1
|
Mareboina M, Bakhl K, Agioti S, Yee NS, Georgakopoulos-Soares I, Zaravinos A. Comprehensive Analysis of Granzymes and Perforin Family Genes in Multiple Cancers. Biomedicines 2025; 13:408. [PMID: 40002821 PMCID: PMC11853441 DOI: 10.3390/biomedicines13020408] [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: 11/05/2024] [Revised: 01/25/2025] [Accepted: 02/05/2025] [Indexed: 02/27/2025] Open
Abstract
Background/Objectives: Cancer remains a significant global health concern, with immunotherapies emerging as promising treatments. This study explored the role of perforin-1 (PRF1) and granzymes A, B and K (GZMA, GZMB and GZMK) in cancer biology, focusing on their impact on tumor cell death and immune response modulation. Methods: Through a comprehensive genomic analysis across various cancer types, we explored the differential expression, mutation profiles and methylation patterns of these genes, providing insights into their potential as therapeutic targets. Furthermore, we investigated their association with immune cell infiltration and pathway activation within the tumor microenvironment in each tumor type. Results: Our findings revealed distinct expression patterns and prognostic implications for PRF1, GZMA, GZMB and GZMK across different cancers, highlighting their multifaceted roles in tumor immunity. We found increased immune infiltration across all tumor types and significant correlations between the genes of interest and cytotoxic T cells, as well as the most significant survival outcomes in breast cancer. We also show that granzymes and perforin-1 are significantly associated with indicators of immunosuppression and T cell dysfunction within patient cohorts. In skin melanoma, glioblastoma, kidney and bladder cancers, we found significant correlations between the genes of interest and patient survival after receiving immune-checkpoint inhibition therapy. Additionally, we identified potential associations between the mRNA expression levels of these genes and drug sensitivity. Conclusions: Overall, this study enhances our understanding of the molecular mechanisms underlying tumor immunity and provides valuable insights into the potential therapeutic implications of PRF1, GZMA, GZMB and GZMK in cancer treatment.
Collapse
Affiliation(s)
- Manvita Mareboina
- Institute for Personalized Medicine, Department of Biochemistry and Molecular Biology, The Pennsylvania State University College of Medicine, Hershey, PA 17033, USA; (M.M.); (K.B.)
| | - Katrina Bakhl
- Institute for Personalized Medicine, Department of Biochemistry and Molecular Biology, The Pennsylvania State University College of Medicine, Hershey, PA 17033, USA; (M.M.); (K.B.)
| | - Stephanie Agioti
- Department of Life Sciences, School of Sciences, European University Cyprus, Nicosia 1516, Cyprus;
- Cancer Genetics, Genomics and Systems Biology Laboratory, Basic and Translational Cancer Research Center (BTCRC), Nicosia 1516, Cyprus
| | - Nelson S. Yee
- Department of Medicine, Division of Hematology-Oncology, Penn State Health Milton S. Hershey Medical Center, Next-Generation Therapies Program, Penn State Cancer Institute, Hershey, PA 17033, USA;
| | - Ilias Georgakopoulos-Soares
- Institute for Personalized Medicine, Department of Biochemistry and Molecular Biology, The Pennsylvania State University College of Medicine, Hershey, PA 17033, USA; (M.M.); (K.B.)
| | - Apostolos Zaravinos
- Department of Life Sciences, School of Sciences, European University Cyprus, Nicosia 1516, Cyprus;
- Cancer Genetics, Genomics and Systems Biology Laboratory, Basic and Translational Cancer Research Center (BTCRC), Nicosia 1516, Cyprus
| |
Collapse
|
2
|
Xie Y, Peng H, Hu Y, Jia K, Yuan J, Liu D, Li Y, Feng X, Li J, Zhang X, Sun Y, Shen L, Chen Y. Immune microenvironment spatial landscapes of tertiary lymphoid structures in gastric cancer. BMC Med 2025; 23:59. [PMID: 39901202 PMCID: PMC11792408 DOI: 10.1186/s12916-025-03889-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2024] [Accepted: 01/22/2025] [Indexed: 02/05/2025] Open
Abstract
BACKGROUND Tertiary lymphoid structures (TLS) correlate with tumour prognosis and immunotherapy responses in gastric cancer (GC) studies. However, understanding the complex and diverse immune microenvironment within TLS requires comprehensive analysis. METHODS We examined the prognostic impact of TLS within the tumour core (TC) of 59 GC patients undergoing immunotherapy. Multispectral fluorescence imaging was employed to evaluate variations in immune cell infiltration across different TLS sites among 110 GC patients, by quantifying immune cell density and spatial characteristics. We also generated a single-cell transcriptomic atlas of TLS-positive (n = 4) and TLS-negative (n = 8) microenvironments and performed spatial transcriptomics (ST) analysis on two samples. RESULTS TLS presence in the TC significantly correlated with improved immune-related overall survival (P = 0.049). CD8+LAG-3-PD-1+TIM-3-, CD4+PD-L1+, and CD4+FoxP3- T cell densities were significantly higher in the TLS within TC compared to tumour and stromal regions. Immune cells within TLS exhibited closer intercellular proximity than those outside TLS. Five key density and spatial characteristics of immune cells within TLS in the TC were selected to develop the Density and Spatial Score risk model. Single-cell RNA sequencing revealed strong intercellular interactions in the presence of TLS within the microenvironment. However, TLS-absent environment facilitated tumour cell interactions with immune cells through MIF- and galectin-dependent pathways, recruiting immunosuppressive cells. ST analysis confirmed that T and B cells co-localise within TLS, enhancing immune response activation compared to cancer nests and exerting a strong anti-tumour effect. CONCLUSIONS TLS presence facilitates frequent cell-to-cell communication, forming an active immune microenvironment, highlighting the prognostic value of TLS.
Collapse
Affiliation(s)
- Yi Xie
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education, Beijing), Peking University Cancer Hospital and Institute, Beijing, 100142, China
| | - Haoxin Peng
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education, Beijing), Peking University Cancer Hospital and Institute, Beijing, 100142, China
| | - Yajie Hu
- Department of Pathology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital and Institute, Beijing, 100142, China
| | - Keren Jia
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education, Beijing), Peking University Cancer Hospital and Institute, Beijing, 100142, China
| | - Jiajia Yuan
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education, Beijing), Peking University Cancer Hospital and Institute, Beijing, 100142, China
| | - Dan Liu
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education, Beijing), Peking University Cancer Hospital and Institute, Beijing, 100142, China
| | - Yanyan Li
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education, Beijing), Peking University Cancer Hospital and Institute, Beijing, 100142, China
| | - Xujiao Feng
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education, Beijing), Peking University Cancer Hospital and Institute, Beijing, 100142, China
| | - Jian Li
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education, Beijing), Peking University Cancer Hospital and Institute, Beijing, 100142, China
| | - Xiaotian Zhang
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education, Beijing), Peking University Cancer Hospital and Institute, Beijing, 100142, China
| | - Yu Sun
- Department of Pathology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital and Institute, Beijing, 100142, China.
| | - Lin Shen
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education, Beijing), Peking University Cancer Hospital and Institute, Beijing, 100142, China.
| | - Yang Chen
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education, Beijing), Peking University Cancer Hospital and Institute, Beijing, 100142, China.
- Department of Gastrointestinal Cancer, Beijing GoBroad Hospital, Beijing, 102200, China.
| |
Collapse
|
3
|
Wu K, Xu X, Wei W, Wen J, Hu H. c-JUN interacts with HDAC1 as a potential combinatorial therapeutic target in acute myeloid leukemia. Int Immunopharmacol 2025; 146:113927. [PMID: 39721452 DOI: 10.1016/j.intimp.2024.113927] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2024] [Revised: 12/10/2024] [Accepted: 12/19/2024] [Indexed: 12/28/2024]
Abstract
Acute myeloid leukemia (AML) is a biologically heterogeneous disease originating from the clonal expansion of hematopoietic stem cells (HSCs). Clonal expansion of hematopoietic stem cell progenitors (HSC-Prog), along with a block in differentiation, are hallmark features of AML. The disease is characterized by poor clinical outcomes, highlighting the urgent need for effective therapeutic strategies and suitable drug targets. We conducted multi-omics analyses, including single-cell RNA sequencing (scRNA-seq), Mendelian randomization (MR), and bulk RNA-seq, to investigate HDAC1's oncogenic role in AML. We identified specific gene signatures at the single-cell level. MR with eQTL data established causal links, and TCGA-LAML RNA-seq provided prognostic insights. Analysis of cellular communication and transcription factors revealed high c-JUN activity in HSC-Prog. We confirmed the association of c-JUN with HDAC1 through Western blotting and Co-immunoprecipitation (Co-IP). Functional validation of c-JUN in AML cells was performed via flow cytometry in vitro. The effectiveness of drugs targeting c-JUN and HDAC1 was assessed in mouse models using live imaging methods like in vivo imaging system (IVIS) and iSMAART. We identified the activity of c-JUN is specifically enhanced in HSC-Prog in AML patients. We suggest a potential regulatory relationship between c-JUN and HDAC1 in AML tumor cells. Inhibition of c-JUN can suppress cell proliferation and CD33 expression in AML, enhancing susceptibility to natural killer (NK) cell-mediated cytotoxicity. The combination of agents targeting c-JUN (Ailanthone) and HDAC1 (Panobinostat) showed robust efficacy in treating AML in xenograft mouse models, outperforming monotherapy. We also observed that the combination of Ailanthone and Panobinostat therapy displayed a safe pharmacological profile without dose-dependent toxicity, suggesting its potential as a therapeutic strategy.
Collapse
Affiliation(s)
- Ke Wu
- Department of Radiology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China
| | - Xiaoyu Xu
- Chinese Academy of Sciences Key Laboratory of Innate Immunity and Chronic Disease, Key Laboratory of Immune Response and Immunotherapy, Institute of Immunology, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230027, China
| | - Wei Wei
- Department of Radiology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China.
| | - Jie Wen
- Department of Radiology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China.
| | - Haixi Hu
- Department of Scientific Research, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230001, China.
| |
Collapse
|
4
|
Maurer K, Park CY, Mani S, Borji M, Raths F, Gouin KH, Penter L, Jin Y, Zhang JY, Shin C, Brenner JR, Southard J, Krishna S, Lu W, Lyu H, Abbondanza D, Mangum C, Olsen LR, Lawson MJ, Fabani M, Neuberg DS, Bachireddy P, Glezer EN, Farhi SL, Li S, Livak KJ, Ritz J, Soiffer RJ, Wu CJ, Azizi E. Coordinated immune networks in leukemia bone marrow microenvironments distinguish response to cellular therapy. Sci Immunol 2025; 10:eadr0782. [PMID: 39854478 DOI: 10.1126/sciimmunol.adr0782] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Accepted: 12/18/2024] [Indexed: 01/26/2025]
Abstract
Understanding how intratumoral immune populations coordinate antitumor responses after therapy can guide treatment prioritization. We systematically analyzed an established immunotherapy, donor lymphocyte infusion (DLI), by assessing 348,905 single-cell transcriptomes from 74 longitudinal bone marrow samples of 25 patients with relapsed leukemia; a subset was evaluated by both protein- and transcriptome-based spatial analysis. In acute myeloid leukemia (AML) DLI responders, we identified clonally expanded ZNF683+ CD8+ cytotoxic T lymphocytes with in vitro specificity for patient-matched AML. These cells originated primarily from the DLI product and appeared to coordinate antitumor immune responses through interaction with diverse immune cell types within the marrow microenvironment. Nonresponders lacked this cross-talk and had cytotoxic T lymphocytes with elevated TIGIT expression. Our study identifies recipient bone marrow microenvironment differences as a determinant of an effective antileukemia response and opens opportunities to modulate cellular therapy.
Collapse
Affiliation(s)
- Katie Maurer
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
- Harvard Medical School, Boston, MA 02115, USA
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Cameron Y Park
- Irving Institute for Cancer Dynamics, Columbia University, New York, NY 10027, USA
- Department of Biomedical Engineering, Columbia University, New York, NY 10027, USA
| | - Shouvik Mani
- Irving Institute for Cancer Dynamics, Columbia University, New York, NY 10027, USA
- Department of Computer Science, Columbia University, New York, NY 10027, USA
| | - Mehdi Borji
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | | | | | - Livius Penter
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
- Harvard Medical School, Boston, MA 02115, USA
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
- Department of Hematology, Oncology, and Tumorimmunology, Campus Virchow Klinikum, Berlin, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 13353 Berlin, Germany
| | - Yinuo Jin
- Irving Institute for Cancer Dynamics, Columbia University, New York, NY 10027, USA
- Department of Biomedical Engineering, Columbia University, New York, NY 10027, USA
| | - Jia Yi Zhang
- Irving Institute for Cancer Dynamics, Columbia University, New York, NY 10027, USA
- Department of Biomedical Engineering, Columbia University, New York, NY 10027, USA
| | - Crystal Shin
- Irving Institute for Cancer Dynamics, Columbia University, New York, NY 10027, USA
- Department of Biomedical Engineering, Columbia University, New York, NY 10027, USA
| | - James R Brenner
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
- Harvard Medical School, Boston, MA 02115, USA
| | - Jackson Southard
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
- Translational Immunogenomics Laboratory, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Sachi Krishna
- Spatial Technology Platform, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Wesley Lu
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
- Translational Immunogenomics Laboratory, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Haoxiang Lyu
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
- Translational Immunogenomics Laboratory, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Domenic Abbondanza
- Spatial Technology Platform, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
- Department of Medicine, Section of Rheumatology, University of Chicago, Chicago, IL 60637, USA
| | - Chanell Mangum
- Spatial Technology Platform, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Lars Rønn Olsen
- Department of Health Technology, Technical University of Denmark, 2800 Kongens Lyngby, Denmark
| | | | | | - Donna S Neuberg
- Department of Data Science, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Pavan Bachireddy
- Department of Hematopoietic Biology & Malignancy, MD Anderson Cancer Center, Houston, TX 77030, USA
| | | | - Samouil L Farhi
- Spatial Technology Platform, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Shuqiang Li
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
- Translational Immunogenomics Laboratory, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Kenneth J Livak
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
- Translational Immunogenomics Laboratory, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Jerome Ritz
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
- Harvard Medical School, Boston, MA 02115, USA
| | - Robert J Soiffer
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
- Harvard Medical School, Boston, MA 02115, USA
| | - Catherine J Wu
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
- Harvard Medical School, Boston, MA 02115, USA
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Elham Azizi
- Irving Institute for Cancer Dynamics, Columbia University, New York, NY 10027, USA
- Department of Biomedical Engineering, Columbia University, New York, NY 10027, USA
- Department of Computer Science, Columbia University, New York, NY 10027, USA
| |
Collapse
|
5
|
Chen J, Xie J, Deng F, Cai J, Chen S, Song X, Xia S, Shen Q, Guo X, Tang Y. Expansion of peripheral cytotoxic CD4+ T cells in Alzheimer's disease: New insights from multi-omics evidence. Genomics 2025; 117:110976. [PMID: 39657893 DOI: 10.1016/j.ygeno.2024.110976] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Revised: 11/19/2024] [Accepted: 12/04/2024] [Indexed: 12/12/2024]
Abstract
The significance of the adaptive immune response in Alzheimer's disease (AD) is increasingly recognized. We analyzed scRNA-Seq data from AD patients, revealing a notable rise in CD4 cytotoxic T cells (CD4-CTLs) in peripheral blood mononuclear cells (PBMCs), validated in vivo and in vitro. This rise correlates with cognitive decline in AD patients. We also identified transcription factors TBX21 and MYBL1 as key drivers of CD4-CTL expansion. Further analyses indicate these cells are terminally differentiated, showing clonal expansion, metabolic changes, and unique communication patterns. Mendelian randomization identified risk genes SRGN and ITGB1, suggesting their genetic regulation in CD4-CTLs may contribute to AD. To summarize, our findings characterize the expansion of CD4-CTLs in the PBMCs of AD patients, providing valuable understanding into the possible mechanisms involved in the expansion of CD4-CTLs in AD.
Collapse
Affiliation(s)
- Jiongxue Chen
- Department of Neurology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China
| | - Jiatian Xie
- Department of Neurology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China
| | - Fuyin Deng
- Department of Neurology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China
| | - Jinhua Cai
- Department of Neurology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China
| | - Sitai Chen
- Department of Neurology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China
| | - Xingrong Song
- Department of Anesthesiology, Guangzhou Women and Children Medical Center, Guangzhou 510623, China
| | - Shangzhou Xia
- Center for Neurodegeneration and Regeneration, Zilkha Neurogenetic Institute and Department of Physiology and Neuroscience, Keck School of Medicine, University of Southern California, Los Angeles, California 90033, USA
| | - Qingyu Shen
- Department of Neurology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China
| | - Xinying Guo
- Department of Anesthesiology, Guangzhou Women and Children Medical Center, Guangzhou 510623, China.
| | - Yamei Tang
- Department of Neurology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China; Brain Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China; Guangdong Provincial Key Laboratory of Epigenetics and Gene Regulation of Malignant Tumors, Sun Yat-sen Memorial Hospital, Guangzhou, China; Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China.
| |
Collapse
|
6
|
Dong YM, Bao GQ. Coagulation factor II thrombin receptor as a promising biomarker in breast cancer management. Open Life Sci 2024; 19:20221001. [PMID: 39655193 PMCID: PMC11627072 DOI: 10.1515/biol-2022-1001] [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: 05/27/2024] [Revised: 10/10/2024] [Accepted: 10/16/2024] [Indexed: 12/12/2024] Open
Abstract
This study aims to comprehensively investigate the role of coagulation factor II thrombin receptor (F2R) in breast cancer (BC) and to evaluate its potential as a biomarker in this context. Data on female BC were retrieved from the TCGA database. Comparative analyses were performed, including enrichment analysis, tumor immune microenvironment analysis, drug sensitivity testing, molecular docking, and cell-based experiments, to assess the expression and function of F2R in BC. Statistical analyses and graphical representations were conducted using R software. The study confirmed a significant upregulation of F2R in BC, which was associated with a more favorable prognosis. Clinical correlation analysis revealed a strong association between F2R expression and key clinical parameters, such as estrogen receptor and progesterone receptor status. Additionally, genes co-expressed with F2R were significantly linked to various biological processes, including cell cycle regulation, oxidative phosphorylation, ribosomal function, and extracellular matrix interactions. F2R also showed associations with immune modulators, particularly CD200 and NRP1. Drug sensitivity analysis, molecular docking, and cell experiments consistently demonstrated positive correlations between F2R expression and sensitivity to dasatinib. This study underscores the potential of F2R as a valuable biomarker in BC, providing insights into the molecular mechanisms underlying tumorigenesis.
Collapse
Affiliation(s)
- Yan-Ming Dong
- Department of General Surgery, The Second Affiliated Hospital of Air Force Medical University, No. 356 of Xinsi Road, Baqiao District, Xi’an, 710038, China
| | - Guo-Qiang Bao
- Department of General Surgery, The Second Affiliated Hospital of Air Force Medical University, No. 356 of Xinsi Road, Baqiao District, Xi’an, 710038, China
| |
Collapse
|
7
|
Kim M, Renert-Yuval Y, Stepensky P, Even-Or E, Zaidman I, Fachler T, Neumark M, Zamir M, NandyMazumdar M, Gour D, Facheris P, Carroll B, Liu Y, Yu Ekey ML, Andrews E, Meariman M, Angelov M, Bose S, Estrada YD, Molho-Pessach V, Guttman-Yassky E. Sclerotic-Type Cutaneous Chronic Graft-Versus-Host Disease Exhibits Activation of T Helper 1 and OX40 Cytokines. J Invest Dermatol 2024; 144:563-572.e9. [PMID: 37742913 PMCID: PMC11447555 DOI: 10.1016/j.jid.2023.08.026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Revised: 08/09/2023] [Accepted: 08/26/2023] [Indexed: 09/26/2023]
Abstract
Sclerotic-type cutaneous chronic graft-versus-host disease is a severe complication of allogeneic hematopoietic stem cell transplantation, with profound morbidity. A dearth of effective, targeted treatment options necessitates further investigation into the molecular mechanisms underlying this T-cell-mediated disease. In this study, we compared the transcriptome in skin biopsies from pediatric and young adult (aged <25 years) patients with sclerotic-type cutaneous chronic graft-versus-host disease (n = 7) with that in demographically matched healthy controls (n = 8) and patients with atopic dermatitis (n = 10) using RNA sequencing with RT-PCR and immunohistochemistry validation. Differential expression was defined as fold change > 1.5 and false discovery rate < 0.05. Sclerotic-type cutaneous chronic graft-versus-host disease exhibited strong and significant T helper (Th)1 skewing through key related cytokines and chemokines (CXCL9/10/11, IFNG/IFN-γ, STAT1/signal transducer and activator of transcription 1). Several markers related to the TSLP-OX40 axis were significantly upregulated relative to those in both controls and lesional atopic dermatitis, including TNFSF4/OX40L, TSLP, and IL33, as well as fibroinflammatory signatures characterized in a prior study in systemic sclerosis. Gene set variation analysis reflected marker-level findings, showing the greatest enrichment of the Th1 and fibroinflammatory pathways, with no global activation identified in Th2 or Th17/Th22. Cell-type deconvolution revealed a significant representation of macrophages and vascular endothelial cells. Sclerotic-type cutaneous chronic graft-versus-host disease in young patients may therefore be characterized by strong Th1-related upregulation with a unique TSLP-OX40 signature, suggesting new therapeutic avenues for this devastating disease.
Collapse
Affiliation(s)
- Madeline Kim
- Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Yael Renert-Yuval
- Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York, New York, USA; Pediatric Dermatology Unit, Schneider Children's Medical Center of Israel, Petah Tikva, Israel; Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Polina Stepensky
- Department of Bone Marrow Transplantation and Cancer Immunotherapy, Hadassah Medical Center, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Ehud Even-Or
- Department of Bone Marrow Transplantation and Cancer Immunotherapy, Hadassah Medical Center, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Irina Zaidman
- Department of Bone Marrow Transplantation and Cancer Immunotherapy, Hadassah Medical Center, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Tahel Fachler
- Department of Dermatology, Hadassah Medical Center, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Michal Neumark
- Department of Dermatology, Hadassah Medical Center, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Mariana Zamir
- Department of Dermatology, Sheba Medical Center, Tel Hashomer, Israel
| | - Monali NandyMazumdar
- Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Digpal Gour
- Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Paola Facheris
- Department of Dermatology, IRCCS Humanitas Research Hospital, Milano, Italy
| | - Britta Carroll
- Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Ying Liu
- Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Mitchelle L Yu Ekey
- Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Elizabeth Andrews
- Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Marguerite Meariman
- Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Michael Angelov
- Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Swaroop Bose
- Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Yeriel D Estrada
- Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Vered Molho-Pessach
- Department of Dermatology, Hadassah Medical Center, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel.
| | - Emma Guttman-Yassky
- Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York, New York, USA.
| |
Collapse
|
8
|
Maurer K, Park CY, Mani S, Borji M, Penter L, Jin Y, Zhang JY, Shin C, Brenner JR, Southard J, Krishna S, Lu W, Lyu H, Abbondanza D, Mangum C, Olsen LR, Neuberg DS, Bachireddy P, Farhi SL, Li S, Livak KJ, Ritz J, Soiffer RJ, Wu CJ, Azizi E. Coordinated Immune Cell Networks in the Bone Marrow Microenvironment Define the Graft versus Leukemia Response with Adoptive Cellular Therapy. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.09.579677. [PMID: 38405900 PMCID: PMC10888840 DOI: 10.1101/2024.02.09.579677] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/27/2024]
Abstract
Understanding how intra-tumoral immune populations coordinate to generate anti-tumor responses following therapy can guide precise treatment prioritization. We performed systematic dissection of an established adoptive cellular therapy, donor lymphocyte infusion (DLI), by analyzing 348,905 single-cell transcriptomes from 74 longitudinal bone-marrow samples of 25 patients with relapsed myeloid leukemia; a subset was evaluated by protein-based spatial analysis. In acute myelogenous leukemia (AML) responders, diverse immune cell types within the bone-marrow microenvironment (BME) were predicted to interact with a clonally expanded population of ZNF683 + GZMB + CD8+ cytotoxic T lymphocytes (CTLs) which demonstrated in vitro specificity for autologous leukemia. This population, originating predominantly from the DLI product, expanded concurrently with NK and B cells. AML nonresponder BME revealed a paucity of crosstalk and elevated TIGIT expression in CD8+ CTLs. Our study highlights recipient BME differences as a key determinant of effective anti-leukemia response and opens new opportunities to modulate cell-based leukemia-directed therapy.
Collapse
|
9
|
Dong L, Wei X, Yu L, Li Y, Chen L. Inhibition of SIRT7 promotes STAT1 activation and STAT1-dependent signaling in hepatocellular carcinoma. Cell Signal 2024; 114:111005. [PMID: 38070755 DOI: 10.1016/j.cellsig.2023.111005] [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: 09/08/2023] [Revised: 11/27/2023] [Accepted: 12/04/2023] [Indexed: 12/21/2023]
Abstract
The signal transducer and activator of transcription 1 (STAT1) plays a crucial role in regulating tumor progression. However, the mechanisms governing its phosphorylation and biological functions remain incompletely understood. Here, we present compelling evidence indicating that knockdown of SIRT7 inhibits Smurf1-induced ubiquitination of STAT1, consequently impeding the proteasome pathway degradation of STAT1. This inhibition leads to increased stability of STAT1 and enhanced binding to JAK1. Importantly, SIRT7 exerts a negative regulatory effect on STAT1 activation and IFN-γ/STAT1 signaling in hepatocellular carcinoma (HCC). Etoposide treatment not only facilitates STAT1 activation but also downregulates SIRT7 expression. Notably, knockdown of STAT1 in SIRT7-deficient cells attenuates the increase in cell apoptosis induced by Etoposide treatment. In conclusion, our data shed light on the intricate interplay between ubiquitination, STAT1, SIRT7, and Smurf1, elucidating their impact on STAT1-related signaling. These insights contribute to a more comprehensive understanding of the molecular mechanisms involved in STAT1 regulation and suggest potential avenues for the development of targeted therapies against cancer.
Collapse
Affiliation(s)
- Ling Dong
- Laboratory Research Center, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, People's Republic of China.
| | - Xufu Wei
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, People's Republic of China
| | - Le Yu
- School of Life Sciences, Chongqing University, Chongqing 401331, People's Republic of China
| | - Yixin Li
- Department of Pathology, College of Basic Medicine, Chongqing Medical University, Chongqing 400016, People's Republic of China
| | - Lixue Chen
- Laboratory Research Center, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, People's Republic of China.
| |
Collapse
|
10
|
Wang X, Yang T, Shi S, Xu C, Wang F, Dai D, Guan G, Zhang Y, Wang S, Wang J, Zhang B, Liu P, Bai X, Jin Y, Li X, Zhu C, Chen D, Xu Q, Guo Y. Heterogeneity-induced NGF-NGFR communication inefficiency promotes mitotic spindle disorganization in exhausted T cells through PREX1 suppression to impair the anti-tumor immunotherapy with PD-1 mAb in hepatocellular carcinoma. Cancer Med 2024; 13:e6736. [PMID: 38204220 PMCID: PMC10905245 DOI: 10.1002/cam4.6736] [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: 04/22/2023] [Revised: 09/20/2023] [Accepted: 10/20/2023] [Indexed: 01/12/2024] Open
Abstract
BACKGROUND The mechanism of decreased T cells infiltrating tumor tissues in hepatocellular carcinoma is poorly understood. METHODS Cells were separated from the single-cell RNA-sequence dataset of hepatocellular carcinoma patients (GSE149614) for cell-cell communication. Flow cytometry, EDU staining, H3-Ser28 staining, confocal immunofluorescence staining, western blotting and naked microsubcutaneous tumors were performed for the mechanism of NGF-NGFR promoting proliferation. RESULTS The present study has revealed that during the process of T-cell infiltration from adjacent tissues to tumor tissues, an inefficiency in NGF-NGFR communication occurs in the tumor tissues. Importantly, NGF secreted by tumor cells interacts with NGFR present on the membranes of the infiltrated T cells, thereby promoting the proliferation through the activation of mitotic spindle signals. Mechanistically, the mediation of mitotic spindle signal activation promoting proliferation is executed by HDAC1-mediated inhibition of unclear trans-localization of PREX1. Furthermore, PD-1 mAb acts synergistically with the NGF-NGFR communication to suppress tumor progression in both mouse models and HCC patients. Additionally, NGF-NGFR communication was positively correlates with the PD-1/PDL-1 expression. However, expressions of NGF and NGFR are low in tumor tissues, which is responsible for the invasive clinicopathological features and the disappointing prognosis in HCC patients. CONCLUSION Inefficiency in NGF-NGFR communication impairs PD-1 mAb immunotherapy and could thus be utilized as a novel therapeutic target in the treatment of HCC patients in clinical practice.
Collapse
Affiliation(s)
- Xin Wang
- Liver Disease CenterThe Affiliated Hospital of Qingdao UniversityQingdaoChina
| | - Tongwang Yang
- Liver Disease CenterThe Affiliated Hospital of Qingdao UniversityQingdaoChina
- Academician WorkstationChangsha Medical UniversityChangshaChina
- Hunan Key Laboratory of the Research and Development of Novel Pharmaceutical PreparationsChangsha Medical UniversityChangshaChina
| | - Shangheng Shi
- Liver Disease CenterThe Affiliated Hospital of Qingdao UniversityQingdaoChina
| | - Chuanshen Xu
- Liver Disease CenterThe Affiliated Hospital of Qingdao UniversityQingdaoChina
| | - Feng Wang
- Liver Disease CenterThe Affiliated Hospital of Qingdao UniversityQingdaoChina
| | - Deshu Dai
- Liver Disease CenterThe Affiliated Hospital of Qingdao UniversityQingdaoChina
| | - Ge Guan
- Liver Disease CenterThe Affiliated Hospital of Qingdao UniversityQingdaoChina
| | - Yong Zhang
- Liver Disease CenterThe Affiliated Hospital of Qingdao UniversityQingdaoChina
| | - Shuxian Wang
- Liver Disease CenterThe Affiliated Hospital of Qingdao UniversityQingdaoChina
| | - Jianhong Wang
- Liver Disease CenterThe Affiliated Hospital of Qingdao UniversityQingdaoChina
| | - Bingliang Zhang
- Liver Disease CenterThe Affiliated Hospital of Qingdao UniversityQingdaoChina
| | - Peng Liu
- Liver Disease CenterThe Affiliated Hospital of Qingdao UniversityQingdaoChina
| | - Xiaoshuai Bai
- Liver Disease CenterThe Affiliated Hospital of Qingdao UniversityQingdaoChina
| | - Yan Jin
- Liver Disease CenterThe Affiliated Hospital of Qingdao UniversityQingdaoChina
| | - Xinqiang Li
- Liver Disease CenterThe Affiliated Hospital of Qingdao UniversityQingdaoChina
| | - Cunle Zhu
- Liver Disease CenterThe Affiliated Hospital of Qingdao UniversityQingdaoChina
| | - Dexi Chen
- Liver Disease CenterThe Affiliated Hospital of Qingdao UniversityQingdaoChina
- Beijing Institute of HepatologyCapital Medical UniversityBeijingChina
| | - Qingguo Xu
- Liver Disease CenterThe Affiliated Hospital of Qingdao UniversityQingdaoChina
- Academician WorkstationChangsha Medical UniversityChangshaChina
| | - Yuan Guo
- Liver Disease CenterThe Affiliated Hospital of Qingdao UniversityQingdaoChina
| |
Collapse
|
11
|
Lu H, Yan H, Li X, Xing Y, Ye Y, Jiang S, Ma L, Ping J, Zuo H, Hao Y, Yu C, Li Y, Zhou G, Lu Y. Single-cell map of dynamic cellular microenvironment of radiation-induced intestinal injury. Commun Biol 2023; 6:1248. [PMID: 38071238 PMCID: PMC10710489 DOI: 10.1038/s42003-023-05645-w] [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: 04/05/2023] [Accepted: 11/29/2023] [Indexed: 12/18/2023] Open
Abstract
Intestine is a highly radiation-sensitive organ that could be injured during the radiotherapy for pelvic, abdominal, and retroperitoneal tumors. However, the dynamic change of the intestinal microenvironment related to radiation-induced intestine injury (RIII) is still unclear. Using single-cell RNA sequencing, we pictured a dynamic landscape of the intestinal microenvironment during RIII and regeneration. We showed that the various cell types of intestine exhibited heterogeneous radiosensitivities. We revealed the distinct dynamic patterns of three subtypes of intestinal stem cells (ISCs), and the cellular trajectory analysis suggested a complex interconversion pattern among them. For the immune cells, we found that Ly6c+ monocytes can give rise to both pro-inflammatory macrophages and resident macrophages after RIII. Through cellular communication analysis, we identified a positive feedback loop between the macrophages and endothelial cells, which could amplify the inflammatory response induced by radiation. Besides, we identified different T cell subtypes and revealed their role in immunomodulation during the early stage of RIII through inflammation and defense response relevant signaling pathways. Overall, our study provides a valuable single-cell map of the multicellular dynamics during RIII and regeneration, which may facilitate the understanding of the mechanism of RIII.
Collapse
Affiliation(s)
- Hao Lu
- Beijing Institute of Radiation Medicine, Beijing, 100850, China
| | - Hua Yan
- Beijing Institute of Radiation Medicine, Beijing, 100850, China
| | - Xiaoyu Li
- Beijing Institute of Radiation Medicine, Beijing, 100850, China
| | - Yuan Xing
- Beijing Institute of Radiation Medicine, Beijing, 100850, China
| | - Yumeng Ye
- Beijing Institute of Radiation Medicine, Beijing, 100850, China
| | - Siao Jiang
- Beijing Institute of Radiation Medicine, Beijing, 100850, China
- College of Life Sciences, Hebei University, Baoding City, Hebei Province, 071002, China
| | - Luyu Ma
- Beijing Institute of Radiation Medicine, Beijing, 100850, China
| | - Jie Ping
- Beijing Institute of Radiation Medicine, Beijing, 100850, China
| | - Hongyan Zuo
- Beijing Institute of Radiation Medicine, Beijing, 100850, China
| | - Yanhui Hao
- Beijing Institute of Radiation Medicine, Beijing, 100850, China
| | - Chao Yu
- Beijing Institute of Radiation Medicine, Beijing, 100850, China
| | - Yang Li
- Beijing Institute of Radiation Medicine, Beijing, 100850, China.
- Academy of Life Sciences, Anhui Medical University, Hefei City, Anhui Province, 230032, China.
| | - Gangqiao Zhou
- Beijing Institute of Radiation Medicine, Beijing, 100850, China.
- Collaborative Innovation Center for Personalized Cancer Medicine, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing City, Jiangsu Province, 211166, China.
| | - Yiming Lu
- Beijing Institute of Radiation Medicine, Beijing, 100850, China.
- College of Life Sciences, Hebei University, Baoding City, Hebei Province, 071002, China.
| |
Collapse
|
12
|
Zhang R, Liu H, Lin J, Ding J, You J, Geng J. AhR may be involved in Th17 cell differentiation in chronic hepatitis B. J Viral Hepat 2023; 30:939-950. [PMID: 37608767 DOI: 10.1111/jvh.13883] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 07/19/2023] [Accepted: 08/09/2023] [Indexed: 08/24/2023]
Abstract
Th17 cells which are crucial for host immunity have been demonstrated to increase HBV infection. However, the mechanism of the Th17 cell increase is unknown. Hence, the mechanism of Th17 cell enhancement is important to provide a theoretical foundation for chronic hepatitis B immunotherapy. This study included 15 instances in the healthy control (HC) and 15 cohorts in the chronic hepatitis B (CHB). Their CD4+ T cells were isolated from their peripheral blood and then subjected to RNA transcriptome sequencing. Then, to identify target genes linked to Th17-cell differentiation, DEGs associated with CHB were convergent with the Th17-cell-associated genes from the KEGG database. Hub genes of DEG and target genes linked to Th17 cells were analysed for correlation. The AhR-related genes were located using the GeneMANIA database. To analyse the function of the genes, GO and KEGG pathways were employed. Protein-protein interaction network analysis employed the Metascape, STRING and Cytoscape databases. Finally, Western blotting and RT-qPCR were used to validate AhR. A total of 348 differential genes were identified in CHB patients. CytoHubba was used for screening five hub genes associated with CHB: CXCL10, RACGAP1, TPX2, FN1 and GZMA. This study aimed to determine the mechanism of elevated Th17 cells in CHB. As a result, further investigation using the convergence of DGEs and Th17 cell-related genes identified three target genes: AhR, HLA-DQA1 and HLA-DQB1, all of which were elevated in CHB. The three genes were primarily involved in immune response-related processes, according to the GO enrichment analysis. Correlation analysis of CXCL10, RACGAP1, TPX2, FN1 and GZMA genes with AhR, HLA-DQA1 and HLA-DQB1 revealed that AhR was positively associated with CXCL10 and GZMA genes, which best respond to the severity of CHB disease. Combined with the role of AhR in Th17 cell differentiation, the genes AhR was chosen for confirmation by RT-qPCR and WB in this study. The results showed that the CHB group had higher expression levels of AhR at both RT-qPCR and WB levels. Furthermore, this study's findings revealed that AhR may contribute to the development of CHB by affecting the differentiation of Th17 cells.
Collapse
Affiliation(s)
- Ruyi Zhang
- Department of Infectious Diseases and Hepatology, The First Affiliated Hospital of Kunming Medical University, Kunming, China
- Department of Infectious Diseases and Hepatology, The First People's Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, China
| | - Huaie Liu
- Department of Infectious Diseases and Hepatology, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Jie Lin
- Department of Infectious Diseases and Hepatology, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Jie Ding
- The Third People's Hospital of Kunming, Kunming, China
| | - Jing You
- Department of Infectious Diseases and Hepatology, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Jiawei Geng
- Department of Infectious Diseases and Hepatology, The First People's Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, China
| |
Collapse
|
13
|
Zhao Y, Wei Y, Fan L, Nie Y, Li J, Zeng R, Li J, Zhan X, Lei L, Kang Z, Li J, Zhang W, Yang Z. Leveraging a disulfidptosis-related signature to predict the prognosis and immunotherapy effectiveness of cutaneous melanoma based on machine learning. Mol Med 2023; 29:145. [PMID: 37884883 PMCID: PMC10601311 DOI: 10.1186/s10020-023-00739-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Accepted: 10/13/2023] [Indexed: 10/28/2023] Open
Abstract
BACKGROUND Disulfidptosis is a recently discovered programmed cell death pathway. However, the exact molecular mechanism of disulfidptosis in cutaneous melanoma remains unclear. METHODS In this study, clustering analysis was performed using data from public databases to construct a prognostic model, which was subsequently externally validated. The biological functions of the model genes were then investigated through various experimental techniques, including qRT-PCR, Western blotting, CCK-8 assay, wound healing assay, and Transwell assay. RESULTS We constructed a signature using cutaneous melanoma (CM) data, which accurately predicts the overall survival (OS) of patients. The predictive value of this signature for prognosis and immune therapy response was validated using multiple external datasets. High-risk CM subgroups may exhibit decreased survival rates, alterations in the tumor microenvironment (TME), and increased tumor mutation burden. We initially verified the expression levels of five optimum disulfidptosis-related genes (ODRGs) in normal tissues and CM. The expression levels of these genes were further confirmed in HaCaT cells and three melanoma cell lines using qPCR and protein blotting analysis. HLA-DQA1 emerged as the gene with the highest regression coefficient in our risk model, highlighting its role in CM. Mechanistically, HLA-DQA1 demonstrated the ability to suppress CM cell growth, proliferation, and migration. CONCLUSION In this study, a novel signature related to disulfidptosis was constructed, which accurately predicts the survival rate and treatment sensitivity of CM patients. Additionally, HLA-DQA1 is expected to be a feasible therapeutic target for effective clinical treatment of CM.
Collapse
Affiliation(s)
- Yi Zhao
- Tumor Research and Therapy Center, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Yanjun Wei
- Department of Radiation Oncology, Weifang People's Hospital, Weifang, China
| | - Lingjia Fan
- Department of Orthopaedic Surgery, Taian Central Hospital, Taian, Shandong, China
| | - Yuanliu Nie
- Tumor Research and Therapy Center, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Jianan Li
- Tumor Research and Therapy Center, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Renya Zeng
- Tumor Research and Therapy Center, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Jixian Li
- Tumor Research and Therapy Center, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Xiang Zhan
- Tumor Research and Therapy Center, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Lingli Lei
- Tumor Research and Therapy Center, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Zhichao Kang
- Tumor Research and Therapy Center, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Jiaxin Li
- Tumor Research and Therapy Center, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Wentao Zhang
- Tumor Research and Therapy Center, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China.
| | - Zhe Yang
- Tumor Research and Therapy Center, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China.
| |
Collapse
|
14
|
Hao L, Li S, Hu X. New insights into T-cell exhaustion in liver cancer: from mechanism to therapy. J Cancer Res Clin Oncol 2023; 149:12543-12560. [PMID: 37423958 DOI: 10.1007/s00432-023-05083-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Accepted: 06/29/2023] [Indexed: 07/11/2023]
Abstract
Liver cancer is one of the most common malignancies. T-cell exhaustion is associated with immunosuppression of tumor and chronic infection. Although immunotherapies that enhance the immune response by targeting programmed cell death-1(PD-1)/programmed cell death ligand 1 (PD-L1) have been applied to malignancies, these treatments have shown limited response rates. This suggested that additional inhibitory receptors (IRs) also contributed to T-cell exhaustion and tumor prognosis. Exhausted T-cells (Tex) in the tumor immune microenvironment (TME) are usually in a dysfunctional state of exhaustion, such as impaired activity and proliferative ability, increased apoptosis rate, and reduced production of effector cytokines. Tex cells participate in the negative regulation of tumor immunity mainly through IRs on the cell surface, changes in cytokines and immunomodulatory cell types, causing tumor immune escape. However, T-cell exhaustion is not irreversible and targeted immune checkpoint inhibitors (ICIs) can effectively reverse the exhaustion of T-cells and restore the anti-tumor immune response. Therefore, the research on the mechanism of T-cell exhaustion in liver cancer, aimed at maintaining or restoring the effector function of Tex cells, might provide a new method for the treatment of liver cancer. In this review, we summarized the basic characteristics of Tex cells (such as IRs and cytokines), discussed the mechanisms associated with T-cell exhaustion, and specifically discussed how these exhaustion characteristics were acquired and shaped by key factors within TME. Then new insights into the molecular mechanism of T-cell exhaustion suggested a potential way to improve the efficacy of cancer immunotherapy, namely to restore the effector function of Tex cells. In addition, we also reviewed the research progress of T-cell exhaustion in recent years and provided suggestions for further research.
Collapse
Affiliation(s)
- Liyuan Hao
- Chengdu University of Traditional Chinese Medicine, No. 37 Shi-Er-Qiao Road, Chengdu, 610075, Sichuan Province, People's Republic of China
- Hospital of Chengdu University of Traditional Chinese Medicine, No. 39 Shi-Er-Qiao Road, Chengdu, 610072, Sichuan Province, People's Republic of China
| | - Shenghao Li
- Chengdu University of Traditional Chinese Medicine, No. 37 Shi-Er-Qiao Road, Chengdu, 610075, Sichuan Province, People's Republic of China
- Hospital of Chengdu University of Traditional Chinese Medicine, No. 39 Shi-Er-Qiao Road, Chengdu, 610072, Sichuan Province, People's Republic of China
| | - Xiaoyu Hu
- Hospital of Chengdu University of Traditional Chinese Medicine, No. 39 Shi-Er-Qiao Road, Chengdu, 610072, Sichuan Province, People's Republic of China.
| |
Collapse
|
15
|
Liu M, Lai M, Li D, Zhang R, Wang L, Peng W, Yang J, He W, Sheng Y, Xiao S, Nan A, Zeng X. Nucleus-localized circSLC39A5 suppresses hepatocellular carcinoma development by binding to STAT1 to regulate TDG transcription. Cancer Sci 2023; 114:3884-3899. [PMID: 37549641 PMCID: PMC10551608 DOI: 10.1111/cas.15906] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Revised: 06/16/2023] [Accepted: 06/26/2023] [Indexed: 08/09/2023] Open
Abstract
Accumulating evidence indicates that circular RNAs (circRNAs) are inextricably linked to cancer development. However, the function and mechanism of nucleus-localized circRNAs in hepatocellular carcinoma (HCC) still require investigation. Here, qRT-PCR and receiver-operating characteristic curve were used to detect the expression and diagnostic potential of circSLC39A5 for HCC. The biological function of circSLC39A5 in HCC was investigated in vitro and in vivo. Nucleoplasmic separation assay, fluorescence in situ hybridization, RNA pulldown, RNA immunoprecipitation, the HDOCK Server, the NucleicNet Webserver, crosslinking-immunoprecipitation, MG132 treatment, and chromatin immunoprecipitation were utilized to explore the potential molecular mechanism of circSLC39A5 in HCC. The results showed that circSLC39A5 was downregulated in both HCC tissues and plasma and was associated with satellite nodules and lymph node metastasis/vascular invasion. CircSLC39A5 was stably expressed in plasma samples under different storage conditions, showing good diagnostic potential for HCC (AUC = 0.915). CircSLC39A5 inhibited proliferation, migration, and invasion, facilitated the apoptosis of HCC cells, and was associated with low expression of Ki67 and CD34. Remarkably, circSLC39A5 is mainly localized in the nucleus and binds to the transcription factor signal transducer and activator of transcription 1 (STAT1), affecting its stabilization and expression. STAT1 binds to the promoter of thymine DNA glycosylase (TDG). Overexpression of circSLC39A5 elevates TDG expression and reverses the increase of proliferating cell nuclear antigen (PCNA) expression and the overactive cell proliferation caused by TDG silencing. Our findings uncovered a novel plasma circRNA, circSLC39A5, which may be a potential circulating diagnostic marker for HCC, and the mechanism by which nucleus-localized circSLC39A5 exerts a transcriptional regulatory role in HCC by affecting STAT1/TDG/PCNA provides new insights into the mechanism of circRNAs.
Collapse
Affiliation(s)
- Meiliang Liu
- Department of Epidemiology and Health Statistics, School of Public HealthGuangxi Medical UniversityNanningChina
- Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent DiseasesGuangxi Medical UniversityNanningChina
| | - Mingshuang Lai
- Department of Epidemiology and Health Statistics, School of Public HealthGuangxi Medical UniversityNanningChina
- Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent DiseasesGuangxi Medical UniversityNanningChina
| | - Deyuan Li
- Department of Epidemiology and Health Statistics, School of Public HealthGuangxi Medical UniversityNanningChina
- Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent DiseasesGuangxi Medical UniversityNanningChina
| | - Ruirui Zhang
- Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent DiseasesGuangxi Medical UniversityNanningChina
- Department of Toxicology, School of Public HealthGuangxi Medical UniversityNanningChina
| | - Lijun Wang
- Department of Epidemiology and Health Statistics, School of Public HealthGuangxi Medical UniversityNanningChina
- Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent DiseasesGuangxi Medical UniversityNanningChina
| | - Wenyi Peng
- Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent DiseasesGuangxi Medical UniversityNanningChina
- Department of Toxicology, School of Public HealthGuangxi Medical UniversityNanningChina
| | - Jialei Yang
- Department of Epidemiology and Health Statistics, School of Public HealthGuangxi Medical UniversityNanningChina
- Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent DiseasesGuangxi Medical UniversityNanningChina
| | - Wanting He
- Department of Epidemiology and Health Statistics, School of Public HealthGuangxi Medical UniversityNanningChina
- Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent DiseasesGuangxi Medical UniversityNanningChina
| | - Yonghong Sheng
- Department of Epidemiology and Health Statistics, School of Public HealthGuangxi Medical UniversityNanningChina
- Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent DiseasesGuangxi Medical UniversityNanningChina
| | - Suyang Xiao
- Department of Epidemiology and Health Statistics, School of Public HealthGuangxi Medical UniversityNanningChina
- Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent DiseasesGuangxi Medical UniversityNanningChina
| | - Aruo Nan
- Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent DiseasesGuangxi Medical UniversityNanningChina
- Department of Toxicology, School of Public HealthGuangxi Medical UniversityNanningChina
| | - Xiaoyun Zeng
- Department of Epidemiology and Health Statistics, School of Public HealthGuangxi Medical UniversityNanningChina
- Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent DiseasesGuangxi Medical UniversityNanningChina
- Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor (Guangxi Medical University), Ministry of EducationNanningChina
- Guangxi Key Laboratory of Early Prevention and Treatment for Regional High Frequency TumorNanningChina
| |
Collapse
|
16
|
Wang H, Wang N, Tang Z, Liu Q, Nie S, Tao W. An 8-gene predicting survival model of hepatocellular carcinoma (HCC) related to pyroptosis and cuproptosis. Hereditas 2023; 160:30. [PMID: 37464443 DOI: 10.1186/s41065-023-00288-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2022] [Accepted: 05/17/2023] [Indexed: 07/20/2023] Open
Abstract
BACKGROUND The study aimed to establish a prognostic survival model with 8 pyroptosis-and-cuproptosis-related genes to examine the prognostic effect in patients of hepatocellular carcinoma (HCC). METHODS We downloaded gene expression data and clinical information of HCC patients from The Cancer Genome Atlas (TCGA), International Cancer Genome Consortium (ICGC) and Gene Expression Omnibus (GEO). The clustering analysis and cox regression with LASSO were used for constructing an 8 PCmRNAs survival model. Using TCGA, ICGC and GEO cohort, the overall survival (OS) between high- and low- risk group was determined. We also evaluated independent prognostic indicators using univariate and multivariate analyses. The relatively bioinformatics analysis, including immune cell infiltration, function enrichment and drug sensitivity analyses, was performed as well. The gene expression of 8 PCmRNAs in vitro were validated in several HCC cell lines by qRT-PCR and Western blot. The relationship between GZMA and Fludarabine were further checked by CCK-8 assay. RESULTS The survival prognostic model was constructed with ATP7A, GLS, CDKN2A, BAK1, CHMP4B, NLRP6, NOD1 and GZMA using data from TCGA cohort. The ICGC and GEO cohort were used for model validation. Receiver operating characteristic (ROC) curves showed a good survival prediction by this model. Risk scores had the highest predictable value for survival among Stage, Age, Gender and Grade. Most Immune cells and immune functions were decreased in high-risk group. Besides, function enrichment analyses showed that steroid metabolic process, hormone metabolic process, collagen - containing extracellular matrix, oxidoreductase activity and pyruvate metabolism were enriched. Potential drugs targeted different PCDEGs like Nelarabine, Dexamethasone and Fludarabine were found as well. ATP7A, GLS, CDKN2A, BAK1, CHMP4B, NOD1 were upregulated while NLRP6 and GZMA were downregulated in most HCC cell lines. The potential therapy of Fludarabine was demonstrated when GZMA was low expressed in Huh7 cell line. CONCLUSION We constructed a novel 8-gene (ATP7A, GLS, CDKN2A, BAK1, CHMP4B, NLRP6, NOD1 and GZMA) prognostic model and explored potential functional information and microenvironment of HCC, which might be worthy of clinical application. In addition, several potential chemotherapy drugs were screened and Fludarabine might be effective for HCC patients whose GZMA was low expressed.
Collapse
Affiliation(s)
- Hongjin Wang
- Department of Critical Care Medicine, Yongchuan Hospital, Chongqing Medical University, Yong Chuan, Chongqing, 402160, China
| | - Nian Wang
- Department of Critical Care Medicine, Yongchuan Hospital, Chongqing Medical University, Yong Chuan, Chongqing, 402160, China
| | - Ze Tang
- Department of Critical Care Medicine, Yongchuan Hospital, Chongqing Medical University, Yong Chuan, Chongqing, 402160, China
| | - Qiuyu Liu
- Department of Critical Care Medicine, Yongchuan Hospital, Chongqing Medical University, Yong Chuan, Chongqing, 402160, China
| | - Shiyu Nie
- Department of Critical Care Medicine, Yongchuan Hospital, Chongqing Medical University, Yong Chuan, Chongqing, 402160, China
| | - Wu Tao
- Department of Critical Care Medicine, Yongchuan Hospital, Chongqing Medical University, Yong Chuan, Chongqing, 402160, China.
| |
Collapse
|
17
|
Cao T, Xie F, Shi Y, Xu J, Liu Y, Cui D, Zhang F, Lin L, Li W, Gao Y, Ruan Y, Wang X, Zhu Y, Han B, Xia S, Guo W, Li B, Jing Y. Rapamycin and Low-dose IL-2 Mediate an Immunosuppressive Microenvironment to Inhibit Benign Prostatic Hyperplasia. Int J Biol Sci 2023; 19:3441-3455. [PMID: 37497009 PMCID: PMC10367549 DOI: 10.7150/ijbs.85089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2023] [Accepted: 06/20/2023] [Indexed: 07/28/2023] Open
Abstract
Benign prostatic hyperplasia (BPH) is a condition that becomes more common with age and manifests itself primarily as the expansion of the prostate and surrounding tissues. However, to date, the etiology of BPH remains unclear. In this respect, we performed single-cell RNA sequencing of prostate transition zone tissues from elderly individuals with different prostate volumes to reveal their distinct tissue microenvironment. Ultimately, we demonstrated that a reduced Treg/CD4+ T-cell ratio in the large-volume prostate and a relatively activated immune microenvironment were present, characterized partially by increased expression levels of granzymes, which may promote vascular growth and profibrotic processes and further exacerbate BPH progression. Consistently, we observed that the prostate gland of patients taking immunosuppressive drugs usually remained at a smaller volume. Furthermore, in mouse models, we confirmed that both suppression of the immune system with rapamycin and induction of Treg proliferation with low doses of IL-2 therapy indeed prevented the progression of BPH. Taken together, our findings suggest that an activated immune microenvironment is necessary for prostate volume growth and that Tregs can reverse this immune activation state, thereby inhibiting the progression of BPH.
Collapse
Affiliation(s)
- Tianyu Cao
- Department of Urology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Feng Xie
- Department of Immunology and Microbiology, Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Youwei Shi
- Department of Urology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Junhao Xu
- Department of Urology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yi Liu
- Department of Plastic Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Di Cui
- Department of Urology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Fang Zhang
- Department of Urology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lihui Lin
- Department of Laboratory Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Weize Li
- Department of Laboratory Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yanting Gao
- Department of Laboratory Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yuan Ruan
- Department of Urology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaohai Wang
- Department of Urology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yiping Zhu
- Department of Urology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Bangmin Han
- Department of Urology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shujie Xia
- Department of Urology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wenhuan Guo
- Department of Pathology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Bin Li
- Center for Immune-Related Diseases at Shanghai Institute of Immunology, Department of Respiratory and Critical Care Medicine of Ruijin Hospital, Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Department of Thoracic Surgery, Clinical Translational Research Center, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
- Institute of Arthritis Research, Guanghua Integrative Medicine Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Department of Integrated TCM & Western Medicine, Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, China
- Henan Key Laboratory for Digestive Organ Transplantation, Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Shenzhen Key Laboratory of Immunity and Inflammatory Diseases, Shenzhen, China
| | - Yifeng Jing
- Department of Urology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
18
|
Kuang T, Zhang L, Chai D, Chen C, Wang W. Construction of a T-cell exhaustion-related gene signature for predicting prognosis and immune response in hepatocellular carcinoma. Aging (Albany NY) 2023; 15:5751-5774. [PMID: 37354485 PMCID: PMC10333082 DOI: 10.18632/aging.204830] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Accepted: 06/12/2023] [Indexed: 06/26/2023]
Abstract
BACKGROUND Hepatocellular carcinoma (HCC) is a heterogeneous malignancy with a rising prevalence worldwide. Immunotherapy has been shown to improve treatment outcomes for HCC. We aimed to construct a T-cell exhaustion-related gene prognostic model (TEXPM) for HCC and to elucidate the immunologic characteristics and advantages of immunotherapy in T-cell exhaustion-Related Gene-defined HCC groups. METHODS Single-cell RNA sequencing data were used in conjunction with TCGA Differentially expressed genes (DEGs) to screen for T-cell exhaustion-Related Genes (TEXGs) for subsequent evaluation. Using univariate Cox regression analysis and LASSO regression analysis, five genes (FTL, GZMA, CD14, NPC2, and IER3) were subsequently selected for the construction of a TEXPM. Then, we evaluated the immunologic characteristics and advantages of immunotherapy in groups identified by TEXPM. RESULTS The TEXPM was formed with FTL, GZMA, CD14, NPC2, and IER3. The results of the training and validation team studies were consistent, with the low TEXPM group surviving longer than the high TEXPM group (P < 0.001). Multivariate Cox regression analysis demonstrated that TEXPM (HR: 2.347, 95%CI: 1.844-2.987; HR: 2.172, 95% CI: 1.689-2.793) was an independent prognostic variable for HCC patients. The low-TEXPM group was linked to active immunity, less aggressive phenotypes, strong infiltration of CD8+ T cells, CD4 + T cells, and M1 macrophages, and a better response to ICI treatment. A high TEXPM group, on the other hand, was associated with suppressive immunity, more aggressive phenotypes, a significant infiltration of B cells, M0 macrophages, and M2 macrophages, and a reduced response to ICI treatment. FTL is an independent prognostic variable in HCC patients and the knockdown of FTL can affect the biological behavior of hepatocellular carcinoma cells. CONCLUSIONS TEXPM is a promising prognostic biomarker connected to the immune system. Differentiating immunological and molecular features and predicting patient outcomes may be facilitated by TEXPM grouping. Furthermore, the expression of FTL was found to be an independent prognostic factor for HCC. Knockdown of FTL significantly inhibited proliferation, migration, and invasive activity in liver cancer cells.
Collapse
Affiliation(s)
- Tianrui Kuang
- Department of General Surgery, Renmin Hospital of Wuhan University, Wuhan, China
- Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, China
| | - Lilong Zhang
- Department of General Surgery, Renmin Hospital of Wuhan University, Wuhan, China
- Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, China
| | - Dongqi Chai
- Department of General Surgery, Renmin Hospital of Wuhan University, Wuhan, China
- Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, China
| | - Chen Chen
- Department of General Surgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Weixing Wang
- Department of General Surgery, Renmin Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
19
|
De Re V, Tornesello ML, Racanelli V, Prete M, Steffan A. Non-Classical HLA Class 1b and Hepatocellular Carcinoma. Biomedicines 2023; 11:1672. [PMID: 37371767 PMCID: PMC10296335 DOI: 10.3390/biomedicines11061672] [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: 04/28/2023] [Revised: 06/02/2023] [Accepted: 06/06/2023] [Indexed: 06/29/2023] Open
Abstract
A number of studies are underway to gain a better understanding of the role of immunity in the pathogenesis of hepatocellular carcinoma and to identify subgroups of individuals who may benefit the most from systemic therapy according to the etiology of their tumor. Human leukocyte antigens play a key role in antigen presentation to T cells. This is fundamental to the host's defense against pathogens and tumor cells. In addition, HLA-specific interactions with innate lymphoid cell receptors, such those present on natural killer cells and innate lymphoid cell type 2, have been shown to be important activators of immune function in the context of several liver diseases. More recent studies have highlighted the key role of members of the non-classical HLA-Ib and the transcript adjacent to the HLA-F locus, FAT10, in hepatocarcinoma. The present review analyzes the major contribution of these molecules to hepatic viral infection and hepatocellular prognosis. Particular attention has been paid to the association of natural killer and Vδ2 T-cell activation, mediated by specific HLA class Ib molecules, with risk assessment and novel treatment strategies to improve immunotherapy in HCC.
Collapse
Affiliation(s)
- Valli De Re
- Immunopathology and Cancer Biomarkers Unit, Centro di Riferimento Oncologico di Aviano (CRO), Istituti di Ricovero e Cura a Carattere Scientifico (IRCCS), 33081 Aviano, Italy;
| | - Maria Lina Tornesello
- Molecular Biology and Viral Oncology Unit, Istituto Nazionale Tumori IRCCS “Fondazione G. Pascale”, 80131 Naples, Italy;
| | - Vito Racanelli
- Department of Interdisciplinary Medicine, School of Medicine, ‘Aldo Moro’ University of Bari, 70124 Bari, Italy; (V.R.); (M.P.)
| | - Marcella Prete
- Department of Interdisciplinary Medicine, School of Medicine, ‘Aldo Moro’ University of Bari, 70124 Bari, Italy; (V.R.); (M.P.)
| | - Agostino Steffan
- Immunopathology and Cancer Biomarkers Unit, Centro di Riferimento Oncologico di Aviano (CRO), Istituti di Ricovero e Cura a Carattere Scientifico (IRCCS), 33081 Aviano, Italy;
| |
Collapse
|
20
|
Hu M, Li X, Jiang Z, Xia Q, Hu Y, Guo J, Fu L. Exosomes and circular RNAs: promising partners in hepatocellular carcinoma from bench to bedside. Discov Oncol 2023; 14:60. [PMID: 37154831 PMCID: PMC10167081 DOI: 10.1007/s12672-023-00672-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2023] [Accepted: 04/26/2023] [Indexed: 05/10/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is characterized by high morbidity and mortality, and a low 5-year survival rate. Exploring the potential molecular mechanisms, finding diagnostic biomarkers with high sensitivity and specificity, and determining new therapeutic targets for HCC are urgently needed. Circular RNAs (circRNAs) have been found to play a key role in the occurrence and development of HCC, while exosomes play an important role in intercellular communication; thus, the combination of circRNAs and exosomes may have inestimable potential in early diagnosis and curative therapy. Previous studies have shown that exosomes can transfer circRNAs from normal or abnormal cells to surrounding or distant cells; thereafter, circRNAs influence target cells. This review summarizes the recent progress regarding the roles of exosomal circRNAs in the diagnosis, prognosis, occurrence and development and immune checkpoint inhibitor and tyrosine kinase inhibitor resistance of HCC to provide inspiration for further research.
Collapse
Affiliation(s)
- Mengyuan Hu
- School of Medicine, Ningbo University, Ningbo, 315211, China
- Department of Infection and Hepatology, Ningbo No. 2 Hospital, Ningbo, 315010, China
| | - Xue Li
- Wenzhou Medical University, Wenzhou, 325035, China
| | - Zhenluo Jiang
- Department of Emergency, Ningbo No. 2 Hospital, Ningbo, 315010, China
| | - Qing Xia
- Department of Hepatopancreatobiliary Surgery, Ningbo No. 2 Hospital, Ningbo, 315010, China
| | - Yaoren Hu
- School of Medicine, Ningbo University, Ningbo, 315211, China
- Department of Infection and Hepatology, Ningbo No. 2 Hospital, Ningbo, 315010, China
- Key Laboratory of Diagnosis and Treatment of Digestive System Tumors of Zhejiang Province, Ningbo, 315010, China
| | - Junming Guo
- School of Medicine, Ningbo University, Ningbo, 315211, China
- Department of Biochemistry and Molecular Biology, and Zhejiang Key Laboratory of Pathophysiology, Medical School of Ningbo University, Ningbo, 315211, China
| | - Liyun Fu
- School of Medicine, Ningbo University, Ningbo, 315211, China.
- Department of Infection and Hepatology, Ningbo No. 2 Hospital, Ningbo, 315010, China.
- Wenzhou Medical University, Wenzhou, 325035, China.
- Key Laboratory of Diagnosis and Treatment of Digestive System Tumors of Zhejiang Province, Ningbo, 315010, China.
- Ningbo Clinical Research Center for Digestive System Tumors, Ningbo, 315010, China.
| |
Collapse
|
21
|
Gao YX, Ning QQ, Yang PX, Guan YY, Liu PX, Liu ML, Qiao LX, Guo XH, Yang TW, Chen DX. Recent advances in recurrent hepatocellular carcinoma therapy. World J Hepatol 2023; 15:460-476. [PMID: 37206651 PMCID: PMC10190692 DOI: 10.4254/wjh.v15.i4.460] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Revised: 12/20/2022] [Accepted: 03/24/2023] [Indexed: 04/20/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is the most prevalent form of primary liver cancer, accounting for 75%-85% of cases. Although treatments are given to cure early-stage HCC, up to 50%-70% of individuals may experience a relapse of the illness in the liver after 5 years. Research on the fundamental treatment modalities for recurrent HCC is moving significantly further. The precise selection of individuals for therapy strategies with established survival advantages is crucial to ensuring better outcomes. These strategies aim to minimize substantial morbidity, support good life quality, and enhance survival for patients with recurrent HCC. For individuals with recurring HCC after curative treatment, no approved therapeutic regimen is currently available. A recent study presented novel approaches, like immunotherapy and antiviral medication, to improve the prognosis of patients with recurring HCC with the apparent lack of data to guide the clinical treatment. The data supporting several neoadjuvant and adjuvant therapies for patients with recurring HCC are outlined in this review. We also discuss the potential for future clinical and translational investigations.
Collapse
Affiliation(s)
- Yu-Xue Gao
- Beijing Institute of Hepatology, Beijing You An Hospital, Capital Medical University, Beijing 100069, China
- Beijing Precision Medicine and Transformation Engineering Technology Research Center of Hepatitis and Liver Cancer, Beijing 100069, China
| | - Qi-Qi Ning
- Beijing Institute of Hepatology, Beijing You An Hospital, Capital Medical University, Beijing 100069, China
- Beijing Precision Medicine and Transformation Engineering Technology Research Center of Hepatitis and Liver Cancer, Beijing 100069, China
| | - Peng-Xiang Yang
- Beijing Institute of Hepatology, Beijing You An Hospital, Capital Medical University, Beijing 100069, China
- Beijing Precision Medicine and Transformation Engineering Technology Research Center of Hepatitis and Liver Cancer, Beijing 100069, China
| | - Yuan-Yue Guan
- Beijing Institute of Hepatology, Beijing You An Hospital, Capital Medical University, Beijing 100069, China
- Beijing Precision Medicine and Transformation Engineering Technology Research Center of Hepatitis and Liver Cancer, Beijing 100069, China
| | - Peng-Xiang Liu
- Beijing Institute of Hepatology, Beijing You An Hospital, Capital Medical University, Beijing 100069, China
- Beijing Precision Medicine and Transformation Engineering Technology Research Center of Hepatitis and Liver Cancer, Beijing 100069, China
| | - Meng-Lu Liu
- Beijing Institute of Hepatology, Beijing You An Hospital, Capital Medical University, Beijing 100069, China
- Beijing Precision Medicine and Transformation Engineering Technology Research Center of Hepatitis and Liver Cancer, Beijing 100069, China
| | - Lu-Xin Qiao
- Beijing Institute of Hepatology, Beijing You An Hospital, Capital Medical University, Beijing 100069, China
- Beijing Precision Medicine and Transformation Engineering Technology Research Center of Hepatitis and Liver Cancer, Beijing 100069, China
| | - Xiang-Hua Guo
- Beijing Institute of Hepatology, Beijing You An Hospital, Capital Medical University, Beijing 100069, China
- Beijing Precision Medicine and Transformation Engineering Technology Research Center of Hepatitis and Liver Cancer, Beijing 100069, China
| | - Tong-Wang Yang
- Beijing Institute of Hepatology, Beijing You An Hospital, Capital Medical University, Beijing 100069, China
- Academician Workstation, Changsha Medical University, Changsha 410219, Hunan Province, China
- Hunan Key Laboratory of the Research and Development of Novel Pharmaceutical Preparations, Changsha Medical University, Changsha 410219, Hunan Province, China
| | - De-Xi Chen
- Beijing Institute of Hepatology, Beijing You An Hospital, Capital Medical University, Beijing 100069, China
- Beijing Precision Medicine and Transformation Engineering Technology Research Center of Hepatitis and Liver Cancer, Beijing 100069, China
| |
Collapse
|
22
|
Liu X, Liu Y, Xu J, Zhang Y, Ruan Y, Zhao Y, Wu L, Hu J, Zhang Z, He M, Chen T, Xu X, Zhang J, Zhang Y, Zhou P. Single-cell transcriptomic analysis deciphers key transitional signatures associated with oncogenic evolution in human intramucosal oesophageal squamous cell carcinoma. Clin Transl Med 2023; 13:e1203. [PMID: 36855810 PMCID: PMC9975454 DOI: 10.1002/ctm2.1203] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2022] [Revised: 02/07/2023] [Accepted: 02/13/2023] [Indexed: 03/02/2023] Open
Abstract
BACKGROUND AND AIMS The early diagnosis and intervention of oesophageal squamous cell carcinoma (ESCC) are particularly important because of the lack of effective therapies and poor prognosis. Comprehensive research on early ESCC at the single-cell level is rare due to the need for fresh and high-quality specimens obtained from ESD. This study aims to systematically describe the cellular atlas of human intramucosal ESCC. METHODS Five paired samples of intramucosal ESCC, para-ESCC oesophageal tissues from endoscopically resected specimens and peripheral blood mononuclear cells were adopted for scRNA-seq analysis. Computational pipeline scMetabolism was applied to quantify the metabolic diversity of single cells. RESULTS A total of 164 715 cells were profiled. Epithelial cells exhibited high intra-tumoural heterogeneity and two evolutionary trajectories during ESCC tumorigenesis initiated from proliferative cells, and then through an intermediate state, to two different terminal states of normally differentiated epithelial cells or malignant cells, respectively. The abundance of CD8+ TEX s, Tregs and PD1+ CD4+ T cells suggested an exhausted and suppressive immune microenvironment. Several genes in immune cells, such as CXCL13, CXCR5 and PADI4, were identified as new biomarkers for poor prognosis. A new subcluster of malignant cells associated with metastasis and angiogenesis that appeared at an early stage compared with progressive ESCC was also identified in this study. Intercellular interaction analysis based on ligand-receptor pairs revealed the subcluster of malignant cells interacting with CAFs via the MDK-NCL pathway, which was verified by cell proliferation assay and IHC. This indicates that the interaction may be an important hallmark in the early change of tumour microenvironment and serves as a sign of CAF activation to stimulate downstream pathways for facilitating tumour invasion. CONCLUSION This study demonstrates the changes of cell subsets and transcriptional levels in human intramucosal ESCC, which may provide unique insights into the development of novel biomarkers and potential intervention strategies.
Collapse
Affiliation(s)
- Xin‐Yang Liu
- Department of Endoscopy Center and Endoscopy Research InstituteZhongshan HospitalFudan UniversityShanghaiChina
- Department of EndoscopyShanghai Collaborative Innovation CenterShanghaiChina
| | - Yan‐Bo Liu
- Department of Endoscopy Center and Endoscopy Research InstituteZhongshan HospitalFudan UniversityShanghaiChina
- Department of EndoscopyShanghai Collaborative Innovation CenterShanghaiChina
| | - Jia‐Cheng Xu
- Department of Endoscopy Center and Endoscopy Research InstituteZhongshan HospitalFudan UniversityShanghaiChina
- Department of EndoscopyShanghai Collaborative Innovation CenterShanghaiChina
| | - Yi‐Fei Zhang
- Department of Endoscopy Center and Endoscopy Research InstituteZhongshan HospitalFudan UniversityShanghaiChina
- Department of EndoscopyShanghai Collaborative Innovation CenterShanghaiChina
| | - Yuan‐Yuan Ruan
- Department of Biochemistry and Molecular Biology, School of Basic Medical SciencesFudan UniversityShanghaiChina
| | | | - Lin‐Feng Wu
- Department of Endoscopy Center and Endoscopy Research InstituteZhongshan HospitalFudan UniversityShanghaiChina
- Department of EndoscopyShanghai Collaborative Innovation CenterShanghaiChina
| | - Jian‐Wei Hu
- Department of Endoscopy Center and Endoscopy Research InstituteZhongshan HospitalFudan UniversityShanghaiChina
- Department of EndoscopyShanghai Collaborative Innovation CenterShanghaiChina
| | - Zhen Zhang
- Department of Endoscopy Center and Endoscopy Research InstituteZhongshan HospitalFudan UniversityShanghaiChina
- Department of EndoscopyShanghai Collaborative Innovation CenterShanghaiChina
| | - Meng‐Jiang He
- Department of Endoscopy Center and Endoscopy Research InstituteZhongshan HospitalFudan UniversityShanghaiChina
- Department of EndoscopyShanghai Collaborative Innovation CenterShanghaiChina
| | - Tian‐Yin Chen
- Department of Endoscopy Center and Endoscopy Research InstituteZhongshan HospitalFudan UniversityShanghaiChina
- Department of EndoscopyShanghai Collaborative Innovation CenterShanghaiChina
| | - Xiao‐Yue Xu
- Department of Endoscopy Center and Endoscopy Research InstituteZhongshan HospitalFudan UniversityShanghaiChina
- Department of EndoscopyShanghai Collaborative Innovation CenterShanghaiChina
| | - Jing‐Wei Zhang
- Department of Genetic Engineering State Key LaboratorySchool of Life SciencesFudan UniversityShanghaiChina
| | - Yi‐Qun Zhang
- Department of Endoscopy Center and Endoscopy Research InstituteZhongshan HospitalFudan UniversityShanghaiChina
- Department of EndoscopyShanghai Collaborative Innovation CenterShanghaiChina
| | - Ping‐Hong Zhou
- Department of Endoscopy Center and Endoscopy Research InstituteZhongshan HospitalFudan UniversityShanghaiChina
- Department of EndoscopyShanghai Collaborative Innovation CenterShanghaiChina
| |
Collapse
|