1
|
Li J, Gao X, Lv L, Huang Y, Zhang H, Sun X, Zhu L. Development of a coagulation‑related gene model for prognostication, immune response and treatment prediction in lung adenocarcinoma. Oncol Lett 2025; 29:290. [PMID: 40276086 PMCID: PMC12018795 DOI: 10.3892/ol.2025.15035] [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: 10/15/2024] [Accepted: 03/13/2025] [Indexed: 04/26/2025] Open
Abstract
Lung adenocarcinoma (LUAD) is the most prevalent form of lung cancer worldwide. Due to the lack of clinically useful molecular biomarkers, the diagnosis and prognosis of patients with LUAD remain poor. Patients with LUAD often exhibit abnormalities in the levels of coagulation factors. Therefore, the objective of the present study was to develop a model based on coagulation-related factors in LUAD. Gene expression data and clinical information from 582 patients with LUAD were obtained from The Cancer Genome Atlas (TCGA). A set of 138 coagulation-related genes (CRGs) was retrieved from The Molecular Signatures Database, and their expression levels were examined in TCGA dataset to identify differentially expressed CRGs. Predictive models were constructed using least absolute shrinkage and selection operator-Cox regression. The risk score from the model was used to establish high- and low-risk patient groups. Additionally, Kaplan-Meier analyses were performed to evaluate the differences in overall survival (OS) and progression-free survival between the two groups. The accuracy of the model was verified through receiver operating characteristic and principal component analysis. In addition, the tumor immune dysfunction and exclusion algorithm was used to assess immune escape and immunotherapy responses in relation to the CRGs. A predictive model comprising four genes, namely matrix metalloproteinase (MMP) 1, MMP10, cathepsin V and thrombin, was established to estimate the survival rate of patients with LUAD. The OS rates of patients in the high-risk group were lower compared with those in the low-risk group. Furthermore, a combination of high-risk score and low tumor mutation burden was associated with the poorest survival in patients with LUAD. Patients in different risk groups exhibited different drug sensitivities based on their risk scores. In conclusion, the four-gene based prognostic model served as an independent predictor of survival rates in patients with LUAD and may offer a novel approach for prognosis and treatment.
Collapse
Affiliation(s)
- Jia Li
- Department of Thoracic Surgery, Jinan Central Hospital, Jinan, Shandong 250013, P.R. China
- Department of Cardiac Surgery, Beijing Anzhen Hospital, Capital Medical University, Beijing 100000, P.R. China
- Department of Cardiac Surgery, Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing 100000, P.R. China
| | - Xuedi Gao
- Department of Surgery, The Fourth People's Hospital of Jinan, Jinan, Shandong 250013, P.R. China
| | - Lin Lv
- Department of Thoracic Surgery, Jinan Central Hospital, Jinan, Shandong 250013, P.R. China
| | - Yubin Huang
- Department of Thoracic Surgery, Jinan Central Hospital, Jinan, Shandong 250013, P.R. China
| | - Houlu Zhang
- Department of Thoracic Surgery, Jinan Central Hospital, Jinan, Shandong 250013, P.R. China
| | - Xiaoming Sun
- Department of Thoracic Surgery, Jinan Central Hospital, Jinan, Shandong 250013, P.R. China
| | - Liangming Zhu
- Department of Thoracic Surgery, Jinan Central Hospital, Jinan, Shandong 250013, P.R. China
- Department of Thoracic Surgery, Medical Integration and Practice Center, Shandong University, Jinan, Shandong 250013, P.R. China
| |
Collapse
|
2
|
Klawon DE, Pagane N, Walker MT, Ganci NK, Miller CH, Gai E, Rodriguez DM, Ryan-Payseur BK, Duncombe RK, Adams EJ, Maienschein-Cline M, Freitag NE, Germain RN, Wong HS, Savage PA. Regulatory T cells constrain T cells of shared specificity to enforce tolerance during infection. Science 2025; 387:eadk3248. [PMID: 40014689 PMCID: PMC12006836 DOI: 10.1126/science.adk3248] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Revised: 09/16/2024] [Accepted: 12/17/2024] [Indexed: 03/01/2025]
Abstract
During infections, CD4+ Foxp3+ regulatory T (Treg) cells must control autoreactive CD4+ conventional T (Tconv) cell responses against self-peptide antigens while permitting those against pathogen-derived "nonself" peptides. We defined the basis of this selectivity using mice in which Treg cells reactive to a single prostate-specific self-peptide were selectively depleted. We found that self-peptide-specific Treg cells were dispensable for the control of Tconv cells of matched specificity at homeostasis. However, they were required to control such Tconv cells and prevent autoimmunity toward the prostate after exposure to elevated self-peptide during infection. Notably, the Treg cell response to self-peptide did not affect protective Tconv cell responses to a pathogen-derived peptide. Thus, self-peptide-specific Treg cells promoted self-nonself discrimination during infection by selectively controlling Tconv cells of shared self-specificity.
Collapse
Affiliation(s)
- David E.J. Klawon
- Department of Pathology, University of Chicago; Chicago, IL 60637, USA
- Present address: Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Nicole Pagane
- The Ragon Institute of Mass General, MIT and Harvard; Cambridge, MA 02139, USA
- Program in Computational and Systems Biology, Massachusetts Institute of Technology; Cambridge, MA 02139, USA
| | - Matthew T. Walker
- Department of Pathology, University of Chicago; Chicago, IL 60637, USA
| | - Nicole K. Ganci
- Department of Pathology, University of Chicago; Chicago, IL 60637, USA
| | - Christine H. Miller
- Department of Pathology, University of Chicago; Chicago, IL 60637, USA
- Interdisciplinary Scientist Training Program, University of Chicago; Chicago, IL 60637, USA
- Present address: Department of Pathology, University of California, San Francisco School of Medicine, San Francisco, CA 94117, USA
| | - Eric Gai
- The Ragon Institute of Mass General, MIT and Harvard; Cambridge, MA 02139, USA
- Program in Computational and Systems Biology, Massachusetts Institute of Technology; Cambridge, MA 02139, USA
| | - Donald M. Rodriguez
- Department of Pathology, University of Chicago; Chicago, IL 60637, USA
- Interdisciplinary Scientist Training Program, University of Chicago; Chicago, IL 60637, USA
| | - Bridgett K. Ryan-Payseur
- Department of Microbiology and Immunology, University of Illinois Chicago; Chicago, Illinois 60612 USA
| | - Ryan K. Duncombe
- Department of Biochemistry and Molecular Biology, University of Chicago; Chicago, IL 60637, USA
| | - Erin J. Adams
- Department of Biochemistry and Molecular Biology, University of Chicago; Chicago, IL 60637, USA
| | - Mark Maienschein-Cline
- Research Informatics Core, Research Resources Center, University of Illinois Chicago; Chicago, IL 60612 USA
| | - Nancy E. Freitag
- Department of Pharmaceutical Sciences, University of Illinois Chicago; Chicago, IL 60612, USA
| | - Ronald N. Germain
- Lymphocyte Biology Section, Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health; Bethesda, MD 20892, USA
| | - Harikesh S. Wong
- The Ragon Institute of Mass General, MIT and Harvard; Cambridge, MA 02139, USA
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Peter A. Savage
- Department of Pathology, University of Chicago; Chicago, IL 60637, USA
| |
Collapse
|
3
|
Wang X, Zhang J, Fang L, Tang X. Angel and devil: the protective immunity and pathogenic inflammation of tissue resident memory T cells in ulcerative colitis. Front Immunol 2025; 16:1518339. [PMID: 40124381 PMCID: PMC11925784 DOI: 10.3389/fimmu.2025.1518339] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Accepted: 02/14/2025] [Indexed: 03/25/2025] Open
Abstract
Ulcerative colitis (UC) is an incurable autoimmune disease. Patients with UC endure the burden of recurrent flare-ups and face a substantial economic burden due to long-term medication. The complex etiology and unclear pathogenesis pose a significant challenge to the development of effective and curative treatments. Recent research indicates that local memory at the site of inflammatory intestinal mucosa in UC is closely associated with the persistent presence of tissue-resident memory T (TRM) cells. TRM cells, a subset of memory T cells, exhibit long-lived, low-migration characteristics. These cells reside in tissues, where they provide immediate immune protection while also contributing to chronic, localized inflammation. The presence of TRM cells in the inflamed intestinal mucosa of UC patients is a crucial factor in the recurrence of the disease. However, the process involved in the formation and differentiation of TRM cells within the intestinal mucosa remains poorly understood. Various surface markers, transcriptional networks, and signaling pathways regulate the formation and maintenance of TRM cells in the intestine. To further understand the role of TRM cells in UC pathogenesis, we have summarized the latest findings to pave the way for the development of future targeted therapies.
Collapse
Affiliation(s)
- Xintong Wang
- Institute of Digestive Diseases, Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, China
- Graduate School, China Academy of Chinese Medical Sciences, Beijing, China
| | - Jiaqi Zhang
- Institute of Digestive Diseases, Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, China
- Department of Gastroenterology, Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, China
| | - Lihui Fang
- Institute of Digestive Diseases, Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, China
- Graduate School, Beijing University of Chinese Medicine, Beijing, China
| | - Xudong Tang
- Institute of Digestive Diseases, Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
4
|
Meng M, Wang J, Yang J, Zhang Y, Tu X, Hu P. PRR13 expression as a prognostic biomarker in breast cancer: correlations with immune infiltration and clinical outcomes. Front Mol Biosci 2025; 12:1518031. [PMID: 40099041 PMCID: PMC11911201 DOI: 10.3389/fmolb.2025.1518031] [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: 10/27/2024] [Accepted: 01/08/2025] [Indexed: 03/19/2025] Open
Abstract
Introduction Breast cancer continues to be a primary cause of cancer-related mortality among women globally. Identifying novel biomarkers is essential for enhancing patient prognosis and informing therapeutic decisions. The PRR13 gene, associated with taxol resistance and the progression of various cancers, remains under-characterized in breast cancer. This study aimed to investigate the role of PRR13 in breast cancer and its potential as a prognostic biomarker. Methods We performed a comparative analysis of PRR13 gene expression utilizing the TCGA database against non-cancerous tissues and employed STRING to evaluate PRR13's protein-protein interactions and associated pathways. Additionally, we investigated the relationship between PRR13 mRNA expression and immune cell infiltration in breast cancer (BRCA) using two methodologies. Furthermore, a retrospective analysis of 160 patients was conducted, wherein clinical data were collected and PRR13 expression was evaluated through immunohistochemistry and qRT-PCR to determine its association with clinicopathological features and patient survival. Results Analysis of the TCGA database revealed significant upregulation of PRR13 expression across 12 different cancer types, including breast cancer. High PRR13 expression was positively correlated with various immune cells, including NK cells, eosinophils, Th17 cells, and mast cells, whereas a negative correlation was observed with B cells, macrophages, and other immune subsets. Enrichment analysis of PRR13 and its 50 interacting proteins revealed significant associations with biological processes such as cell adhesion and migration, and pathways including ECMreceptor interaction and PI3K-Akt signaling. Single-cell analysis demonstrated associations between PRR13 and pathways pertinent to inflammation and apoptosis. Validation studies confirmed elevated PRR13 expression in tumor tissue compared to adjacent non-cancerous tissue. Immunohistochemistry demonstrated high PRR13 expression in 55.6% of cancer cases, particularly associated with advanced clinical stage and lymph node metastasis. Moreover, high PRR13 expression significantly correlated with shorter overall survival and served as an independent prognostic factor. Subgroup analysis underscored the prognostic significance of PRR13 in aggressive tumor subtypes, with particularly strong associations observed in T3, N1-3, and moderately to poorly differentiated tumors. Discussion In conclusion, PRR13 expression is upregulated in breast cancer tissues and may serve as a valuable prognostic indicator for breast cancer patients, potentially impacting patient survival and therapeutic strategies.
Collapse
Affiliation(s)
- Mingjing Meng
- Department of Research and Foreign Affairs, The Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou, China
| | - Jiani Wang
- Breast Cancer Center, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Jiumei Yang
- Guangdong Second Provincial General Hospital of Jinan University, Guangzhou, China
| | - Yangming Zhang
- Equipment Department, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Xusheng Tu
- Emergency Department, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Pan Hu
- Breast Cancer Center, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
5
|
Wei X, Sui K, Peng Y, Li S, Fang Y, Chen Z, Du X, Xie X, Tang H, Wen Q, Li J, He M, Cheng Q, Zhang W. Human Umbilical Cord Mesenchymal Stem Cell-Derived Exosomes Loaded Mir-29-3p Targets AhR to Improve Juvenile Idiopathic Arthritis via Inhibiting the Expression of IL-22 in CD4 + T Cell. Stem Cell Rev Rep 2025; 21:536-553. [PMID: 39621151 DOI: 10.1007/s12015-024-10827-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/21/2024] [Indexed: 01/03/2025]
Abstract
BACKGROUND Juvenile idiopathic arthritis (JIA) is one of the most common chronic inflammatory rheumatic diseases in children. Human umbilical cord mesenchymal stem cells (HUCMSCs)-derived exosomes (HUCMSCs-Exos) are involved in autoimmune diseases. This study investigates the mechanism of HUCMSC-Exos in improving JIA by targeting AhR through delivery of miR-29-3p to inhibit IL-22 expression in CD4+ T cells. METHODS Collagen induced arthritis (CIA) mouse model was established, and mice were treated with HUCMSCs-Exos and miR-29-3p antagomir, respectively. CD4+ T cells from JIA patients were used for cell experiments. The mechanism was elucidated by histopathological staining, transmission electron microscopy (TEM), immunohistochemistry, CCK-8 assay, flow cytometry, Western blotting, real-time PCR, and enzyme-linked immunosorbent assay (ELISA), laser confocal microscopy, and luciferase assay. RESULT JIA-CD4+ T cells showed higher expression of IL-22 and lower the levels of miR-29-3p, while HUCMSCs-Exos significantly inhibited the expression of IL-22 and increased the levels of miR-29a-3p, miR-29b-3p, and miR-29c-3p in CD4+ T cells from JIA patients. The expression of miR-29a-3p, miR-29b-3p, miR-29c-3p, AhR, and IL-22 in CD4+ T cells was significantly reversed when co-cultured with HUCMSCs transfected with miR-29-3p mimic or miR-29-3p inhibitor. In vivo experiment, HUCMSCs-Exos ameliorated CIA mice by delivering miR-29-3p to inhibit AhR, IL-22, IL-22R1, MMP3, and MMP13 expression. Furthermore, HUCMSCs-Exos also deliver miR-29-3p targeting AhR expression to inhibit IL-22 in JIA-CD4 + T cells through alleviating arthritic synovial fibroblast activation. CONCLUSION HUCMSCs-Exos loaded miR-29-3p targets AhR to improve JIA via inhibiting the expression of IL-22 in CD4+ T cell, which provides a scientific basis for the treatment of JIA.
Collapse
Affiliation(s)
- Xinyi Wei
- Pediatric Immunology and Rheumatology Department, School of Medicine, Chengdu Women's and Children's Central Hospital, University of Electronic Science and Technology of China, Chengdu, 611731, China
| | - Kunpeng Sui
- Pediatric Immunology and Rheumatology Department, School of Medicine, Chengdu Women's and Children's Central Hospital, University of Electronic Science and Technology of China, Chengdu, 611731, China
| | - Yuanyuan Peng
- Pediatric Immunology and Rheumatology Department, School of Medicine, Chengdu Women's and Children's Central Hospital, University of Electronic Science and Technology of China, Chengdu, 611731, China
| | - Sha Li
- Pediatric Immunology and Rheumatology Department, School of Medicine, Chengdu Women's and Children's Central Hospital, University of Electronic Science and Technology of China, Chengdu, 611731, China
| | - Yu Fang
- Pediatric Immunology and Rheumatology Department, School of Medicine, Chengdu Women's and Children's Central Hospital, University of Electronic Science and Technology of China, Chengdu, 611731, China
| | - Zhi Chen
- Pediatric Immunology and Rheumatology Department, School of Medicine, Chengdu Women's and Children's Central Hospital, University of Electronic Science and Technology of China, Chengdu, 611731, China
| | - Xiao Du
- Pediatric Immunology and Rheumatology Department, School of Medicine, Chengdu Women's and Children's Central Hospital, University of Electronic Science and Technology of China, Chengdu, 611731, China
| | - Xue Xie
- Pediatric Immunology and Rheumatology Department, School of Medicine, Chengdu Women's and Children's Central Hospital, University of Electronic Science and Technology of China, Chengdu, 611731, China
| | - Haiming Tang
- Pediatric Immunology and Rheumatology Department, School of Medicine, Chengdu Women's and Children's Central Hospital, University of Electronic Science and Technology of China, Chengdu, 611731, China
| | - QiuYue Wen
- Pediatric Immunology and Rheumatology Department, School of Medicine, Chengdu Women's and Children's Central Hospital, University of Electronic Science and Technology of China, Chengdu, 611731, China
| | - JingWei Li
- Pediatric Immunology and Rheumatology Department, School of Medicine, Chengdu Women's and Children's Central Hospital, University of Electronic Science and Technology of China, Chengdu, 611731, China
| | - Meilin He
- Pediatric Immunology and Rheumatology Department, School of Medicine, Chengdu Women's and Children's Central Hospital, University of Electronic Science and Technology of China, Chengdu, 611731, China
| | - Qin Cheng
- Pediatric Immunology and Rheumatology Department, School of Medicine, Chengdu Women's and Children's Central Hospital, University of Electronic Science and Technology of China, Chengdu, 611731, China
| | - Wei Zhang
- Pediatric Immunology and Rheumatology Department, School of Medicine, Chief Physician, Chengdu Women's and Children's Central Hospital, University of Electronic Science and Technology of China, No.1617, Riyue Avenue, Qingyang District, Chengdu, Sichuan, China.
| |
Collapse
|
6
|
Richards KA, Changrob S, Thomas PG, Wilson PC, Sant AJ. Lack of memory recall in human CD4 T cells elicited by the first encounter with SARS-CoV-2. iScience 2024; 27:109992. [PMID: 38868209 PMCID: PMC11166706 DOI: 10.1016/j.isci.2024.109992] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Revised: 04/11/2024] [Accepted: 05/13/2024] [Indexed: 06/14/2024] Open
Abstract
The studies reported here focus on the impact of pre-existing CD4 T cell immunity on the first encounter with SARS-CoV-2. They leverage PBMC samples from plasma donors collected after a first SARS-CoV-2 infection, prior to vaccine availability and compared to samples collected prior to the emergence of SARS-CoV-2. Analysis of CD4 T cell specificity across the entire SARS-CoV-2 proteome revealed that the recognition of SARS-CoV-2-derived epitopes by CD4 memory cells prior to the pandemic are enriched for reactivity toward non-structural proteins conserved across endemic CoV strains. However, CD4 T cells after primary infection with SARS-CoV-2 focus on epitopes from structural proteins. We observed little evidence for preferential recall to epitopes conserved between SARS-CoV-2 and seasonal CoV, a finding confirmed through use of selectively curated conserved and SARS-unique peptides. Our data suggest that SARS-CoV-2 CD4 T cells elicited by the first infection are primarily established from the naive CD4 T cell pool.
Collapse
Affiliation(s)
- Katherine A. Richards
- David H. Smith Center for Vaccine Biology and Immunology, University of Rochester Medical Center, Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Siriruk Changrob
- Drukier Institute for Children’s Health, Department of Pediatrics, Weill Cornell Medicine, New York, NY 10065, USA
| | - Paul G. Thomas
- Department of Immunology, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA
| | - Patrick C. Wilson
- Drukier Institute for Children’s Health, Department of Pediatrics, Weill Cornell Medicine, New York, NY 10065, USA
| | - Andrea J. Sant
- David H. Smith Center for Vaccine Biology and Immunology, University of Rochester Medical Center, Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY 14642, USA
| |
Collapse
|
7
|
Li Y, Ye X, Huang H, Cao R, Huang F, Chen L. Construction of a prognostic model based on memory CD4+ T cell-associated genes for lung adenocarcinoma and its applications in immunotherapy. CPT Pharmacometrics Syst Pharmacol 2024; 13:837-852. [PMID: 38594917 PMCID: PMC11098152 DOI: 10.1002/psp4.13122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Revised: 02/20/2024] [Accepted: 02/26/2024] [Indexed: 04/11/2024] Open
Abstract
The association between memory CD4+ T cells and cancer prognosis is increasingly recognized, but their impact on lung adenocarcinoma (LUAD) prognosis remains unclear. In this study, using the cell-type identification by estimating relative subsets of RNA transcripts algorithm, we analyzed immune cell composition and patient survival in LUAD. Weighted gene coexpression network analysis helped identify memory CD4+ T cell-associated gene modules. Combined with module genes, a five-gene LUAD prognostic risk model (HOXB7, MELTF, ABCC2, GNPNAT1, and LDHA) was constructed by regression analysis. The model was validated using the GSE31210 data set. The validation results demonstrated excellent predictive performance of the risk scoring model. Correlation analysis was conducted between the clinical information and risk scores of LUAD samples, revealing that LUAD patients with disease progression exhibited higher risk scores. Furthermore, univariate and multivariate regression analyses demonstrated the model independent prognostic capability. The constructed nomogram results demonstrated that the predictive performance of the nomogram was superior to the prognostic model and outperformed individual clinical factors. Immune landscape assessment was performed to compare different risk score groups. The results revealed a better prognosis in the low-risk group with higher immune infiltration. The low-risk group also showed potential benefits from immunotherapy. Our study proposes a memory CD4+ T cell-associated gene risk model as a reliable prognostic biomarker for personalized treatment in LUAD patients.
Collapse
Affiliation(s)
- Yong Li
- Pulmonary and Critical Care MedicineFujian Medical University Union HospitalFuzhouChina
| | - Xiangli Ye
- Pulmonary and Critical Care MedicineFujian Medical University Union HospitalFuzhouChina
| | - Huiqin Huang
- Fujian Provincial Key Laboratory of Medical TestingFujian Academy of Medical SciencesFuzhouChina
| | - Rongxiang Cao
- Pulmonary and Critical Care MedicineFujian Medical University Union HospitalFuzhouChina
| | - Feijian Huang
- Pulmonary and Critical Care MedicineFujian Medical University Union HospitalFuzhouChina
| | - Limin Chen
- Pulmonary and Critical Care MedicineFujian Medical University Union HospitalFuzhouChina
| |
Collapse
|
8
|
Bravo M, Dileepan T, Dolan M, Hildebrand J, Wolford J, Hanson ID, Hamilton SE, Frosch AE, Burrack KS. IL-15 Complex-Induced IL-10 Enhances Plasmodium-specific CD4+ T Follicular Helper Differentiation and Antibody Production. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2024; 212:992-1001. [PMID: 38305633 PMCID: PMC10932862 DOI: 10.4049/jimmunol.2300525] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Accepted: 01/09/2024] [Indexed: 02/03/2024]
Abstract
Malaria, which results from infection with Plasmodium parasites, remains a major public health problem. Although humans do not develop long-lived, sterilizing immunity, protection against symptomatic disease develops after repeated exposure to Plasmodium parasites and correlates with the acquisition of humoral immunity. Despite the established role Abs play in protection from malaria disease, dysregulated inflammation is thought to contribute to the suboptimal immune response to Plasmodium infection. Plasmodium berghei ANKA (PbA) infection results in a fatal severe malaria disease in mice. We previously demonstrated that treatment of mice with IL-15 complex (IL-15C; IL-15 bound to an IL-15Rα-Fc fusion protein) induces IL-10 expression in NK cells, which protects mice from PbA-induced death. Using a novel MHC class II tetramer to identify PbA-specific CD4+ T cells, in this study we demonstrate that IL-15C treatment enhances T follicular helper (Tfh) differentiation and modulates cytokine production by CD4+ T cells. Moreover, genetic deletion of NK cell-derived IL-10 or IL-10R expression on T cells prevents IL-15C-induced Tfh differentiation. Additionally, IL-15C treatment results in increased anti-PbA IgG Ab levels and improves survival following reinfection. Overall, these data demonstrate that IL-15C treatment, via its induction of IL-10 from NK cells, modulates the dysregulated inflammation during Plasmodium infection to promote Tfh differentiation and Ab generation, correlating with improved survival from reinfection. These findings will facilitate improved control of malaria infection and protection from disease by informing therapeutic strategies and vaccine design.
Collapse
Affiliation(s)
| | | | | | - Jacob Hildebrand
- Center for Immunology, University of Minnesota
- Department of Laboratory Medicine and Pathology, University of Minnesota
| | | | | | - Sara E. Hamilton
- Center for Immunology, University of Minnesota
- Department of Laboratory Medicine and Pathology, University of Minnesota
| | - Anne E. Frosch
- Hennepin Healthcare Research Institute
- Center for Immunology, University of Minnesota
| | - Kristina S. Burrack
- Hennepin Healthcare Research Institute
- Center for Immunology, University of Minnesota
| |
Collapse
|
9
|
Li K, Deng Z, Lei C, Ding X, Li J, Wang C. The Role of Oxidative Stress in Tumorigenesis and Progression. Cells 2024; 13:441. [PMID: 38474405 PMCID: PMC10931308 DOI: 10.3390/cells13050441] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Revised: 02/20/2024] [Accepted: 02/29/2024] [Indexed: 03/14/2024] Open
Abstract
Oxidative stress refers to the imbalance between the production of reactive oxygen species (ROS) and the endogenous antioxidant defense system. Its involvement in cell senescence, apoptosis, and series diseases has been demonstrated. Advances in carcinogenic research have revealed oxidative stress as a pivotal pathophysiological pathway in tumorigenesis and to be involved in lung cancer, glioma, hepatocellular carcinoma, leukemia, and so on. This review combs the effects of oxidative stress on tumorigenesis on each phase and cell fate determination, and three features are discussed. Oxidative stress takes part in the processes ranging from tumorigenesis to tumor death via series pathways and processes like mitochondrial stress, endoplasmic reticulum stress, and ferroptosis. It can affect cell fate by engaging in the complex relationships between senescence, death, and cancer. The influence of oxidative stress on tumorigenesis and progression is a multi-stage interlaced process that includes two aspects of promotion and inhibition, with mitochondria as the core of regulation. A deeper and more comprehensive understanding of the effects of oxidative stress on tumorigenesis is conducive to exploring more tumor therapies.
Collapse
Affiliation(s)
| | | | | | | | | | - Changshan Wang
- Laboratory of Reproductive Regulation & Breeding of Grassland Livestock, School of Life Science, Inner Mongolia University, 49 Xilingol South Road, Yu Quan District, Hohhot 010020, China; (K.L.); (Z.D.); (C.L.); (X.D.); (J.L.)
| |
Collapse
|
10
|
Rixon JA, Fong KD, Morris C, Nguyen AT, Depew CE, McSorley SJ. Elimination of Chlamydia muridarum from the female reproductive tract is IL-12p40 dependent, but independent of Th1 and Th2 cells. PLoS Pathog 2024; 20:e1011914. [PMID: 38166152 PMCID: PMC10786385 DOI: 10.1371/journal.ppat.1011914] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Revised: 01/12/2024] [Accepted: 12/19/2023] [Indexed: 01/04/2024] Open
Abstract
Chlamydia vaccine approaches aspire to induce Th1 cells for optimal protection, despite the fact that there is no direct evidence demonstrating Th1-mediated Chlamydia clearance from the female reproductive tract (FRT). We recently reported that T-bet-deficient mice can resolve primary Chlamydia infection normally, undermining the potentially protective role of Th1 cells in Chlamydia immunity. Here, we show that T-bet-deficient mice develop robust Th17 responses and that mice deficient in Th17 cells exhibit delayed bacterial clearance, demonstrating that Chlamydia-specific Th17 cells represent an underappreciated protective population. Additionally, Th2-deficient mice competently clear cervicovaginal infection. Furthermore, we show that sensing of IFN-γ by non-hematopoietic cells is essential for Chlamydia immunity, yet bacterial clearance in the FRT does not require IFN-γ secretion by CD4 T cells. Despite the fact that Th1 cells are not necessary for Chlamydia clearance, protective immunity to Chlamydia is still dependent on MHC class-II-restricted CD4 T cells and IL-12p40. Together, these data point to IL-12p40-dependent CD4 effector maturation as essential for Chlamydia immunity, and Th17 cells to a lesser extent, yet neither Th1 nor Th2 cell development is critical. Future Chlamydia vaccination efforts will be more effective if they focus on induction of this protective CD4 T cell population.
Collapse
Affiliation(s)
- Jordan A. Rixon
- Department of Anatomy, Physiology and Cell Biology, School of Veterinary Medicine, University of California Davis, Davis, California, United States of America
| | - Kevin D. Fong
- Department of Anatomy, Physiology and Cell Biology, School of Veterinary Medicine, University of California Davis, Davis, California, United States of America
| | - Claire Morris
- Department of Anatomy, Physiology and Cell Biology, School of Veterinary Medicine, University of California Davis, Davis, California, United States of America
| | - Alana T. Nguyen
- Department of Anatomy, Physiology and Cell Biology, School of Veterinary Medicine, University of California Davis, Davis, California, United States of America
| | - Claire E. Depew
- Department of Anatomy, Physiology and Cell Biology, School of Veterinary Medicine, University of California Davis, Davis, California, United States of America
| | - Stephen J. McSorley
- Department of Anatomy, Physiology and Cell Biology, School of Veterinary Medicine, University of California Davis, Davis, California, United States of America
| |
Collapse
|
11
|
Abstract
Specialized subpopulations of CD4+ T cells survey major histocompatibility complex class II-peptide complexes to control phagosomal infections, help B cells, regulate tissue homeostasis and repair or perform immune regulation. Memory CD4+ T cells are positioned throughout the body and not only protect the tissues from reinfection and cancer, but also participate in allergy, autoimmunity, graft rejection and chronic inflammation. Here we provide updates on our understanding of the longevity, functional heterogeneity, differentiation, plasticity, migration and human immunodeficiency virus reservoirs as well as key technological advances that are facilitating the characterization of memory CD4+ T cell biology.
Collapse
Affiliation(s)
- Marco Künzli
- Center for Immunology, Department of Microbiology and Immunology, University of Minnesota, Minneapolis, MN, USA
| | - David Masopust
- Center for Immunology, Department of Microbiology and Immunology, University of Minnesota, Minneapolis, MN, USA.
| |
Collapse
|
12
|
Kheradmand F, Zhang Y, Corry DB. Contribution of adaptive immunity to human COPD and experimental models of emphysema. Physiol Rev 2023; 103:1059-1093. [PMID: 36201635 PMCID: PMC9886356 DOI: 10.1152/physrev.00036.2021] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Revised: 09/15/2022] [Accepted: 09/20/2022] [Indexed: 02/01/2023] Open
Abstract
The pathophysiology of chronic obstructive pulmonary disease (COPD) and the undisputed role of innate immune cells in this condition have dominated the field in the basic research arena for many years. Recently, however, compelling data suggesting that adaptive immune cells may also contribute to the progressive nature of lung destruction associated with COPD in smokers have gained considerable attention. The histopathological changes in the lungs of smokers can be limited to the large or small airways, but alveolar loss leading to emphysema, which occurs in some individuals, remains its most significant and irreversible outcome. Critically, however, the question of why emphysema progresses in a subset of former smokers remained a mystery for many years. The recognition of activated and organized tertiary T- and B-lymphoid aggregates in emphysematous lungs provided the first clue that adaptive immune cells may play a crucial role in COPD pathophysiology. Based on these findings from human translational studies, experimental animal models of emphysema were used to determine the mechanisms through which smoke exposure initiates and orchestrates adaptive autoreactive inflammation in the lungs. These models have revealed that T helper (Th)1 and Th17 subsets promote a positive feedback loop that activates innate immune cells, confirming their role in emphysema pathogenesis. Results from genetic studies and immune-based discoveries have further provided strong evidence for autoimmunity induction in smokers with emphysema. These new findings offer a novel opportunity to explore the mechanisms underlying the inflammatory landscape in the COPD lung and offer insights for development of precision-based treatment to halt lung destruction.
Collapse
Affiliation(s)
- Farrah Kheradmand
- Department of Medicine, Baylor College of Medicine, Houston, Texas
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, Texas
- Biology of Inflammation Center, Baylor College of Medicine, Houston, Texas
- Center for Translational Research on Inflammatory Diseases (CTRID), Michael E. DeBakey Department of Veterans Affairs Medical Center, Houston, Texas
| | - Yun Zhang
- Department of Medicine, Baylor College of Medicine, Houston, Texas
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, Texas
| | - David B Corry
- Department of Medicine, Baylor College of Medicine, Houston, Texas
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, Texas
- Biology of Inflammation Center, Baylor College of Medicine, Houston, Texas
- Center for Translational Research on Inflammatory Diseases (CTRID), Michael E. DeBakey Department of Veterans Affairs Medical Center, Houston, Texas
| |
Collapse
|
13
|
Shanmuganad S, Ferguson A, Paranjpe A, Cianciolo EE, Katz JD, Herold MJ, Hildeman DA. Subset-specific and temporal control of effector and memory CD4+ T cell survival. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.01.530323. [PMID: 36909576 PMCID: PMC10002744 DOI: 10.1101/2023.03.01.530323] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/07/2023]
Abstract
Following their proliferative expansion and differentiation into effector cells like Th1, Tfh, and T central memory precursors (Tcmp), most effector CD4+ T cells die, while some survive and become memory cells. Here, we explored how Bcl-2 family members controlled the survival of CD4+ T cells during distinct phases of mouse acute LCMV infection. During expansion, we found that Th1 cells dominated the response, downregulated expression of Bcl-2, and did not require Bcl-2 for survival. Instead, they relied on the anti-apoptotic protein, A1 for survival. Similarly, Th17 cells in an EAE model also depended on A1 for survival. However, after the peak of the response, CD4+ effector T cells required Bcl-2 to counteract Bim to aid their transition into memory. This Bcl-2 dependence persisted in established memory CD4+ T cells. Combined, these data show a temporal switch in Bcl-2 family-mediated survival of CD4+ T cells over the course of an immune response. This knowledge can help improve T cell survival to boost immunity and conversely, target pathogenic T cells.
Collapse
|
14
|
Ma R, Su H, Jiao K, Liu J. Role of Th17 cells, Treg cells, and Th17/Treg imbalance in immune homeostasis disorders in patients with chronic obstructive pulmonary disease. Immun Inflamm Dis 2023; 11:e784. [PMID: 36840492 PMCID: PMC9950879 DOI: 10.1002/iid3.784] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Revised: 01/21/2023] [Accepted: 01/30/2023] [Indexed: 02/26/2023] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is the third leading cause of death worldwide, following strokes and cardiovascular diseases. Chronic lung inflammation is believed to play a role in the development of COPD. In addition, accumulating evidence shows that the immune system plays a crucial role in the pathogenesis of COPD. Significant advancements have been made in research on the pathogenesis of immune diseases and chronic inflammation in recent years, and T helper 17 (Th17) cells and regulatory T (Treg) cells have been found to play a crucial role in the autoimmune response. Th17 cells are a proinflammatory subpopulation that causes autoimmune disease and tissue damage. Treg cells, on the other hand, have a negative effect but can contribute to the occurrence of the same disease when their antagonism fails. This review mainly summarizes the biological characteristics of Th17 cells and Treg cells, their roles in chronic inflammatory diseases of COPD, and the role of the Th17/Treg ratio in the onset, development, and outcome of inflammatory disorders, as well as recent advancements in immunomodulatory treatment targeting Th17/Treg cells in COPD.
Collapse
Affiliation(s)
- Ru Ma
- Department of The First Clinical School of MedicineLanzhou UniversityLanzhouChina
- Department of Gansu Provincial People's HospitalLanzhouChina
| | - Hongling Su
- Department of The First Clinical School of MedicineLanzhou UniversityLanzhouChina
- Department of Gansu Provincial People's HospitalLanzhouChina
| | - Keping Jiao
- Department of The First Clinical School of MedicineLanzhou UniversityLanzhouChina
- Department of Gansu Provincial People's HospitalLanzhouChina
| | - Jian Liu
- Department of The First Clinical School of MedicineLanzhou UniversityLanzhouChina
| |
Collapse
|
15
|
Panda AK, Kim YH, Shevach EM. Control of Memory Phenotype T Lymphocyte Homeostasis: Role of Costimulation. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2022; 208:851-860. [PMID: 35039334 DOI: 10.4049/jimmunol.2100653] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Accepted: 12/06/2021] [Indexed: 11/19/2022]
Abstract
Foxp3+ T regulatory cells (Tregs), CD4+Foxp3- T cells, and CD8+ T cells are composed of naive phenotype (NP) and memory phenotype (MP) subsets. Ten to 20% of each MP T cell population are cycling (Ki-67+) in vivo. We investigated the contribution of costimulatory (CD28) and coinhibitory (CTLA-4, PD-1) receptors on MP T cell homeostatic proliferation in vivo in the mouse. Blockade of CD28-CD80/CD86 signaling completely abolished MP Tregs and profoundly inhibited MP CD4+Foxp3- T cell proliferation, but it did not affect MP CD8+ T cell proliferation. Marked enhancement of homeostatic proliferation of MP Tregs and MP CD4+Foxp3- T cells was seen after blocking CTLA4-CD80/CD86 interactions and PD-1-PD-L1/2 interactions, and greater enhancement was seen with blockade of both pathways. The CD28 pathway also played an important role in the expansion of Tregs and MP T cells after treatment of mice with agonistic Abs to members of the TNF receptor superfamily, which can act directly (anti-GITR, anti-OX40, anti-4-1BB) or indirectly (anti-CD40) on T cells. Induction of a cytokine storm by blocking the interaction of NK inhibitory receptors with MHC class I had no effect on Treg homeostasis, enhanced MP CD4+ proliferation, and expansion in a CD28-dependent manner, but it enhanced MP CD8+ T cell proliferation in a CD28-independent manner. Because MP T cells exert potent biologic effects primarily before the induction of adaptive immune responses, these findings have important implications for the use of biologic agents designed to suppress autoimmune disease or enhance T effector function in cancer that may have negative effects on MP T cells.
Collapse
Affiliation(s)
- Abir K Panda
- Cellular Immunology Section, Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
| | - Yong-Hee Kim
- Cellular Immunology Section, Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
| | - Ethan M Shevach
- Cellular Immunology Section, Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
| |
Collapse
|
16
|
Read KA, Oestreich KJ. Making memories with Bcl-6. J Exp Med 2022; 219:212863. [PMID: 34792529 PMCID: PMC8605494 DOI: 10.1084/jem.20212177] [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] [Indexed: 11/23/2022] Open
Abstract
For over a decade, mutual antagonism between the transcriptional repressors Bcl-6 and Blimp-1 has been appreciated as a key mechanistic determinant of lymphoid differentiation programs. Now, in this issue of JEM, Ciucci et al. (2021. J. Exp. Med.https://doi.org/10.1084/jem.20202343) demonstrate that this relationship is "central" to the generation of T cell memory.
Collapse
Affiliation(s)
- Kaitlin A Read
- Department of Microbial Infection and Immunity, The Ohio State University College of Medicine and Wexner Medical Center, Columbus, OH
| | - Kenneth J Oestreich
- Department of Microbial Infection and Immunity, The Ohio State University College of Medicine and Wexner Medical Center, Columbus, OH
| |
Collapse
|
17
|
Huang C, Zhang C, Sheng J, Wang D, Zhao Y, Qian L, Xie L, Meng Z. Identification and Validation of a Tumor Microenvironment-Related Gene Signature in Hepatocellular Carcinoma Prognosis. Front Genet 2021; 12:717319. [PMID: 34899826 PMCID: PMC8662347 DOI: 10.3389/fgene.2021.717319] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Accepted: 10/25/2021] [Indexed: 12/21/2022] Open
Abstract
Background: Hepatocellular carcinoma (HCC) is a typical inflammatory-related malignant tumor with complex immune tolerance microenvironment and poor prognosis. In this study, we aimed to construct a novel immune-related gene signature for the prognosis of HCC patients, exploring tumor microenvironment (TME) cell infiltration characterization and potential mechanisms. Methods: A total of 364 HCC samples with follow-up information in the TCGA-LIHC dataset were analyzed for the training of the prognostic signature. The Least Absolute Shrinkage and Selector Operation (LASSO) regression based on the IRGs was conducted to identify the prognostic genes and establish an immune risk signature. The immune cell infiltration in TME was estimated via the CIBERSORT method. Gene Set Variation Analysis (GSVA) was conducted to compare the biological pathways involved in the low-risk and high-risk groups. Furthermore, paraffin sections of HCC tissue microarrays containing 77 patients from Fudan University Shanghai Cancer Center were used for IHC staining. The clinical characteristics of the 77 HCC patients were collected and summarized for survival analysis validation via the Kaplan-Meier (KM) method. Results: Three-gene signature with close immune correlation (Risk score = EPO * 0.02838 + BIRC5 * 0.02477 + SPP1 * 0.0002044) was constructed eventually and proven to be an effective prognostic factor for HCC patients. The patients were divided into a high-risk and a low-risk group according to the optimal cutoff, and the survival analysis revealed that HCC samples with high-risk immuno-score had significantly poorer outcomes than the low-risk group (p < 0.0001). The results of CIBERSORT suggested that the immune cell activation was relatively higher in the low-risk group with better prognosis. Besides, GSVA analysis showed multiple signaling differences between the high- and low-risk group, indicating that the three-gene prognostic model can affect the prognosis of patients by affecting immune-related mechanisms. Tissue microarray (TMA) results further confirmed that the expression of three genes in HCC tissues was closely related to the prognosis of patients, respectively. Conclusion: In this study, we constructed and validated a robust three-gene signature with close immune correlation in HCC, which presented a reliable performance in the prediction of HCC patients' survival.
Collapse
Affiliation(s)
- Changjing Huang
- Minimally Invasive Therapy Center, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Chenyue Zhang
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.,Department of Integrated Therapy, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Jie Sheng
- Minimally Invasive Therapy Center, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Dan Wang
- Minimally Invasive Therapy Center, Fudan University Shanghai Cancer Center, Shanghai, China.,Cancer Institute, Fudan University Shanghai Cancer Center, Fudan University, Shanghai, China
| | - Yingke Zhao
- Minimally Invasive Therapy Center, Fudan University Shanghai Cancer Center, Shanghai, China.,Cancer Institute, Fudan University Shanghai Cancer Center, Fudan University, Shanghai, China
| | - Ling Qian
- Minimally Invasive Therapy Center, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Lin Xie
- Minimally Invasive Therapy Center, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Zhiqiang Meng
- Minimally Invasive Therapy Center, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| |
Collapse
|