1
|
Liu Q, Wang Y, Song M, Huang J, Shi J, Sun W, Ji X, Chang Y, Ma B, Zhang P, Yan Y, Zhang H. CCL20/CXCL5 Drives Crosstalk Between Anaplastic Thyroid Cancer Stem Cells and Tumor-Associated Macrophages to Promote Tumor Progression. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2405399. [PMID: 40091357 PMCID: PMC12061268 DOI: 10.1002/advs.202405399] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Revised: 02/26/2025] [Indexed: 03/19/2025]
Abstract
The dynamic interplay between tumor-associated macrophages (TAMs) and anaplastic thyroid cancer (ATC) shapes the tumor microenvironment and facilitates ATC progression. However, the mechanisms of communication between TAMs and anaplastic thyroid cancer stem cells (ATCSCs) remain largely unelucidated. Integrative analyses of single-cell RNA sequencing, cytokine/chemokine arrays, proteomics, and mRNA expression datasets are performed to reveal crosstalk between TAMs and ATCSCs and signaling pathways in ATCSCs. Subsequently, in vitro experiments are performed to validate the regulatory effects of key cytokines on ATCSC stemness. Last, xenogeneic orthotopic thyroid ATCSCs transplantation models are utilized to corroborate the regulatory effect of cytokines on stemness. CCL20 derived from THP-1-M2 activates the IRAK-1/NF-κB1/2 signaling pathway in ATCSCs, thereby positively regulating stemness characteristics and upregulating CXCL5 secretion. ATCSCs not only exhibit autocrine CXCL5 participation in the regulation of stemness but also demonstrate paracrine CXCL5 activity to recruit THP-1-Mφ and maintain the M2 phenotype. CCL20 and CXCL5 are involved in the crosstalk between TAMs and ATCSCs. The CCL20/CXCL5 axis plays a crucial role in the interaction between TAMs and ATCSCs, establishing a progressive tumor microenvironment.
Collapse
Affiliation(s)
- Qi Liu
- Department of Thyroid SurgeryThe First Hospital of China Medical UniversityShenyang110801P. R. China
| | - Yan Wang
- Department of PharmacologySchool of PharmacyChina Medical UniversityShenyang110122P. R. China
| | - Mingyuan Song
- Department of Thyroid SurgeryThe First Hospital of China Medical UniversityShenyang110801P. R. China
| | - Jiapeng Huang
- Department of Thyroid SurgeryThe First Hospital of China Medical UniversityShenyang110801P. R. China
| | - Jinyuan Shi
- Department of Thyroid SurgeryGeneral SurgeryQilu Hospital of Shandong UniversityJinan250012P. R. China
| | - Wei Sun
- Department of Thyroid SurgeryThe First Hospital of China Medical UniversityShenyang110801P. R. China
| | - Xiaoyu Ji
- Department of Thyroid SurgeryThe First Hospital of China Medical UniversityShenyang110801P. R. China
| | - Yuang Chang
- Department of Thyroid SurgeryThe First Hospital of China Medical UniversityShenyang110801P. R. China
| | - Bing Ma
- Department of Clinical Epidemiology and Evidence‐based MedicineThe First Hospital of China Medical UniversityShenyang110801P. R. China
| | - Ping Zhang
- Department of Thyroid SurgeryThe First Hospital of China Medical UniversityShenyang110801P. R. China
| | - Yuanyuan Yan
- Department of PharmacologySchool of PharmacyChina Medical UniversityShenyang110122P. R. China
| | - Hao Zhang
- Department of Thyroid SurgeryThe First Hospital of China Medical UniversityShenyang110801P. R. China
| |
Collapse
|
2
|
Wang C, Zai W, Zhao K, Li Y, Shi B, Wu M, Zhou X, Kozlowski M, Zhang X, Fang Z, Yuan Z. Potential role of liver-resident CD3 + macrophages in HBV clearance in a mouse hepatitis B model. JHEP Rep 2025; 7:101323. [PMID: 40143948 PMCID: PMC11937660 DOI: 10.1016/j.jhepr.2024.101323] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Revised: 12/18/2024] [Accepted: 12/23/2024] [Indexed: 03/28/2025] Open
Abstract
Background & Aims Chronic HBV infection usually causes cirrhosis and hepatocellular carcinoma. Comparative investigations of acute and chronic HBV cases would help determine the immune responses crucial for viral clearance. Methods A fast-cleared HBV mouse model was established in Alb-Cre mice via hydrodynamic injection of HBV plasmid, while persistent HBV model mice were generated via recombinant covalently closed circular DNA-adeno-associated virus 8 infection. The single-cell transcriptomes of CD45+ intrahepatic non-parenchymal cells from these mice were conducted. Multiplexed immunohistochemistry and flow cytometry were used to confirm the findings from single-cell transcriptomes. Transwell, coculture, and adoptive transfer experiments were performed to study the generation and functions of macrophages. Results Twenty-four clusters of immune cells were identified. Myeloid cells, including granulocytes, monocytes, and dendritic cells, are activated early in HBV fast-cleared mice. Significantly, a cluster of CD3+ macrophages was found in the viral clearance phase, which was confirmed in liver tissue from five acute patients with HBV. These cells highly expressed CXCL1, tumor necrosis factor alpha, and HBsAg-specific T cell receptors. The transwell assay revealed that CD3+ macrophages originate from macrophages (n = 6). T cells and anti-HBsAg antibodies are indispensable for their differentiation, which was further confirmed in T- and/or B-cell-deficient mice. Interestingly, these CD3+ macrophages capable of killing peptide-loaded hepatocytes and engulfing IgG-coated beads were persistently detectable in the mouse liver for 10 weeks after HBV clearance. The expression levels of CD5L and Bcl2, two classical antiapoptotic proteins, increased (p <0.001), suggesting that the CD3+ macrophages are long-term resident populations. Finally, adoptive transfer of CD3+ macrophages accelerated HBV clearance in mice (n = 5, p <0.01). Conclusions We identified long-term polyfunctional CD3+ macrophages residing in HBV fast-cleared livers that could help elucidate the immune responses involved in eliminating HBV. Impact and implications The liver is a special organ with unique immune characteristics and tolerance to foodborne antigens. Chronic infections can develop in newborns after exposure to HBV; however, acute infections usually occur in adults, indicating that immune cells in the liver tissue microenvironment can also effectively fight against the virus. Nevertheless, the mechanisms involved in acute HBV infection have rarely been studied. In this study, we identified a macrophage population with both T cell and macrophage characteristics in the livers of acute HBV model mice and revealed that these macrophages play important roles in HBV clearance. Moreover, we confirmed that this population is derived from macrophages in the presence of virus-specific T cells and antibodies. This finding highlights the complexity of antiviral immune responses in liver microenvironments.
Collapse
Affiliation(s)
- Cong Wang
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), Research Unit of Cure of Chronic Hepatitis B Virus Infection (CAMS), Shanghai Frontiers Science Center of Pathogenic Microbes and Infection, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
- Shanghai Public Health Clinical Center, Shanghai Medical College of Fudan University, Shanghai, China
| | - Wenjing Zai
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), Research Unit of Cure of Chronic Hepatitis B Virus Infection (CAMS), Shanghai Frontiers Science Center of Pathogenic Microbes and Infection, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
| | - Kuangjie Zhao
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), Research Unit of Cure of Chronic Hepatitis B Virus Infection (CAMS), Shanghai Frontiers Science Center of Pathogenic Microbes and Infection, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yaming Li
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), Research Unit of Cure of Chronic Hepatitis B Virus Infection (CAMS), Shanghai Frontiers Science Center of Pathogenic Microbes and Infection, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
| | - Bisheng Shi
- Shanghai Public Health Clinical Center, Shanghai Medical College of Fudan University, Shanghai, China
| | - Min Wu
- Shanghai Public Health Clinical Center, Shanghai Medical College of Fudan University, Shanghai, China
| | - Xiaohui Zhou
- Shanghai Public Health Clinical Center, Shanghai Medical College of Fudan University, Shanghai, China
| | - Maya Kozlowski
- Shanghai Public Health Clinical Center, Shanghai Medical College of Fudan University, Shanghai, China
| | - Xiaonan Zhang
- Shanghai Public Health Clinical Center, Shanghai Medical College of Fudan University, Shanghai, China
| | - Zhong Fang
- Liver Cancer Institute of Zhongshan Hospital and Key Laboratory of Carcinogenesis and Cancer Invasion (Ministry of Education), Fudan University, Shanghai, China
| | - Zhenghong Yuan
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), Research Unit of Cure of Chronic Hepatitis B Virus Infection (CAMS), Shanghai Frontiers Science Center of Pathogenic Microbes and Infection, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
| |
Collapse
|
3
|
Brooke G, Wendel S, Banerjee A, Wallace N. Opportunities to advance cervical cancer prevention and care. Tumour Virus Res 2024; 18:200292. [PMID: 39490532 PMCID: PMC11566706 DOI: 10.1016/j.tvr.2024.200292] [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: 09/13/2024] [Revised: 10/21/2024] [Accepted: 10/22/2024] [Indexed: 11/05/2024] Open
Abstract
Cervical cancer (CaCx) is a major public health issue, with over 600,000 women diagnosed annually. CaCx kills someone every 90 s, mostly in low- and middle-income countries. There are effective yet imperfect mechanisms to prevent CaCx. Since human papillomavirus (HPV) infections cause most CaCx, they can be prevented by vaccination. Screening methodologies can identify premalignant lesions and allow interventions before a CaCx develops. However, these tools are less feasible in resource-poor environments. Additionally, current screening modalities cannot triage lesions based on their relative risk of progression, which results in overtreatment. CaCx care relies heavily on genotoxic agents that cause severe side effects. This review discusses ways that recent technological advancements could be leveraged to improve CaCx care and prevention.
Collapse
Affiliation(s)
- Grant Brooke
- Division of Biology, Kansas State University, Manhattan, KS 66506, USA
| | - Sebastian Wendel
- Department of Kinesiology, Kansas State University, Manhattan, KS 66506, USA
| | - Abhineet Banerjee
- Division of Biology, Kansas State University, Manhattan, KS 66506, USA
| | - Nicholas Wallace
- Department of Kinesiology, Kansas State University, Manhattan, KS 66506, USA.
| |
Collapse
|
4
|
Conde-Lopez C, Marripati D, Elkabets M, Hess J, Kurth I. Unravelling the Complexity of HNSCC Using Single-Cell Transcriptomics. Cancers (Basel) 2024; 16:3265. [PMID: 39409886 PMCID: PMC11475296 DOI: 10.3390/cancers16193265] [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: 07/17/2024] [Revised: 09/23/2024] [Accepted: 09/23/2024] [Indexed: 10/20/2024] Open
Abstract
BACKGROUND/OBJECTIVES Head and neck squamous cell carcinoma (HNSCC) is a highly heterogeneous and the most common form of head and neck cancer, posing significant challenges for disease management. The objective of this review is to assess the utility of single-cell RNA sequencing (scRNAseq) in addressing these challenges by enabling a detailed characterization of the tumor microenvironment (TME) at the cellular level. METHODS This review compiles and analyzes current strategies that utilize scRNAseq and other single-cell technologies in HNSCC research. RESULTS For HNSCC etiology, scRNAseq allows for the construction of cellular atlases, characterization of different cell types, and investigation of genes and processes involved in cancer initiation, development, and progression within the TME. In terms of HNSCC diagnosis and prognosis, the resolution offered by scRNAseq enables the identification of cell type-specific signatures, enhancing prognostic models and disease stratifiers for patient outcome assessments. Regarding HNSCC treatment, scRNAseq provides insights into cellular responses to various treatments, including radiotherapy, chemotherapy, and immunotherapy, contributing to a better understanding of treatment efficacy and patient outcomes. CONCLUSIONS This review highlights the contributions of scRNAseq to HNSCC research, addressing its cellular and biological complexity, and emphasizes its potential for advancing research and clinical practice in other cancer types.
Collapse
Affiliation(s)
- Cristina Conde-Lopez
- Division Radiooncology/Radiobiology, German Cancer Research Center (DKFZ) Heidelberg, 69120 Heidelberg, Germany; (J.H.); (I.K.)
| | - Divyasree Marripati
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel; (D.M.); (M.E.)
- Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel
| | - Moshe Elkabets
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel; (D.M.); (M.E.)
- Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel
| | - Jochen Hess
- Division Radiooncology/Radiobiology, German Cancer Research Center (DKFZ) Heidelberg, 69120 Heidelberg, Germany; (J.H.); (I.K.)
- Department of Otorhinolaryngology, Head and Neck Surgery, Heidelberg University Hospital, 69120 Heidelberg, Germany
| | - Ina Kurth
- Division Radiooncology/Radiobiology, German Cancer Research Center (DKFZ) Heidelberg, 69120 Heidelberg, Germany; (J.H.); (I.K.)
| |
Collapse
|
5
|
Xu L, Li W, Liu D, Cao J, Ge J, Liu X, Wang Y, Teng Y, Liu P, Guo X, He C, Liu M, Tian L. ANXA3-Rich Exosomes Derived from Tumor-Associated Macrophages Regulate Ferroptosis and Lymphatic Metastasis of Laryngeal Squamous Cell Carcinoma. Cancer Immunol Res 2024; 12:614-630. [PMID: 38393971 DOI: 10.1158/2326-6066.cir-23-0595] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2023] [Revised: 12/02/2023] [Accepted: 02/20/2024] [Indexed: 02/25/2024]
Abstract
Tumor-associated macrophages (TAM) induce immunosuppression in laryngeal squamous cell carcinoma (LSCC). The interaction between LSCC cells and TAMs affects the progression of laryngeal cancer through exosomes, but the underlying molecular mechanism remains unclear. Proteomics analysis of TAMs isolated from human laryngeal tumor tissues obtained from patients with confirmed lymphatic metastasis revealed an upregulation of annexin A3 (ANXA3). In TAMs, ANXA3 promoted macrophages to polarize to an M2-like phenotype by activating the AKT-GSK3β-β-catenin pathway. In addition, ANXA3-rich exosomes derived from TAMs inhibited ferroptosis in laryngeal cancer cells through an ATF2-CHAC1 axis, and this process was associated with lymphatic metastasis. Mechanistically, ANXA3 in exosomes inhibited the ubiquitination of ATF2, whereas ATF2 acted as a transcription factor to regulate the expression of CHAC1, thus inhibiting ferroptosis in LSCC cells. These data indicate that abnormal ANXA3 expression can drive TAM reprogramming and promote an immunosuppressive microenvironment in LSCC. Meanwhile, ANXA3-rich exosomes inhibit ferroptosis of LSCC cells and promote lymphatic metastasis, thus promoting tumor progression.
Collapse
Affiliation(s)
- Licheng Xu
- Department of Otorhinolaryngology, Head and Neck Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
- The Key Laboratory of Myocardial Ischemia, Ministry of Education, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Wenjing Li
- Department of Otorhinolaryngology, Head and Neck Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
- The Key Laboratory of Myocardial Ischemia, Ministry of Education, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Danxi Liu
- Department of Pancreatic and Biliary Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
- The Key Laboratory of Hepatosplenic Surgery Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Jing Cao
- Department of Otorhinolaryngology, Head and Neck Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Jingchun Ge
- Department of Otorhinolaryngology, Head and Neck Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Xinyu Liu
- Department of Otorhinolaryngology, Head and Neck Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yue Wang
- Department of Otorhinolaryngology, Head and Neck Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yujian Teng
- Department of Otorhinolaryngology, Head and Neck Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Pengyan Liu
- Department of Otorhinolaryngology, Head and Neck Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Xinyue Guo
- Department of Otorhinolaryngology, Head and Neck Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Chen He
- Department of Otorhinolaryngology, Head and Neck Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
- The Key Laboratory of Myocardial Ischemia, Ministry of Education, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Ming Liu
- Department of Otorhinolaryngology, Head and Neck Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Linli Tian
- Department of Otorhinolaryngology, Head and Neck Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| |
Collapse
|
6
|
Kurkalang S, Roy S, Acharya A, Mazumder P, Mazumder S, Patra S, Ghosh S, Sarkar S, Kundu S, Biswas NK, Ghose S, Majumder PP, Maitra A. Single-cell transcriptomic analysis of gingivo-buccal oral cancer reveals two dominant cellular programs. Cancer Sci 2023; 114:4732-4746. [PMID: 37792582 PMCID: PMC10728019 DOI: 10.1111/cas.15979] [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: 06/23/2023] [Revised: 09/02/2023] [Accepted: 09/13/2023] [Indexed: 10/06/2023] Open
Abstract
Oral squamous cell carcinoma of the gingivo-buccal region (OSCC-GB) is the most common cancer among men in India, and is associated with poor prognosis and frequent recurrence. Cellular heterogeneity in OSCC-GB was investigated by single-cell RNA sequencing of tumors derived from the oral cavity of 12 OSCC-GB patients, 3 of whom had concomitant presence of a precancerous lesion (oral submucous fibrosis [OSMF]). Unique malignant cell types, features, and phenotypic shifts in the stromal cell population were identified in oral tumors with associated submucous fibrosis. Expression levels of FOS, ATP1A, and DUSP1 provided robust discrimination between tumors with or without the concomitant presence of OSMF. Malignant cell populations shared between tumors with and without OSMF were enriched with the expression of partial epithelial-mesenchymal transition (pEMT) or fetal cell type signatures indicative of two dominant cellular programs in OSCC-GB-pEMT and fetal cellular reprogramming. Malignant cells exhibiting fetal cellular and pEMT programs were enriched with the expression of immune-related pathway genes known to be involved in antitumor immune response. In the tumor microenvironment, higher infiltration of immune cells than the stromal cells was observed. The T cell population was large in tumors and diverse subtypes of T cells with varying levels of infiltration were found. We also detected double-negative PLCG2+ T cells and cells with intermediate M1-M2 macrophage polarization. Our findings shed light on unique aspects of cellular heterogeneity and cell states in OSCC-GB.
Collapse
Affiliation(s)
| | - Sumitava Roy
- National Institute of Biomedical GenomicsKalyaniIndia
- Regional Centre for BiotechnologyFaridabadIndia
| | - Arunima Acharya
- National Institute of Biomedical GenomicsKalyaniIndia
- Regional Centre for BiotechnologyFaridabadIndia
| | - Paramita Mazumder
- Department of Oral PathologyDr. R. Ahmed Dental College and HospitalKolkataIndia
| | | | - Subrata Patra
- National Institute of Biomedical GenomicsKalyaniIndia
| | - Shekhar Ghosh
- National Institute of Biomedical GenomicsKalyaniIndia
| | | | - Sudip Kundu
- National Institute of Biomedical GenomicsKalyaniIndia
| | | | - Sandip Ghose
- Department of Oral PathologyDr. R. Ahmed Dental College and HospitalKolkataIndia
| | | | | |
Collapse
|
7
|
HELLER GERWIN, FUEREDER THORSTEN, GRANDITS ALEXANDERMICHAEL, WIESER ROTRAUD. New perspectives on biology, disease progression, and therapy response of head and neck cancer gained from single cell RNA sequencing and spatial transcriptomics. Oncol Res 2023; 32:1-17. [PMID: 38188682 PMCID: PMC10767240 DOI: 10.32604/or.2023.044774] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Accepted: 10/12/2023] [Indexed: 01/09/2024] Open
Abstract
Head and neck squamous cell carcinoma (HNSCC) is one of the most frequent cancers worldwide. The main risk factors are consumption of tobacco products and alcohol, as well as infection with human papilloma virus. Approved therapeutic options comprise surgery, radiation, chemotherapy, targeted therapy through epidermal growth factor receptor inhibition, and immunotherapy, but outcome has remained unsatisfactory due to recurrence rates of ~50% and the frequent occurrence of second primaries. The availability of the human genome sequence at the beginning of the millennium heralded the omics era, in which rapid technological progress has advanced our knowledge of the molecular biology of malignant diseases, including HNSCC, at an unprecedented pace. Initially, microarray-based methods, followed by approaches based on next-generation sequencing, were applied to study the genetics, epigenetics, and gene expression patterns of bulk tumors. More recently, the advent of single-cell RNA sequencing (scRNAseq) and spatial transcriptomics methods has facilitated the investigation of the heterogeneity between and within different cell populations in the tumor microenvironment (e.g., cancer cells, fibroblasts, immune cells, endothelial cells), led to the discovery of novel cell types, and advanced the discovery of cell-cell communication within tumors. This review provides an overview of scRNAseq, spatial transcriptomics, and the associated bioinformatics methods, and summarizes how their application has promoted our understanding of the emergence, composition, progression, and therapy responsiveness of, and intercellular signaling within, HNSCC.
Collapse
Affiliation(s)
- GERWIN HELLER
- Division of Oncology, Department of Medicine I, Medical University of Vienna, Vienna, 1090, Austria
| | - THORSTEN FUEREDER
- Division of Oncology, Department of Medicine I, Medical University of Vienna, Vienna, 1090, Austria
| | | | - ROTRAUD WIESER
- Division of Oncology, Department of Medicine I, Medical University of Vienna, Vienna, 1090, Austria
- Ludwig Boltzmann Institute for Hematology and Oncology, Medical University of Vienna, Vienna, 1090, Austria
| |
Collapse
|
8
|
Huo X, Ma S, Wang C, Song L, Yao B, Zhu S, Li P, Wang L, Wu Z, Wang K. Unravelling the role of immune cells and FN1 in the recurrence and therapeutic process of skull base chordoma. Clin Transl Med 2023; 13:e1429. [PMID: 37784253 PMCID: PMC10545891 DOI: 10.1002/ctm2.1429] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Revised: 08/23/2023] [Accepted: 09/25/2023] [Indexed: 10/04/2023] Open
Abstract
BACKGROUND Skull base chordoma is a rare and aggressive tumour of the bone that has a high likelihood of recurrence. The fundamental differences in single cells between primary and recurrent lesions remain poorly understood, impeding development of effective treatment approaches. METHODS To obtain an understanding of the differences in single cells between primary and recurrent chordomas, we performed single-cell RNA sequencing and T-cell/B-cell receptor (BCR) sequencing. This allowed us to delineate the differences between the two types of tumour cells, tumour-infiltrating lymphocytes, myeloid cells, fibroblasts and B cells. Copy number variants (CNVs) were detected and compared between the tumour types to assess heterogeneity. Selected samples were subjected to immunohistochemistry to validate protein expression. Fluorescence in situ hybridisation experiments, Transwell assays and xenograft mouse models helped verify the role of fibronectin 1 (FN1) in chordoma. RESULTS Promoting natural killer (NK) cell and CD8_GZMK T-cell function or inhibiting the transformation of CD8_GZMK T cells to CD8_ZNF683 T cells and promoting the transformation of natural killer T (NKT) cells to NK cells are promising strategies for preventing chordoma recurrence. Additionally, inhibiting the M2-like activity of tumour-associated macrophages (TAMs) could be an effective approach. Antigen-presenting cancer-associated fibroblasts (apCAFs) and dendritic cells (DCs) with high enrichment of the antigen-presenting signature were enriched in primary chordomas. There were fewer plasma cells and BCR clonotypes in recurrent chordomas. Remarkably, FN1 was upregulated, had more CNVs, and was more highly secreted by tumours, macrophages, CD4 T cells, CD8 T cells and fibroblasts in recurrent chordoma than in primary chordoma. Finally, FN1 enhanced the invasion and proliferation of chordomas in vivo and in vitro. CONCLUSION Our comprehensive picture of the microenvironment of primary and recurrent chordomas provides deep insights into the mechanisms of chordoma recurrence. FN1 is an important target for chordoma therapy.
Collapse
Affiliation(s)
- Xulei Huo
- Department of NeurosurgeryBeijing Tiantan HospitalCapital Medical UniversityBeijingChina
| | - Sihan Ma
- Department of NeurosurgeryBeijing Tiantan HospitalCapital Medical UniversityBeijingChina
| | - Can Wang
- Department of Neuro‐oncologyCancer CenterBeijing Tiantan HospitalCapital Medical UniversityBeijingChina
| | - Lairong Song
- Department of NeurosurgeryBeijing Tiantan HospitalCapital Medical UniversityBeijingChina
| | - Bohan Yao
- Department of NeurosurgeryBeijing Tiantan HospitalCapital Medical UniversityBeijingChina
| | - Sipeng Zhu
- Department of NeurosurgeryBeijing Tiantan HospitalCapital Medical UniversityBeijingChina
| | - Peiran Li
- Department of NeurosurgeryBeijing Tiantan HospitalCapital Medical UniversityBeijingChina
| | - Liang Wang
- Department of NeurosurgeryBeijing Tiantan HospitalCapital Medical UniversityBeijingChina
| | - Zhen Wu
- Department of NeurosurgeryBeijing Tiantan HospitalCapital Medical UniversityBeijingChina
| | - Ke Wang
- Department of NeurosurgeryBeijing Tiantan HospitalCapital Medical UniversityBeijingChina
| |
Collapse
|
9
|
Gong X, Chi H, Xia Z, Yang G, Tian G. Advances in HPV-associated tumor management: Therapeutic strategies and emerging insights. J Med Virol 2023; 95:e28950. [PMID: 37465863 DOI: 10.1002/jmv.28950] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Revised: 06/23/2023] [Accepted: 06/29/2023] [Indexed: 07/20/2023]
Abstract
With the rapid increase in the incidence of cervical cancer, anal cancer and other cancers, human papillomavirus (HPV) infection has become a growing concern. Persistent infection with high-risk HPV is a major cause of malignant tumors. In addition, microbiota and viruses such as human immunodeficiency virus, herpes simplex virus, and Epstein-Barr virus are closely associated with HPV infection. The limited effectiveness of existing treatments for HPV-associated tumors and the high rates of recurrence and metastasis in patients create an urgent need for novel and effective approaches. In recent years, HPV vaccine coverage has increased and can reduce the incidence of serious adverse events. Overall, this article provides a comprehensive overview of HPV biology, microbiome, and other viral interactions in cancer development, highlighting the need for a more comprehensive approach to cancer prevention and treatment. Current and emerging HPV-related cancer control and treatment strategies are also further explored.
Collapse
Affiliation(s)
- Xiangjin Gong
- Department of Sports Rehabilitation, Southwest Medical University, Luzhou, China
| | - Hao Chi
- Department of Clinical Medicine, Southwest Medical University, Luzhou, China
| | - Zhijia Xia
- Department of General, Visceral, and Transplant Surgery, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Guanhu Yang
- Department of Specialty Medicine, Ohio University, Athens, Ohio, USA
| | - Gang Tian
- Department of Laboratory Medicine, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| |
Collapse
|
10
|
Wang L, Wang D, Zhang T, Ma Y, Tong X, Fan H. The role of immunometabolism in macrophage polarization and its impact on acute lung injury/acute respiratory distress syndrome. Front Immunol 2023; 14:1117548. [PMID: 37020557 PMCID: PMC10067752 DOI: 10.3389/fimmu.2023.1117548] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Accepted: 03/03/2023] [Indexed: 04/07/2023] Open
Abstract
Lung macrophages constitute the first line of defense against airborne particles and microbes and are key to maintaining pulmonary immune homeostasis. There is increasing evidence suggesting that macrophages also participate in the pathogenesis of acute lung injury (ALI)/acute respiratory distress syndrome (ARDS), including the modulation of inflammatory responses and the repair of damaged lung tissues. The diversity of their functions may be attributed to their polarized states. Classically activated or inflammatory (M1) macrophages and alternatively activated or anti-inflammatory (M2) macrophages are the two main polarized macrophage phenotypes. The precise regulatory mechanism of macrophage polarization is a complex process that is not completely understood. A growing body of literature on immunometabolism has demonstrated the essential role of immunometabolism and its metabolic intermediates in macrophage polarization. In this review, we summarize macrophage polarization phenotypes, the role of immunometabolism, and its metabolic intermediates in macrophage polarization and ALI/ARDS, which may represent a new target and therapeutic direction.
Collapse
Affiliation(s)
- Lian Wang
- Department of Respiratory and Critical Care Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Dongguang Wang
- Department of Respiratory and Critical Care Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Tianli Zhang
- Department of Respiratory and Critical Care Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Yao Ma
- Department of Geriatrics and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Xiang Tong
- Department of Respiratory and Critical Care Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Hong Fan
- Department of Respiratory and Critical Care Medicine, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|