of GBM are individually examined. The collaborative mechanisms of tumor cell-immune cell interactions within the tumor microenvironment synergistically promote the growth, infiltration, and metastasis of gliomas, while also influencing the immune status and therapeutic response of the tumor microenvironment. As immunotherapy continues to progress, targeting the cells within the inter-tumor microenvironment emerges as a promising novel therapeutic approach for GBM. By comprehensively understanding and intervening in the intricate cellular interactions within the tumor microenvironment, novel therapeutic modalities may be developed to enhance treatment outcomes for patients with GBM.
Collapse
Affiliation(s)
- Jinwei Li
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Department of Neurosurgery, Liuzhou Workers Hospital, Liuzhou, Guangxi, China
| | - Yang Zhang
- Graduate School of Medicine, Kunming Medical University, Kunming, Yunnan, China
| | - Cong Liang
- Department of Pharmacy, Liuzhou Workers Hospital, Liuzhou, Guangxi, China
| | - Xianlei Yan
- Department of Neurosurgery, Liuzhou Workers Hospital, Liuzhou, Guangxi, China
| | - Xuhui Hui
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Quan Liu
- Department of Neurosurgery, Liuzhou Workers Hospital, Liuzhou, Guangxi, China
| |
Collapse
|
47
|
Coursier D, Calvo F. CAFs vs. TECs: when blood feuds fuel cancer progression, dissemination and therapeutic resistance. Cell Oncol (Dordr) 2024; 47:1091-1112. [PMID: 38453816 PMCID: PMC11322395 DOI: 10.1007/s13402-024-00931-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] [Accepted: 02/23/2024] [Indexed: 03/09/2024] Open
Abstract
Neoplastic progression involves complex interactions between cancer cells and the surrounding stromal milieu, fostering microenvironments that crucially drive tumor progression and dissemination. Of these stromal constituents, cancer-associated fibroblasts (CAFs) emerge as predominant inhabitants within the tumor microenvironment (TME), actively shaping multiple facets of tumorigenesis, including cancer cell proliferation, invasiveness, and immune evasion. Notably, CAFs also orchestrate the production of pro-angiogenic factors, fueling neovascularization to sustain the metabolic demands of proliferating cancer cells. Moreover, CAFs may also directly or indirectly affect endothelial cell behavior and vascular architecture, which may impact in tumor progression and responses to anti-cancer interventions. Conversely, tumor endothelial cells (TECs) exhibit a corrupted state that has been shown to affect cancer cell growth and inflammation. Both CAFs and TECs are emerging as pivotal regulators of the TME, engaging in multifaceted biological processes that significantly impact cancer progression, dissemination, and therapeutic responses. Yet, the intricate interplay between these stromal components and the orchestrated functions of each cell type remains incompletely elucidated. In this review, we summarize the current understanding of the dynamic interrelationships between CAFs and TECs, discussing the challenges and prospects for leveraging their interactions towards therapeutic advancements in cancer.
Collapse
Affiliation(s)
- Diane Coursier
- Instituto de Biomedicina y Biotecnología de Cantabria (Consejo Superior de Investigaciones Científicas, Universidad de Cantabria), Santander, Spain
| | - Fernando Calvo
- Instituto de Biomedicina y Biotecnología de Cantabria (Consejo Superior de Investigaciones Científicas, Universidad de Cantabria), Santander, Spain.
| |
Collapse
|
48
|
Fu D, Zhang T, Liu J, Chang B, Zhang Q, Tan Y, Chen X, Tan L. Identification of adipocyte infiltration-related gene subtypes for predicting colorectal cancer prognosis and responses of immunotherapy/chemotherapy. Heliyon 2024; 10:e33616. [PMID: 39050460 PMCID: PMC11266998 DOI: 10.1016/j.heliyon.2024.e33616] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Revised: 06/22/2024] [Accepted: 06/24/2024] [Indexed: 07/27/2024] Open
Abstract
Colorectal cancer (CRC) is a prevalent and aggressive malignancy characterized by a complex tumor microenvironment (TME). Given the variations in the level of adipocyte infiltration in TME, the prognosis may differ among CRC patients. Thus, there is an urgent need to establish a reliable method for identifying adipocyte subtypes in CRC in order to elucidate the impact of adipocyte infiltration on CRC treatment and prognosis. Herein, 144 adipocyte-infiltration-related genes (AIRGs) were identified as predictive markers for the immune-associated features and prognosis of CRC patients. Based on the 144 genes, the unsupervised clustering algorithm identified two distinct clusters of CRC patients with variations in molecular and signaling pathways, clinicopathological characteristics and responses to CRC chemotherapy and immunotherapy. Furthermore, an AIRG prognostic signature was constructed and validated in independent datasets. Overall, this study developed a prognostic signature based on AIRGs in CRC, which may contribute to the development of personalized treatment strategies and enhance prognostic prediction for CRC patients.
Collapse
Affiliation(s)
- Daan Fu
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Key Laboratory of Anesthesiology and Resuscitation (Huazhong University of Science and Technology), Ministry of Education, China
- Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Tianhao Zhang
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Jia Liu
- Research Center for Tissue Engineering and Regenerative Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Bingcheng Chang
- The Second Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, 550003, China
| | - Qingqing Zhang
- Haiyan County Hospital of Traditional Chinese Medicine, JiaXing, 314399, China
| | - Yuyan Tan
- Department of Breast and Thyroid Surgery, The First College of Clinical Medical Science, China Three Gorges University, Yichang, 443000, China
| | - Xiangdong Chen
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Key Laboratory of Anesthesiology and Resuscitation (Huazhong University of Science and Technology), Ministry of Education, China
- Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Lulu Tan
- Department of Breast and Thyroid Surgery, The First College of Clinical Medical Science, China Three Gorges University, Yichang, 443000, China
| |
Collapse
|
49
|
Fu Y, Tao J, Liu T, Liu Y, Qiu J, Su D, Wang R, Luo W, Cao Z, Weng G, Zhang T, Zhao Y. Unbiasedly decoding the tumor microenvironment with single-cell multiomics analysis in pancreatic cancer. Mol Cancer 2024; 23:140. [PMID: 38982491 PMCID: PMC11232163 DOI: 10.1186/s12943-024-02050-7] [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: 05/07/2024] [Accepted: 06/21/2024] [Indexed: 07/11/2024] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a highly aggressive malignancy with a poor prognosis and limited therapeutic options. Research on the tumor microenvironment (TME) of PDAC has propelled the development of immunotherapeutic and targeted therapeutic strategies with a promising future. The emergence of single-cell sequencing and mass spectrometry technologies, coupled with spatial omics, has collectively revealed the heterogeneity of the TME from a multiomics perspective, outlined the development trajectories of cell lineages, and revealed important functions of previously underrated myeloid cells and tumor stroma cells. Concurrently, these findings necessitated more refined annotations of biological functions at the cell cluster or single-cell level. Precise identification of all cell clusters is urgently needed to determine whether they have been investigated adequately and to identify target cell clusters with antitumor potential, design compatible treatment strategies, and determine treatment resistance. Here, we summarize recent research on the PDAC TME at the single-cell multiomics level, with an unbiased focus on the functions and potential classification bases of every cellular component within the TME, and look forward to the prospects of integrating single-cell multiomics data and retrospectively reusing bulk sequencing data, hoping to provide new insights into the PDAC TME.
Collapse
Affiliation(s)
- Yifan Fu
- General Surgery Department, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
- 4+4 Medical Doctor Program, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Jinxin Tao
- General Surgery Department, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Tao Liu
- General Surgery Department, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Yueze Liu
- General Surgery Department, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Jiangdong Qiu
- General Surgery Department, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Dan Su
- General Surgery Department, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Ruobing Wang
- General Surgery Department, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Wenhao Luo
- General Surgery Department, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Zhe Cao
- General Surgery Department, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Guihu Weng
- General Surgery Department, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Taiping Zhang
- General Surgery Department, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China.
- Clinical Immunology Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China.
| | - Yupei Zhao
- General Surgery Department, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China.
| |
Collapse
|
50
|
Huang S, Hou Y, Tang Z, Suhail M, Cui M, Iqbal MZ, Kong X. Near-infrared-II responsive ovalbumin functionalized gold-genipin nanosystem cascading photo-immunotherapy of cancer. NANOTECHNOLOGY 2024; 35:365102. [PMID: 38861966 DOI: 10.1088/1361-6528/ad568c] [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: 03/05/2024] [Accepted: 06/11/2024] [Indexed: 06/13/2024]
Abstract
Synergistic cancer therapies have attracted wide attention owing to their multi-mode tumor inhibition properties. Especially, photo-responsive photoimmunotherapy demonstrates an emerging cancer treatment paradigm that significantly improved treatment efficiency. Herein, near-infrared-II responsive ovalbumin functionalized Gold-Genipin nanosystem (Au-G-OVA NRs) was designed for immunotherapy and deep photothermal therapy of breast cancer. A facile synthesis method was employed to prepare the homogeneous Au nanorods (Au NRs) with good dispersion. The nanovaccine was developed further by the chemical cross-linking of Au-NRs, genipin and ovalbumin. The Au-G-OVA NRs outstanding aqueous solubility, and biocompatibility against normal and cancer cells. The designed NRs possessed enhanced localized surface plasmon resonance (LSPR) effect, which extended the NIR absorption in the second window, enabling promising photothermal properties. Moreover, genipin coating provided complimentary red fluorescent and prepared Au-G-OVA NRs showed significant intracellular encapsulation for efficient photoimmunotherapy outcomes. The designed nanosystem possessed deep photothermal therapy of breast cancer and 90% 4T1 cells were ablated by Au-G-OVA NRs (80μg ml-1concentration) after 1064 nm laser irradiation. In addition, Au-G-OVA NRs demonstrated outstanding vaccination phenomena by facilitating OVA delivery, antigen uptake, maturation of bone marrow dendritic cells, and cytokine IFN-γsecretion for tumor immunosurveillance. The aforementioned advantages permit the utilization of fluorescence imaging-guided photo-immunotherapy for cancers, demonstrating a straightforward approach for developing nanovaccines tailored to precise tumor treatment.
Collapse
Affiliation(s)
- Shuqi Huang
- Institute of Smart Biomedical Materials, School of Materials Science and Engineering, Zhejiang Sci-Tech University, Hangzhou 310018, People's Republic of China
| | - Yike Hou
- Institute of Smart Biomedical Materials, School of Materials Science and Engineering, Zhejiang Sci-Tech University, Hangzhou 310018, People's Republic of China
| | - Zhe Tang
- Institute of Smart Biomedical Materials, School of Materials Science and Engineering, Zhejiang Sci-Tech University, Hangzhou 310018, People's Republic of China
- South China Advanced Institute for Soft Matter Science and Technology, School of Emergent Soft Matter, South China University of Technology, Guangzhou 510640, People's Republic of China
| | - Muhamamd Suhail
- Institute of Smart Biomedical Materials, School of Materials Science and Engineering, Zhejiang Sci-Tech University, Hangzhou 310018, People's Republic of China
| | - Mingyue Cui
- Institute of Smart Biomedical Materials, School of Materials Science and Engineering, Zhejiang Sci-Tech University, Hangzhou 310018, People's Republic of China
| | - M Zubair Iqbal
- Institute of Smart Biomedical Materials, School of Materials Science and Engineering, Zhejiang Sci-Tech University, Hangzhou 310018, People's Republic of China
| | - Xiangdong Kong
- Institute of Smart Biomedical Materials, School of Materials Science and Engineering, Zhejiang Sci-Tech University, Hangzhou 310018, People's Republic of China
| |
Collapse
|