1
|
Hatten H, Colyn L, Volkert I, Gaßler N, Lammers T, Hofmann U, Hengstler JG, Schneider KM, Trautwein C. Loss of Toll-like receptor 9 protects from hepatocellular carcinoma in murine models of chronic liver disease. Biochim Biophys Acta Mol Basis Dis 2024; 1870:167321. [PMID: 38943920 DOI: 10.1016/j.bbadis.2024.167321] [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: 03/21/2024] [Revised: 06/19/2024] [Accepted: 06/20/2024] [Indexed: 07/01/2024]
Abstract
BACKGROUND & AIMS Toll-like receptor 9 (Tlr9) is a pathogen recognition receptor detecting unmethylated DNA derivatives of pathogens and damaged host cells. It is therefore an important modulator of innate immunity. Here we investigated the role of Tlr9 in fibrogenesis and progression of hepatocellular carcinoma in chronic liver disease. MATERIALS AND METHODS We treated mice with a constitutive deletion of Tlr9 (Tlr9-/-) with DEN/CCl4 for 24 weeks. As a second model, we used hepatocyte-specific Nemo knockout (NemoΔhepa) mice and generated double knockout (NemoΔhepaTlr9-/-) animals. RESULTS We show that Tlr9 is in the liver primarily expressed in Kupffer cells, suggesting a key role of Tlr9 in intercellular communication during hepatic injury. Tlr9 deletion resulted in reduced liver fibrosis as well as tumor burden. We observed down-regulation of hepatic stellate cell activation and consequently decreased collagen production in both models. Tlr9 deletion was associated with decreased apoptosis and compensatory proliferation of hepatocytes, modulating the initiation and progression of hepatocarcinogenesis. These findings were accompanied by a decrease in interferon-β and an increase in chemokines having an anti-tumoral effect. CONCLUSIONS Our data define Tlr9 as an important receptor involved in fibrogenesis, but also in the initiation and progression of hepatocellular carcinoma during chronic liver diseases.
Collapse
Affiliation(s)
- Hannes Hatten
- University Hospital RWTH Aachen, Department of Internal Medicine III, Aachen, Germany
| | - Leticia Colyn
- University Hospital RWTH Aachen, Department of Internal Medicine III, Aachen, Germany.
| | - Ines Volkert
- University Hospital RWTH Aachen, Department of Internal Medicine III, Aachen, Germany
| | - Nikolaus Gaßler
- Institute of Forensic Medicine, Section Pathology, University Hospital of Jena, Jena, Germany
| | - Twan Lammers
- University Hospital RWTH Aachen, Institute for Experimental Molecular Imaging (ExMI), Aachen, Germany
| | - Ute Hofmann
- Dr. Margarete Fischer-Bosch-Institute of Clinical Pharmacology, Stuttgart, Germany; University of Tuebingen, Tuebingen, Germany
| | - Jan G Hengstler
- Department of Toxicology, Leibniz Research Centre for Working Environment and Human Factors (IfADo), Dortmund, Germany
| | - Kai Markus Schneider
- University Hospital RWTH Aachen, Department of Internal Medicine III, Aachen, Germany
| | - Christian Trautwein
- University Hospital RWTH Aachen, Department of Internal Medicine III, Aachen, Germany; Department of Toxicology, Leibniz Research Centre for Working Environment and Human Factors (IfADo), Dortmund, Germany.
| |
Collapse
|
2
|
Yi Q, Zhu G, Zhu W, Wang J, Ouyang X, Yang K, Fan Y, Zhong J. LINC01094: A key long non-coding RNA in the regulation of cancer progression and therapeutic targets. Heliyon 2024; 10:e37527. [PMID: 39309878 PMCID: PMC11415682 DOI: 10.1016/j.heliyon.2024.e37527] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2024] [Revised: 07/25/2024] [Accepted: 09/04/2024] [Indexed: 09/25/2024] Open
Abstract
LINC01094 is a long non-coding RNA that plays a crucial role in cancer progression by modulating key signaling pathways, such as PI3K/AKT, Wnt/β-catenin and TGF-β Signaling Pathway Feedback Loop. In this review we summarize the recent research on the functional mechanisms of LINC01094 in various cancers, including its impact on tumor growth, metastasis, and resistance to therapy. We also discuss the therapeutic potential of targeting LINC01094 and highlight the current strategies and challenges in this area. Perspectives on future development of LINC01094-based therapies are also provided.
Collapse
Affiliation(s)
- Qiang Yi
- The First Clinical Medical College, Gannan Medical University, Ganzhou, Jiangxi Province, 341000, China
| | - Gangfeng Zhu
- The First Clinical Medical College, Gannan Medical University, Ganzhou, Jiangxi Province, 341000, China
| | - Weijian Zhu
- The First Clinical Medical College, Gannan Medical University, Ganzhou, Jiangxi Province, 341000, China
| | - Jiaqi Wang
- The First Clinical Medical College, Gannan Medical University, Ganzhou, Jiangxi Province, 341000, China
| | - Xinting Ouyang
- The First Clinical Medical College, Gannan Medical University, Ganzhou, Jiangxi Province, 341000, China
| | - Kuan Yang
- The First Clinical Medical College, Gannan Medical University, Ganzhou, Jiangxi Province, 341000, China
| | - Yu Fan
- The First Clinical Medical College, Gannan Medical University, Ganzhou, Jiangxi Province, 341000, China
| | - Jinghua Zhong
- Department of Oncology, The First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi Province, 341000, China
| |
Collapse
|
3
|
Huang Y, Peng M, Yu W, Li H. Activation of Wnt/β-catenin signaling promotes immune evasion via the β-catenin/IKZF1/CCL5 axis in hepatocellular carcinoma. Int Immunopharmacol 2024; 138:112534. [PMID: 38941667 DOI: 10.1016/j.intimp.2024.112534] [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: 05/08/2024] [Revised: 06/10/2024] [Accepted: 06/18/2024] [Indexed: 06/30/2024]
Abstract
Immune checkpoint therapy (ICT) has been shown to produce durable responses in various cancer patients. However, its efficacy is notably limited in hepatocellular carcinoma (HCC), with only a small percentage of patients responding positively to treatment. The mechanism underlying resistance to ICT in HCC remains poorly understood. Here, we showed that combination treatment of ICG-001, an inhibitor of the Wnt/β-catenin signaling pathway, with anti-PD-1 antibody effectively suppresses tumor growth and promotes the infiltration of immune cells such as DCs and CD8+ T cells in the tumor microenvironment (TME). By inhibiting the activity of β-catenin and blocking its binding to the transcription factor IKAROS family zinc finger 1 (IKZF1), ICG-001 upregulated the expression of CCL5. Moreover, IKZF1 regulated the activity of the CCL5 promoter and its endogenous expression. Through inhibition of the WNT/β-catenin signaling pathway, upregulation of the expression of CCL5 was achieved, which subsequently recruited more DCs into the TME via C-C motif chemokine receptor 5 (CCR5). This, in turn, resulted in an increase in the infiltration of CD8+ T cells in the TME, thereby enhancing the antitumor immune response. Analysis of a tissue microarray derived from HCC patient samples revealed a positive correlation between survival rate and prognosis and the expression levels of CCL5/CD8. In conclusion, our findings suggest that combined application of ICG-001 and anti-PD-1 antibody exhibits significantly enhanced antitumor efficacy. Hence, combining a WNT/β-catenin signaling pathway inhibitor with anti-PD-1 therapy may be a promising treatment strategy for patients with HCC.
Collapse
Affiliation(s)
- Yamei Huang
- Department of Pathology and Pathophysiology, Medical School of Southeast University, China
| | - Min Peng
- Department of Pathogenic Biology and Immunology, Medical School of Southeast University, China
| | - Weiping Yu
- Department of Pathology and Pathophysiology, Medical School of Southeast University, China.
| | - Hui Li
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University, China.
| |
Collapse
|
4
|
Chang H, Li M, Zhang L, Li M, Ong SH, Zhang Z, Zheng J, Xu X, Zhang Y, Wang J, Liu X, Li K, Luo Y, Wang H, Miao Z, Chen X, Zha J, Yu Y. Loss of histone deubiquitinase Bap1 triggers anti-tumor immunity. Cell Oncol (Dordr) 2024:10.1007/s13402-024-00978-y. [PMID: 39141316 DOI: 10.1007/s13402-024-00978-y] [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] [Accepted: 07/26/2024] [Indexed: 08/15/2024] Open
Abstract
PURPOSE Immunotherapy using PD-L1 blockade is effective in only a small group of cancer patients, and resistance is common. This emphasizes the importance of understanding the mechanisms of cancer immune evasion and resistance. METHODS A genome-scale CRISPR-Cas9 screen identified Bap1 as a regulator of PD-L1 expression. To measure tumor size and survival, tumor cells were subcutaneously injected into both syngeneic WT mice and immunocompromised mice. The phenotypic and transcriptional characteristics of Bap1-deleted tumors were examined using flow cytometry, RNA-seq, and CUT&Tag-seq analysis. RESULTS We found that loss of histone deubiquitinase Bap1 in cancer cells activates a cDC1-CD8+ T cell-dependent anti-tumor immunity. The absence of Bap1 leads to an increase in genes associated with anti-tumor immune response and a decrease in genes related to immune evasion. As a result, the tumor microenvironment becomes inflamed, with more cDC1 cells and effector CD8+ T cells, but fewer neutrophils and regulatory T cells. We also found that the elimination of Bap1-deleted tumors depends on the tumor MHCI molecule and Fas-mediated CD8+ T cell cytotoxicity. Our analysis of TCGA data further supports these findings, showing a reverse correlation between BAP1 expression and mRNA signatures of activated DCs and T-cell cytotoxicity in various human cancers. CONCLUSION The histone deubiquitinase Bap1 could be used as a biomarker for tumor stratification and as a potential therapeutic target for cancer immunotherapies.
Collapse
Affiliation(s)
- Hong Chang
- Department of Hematology, Tongji Hospital, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, 1239 Siping Road, Shanghai, 200092, China
| | - Mingxia Li
- Department of Hematology, Tongji Hospital, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, 1239 Siping Road, Shanghai, 200092, China
| | - Linlin Zhang
- Department of Hematology, Tongji Hospital, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, 1239 Siping Road, Shanghai, 200092, China
| | - Meng Li
- Wellcome Sanger Institute, Hinxton, Cambridge, CB10 1HH, UK
| | - Swee Hoe Ong
- Wellcome Sanger Institute, Hinxton, Cambridge, CB10 1HH, UK
| | - Zhiwei Zhang
- Department of Hematology, Tongji Hospital, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, 1239 Siping Road, Shanghai, 200092, China
| | - Jie Zheng
- Department of Hematology, Tongji Hospital, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, 1239 Siping Road, Shanghai, 200092, China
| | - Xiang Xu
- Department of Hematology, Tongji Hospital, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, 1239 Siping Road, Shanghai, 200092, China
| | - Yu Zhang
- Department of Hematology, Tongji Hospital, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, 1239 Siping Road, Shanghai, 200092, China
| | - Jing Wang
- Department of Hematology, Tongji Hospital, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, 1239 Siping Road, Shanghai, 200092, China
| | - Xingjie Liu
- Department of Hematology, Tongji Hospital, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, 1239 Siping Road, Shanghai, 200092, China
| | - Kairui Li
- Department of Hematology, Tongji Hospital, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, 1239 Siping Road, Shanghai, 200092, China
| | - Yao Luo
- Department of Hematology, Tongji Hospital, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, 1239 Siping Road, Shanghai, 200092, China
| | - Haiyun Wang
- Department of Hematology, Tongji Hospital, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, 1239 Siping Road, Shanghai, 200092, China
| | - Zhichao Miao
- Translational Research Institute of Brain and Brain-Like Intelligence, Department of Anesthesiology, Shanghai Fourth People's Hospital Affiliated to Tongji University School of Medicine, Shanghai, 200081, China
- GMU-GIBH Joint School of Life Sciences, The Guangdong-Hong Kong-Macau Joint Laboratory for Cell Fate Regulation and Diseases, Guangzhou Laboratory, Guangzhou Medical University, Guangzhou, China
| | - Xi Chen
- Department of Biology, Southern University of Science and Technology, 1088 Xueyuan Avenue, Shenzhen, 518055, China
| | - Jie Zha
- Department of Hematology, The First Affiliated Hospital of Xiamen University and Institute of Hematology, School of Medicine, Xiamen University, Xiamen, China.
- Key Laboratory of Xiamen for Diagnosis and Treatment of Hematological Malignancy, Xiamen, China.
| | - Yong Yu
- Department of Hematology, Tongji Hospital, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, 1239 Siping Road, Shanghai, 200092, China.
| |
Collapse
|
5
|
Snyder M, Wang Z, Lara B, Fimbres J, Pichardo T, Mazzilli S, Khan MM, Duggineni VK, Monti S, Sherr DH. The Aryl Hydrocarbon Receptor Controls IFNγ-Induced Immune Checkpoints PD-L1 and IDO via the JAK/STAT Pathway in Lung Adenocarcinoma. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.12.607602. [PMID: 39185148 PMCID: PMC11343147 DOI: 10.1101/2024.08.12.607602] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 08/27/2024]
Abstract
While immunotherapy has shown efficacy in lung adenocarcinoma (LUAD) patients, many respond only partially or not at all. One limitation in improving outcomes is the lack of a complete understanding of immune checkpoint regulation. Here, we investigated a possible link between an environmental chemical receptor implicated in lung cancer and immune regulation, (the aryl hydrocarbon receptor/AhR), a known but counterintuitive mediator of immunosuppression (IFNγ), and regulation of two immune checkpoints (PD-L1 and IDO). AhR gene-edited LUAD cell lines, a syngeneic LUAD mouse model, bulk- and scRNA sequencing of LUADs and tumor-infiltrating leukocytes were used to map out a signaling pathway leading from IFNγ through the AhR to JAK/STAT, PD-L1, IDO, and tumor-mediated immunosuppression. The data demonstrate that: 1) IFNγ activation of the JAK/STAT pathway leading to PD-L1 and IDO1 upregulation is mediated by the AhR in murine and human LUAD cells, 2) AhR-driven IDO1 induction results in the production of Kynurenine (Kyn), an AhR ligand, which likely mediates an AhR→IDO1→Kyn→AhR amplification loop, 3) transplantation of AhR-knockout LUAD cells results in long-term tumor immunity in most recipients. 4) The 23% of AhR-knockout tumors that do grow do so at a much slower pace than controls and exhibit higher densities of CD8+ T cells expressing markers of immunocompetence, increased activity, and increased cell-cell communication. The data definitively link the AhR to IFNγ-induced JAK/STAT pathway and immune checkpoint-mediated immunosuppression and support the targeting of the AhR in the context of LUAD.
Collapse
Affiliation(s)
- Megan Snyder
- Graduate Program in Genetics and Genomics, Boston University School of Medicine
| | - Zhongyan Wang
- Department of Environmental Health, Boston University School of Public Health
| | - Brian Lara
- Department of Environmental Health, Boston University School of Public Health
| | - Jocelyn Fimbres
- Department of Environmental Health, Boston University School of Public Health
| | | | | | - Mohammed Muzamil Khan
- Section of Computational Biomedicine, Boston University Chobanian & Avedisian School of Medicine
| | - Vinay K. Duggineni
- Department of Environmental Health, Boston University School of Public Health
| | - Stefano Monti
- Section of Computational Biomedicine, Boston University Chobanian & Avedisian School of Medicine
| | - David H. Sherr
- Department of Environmental Health, Boston University School of Public Health
| |
Collapse
|
6
|
Giammona A, De Vellis C, Crivaro E, Maresca L, Amoriello R, Ricci F, Anichini G, Pietrobono S, Pease DR, Fernandez-Zapico ME, Ballerini C, Stecca B. Tumor-derived GLI1 promotes remodeling of the immune tumor microenvironment in melanoma. J Exp Clin Cancer Res 2024; 43:214. [PMID: 39090759 PMCID: PMC11295348 DOI: 10.1186/s13046-024-03138-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Accepted: 07/24/2024] [Indexed: 08/04/2024] Open
Abstract
BACKGROUND Melanoma progression is based on a close interaction between cancer cells and immune cells in the tumor microenvironment (TME). Thus, a better understanding of the mechanisms controlling TME dynamics and composition will help improve the management of this dismal disease. Work from our and other groups has reported the requirement of an active Hedgehog-GLI (HH-GLI) signaling for melanoma growth and stemness. However, the role of the downstream GLI1 transcription factor in melanoma TME remains largely unexplored. METHODS The immune-modulatory activity of GLI1 was evaluated in a syngeneic B16F10 melanoma mouse model assessing immune populations by flow cytometry. Murine polymorphonuclear myeloid-derived suppressor cells (PMN-MDSCs) were differentiated from bone marrow cells and their immunosuppressive ability was assessed by inhibition of T cells. Conditioned media (CM) from GLI1-overexpressing mouse melanoma cells was used to culture PMN-MDSCs, and the effects of CM were evaluated by Transwell invasion assay and T cell inhibition. Cytokine array analysis, qPCR and chromatin immunoprecipitation were performed to explore the regulation of CX3CL1 expression by GLI1. Human monocyte-derived dendritic cells (moDCs) were cultured in CM from GLI1-silenced patient-derived melanoma cells to assess their activation and recruitment. Blocking antibodies anti-CX3CL1, anti-CCL7 and anti-CXCL8 were used for in vitro functional assays. RESULTS Melanoma cell-intrinsic activation of GLI1 promotes changes in the infiltration of immune cells, leading to accumulation of immunosuppressive PMN-MDSCs and regulatory T cells, and to decreased infiltration of dendric cells (DCs), CD8 + and CD4 + T cells in the TME. In addition, we show that ectopic expression of GLI1 in melanoma cells enables PMN-MDSC expansion and recruitment, and increases their ability to inhibit T cells. The chemokine CX3CL1, a direct transcriptional target of GLI1, contributes to PMN-MDSC expansion and recruitment. Finally, silencing of GLI1 in patient-derived melanoma cells promotes the activation of human monocyte-derived dendritic cells (moDCs), increasing cytoskeleton remodeling and invasion ability. This phenotype is partially prevented by blocking the chemokine CCL7, but not CXCL8. CONCLUSION Our findings highlight the relevance of tumor-derived GLI1 in promoting an immune-suppressive TME, which allows melanoma cells to evade the immune system, and pave the way for the design of new combination treatments targeting GLI1.
Collapse
Affiliation(s)
- Alessandro Giammona
- Core Research Laboratory - Institute for Cancer Research and Prevention (ISPRO), Viale Pieraccini 6, 50139, Florence, Italy
| | - Chiara De Vellis
- Core Research Laboratory - Institute for Cancer Research and Prevention (ISPRO), Viale Pieraccini 6, 50139, Florence, Italy
| | - Enrica Crivaro
- Core Research Laboratory - Institute for Cancer Research and Prevention (ISPRO), Viale Pieraccini 6, 50139, Florence, Italy
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Siena, Italy
| | - Luisa Maresca
- Core Research Laboratory - Institute for Cancer Research and Prevention (ISPRO), Viale Pieraccini 6, 50139, Florence, Italy
- Department of Experimental and Clinical Medicine, University of Florence, Viale Pieraccini 6, 50139, Florence, Italy
| | - Roberta Amoriello
- Department of Experimental and Clinical Medicine, University of Florence, Viale Pieraccini 6, 50139, Florence, Italy
| | - Federica Ricci
- Core Research Laboratory - Institute for Cancer Research and Prevention (ISPRO), Viale Pieraccini 6, 50139, Florence, Italy
| | - Giulia Anichini
- Core Research Laboratory - Institute for Cancer Research and Prevention (ISPRO), Viale Pieraccini 6, 50139, Florence, Italy
| | - Silvia Pietrobono
- Core Research Laboratory - Institute for Cancer Research and Prevention (ISPRO), Viale Pieraccini 6, 50139, Florence, Italy
| | - David R Pease
- Schulze Center for Novel Therapeutics, Division of Oncology Research, Department of Oncology, Mayo Clinic, Rochester, MN, 55905, USA
| | - Martin E Fernandez-Zapico
- Schulze Center for Novel Therapeutics, Division of Oncology Research, Department of Oncology, Mayo Clinic, Rochester, MN, 55905, USA
| | - Clara Ballerini
- Department of Experimental and Clinical Medicine, University of Florence, Viale Pieraccini 6, 50139, Florence, Italy
| | - Barbara Stecca
- Core Research Laboratory - Institute for Cancer Research and Prevention (ISPRO), Viale Pieraccini 6, 50139, Florence, Italy.
| |
Collapse
|
7
|
Sun D, Tan L, Chen Y, Yuan Q, Jiang K, Liu Y, Xue Y, Zhang J, Cao X, Xu M, Luo Y, Xu Z, Xu Z, Xu W, Shen M. CXCL5 impedes CD8 + T cell immunity by upregulating PD-L1 expression in lung cancer via PXN/AKT signaling phosphorylation and neutrophil chemotaxis. J Exp Clin Cancer Res 2024; 43:202. [PMID: 39034411 PMCID: PMC11264977 DOI: 10.1186/s13046-024-03122-8] [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: 04/22/2024] [Accepted: 07/09/2024] [Indexed: 07/23/2024] Open
Abstract
BACKGROUND Lung cancer remains one of the most prevalent cancer types worldwide, with a high mortality rate. Upregulation of programmed cell death protein 1 (PD-1) and its ligand (PD-L1) may represent a key mechanism for evading immune surveillance. Immune checkpoint blockade (ICB) antibodies against PD-1 or PD-L1 are therefore widely used to treat patients with lung cancer. However, the mechanisms by which lung cancer and neutrophils in the microenvironment sustain PD-L1 expression and impart stronger inhibition of CD8+ T cell function remain unclear. METHODS We investigated the role and underlying mechanism by which PD-L1+ lung cancer and PD-L1+ neutrophils impede the function of CD8+ T cells through magnetic bead cell sorting, quantitative real-time polymerase chain reaction (RT-PCR), western blotting, enzyme-linked immunosorbent assays, confocal immunofluorescence, gene silencing, flow cytometry, etc. In vivo efficacy and safety studies were conducted using (Non-obeseDiabetes/severe combined immune deficiency) SCID/NOD mice. Additionally, we collected clinical and prognostic data from 208 patients who underwent curative lung cancer resection between 2017 and 2018. RESULTS We demonstrated that C-X-C motif chemokine ligand 5 (CXCL5) is markedly overexpressed in lung cancer cells and is positively correlated with a poor prognosis in patients with lung cancer. Mechanistically, CXCL5 activates the phosphorylation of the Paxillin/AKT signaling cascade, leading to upregulation of PD-L1 expression and the formation of a positive feedback loop. Moreover, CXCL5 attracts neutrophils, compromising CD8+ T cell-dependent antitumor immunity. These PD-L1+ neutrophils aggravate CD8+ T cell exhaustion following lung cancer domestication. Combined treatment with anti-CXCL5 and anti-PD-L1 antibodies significantly inhibits tumor growth in vivo. CONCLUSIONS Our findings collectively demonstrate that CXCL5 promotes immune escape through PD-L1 upregulation in lung cancer and neutrophils chemotaxis through autocrine and paracrine mechanisms. CXCL5 may serve as a potential therapeutic target in synergy with ICBs in lung cancer immunotherapy.
Collapse
Affiliation(s)
- Dantong Sun
- Department of Thoracic and Cardiac Surgery, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, China
| | - Lipin Tan
- Department of nursing administration, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, China
| | - Yongbing Chen
- Department of Thoracic and Cardiac Surgery, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, China
| | - Qiang Yuan
- Department of interventional medicine, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, China
| | - Kanqiu Jiang
- Department of Thoracic and Cardiac Surgery, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, China
| | - Yangyang Liu
- Department of Vascular Surgery, Hospital of Zhangjiagang, Suzhou, 215600, China
| | - Yuhang Xue
- Department of Thoracic Surgery, Hospital of Yancheng, Yancheng, 224000, China
| | - Jinzhi Zhang
- Department of Thoracic and Cardiac Surgery, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, China
| | - Xianbao Cao
- Department of Thoracic and Cardiac Surgery, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, China
| | - Minzhao Xu
- Department of Thoracic and Cardiac Surgery, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, China
| | - Yang Luo
- Department of Thoracic and Cardiac Surgery, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, China
| | - Zhonghua Xu
- Department of Thoracic and Cardiac Surgery, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, China
| | - Zhonghen Xu
- Department of Thoracic and Cardiac Surgery, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, China
| | - Weihua Xu
- Department of Thoracic and Cardiac Surgery, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, China
| | - Mingjing Shen
- Department of Thoracic and Cardiac Surgery, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, China.
| |
Collapse
|
8
|
Jung H, Paust S. Chemokines in the tumor microenvironment: implications for lung cancer and immunotherapy. Front Immunol 2024; 15:1443366. [PMID: 39114657 PMCID: PMC11304008 DOI: 10.3389/fimmu.2024.1443366] [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: 06/03/2024] [Accepted: 07/01/2024] [Indexed: 08/10/2024] Open
Abstract
The tumor microenvironment (TME) is a complex interconnected network of immune cells, fibroblasts, blood vessels, and extracellular matrix surrounding the tumor. Because of its immunosuppressive nature, the TME can pose a challenge for cancer immunotherapies targeting solid tumors. Chemokines have emerged as a crucial element in enhancing the efficacy of cancer immunotherapy, playing a direct role in immune cell signaling within the TME and facilitating immune cell migration towards cancer cells. However, chemokine ligands and their receptors exhibit context-dependent diversity, necessitating evaluation of their tumor-promoting or inhibitory effects based on tumor type and immune cell characteristics. This review explores the role of chemokines in tumor immunity and metastasis in the context of the TME. We also discuss current chemokine-related advances in cancer immunotherapy research, with a particular focus on lung cancer, a common cancer with a low survival rate and limited immunotherapy options.
Collapse
Affiliation(s)
| | - Silke Paust
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, United States
| |
Collapse
|
9
|
Zhang LL, Du MY, Du X, Duan J, Yao DM, Jing J, Feng C, Song L. Correlation analysis of human papillomavirus E6/E7 mRNA detection with diagnosis, prognosis and recurrence risk in patients with cervical epithelioma. World J Clin Cases 2024; 12:4146-4153. [PMID: 39015927 PMCID: PMC11235549 DOI: 10.12998/wjcc.v12.i20.4146] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Revised: 05/13/2024] [Accepted: 05/20/2024] [Indexed: 06/30/2024] Open
Abstract
BACKGROUND Cervical intraepithelial neoplasia (CIN) is an important precursor of cervical cancer. Early detection and treatment can reduce the incidence of cervical cancer. AIM To investigate the detection rate of human papillomavirus (HPV) E6/E7 mRNA in cervical tissue of patients with different types of epithelial cell neoplasia (CIN) and its relationship with CIN progression and diagnosis. METHODS One hundred women with HPV infection detected by cervical exfoliation cytology between January 2022 and January 2023 were retrospectively selected. These patients were graded CIN based on colposcopy and cervical pathology. The positive expression rates of HPV E6/E7 mRNA and HPV [polymerase chain reaction (PCR)-reverse dot crossing] were compared among all groups. Patients with HPV E6/E7 mRNA expression in the grade 1 CIN group were followed up for 1 yr. The relationship between atypical squamous epithelium and high malignant epithelial neoplasia was investigated by univariate and multivariate analysis. RESULTS The diagnostic sensitivity, specificity, and sensitivity of PCR-reverse point hybridization technology for secondary CIN were 70.41%, 70.66%, and 0.714, respectively. Sensitivity and specificity for secondary CIN were 752% and 7853%, respectively, the area under the curve value was 0.789. Logistic Multifactorial model analysis revealed that the HPV positive rates and the HPV E6/E7 mRNA positive rates were independent risk factors of CIN grade I (P < 0.05). In CIN grade I patients with positive for HPV E6/E7 mRNA, in its orientation to grade CIN patients, in its orientation to grade CIN patients, at 69.2%, compared with patients negative for HPV E6/E7 mRNA (30.8%), significant difference (P < 0.05). CONCLUSION HPV E6/E7 mRNA and HPV (PCR-reverse dot hybrid) positive expression have a close relationship with CIN-grade disease progression and is an independent risk factor for high-grade CIN lesions.
Collapse
Affiliation(s)
- Ling-Li Zhang
- Department of Gynaecology, Maternal and Child Health Hospital of Hubei Province, Wuhan 430075, Hubei Province, China
| | - Ming-Yan Du
- Department of Gynaecology, China Resources WISCO General Hospital, Wuhan 430080, Hubei Province, China
| | - Xin Du
- Department of Gynaecology, Maternal and Child Health Hospital of Hubei Province, Wuhan 430075, Hubei Province, China
| | - Jie Duan
- Department of Gynaecology, Maternal and Child Health Hospital of Hubei Province, Wuhan 430075, Hubei Province, China
| | - Dong-Mei Yao
- Department of Gynaecology, Maternal and Child Health Hospital of Hubei Province, Wuhan 430075, Hubei Province, China
| | - Jing Jing
- Department of Gynaecology, Maternal and Child Health Hospital of Hubei Province, Wuhan 430075, Hubei Province, China
| | - Chun Feng
- Department of Gynaecology, Maternal and Child Health Hospital of Hubei Province, Wuhan 430075, Hubei Province, China
| | - Lin Song
- Department of Surgery, Maternal and Child Health Hospital of Hubei Province, Wuhan 430075, Hubei Province, China
| |
Collapse
|
10
|
McAndrews KM, Mahadevan KK, Kalluri R. Mouse Models to Evaluate the Functional Role of the Tumor Microenvironment in Cancer Progression and Therapy Responses. Cold Spring Harb Perspect Med 2024; 14:a041411. [PMID: 38191175 PMCID: PMC11216184 DOI: 10.1101/cshperspect.a041411] [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: 01/10/2024]
Abstract
The tumor microenvironment (TME) is a complex ecosystem of both cellular and noncellular components that functions to impact the evolution of cancer. Various aspects of the TME have been targeted for the control of cancer; however, TME composition is dynamic, with the overall abundance of immune cells, endothelial cells (ECs), fibroblasts, and extracellular matrix (ECM) as well as subsets of TME components changing at different stages of progression and in response to therapy. To effectively treat cancer, an understanding of the functional role of the TME is needed. Genetically engineered mouse models have enabled comprehensive insight into the complex interactions within the TME ecosystem that regulate disease progression. Here, we review recent advances in mouse models that have been employed to understand how the TME regulates cancer initiation, progression, metastasis, and response to therapy.
Collapse
Affiliation(s)
- Kathleen M McAndrews
- Department of Cancer Biology, Metastasis Research Center, University of Texas MD Anderson Cancer Center, Houston, Texas 77054, USA
| | - Krishnan K Mahadevan
- Department of Cancer Biology, Metastasis Research Center, University of Texas MD Anderson Cancer Center, Houston, Texas 77054, USA
| | - Raghu Kalluri
- Department of Cancer Biology, Metastasis Research Center, University of Texas MD Anderson Cancer Center, Houston, Texas 77054, USA
- Department of Bioengineering, Rice University, Houston, Texas 77251, USA
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas 77030, USA
| |
Collapse
|
11
|
Chen X, Peng H, Zhang Z, Yang C, Liu Y, Chen Y, Yu F, Wu S, Cao L. SPDYC serves as a prognostic biomarker related to lipid metabolism and the immune microenvironment in breast cancer. Immunol Res 2024:10.1007/s12026-024-09505-5. [PMID: 38890248 DOI: 10.1007/s12026-024-09505-5] [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: 04/23/2024] [Accepted: 06/04/2024] [Indexed: 06/20/2024]
Abstract
Breast cancer remains the most common malignant carcinoma among women globally and is resistant to several therapeutic agents. There is a need for novel targets to improve the prognosis of patients with breast cancer. Bioinformatics analyses were conducted to explore potentially relevant prognostic genes in breast cancer using The Cancer Genome Atlas (TCGA) and The Gene Expression Omnibus (GEO) databases. Gene subtypes were categorized by machine learning algorithms. The machine learning-related breast cancer (MLBC) score was evaluated through principal component analysis (PCA) of clinical patients' pathological statuses and subtypes. Immune cell infiltration was analyzed using the xCell and CIBERSORT algorithms. Kyoto Encyclopedia of Genes and Genomes enrichment analysis elucidated regulatory pathways related to speedy/RINGO cell cycle regulator family member C (SPDYC) in breast cancer. The biological functions and lipid metabolic status of breast cancer cell lines were validated via quantitative real-time polymerase chain reaction (RT‒qPCR) assays, western blotting, CCK-8 assays, PI‒Annexin V fluorescence staining, transwell assays, wound healing assays, and Oil Red O staining. Key differentially expressed genes (DEGs) in breast cancer from the TCGA and GEO databases were screened and utilized to establish the MLBC score. Moreover, the MLBC score we established was negatively correlated with poor prognosis in breast cancer patients. Furthermore, the impacts of SPDYC on the tumor immune microenvironment and lipid metabolism in breast cancer were revealed and validated. SPDYC is closely related to activated dendritic cells and macrophages and is simultaneously correlated with the immune checkpoints CD47, cytotoxic T lymphocyte antigen-4 (CTLA-4), and poliovirus receptor (PVR). SPDYC strongly correlated with C-C motif chemokine ligand 7 (CCL7), a chemokine that influences breast cancer patient prognosis. A significant relationship was discovered between key genes involved in lipid metabolism and SPDYC, such as ELOVL fatty acid elongase 2 (ELOVL2), malic enzyme 1 (ME1), and squalene epoxidase (SQLE). Potent inhibitors targeting SPDYC in breast cancer were also discovered, including JNK inhibitor VIII, AICAR, and JW-7-52-1. Downregulation of SPDYC expression in vitro decreased proliferation, increased the apoptotic rate, decreased migration, and reduced lipid droplets. SPDYC possibly influences the tumor immune microenvironment and regulates lipid metabolism in breast cancer. Hence, this study identified SPDYC as a pivotal biomarker for developing therapeutic strategies for breast cancer.
Collapse
Affiliation(s)
- Xinxin Chen
- Department of Breast Surgery, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Haojie Peng
- Department of Breast Surgery, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Zhentao Zhang
- The First Clinical College, Guangdong Medical University, Zhanjiang, Guangdong, China
| | - Changnian Yang
- The First Clinical College, Guangdong Medical University, Zhanjiang, Guangdong, China
| | - Yingqi Liu
- The First Clinical College, Guangdong Medical University, Zhanjiang, Guangdong, China
| | - Yanzhen Chen
- Department of Gynecology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China
| | - Fei Yu
- Medical Research Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, China
| | - Shanshan Wu
- Department of Biology, School of Basic Medical Science, Guangdong Medical University, Zhanjiang, Guangdong, China.
| | - Lixue Cao
- Medical Research Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, China.
| |
Collapse
|
12
|
Yu H, Liu J, Bu X, Ma Z, Yao Y, Li J, Zhang T, Song W, Xiao X, Sun Y, Xiong W, Shi J, Dai P, Xiang B, Duan H, Yan X, Wu F, Zhang WC, Lin D, Hu H, Zhang H, Slack FJ, He HH, Freeman GJ, Wei W, Zhang J. Targeting METTL3 reprograms the tumor microenvironment to improve cancer immunotherapy. Cell Chem Biol 2024; 31:776-791.e7. [PMID: 37751743 PMCID: PMC10954589 DOI: 10.1016/j.chembiol.2023.09.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 07/02/2023] [Accepted: 09/01/2023] [Indexed: 09/28/2023]
Abstract
The tumor microenvironment (TME) is a heterogeneous ecosystem containing cancer cells, immune cells, stromal cells, cytokines, and chemokines which together govern tumor progression and response to immunotherapies. Methyltransferase-like 3 (METTL3), a core catalytic subunit for RNA N6-methyladenosine (m6A) modification, plays a crucial role in regulating various physiological and pathological processes. Whether and how METTL3 regulates the TME and anti-tumor immunity in non-small-cell lung cancer (NSCLC) remain poorly understood. Here, we report that METTL3 elevates expression of pro-tumorigenic chemokines including CXCL1, CXCL5, and CCL20, and destabilizes PD-L1 mRNA in an m6A-dependent manner, thereby shaping a non-inflamed TME. Thus, inhibiting METTL3 reprograms a more inflamed TME that renders anti-PD-1 therapy more effective in several murine lung tumor models. Clinically, NSCLC patients who exhibit low-METTL3 expression have a better prognosis when receiving anti-PD-1 therapy. Collectively, our study highlights targeting METTL3 as a promising strategy to improve immunotherapy in NSCLC patients.
Collapse
Affiliation(s)
- Haisheng Yu
- Department of Radiation and Medical Oncology, Medical Research Institute, Frontier Science Center of Immunology and Metabolism, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan 430071, China; Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan 430071, China
| | - Jing Liu
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA; Department of Urology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China
| | - Xia Bu
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Zhiqiang Ma
- Department of Medical Oncology, Senior Department of Oncology, Chinese PLA General Hospital, The Fifth Medical Center, 28 Fuxing Road, Beijing 100853, China
| | - Yingmeng Yao
- Department of Radiation and Medical Oncology, Medical Research Institute, Frontier Science Center of Immunology and Metabolism, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan 430071, China; Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan 430071, China
| | - Jinfeng Li
- Institute of Oncology, Chinese PLA General Hospital, The Fifth Medical Center, 28 Fuxing Road, Beijing 100853, China
| | - Tiantian Zhang
- Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan 430071, China; Medical Research Institute, Frontier Science Center of Immunology and Metabolism, Wuhan University, Wuhan 430071, China
| | - Wenjing Song
- Department of Hepatobiliary and Pancreatic Surgery, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Xiangling Xiao
- Department of Radiation and Medical Oncology, Medical Research Institute, Frontier Science Center of Immunology and Metabolism, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan 430071, China; Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan 430071, China
| | - Yishuang Sun
- Department of Radiation and Medical Oncology, Medical Research Institute, Frontier Science Center of Immunology and Metabolism, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan 430071, China; Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan 430071, China
| | - Wenjun Xiong
- Department of Radiation and Medical Oncology, Medical Research Institute, Frontier Science Center of Immunology and Metabolism, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan 430071, China; Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan 430071, China
| | - Jie Shi
- Department of Radiation and Medical Oncology, Medical Research Institute, Frontier Science Center of Immunology and Metabolism, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan 430071, China; Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan 430071, China
| | - Panpan Dai
- Department of Radiation and Medical Oncology, Medical Research Institute, Frontier Science Center of Immunology and Metabolism, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan 430071, China
| | - Bolin Xiang
- Department of Radiation and Medical Oncology, Medical Research Institute, Frontier Science Center of Immunology and Metabolism, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan 430071, China; Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan 430071, China
| | - Hongtao Duan
- Department of Thoracic Surgery, Tangdu Hospital, The Fourth Military Medical University, 1 Xinsi Road, Xi'an 710038, China
| | - Xiaolong Yan
- Department of Thoracic Surgery, Tangdu Hospital, The Fourth Military Medical University, 1 Xinsi Road, Xi'an 710038, China
| | - Fei Wu
- Department of Urology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan 250021, P.R.China
| | - Wen Cai Zhang
- Department of Cancer Division, Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida. Orlando, FL 32827, USA
| | - Dandan Lin
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan 430061, China
| | - Hankun Hu
- Department of Pharmacy, Zhongnan Hospital of Wuhan University, School of Pharmaceutical Sciences, Wuhan University, Wuhan 430071, China
| | - Haojian Zhang
- Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan 430071, China; Medical Research Institute, Frontier Science Center of Immunology and Metabolism, Wuhan University, Wuhan 430071, China
| | - Frank J Slack
- Harvard Medical School Initiative for RNA Medicine, Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Housheng Hansen He
- Princess Margaret Cancer Center, University Health Network, Toronto, ON, Canada; Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada
| | - Gordon J Freeman
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02115, USA.
| | - Wenyi Wei
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA.
| | - Jinfang Zhang
- Department of Radiation and Medical Oncology, Medical Research Institute, Frontier Science Center of Immunology and Metabolism, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan 430071, China; Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan 430071, China.
| |
Collapse
|
13
|
Li H, Sun Y, Yao Y, Ke S, Zhang N, Xiong W, Shi J, He C, Xiao X, Yu H, Dai P, Xiang B, Xing X, Xu G, Song W, Song J, Zhang J. USP8-governed GPX4 homeostasis orchestrates ferroptosis and cancer immunotherapy. Proc Natl Acad Sci U S A 2024; 121:e2315541121. [PMID: 38598341 PMCID: PMC11032464 DOI: 10.1073/pnas.2315541121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Accepted: 03/18/2024] [Indexed: 04/12/2024] Open
Abstract
Ferroptosis is an iron-dependent type of regulated cell death resulting from extensive lipid peroxidation and plays a critical role in various physiological and pathological processes. However, the regulatory mechanisms for ferroptosis sensitivity remain incompletely understood. Here, we report that homozygous deletion of Usp8 (ubiquitin-specific protease 8) in intestinal epithelial cells (IECs) leads to architectural changes in the colonic epithelium and shortens mouse lifespan accompanied by increased IEC death and signs of lipid peroxidation. However, mice with heterozygous deletion of Usp8 in IECs display normal phenotype and become resistant to azoxymethane/dextran sodium sulfate-induced colorectal tumorigenesis. Mechanistically, USP8 interacts with and deubiquitinates glutathione peroxidase 4 (GPX4), leading to GPX4 stabilization. Thus, USP8 inhibition destabilizes GPX4 and sensitizes cancer cells to ferroptosis in vitro. Notably, USP8 inhibition in combination with ferroptosis inducers retards tumor growth and enhances CD8+ T cell infiltration, which potentiates tumor response to anti-PD-1 immunotherapy in vivo. These findings uncover that USP8 counteracts ferroptosis by stabilizing GPX4 and highlight targeting USP8 as a potential therapeutic strategy to boost ferroptosis for enhancing cancer immunotherapy.
Collapse
Affiliation(s)
- Haiou Li
- Department of Dermatology, Medical Research Institute, Frontier Science Center of Immunology and Metabolism, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan430071, China
| | - Yishuang Sun
- Department of Dermatology, Medical Research Institute, Frontier Science Center of Immunology and Metabolism, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan430071, China
- Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan430071, China
| | - Yingmeng Yao
- Department of Dermatology, Medical Research Institute, Frontier Science Center of Immunology and Metabolism, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan430071, China
- Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan430071, China
| | - Shanwen Ke
- Department of Dermatology, Medical Research Institute, Frontier Science Center of Immunology and Metabolism, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan430071, China
- Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan430071, China
| | - Nannan Zhang
- Department of Radiation Oncology, The Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou450008, China
| | - Wenjun Xiong
- Department of Dermatology, Medical Research Institute, Frontier Science Center of Immunology and Metabolism, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan430071, China
- Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan430071, China
| | - Jie Shi
- Department of Dermatology, Medical Research Institute, Frontier Science Center of Immunology and Metabolism, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan430071, China
- Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan430071, China
| | - Chuan He
- Department of Dermatology, Medical Research Institute, Frontier Science Center of Immunology and Metabolism, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan430071, China
- Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan430071, China
| | - Xiangling Xiao
- Department of Dermatology, Medical Research Institute, Frontier Science Center of Immunology and Metabolism, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan430071, China
- Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan430071, China
| | - Haisheng Yu
- Department of Dermatology, Medical Research Institute, Frontier Science Center of Immunology and Metabolism, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan430071, China
- Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan430071, China
| | - Panpan Dai
- Department of Radiation and Medical Oncology, Hubei Key Laboratory of Tumor Biological Behaviors, Hubei Cancer Clinical Study Center, Zhongnan Hospital of Wuhan University, Wuhan430071, China
| | - Bolin Xiang
- Department of Dermatology, Medical Research Institute, Frontier Science Center of Immunology and Metabolism, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan430071, China
- Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan430071, China
| | - Xixin Xing
- Department of Dermatology, Medical Research Institute, Frontier Science Center of Immunology and Metabolism, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan430071, China
- Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan430071, China
| | - Gaoshan Xu
- Department of Dermatology, Medical Research Institute, Frontier Science Center of Immunology and Metabolism, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan430071, China
- Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan430071, China
| | - Wenjing Song
- Department of Hepatobiliary and Pancreatic Surgery, Zhongnan Hospital of Wuhan University, Wuhan430071, China
| | - Jiquan Song
- Department of Dermatology, Medical Research Institute, Frontier Science Center of Immunology and Metabolism, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan430071, China
| | - Jinfang Zhang
- Department of Dermatology, Medical Research Institute, Frontier Science Center of Immunology and Metabolism, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan430071, China
- Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan430071, China
- Department of Radiation and Medical Oncology, Hubei Key Laboratory of Tumor Biological Behaviors, Hubei Cancer Clinical Study Center, Zhongnan Hospital of Wuhan University, Wuhan430071, China
| |
Collapse
|
14
|
Dabbaghipour R, Ahmadi E, Entezam M, Farzam OR, Sohrabi S, Jamali S, Sichani AS, Paydar H, Baradaran B. Concise review: The heterogenous roles of BATF3 in cancer oncogenesis and dendritic cells and T cells differentiation and function considering the importance of BATF3-dependent dendritic cells. Immunogenetics 2024; 76:75-91. [PMID: 38358555 DOI: 10.1007/s00251-024-01335-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Accepted: 12/23/2023] [Indexed: 02/16/2024]
Abstract
The transcription factor, known as basic leucine zipper ATF-like 3 (BATF3), is a crucial contributor to the development of conventional type 1 dendritic cells (cDC1), which is definitely required for priming CD8 + T cell-mediated immunity against intracellular pathogens and malignancies. In this respect, BATF3-dependent cDC1 can bring about immunological tolerance, an autoimmune response, graft immunity, and defense against infectious agents such as viruses, microbes, parasites, and fungi. Moreover, the important function of cDC1 in stimulating CD8 + T cells creates an excellent opportunity to develop a highly effective target for vaccination against intracellular pathogens and diseases. BATF3 has been clarified to control the development of CD8α+ and CD103+ DCs. The presence of BATF3-dependent cDC1 in the tumor microenvironment (TME) reinforces immunosurveillance and improves immunotherapy approaches, which can be beneficial for cancer immunotherapy. Additionally, BATF3 acts as a transcriptional inhibitor of Treg development by decreasing the expression of the transcription factor FOXP3. However, when overexpressed in CD8 + T cells, it can enhance their survival and facilitate their transition to a memory state. BATF3 induces Th9 cell differentiation by binding to the IL-9 promoter through a BATF3/IRF4 complex. One of the latest research findings is the oncogenic function of BATF3, which has been approved and illustrated in several biological processes of proliferation and invasion.
Collapse
Affiliation(s)
- Reza Dabbaghipour
- Department of Medical Genetics, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Elham Ahmadi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mona Entezam
- Department of Medical Genetics, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Omid Rahbar Farzam
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Sepideh Sohrabi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Sajjad Jamali
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ali Saber Sichani
- Department of Medical Genetics, Shiraz University of Medical Sciences, Shiraz, Iran
- Department of Biology, Texas A&M University, College Station, TX, 77843, USA
| | - Hadi Paydar
- Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
- Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
15
|
Lee J, Kim D, Kong J, Ha D, Kim I, Park M, Lee K, Im SH, Kim S. Cell-cell communication network-based interpretable machine learning predicts cancer patient response to immune checkpoint inhibitors. SCIENCE ADVANCES 2024; 10:eadj0785. [PMID: 38295179 PMCID: PMC10830106 DOI: 10.1126/sciadv.adj0785] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Accepted: 12/28/2023] [Indexed: 02/02/2024]
Abstract
Immune checkpoint inhibitors (ICIs) have revolutionized cancer treatment. However, only some patients respond to ICIs, and current biomarkers for ICI efficacy have limited performance. Here, we devised an interpretable machine learning (ML) model trained using patient-specific cell-cell communication networks (CCNs) decoded from the patient's bulk tumor transcriptome. The model could (i) predict ICI efficacy for patients across four cancer types (median AUROC: 0.79) and (ii) identify key communication pathways with crucial players responsible for patient response or resistance to ICIs by analyzing more than 700 ICI-treated patient samples from 11 cohorts. The model prioritized chemotaxis communication of immune-related cells and growth factor communication of structural cells as the key biological processes underlying response and resistance to ICIs, respectively. We confirmed the key communication pathways and players at the single-cell level in patients with melanoma. Our network-based ML approach can be used to expand ICIs' clinical benefits in cancer patients.
Collapse
Affiliation(s)
- Juhun Lee
- Department of Life Sciences, Pohang University of Science and Technology, Pohang 790-784, Korea
| | - Donghyo Kim
- Department of Life Sciences, Pohang University of Science and Technology, Pohang 790-784, Korea
| | - JungHo Kong
- Department of Life Sciences, Pohang University of Science and Technology, Pohang 790-784, Korea
| | - Doyeon Ha
- Department of Life Sciences, Pohang University of Science and Technology, Pohang 790-784, Korea
| | - Inhae Kim
- ImmunoBiome Inc., Pohang 166-20, Korea
| | - Minhyuk Park
- Department of Life Sciences, Pohang University of Science and Technology, Pohang 790-784, Korea
| | - Kwanghwan Lee
- Department of Life Sciences, Pohang University of Science and Technology, Pohang 790-784, Korea
| | - Sin-Hyeog Im
- Department of Life Sciences, Pohang University of Science and Technology, Pohang 790-784, Korea
- ImmunoBiome Inc., Pohang 166-20, Korea
- Institute of Convergence Science, Yonsei University, Seoul 120-749, Korea
| | - Sanguk Kim
- Department of Life Sciences, Pohang University of Science and Technology, Pohang 790-784, Korea
- Institute of Convergence Science, Yonsei University, Seoul 120-749, Korea
| |
Collapse
|
16
|
Yin J, Gu T, Chaudhry N, Davidson NE, Huang Y. Epigenetic modulation of antitumor immunity and immunotherapy response in breast cancer: biological mechanisms and clinical implications. Front Immunol 2024; 14:1325615. [PMID: 38268926 PMCID: PMC10806158 DOI: 10.3389/fimmu.2023.1325615] [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: 10/21/2023] [Accepted: 12/22/2023] [Indexed: 01/26/2024] Open
Abstract
Breast cancer (BC) is the most common non-skin cancer and the second leading cause of cancer death in American women. The initiation and progression of BC can proceed through the accumulation of genetic and epigenetic changes that allow transformed cells to escape the normal cell cycle checkpoint control. Unlike nucleotide mutations, epigenetic changes such as DNA methylation, histone posttranslational modifications (PTMs), nucleosome remodeling and non-coding RNAs are generally reversible and therefore potentially responsive to pharmacological intervention. Epigenetic dysregulations are critical mechanisms for impaired antitumor immunity, evasion of immune surveillance, and resistance to immunotherapy. Compared to highly immunogenic tumor types, such as melanoma or lung cancer, breast cancer has been viewed as an immunologically quiescent tumor which displays a relatively low population of tumor-infiltrating lymphocytes (TIL), low tumor mutational burden (TMB) and modest response rates to immune checkpoint inhibitors (ICI). Emerging evidence suggests that agents targeting aberrant epigenetic modifiers may augment host antitumor immunity in BC via several interrelated mechanisms such as enhancing tumor antigen presentation, activation of cytotoxic T cells, inhibition of immunosuppressive cells, boosting response to ICI, and induction of immunogenic cell death (ICD). These discoveries have established a highly promising basis for using combinatorial approaches of epigenetic drugs with immunotherapy as an innovative paradigm to improve outcomes of BC patients. In this review, we summarize the current understanding of how epigenetic processes regulate immune cell function and antitumor immunogenicity in the context of the breast tumor microenvironment. Moreover, we discuss the therapeutic potential and latest clinical trials of the combination of immune checkpoint blockers with epigenetic agents in breast cancer.
Collapse
Affiliation(s)
- Jun Yin
- The University of Pittsburgh Medical Center (UPMC) Hillman Cancer Center, School of Medicine, University of Pittsburgh, Pittsburgh, PA, United States
| | - Tiezheng Gu
- The University of Pittsburgh Medical Center (UPMC) Hillman Cancer Center, School of Medicine, University of Pittsburgh, Pittsburgh, PA, United States
| | - Norin Chaudhry
- Department of Internal Medicine, Division of Hematology, Oncology, and Blood and Marrow Transplantation, Carver College of Medicine, University of Iowa, Iowa City, IA, United States
| | - Nancy E. Davidson
- Fred Hutchinson Cancer Center, University of Washington, Seattle, WA, United States
| | - Yi Huang
- Department of Internal Medicine, Division of Hematology, Oncology, and Blood and Marrow Transplantation, Carver College of Medicine, University of Iowa, Iowa City, IA, United States
- Holden Comprehensive Cancer Center, University of Iowa, Iowa City, IA, United States
| |
Collapse
|
17
|
Tian Y, Xie Y, Yi G, Wu F, Dang X, Bai F, Wang J, Zhang D. Prognostic Value and Therapeutic Significance of CCL Chemokines in Gastric Cancer. Curr Med Chem 2024; 31:7043-7058. [PMID: 39129286 DOI: 10.2174/0109298673315146240731100101] [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: 03/07/2024] [Revised: 06/10/2024] [Accepted: 07/02/2024] [Indexed: 08/13/2024]
Abstract
BACKGROUND Gastric cancer is one of the most common malignant tumours of the gastrointestinal tract, which has a significant negative impact on human health. AIMS CCL chemokines play important roles in a variety of tumor microenvironments; nevertheless, gastric cancer has surprisingly limited associations with CCL chemokines. METHODS In our study, we comprehensively utilized bioinformatics analysis tools and databases such as cBioPortal, UALCAN, GEPIA, GeneMANIA, STRING, and TRRUST to clarify the clinical significance and biology function of CCL chemokines in gastric cancer. RESULTS The mRNA expression levels of CCL1/3/4/5/7/8/14/15/18/20/21/22/26 were up-regulated, while the mRNA expression levels of CCL2/11/13/16/17/19/23/24/25/28 were down-regulated. The chemokine significantly associated with the pathological stage of gastric cancer is CCL2/11/19/21. In gastric cancer, the expression level of CCL chemokines was not associated with disease-free survival, but low expression of CCL14 was significantly associated with longer overall survival. Therein, associated with the regulation of CCL chemokines are only 10 transcription factors (RELA, NFKB1, STAT6, IRF3, REL, SPI1, STAT1, STAT3, JUN and SP1). The major biological process and functional enrichment of CCL chemokines are to induce cell-directed migration. CONCLUSION These results may indicate that CCL chemokines may be immunotherapeutic targets and promising prognostic biomarkers for gastric cancer.
Collapse
Affiliation(s)
- Yonggang Tian
- Department of Gastroenterology, Lanzhou University Second Hospital, Lanzhou, Gansu Province, China
| | - Yunqian Xie
- Department of Gastroenterology, The Gastroenterology Clinical Medical Center of Hainan Province, The Second Affiliated Hospital of Hainan Medical University, Haikou City, Hainan Province, China
| | - Guirong Yi
- Department of Gastroenterology, Lanzhou University Second Hospital, Lanzhou, Gansu Province, China
| | - Fanqi Wu
- The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, Gansu Province, China
| | - Xiaoyu Dang
- The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, Gansu Province, China
| | - Feihu Bai
- Department of Gastroenterology, The Gastroenterology Clinical Medical Center of Hainan Province, The Second Affiliated Hospital of Hainan Medical University, Haikou City, Hainan Province, China
| | - Jun Wang
- Department of Gastroenterology, 986 Hospital, Xijing Hospital, Air Force Military Medical University, Xi'an, Shaanxi Province, China
| | - Dekui Zhang
- Department of Gastroenterology, Lanzhou University Second Hospital, Lanzhou, Gansu Province, China
| |
Collapse
|
18
|
Cheng D, Wang J, Wang Y, Xue Y, Yang Q, Yang Q, Zhao H, Huang J, Peng X. Chemokines: Function and therapeutic potential in bone metastasis of lung cancer. Cytokine 2023; 172:156403. [PMID: 37871366 DOI: 10.1016/j.cyto.2023.156403] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Revised: 10/15/2023] [Accepted: 10/16/2023] [Indexed: 10/25/2023]
Abstract
Lung cancer is a rapidly progressing disease with a poor prognosis. Bone metastasis is commonly found in 40.6% of advanced-stage patients. The mortality rate of lung cancer patients with bone metastasis can be significantly decreased by implementing novel diagnostic techniques, improved staging and classification systems, precise surgical interventions, and advanced treatment modalities. However, it is important to note that there is currently a lack of radical procedures available for these patients due to the development of drug resistance. Consequently, palliative care approaches are commonly employed in clinical practice. Therefore, new understandings of the process of bone metastasis of lung cancer are critical for developing better treatment strategies to improve patient's clinical cure rate and quality of life. Chemokines are cell-secreted small signaling proteins in cancer occurrence, proliferation, invasion, and metastasis. In this study, we review the development of bone metastasis in lung cancer and discuss the mechanisms of specific chemokine families (CC, CXC, CX3C, and XC) in regulating the biological activities of tumors and promoting bone metastasis. We also highlight some preclinical studies and clinical trials on chemokines for lung cancer and bone metastasis.
Collapse
Affiliation(s)
- Dezhou Cheng
- Department of Medical Imaging, Health Science Center, Yangtze University, Jingzhou, Hubei, China
| | - Jiancheng Wang
- Department of Radiology, The Second People's Hospital of Jingzhou, China
| | - Yiling Wang
- Department of Medical Imaging, Health Science Center, Yangtze University, Jingzhou, Hubei, China
| | - Yanfang Xue
- Department of Medical Imaging, Health Science Center, Yangtze University, Jingzhou, Hubei, China
| | - Qing Yang
- Department of Medical Imaging, Health Science Center, Yangtze University, Jingzhou, Hubei, China
| | - Qun Yang
- Department of Medical Imaging, Health Science Center, Yangtze University, Jingzhou, Hubei, China
| | - Huichuan Zhao
- Department of Pathology of the First Affiliated Hospital of Yangtze University, Jingzhou, Hubei, China
| | - Jinbai Huang
- Department of Medical Imaging, Health Science Center, Yangtze University, Jingzhou, Hubei, China; Department of Medical Imaging, the First Affiliated Hospital of Yangtze University, and School of Medicine of Yangtze University, Jingzhou, Hubei, China.
| | - Xiaochun Peng
- Department of Pathophysiology, School of Basic Medicine, Health Science Center, Yangtze University, Jingzhou, Hubei, China.
| |
Collapse
|
19
|
Yi M, Li T, Niu M, Mei Q, Zhao B, Chu Q, Dai Z, Wu K. Exploiting innate immunity for cancer immunotherapy. Mol Cancer 2023; 22:187. [PMID: 38008741 PMCID: PMC10680233 DOI: 10.1186/s12943-023-01885-w] [Citation(s) in RCA: 31] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Accepted: 10/23/2023] [Indexed: 11/28/2023] Open
Abstract
Immunotherapies have revolutionized the treatment paradigms of various types of cancers. However, most of these immunomodulatory strategies focus on harnessing adaptive immunity, mainly by inhibiting immunosuppressive signaling with immune checkpoint blockade, or enhancing immunostimulatory signaling with bispecific T cell engager and chimeric antigen receptor (CAR)-T cell. Although these agents have already achieved great success, only a tiny percentage of patients could benefit from immunotherapies. Actually, immunotherapy efficacy is determined by multiple components in the tumor microenvironment beyond adaptive immunity. Cells from the innate arm of the immune system, such as macrophages, dendritic cells, myeloid-derived suppressor cells, neutrophils, natural killer cells, and unconventional T cells, also participate in cancer immune evasion and surveillance. Considering that the innate arm is the cornerstone of the antitumor immune response, utilizing innate immunity provides potential therapeutic options for cancer control. Up to now, strategies exploiting innate immunity, such as agonists of stimulator of interferon genes, CAR-macrophage or -natural killer cell therapies, metabolic regulators, and novel immune checkpoint blockade, have exhibited potent antitumor activities in preclinical and clinical studies. Here, we summarize the latest insights into the potential roles of innate cells in antitumor immunity and discuss the advances in innate arm-targeted therapeutic strategies.
Collapse
Affiliation(s)
- Ming Yi
- Cancer Center, Shanxi Bethune Hospital, Shanxi Academy of Medical Science, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, 030032, People's Republic of China
- Department of Breast Surgery, College of Medicine, The First Affiliated Hospital, Zhejiang University, Hangzhou, 310000, People's Republic of China
| | - Tianye Li
- Department of Gynecology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 310000, People's Republic of China
| | - Mengke Niu
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030, People's Republic of China
| | - Qi Mei
- Cancer Center, Shanxi Bethune Hospital, Shanxi Academy of Medical Science, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, 030032, People's Republic of China
| | - Bin Zhao
- Department of Breast Surgery, College of Medicine, The First Affiliated Hospital, Zhejiang University, Hangzhou, 310000, People's Republic of China
| | - Qian Chu
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030, People's Republic of China.
| | - Zhijun Dai
- Department of Breast Surgery, College of Medicine, The First Affiliated Hospital, Zhejiang University, Hangzhou, 310000, People's Republic of China.
| | - Kongming Wu
- Cancer Center, Shanxi Bethune Hospital, Shanxi Academy of Medical Science, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, 030032, People's Republic of China.
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030, People's Republic of China.
| |
Collapse
|
20
|
Chen Z, Giotti B, Kaluzova M, Vallcorba MP, Rawat K, Price G, Herting CJ, Pinero G, Cristea S, Ross JL, Ackley J, Maximov V, Szulzewsky F, Thomason W, Marquez-Ropero M, Angione A, Nichols N, Tsankova NM, Michor F, Shayakhmetov DM, Gutmann DH, Tsankov AM, Hambardzumyan D. A paracrine circuit of IL-1β/IL-1R1 between myeloid and tumor cells drives genotype-dependent glioblastoma progression. J Clin Invest 2023; 133:e163802. [PMID: 37733448 PMCID: PMC10645395 DOI: 10.1172/jci163802] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Accepted: 09/19/2023] [Indexed: 09/23/2023] Open
Abstract
Monocytes and monocyte-derived macrophages (MDMs) from blood circulation infiltrate glioblastoma (GBM) and promote growth. Here, we show that PDGFB-driven GBM cells induce the expression of the potent proinflammatory cytokine IL-1β in MDM, which engages IL-1R1 in tumor cells, activates the NF-κB pathway, and subsequently leads to induction of monocyte chemoattractant proteins (MCPs). Thus, a feedforward paracrine circuit of IL-1β/IL-1R1 between tumors and MDM creates an interdependence driving PDGFB-driven GBM progression. Genetic loss or locally antagonizing IL-1β/IL-1R1 leads to reduced MDM infiltration, diminished tumor growth, and reduced exhausted CD8+ T cells and thereby extends the survival of tumor-bearing mice. In contrast to IL-1β, IL-1α exhibits antitumor effects. Genetic deletion of Il1a/b is associated with decreased recruitment of lymphoid cells and loss-of-interferon signaling in various immune populations and subsets of malignant cells and is associated with decreased survival time of PDGFB-driven tumor-bearing mice. In contrast to PDGFB-driven GBM, Nf1-silenced tumors have a constitutively active NF-κB pathway, which drives the expression of MCPs to recruit monocytes into tumors. These results indicate local antagonism of IL-1β could be considered as an effective therapy specifically for proneural GBM.
Collapse
Affiliation(s)
- Zhihong Chen
- Department of Oncological Sciences, The Tisch Cancer Institute, Mount Sinai Icahn School of Medicine, New York, New York, USA
- Department of Pediatrics, AFLAC Cancer and Blood Disorders Center, Children’s Healthcare of Atlanta and Winship Cancer Institute, and
- Winship Cancer Institute, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Bruno Giotti
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Milota Kaluzova
- Department of Oncological Sciences, The Tisch Cancer Institute, Mount Sinai Icahn School of Medicine, New York, New York, USA
- Department of Pediatrics, AFLAC Cancer and Blood Disorders Center, Children’s Healthcare of Atlanta and Winship Cancer Institute, and
- Department of Neurology, Rutgers University, New Brunswick, New Jersey, USA
| | - Montse Puigdelloses Vallcorba
- Department of Oncological Sciences, The Tisch Cancer Institute, Mount Sinai Icahn School of Medicine, New York, New York, USA
| | - Kavita Rawat
- Department of Oncological Sciences, The Tisch Cancer Institute, Mount Sinai Icahn School of Medicine, New York, New York, USA
| | - Gabrielle Price
- Department of Oncological Sciences, The Tisch Cancer Institute, Mount Sinai Icahn School of Medicine, New York, New York, USA
| | - Cameron J. Herting
- Department of Pediatrics, AFLAC Cancer and Blood Disorders Center, Children’s Healthcare of Atlanta and Winship Cancer Institute, and
| | - Gonzalo Pinero
- Department of Oncological Sciences, The Tisch Cancer Institute, Mount Sinai Icahn School of Medicine, New York, New York, USA
| | - Simona Cristea
- Department of Data Science, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
- Department of Medicine, Brigham and Women’s Hospital, Boston, Massachusetts, USA
- Department of Medicine, Harvard Medical School, Boston, Massachusetts, USA
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, Massachusetts, USA
| | - James L. Ross
- Department of Pediatrics, AFLAC Cancer and Blood Disorders Center, Children’s Healthcare of Atlanta and Winship Cancer Institute, and
- Emory University Department of Microbiology and Immunology, Emory Vaccine Center, Atlanta, Georgia, USA
| | - James Ackley
- Department of Pediatrics, AFLAC Cancer and Blood Disorders Center, Children’s Healthcare of Atlanta and Winship Cancer Institute, and
| | - Victor Maximov
- Department of Pediatrics, AFLAC Cancer and Blood Disorders Center, Children’s Healthcare of Atlanta and Winship Cancer Institute, and
| | - Frank Szulzewsky
- Department of Human Biology, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Wes Thomason
- Department of Oncological Sciences, The Tisch Cancer Institute, Mount Sinai Icahn School of Medicine, New York, New York, USA
| | - Mar Marquez-Ropero
- Department of Oncological Sciences, The Tisch Cancer Institute, Mount Sinai Icahn School of Medicine, New York, New York, USA
| | - Angelo Angione
- Department of Oncological Sciences, The Tisch Cancer Institute, Mount Sinai Icahn School of Medicine, New York, New York, USA
| | | | - Nadejda M. Tsankova
- Department of Pathology and Molecular and Cell-Based Medicine, Mount Sinai Icahn School of Medicine, New York, New York, USA
| | - Franziska Michor
- Department of Data Science, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, Massachusetts, USA
- The Ludwig Center at Harvard, Boston, Massachusetts, USA
- The Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Dmitry M. Shayakhmetov
- Winship Cancer Institute, Emory University School of Medicine, Atlanta, Georgia, USA
- Lowance Center for Human Immunology and Emory Vaccine Center, Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia, USA
| | - David H. Gutmann
- Department of Neurology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Alexander M. Tsankov
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Dolores Hambardzumyan
- Department of Oncological Sciences, The Tisch Cancer Institute, Mount Sinai Icahn School of Medicine, New York, New York, USA
- Department of Pediatrics, AFLAC Cancer and Blood Disorders Center, Children’s Healthcare of Atlanta and Winship Cancer Institute, and
- Winship Cancer Institute, Emory University School of Medicine, Atlanta, Georgia, USA
- Department of Neurosurgery and
| |
Collapse
|
21
|
Wang S, Zhang G, Cui Q, Yang Y, Wang D, Liu A, Xia Y, Li W, Liu Y, Yu J. The DC-T cell axis is an effective target for the treatment of non-small cell lung cancer. Immun Inflamm Dis 2023; 11:e1099. [PMID: 38018578 PMCID: PMC10681037 DOI: 10.1002/iid3.1099] [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: 04/18/2023] [Revised: 11/06/2023] [Accepted: 11/09/2023] [Indexed: 11/30/2023] Open
Abstract
The dendritic cell (DC)-T cell axis is a bridge that connects innate and adaptive immunities. The initial immune response against tumors is mainly induced by mature antigen-presenting DCs. Enhancing the crosstalk between DCs and T cells may be an effective approach to improve the immune response to non-small cell lung cancer (NSCLC). In this article, a review was made of the interaction between DCs and T cells in the treatment of NSCLC and how this interaction affects the treatment outcome.
Collapse
Affiliation(s)
- Shuangcui Wang
- Department of OncologyFirst Teaching Hospital of Tianjin University of Traditional Chinese MedicineTianjinChina
- National Clinical Research Center for Chinese Medicine Acupuncture and MoxibustionTianjinChina
- Graduate SchoolTianjin University of Traditional Chinese MedicineTianjinChina
| | - Guan Zhang
- Department of OncologyFirst Teaching Hospital of Tianjin University of Traditional Chinese MedicineTianjinChina
- National Clinical Research Center for Chinese Medicine Acupuncture and MoxibustionTianjinChina
- Graduate SchoolTianjin University of Traditional Chinese MedicineTianjinChina
| | - Qian Cui
- Department of OncologyFirst Teaching Hospital of Tianjin University of Traditional Chinese MedicineTianjinChina
- National Clinical Research Center for Chinese Medicine Acupuncture and MoxibustionTianjinChina
- Graduate SchoolTianjin University of Traditional Chinese MedicineTianjinChina
| | - Yanjie Yang
- Department of OncologyFirst Teaching Hospital of Tianjin University of Traditional Chinese MedicineTianjinChina
- National Clinical Research Center for Chinese Medicine Acupuncture and MoxibustionTianjinChina
- Graduate SchoolTianjin University of Traditional Chinese MedicineTianjinChina
| | - Dong Wang
- Department of OncologyFirst Teaching Hospital of Tianjin University of Traditional Chinese MedicineTianjinChina
- National Clinical Research Center for Chinese Medicine Acupuncture and MoxibustionTianjinChina
- Graduate SchoolTianjin University of Traditional Chinese MedicineTianjinChina
| | - Aqing Liu
- Department of OncologyFirst Teaching Hospital of Tianjin University of Traditional Chinese MedicineTianjinChina
- National Clinical Research Center for Chinese Medicine Acupuncture and MoxibustionTianjinChina
- Graduate SchoolTianjin University of Traditional Chinese MedicineTianjinChina
| | - Ying Xia
- Department of OncologyFirst Teaching Hospital of Tianjin University of Traditional Chinese MedicineTianjinChina
- National Clinical Research Center for Chinese Medicine Acupuncture and MoxibustionTianjinChina
- Graduate SchoolTianjin University of Traditional Chinese MedicineTianjinChina
| | - Wentao Li
- Department of OncologyFirst Teaching Hospital of Tianjin University of Traditional Chinese MedicineTianjinChina
| | - Yunhe Liu
- Department of OncologyFirst Teaching Hospital of Tianjin University of Traditional Chinese MedicineTianjinChina
| | - Jianchun Yu
- Department of OncologyFirst Teaching Hospital of Tianjin University of Traditional Chinese MedicineTianjinChina
| |
Collapse
|
22
|
Chen Y, Shi J, Wang X, Zhou L, Wang Q, Xie Y, Peng C, Kuang L, Yang D, Yang J, Yang C, Li X, Yuan Y, Zhou Y, Peng A, Zhang Y, Chen H, Liu X, Zheng L, Huang K, Li Y. An antioxidant feedforward cycle coordinated by linker histone variant H1.2 and NRF2 that drives nonsmall cell lung cancer progression. Proc Natl Acad Sci U S A 2023; 120:e2306288120. [PMID: 37729198 PMCID: PMC10523483 DOI: 10.1073/pnas.2306288120] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Accepted: 08/11/2023] [Indexed: 09/21/2023] Open
Abstract
Nonsmall cell lung cancer (NSCLC) is highly malignant with limited treatment options, platinum-based chemotherapy is a standard treatment for NSCLC with resistance commonly seen. NSCLC cells exploit enhanced antioxidant defense system to counteract excessive reactive oxygen species (ROS), which contributes largely to tumor progression and resistance to chemotherapy, yet the mechanisms are not fully understood. Recent studies have suggested the involvement of histones in tumor progression and cellular antioxidant response; however, whether a major histone variant H1.2 (H1C) plays roles in the development of NSCLC remains unclear. Herein, we demonstrated that H1.2 was increasingly expressed in NSCLC tumors, and its expression was correlated with worse survival. When crossing the H1c knockout allele with a mouse NSCLC model (KrasLSL-G12D/+), H1.2 deletion suppressed NSCLC progression and enhanced oxidative stress and significantly decreased the levels of key antioxidant glutathione (GSH) and GCLC, the catalytic subunit of rate-limiting enzyme for GSH synthesis. Moreover, high H1.2 was correlated with the IC50 of multiple chemotherapeutic drugs and with worse prognosis in NSCLC patients receiving chemotherapy; H1.2-deficient NSCLC cells presented reduced survival and increased ROS levels upon cisplatin treatment, while ROS scavenger eliminated the survival inhibition. Mechanistically, H1.2 interacted with NRF2, a master regulator of antioxidative response; H1.2 enhanced the nuclear level and stability of NRF2 and, thus, promoted NRF2 binding to GCLC promoter and the consequent transcription; while NRF2 also transcriptionally up-regulated H1.2. Collectively, these results uncovered a tumor-driving role of H1.2 in NSCLC and indicate an "H1.2-NRF2" antioxidant feedforward cycle that promotes tumor progression and chemoresistance.
Collapse
Affiliation(s)
- Yuchen Chen
- Tongji School of Pharmacy, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan430030, China
| | - Jiajian Shi
- Tongji School of Pharmacy, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan430030, China
| | - Xiaomu Wang
- Department of Pharmacy, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang441000, China
| | - Lin Zhou
- Department of Thoracic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan430030, China
| | - Qing Wang
- College of Life Sciences, Wuhan University, Wuhan430072, China
| | - Yunhao Xie
- College of Life Sciences, Wuhan University, Wuhan430072, China
| | - Chentai Peng
- Tongji School of Pharmacy, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan430030, China
| | - Linwu Kuang
- Department of Thoracic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan430030, China
| | - Dong Yang
- Tongji School of Pharmacy, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan430030, China
| | - Jing Yang
- Tongji School of Pharmacy, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan430030, China
| | - Chen Yang
- Tongji School of Pharmacy, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan430030, China
| | - Xi Li
- Tongji School of Pharmacy, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan430030, China
| | - Yangmian Yuan
- College of Life Sciences, Wuhan University, Wuhan430072, China
| | - Yihao Zhou
- College of Life Sciences, Wuhan University, Wuhan430072, China
| | - Anlin Peng
- Department of Pharmacy, Wuhan Third Hospital and Tongren Hospital of Wuhan University, Wuhan430060, China
| | - Yu Zhang
- Tongji School of Pharmacy, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan430030, China
| | - Hong Chen
- Tongji School of Pharmacy, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan430030, China
| | - Xinran Liu
- Tongji School of Pharmacy, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan430030, China
| | - Ling Zheng
- College of Life Sciences, Wuhan University, Wuhan430072, China
| | - Kun Huang
- Tongji School of Pharmacy, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan430030, China
- Tongji-RongCheng Biomedical Center, Tongji Medical College, Huazhong University of Science and Technology, Wuhan430030, China
| | - Yangkai Li
- Department of Thoracic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan430030, China
| |
Collapse
|
23
|
Li W, Li M, Huang Q, He X, Shen C, Hou X, Xue F, Deng Z, Luo Y. Advancement of regulating cellular signaling pathways in NSCLC target therapy via nanodrug. Front Chem 2023; 11:1251986. [PMID: 37744063 PMCID: PMC10512551 DOI: 10.3389/fchem.2023.1251986] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Accepted: 08/29/2023] [Indexed: 09/26/2023] Open
Abstract
Lung cancer (LC) is one of the leading causes of high cancer-associated mortality worldwide. Non-small cell lung cancer (NSCLC) is the most common type of LC. The mechanisms of NSCLC evolution involve the alterations of multiple complex signaling pathways. Even with advances in biological understanding, early diagnosis, therapy, and mechanisms of drug resistance, many dilemmas still need to face in NSCLC treatments. However, many efforts have been made to explore the pathological changes of tumor cells based on specific molecular signals for drug therapy and targeted delivery. Nano-delivery has great potential in the diagnosis and treatment of tumors. In recent years, many studies have focused on different combinations of drugs and nanoparticles (NPs) to constitute nano-based drug delivery systems (NDDS), which deliver drugs regulating specific molecular signaling pathways in tumor cells, and most of them have positive implications. This review summarized the recent advances of therapeutic targets discovered in signaling pathways in NSCLC as well as the related NDDS, and presented the future prospects and challenges.
Collapse
Affiliation(s)
- Wenqiang Li
- Zigong First People’s Hospital, Zigong, Sichuan, China
| | - Mei Li
- West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Qian Huang
- Sichuan North Medical College, Nanchong, Sichuan, China
| | - Xiaoyu He
- Sichuan North Medical College, Nanchong, Sichuan, China
| | - Chen Shen
- West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Xiaoming Hou
- West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Fulai Xue
- West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Zhiping Deng
- Zigong First People’s Hospital, Zigong, Sichuan, China
| | - Yao Luo
- Zigong First People’s Hospital, Zigong, Sichuan, China
- West China Hospital, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
24
|
Pan H, Zou N, Tian Y, Zhu H, Zhang J, Jin W, Gu Z, Ning J, Li Z, Kong W, Jiang L, Huang J, Luo Q. Short-term outcomes of robot-assisted versus video-assisted thoracoscopic surgery for non-small cell lung cancer patients with neoadjuvant immunochemotherapy: a single-center retrospective study. Front Immunol 2023; 14:1228451. [PMID: 37497221 PMCID: PMC10366598 DOI: 10.3389/fimmu.2023.1228451] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Accepted: 06/27/2023] [Indexed: 07/28/2023] Open
Abstract
Background Neoadjuvant immunochemotherapy has been increasingly applied to treat non-small cell lung cancer (NSCLC). However, the comparison between robotic-assisted thoracoscopic surgery (RATS) and video-assisted thoracoscopic surgery (VATS) in the feasibility and oncological efficacy following neoadjuvant immunochemotherapy is scarce. This study aims to assess the superiorities of RATS over (VATS) concerning short-term outcomes in treating NSCLC patients with neoadjuvant immunochemotherapy. Methods NSCLC patients receiving RATS or VATS lobectomy following neoadjuvant immunochemotherapy at Shanghai Chest Hospital from 2019 to 2022 were retrospectively identified. Baseline clinical characteristics, perioperative outcomes, and survival profiles were analyzed. Results Forty-six NSCLC patients with neoadjuvant immunochemotherapy were included and divided into the RATS (n=15) and VATS (n=31) groups. The baseline clinical characteristics and induction-related adverse events were comparable between the two groups (all p>0.050). The 30-day mortality in the RATS and VATS groups were 0% and 3.23%, respectively (p=1.000). Patients undergoing RATS were associated with reduced surgical-related intensive unit care (ICU) stay than those receiving VATS (0.0 [0.0-0.0] vs. 0.0 [0.0-1.0] days, p=0.026). Moreover, RATS assessed more N1 LNs (6.27 ± 1.94 vs 4.90 ± 1.92, p=0.042) and LN stations (3.07 ± 1.03 vs 2.52 ± 0.57, p=0.038) compared with VATS. By comparison, no difference was found in surgical outcomes, pathological results, and postoperative complications between the RATS and VATS groups (all p>0.050). Finally, RATS and VATS achieved comparable one-year recurrence-free survival (82.96% vs. 85.23%, p=0.821) and the timing of central nervous system, LN, and bone recurrences (all p>0.050). Conclusion RATS is safe and feasible for NSCLC patients with neoadjuvant immunochemotherapy, reducing surgical-related ICU stay, assessing increased N1 LNs and stations, and achieving similar survival profiles to VATS.
Collapse
Affiliation(s)
- Hanbo Pan
- Department of Thoracic Surgical Oncology, Shanghai Lung Cancer Center, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ningyuan Zou
- Department of Thoracic Surgical Oncology, Shanghai Lung Cancer Center, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yu Tian
- Department of Thoracic Surgical Oncology, Shanghai Lung Cancer Center, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hongda Zhu
- Department of Thoracic Surgical Oncology, Shanghai Lung Cancer Center, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jiaqi Zhang
- Department of Thoracic Surgical Oncology, Shanghai Lung Cancer Center, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Weiqiu Jin
- Department of Thoracic Surgical Oncology, Shanghai Lung Cancer Center, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zenan Gu
- Department of Thoracic Surgical Oncology, Shanghai Lung Cancer Center, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Junwei Ning
- Department of Thoracic Surgical Oncology, Shanghai Lung Cancer Center, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Department of Thoracic Surgery, Shanghai Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ziming Li
- Department of Oncology, Shanghai Lung Cancer Center, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Weicheng Kong
- Department of Thoracic Surgical Oncology, Shanghai Lung Cancer Center, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Department of Thoracic Surgery, Putuo District People’s Hospital, Zhejiang, China
| | - Long Jiang
- Department of Thoracic Surgical Oncology, Shanghai Lung Cancer Center, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jia Huang
- Department of Thoracic Surgical Oncology, Shanghai Lung Cancer Center, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qingquan Luo
- Department of Thoracic Surgical Oncology, Shanghai Lung Cancer Center, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
25
|
Liu X, Zhuang Y, Huang W, Wu Z, Chen Y, Shan Q, Zhang Y, Wu Z, Ding X, Qiu Z, Cui W, Wang Z. Interventional hydrogel microsphere vaccine as an immune amplifier for activated antitumour immunity after ablation therapy. Nat Commun 2023; 14:4106. [PMID: 37433774 PMCID: PMC10336067 DOI: 10.1038/s41467-023-39759-w] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Accepted: 06/28/2023] [Indexed: 07/13/2023] Open
Abstract
The response rate of pancreatic cancer to chemotherapy or immunotherapy pancreatic cancer is low. Although minimally invasive irreversible electroporation (IRE) ablation is a promising option for irresectable pancreatic cancers, the immunosuppressive tumour microenvironment that characterizes this tumour type enables tumour recurrence. Thus, strengthening endogenous adaptive antitumour immunity is critical for improving the outcome of ablation therapy and post-ablation immune therapy. Here we present a hydrogel microsphere vaccine that amplifies post-ablation anti-cancer immune response via releasing its cargo of FLT3L and CD40L at the relatively lower pH of the tumour bed. The vaccine facilitates migration of the tumour-resident type 1 conventional dendritic cells (cDC1) to the tumour-draining lymph nodes (TdLN), thus initiating the cDC1-mediated antigen cross-presentation cascade, resulting in enhanced endogenous CD8+ T cell response. We show in an orthotopic pancreatic cancer model in male mice that the hydrogel microsphere vaccine transforms the immunologically cold tumour microenvironment into hot in a safe and efficient manner, thus significantly increasing survival and inhibiting the growth of distant metastases.
Collapse
Affiliation(s)
- Xiaoyu Liu
- Department of Radiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, No.197, Ruijin 2nd Road, 200025, Shanghai, P. R. China
| | - Yaping Zhuang
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, 200025, Shanghai, P. R. China
| | - Wei Huang
- Department of Radiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, No.197, Ruijin 2nd Road, 200025, Shanghai, P. R. China
| | - Zhuozhuo Wu
- Department of Radiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, No.197, Ruijin 2nd Road, 200025, Shanghai, P. R. China
| | - Yingjie Chen
- Department of Radiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, No.197, Ruijin 2nd Road, 200025, Shanghai, P. R. China
| | - Qungang Shan
- Department of Radiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, No.197, Ruijin 2nd Road, 200025, Shanghai, P. R. China
| | - Yuefang Zhang
- Institute of Neuroscience, CAS Key Laboratory of Primate Neurobiology, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, No.320 Yueyang Road, 200032, Shanghai, P. R. China
| | - Zhiyuan Wu
- Department of Radiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, No.197, Ruijin 2nd Road, 200025, Shanghai, P. R. China
| | - Xiaoyi Ding
- Department of Radiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, No.197, Ruijin 2nd Road, 200025, Shanghai, P. R. China
| | - Zilong Qiu
- Institute of Neuroscience, CAS Key Laboratory of Primate Neurobiology, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, No.320 Yueyang Road, 200032, Shanghai, P. R. China
| | - Wenguo Cui
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, 200025, Shanghai, P. R. China.
| | - Zhongmin Wang
- Department of Radiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, No.197, Ruijin 2nd Road, 200025, Shanghai, P. R. China.
- Department of Radiology, Ruijin Hospital Luwan Branch, Shanghai Jiao Tong University School of Medicine, No.149, South Chongqing Road, 200025, Shanghai, P. R. China.
| |
Collapse
|
26
|
Dong HP, Li Y, Tang Z, Wang P, Zhong B, Chu Q, Lin D. Combined targeting of CCL7 and Flt3L to promote the expansion and infiltration of cDC1s in tumors enhances T-cell activation and anti-PD-1 therapy effectiveness in NSCLC. Cell Mol Immunol 2023; 20:850-853. [PMID: 36894615 PMCID: PMC10310796 DOI: 10.1038/s41423-023-00991-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Accepted: 02/20/2023] [Indexed: 03/11/2023] Open
Affiliation(s)
- Hong-Peng Dong
- Cancer Center, Renmin Hospital of Wuhan University, Medical Research Institute and Frontier Science Center of Immunology and Metabolism, Wuhan University, Wuhan, 430071, China
- College of Life Sciences, Wuhan University, Wuhan, 430072, China
| | - Ying Li
- CAS Key Laboratory of Infection and Immunity, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
| | - Zhen Tang
- Cancer Center, Renmin Hospital of Wuhan University, Medical Research Institute and Frontier Science Center of Immunology and Metabolism, Wuhan University, Wuhan, 430071, China
- College of Life Sciences, Wuhan University, Wuhan, 430072, China
| | - Peng Wang
- Cancer Center, Renmin Hospital of Wuhan University, Medical Research Institute and Frontier Science Center of Immunology and Metabolism, Wuhan University, Wuhan, 430071, China
- College of Life Sciences, Wuhan University, Wuhan, 430072, China
| | - Bo Zhong
- College of Life Sciences, Wuhan University, Wuhan, 430072, China
- TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan, 430071, China
| | - Qian Chu
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Dandan Lin
- Cancer Center, Renmin Hospital of Wuhan University, Medical Research Institute and Frontier Science Center of Immunology and Metabolism, Wuhan University, Wuhan, 430071, China.
| |
Collapse
|
27
|
Suntiparpluacha M, Chanthercrob J, Sa-nguanraksa D, Sitthikornpaiboon J, Chaiboonchoe A, Kueanjinda P, Jinawath N, Sampattavanich S. Retrospective study of transcriptomic profiling identifies Thai triple-negative breast cancer patients who may benefit from immune checkpoint and PARP inhibitors. PeerJ 2023; 11:e15350. [PMID: 37334114 PMCID: PMC10269579 DOI: 10.7717/peerj.15350] [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: 01/13/2023] [Accepted: 04/13/2023] [Indexed: 06/20/2023] Open
Abstract
Background Triple-negative breast cancer (TNBC) is a rare and aggressive breast cancer subtype. Unlike the estrogen receptor-positive subtype, whose recurrence risk can be predicted by gene expression-based signature, TNBC is more heterogeneous, with diverse drug sensitivity levels to standard regimens. This study explored the benefit of gene expression-based profiling for classifying the molecular subtypes of Thai TNBC patients. Methods The nCounter-based Breast 360 gene expression was used to classify Thai TNBC retrospective cohort subgroups. Their expression profiles were then compared against the previously established TNBC classification system. The differential characteristics of the tumor microenvironment and DNA damage repair signatures across subgroups were also explored. Results Thai TNBC cohort could be classified into four main subgroups, corresponding to the LAR, BL-2, and M subtypes based on Lehmann's TNBC classification. The PAM50 gene set classified most samples as basal-like subtypes except for Group 1. Group 1 exhibited similar enrichment of the metabolic and hormone response pathways to the LAR subtype. Group 2 shared pathway activation with the BL-2 subtype. Group 3 showed an increase in the EMT pathway, similar to the M subtype. Group 4 showed no correlation with Lehmann's TNBC. The tumor microenvironment (TME) analysis showed high TME cell abundance with increased expression of immune blockade genes in Group 2. Group 4 exhibited low TME cell abundance and reduced immune blockade gene expressions. We also observed distinct signatures of the DNA double-strand break repair genes in Group 1. Conclusions Our study reported unique characteristics between the four TNBC subgroups and showed the potential use of immune checkpoint and PARP inhibitors in subsets of Thai TNBC patients. Our findings warrant further clinical investigation to validate TNBC's sensitivity to these regimens.
Collapse
Affiliation(s)
- Monthira Suntiparpluacha
- Siriraj Center of Research Excellence for Systems Pharmacology, Department of Pharmacology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Jantappapa Chanthercrob
- Siriraj Center of Research Excellence for Systems Pharmacology, Department of Pharmacology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Doonyapat Sa-nguanraksa
- Division of Head Neck and Breast Surgery, Department of Surgery, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Juthamas Sitthikornpaiboon
- Siriraj Center of Research Excellence for Systems Pharmacology, Department of Pharmacology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Amphun Chaiboonchoe
- Siriraj Center of Research Excellence for Systems Pharmacology, Department of Pharmacology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Patipark Kueanjinda
- Center of Excellence in Immunology and Immune-mediated Diseases, Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Natini Jinawath
- Program in Translational Medicine, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
- Chakri Naruebodindra Medical Institute, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Samut Prakan, Thailand
- Integrative Computational BioScience (ICBS) Center, Mahidol University, Nakhon Pathom, Thailand
| | - Somponnat Sampattavanich
- Siriraj Center of Research Excellence for Systems Pharmacology, Department of Pharmacology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| |
Collapse
|
28
|
Zheng Y, Liu X, Li N, Zhao A, Sun Z, Wang M, Luo J. Radiotherapy combined with immunotherapy could improve the immune infiltration of melanoma in mice and enhance the abscopal effect. Radiat Oncol J 2023; 41:129-139. [PMID: 37403355 DOI: 10.3857/roj.2023.00185] [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: 02/27/2023] [Accepted: 06/08/2023] [Indexed: 07/06/2023] Open
Abstract
PURPOSE To analyze the gene mutation, immune infiltration and tumor growth of primary tumor and distant tumor under different treatment modes. MATERIALS AND METHODS Twenty B16 murine melanoma cells were injected subcutaneously into the of both sides of the thigh, simulating a primary tumor and a secondary tumor impacted by the abscopal effect, respectively. They were divided into blank control group, immunotherapy group, radiotherapy group, and radiotherapy combined immunotherapy group. During this period, tumor volume was measured, and RNA sequencing was performed on tumor samples after the test. R software was used to analyze differentially expressed genes, functional enrichment, and immune infiltration. RESULTS We found that any treatment mode could cause changes in differentially expressed genes, especially the combination treatment. The different therapeutic effects might be caused by gene expression. In addition, the proportions of infiltrating immune cells in the irradiated and abscopal tumors were different. In the combination treatment group, T-cell infiltration in the irradiated site was the most obvious. In the immunotherapy group, CD8+ T-cell infiltration in the abscopal tumor site was obvious, but immunotherapy alone might have a poor prognosis. Whether the irradiated or abscopal tumor was evaluated, radiotherapy combined with anti-programmed cell death protein 1 (anti-PD-1) therapy produced the most obvious tumor control and might have a positive impact on prognosis. CONCLUSION Combination therapy not only improves the immune microenvironment but may also have a positive impact on prognosis.
Collapse
Affiliation(s)
- Yufeng Zheng
- Department of Radiotherapy, The Affiliated Changzhou Second People's Hospital of Nanjing Medical University, Changzhou, China
| | - Xue Liu
- Department of Radiotherapy, The Affiliated Changzhou Second People's Hospital of Nanjing Medical University, Changzhou, China
- Department of Radiotherapy, Dalian Medical University, Dalian, China
| | - Na Li
- Department of Radiotherapy, The Affiliated Changzhou Second People's Hospital of Nanjing Medical University, Changzhou, China
| | - Aimei Zhao
- Department of Obstetrics and Gynecology, Liaocheng Dongchangfu District Maternal and Child Health Hospital, Liaocheng, China
| | - Zhiqiang Sun
- Department of Radiotherapy, The Affiliated Changzhou Second People's Hospital of Nanjing Medical University, Changzhou, China
| | - Meihua Wang
- Department of Pathology, Changzhou Fourth People's Hospital, Changzhou, China
| | - Judong Luo
- Department of Radiotherapy, The Affiliated Changzhou Second People's Hospital of Nanjing Medical University, Changzhou, China
| |
Collapse
|
29
|
Liu X, Zeng L, Zhou Y, Zhao X, Zhu L, Zhang J, Pan Y, Shao C, Fu J. P21 facilitates macrophage chemotaxis by promoting CCL7 in the lung epithelial cell lines treated with radiation and bleomycin. J Transl Med 2023; 21:314. [PMID: 37161570 PMCID: PMC10169365 DOI: 10.1186/s12967-023-04177-5] [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: 03/24/2023] [Accepted: 04/30/2023] [Indexed: 05/11/2023] Open
Abstract
BACKGROUND Interstitial lung diseases (ILDs) can be induced and even exacerbated by radiotherapy in thoracic cancer patients. The roles of immune responses underlying the development of these severe lung injuries are still obscure and need to be investigated. METHODS A severe lung damage murine model was established by delivering 16 Gy X-rays to the chest of mice that had been pre-treated with bleomycin (BLM) and thus hold ILDs. Bioinformatic analyses were performed on the GEO datasets of radiation-induced lung injury (RILI) and BLM-induced pulmonary fibrosis (BIPF), and RNA-sequencing data of the severely damaged lung tissues. The screened differentially expressed genes (DEGs) were verified in lung epithelial cell lines by qRT-PCR assay. The injured lung tissue pathology was analyzed with H&E and Masson's staining, and immunohistochemistry staining. The macrophage chemotaxis and activity promoted by the stressed epithelial cells were determined by using a cell co-culture system. The expressions of p21 in MLE-12 and Beas-2B cells were detected by qRT-PCR, western blot, and immunofluorescence. The concentration of CCL7 in cell supernatant was measured by ELISA assay. In some experiments, Beas-2B cells were transfected with p21-siRNA or CCL7-siRNA before irradiation and/or BLM treatment. RESULTS After the treatment of irradiation and/or BLM, the inflammatory and immune responses, chemokine-mediated signaling pathways were steadily activated in the severely injured lung, and p21 was screened out by the bioinformatic analysis and further verified to be upregulated in both mouse and human lung epithelial cell lines. The expression of P21 was positively correlated with macrophage infiltration in the injured lung tissues. Co-culturing with stressed Beas-2B cells or its conditioned medium containing CCL7 protein, U937 macrophages were actively polarized to M1-phase and their migration ability was obviously increased along with the damage degree of Beas-2B cells. Furthermore, knockdown p21 reduced CCL7 expression in Beas-2B cells and then decreased the chemotaxis of co-cultured macrophages. CONCLUSIONS P21 promoted CCL7 release from the severely injured lung epithelial cell lines and contributed to the macrophage chemotaxis in vitro, which provides new insights for better understanding the inflammatory responses in lung injury.
Collapse
Affiliation(s)
- Xinglong Liu
- Shanghai Institute of Infectious Disease and Biosecurity, and Institute of Radiation Medicine, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Liang Zeng
- Institute of Radiation Medicine, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Yuchuan Zhou
- Institute of Radiation Medicine, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Xinrui Zhao
- Institute of Radiation Medicine, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Lin Zhu
- Institute of Radiation Medicine, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Jianghong Zhang
- Institute of Radiation Medicine, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Yan Pan
- Institute of Radiation Medicine, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Chunlin Shao
- Shanghai Institute of Infectious Disease and Biosecurity, and Institute of Radiation Medicine, Shanghai Medical College, Fudan University, Shanghai, 200032, China.
| | - Jiamei Fu
- Department of Radiation Oncology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, 200433, China.
| |
Collapse
|
30
|
Ito M, Iwama S, Sugiyama D, Yasuda Y, Okuji T, Kobayashi T, Zhou X, Yamagami A, Onoue T, Miyata T, Sugiyama M, Hagiwara D, Suga H, Banno R, Nishikawa H, Arima H. Anti-tumor effects of anti-programmed cell death-1 antibody treatment are attenuated in streptozotocin-induced diabetic mice. Sci Rep 2023; 13:5939. [PMID: 37046033 PMCID: PMC10097709 DOI: 10.1038/s41598-023-33049-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Accepted: 04/06/2023] [Indexed: 04/14/2023] Open
Abstract
Hyperglycemia impairs immune response; however, it remains unknown whether the anti-tumor effects of anti-programmed cell death-1 antibody (PD-1-Ab) treatment are changed in hyperglycemic conditions. We analyzed the effect of PD-1-Ab on tumor growth in streptozotocin-induced diabetic mice (STZ-mice) subcutaneously inoculated with MC38 (a colon carcinoma cell line). Furthermore, we assessed the expression of chemokines by polymerase chain reaction (PCR) array in tumor-draining lymph nodes (dLNs) of these mice and MC38 cells cultured in different glucose concentrations. The suppressive effect of PD-1-Ab on tumor growth was attenuated. This was accompanied by fewer tumor-infiltrating CD8+ T cells, and STZ-mice had fewer tumor-infiltrating CD11c+ dendritic cells (DCs) than normoglycemic mice. mRNA expression levels of CXCL9, a chemokine recruiting CD8+ T cells, were lower in dLNs of STZ-mice than in normoglycemic mice after PD-1-Ab treatment, and its protein was expressed in DCs. In MC38 cells cultured with 25 mM glucose, mRNA expression of CCL7, a chemokine recruiting DCs, was decreased compared to cells cultured with 5 mM glucose. These results suggest that the STZ-induced hyperglycemia impairs the effect of PD-1-Ab treatment on MC38 tumor growth, and is accompanied by reduced infiltration of DCs and CD8+ T cells and decreased expression of CCL7 and CXCL9.
Collapse
Affiliation(s)
- Masaaki Ito
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, Nagoya, 466-8550, Japan
| | - Shintaro Iwama
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, Nagoya, 466-8550, Japan.
| | - Daisuke Sugiyama
- Department of Immunology, Nagoya University Graduate School of Medicine, Nagoya, 466-8550, Japan
| | - Yoshinori Yasuda
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, Nagoya, 466-8550, Japan
| | - Takayuki Okuji
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, Nagoya, 466-8550, Japan
| | - Tomoko Kobayashi
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, Nagoya, 466-8550, Japan
| | - Xin Zhou
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, Nagoya, 466-8550, Japan
| | - Ayana Yamagami
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, Nagoya, 466-8550, Japan
| | - Takeshi Onoue
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, Nagoya, 466-8550, Japan
| | - Takashi Miyata
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, Nagoya, 466-8550, Japan
| | - Mariko Sugiyama
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, Nagoya, 466-8550, Japan
| | - Daisuke Hagiwara
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, Nagoya, 466-8550, Japan
| | - Hidetaka Suga
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, Nagoya, 466-8550, Japan
| | - Ryoichi Banno
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, Nagoya, 466-8550, Japan
- Research Center of Health, Physical Fitness and Sports, Nagoya University, Nagoya, 464-8601, Japan
| | - Hiroyoshi Nishikawa
- Department of Immunology, Nagoya University Graduate School of Medicine, Nagoya, 466-8550, Japan
- Division of Cancer Immunology, Research Institute/Exploratory Oncology Research & Clinical Trial Center (EPOC), National Cancer Center, Tokyo, 104-0045, Japan
| | - Hiroshi Arima
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, Nagoya, 466-8550, Japan
| |
Collapse
|
31
|
Hu L, Liu M, Tang B, Li Q, Pan BS, Xu C, Lin HK. Posttranslational regulation of liver kinase B1 (LKB1) in human cancer. J Biol Chem 2023; 299:104570. [PMID: 36870679 PMCID: PMC10068580 DOI: 10.1016/j.jbc.2023.104570] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2023] [Revised: 02/20/2023] [Accepted: 02/22/2023] [Indexed: 03/06/2023] Open
Abstract
Liver kinase B1 (LKB1) is a serine-threonine kinase that participates in multiple cellular and biological processes, including energy metabolism, cell polarity, cell proliferation, cell migration, and many others. LKB1 is initially identified as a germline-mutated causative gene in Peutz-Jeghers syndrome (PJS) and is commonly regarded as a tumor suppressor due to frequent inactivation in a variety of cancers. LKB1 directly binds and activates its downstream kinases including the AMP-activated protein kinase (AMPK) and AMPK-related kinases by phosphorylation, which has been intensively investigated for the past decades. An increasing number of studies has uncovered the posttranslational modifications (PTMs) of LKB1 and consequent changes in its localization, activity, and interaction with substrates. The alteration in LKB1 function as a consequence of genetic mutations and aberrant upstream signaling regulation leads to tumor development and progression. Here, we review current knowledge about the mechanism of LKB1 in cancer and the contributions of PTMs, such as phosphorylation, ubiquitination, SUMOylation, acetylation, prenylation, and others, to the regulation of LKB1 function, offering new insights into the therapeutic strategies in cancer.
Collapse
Affiliation(s)
- Lanlin Hu
- Department of Oncology & Cancer Institute, Sichuan Academy of Medical Sciences, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China; Department of Laboratory Medicine and Sichuan Provincial Key Laboratory for Human Disease Gene Study, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China; Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, University of Electronic Science and Technology of China, Chengdu, China
| | - Mingxin Liu
- Department of Oncology & Cancer Institute, Sichuan Academy of Medical Sciences, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China; Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, University of Electronic Science and Technology of China, Chengdu, China
| | - Bo Tang
- Department of Oncology & Cancer Institute, Sichuan Academy of Medical Sciences, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China; Department of Laboratory Medicine and Sichuan Provincial Key Laboratory for Human Disease Gene Study, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China; Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, University of Electronic Science and Technology of China, Chengdu, China
| | - Qiang Li
- Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, University of Electronic Science and Technology of China, Chengdu, China
| | - Bo-Syong Pan
- Department of Cancer Biology, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
| | - Chuan Xu
- Department of Oncology & Cancer Institute, Sichuan Academy of Medical Sciences, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China; Department of Laboratory Medicine and Sichuan Provincial Key Laboratory for Human Disease Gene Study, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China; Department of Cancer Biology, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA.
| | - Hui-Kuan Lin
- Department of Cancer Biology, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA.
| |
Collapse
|
32
|
Su X, Wang G, Zheng S, Ge C, Kong F, Wang C. Comprehensive Explorations of CCL28 in Lung Adenocarcinoma Immunotherapy and Experimental Validation. J Inflamm Res 2023; 16:1325-1342. [PMID: 37006812 PMCID: PMC10065022 DOI: 10.2147/jir.s399193] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Accepted: 03/17/2023] [Indexed: 03/29/2023] Open
Abstract
Background Chemokines have been reported to play an important role in cancer immunotherapy. This study aimed to explore the chemokines involved in lung cancer immunotherapy. Methods All the public data were downloaded from The Cancer Genome Atlas Program database. Quantitative real time-PCR was used to detect the mRNA level of specific molecules and Western blot was used for the protein level. Other experiments used include luciferase reporter experiments, flow cytometric analysis, Chromatin immunoprecipitation assay, ELISA and co-cultured system. Results We found that the CCL7, CCL11, CCL14, CCL24, CCL25, CCL26, CCL28 had a higher level, while the CCL17, CCL23 had a lower level in immunotherapy non-responders. Also, we found that immunotherapy non-responders had a higher level of CD56dim NK cells, NK cells, Th1 cells, Th2 cells and Treg, yet a lower level of iDC and Th17 cells. Biological enrichment analysis indicated that in the patients with high Treg infiltration, the pathways of pancreas beta cells, KRAS signaling, coagulation, WNT BETA catenin signaling, bile acid metabolism, interferon alpha response, hedgehog signaling, PI3K/AKT/mTOR signaling, apical surface, myogenesis were significantly enriched in. CCL7, CCL11, CCL26 and CCL28 were selected for further analysis. Compared with the patients with high CCL7, CCL11, CCL26 and CCL28 expression, the patients with low CCL7, CCL11, CCL26 and CCL28 expression had a better performance of immunotherapy response and this effect might partly be due to Treg cells. Furthermore, biological exploration and clinical correlation of CCL7, CCL11, CCL26 and CCL28 were conducted, Finally, CCL28 was selected for validation. Experiments showed that under the hypoxia condition, HIF-1α was upregulated, which can directly bind to the promoter region of CCL28 and lead to its higher level. Also, CCL28 secreted by lung cancer cells could induce Tregs infiltration. Conclusion Our study provides a novel insight focused on the chemokines in lung cancer immunotherapy. Also, CCL28 was identified as an underlying biomarker for lung cancer immunotherapy.
Collapse
Affiliation(s)
- Xiangyu Su
- School of Medicine, Southeast University, Nanjing, 210009, People’s Republic of China
- Department of Oncology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, 210009, People’s Republic of China
| | - Guoqing Wang
- Department of Pathology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, 210009, People’s Republic of China
| | - Shiya Zheng
- Department of Oncology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, 210009, People’s Republic of China
| | - Chang Ge
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, 210028, People’s Republic of China
| | - Fei Kong
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, 210028, People’s Republic of China
| | - Cailian Wang
- School of Medicine, Southeast University, Nanjing, 210009, People’s Republic of China
- Department of Oncology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, 210009, People’s Republic of China
- Correspondence: Cailian Wang, Email
| |
Collapse
|
33
|
Mortezaee K, Majidpoor J, Najafi S, Tasa D. Bypassing anti-PD-(L)1 therapy: Mechanisms and management strategies. Biomed Pharmacother 2023; 158:114150. [PMID: 36577330 DOI: 10.1016/j.biopha.2022.114150] [Citation(s) in RCA: 17] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 12/14/2022] [Accepted: 12/21/2022] [Indexed: 12/28/2022] Open
Abstract
Resistance to immune checkpoint inhibitors (ICIs) is a major issue of the current era in cancer immunotherapy. Immune evasion is a multi-factorial event, which occurs generally at a base of cold immunity. Despite advances in the field, there are still unsolved challenges about how to combat checkpoint hijacked by tumor cells and what are complementary treatment strategies to render durable anti-tumor outcomes. A point is that anti-programed death-1 receptor (PD-1)/anti-programmed death-ligand 1 (PD-L1) is not the solo path of immune escape, and responses in many types of solid tumors to the PD-1/PD-L1 inhibitors are not satisfactory. Thus, seeking mechanisms inter-connecting tumor with its immune ecosystem nearby unravel more about resistance mechanisms so as to develop methods for sustained reinvigoration of immune activity against cancer. In this review, we aimed to discuss about common and specific paths taken by tumor cells to evade immune surveillance, describing novel detection strategies, as well as suggesting some approaches to recover tumor sensitivity to the anti-PD-(L)1 therapy based on the current knowledge.
Collapse
Affiliation(s)
- Keywan Mortezaee
- Department of Anatomy, School of Medicine, Kurdistan University of Medical Sciences, Sanandaj, Iran.
| | - Jamal Majidpoor
- Department of Anatomy, School of Medicine, Infectious Diseases Research Center, Gonabad University of Medical Sciences, Gonabad, Iran
| | - Sajad Najafi
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Cellular and Molecular Biology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Davood Tasa
- Hepatopancreatobiliary Surgery Fellowship, Organ Transplantation Group, Massih Daneshvari Educational Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Department of Surgery, School of Medicine, Kurdistan University of Medical Sciences, Sanandaj, Iran
| |
Collapse
|
34
|
Zhang X, Wang X, Hou L, Xu Z, Liu Y, Wang X. Nanoparticles overcome adaptive immune resistance and enhance immunotherapy via targeting tumor microenvironment in lung cancer. Front Pharmacol 2023; 14:1130937. [PMID: 37033636 PMCID: PMC10080031 DOI: 10.3389/fphar.2023.1130937] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Accepted: 03/16/2023] [Indexed: 04/11/2023] Open
Abstract
Lung cancer is one of the common malignant cancers worldwide. Immune checkpoint inhibitor (ICI) therapy has improved survival of lung cancer patients. However, ICI therapy leads to adaptive immune resistance and displays resistance to PD-1/PD-L1 blockade in lung cancer, leading to less immune response of lung cancer patients. Tumor microenvironment (TME) is an integral tumor microenvironment, which is involved in immunotherapy resistance. Nanomedicine has been used to enhance the immunotherapy in lung cancer. In this review article, we described the association between TME and immunotherapy in lung cancer. We also highlighted the importance of TME in immunotherapy in lung cancer. Moreover, we discussed how nanoparticles are involved in regulation of TME to improve the efficacy of immunotherapy, including Nanomedicine SGT-53, AZD1080, Nanomodulator NRF2, Cisplatin nanoparticles, Au@PG, DPAICP@ME, SPIO NP@M-P, NBTXR3 nanoparticles, ARAC nanoparticles, Nano-DOX, MS NPs, Nab-paclitaxel, GNPs-hPD-L1 siRNA. Furthermore, we concluded that targeting TME by nanoparticles could be helpful to overcome resistance to PD-1/PD-L1 blockade in lung cancer.
Collapse
Affiliation(s)
- Xin Zhang
- Department of Pathology, China-Japan Union Hospital, Jilin University, Changchun, China
| | - Xuemei Wang
- Department of Pathology, China-Japan Union Hospital, Jilin University, Changchun, China
| | - Lijian Hou
- Department of Pathology, China-Japan Union Hospital, Jilin University, Changchun, China
| | - Zheng Xu
- Department of Pathology, China-Japan Union Hospital, Jilin University, Changchun, China
| | - Yu’e Liu
- School of Medicine, Tongji University Cancer Center, Shanghai Tenth People’s Hospital of Tongji University, Tongji University, Shanghai, China
| | - Xueju Wang
- Department of Pathology, China-Japan Union Hospital, Jilin University, Changchun, China
- *Correspondence: Xueju Wang,
| |
Collapse
|
35
|
Wei S, Chen Z, Ling X, Zhang W, Jiang L. Comprehensive analysis illustrating the role of PANoptosis-related genes in lung cancer based on bioinformatic algorithms and experiments. Front Pharmacol 2023; 14:1115221. [PMID: 36874021 PMCID: PMC9977813 DOI: 10.3389/fphar.2023.1115221] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2022] [Accepted: 02/07/2023] [Indexed: 02/18/2023] Open
Abstract
Background: Recently, PANoptosis has aroused the interest of researchers for its role in cancers. However, the studies that investigated PANoptosis in lung cancer are still few. Methods: The public data were mainly collected from The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus database. R software was utilized for the analysis of public data. Quantitative real-time (qRT) polymerase chain reaction (PCR) was used to measure the RNA level of FADD. The cell proliferation ability was evaluated using the CCK8, colony formation, and 5-ethynyl-2'-deoxyuridine (EdU) assays. Western blot was used to detect the protein level of specific molecules. Flow cytometry analysis and TUNEL staining were used to evaluate cell apoptosis. Results: In our study, we collected the PANoptosis-related genes from previous studies. Through series analysis, we identified the FADD, an adaptor of PANoptosis and apoptosis, for further analysis. Results showed that FADD is one of the prominent risk factors in lung cancer, mainly localized in nucleoplasm and cytosol. We next performed immune infiltration analysis and biological enrichment to illustrate the underlying cause of FADD in lung cancer. Subsequently, we discovered that the patients with a high level of FADD might respond worse to immunotherapy but better to AICAR, bortezomib, docetaxel, and gemcitabine. In vitro experiments indicated that inhibiting FADD could reduce significantly the ability of cancerous lung cells to proliferate. Meanwhile, we found that the knockdown of FADD promotes the apoptosis and pyroptosis. Ultimately, a prognosis signature was identified based on the FADD-regulated genes, which showed satisfactory prediction efficiency on patients with lung cancer. Conclusion: Our result can provide a novel direction for future studies focused on the role of PANoptosis in lung cancer.
Collapse
Affiliation(s)
- Shiyou Wei
- Department of Anesthesiology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Zhigang Chen
- Department of Anesthesiology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Xinyu Ling
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Wentian Zhang
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Lei Jiang
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| |
Collapse
|
36
|
CCR3 blockage elicits polyploidization associated with the signatures of epithelial-mesenchymal transition in carcinoma cell lines. Cancer Gene Ther 2023; 30:137-148. [PMID: 36123391 DOI: 10.1038/s41417-022-00532-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Revised: 08/12/2022] [Accepted: 09/02/2022] [Indexed: 01/19/2023]
Abstract
Malignant features such as the acquisition of metastatic ability, stemness of cells, and therapeutic resistance of cancer cells are associated with epithelial-mesenchymal transition (EMT) accompanied by changes in motility and morphology. Recent reports implicated that the formation of polyploid giant cancer cells (PGCCs) in human malignancy correlated with the EMT processes. Chemokines are often involved in the regulation of cancer cell migration into tissues, and various types of human cancers exhibit enhanced expression of chemokine receptors, which could augment intrinsic potentials such as invasive activity, proliferating ability, and survival capacity in cancer cells. Nevertheless, the contribution of CCR3 in malignant cancer cells is controversial because it is a well-known primal receptor for the migration of eosinophils, one of the cells of the innate immune system. Here, we explored the blockage of chemokine receptor CCR3 in carcinoma cell lines and found that inhibition of CCR3 induced the formation of polyploid giant cells and stabilization of β-catenin via the PI3K/Akt/GSK-3β signaling pathway, which are processes associated with EMT. As a result of CCR3 inhibition, converted cells acquired enhanced mobile and proliferation abilities. In summary, these data indicate that modulation of the CCR3/PI3K/Akt/GSK-3β signaling pathway regulates polyploidization associated with the EMT processes.
Collapse
|
37
|
Wang Z, Hou X, Li M, Ji R, Li Z, Wang Y, Guo Y, Liu D, Huang B, Du H. Active fractions of golden-flowered tea ( Camellia nitidissima Chi) inhibit epidermal growth factor receptor mutated non-small cell lung cancer via multiple pathways and targets in vitro and in vivo. Front Nutr 2022; 9:1014414. [PMID: 36386893 PMCID: PMC9649924 DOI: 10.3389/fnut.2022.1014414] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Accepted: 09/29/2022] [Indexed: 08/13/2023] Open
Abstract
As a medicine-food homology (MFH) plant, golden-flowered tea (Camellia nitidissima Chi, CNC) has many different pharmacologic activities and is known as "the queen of the tea family" and "the Panda of the Plant world". Several studies have revealed the pharmacologic effects of CNC crude extract, including anti-tumor, anti-oxidative and hepatoprotective activity. However, there are few studies on the anti-tumor active fractions and components of CNC, yet the underlying mechanism has not been investigated. Thus, we sought to verify the anti-non-small cell lung cancer (NSCLC) effects of four active fractions of CNC. Firstly, we determined the pharmacodynamic material basis of the four active fractions of CNC (Camellia. leave. saponins, Camellia. leave. polyphenols, Camellia. flower. saponins, Camellia. flower. polyphenols) by UPLC-Q-TOF-MS/MS and confirmed the differences in their specific compound contents. Then, MTT, colony formation assay and EdU incorporation assay confirmed that all fractions of CNC exhibit significant inhibitory on NSCLC, especially the Camellia. leave. saponins (CLS) fraction on EGFR mutated NSCLC cell lines. Moreover, transcriptome analysis revealed that the inhibition of NSCLC cell growth by CLS may be via three pathways, including "Cytokine-cytokine receptor interaction," "PI3K-Akt signaling pathway" and "MAPK signaling pathway." Subsequently, quantitative real-time PCR (RT-qPCR) and Western blot (WB) revealed TGFB2, INHBB, PIK3R3, ITGB8, TrkB and CACNA1D as the critical targets for the anti-tumor effects of CLS in vitro. Finally, the xenograft models confirmed that CLS treatment effectively suppressed tumor growth, and the key targets were also verified in vivo. These observations suggest that golden-flowered tea could be developed as a functional tea drink with anti-cancer ability, providing an essential molecular mechanism foundation for MFH medicine treating NSCLC.
Collapse
Affiliation(s)
- Ziling Wang
- Key Laboratory of Traditional Chinese Medicine Resources and Chemistry of Hubei Province, School of Pharmacy, Hubei University of Chinese Medicine, Wuhan, China
| | - Xiaoying Hou
- School of Medicine, Wuhan Institutes of Biomedical Sciences, Jianghan University, Wuhan, China
| | - Min Li
- Shenzhen Luohu Hospital Group Luohu People’s Hospital, The Third Affiliated Hospital of Shenzhen University, Shenzhen, China
| | - Rongsheng Ji
- Key Laboratory of Traditional Chinese Medicine Resources and Chemistry of Hubei Province, School of Pharmacy, Hubei University of Chinese Medicine, Wuhan, China
| | - Zhouyuan Li
- Key Laboratory of Traditional Chinese Medicine Resources and Chemistry of Hubei Province, School of Pharmacy, Hubei University of Chinese Medicine, Wuhan, China
| | - Yuqiao Wang
- Key Laboratory of Traditional Chinese Medicine Resources and Chemistry of Hubei Province, School of Pharmacy, Hubei University of Chinese Medicine, Wuhan, China
| | - Yujie Guo
- Key Laboratory of Traditional Chinese Medicine Resources and Chemistry of Hubei Province, School of Pharmacy, Hubei University of Chinese Medicine, Wuhan, China
| | - Dahui Liu
- Key Laboratory of Traditional Chinese Medicine Resources and Chemistry of Hubei Province, School of Pharmacy, Hubei University of Chinese Medicine, Wuhan, China
| | - Bisheng Huang
- Key Laboratory of Traditional Chinese Medicine Resources and Chemistry of Hubei Province, School of Pharmacy, Hubei University of Chinese Medicine, Wuhan, China
| | - Hongzhi Du
- Key Laboratory of Traditional Chinese Medicine Resources and Chemistry of Hubei Province, School of Pharmacy, Hubei University of Chinese Medicine, Wuhan, China
| |
Collapse
|
38
|
Yang Y, Cui H, Li D, Gao Y, Chen L, Zhou C, Feng M, Tu W, Li S, Chen X, Hao B, Li L, Cao Y. Prognosis and Immunological Characteristics of PGK1 in Lung Adenocarcinoma: A Systematic Analysis. Cancers (Basel) 2022; 14:5228. [PMID: 36358653 PMCID: PMC9653683 DOI: 10.3390/cancers14215228] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Revised: 10/13/2022] [Accepted: 10/21/2022] [Indexed: 01/30/2024] Open
Abstract
Background: Aerobic glycolysis plays a key role in tumor metabolic reprogramming to reshape the immune microenvironment. The phosphoglycerate kinase 1 (PGK1) gene codes a glycolytic enzyme that converts 1,3-diphosphoglycerate to 3-phosphoglycerate. However, in lung adenocarcinoma (LUAD), the role of PGK1 in altering the tumor microenvironment (TME) has not yet been determined. Methods: Raw data, including bulk DNA and mRNA-seq data, methylation modification data, single-cell RNA-seq data, proteomics data, clinical case characteristics survival, immunotherapy data, and so on, were obtained from multiple independent public data sets. These data were reanalyzed to uncover the prognosis and immunological characteristics of PGK1 in LUAD. Results: We found that PGK1 mRNA and protein were considerably over-expressed in LUAD compared to normal tissue and that high PGK1 expression is associated with poorer prognostic outcomes in LUAD. The enrichment analysis of PGK1 co-expressed genes in lung adenocarcinoma revealed that PGK1 may be involved in hypoxia, metabolism, DNA synthesis, cell cycle, PI3K/AKT, and various immune and inflammatory signaling pathways. Furthermore, PGK1 is also linked to the recruitment of numerous immune cells, including aDC (dendritic cells), macrophages, and neutrophils. More importantly, PGK1 was highly expressed in immunosuppressive cells, including M2 macrophages, Tregs, and exhausted T cells, among others. Finally, higher PGK1 expression indicated significant correlations to immune checkpoints, TMB (tumor mutation burden), and high response to immunotherapy. Conclusions: The presented findings imply that PGK1, as a glycolysis core gene, may be important for the modification of the immune microenvironment by interacting with the tumor metabolism. The results of this study provide clues for a potential immunometabolic combination therapy strategy in LUAD, for which more experimental and clinical translational research is needed.
Collapse
Affiliation(s)
- Yuechao Yang
- Department of Neurosurgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Huanhuan Cui
- Department of Neurosurgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Deheng Li
- Department of Neurosurgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Yang Gao
- Department of Neurosurgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Lei Chen
- Department of Neurosurgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Changshuai Zhou
- Department of Neurosurgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Mingtao Feng
- Department of Neurosurgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Wenjing Tu
- Department of Neurosurgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Sen Li
- Department of Neurosurgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Xin Chen
- Department of Neurosurgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Bin Hao
- Department of Neurosurgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Liangdong Li
- Department of Neurosurgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Yiqun Cao
- Department of Neurosurgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| |
Collapse
|
39
|
Clinical relevance and therapeutic aspects of professional antigen-presenting cells in lung cancer. MEDICAL ONCOLOGY (NORTHWOOD, LONDON, ENGLAND) 2022; 39:237. [PMID: 36175603 DOI: 10.1007/s12032-022-01841-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Accepted: 09/01/2022] [Indexed: 10/14/2022]
Abstract
Lung cancer stays the preeminent cause of death worldwide. Despite recent advancements in chemotherapy, radiotherapy, and immunotherapy, the survival rate for people with advanced stages of the disease is still appalling. Moreover, there is a severe lack of reliable prognoses and indicators for classification in newly developed immunotherapies. A better understanding of immune cells is necessary to harness immune response mechanisms for therapeutic effects. Professional antigen-presenting cells are responsible for determining the fate of the immune response through the antigen processing and presentation pathway (APP). The most professional antigen-presenting cells (APC) include the dendritic cells (DC), macrophages, and B cells, which present antigens to the T-helper cells. Dendritic cells are significantly explored as a tool for immunotherapy owing to their precise ability to provoke and alter T-cell responses. Moreover, the role of tumor-associated macrophages (TAMs), an abundant leukocyte in lung cancer, is also a potential target for adjuvant anti-cancer therapies. In this review, we summarize the recent advances in our understanding of the various types of immunotherapy mapped out via professional antigen-presenting cells in lung cancer.
Collapse
|
40
|
Dai R, Tao R, Li X, Shang T, Zhao S, Ren Q. Expression profiling of mRNA and functional network analyses of genes regulated by human papilloma virus E6 and E7 proteins in HaCaT cells. Front Microbiol 2022; 13:979087. [PMID: 36188003 PMCID: PMC9515614 DOI: 10.3389/fmicb.2022.979087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Accepted: 08/16/2022] [Indexed: 11/21/2022] Open
Abstract
Human papillomavirus (HPV) oncogenes E6 and E7 are essential for HPV-related cancer development. Here, we developed a cell line model using lentiviruses for transfection of the HPV16 oncogenes E6 and E7 and investigated the differences in mRNA expression during cell adhesion and chemokine secretion. Subsequently, RNA sequencing (RNA-seq) analysis was performed to explore the differences in mRNA expression. Compared to levels in the control group, 2,905 differentially expressed mRNAs (1,261 downregulated and 1,644 upregulated) were identified in the HaCaT-HPV16E6E7 cell line. To predict the functions of these differentially expressed genes (DEGs) the Gene Ontology and Kyoto Encyclopedia of Genes and Genomes databases were used. Protein–protein interactions were established, and the hub gene was identified based on this network. Real-time quantitative-PCR (RT-qPCR) was conducted to confirm the levels of 14 hub genes, which were consistent with the RNA-seq data. According to this, we found that these DEGs participate in the extracellular matrix (ECM), cell adhesion, immune control, and cancer-related signaling pathways. Currently, an increasing number of clinicians depend on E6/E7mRNA results to make a comprehensive judgment of cervical precancerous lesions. In this study, 14 hub genes closely related to the expression of cell adhesion ability and chemokines were analyzed in HPV16E6E7-stably expressing cell lines, which will open up new research ideas for targeting E6E7 in the treatment of HPV-related cancers.
Collapse
Affiliation(s)
- Renjinming Dai
- The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Ran Tao
- Laboratory of Clinical Applied Anatomy, Department of Human Anatomy, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China
| | - Xiu Li
- The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Tingting Shang
- The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Shixian Zhao
- The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Qingling Ren
- The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
- *Correspondence: Qingling Ren,
| |
Collapse
|
41
|
Papadas A, Deb G, Cicala A, Officer A, Hope C, Pagenkopf A, Flietner E, Morrow ZT, Emmerich P, Wiesner J, Arauz G, Bansal V, Esbona K, Capitini CM, Matkowskyj KA, Deming DA, Politi K, Abrams SI, Harismendy O, Asimakopoulos F. Stromal remodeling regulates dendritic cell abundance and activity in the tumor microenvironment. Cell Rep 2022; 40:111201. [PMID: 35977482 PMCID: PMC9402878 DOI: 10.1016/j.celrep.2022.111201] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Revised: 06/10/2022] [Accepted: 07/20/2022] [Indexed: 11/24/2022] Open
Abstract
Stimulatory type 1 conventional dendritic cells (cDC1s) engage in productive interactions with CD8+ effectors along tumor-stroma boundaries. The paradoxical accumulation of “poised” cDC1s within stromal sheets is unlikely to simply reflect passive exclusion from tumor cores. Drawing parallels with embryonic morphogenesis, we hypothesized that invasive margin stromal remodeling generates developmentally conserved cell fate cues that regulate cDC1 behavior. We find that, in human T cell-inflamed tumors, CD8+ T cells penetrate tumor nests, whereas cDC1s are confined within adjacent stroma that recurrently displays site-specific proteolysis of the matrix proteoglycan versican (VCAN), an essential organ-sculpting modification in development. VCAN is necessary, and its proteolytic fragment (matrikine) versikine is sufficient for cDC1 accumulation. Versikine does not influence tumor-seeding pre-DC differentiation; rather, it orchestrates a distinctive cDC1 activation program conferring exquisite sensitivity to DNA sensing, supported by atypical innate lymphoid cells. Thus, peritumoral stroma mimicking embryonic provisional matrix remodeling regulates cDC1 abundance and activity to elicit T cell-inflamed tumor microenvironments. T cell-inflamed tumor microenvironments are a prerequisite for immunotherapy efficacy; however, why some tumors are inflamed and others not remains poorly understood. Papadas et al. link stromal reaction dynamics with T cell-induced inflammation. Peritumoral stroma emulating embryonic provisional matrix remodeling regulates cDC1-NK-CD8+ crosstalk to promote T cell repriming and penetration into tumor nests.
Collapse
Affiliation(s)
- Athanasios Papadas
- Division of Blood and Marrow Transplantation, Department of Medicine, University of California, San Diego (UCSD), La Jolla, CA, USA; Moores Cancer Center, University of California, San Diego (UCSD), La Jolla, CA, USA; Cellular and Molecular Pathology Graduate Program, University of Wisconsin-Madison, Madison, WI, USA
| | - Gauri Deb
- Division of Blood and Marrow Transplantation, Department of Medicine, University of California, San Diego (UCSD), La Jolla, CA, USA; Moores Cancer Center, University of California, San Diego (UCSD), La Jolla, CA, USA
| | - Alexander Cicala
- Division of Blood and Marrow Transplantation, Department of Medicine, University of California, San Diego (UCSD), La Jolla, CA, USA; Moores Cancer Center, University of California, San Diego (UCSD), La Jolla, CA, USA
| | - Adam Officer
- Moores Cancer Center, University of California, San Diego (UCSD), La Jolla, CA, USA; Division of Biomedical Informatics, Department of Medicine, University of California, San Diego (UCSD), Moores Cancer Center, La Jolla, CA, USA; Bioinformatics and Systems Biology Graduate Program, University of California, San Diego (UCSD), La Jolla, CA, USA
| | - Chelsea Hope
- Cellular and Molecular Pathology Graduate Program, University of Wisconsin-Madison, Madison, WI, USA; Division of Hematology and Oncology, Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA; UW Carbone Cancer Center, University of Wisconsin-Madison, Madison, WI, USA
| | - Adam Pagenkopf
- Division of Hematology and Oncology, Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA; UW Carbone Cancer Center, University of Wisconsin-Madison, Madison, WI, USA
| | - Evan Flietner
- Cellular and Molecular Pathology Graduate Program, University of Wisconsin-Madison, Madison, WI, USA; Division of Hematology and Oncology, Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA; UW Carbone Cancer Center, University of Wisconsin-Madison, Madison, WI, USA
| | - Zachary T Morrow
- Division of Hematology and Oncology, Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA; UW Carbone Cancer Center, University of Wisconsin-Madison, Madison, WI, USA
| | - Philip Emmerich
- Cellular and Molecular Pathology Graduate Program, University of Wisconsin-Madison, Madison, WI, USA; UW Carbone Cancer Center, University of Wisconsin-Madison, Madison, WI, USA
| | - Joshua Wiesner
- Division of Blood and Marrow Transplantation, Department of Medicine, University of California, San Diego (UCSD), La Jolla, CA, USA; Moores Cancer Center, University of California, San Diego (UCSD), La Jolla, CA, USA
| | - Garrett Arauz
- Division of Blood and Marrow Transplantation, Department of Medicine, University of California, San Diego (UCSD), La Jolla, CA, USA; Moores Cancer Center, University of California, San Diego (UCSD), La Jolla, CA, USA
| | - Varun Bansal
- Division of Blood and Marrow Transplantation, Department of Medicine, University of California, San Diego (UCSD), La Jolla, CA, USA; Moores Cancer Center, University of California, San Diego (UCSD), La Jolla, CA, USA
| | - Karla Esbona
- UW Carbone Cancer Center, University of Wisconsin-Madison, Madison, WI, USA; Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, WI, USA
| | - Christian M Capitini
- UW Carbone Cancer Center, University of Wisconsin-Madison, Madison, WI, USA; Division of Hematology and Oncology, Department of Pediatrics, University of Wisconsin-Madison, Madison, WI, USA
| | - Kristina A Matkowskyj
- UW Carbone Cancer Center, University of Wisconsin-Madison, Madison, WI, USA; Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, WI, USA
| | - Dustin A Deming
- Division of Hematology and Oncology, Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA; UW Carbone Cancer Center, University of Wisconsin-Madison, Madison, WI, USA; McArdle Laboratory for Cancer Research, University of Wisconsin-Madison, Madison, WI, USA
| | - Katerina Politi
- Department of Pathology, Yale School of Medicine, New Haven, CT, USA; Department of Medicine, Yale School of Medicine, New Haven, CT, USA; Yale Cancer Center, Yale School of Medicine, New Haven, CT, USA
| | - Scott I Abrams
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Olivier Harismendy
- Moores Cancer Center, University of California, San Diego (UCSD), La Jolla, CA, USA; Division of Biomedical Informatics, Department of Medicine, University of California, San Diego (UCSD), Moores Cancer Center, La Jolla, CA, USA
| | - Fotis Asimakopoulos
- Division of Blood and Marrow Transplantation, Department of Medicine, University of California, San Diego (UCSD), La Jolla, CA, USA; Moores Cancer Center, University of California, San Diego (UCSD), La Jolla, CA, USA.
| |
Collapse
|
42
|
Liu Z, Yu Z, Chen D, Verma V, Yuan C, Wang M, Wang F, Fan Q, Wang X, Li Y, Ma Y, Wu M, Yu J. Pivotal roles of tumor-draining lymph nodes in the abscopal effects from combined immunotherapy and radiotherapy. CANCER COMMUNICATIONS (LONDON, ENGLAND) 2022; 42:971-986. [PMID: 35962977 PMCID: PMC9558691 DOI: 10.1002/cac2.12348] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 05/28/2022] [Accepted: 07/27/2022] [Indexed: 11/10/2022]
Abstract
BACKGROUND Currently, due to synergy enhancement of anti-tumor effects and potent stimulation of abscopal effects, combination therapy with irradiation and programmed cell death protein 1/programmed death-ligand 1 (PD-1/PD-L1) immune checkpoint inhibition (immuno-radiotherapy, iRT) has revolutionized the therapeutic guidelines. It has been demonstrated that tumor-draining lymph nodes (TDLN) are essential for effective antitumor immunity induced by radiotherapy, immunotherapy, or iRT. Given that the function of TDLN in iRT remains unclear, this study aimed to investigate the function and mechanism of TDLN in iRT-induced abscopal effects. METHODS The function of TDLN was evaluated using unilateral or bilateral MC38 and B16F10 subcutaneous tumor models with or without indicated TDLN. The flow cytometry, multiple immunofluorescence analysis, and NanoString analysis were utilized to detect the composition and function of the immune cells in the primary and abscopal tumor microenvironment. Additionally, we tempted to interrogate the possible mechanisms via RNA-sequencing of tumor-infiltrating lymphocytes and TDLN. RESULTS TDLN deficiency impaired the control of tumor growth by monotherapy. Bilateral TDLN removal rather than unilateral TDLN removal substantially curtailed iRT-stimulated anti-tumor and abscopal effects. Furthermore, in the absence of TDLN, the infiltration of CD45+ and CD8+ T cells was substantially reduced in both primary and abscopal tumors, and the anti-tumor function of CD8+ T cells was attenuated as well. Additionally, the polarization of tumor-associated macrophages in primary and abscopal tumors were found to be dependent on intact bilateral TDLN. RNA-sequencing data indicated that impaired infiltration and anti-tumor effects of immune cells partially attributed to the altered secretion of components from the tumor microenvironment. CONCLUSIONS TDLN play a critical role in iRT by promoting the infiltration of CD8+ T cells and maintaining the M1/M2 macrophage ratio.
Collapse
Affiliation(s)
- Zhaoyun Liu
- Department of Oncology, Shandong University Cancer Center, Jinan, Shandong, 250117, P. R. China.,Department of Radiation Oncology and Shandong Provincial Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, 250117, P. R. China.,Breast Cancer Center, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, 250117, P. R. China
| | - Zhiyong Yu
- Breast Cancer Center, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, 250117, P. R. China
| | - Dawei Chen
- Department of Oncology, Shandong University Cancer Center, Jinan, Shandong, 250117, P. R. China.,Department of Radiation Oncology and Shandong Provincial Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, 250117, P. R. China
| | - Vivek Verma
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, 77030, United States
| | - Chenxi Yuan
- Department of Radiation Oncology and Shandong Provincial Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, 250117, P. R. China
| | - Minglei Wang
- Department of Oncology, Shandong University Cancer Center, Jinan, Shandong, 250117, P. R. China.,Department of Radiation Oncology and Shandong Provincial Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, 250117, P. R. China
| | - Fei Wang
- Department of Oncology, Shandong University Cancer Center, Jinan, Shandong, 250117, P. R. China.,Department of Radiation Oncology and Shandong Provincial Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, 250117, P. R. China
| | - Qing Fan
- Department of Pharmacy, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, 250117, P. R. China
| | - Xingwu Wang
- Department of Oncology, Shandong University Cancer Center, Jinan, Shandong, 250117, P. R. China.,Department of Radiation Oncology and Shandong Provincial Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, 250117, P. R. China
| | - Yang Li
- Department of Oncology, Shandong University Cancer Center, Jinan, Shandong, 250117, P. R. China.,Department of Radiation Oncology and Shandong Provincial Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, 250117, P. R. China
| | - Yuequn Ma
- Department of Oncology, Shandong University Cancer Center, Jinan, Shandong, 250117, P. R. China.,Department of Radiation Oncology and Shandong Provincial Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, 250117, P. R. China
| | - Meng Wu
- Department of Oncology, Shandong University Cancer Center, Jinan, Shandong, 250117, P. R. China.,Department of Radiation Oncology and Shandong Provincial Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, 250117, P. R. China
| | - Jinming Yu
- Department of Oncology, Shandong University Cancer Center, Jinan, Shandong, 250117, P. R. China.,Department of Radiation Oncology and Shandong Provincial Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, 250117, P. R. China.,Research Unit of Radiation Oncology, Chinese Academy of Medical Sciences, Jinan, Shandong, 250117, P. R. China
| |
Collapse
|
43
|
Queiroz AL, Dantas E, Ramsamooj S, Murthy A, Ahmed M, Zunica ERM, Liang RJ, Murphy J, Holman CD, Bare CJ, Ghahramani G, Wu Z, Cohen DE, Kirwan JP, Cantley LC, Axelrod CL, Goncalves MD. Blocking ActRIIB and restoring appetite reverses cachexia and improves survival in mice with lung cancer. Nat Commun 2022; 13:4633. [PMID: 35941104 PMCID: PMC9360437 DOI: 10.1038/s41467-022-32135-0] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Accepted: 07/13/2022] [Indexed: 12/30/2022] Open
Abstract
Cancer cachexia is a common, debilitating condition with limited therapeutic options. Using an established mouse model of lung cancer, we find that cachexia is characterized by reduced food intake, spontaneous activity, and energy expenditure accompanied by muscle metabolic dysfunction and atrophy. We identify Activin A as a purported driver of cachexia and treat with ActRIIB-Fc, a decoy ligand for TGF-β/activin family members, together with anamorelin (Ana), a ghrelin receptor agonist, to reverse muscle dysfunction and anorexia, respectively. Ana effectively increases food intake but only the combination of drugs increases lean mass, restores spontaneous activity, and improves overall survival. These beneficial effects are limited to female mice and are dependent on ovarian function. In agreement, high expression of Activin A in human lung adenocarcinoma correlates with unfavorable prognosis only in female patients, despite similar expression levels in both sexes. This study suggests that multimodal, sex-specific, therapies are needed to reverse cachexia.
Collapse
Affiliation(s)
- Andre Lima Queiroz
- Division of Endocrinology, Department of Medicine, Weill Cornell Medicine, New York, NY, 10065, USA
- Meyer Cancer Center, Weill Cornell Medicine, New York, NY, 10065, USA
| | - Ezequiel Dantas
- Division of Endocrinology, Department of Medicine, Weill Cornell Medicine, New York, NY, 10065, USA
- Meyer Cancer Center, Weill Cornell Medicine, New York, NY, 10065, USA
| | - Shakti Ramsamooj
- Division of Endocrinology, Department of Medicine, Weill Cornell Medicine, New York, NY, 10065, USA
- Meyer Cancer Center, Weill Cornell Medicine, New York, NY, 10065, USA
| | - Anirudh Murthy
- Division of Endocrinology, Department of Medicine, Weill Cornell Medicine, New York, NY, 10065, USA
- Meyer Cancer Center, Weill Cornell Medicine, New York, NY, 10065, USA
| | - Mujmmail Ahmed
- Division of Endocrinology, Department of Medicine, Weill Cornell Medicine, New York, NY, 10065, USA
- Meyer Cancer Center, Weill Cornell Medicine, New York, NY, 10065, USA
| | | | - Roger J Liang
- Division of Endocrinology, Department of Medicine, Weill Cornell Medicine, New York, NY, 10065, USA
- Meyer Cancer Center, Weill Cornell Medicine, New York, NY, 10065, USA
| | - Jessica Murphy
- Meyer Cancer Center, Weill Cornell Medicine, New York, NY, 10065, USA
- Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Corey D Holman
- Division of Gastroenterology and Hepatology, Department of Medicine, Weill Cornell Medicine, New York, NY, 10065, USA
| | - Curtis J Bare
- Division of Gastroenterology and Hepatology, Department of Medicine, Weill Cornell Medicine, New York, NY, 10065, USA
| | - Gregory Ghahramani
- Weill Cornell Graduate School of Medical Sciences, Weill Cornell Medicine, New York, NY, 10065, USA
| | - Zhidan Wu
- Internal Medicine Research Unit, Pfizer Global R&D, Cambridge, MA, USA
| | - David E Cohen
- Division of Gastroenterology and Hepatology, Department of Medicine, Weill Cornell Medicine, New York, NY, 10065, USA
| | - John P Kirwan
- Pennington Biomedical Research Center, Baton Rouge, LA, 70808, USA
| | - Lewis C Cantley
- Meyer Cancer Center, Weill Cornell Medicine, New York, NY, 10065, USA
| | | | - Marcus D Goncalves
- Division of Endocrinology, Department of Medicine, Weill Cornell Medicine, New York, NY, 10065, USA.
- Meyer Cancer Center, Weill Cornell Medicine, New York, NY, 10065, USA.
| |
Collapse
|
44
|
Wang W, Lozar T, Golfinos AE, Lee D, Gronski E, Ward-Shaw E, Hayes M, Bruce JY, Kimple RJ, Hu R, Harari PM, Xu J, Keske A, Sondel PM, Fitzpatrick MB, Dinh HQ, Lambert PF. Stress Keratin 17 Expression in Head and Neck Cancer Contributes to Immune Evasion and Resistance to Immune-Checkpoint Blockade. Clin Cancer Res 2022; 28:2953-2968. [PMID: 35621713 PMCID: PMC9250640 DOI: 10.1158/1078-0432.ccr-21-3039] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2021] [Revised: 11/25/2021] [Accepted: 04/20/2022] [Indexed: 01/03/2023]
Abstract
PURPOSE We investigated whether in human head and neck squamous cell carcinoma (HNSCC) high levels of expression of stress keratin 17 (K17) are associated with poor survival and resistance to immunotherapy. EXPERIMENTAL DESIGN We investigated the role of K17 in regulating both the tumor microenvironment and immune responsiveness of HNSCC using a syngeneic mouse HNSCC model, MOC2. MOC2 gives rise to immunologically cold tumors that are resistant to immune-checkpoint blockade (ICB). We engineered multiple, independent K17 knockout (KO) MOC2 cell lines and monitored their growth and response to ICB. We also measured K17 expression in human HNSCC of patients undergoing ICB. RESULTS MOC2 tumors were found to express K17 at high levels. When knocked out for K17 (K17KO MOC2), these cells formed tumors that grew slowly or spontaneously regressed and had a high CD8+ T-cell infiltrate in immunocompetent syngeneic C57BL/6 mice compared with parental MOC2 tumors. This phenotype was reversed when we depleted mice for T cells. Whereas parental MOC2 tumors were resistant to ICB treatment, K17KO MOC2 tumors that did not spontaneously regress were eliminated upon ICB treatment. In a cohort of patients with HNSCC receiving pembrolizumab, high K17 expression correlated with poor response. Single-cell RNA-sequencing analysis revealed broad differences in the immune landscape of K17KO MOC2 tumors compared with parental MOC2 tumors, including differences in multiple lymphoid and myeloid cell types. CONCLUSIONS We demonstrate that K17 expression in HNSCC contributes to immune evasion and resistance to ICB treatment by broadly altering immune landscapes of tumors.
Collapse
Affiliation(s)
- Wei Wang
- McArdle Laboratory for Cancer Research/ Department of Oncology, University of Wisconsin School of Medicine and Public Health, Madison WI, USA
| | - Taja Lozar
- McArdle Laboratory for Cancer Research/ Department of Oncology, University of Wisconsin School of Medicine and Public Health, Madison WI, USA,University of Ljubljana, Ljubljana, Slovenia
| | - Athena E. Golfinos
- McArdle Laboratory for Cancer Research/ Department of Oncology, University of Wisconsin School of Medicine and Public Health, Madison WI, USA
| | - Denis Lee
- McArdle Laboratory for Cancer Research/ Department of Oncology, University of Wisconsin School of Medicine and Public Health, Madison WI, USA
| | - Ellery Gronski
- McArdle Laboratory for Cancer Research/ Department of Oncology, University of Wisconsin School of Medicine and Public Health, Madison WI, USA
| | - Ella Ward-Shaw
- McArdle Laboratory for Cancer Research/ Department of Oncology, University of Wisconsin School of Medicine and Public Health, Madison WI, USA
| | - Mitchell Hayes
- McArdle Laboratory for Cancer Research/ Department of Oncology, University of Wisconsin School of Medicine and Public Health, Madison WI, USA
| | - Justine Y. Bruce
- Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA
| | - Randall J Kimple
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison WI, USA
| | - Rong Hu
- Department of Pathology and Laboratory Medicine, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA
| | - Paul M. Harari
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison WI, USA
| | - Jin Xu
- Department of Pathology and Laboratory Medicine, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA
| | - Aysenur Keske
- Department of Pathology and Laboratory Medicine, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA
| | - Paul M. Sondel
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison WI, USA,University of Wisconsin Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health, Madison
| | - Megan B. Fitzpatrick
- Department of Pathology and Laboratory Medicine, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA
| | - Huy Q. Dinh
- McArdle Laboratory for Cancer Research/ Department of Oncology, University of Wisconsin School of Medicine and Public Health, Madison WI, USA,University of Wisconsin Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health, Madison,Department of Biostatistics and Medical Informatics, University of Wisconsin School of Medicine and Public Health, Madison WI, USA
| | - Paul F. Lambert
- McArdle Laboratory for Cancer Research/ Department of Oncology, University of Wisconsin School of Medicine and Public Health, Madison WI, USA,University of Wisconsin Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health, Madison
| |
Collapse
|
45
|
Guo Q, Liu XL, Liu HS, Luo XY, Yuan Y, Ji YM, Liu T, Guo JL, Zhang J. The Risk Model Based on the Three Oxidative Stress-Related Genes Evaluates the Prognosis of LAC Patients. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:4022896. [PMID: 35783192 PMCID: PMC9246616 DOI: 10.1155/2022/4022896] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Accepted: 05/30/2022] [Indexed: 12/20/2022]
Abstract
Background Oxidative stress plays a role in carcinogenesis. This study explores the roles of oxidative stress-related genes (OSRGs) in lung adenocarcinoma (LAC). Besides, we construct a risk score model of OSRGs that evaluates the prognosis of LAC patients. Methods OSRGs were downloaded from the Gene Set Enrichment Analysis (GSEA) website. The expression levels of OSRGs were confirmed in LAC tissues of the TCGA database. GO and KEGG analyses were used to evaluate the roles and mechanisms of oxidative stress-related differentially expressed genes (DEGs). Survival, ROC, Cox analysis, and AIC method were used to screen the prognostic DEGs in LAC patients. Subsequently, we constructed a risk score model of OSRGs and a nomogram. Further, this work investigated the values of the risk score model in LAC progression and the relationship between the risk score model and immune infiltration. Results We discovered 163 oxidative stress-related DEGs in LAC, involving cellular response to oxidative stress and reactive oxygen species. Besides, the areas under the curve of CCNA2, CDC25C, ERO1A, CDK1, PLK1, ITGB4, and GJB2 were 0.970, 0.984, 0.984, 0.945, 0.984, 0.771, and 0.959, respectively. This indicates that these OSRGs have diagnosis values of LAC and are significantly related to the overall survival of LAC patients. ERO1A, CDC25C, and ITGB4 overexpressions were independent risk factors for the poor prognosis of LAC patients and were associated with risk scores in the risk model. High-risk score levels affected the poor prognosis of LAC patients. Notably, a high-risk score may be implicated in LAC progression via cell cycle, DNA replication, mismatch repair, and other mechanisms. Further, ERO1A, CDC25C, and ITGB4 expression levels were related to the immune infiltrating cells of LAC, including mast cells, NK cells, and CD8 T cells. Conclusion In summary, ERO1A, CDC25C, and ITGB4 of OSRGs are associated with poor prognosis of LAC patients. We confirmed that the risk model based on the ERO1A, CDC25C, and ITGB4 is expected to assess the prognosis of LAC patients.
Collapse
Affiliation(s)
- Qiang Guo
- Department of Cardiothoracic Surgery, Taihe Hospital, Hubei University of Medicine, Shiyan 442012, Hubei Province, China
| | - Xiao-Li Liu
- Department of Ultrasound, The People's Hospital of Jianyang City, Jianyang 641400, Sichuan Province, China
| | - Hua-Song Liu
- Department of Cardiothoracic Surgery, Taihe Hospital, Hubei University of Medicine, Shiyan 442012, Hubei Province, China
| | - Xiang-Yu Luo
- Department of Cardiothoracic Surgery, Taihe Hospital, Hubei University of Medicine, Shiyan 442012, Hubei Province, China
| | - Ye Yuan
- Department of Cardiothoracic Surgery, Taihe Hospital, Hubei University of Medicine, Shiyan 442012, Hubei Province, China
| | - Yan-Mei Ji
- Department of Cardiothoracic Surgery, Taihe Hospital, Hubei University of Medicine, Shiyan 442012, Hubei Province, China
| | - Tao Liu
- Department of Cardiothoracic Surgery, Taihe Hospital, Hubei University of Medicine, Shiyan 442012, Hubei Province, China
| | - Jia-Long Guo
- Department of Cardiothoracic Surgery, Taihe Hospital, Hubei University of Medicine, Shiyan 442012, Hubei Province, China
| | - Jun Zhang
- Department of Cardiothoracic Surgery, Taihe Hospital, Hubei University of Medicine, Shiyan 442012, Hubei Province, China
| |
Collapse
|
46
|
Yang H, Jian L, Jin Q, Xia K, Cai-Ru W, Jun S, Chen H, Wei W, Ben-Jing S, Shi-Hong L, Shi-Wei L, Juan W, Wei Z. CCL7 playing a dominant role in recruiting early OCPs to facilitate osteolysis at metastatic site of colorectal cancer. Cell Commun Signal 2022; 20:94. [PMID: 35715847 PMCID: PMC9205124 DOI: 10.1186/s12964-022-00867-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Accepted: 03/27/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Chemoattractant is critical to recruitment of osteoclast precursors and stimulates tumor bone metastasis. However, the role of chemoattractant in bone metastasis of colorectal cancer (CRC) is still unclear. METHODS Histochemistry analysis and TRAP staining were utilized to detect the bone resorption and activation of osteoclasts (OCs) after administration of CCL7 neutralizing antibody or CCR1 siRNA. qRT-PCR analysis and ELISA assay were performed to detect the mRNA level and protein level of chemoattractant. BrdU assay and Tunel assay were used to detect the proliferation and apoptosis of osteoclast precursors (OCPs). The migration of OCPs was detected by Transwell assay. Western blots assay was performed to examine the protein levels of pathways regulating the expression of CCL7 or CCR1. RESULTS OCPs-derived CCL7 was significantly upregulated in bone marrow after bone metastasis of CRC. Blockage of CCL7 efficiently prevented bone resorption. Administration of CCL7 promoted the migration of OCPs. Lactate promoted the expression of CCL7 through JNK pathway. In addition, CCR1 was the most important receptor of CCL7. CONCLUSION Our study indicates the essential role of CCL7-CCR1 signaling for recruitment of OCPs in early bone metastasis of CRC. Targeting CCL7 or CCR1 could restore the bone volume, which could be a potential therapeutical target. Video Abstract.
Collapse
Affiliation(s)
- He Yang
- Department of Orthopedics, General Hospital of Western Theater Command, Rongdu Avenue No. 270, Chengdu, 610000, People's Republic of China.,Chengdu Medical College, Rongdu Avenue No. 601, Chengdu, 610000, People's Republic of China
| | - Li Jian
- Department of Orthopedics, General Hospital of Western Theater Command, Rongdu Avenue No. 270, Chengdu, 610000, People's Republic of China.,Chengdu Medical College, Rongdu Avenue No. 601, Chengdu, 610000, People's Republic of China
| | - Qian Jin
- Department of Orthopedics, General Hospital of Western Theater Command, Rongdu Avenue No. 270, Chengdu, 610000, People's Republic of China
| | - Kang Xia
- Department of Orthopedics, General Hospital of Western Theater Command, Rongdu Avenue No. 270, Chengdu, 610000, People's Republic of China.
| | - Wang Cai-Ru
- Department of Orthopedics, General Hospital of Western Theater Command, Rongdu Avenue No. 270, Chengdu, 610000, People's Republic of China
| | - Sheng Jun
- Department of Orthopedics, General Hospital of Western Theater Command, Rongdu Avenue No. 270, Chengdu, 610000, People's Republic of China
| | - Huang Chen
- Department of Orthopedics, General Hospital of Western Theater Command, Rongdu Avenue No. 270, Chengdu, 610000, People's Republic of China
| | - Wang Wei
- Department of Orthopedics, General Hospital of Western Theater Command, Rongdu Avenue No. 270, Chengdu, 610000, People's Republic of China
| | - Song Ben-Jing
- Department of Orthopedics, General Hospital of Western Theater Command, Rongdu Avenue No. 270, Chengdu, 610000, People's Republic of China
| | - Li Shi-Hong
- Department of Orthopedics, General Hospital of Western Theater Command, Rongdu Avenue No. 270, Chengdu, 610000, People's Republic of China
| | - Long Shi-Wei
- Department of Orthopedics, General Hospital of Western Theater Command, Rongdu Avenue No. 270, Chengdu, 610000, People's Republic of China
| | - Wu Juan
- Department of Pharmacy, General Hospital of Western Theater Command, Rongdu Avenue No. 270, Chengdu, 610000, People's Republic of China.
| | - Zheng Wei
- Department of Orthopedics, General Hospital of Western Theater Command, Rongdu Avenue No. 270, Chengdu, 610000, People's Republic of China. .,Chengdu Medical College, Rongdu Avenue No. 601, Chengdu, 610000, People's Republic of China.
| |
Collapse
|
47
|
Liu LY, Jiang D, Qu Y, Wang H, Zhang Y, Yang S, Xie X, Wu S, Zhou H, Xu G. Potential and functional prediction of six circular RNAs as diagnostic markers for colorectal cancer. PeerJ 2022; 10:e13420. [PMID: 35611168 PMCID: PMC9124462 DOI: 10.7717/peerj.13420] [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: 12/14/2021] [Accepted: 04/20/2022] [Indexed: 01/20/2023] Open
Abstract
Background Circular RNAs (circRNAs) have been discovered in colorectal cancer (CRC), but there are few reports on the expression distribution and functional mining analysis of circRNAs. Methods Differentially expressed circRNAs in CRC tissues and adjacent normal tissues were screened and identified by microarray and qRT-PCR. ROC curves of the six circRNAs were analyzed. A series of bioinformatics analyses on differentially expressed circRNAs were performed. Results A total of 207 up-regulated and 357 down-regulated circRNAs in CRC were screened, and three top up-regulated and down-regulated circRNAs were chosen to be verified in 33 pairs of CRCs by qRT-PCR. 6 circRNAs showed high diagnostic values (AUC = 0.6860, AUC = 0.8127, AUC = 0.7502, AUC = 0.9945, AUC = 0.9642, AUC = 0.9486 for hsa_circRNA_100833, hsa_circRNA_103828, hsa_circRNA_103831 and hsa_circRNA_103752, hsa_circRNA_071106, hsa_circRNA_102293). A circRNA-miRNA-mRNA regulatory network (cirReNET) including six candidate circRNAs, 19 miRNAs and 210 mRNA was constructed, and the functions of the cirReNET were predicted and displayed via Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analyses on these mRNAs and protein-protein interaction (PPI) network of the hub genes acquired by string and CytoHubba. Conclusion A cirReNET containing potential diagnostic and predictive indicators of CRCs and several critical circRNA-miRNA-mRNA regulatory axes (cirReAXEs) in CRC were mined, and may provide a novel route to study the mechanism and clinical targets of CRC.
Collapse
Affiliation(s)
- Li yuan Liu
- School of Clinical Medicine, The Third Affiliated Hospital of Ningxia Medical University, Ningxia Medical University, Yinchuan, Ningxia, China,Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, School of Medical Technology, Institute of Clinical Laboratory, Guangdong Medical University, Dongguan, Guangdong, China
| | - Dan Jiang
- School of Clinical Medicine, The Third Affiliated Hospital of Ningxia Medical University, Ningxia Medical University, Yinchuan, Ningxia, China,Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, School of Medical Technology, Institute of Clinical Laboratory, Guangdong Medical University, Dongguan, Guangdong, China
| | - Yuliang Qu
- School of Clinical Medicine, The Third Affiliated Hospital of Ningxia Medical University, Ningxia Medical University, Yinchuan, Ningxia, China
| | - Hongxia Wang
- School of Clinical Medicine, The Third Affiliated Hospital of Ningxia Medical University, Ningxia Medical University, Yinchuan, Ningxia, China
| | - Yanting Zhang
- School of Clinical Medicine, The Third Affiliated Hospital of Ningxia Medical University, Ningxia Medical University, Yinchuan, Ningxia, China
| | - Shaoqi Yang
- School of Clinical Medicine, The Third Affiliated Hospital of Ningxia Medical University, Ningxia Medical University, Yinchuan, Ningxia, China
| | - Xiaoliang Xie
- School of Clinical Medicine, The Third Affiliated Hospital of Ningxia Medical University, Ningxia Medical University, Yinchuan, Ningxia, China
| | - Shan Wu
- School of Clinical Medicine, The Third Affiliated Hospital of Ningxia Medical University, Ningxia Medical University, Yinchuan, Ningxia, China
| | - Haijin Zhou
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, School of Medical Technology, Institute of Clinical Laboratory, Guangdong Medical University, Dongguan, Guangdong, China
| | - Guangxian Xu
- School of Clinical Medicine, The Third Affiliated Hospital of Ningxia Medical University, Ningxia Medical University, Yinchuan, Ningxia, China,Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, School of Medical Technology, Institute of Clinical Laboratory, Guangdong Medical University, Dongguan, Guangdong, China
| |
Collapse
|
48
|
Zhang Z, Li H, Gan H, Tang Z, Guo Y, Yao S, Liuyu T, Zhong B, Lin D. RNF115 Inhibits the Post-ER Trafficking of TLRs and TLRs-Mediated Immune Responses by Catalyzing K11-Linked Ubiquitination of RAB1A and RAB13. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2105391. [PMID: 35343654 PMCID: PMC9165487 DOI: 10.1002/advs.202105391] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Revised: 03/07/2022] [Indexed: 05/16/2023]
Abstract
The subcellular localization and intracellular trafficking of Toll-like receptors (TLRs) critically regulate TLRs-mediated antimicrobial immunity and autoimmunity. Here, it is demonstrated that the E3 ubiquitin ligase RNF115 inhibits the post-endoplasmic reticulum (ER) trafficking of TLRs and TLRs-mediated immune responses by catalyzing ubiquitination of the small GTPases RAB1A and RAB13. It is shown that the 14-3-3 chaperones bind to AKT1-phosphorylated RNF115 and facilitate RNF115 localizing on the ER and the Golgi apparatus. RNF115 interacts with RAB1A and RAB13 and catalyzes K11-linked ubiquitination on the Lys49 and Lys61 residues of RAB1A and on the Lys46 and Lys58 residues of RAB13, respectively. Such a modification impairs the recruitment of guanosine diphosphate (GDP) dissociation inhibitor 1 (GDI1) to RAB1A and RAB13, a prerequisite for the reactivation of RAB proteins. Consistently, knockdown of RAB1A and RAB13 in Rnf115+/+ and Rnf115-/- cells markedly inhibits the post-ER and the post-Golgi trafficking of TLRs, respectively. In addition, reconstitution of RAB1AK49/61R or RAB13K46/58R into Rnf115+/+ cells but not Rnf115-/- cells promotes the trafficking of TLRs from the ER to the Golgi apparatus and from the Golgi apparatus to the cell surface, respectively. These findings uncover a common and step-wise regulatory mechanism for the post-ER trafficking of TLRs.
Collapse
Affiliation(s)
- Zhi‐Dong Zhang
- Department of Gastrointestinal SurgeryMedical Research InstituteZhongnan Hospital of Wuhan UniversityWuhan430071China
- Department of Pulmonary and Critical Care MedicineZhongnan Hospital of Wuhan UniversityWuhan430071China
- Frontier Science Center for Immunology and MetabolismWuhan UniversityWuhan430071China
- Cancer CenterRenmin Hospital of Wuhan UniversityWuhan430061China
| | - Hong‐Xu Li
- Department of Gastrointestinal SurgeryMedical Research InstituteZhongnan Hospital of Wuhan UniversityWuhan430071China
- Department of Pulmonary and Critical Care MedicineZhongnan Hospital of Wuhan UniversityWuhan430071China
- Frontier Science Center for Immunology and MetabolismWuhan UniversityWuhan430071China
| | - Hu Gan
- Department of Gastrointestinal SurgeryMedical Research InstituteZhongnan Hospital of Wuhan UniversityWuhan430071China
- Department of Pulmonary and Critical Care MedicineZhongnan Hospital of Wuhan UniversityWuhan430071China
- Frontier Science Center for Immunology and MetabolismWuhan UniversityWuhan430071China
- Department of VirologyCollege of Life SciencesWuhan UniversityWuhan430072China
| | - Zhen Tang
- Department of Gastrointestinal SurgeryMedical Research InstituteZhongnan Hospital of Wuhan UniversityWuhan430071China
- Department of Pulmonary and Critical Care MedicineZhongnan Hospital of Wuhan UniversityWuhan430071China
- Frontier Science Center for Immunology and MetabolismWuhan UniversityWuhan430071China
- Department of VirologyCollege of Life SciencesWuhan UniversityWuhan430072China
| | - Yu‐Yao Guo
- Department of Gastrointestinal SurgeryMedical Research InstituteZhongnan Hospital of Wuhan UniversityWuhan430071China
- Department of Pulmonary and Critical Care MedicineZhongnan Hospital of Wuhan UniversityWuhan430071China
- Frontier Science Center for Immunology and MetabolismWuhan UniversityWuhan430071China
| | - Shu‐Qi Yao
- Department of Gastrointestinal SurgeryMedical Research InstituteZhongnan Hospital of Wuhan UniversityWuhan430071China
- Department of Pulmonary and Critical Care MedicineZhongnan Hospital of Wuhan UniversityWuhan430071China
- Frontier Science Center for Immunology and MetabolismWuhan UniversityWuhan430071China
- Department of VirologyCollege of Life SciencesWuhan UniversityWuhan430072China
| | - Tianzi Liuyu
- Department of Gastrointestinal SurgeryMedical Research InstituteZhongnan Hospital of Wuhan UniversityWuhan430071China
| | - Bo Zhong
- Department of Gastrointestinal SurgeryMedical Research InstituteZhongnan Hospital of Wuhan UniversityWuhan430071China
- Department of Pulmonary and Critical Care MedicineZhongnan Hospital of Wuhan UniversityWuhan430071China
- Frontier Science Center for Immunology and MetabolismWuhan UniversityWuhan430071China
- Department of VirologyCollege of Life SciencesWuhan UniversityWuhan430072China
- Wuhan Research Center for Infectious Diseases and CancerChinese Academy of Medical SciencesWuhan430071China
| | - Dandan Lin
- Cancer CenterRenmin Hospital of Wuhan UniversityWuhan430061China
| |
Collapse
|
49
|
Mi T, Jin L, Zhang Z, Wang J, Li M, Zhanghuang C, Tan X, Wang Z, Tian X, Xiang B, He D. DNA Hypermethylation-Regulated CX3CL1 Reducing T Cell Infiltration Indicates Poor Prognosis in Wilms Tumour. Front Oncol 2022; 12:882714. [PMID: 35530333 PMCID: PMC9072742 DOI: 10.3389/fonc.2022.882714] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Accepted: 03/21/2022] [Indexed: 12/14/2022] Open
Abstract
Objective To investigate the role of chemokines in Wilms tumours, especially their chemotaxis to immune cells and the role of DNA methylation in regulating the expression level of chemokines. Methods RNAseqV2 gene expression and clinical data were downloaded from the TARGET database. DNA methylation data were downloaded from the GEO and cBioPortal database. The difference analysis and Kaplan-Meier(KM) analysis of chemokines were performed by edgeR package. Then predictive model based on chemokines was constructed by lasso regression and multivariate COX regression. ROC curve, DCA curve, Calibration curve, and Nomogram were used to evaluate the prognostic model. MCPcounter and Cibersort algorithm was used to calculate the infiltration of immune cells in Wilms tumour and para-tumour samples. Then the difference analysis of the immune cells was performed. The relationship between chemokines and immune cells were calculated by Pearson correlation. In addition, DNA methylation differences between Wilms tumour and para-tumour samples was performed. The correlation between DNA methylation and mRNA expression was calculated by Pearson correlation. Western blot(WB)and immunofluorescence were used to confirm the differential expression of CX3CL1 and T cells, and the correlation between them. Results A total of 16 chemokines were differentially expressed in tumour and para-tumour samples. A total of seven chemokines were associated with survival. CCL2 and CX3CL1 were positively correlated with prognosis, while high expression of CCL3, CCL8, CCL15, CCL18 and CXCL9 predicted poor prognosis. By lasso regression and multivariate COX regression, CCL3, CCL15, CXCL9 and CX3CL1 were finally included to construct a prediction model. The model shows good prediction ability. MCPcounter and Cibersort algorithm both showed that T cells were higher in para-tumour tissues than cancer tissues. Correlation analysis showed that CX3CL1 had a strong correlation with T cells. These were verified by Weston blot and immunofluorescence. DNA methylation analysis showed that various chemokines were different in para-tumours and tumours. CX3CL1 was hypermethylated in tumours, and the degree of methylation was negatively correlated with mRNA expression. Conclusion 1. There is low T cell infiltration in nephroblastoma. 2. Chemokines such as CX3CL1 indicate a favourable prognosis and positively correlate with the number of T cells. 3. chemokines such as CX3CL1 are negatively regulated by DNA hypermethylation.
Collapse
Affiliation(s)
- Tao Mi
- Department of Urology, Children’s Hospital of Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Children Urogenital Development and Tissue Engineering, Chongqing, China
- China International Science and Technology Cooperation Base of Child Development and Critical; National Clinical Research Center for Child Health and Disorders, Chongqing; Ministry of Education Key Laboratory of Child Development and Disorders; Chongqing Key Laboratory of Pediatrics, Chongqing, China
| | - Liming Jin
- Department of Urology, Children’s Hospital of Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Children Urogenital Development and Tissue Engineering, Chongqing, China
- China International Science and Technology Cooperation Base of Child Development and Critical; National Clinical Research Center for Child Health and Disorders, Chongqing; Ministry of Education Key Laboratory of Child Development and Disorders; Chongqing Key Laboratory of Pediatrics, Chongqing, China
| | - Zhaoxia Zhang
- Department of Urology, Children’s Hospital of Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Children Urogenital Development and Tissue Engineering, Chongqing, China
- China International Science and Technology Cooperation Base of Child Development and Critical; National Clinical Research Center for Child Health and Disorders, Chongqing; Ministry of Education Key Laboratory of Child Development and Disorders; Chongqing Key Laboratory of Pediatrics, Chongqing, China
| | - Jinkui Wang
- Department of Urology, Children’s Hospital of Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Children Urogenital Development and Tissue Engineering, Chongqing, China
- China International Science and Technology Cooperation Base of Child Development and Critical; National Clinical Research Center for Child Health and Disorders, Chongqing; Ministry of Education Key Laboratory of Child Development and Disorders; Chongqing Key Laboratory of Pediatrics, Chongqing, China
| | - Mujie Li
- Department of Urology, Children’s Hospital of Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Children Urogenital Development and Tissue Engineering, Chongqing, China
- China International Science and Technology Cooperation Base of Child Development and Critical; National Clinical Research Center for Child Health and Disorders, Chongqing; Ministry of Education Key Laboratory of Child Development and Disorders; Chongqing Key Laboratory of Pediatrics, Chongqing, China
| | - Chenghao Zhanghuang
- Department of Urology, Children’s Hospital of Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Children Urogenital Development and Tissue Engineering, Chongqing, China
- China International Science and Technology Cooperation Base of Child Development and Critical; National Clinical Research Center for Child Health and Disorders, Chongqing; Ministry of Education Key Laboratory of Child Development and Disorders; Chongqing Key Laboratory of Pediatrics, Chongqing, China
| | - Xiaojun Tan
- Department of Urology, Children’s Hospital of Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Children Urogenital Development and Tissue Engineering, Chongqing, China
- China International Science and Technology Cooperation Base of Child Development and Critical; National Clinical Research Center for Child Health and Disorders, Chongqing; Ministry of Education Key Laboratory of Child Development and Disorders; Chongqing Key Laboratory of Pediatrics, Chongqing, China
| | - Zhang Wang
- Department of Urology, Children’s Hospital of Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Children Urogenital Development and Tissue Engineering, Chongqing, China
- China International Science and Technology Cooperation Base of Child Development and Critical; National Clinical Research Center for Child Health and Disorders, Chongqing; Ministry of Education Key Laboratory of Child Development and Disorders; Chongqing Key Laboratory of Pediatrics, Chongqing, China
| | - Xiaomao Tian
- Department of Urology, Children’s Hospital of Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Children Urogenital Development and Tissue Engineering, Chongqing, China
- China International Science and Technology Cooperation Base of Child Development and Critical; National Clinical Research Center for Child Health and Disorders, Chongqing; Ministry of Education Key Laboratory of Child Development and Disorders; Chongqing Key Laboratory of Pediatrics, Chongqing, China
| | - Bin Xiang
- Department of Urology, Children’s Hospital of Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Children Urogenital Development and Tissue Engineering, Chongqing, China
- China International Science and Technology Cooperation Base of Child Development and Critical; National Clinical Research Center for Child Health and Disorders, Chongqing; Ministry of Education Key Laboratory of Child Development and Disorders; Chongqing Key Laboratory of Pediatrics, Chongqing, China
| | - Dawei He
- Department of Urology, Children’s Hospital of Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Children Urogenital Development and Tissue Engineering, Chongqing, China
- China International Science and Technology Cooperation Base of Child Development and Critical; National Clinical Research Center for Child Health and Disorders, Chongqing; Ministry of Education Key Laboratory of Child Development and Disorders; Chongqing Key Laboratory of Pediatrics, Chongqing, China
- *Correspondence: Dawei He,
| |
Collapse
|
50
|
Wang L, Jiang J, Chen Y, Jia Q, Chu Q. The roles of CC chemokines in response to radiation. Radiat Oncol 2022; 17:63. [PMID: 35365161 PMCID: PMC8974090 DOI: 10.1186/s13014-022-02038-x] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Accepted: 03/20/2022] [Indexed: 01/21/2023] Open
Abstract
Radiotherapy is an effective regimen for cancer treatment alone or combined with chemotherapy or immunotherapy. The direct effect of radiotherapy involves radiation-induced DNA damage, and most studies have focused on this area to improve the efficacy of radiotherapy. Recently, the immunomodulatory effect of radiation on the tumour microenvironment has attracted much interest. Dying tumour cells can release multiple immune-related molecules, including tumour-associated antigens, chemokines, and inflammatory mediators. Then, immune cells are attracted to the irradiated site, exerting immunostimulatory or immunosuppressive effects. CC chemokines play pivotal roles in the trafficking process. The CC chemokine family includes 28 members that attract different immune subsets. Upon irradiation, tumour cells or immune cells can release different CC chemokines. Here, we mainly discuss the importance of CCL2, CCL3, CCL5, CCL8, CCL11, CCL20 and CCL22 in radiotherapy. In irradiated normal tissues, released chemokines induce epithelial to mesenchymal transition, thus promoting tissue injury. In the tumour microenvironment, released chemokines recruit cancer-associated cells, such as tumour-infiltrating lymphocytes, myeloid-derived suppressor cells and tumour-associated macrophages, to the tumour niche. Thus, CC chemokines have protumour and antitumour properties. Based on the complex roles of CC chemokines in the response to radiation, it would be promising to target specific chemokines to alleviate radiation-induced injury or promote tumour control.
Collapse
|