1
|
Yang Q, Cao Y, Wang L, Dong Y, Zhao L, Geng Z, Bi Y, Liu G. Mechanical force receptor Piezo1 regulates T H9 cell differentiation. Cell Rep 2025; 44:115136. [PMID: 39932192 DOI: 10.1016/j.celrep.2024.115136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Revised: 08/30/2024] [Accepted: 12/11/2024] [Indexed: 05/02/2025] Open
Abstract
Interleukin (IL)-9-producing CD4+ T cells (TH9) are essential for mediating antitumor immunity, but the mechanisms of TH9 cell differentiation remain unclear. Here, we found that the mechanical force receptor Piezo1 is critical for regulating TH9 cell differentiation. Piezo1 deficiency in CD4+ T cells intrinsically inhibited TH9 cell differentiation, whereas ectopic Piezo1 expression promoted this process. Notably, Piezo1 deficiency inhibited TH9 cell differentiation and contributed to tumor development. Mechanistically, Piezo1 deficiency inhibits TH9 cell differentiation mainly through the SIRT3-succinate dehydrogenase A (SDHA)-oxidative phosphorylation (OXPHOS) pathway. SIRT3 deficiency or blockade of SDHA-OXPHOS signaling activity reversed the TH9 cell differentiation induced by Piezo1 deficiency. Moreover, HIF1α signaling is responsible for the TH9 cell differentiation induced by Piezo1 deficiency. Thus, our findings identify a redox metabolism signaling mechanism regulated by the mechanical force receptor Piezo1 that limits the mitochondrial SIRT3-SDHA-dependent OXPHOS pathway and triggers HIF1α-IL-9 to reprogram TH9 cell differentiation, with implications for future immunotherapy approaches.
Collapse
Affiliation(s)
- Qiuli Yang
- Key Laboratory of Cell Proliferation and Regulation Biology, Ministry of Education, College of Life Sciences, Beijing Normal University, Beijing 100875, China
| | - Yejin Cao
- Key Laboratory of Cell Proliferation and Regulation Biology, Ministry of Education, College of Life Sciences, Beijing Normal University, Beijing 100875, China
| | - Likun Wang
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Science, Beijing 100080, China
| | - Yingjie Dong
- Key Laboratory of Cell Proliferation and Regulation Biology, Ministry of Education, College of Life Sciences, Beijing Normal University, Beijing 100875, China
| | - Longhao Zhao
- Key Laboratory of Cell Proliferation and Regulation Biology, Ministry of Education, College of Life Sciences, Beijing Normal University, Beijing 100875, China
| | - Zi Geng
- Key Laboratory of Cell Proliferation and Regulation Biology, Ministry of Education, College of Life Sciences, Beijing Normal University, Beijing 100875, China
| | - Yujing Bi
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Science, Beijing 100080, China.
| | - Guangwei Liu
- Key Laboratory of Cell Proliferation and Regulation Biology, Ministry of Education, College of Life Sciences, Beijing Normal University, Beijing 100875, China.
| |
Collapse
|
2
|
Tan B, Tu C, Xiong H, Xu Y, Shi X, Zhang X, Yang R, Zhang N, Lin B, Liu M, Qin J, Du B. GITRL enhances cytotoxicity and persistence of CAR-T cells in cancer therapy. Mol Ther 2025:S1525-0016(25)00040-1. [PMID: 39863927 DOI: 10.1016/j.ymthe.2025.01.036] [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: 07/23/2024] [Revised: 01/05/2025] [Accepted: 01/22/2025] [Indexed: 01/27/2025] Open
Abstract
Chimeric antigen receptor (CAR)-T cell therapy has achieved remarkable clinical success in treating hematological malignancies. However, its clinical efficacy in solid tumors is less satisfactory, partially due to poor in vivo expansion and the limited persistence of CAR-T cells. Here, we demonstrated that the overexpression of GITR ligand enhances the anti-tumor activity of CAR-T cells. Compared to prostate-specific membrane antigen-BB-Z (PSMA-BB-Z) CAR-T, PSMA-BB-Z-GITRL CAR-T cells have much more interferon (IFN)-γ, TNF-α, and interleukin (IL)-9 secretion, a higher proportion of central memory T (TCM) cells and T helper 9 (Th9) cells, less expression of exhaustion markers, and robust proliferation capacity. Consequently, PSMA-BB-Z-GITRL CAR-T cells exhibited more potent anti-tumor activity against established solid tumors in vivo than PSMA-BB-Z CAR-T cells. The results of the in vivo persistence experiment also indicated that PSMA-BB-Z-GITRL CAR-T cells exhibited much more retention in mouse blood, spleen, and tumor tissue than PSMA-BB-Z CAR-T cells 15 days after CAR-T cell therapy. In addition, PSMA-BB-Z-GITRL CAR-T cells produce higher levels of IFN-γ, TNF-α, and IL-9 in mouse blood, exhibiting a higher proportion of TCM cells and a lower proportion of Treg cells compared to PSMA-BB-Z CAR-T cells. Our results demonstrate that the overexpression of GITRL has important implications for improving CAR-T cell-based human solid tumor immunotherapy.
Collapse
Affiliation(s)
- Binghe Tan
- Shanghai Frontiers Science Center of Genome Editing and Cell Therapy, Shanghai Key Laboratory of Regulatory Biology and School of Life Sciences, East China Normal University, Shanghai 200241, China; BRL Medicine, Inc., Shanghai 201109, China
| | - Chuntian Tu
- Shanghai Frontiers Science Center of Genome Editing and Cell Therapy, Shanghai Key Laboratory of Regulatory Biology and School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Hao Xiong
- Shanghai Frontiers Science Center of Genome Editing and Cell Therapy, Shanghai Key Laboratory of Regulatory Biology and School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Yongqian Xu
- Shanghai Frontiers Science Center of Genome Editing and Cell Therapy, Shanghai Key Laboratory of Regulatory Biology and School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Xiujuan Shi
- Shanghai Frontiers Science Center of Genome Editing and Cell Therapy, Shanghai Key Laboratory of Regulatory Biology and School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Xiaolin Zhang
- Shanghai Frontiers Science Center of Genome Editing and Cell Therapy, Shanghai Key Laboratory of Regulatory Biology and School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Ruijie Yang
- Shanghai Frontiers Science Center of Genome Editing and Cell Therapy, Shanghai Key Laboratory of Regulatory Biology and School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Na Zhang
- Shanghai Frontiers Science Center of Genome Editing and Cell Therapy, Shanghai Key Laboratory of Regulatory Biology and School of Life Sciences, East China Normal University, Shanghai 200241, China; BRL Medicine, Inc., Shanghai 201109, China
| | - Boxu Lin
- Shanghai Frontiers Science Center of Genome Editing and Cell Therapy, Shanghai Key Laboratory of Regulatory Biology and School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Mingyao Liu
- Shanghai Frontiers Science Center of Genome Editing and Cell Therapy, Shanghai Key Laboratory of Regulatory Biology and School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Juliang Qin
- Shanghai Frontiers Science Center of Genome Editing and Cell Therapy, Shanghai Key Laboratory of Regulatory Biology and School of Life Sciences, East China Normal University, Shanghai 200241, China.
| | - Bing Du
- Shanghai Frontiers Science Center of Genome Editing and Cell Therapy, Shanghai Key Laboratory of Regulatory Biology and School of Life Sciences, East China Normal University, Shanghai 200241, China.
| |
Collapse
|
3
|
Kalim M, Jing R, Guo W, Xing H, Lu Y. Functional diversity and regulation of IL-9-producing T cells in cancer immunotherapy. Cancer Lett 2024; 606:217306. [PMID: 39426662 PMCID: PMC11675864 DOI: 10.1016/j.canlet.2024.217306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Revised: 10/15/2024] [Accepted: 10/17/2024] [Indexed: 10/21/2024]
Abstract
IL-9-producing T cells (T9) regulate immunological responses that affect various cellular biological processes, though their precise function remains fully understood. Previous studies have linked T9 cells to conditions such as allergic disorders, parasitic infection clearance, and various types of cancers. While the functional heterogeneity of IL-9 and T9 cells in cancer development has been documented, these cells present promising therapeutic opportunities for treating solid tumors. This review highlights the roles of IL-9 and T9 cells in cancer progression and treatment responses, focusing on potential discrepancies in IL-9/IL-9R signaling between murine tumors and cancer patients. Additionally, we discuss the regulation of tumor-specific Th9/Tc9 cell differentiation, the therapeutic potential of these cells, and current strategies to enhance their anti-tumor activities.
Collapse
Affiliation(s)
- Muhammad Kalim
- Houston Methodist Cancer Center/Weill Cornell Medicine, Houston, TX, 77030, USA
| | - Rui Jing
- Houston Methodist Cancer Center/Weill Cornell Medicine, Houston, TX, 77030, USA
| | - Wei Guo
- Houston Methodist Cancer Center/Weill Cornell Medicine, Houston, TX, 77030, USA
| | - Hui Xing
- Houston Methodist Cancer Center/Weill Cornell Medicine, Houston, TX, 77030, USA
| | - Yong Lu
- Houston Methodist Cancer Center/Weill Cornell Medicine, Houston, TX, 77030, USA.
| |
Collapse
|
4
|
Poveda-Garavito N, Orozco Castaño CA, Torres-Llanos Y, Cruz-Rodriguez N, Parra-Medina R, Quijano S, Zabaleta J, Combita AL. ID1 and ID3 functions in the modulation of the tumour immune microenvironment in adult patients with B-cell acute lymphoblastic leukaemia. Front Immunol 2024; 15:1473909. [PMID: 39676870 PMCID: PMC11638060 DOI: 10.3389/fimmu.2024.1473909] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Accepted: 11/07/2024] [Indexed: 12/17/2024] Open
Abstract
Introduction B-cell acute lymphoblastic leukemia (B-ALL) in adults often presents a poor prognosis. ID1 and ID3 genes have been identified as predictors of poor response in Colombian adult B-ALL patients, contributing to cancer development. In various cancer models, these genes have been associated with immune regulatory populations within the tumor immune microenvironment (TIME). B-ALL progression alters immune cell composition and the bone marrow (BM) microenvironment, impacting disease progression and therapy response. This study investigates the relationship between ID1 and ID3 expression, TIME dynamics, and immune evasion mechanisms in adult B-ALL patients. Methods This exploratory study analysed BM samples from 10 B-ALL adult patients diagnosed at the National Cancer Institute of Colombia. First, RT-qPCR was used to assess ID1 and ID3 expression in BM tumour cells. Flow cytometry characterised immune populations in the TIME. RNA-seq evaluated immune genes associatedwith B-ALL immune response, while xCell and CytoSig analysed TIME cell profiles and cytokines. Pathway analysis, gene ontology, and differential gene expression (DEGs) were examined, with functional enrichment analysis performed using KEGG ontology. Results Patients were divided into two groups based on ID1 and ID3 expression, namely basal and overexpression. A total of 94 differentially expressed genes were identified between these groups, with top overexpressed genes associated with neutrophil pathways. Gene set enrichment analysis revealed increased expression of genes associated with neutrophil degranulation, immune response-related neutrophil activation, and neutrophil-mediated immunity. These findings correlated with xCell data. Overexpression group showed significant differences in neutrophils, monocytes and CD4+ naive T cells compared to basal group patients. Microenvironment and immune scores were also significantly different, consistent with the flow cytometry results. Elevated cytokine levels associated with neutrophil activation supported these findings. Validation was performed using the Therapeutically Applicable Research to Generate Effective Treatments (TARGET) TCGA B-ALL cohorts. Discussion These findings highlight significant differences in ID1 and ID3 expression levels and their impact on TIME populations, particularly neutrophil-related pathways. The results suggest a potential role for ID1 and ID3 in immune evasion in adult B-ALL, mediated through neutrophil activation and immune regulation.
Collapse
Affiliation(s)
- Nathaly Poveda-Garavito
- Grupo de Investigación en Biología del Cáncer - Instituto Nacional de Cancerología, Bogotá, Colombia
- Grupo de Investigación Traslacional en Oncología - Instituto Nacional de Cancerología, Bogotá, Colombia
- Maestría en Inmunología, Departamento de Microbiología - Universidad Nacional de Colombia, Bogotá, Colombia
| | - Carlos A Orozco Castaño
- Grupo de Investigación en Biología del Cáncer - Instituto Nacional de Cancerología, Bogotá, Colombia
- Grupo de Investigación Traslacional en Oncología - Instituto Nacional de Cancerología, Bogotá, Colombia
| | - Yulieth Torres-Llanos
- Grupo de Investigación en Biología del Cáncer - Instituto Nacional de Cancerología, Bogotá, Colombia
- Grupo de Investigación Traslacional en Oncología - Instituto Nacional de Cancerología, Bogotá, Colombia
- Laboratorio clínico, Hospital Universitario San Ignacio, Bogotá, Colombia
| | | | - Rafael Parra-Medina
- Departamento de Patología, Instituto Nacional de Cancerología, Bogotá, Colombia
- Research Institute, Fundación Universitaria de Ciencias de la Salud - FUCS, Bogotá, Colombia
| | - Sandra Quijano
- Grupo de Inmunobiología y Biología Celular, Departamento de Microbiología, Pontificia Universidad Javeriana, Bogotá, Colombia
| | - Jovanny Zabaleta
- Department of Interdisciplinary Oncology, Louisiana State University Health Sciences Center, New Orleans, LA, United States
| | - Alba Lucia Combita
- Grupo de Investigación en Biología del Cáncer - Instituto Nacional de Cancerología, Bogotá, Colombia
- Grupo de Investigación Traslacional en Oncología - Instituto Nacional de Cancerología, Bogotá, Colombia
- Maestría en Inmunología, Departamento de Microbiología - Universidad Nacional de Colombia, Bogotá, Colombia
| |
Collapse
|
5
|
Nakajima T, Kanno T, Ueda Y, Miyako K, Endo T, Yoshida S, Yokoyama S, Asou HK, Yamada K, Ikeda K, Togashi Y, Endo Y. Fatty acid metabolism constrains Th9 cell differentiation and antitumor immunity via the modulation of retinoic acid receptor signaling. Cell Mol Immunol 2024; 21:1266-1281. [PMID: 39187636 PMCID: PMC11528006 DOI: 10.1038/s41423-024-01209-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] [Received: 01/12/2024] [Accepted: 08/05/2024] [Indexed: 08/28/2024] Open
Abstract
T helper 9 (Th9) cells are interleukin 9 (IL-9)-producing cells that have diverse functions ranging from antitumor immune responses to allergic inflammation. Th9 cells differentiate from naïve CD4+ T cells in the presence of IL-4 and transforming growth factor-beta (TGF-β); however, our understanding of the molecular basis of their differentiation remains incomplete. Previously, we reported that the differentiation of another subset of TGF-β-driven T helper cells, Th17 cells, is highly dependent on de novo lipid biosynthesis. On the basis of these findings, we hypothesized that lipid metabolism may also be important for Th9 cell differentiation. We therefore investigated the differentiation and function of mouse and human Th9 cells in vitro under conditions of pharmacologically or genetically induced deficiency of the intracellular fatty acid content and in vivo in mice genetically deficient in acetyl-CoA carboxylase 1 (ACC1), an important enzyme for fatty acid biosynthesis. Both the inhibition of de novo fatty acid biosynthesis and the deprivation of environmental lipids augmented differentiation and IL-9 production in mouse and human Th9 cells. Mechanistic studies revealed that the increase in Th9 cell differentiation was mediated by the retinoic acid receptor and the TGF-β-SMAD signaling pathways. Upon adoptive transfer, ACC1-inhibited Th9 cells suppressed tumor growth in murine models of melanoma and adenocarcinoma. Together, our findings highlight a novel role of fatty acid metabolism in controlling the differentiation and in vivo functions of Th9 cells.
Collapse
Affiliation(s)
- Takahiro Nakajima
- Department of Frontier Research and Development, Laboratory of Medical Omics Research, Kazusa DNA Research Institute, 2-6-7 Kazusa Kamatari, Kisarazu, Chiba, 292-0818, Japan
| | - Toshio Kanno
- Department of Frontier Research and Development, Laboratory of Medical Omics Research, Kazusa DNA Research Institute, 2-6-7 Kazusa Kamatari, Kisarazu, Chiba, 292-0818, Japan
| | - Yuki Ueda
- Department of Tumor Microenvironment, Faculty of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, 2-5-1, Shikata-cho, Kita-ku, Okayama, 700-8558, Japan
| | - Keisuke Miyako
- Department of Frontier Research and Development, Laboratory of Medical Omics Research, Kazusa DNA Research Institute, 2-6-7 Kazusa Kamatari, Kisarazu, Chiba, 292-0818, Japan
- Department of Applied Genomics, Kazusa DNA Research Institute, 2-6-7 Kazusa Kamatari, Kisarazu, Chiba, 292-0818, Japan
| | - Takeru Endo
- Department of Frontier Research and Development, Laboratory of Medical Omics Research, Kazusa DNA Research Institute, 2-6-7 Kazusa Kamatari, Kisarazu, Chiba, 292-0818, Japan
| | - Souta Yoshida
- Department of Frontier Research and Development, Laboratory of Medical Omics Research, Kazusa DNA Research Institute, 2-6-7 Kazusa Kamatari, Kisarazu, Chiba, 292-0818, Japan
| | - Satoru Yokoyama
- Department of Frontier Research and Development, Laboratory of Medical Omics Research, Kazusa DNA Research Institute, 2-6-7 Kazusa Kamatari, Kisarazu, Chiba, 292-0818, Japan
| | - Hikari K Asou
- Department of Frontier Research and Development, Laboratory of Medical Omics Research, Kazusa DNA Research Institute, 2-6-7 Kazusa Kamatari, Kisarazu, Chiba, 292-0818, Japan
| | - Kazuko Yamada
- Department of Frontier Research and Development, Laboratory of Medical Omics Research, Kazusa DNA Research Institute, 2-6-7 Kazusa Kamatari, Kisarazu, Chiba, 292-0818, Japan
| | - Kazutaka Ikeda
- Department of Applied Genomics, Kazusa DNA Research Institute, 2-6-7 Kazusa Kamatari, Kisarazu, Chiba, 292-0818, Japan
| | - Yosuke Togashi
- Department of Tumor Microenvironment, Faculty of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, 2-5-1, Shikata-cho, Kita-ku, Okayama, 700-8558, Japan
- Division of Cell Therapy, Chiba Cancer Center Research Institute, 666-2 Nitona-cho, Chuo-ku, Chiba, 260-8717, Japan
| | - Yusuke Endo
- Department of Frontier Research and Development, Laboratory of Medical Omics Research, Kazusa DNA Research Institute, 2-6-7 Kazusa Kamatari, Kisarazu, Chiba, 292-0818, Japan.
- Department of Omics Medicine, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba, 260-8670, Japan.
| |
Collapse
|
6
|
Lim YJ, Park SA, Wang D, Jin W, Ku WL, Zhang D, Xu J, Patiño LC, Liu N, Chen W, Kazmi R, Zhao K, Zhang YE, Sun L, Chen W. MicroRNA-19b exacerbates systemic sclerosis through promoting Th9 cells. Cell Rep 2024; 43:114565. [PMID: 39083380 PMCID: PMC11440512 DOI: 10.1016/j.celrep.2024.114565] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Revised: 06/19/2024] [Accepted: 07/16/2024] [Indexed: 08/02/2024] Open
Abstract
Systemic sclerosis (SSc) is a chronic autoimmune disease characterized by fibrosis of the skin and multiple vital organs, but the immunological pathogenesis of SSc remains unclear. We show here that miR-19b promotes Th9 cells that exacerbate SSc. Specifically, miR-19b and interleukin (IL)-9 increase in CD4+ T cells in experimental SSc in mice induced with bleomycin. Inhibiting miR-19b reduces Th9 cells and ameliorates the disease. Mechanistically, transforming growth factor beta (TGF-β) plus IL-4 activates pSmad3-Ser213 and TRAF6-K63 ubiquitination by suppressing NLRC3. Activated TRAF6 sequentially promotes TGF-β-activated kinase 1 (TAK1) and nuclear factor κB (NF-κB) p65 phosphorylation, leading to the upregulation of miR-19b. Notably, miR-19b activated Il9 gene expression by directly suppressing atypical E2F family member E2f8. In patients with SSc, higher levels of IL9 and MIR-19B correlate with worse disease progression. Our findings reveal miR-19b as a key factor in Th9 cell-mediated SSc pathogenesis and should have clinical implications for patients with SSc.
Collapse
Affiliation(s)
- Yun-Ji Lim
- Mucosal Immunology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, 30 Convent Drive, Bethesda, MD 20892, USA
| | - Sang-A Park
- Mucosal Immunology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, 30 Convent Drive, Bethesda, MD 20892, USA
| | - Dandan Wang
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210008, China
| | - Wenwen Jin
- Mucosal Immunology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, 30 Convent Drive, Bethesda, MD 20892, USA
| | - Wai Lim Ku
- Systemic Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, 31 Center Drive, Bethesda, MD 20892, USA
| | - Dunfang Zhang
- Mucosal Immunology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, 30 Convent Drive, Bethesda, MD 20892, USA
| | - Junji Xu
- Mucosal Immunology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, 30 Convent Drive, Bethesda, MD 20892, USA
| | - Liliana C Patiño
- Mucosal Immunology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, 30 Convent Drive, Bethesda, MD 20892, USA
| | - Na Liu
- Mucosal Immunology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, 30 Convent Drive, Bethesda, MD 20892, USA
| | - Weiwei Chen
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210008, China
| | - Rida Kazmi
- Mucosal Immunology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, 30 Convent Drive, Bethesda, MD 20892, USA
| | - Keji Zhao
- Systemic Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, 31 Center Drive, Bethesda, MD 20892, USA
| | - Ying E Zhang
- Laboratory of Cellular and Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, 37 Convent Drive, Bethesda, MD 20892, USA
| | - Lingyun Sun
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210008, China.
| | - WanJun Chen
- Mucosal Immunology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, 30 Convent Drive, Bethesda, MD 20892, USA.
| |
Collapse
|
7
|
Khokhar M, Purohit P. The emerging role of T helper 9 (Th9) cells in immunopathophysiology: A comprehensive review of their effects and responsiveness in various disease states. Int Rev Immunol 2024; 43:341-360. [PMID: 38864109 DOI: 10.1080/08830185.2024.2364586] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Revised: 05/10/2024] [Accepted: 05/31/2024] [Indexed: 06/13/2024]
Abstract
Th9 cells, a subset of T-helper cells producing interleukin-9 (IL-9), play a vital role in the adaptive immune response and have diverse effects in different diseases. Regulated by transcription factors like PU.1 and IRF4, and cytokines such as IL-4 and TGF-β, Th9 cells drive tissue inflammation. This review focuses on their emerging role in immunopathophysiology. Th9 cells exhibit immune-mediated cancer cell destruction, showing promise in glioma and cervical cancer treatment. However, their role in breast and lung cancer is intricate, requiring a deeper understanding of pro- and anti-tumor aspects. Th9 cells, along with IL-9, foster T cell and immune cell proliferation, contributing to autoimmune disorders. They are implicated in psoriasis, atopic dermatitis, and infections. In allergic reactions and asthma, Th9 cells fuel pro-inflammatory responses. Targeting Foxo1 may regulate innate and adaptive immune responses, alleviating disease symptoms. This comprehensive review outlines Th9 cells' evolving immunopathophysiological role, emphasizing the necessity for further research to grasp their effects and potential therapeutic applications across diseases.
Collapse
Affiliation(s)
- Manoj Khokhar
- Department of Biochemistry, All India Institute of Medical Sciences (AIIMS), Jodhpur, India
| | - Purvi Purohit
- Department of Biochemistry, All India Institute of Medical Sciences (AIIMS), Jodhpur, India
| |
Collapse
|
8
|
Son A, Baral I, Falduto GH, Schwartz DM. Locus of (IL-9) control: IL9 epigenetic regulation in cellular function and human disease. Exp Mol Med 2024; 56:1331-1339. [PMID: 38825637 PMCID: PMC11263352 DOI: 10.1038/s12276-024-01241-y] [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: 11/16/2023] [Revised: 03/06/2024] [Accepted: 03/07/2024] [Indexed: 06/04/2024] Open
Abstract
Interleukin-9 (IL-9) is a multifunctional cytokine with roles in a broad cross-section of human diseases. Like many cytokines, IL-9 is transcriptionally regulated by a group of noncoding regulatory elements (REs) surrounding the IL9 gene. These REs modulate IL-9 transcription by forming 3D loops that recruit transcriptional machinery. IL-9-promoting transcription factors (TFs) can bind REs to increase locus accessibility and permit chromatin looping, or they can be recruited to already accessible chromatin to promote transcription. Ample mechanistic and genome-wide association studies implicate this interplay between IL-9-modulating TFs and IL9 cis-REs in human physiology, homeostasis, and disease.
Collapse
Affiliation(s)
- Aran Son
- Neuroscience Department, International School for Advanced Studies (SISSA), via Bonomea 265, Trieste, 34136, Italy
| | - Ishita Baral
- Division of Rheumatology and Clinical Immunology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Guido H Falduto
- Division of Rheumatology and Clinical Immunology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Daniella M Schwartz
- Division of Rheumatology and Clinical Immunology, University of Pittsburgh, Pittsburgh, PA, USA.
| |
Collapse
|
9
|
Li H, Bradbury JA, Edin ML, Gruzdev A, Li H, Graves JP, DeGraff LM, Lih FB, Feng C, Wolf ER, Bortner CD, London SJ, Sparks MA, Coffman TM, Zeldin DC. TXA2 attenuates allergic lung inflammation through regulation of Th2, Th9, and Treg differentiation. J Clin Invest 2024; 134:e165689. [PMID: 38483511 PMCID: PMC11060738 DOI: 10.1172/jci165689] [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: 01/03/2023] [Accepted: 03/12/2024] [Indexed: 05/02/2024] Open
Abstract
In lung, thromboxane A2 (TXA2) activates the TP receptor to induce proinflammatory and bronchoconstrictor effects. Thus, TP receptor antagonists and TXA2 synthase inhibitors have been tested as potential asthma therapeutics in humans. Th9 cells play key roles in asthma and regulate the lung immune response to allergens. Herein, we found that TXA2 reduces Th9 cell differentiation during allergic lung inflammation. Th9 cells were decreased approximately 2-fold and airway hyperresponsiveness was attenuated in lungs of allergic mice treated with TXA2. Naive CD4+ T cell differentiation to Th9 cells and IL-9 production were inhibited dose-dependently by TXA2 in vitro. TP receptor-deficient mice had an approximately 2-fold increase in numbers of Th9 cells in lungs in vivo after OVA exposure compared with wild-type mice. Naive CD4+ T cells from TP-deficient mice exhibited increased Th9 cell differentiation and IL-9 production in vitro compared with CD4+ T cells from wild-type mice. TXA2 also suppressed Th2 and enhanced Treg differentiation both in vitro and in vivo. Thus, in contrast to its acute, proinflammatory effects, TXA2 also has longer-lasting immunosuppressive effects that attenuate the Th9 differentiation that drives asthma progression. These findings may explain the paradoxical failure of anti-thromboxane therapies in the treatment of asthma.
Collapse
Affiliation(s)
- Hong Li
- Division of Intramural Research, National Institute of Environmental Health Sciences/NIH, Research Triangle Park, North Carolina, USA
| | - J. Alyce Bradbury
- Division of Intramural Research, National Institute of Environmental Health Sciences/NIH, Research Triangle Park, North Carolina, USA
| | - Matthew L. Edin
- Division of Intramural Research, National Institute of Environmental Health Sciences/NIH, Research Triangle Park, North Carolina, USA
| | - Artiom Gruzdev
- Division of Intramural Research, National Institute of Environmental Health Sciences/NIH, Research Triangle Park, North Carolina, USA
| | - Huiling Li
- Division of Intramural Research, National Institute of Environmental Health Sciences/NIH, Research Triangle Park, North Carolina, USA
| | - Joan P. Graves
- Division of Intramural Research, National Institute of Environmental Health Sciences/NIH, Research Triangle Park, North Carolina, USA
| | - Laura M. DeGraff
- Division of Intramural Research, National Institute of Environmental Health Sciences/NIH, Research Triangle Park, North Carolina, USA
| | - Fred B. Lih
- Division of Intramural Research, National Institute of Environmental Health Sciences/NIH, Research Triangle Park, North Carolina, USA
| | - Chiguang Feng
- Division of Intramural Research, National Institute of Environmental Health Sciences/NIH, Research Triangle Park, North Carolina, USA
| | - Erin R. Wolf
- Department of Nephrology, Duke University Medical Center, Durham, North Carolina, USA
| | - Carl D. Bortner
- Division of Intramural Research, National Institute of Environmental Health Sciences/NIH, Research Triangle Park, North Carolina, USA
| | - Stephanie J. London
- Division of Intramural Research, National Institute of Environmental Health Sciences/NIH, Research Triangle Park, North Carolina, USA
| | - Matthew A. Sparks
- Department of Nephrology, Duke University Medical Center, Durham, North Carolina, USA
| | - Thomas M. Coffman
- Department of Nephrology, Duke University Medical Center, Durham, North Carolina, USA
- Program in Cardiovascular and Metabolic Disorders, Duke-NUS Medical School, Singapore
| | - Darryl C. Zeldin
- Division of Intramural Research, National Institute of Environmental Health Sciences/NIH, Research Triangle Park, North Carolina, USA
| |
Collapse
|
10
|
Lee WH, Hong KJ, Li H, Lee GR. Transcription Factor Id1 Plays an Essential Role in Th9 Cell Differentiation by Inhibiting Tcf3 and Tcf4. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2305527. [PMID: 37867222 PMCID: PMC10724384 DOI: 10.1002/advs.202305527] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Revised: 09/08/2023] [Indexed: 10/24/2023]
Abstract
T helper type 9 (Th9) cells play important roles in immune responses by producing interleukin-9 (IL-9). Several transcription factors are responsible for Th9 cell differentiation; however, transcriptional regulation of Th9 cells is not fully understood. Here, it is shown that Id1 is an essential transcriptional regulator of Th9 cell differentiation. Id1 is induced by IL-4 and TGF-β. Id1-deficient naïve CD4 T cells fail to differentiate into Th9 cells, and overexpression of Id1 induce expression of IL-9. Mass spectrometry analysis reveals that Id1 interacts with Tcf3 and Tcf4 in Th9 cells. In addition, RNA-sequencing, chromatin immunoprecipitation, and transient reporter assay reveal that Tcf3 and Tcf4 bind to the promoter region of the Il9 gene to suppress its expression, and that Id1 inhibits their function, leading to Th9 differentiation. Finally, Id1-deficient Th9 cells ameliorate airway inflammation in an animal model of asthma. Thus, Id1 is a transcription factor that plays an essential role in Th9 cell differentiation by inhibiting Tcf3 and Tcf4.
Collapse
Affiliation(s)
- Woo Ho Lee
- Department of Life ScienceSogang University35 Baekbeom‐roMapo‐guSeoul04107South Korea
| | - Kyung Jin Hong
- Department of Life ScienceSogang University35 Baekbeom‐roMapo‐guSeoul04107South Korea
| | - Hua‐Bing Li
- Shanghai Institute of Immunology, State Key Laboratory of Oncogenes and Related GenesShanghai Jiao Tong University School of Medicine280 Chongqing South Rd, Building #5‐602Shanghai200025China
| | - Gap Ryol Lee
- Department of Life ScienceSogang University35 Baekbeom‐roMapo‐guSeoul04107South Korea
| |
Collapse
|
11
|
Laudisi F, Stolfi C, Monteleone I, Monteleone G. TGF-β1 signaling and Smad7 control T-cell responses in health and immune-mediated disorders. Eur J Immunol 2023; 53:e2350460. [PMID: 37611637 DOI: 10.1002/eji.202350460] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 07/14/2023] [Accepted: 08/22/2023] [Indexed: 08/25/2023]
Abstract
Transforming growth factor (TGF)-β1, a member of the TGF-β superfamily, is produced by many immune and nonimmune cells and has pleiotropic effects on both innate and adaptive immunity, especially in the control of T-cell differentiation and function. Consistently, loss of TGF-β1 function is associated with exacerbated T-cell-dependent inflammatory responses that culminate in pathological processes in allergic and immune-mediated diseases. In this review, we highlight the roles of TGF-β1 in immunity, focusing mainly on its ability to promote differentiation of regulatory T cells, T helper (Th)-17, and Th9 cells, thus contributing to amplifying or restricting T-cell responses in health and human diseases (e.g., inflammatory bowel diseases, type 1 diabetes, asthma, and MS). In addition, we discuss the involvement of Smad7, an inhibitor of TGF-β1 signaling, in immune-mediated disorders (e.g., psoriasis, rheumatoid arthritis, MS, and inflammatory bowel diseases), as well as the discordant results of clinical trials with mongersen, an oral pharmaceutical compound containing a Smad7 antisense oligonucleotide, in patients with Crohn's disease. Further work is needed to ascertain the reasons for such a discrepancy as well as to identify better candidates for treatment with Smad7 inhibitors.
Collapse
Affiliation(s)
- Federica Laudisi
- Department of Systems Medicine, University of Rome "Tor Vergata", Rome, Italy
| | - Carmine Stolfi
- Department of Systems Medicine, University of Rome "Tor Vergata", Rome, Italy
| | - Ivan Monteleone
- Department of Biomedicine and Prevention, University of Rome "Tor Vergata", Rome, Italy
| | - Giovanni Monteleone
- Department of Systems Medicine, University of Rome "Tor Vergata", Rome, Italy
- Gastroenterology Unit, Azienda Ospedaliera Policlinico Tor Vergata, Rome, Italy
| |
Collapse
|
12
|
Cannon A, Pajulas A, Kaplan MH, Zhang J. The Dichotomy of Interleukin-9 Function in the Tumor Microenvironment. J Interferon Cytokine Res 2023; 43:229-245. [PMID: 37319357 PMCID: PMC10282829 DOI: 10.1089/jir.2023.0035] [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: 03/13/2023] [Accepted: 04/25/2023] [Indexed: 06/17/2023] Open
Abstract
Interleukin 9 (IL-9) is a cytokine with potent proinflammatory properties that plays a central role in pathologies such as allergic asthma, immunity to parasitic infection, and autoimmunity. More recently, IL-9 has garnered considerable attention in tumor immunity. Historically, IL-9 has been associated with a protumor function in hematological malignancies and an antitumor function in solid malignancies. However, recent discoveries of the dynamic role of IL-9 in cancer progression suggest that IL-9 can act as both a pro- or antitumor factor in various hematological and solid malignancies. This review summarizes IL-9-dependent control of tumor growth, regulation, and therapeutic applicability of IL-9 blockade and IL-9-producing cells in cancer.
Collapse
Affiliation(s)
- Anthony Cannon
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Abigail Pajulas
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Mark H. Kaplan
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, Indiana, USA
- Brown Center for Immunotherapy, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Jilu Zhang
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| |
Collapse
|
13
|
Son A, Meylan F, Gomez-Rodriguez J, Kaul Z, Sylvester M, Falduto GH, Vazquez E, Haque T, Kitakule MM, Wang C, Manthiram K, Qi CF, Cheng J, Gurram RK, Zhu J, Schwartzberg P, Milner JD, Frischmeyer-Guerrerio PA, Schwartz DM. Dynamic chromatin accessibility licenses STAT5- and STAT6-dependent innate-like function of T H9 cells to promote allergic inflammation. Nat Immunol 2023; 24:1036-1048. [PMID: 37106040 PMCID: PMC10247433 DOI: 10.1038/s41590-023-01501-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Accepted: 03/27/2023] [Indexed: 04/29/2023]
Abstract
Allergic diseases are a major global health issue. Interleukin (IL)-9-producing helper T (TH9) cells promote allergic inflammation, yet TH9 cell effector functions are incompletely understood because their lineage instability makes them challenging to study. Here we found that resting TH9 cells produced IL-9 independently of T cell receptor (TCR) restimulation, due to STAT5- and STAT6-dependent bystander activation. This mechanism was seen in circulating cells from allergic patients and was restricted to recently activated cells. STAT5-dependent Il9/IL9 regulatory elements underwent remodeling over time, inactivating the locus. A broader 'allergic TH9' transcriptomic and epigenomic program was also unstable. In vivo, TH9 cells induced airway inflammation via TCR-independent, STAT-dependent mechanisms. In allergic patients, TH9 cell expansion was associated with responsiveness to JAK inhibitors. These findings suggest that TH9 cell instability is a negative checkpoint on bystander activation that breaks down in allergy and that JAK inhibitors should be considered for allergic patients with TH9 cell expansion.
Collapse
Affiliation(s)
- Aran Son
- Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Francoise Meylan
- Office of Science and Technology, National Institute of Arthritis, Musculoskeletal, and Skin Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Julio Gomez-Rodriguez
- Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
- TCR Therapeutics, Cambridge, MA, USA
| | - Zenia Kaul
- Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - McKella Sylvester
- Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Guido H Falduto
- Division of Rheumatology and Clinical Immunology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Estefania Vazquez
- Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Tamara Haque
- Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Moses M Kitakule
- Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
- Division of Pediatric Allergy Immunology and Rheumatology, Department of Pediatrics, Columbia University Medical Center, New York, NY, USA
| | - Chujun Wang
- Division of Rheumatology and Clinical Immunology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Kalpana Manthiram
- Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Chen-Feng Qi
- Pathology Core, Laboratory of Immunogenetics, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Jun Cheng
- Embryonic Stem Cell and Transgenic Core, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | - Rama K Gurram
- Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Jinfang Zhu
- Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Pamela Schwartzberg
- Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Joshua D Milner
- Division of Pediatric Allergy Immunology and Rheumatology, Department of Pediatrics, Columbia University Medical Center, New York, NY, USA
| | - Pamela A Frischmeyer-Guerrerio
- Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Daniella M Schwartz
- Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA.
- Division of Rheumatology and Clinical Immunology, University of Pittsburgh, Pittsburgh, PA, USA.
| |
Collapse
|
14
|
Qiaolongbatu X, Zhao W, Huang X, Qian F, Yang X, Wu J, Ma C, Qu H, Wang L, Fan G, Wu Z. The Therapeutic Mechanism of Schisandrol A and Its Metabolites on Pulmonary Fibrosis Based on Plasma Metabonomics and Network Analysis. Drug Des Devel Ther 2023; 17:477-496. [PMID: 36814892 PMCID: PMC9939797 DOI: 10.2147/dddt.s391503] [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: 10/20/2022] [Accepted: 01/25/2023] [Indexed: 02/16/2023] Open
Abstract
Background Schisandrol A (Sch A) is the main active ingredient of Schisandra chinensis (Turcz.) Baill. Our previous study showed that Sch A has anti-pulmonary fibrosis (PF) activity, but its metabolic-related mechanisms of action are not clear. Methods Here, we explored the therapeutic mechanisms of Sch A on PF by ultra-high performance liquid chromatography coupled with tandem mass spectrometry (UPLC-MS/MS) metabolomics approach and network analysis. The metabolites of Sch A in mice (bleomycin + Sch A high-dose group) plasma were identified based on ultra-high performance liquid chromatography-quadrupole time-of-flight mass spectrometry (UPLC-Q-TOF/MS). Results 32 metabolites were detected reversed to normal level after treating bleomycin (BLM)-induced PF mice with Sch A. The 32 biomarkers were enriched in energy metabolism and several amino acid metabolisms, which was the first report on the therapeutic effects of Sch A on PF through rescuing the disordered energy metabolism. The UPLC-Q-TOF/MS analysis identified 17 possible metabolites (including isomers) of Sch A in mice plasma. Network analysis revealed that Sch A and 17 metabolites were related to 269 genes, and 1109 disease genes were related to PF. The construction of the Sch A/metabolites-target-PF network identified a total of 79 intersection genes and the TGF-β signaling pathway was determined to be the main signaling pathway related to the treatment of PF by Sch A. The integrated approach involving metabolomics and network analysis revealed that the TGF-β1-ID3-creatine pathway, TGF-β1-VIM-carnosine pathway were two of the possible pathways Sch A regulated to modulate metabolic disorders, especially energy metabolism, and the metabolite of Sch A M5 was identified as a most likely active metabolite. Conclusion The results suggested the feasibility of combining metabolomics and network analysis to reflect the effects of Sch A on the biological network and the metabolic state of PF and to evaluate the drug efficacy of Sch A and its related mechanisms.
Collapse
Affiliation(s)
- Xijier Qiaolongbatu
- Engineering Research Center of Cell & Therapeutic Antibody, Ministry of Education, Pharm-X Center, School of Pharmacy, Shanghai Jiao Tong University, Shanghai, People’s Republic of China
| | - Wenjuan Zhao
- Engineering Research Center of Cell & Therapeutic Antibody, Ministry of Education, Pharm-X Center, School of Pharmacy, Shanghai Jiao Tong University, Shanghai, People’s Republic of China
| | - Xucong Huang
- Department of Clinical Pharmacy, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, People’s Republic of China,School of Pharmacy, Chongqing Medical University, Chongqing, People’s Republic of China
| | - Feng Qian
- Engineering Research Center of Cell & Therapeutic Antibody, Ministry of Education, Pharm-X Center, School of Pharmacy, Shanghai Jiao Tong University, Shanghai, People’s Republic of China
| | - Xinyi Yang
- Engineering Research Center of Cell & Therapeutic Antibody, Ministry of Education, Pharm-X Center, School of Pharmacy, Shanghai Jiao Tong University, Shanghai, People’s Republic of China
| | - Jiaqi Wu
- Department of Clinical Pharmacy, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, People’s Republic of China
| | - Cui Ma
- Department of Clinical Pharmacy, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, People’s Republic of China
| | - Han Qu
- Engineering Research Center of Cell & Therapeutic Antibody, Ministry of Education, Pharm-X Center, School of Pharmacy, Shanghai Jiao Tong University, Shanghai, People’s Republic of China,Department of Clinical Pharmacy, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, People’s Republic of China
| | - Li Wang
- School of Pharmacy, Chongqing Medical University, Chongqing, People’s Republic of China
| | - Guorong Fan
- Department of Clinical Pharmacy, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, People’s Republic of China
| | - Zhenghua Wu
- Department of Clinical Pharmacy, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, People’s Republic of China,Correspondence: Zhenghua Wu; Guorong Fan, Department of Clinical Pharmacy, Shanghai General Hospital, School of Medicine, Shanghai Jiaotong University, No. 85 Wujin Road, Shanghai, 200080, People’s Republic of China, Tel +86-133-0177-7863; +86-21-36123711, Email ;
| |
Collapse
|
15
|
CD4+IL9+ (Th9) cells as the major source of IL-9, potentially modulate Th17/Treg mediated host immune response during experimental cerebral malaria. Mol Immunol 2022; 152:240-254. [DOI: 10.1016/j.molimm.2022.11.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Revised: 10/18/2022] [Accepted: 11/07/2022] [Indexed: 11/15/2022]
|
16
|
Park SA, Lim YJ, Ku WL, Zhang D, Cui K, Tang LY, Chia C, Zanvit P, Chen Z, Jin W, Wang D, Xu J, Liu O, Wang F, Cain A, Guo N, Nakatsukasa H, Wu C, Zhang YE, Zhao K, Chen W. Opposing functions of circadian protein DBP and atypical E2F family E2F8 in anti-tumor Th9 cell differentiation. Nat Commun 2022; 13:6069. [PMID: 36241625 PMCID: PMC9568563 DOI: 10.1038/s41467-022-33733-8] [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: 10/08/2021] [Accepted: 09/30/2022] [Indexed: 12/24/2022] Open
Abstract
Interleukin-9 (IL-9)-producing CD4+ T helper cells (Th9) have been implicated in allergy/asthma and anti-tumor immunity, yet molecular insights on their differentiation from activated T cells, driven by IL-4 and transforming growth factor-beta (TGF-β), is still lacking. Here we show opposing functions of two transcription factors, D-binding protein (DBP) and E2F8, in controlling Th9 differentiation. Specifically, TGF-β and IL-4 signaling induces phosphorylation of the serine 213 site in the linker region of the Smad3 (pSmad3L-Ser213) via phosphorylated p38, which is necessary and sufficient for Il9 gene transcription. We identify DBP and E2F8 as an activator and repressor, respectively, for Il9 transcription by pSmad3L-Ser213. Notably, Th9 cells with siRNA-mediated knockdown for Dbp or E2f8 promote and suppress tumor growth, respectively, in mouse tumor models. Importantly, DBP and E2F8 also exhibit opposing functions in regulating human TH9 differentiation in vitro. Thus, our data uncover a molecular mechanism of Smad3 linker region-mediated, opposing functions of DBP and E2F8 in Th9 differentiation.
Collapse
Affiliation(s)
- Sang-A Park
- grid.94365.3d0000 0001 2297 5165Mucosal Immunology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, 30 Convent Drive, Bethesda, 20892 MD USA
| | - Yun-Ji Lim
- grid.94365.3d0000 0001 2297 5165Mucosal Immunology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, 30 Convent Drive, Bethesda, 20892 MD USA
| | - Wai Lim Ku
- grid.94365.3d0000 0001 2297 5165Systemic Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, 31 Center Drive, Bethesda, 20892 MD USA
| | - Dunfang Zhang
- grid.94365.3d0000 0001 2297 5165Mucosal Immunology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, 30 Convent Drive, Bethesda, 20892 MD USA
| | - Kairong Cui
- grid.94365.3d0000 0001 2297 5165Systemic Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, 31 Center Drive, Bethesda, 20892 MD USA
| | - Liu-Ya Tang
- grid.94365.3d0000 0001 2297 5165Laboratory of Cellular and Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, 37 Convent Drive, Bethesda, 20892 MD USA
| | - Cheryl Chia
- grid.94365.3d0000 0001 2297 5165Mucosal Immunology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, 30 Convent Drive, Bethesda, 20892 MD USA
| | - Peter Zanvit
- grid.94365.3d0000 0001 2297 5165Mucosal Immunology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, 30 Convent Drive, Bethesda, 20892 MD USA
| | - Zuojia Chen
- grid.94365.3d0000 0001 2297 5165Experimental Immunology Branch, National Cancer Institute, National Institutes of Health, 37 Convent Drive, Bethesda, 20892 MD USA
| | - Wenwen Jin
- grid.94365.3d0000 0001 2297 5165Mucosal Immunology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, 30 Convent Drive, Bethesda, 20892 MD USA
| | - Dandan Wang
- grid.94365.3d0000 0001 2297 5165Mucosal Immunology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, 30 Convent Drive, Bethesda, 20892 MD USA
| | - Junji Xu
- grid.94365.3d0000 0001 2297 5165Mucosal Immunology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, 30 Convent Drive, Bethesda, 20892 MD USA
| | - Ousheng Liu
- grid.94365.3d0000 0001 2297 5165Mucosal Immunology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, 30 Convent Drive, Bethesda, 20892 MD USA
| | - Fu Wang
- grid.94365.3d0000 0001 2297 5165Mucosal Immunology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, 30 Convent Drive, Bethesda, 20892 MD USA
| | - Alexander Cain
- grid.94365.3d0000 0001 2297 5165Mucosal Immunology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, 30 Convent Drive, Bethesda, 20892 MD USA
| | - Nancy Guo
- grid.94365.3d0000 0001 2297 5165Mucosal Immunology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, 30 Convent Drive, Bethesda, 20892 MD USA
| | - Hiroko Nakatsukasa
- grid.94365.3d0000 0001 2297 5165Mucosal Immunology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, 30 Convent Drive, Bethesda, 20892 MD USA
| | - Chuan Wu
- grid.94365.3d0000 0001 2297 5165Experimental Immunology Branch, National Cancer Institute, National Institutes of Health, 37 Convent Drive, Bethesda, 20892 MD USA
| | - Ying E. Zhang
- grid.94365.3d0000 0001 2297 5165Laboratory of Cellular and Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, 37 Convent Drive, Bethesda, 20892 MD USA
| | - Keji Zhao
- grid.94365.3d0000 0001 2297 5165Systemic Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, 31 Center Drive, Bethesda, 20892 MD USA
| | - WanJun Chen
- grid.94365.3d0000 0001 2297 5165Mucosal Immunology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, 30 Convent Drive, Bethesda, 20892 MD USA
| |
Collapse
|
17
|
Anderson MK. Shifting gears: Id3 enables recruitment of E proteins to new targets during T cell development and differentiation. Front Immunol 2022; 13:956156. [PMID: 35983064 PMCID: PMC9378783 DOI: 10.3389/fimmu.2022.956156] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2022] [Accepted: 07/08/2022] [Indexed: 11/13/2022] Open
Abstract
Shifting levels of E proteins and Id factors are pivotal in T cell commitment and differentiation, both in the thymus and in the periphery. Id2 and Id3 are two different factors that prevent E proteins from binding to their target gene cis-regulatory sequences and inducing gene expression. Although they use the same mechanism to suppress E protein activity, Id2 and Id3 play very different roles in T cell development and CD4 T cell differentiation. Id2 imposes an irreversible choice in early T cell precursors between innate and adaptive lineages, which can be thought of as a railway switch that directs T cells down one path or another. By contrast, Id3 acts in a transient fashion downstream of extracellular signals such as T cell receptor (TCR) signaling. TCR-dependent Id3 upregulation results in the dislodging of E proteins from their target sites while chromatin remodeling occurs. After the cessation of Id3 expression, E proteins can reassemble in the context of a new genomic landscape and molecular context that allows induction of different E protein target genes. To describe this mode of action, we have developed the “Clutch” model of differentiation. In this model, Id3 upregulation in response to TCR signaling acts as a clutch that stops E protein activity (“clutch in”) long enough to allow shifting of the genomic landscape into a different “gear”, resulting in accessibility to different E protein target genes once Id3 decreases (“clutch out”) and E proteins can form new complexes on the DNA. While TCR signal strength and cytokine signaling play a role in both peripheral and thymic lineage decisions, the remodeling of chromatin and E protein target genes appears to be more heavily influenced by the cytokine milieu in the periphery, whereas the outcome of Id3 activity during T cell development in the thymus appears to depend more on the TCR signal strength. Thus, while the Clutch model applies to both CD4 T cell differentiation and T cell developmental transitions within the thymus, changes in chromatin accessibility are modulated by biased inputs in these different environments. New emerging technologies should enable a better understanding of the molecular events that happen during these transitions, and how they fit into the gene regulatory networks that drive T cell development and differentiation.
Collapse
Affiliation(s)
- Michele K. Anderson
- Department of Biological Sciences, Sunnybrook Research Institute, Toronto, ON, Canada
- Department of Immunology, University of Toronto, Toronto, ON, Canada
- *Correspondence: Michele K. Anderson,
| |
Collapse
|
18
|
Chen B, Mu C, Zhang Z, He X, Liu X. The Love-Hate Relationship Between TGF-β Signaling and the Immune System During Development and Tumorigenesis. Front Immunol 2022; 13:891268. [PMID: 35720407 PMCID: PMC9204485 DOI: 10.3389/fimmu.2022.891268] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Accepted: 04/25/2022] [Indexed: 11/20/2022] Open
Abstract
Since TGF-β was recognized as an essential secreted cytokine in embryogenesis and adult tissue homeostasis a decade ago, our knowledge of the role of TGF-β in mammalian development and disease, particularly cancer, has constantly been updated. Mounting evidence has confirmed that TGF-β is the principal regulator of the immune system, as deprivation of TGF-β signaling completely abrogates adaptive immunity. However, enhancing TGF-β signaling constrains the immune response through multiple mechanisms, including boosting Treg cell differentiation and inducing CD8+ T-cell apoptosis in the disease context. The love-hate relationship between TGF-β signaling and the immune system makes it challenging to develop effective monotherapies targeting TGF-β, especially for cancer treatment. Nonetheless, recent work on combination therapies of TGF-β inhibition and immunotherapy have provide insights into the development of TGF-β-targeted therapies, with favorable outcomes in patients with advanced cancer. Hence, we summarize the entanglement between TGF-β and the immune system in the developmental and tumor contexts and recent progress on hijacking crucial TGF-β signaling pathways as an emerging area of cancer therapy.
Collapse
Affiliation(s)
- Baode Chen
- Department of Laboratory Medicine, Key Laboratory of Clinical In Vitro Diagnostic Techniques of Zhejiang Province, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Chenglin Mu
- Institute for Intelligent Bio/Chem Manufacturing (iBCM), Zhejiang University (ZJU)-Hangzhou Global Scientific and Technological Innovation Center, Hangzhou, China
| | - Zhiwei Zhang
- Institute for Intelligent Bio/Chem Manufacturing (iBCM), Zhejiang University (ZJU)-Hangzhou Global Scientific and Technological Innovation Center, Hangzhou, China
| | - Xuelin He
- Kidney Disease Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Xia Liu
- Institute for Intelligent Bio/Chem Manufacturing (iBCM), Zhejiang University (ZJU)-Hangzhou Global Scientific and Technological Innovation Center, Hangzhou, China
| |
Collapse
|
19
|
Gumberger P, Bjornsson B, Sandström P, Bojmar L, Zambirinis CP. The Liver Pre-Metastatic Niche in Pancreatic Cancer: A Potential Opportunity for Intervention. Cancers (Basel) 2022; 14:3028. [PMID: 35740692 PMCID: PMC9221452 DOI: 10.3390/cancers14123028] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Revised: 05/30/2022] [Accepted: 06/11/2022] [Indexed: 12/16/2022] Open
Abstract
Cancer-related mortality is primarily a consequence of metastatic dissemination and associated complications. Pancreatic ductal adenocarcinoma (PDAC) is one of the most lethal malignancies and tends to metastasize early, especially in the liver. Emerging evidence suggests that organs that develop metastases exhibit microscopic changes that favor metastatic growth, collectively known as "pre-metastatic niches". By definition, a pre-metastatic niche is chronologically established before overt metastatic outgrowth, and its generation involves the release of tumor-derived secreted factors that modulate cells intrinsic to the recipient organ, as well as recruitment of additional cells from tertiary sites, such as bone marrow-all orchestrated by the primary tumor. The pre-metastatic niche is characterized by tumor-promoting inflammation with tumor-supportive and immune-suppressive features, remodeling of the extracellular matrix, angiogenic modulation and metabolic alterations that support growth of disseminated tumor cells. In this paper, we review the current state of knowledge of the hepatic pre-metastatic niche in PDAC and attempt to create a framework to guide future diagnostic and therapeutic studies.
Collapse
Affiliation(s)
- Peter Gumberger
- Department of Surgery, Linköping University, 58183 Linköping, Sweden; (P.G.); (B.B.); (P.S.)
- Department of Biomedical and Clinical Sciences, Linköping University, 58183 Linköping, Sweden;
| | - Bergthor Bjornsson
- Department of Surgery, Linköping University, 58183 Linköping, Sweden; (P.G.); (B.B.); (P.S.)
- Department of Biomedical and Clinical Sciences, Linköping University, 58183 Linköping, Sweden;
| | - Per Sandström
- Department of Surgery, Linköping University, 58183 Linköping, Sweden; (P.G.); (B.B.); (P.S.)
- Department of Biomedical and Clinical Sciences, Linköping University, 58183 Linköping, Sweden;
| | - Linda Bojmar
- Department of Biomedical and Clinical Sciences, Linköping University, 58183 Linköping, Sweden;
- Children’s Cancer and Blood Foundation Laboratories, Departments of Pediatrics, and Cell and Developmental Biology, Drukier Institute for Children’s Health, Meyer Cancer Center, Weill Cornell Medicine, New York, NY 10021, USA
| | | |
Collapse
|
20
|
Tang M, Jia F, Nan F, Zuo F, Yuan Z, Zhang D. Role of Cytokines in Thymic Regulatory T Cell Generation: Overview and Updates. Front Immunol 2022; 13:883560. [PMID: 35432378 PMCID: PMC9008509 DOI: 10.3389/fimmu.2022.883560] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Accepted: 03/11/2022] [Indexed: 02/05/2023] Open
Abstract
CD4+CD25+Foxp3+ Regulatory (Treg) T cells are mainly generated within the thymus. However, the mechanism of thymic Treg cell (tTreg cell) generation remains to be fully revealed. Although the functions of TCR/CD28 co-stimulation have been widely accepted, the functions of cytokines in the generation of tTreg cells remain highly controversial. In this review, we summarize the existing studies on cytokine regulation of tTreg cell generation. By integrating the key findings of cytokines in tTreg cell generation, we have concluded that four members of γc family cytokines (IL-2, IL-4, IL-7 and IL-15), transforming growth factor β (TGF-β), and three members of TNF superfamily cytokines (GITRL, OX40L and TNF-α) play vitally important roles in regulating tTreg cell generation. We also point out all disputed points and highlight critical scientific questions that need to be addressed in the future.
Collapse
Affiliation(s)
- Mei Tang
- Department of Biotherapy, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Fuya Jia
- West China School of Pharmacy, Sichuan University, Chengdu, China
| | - Fang Nan
- Department of Biotherapy, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Fengqiong Zuo
- Department of Immunology, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, China
| | - Zhu Yuan
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Dunfang Zhang
- Department of Biotherapy, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
21
|
Lipp JJ, Wang L, Yang H, Yao F, Harrer N, Müller S, Berezowska S, Dorn P, Marti TM, Schmid RA, Hegedüs B, Souabni A, Carotta S, Pearson MA, Sommergruber W, Kocher GJ, Hall SR. Functional and molecular characterization of PD1+ tumor-infiltrating lymphocytes from lung cancer patients. Oncoimmunology 2022; 11:2019466. [PMID: 35154905 PMCID: PMC8837234 DOI: 10.1080/2162402x.2021.2019466] [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
Antibody-mediated cancer immunotherapy targets inhibitory surface molecules, such as PD1, PD-L1, and CTLA-4, aiming to re-invigorate dysfunctional T cells. We purified and characterized tumor-infiltrating lymphocytes (TILs) and their patient-matched non-tumor counterparts from treatment-naïve NSCLC patient biopsies to evaluate the effect of PD1 expression on the functional and molecular profiles of tumor-resident T cells. We show that PD1+ CD8+ TILs have elevated expression of the transcriptional regulator ID3 and that the cytotoxic potential of CD8 T cells can be improved by knocking down ID3, defining it as a potential regulator of T cell effector function. PD1+ CD4+ memory TILs display transcriptional patterns consistent with both helper and regulator function, but can robustly facilitate B cell activation and expansion. Furthermore, we show that expanding ex vivo-prepared TILs in vitro broadly preserves their functionality with respect to tumor cell killing, B cell help, and TCR repertoire. Although purified PD1+ CD8+ TILs generally maintain an exhausted phenotype upon expansion in vitro, transcriptional analysis reveals a downregulation of markers of T-cell dysfunction, including the co-inhibitory molecules PD1 and CTLA-4 and transcription factors ID3, TOX and TOX2, while genes involved in cell cycle and DNA repair are upregulated. We find reduced expression of WNT signaling components to be a hallmark of PD1+ CD8+ exhausted T cells in vivo and in vitro and demonstrate that restoring WNT signaling, by pharmacological blockade of GSK3β, can improve effector function. These data unveil novel targets for tumor immunotherapy and have promising implications for the development of a personalized TIL-based cell therapy for lung cancer.
Collapse
Affiliation(s)
- Jesse J. Lipp
- Boehringer Ingelheim, Rcv GmbH & Co Kg, Vienna, Austria
| | - Limei Wang
- Division of General Thoracic Surgery, Bern University Hospital, Bern, Switzerland
- Department of BioMedical Research, University of Bern, Bern, Switzerland
| | - Haitang Yang
- Department of Thoracic Surgery, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, People’s Republic of China
| | - Feng Yao
- Department of Thoracic Surgery, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, People’s Republic of China
| | | | - Stefan Müller
- Department of BioMedical Research, University of Bern, Bern, Switzerland
| | | | - Patrick Dorn
- Division of General Thoracic Surgery, Bern University Hospital, Bern, Switzerland
| | - Thomas M. Marti
- Division of General Thoracic Surgery, Bern University Hospital, Bern, Switzerland
- Department of BioMedical Research, University of Bern, Bern, Switzerland
| | - Ralph A. Schmid
- Division of General Thoracic Surgery, Bern University Hospital, Bern, Switzerland
- Department of BioMedical Research, University of Bern, Bern, Switzerland
| | - Belazs Hegedüs
- Department of Thoracic Surgery, University Medicine Essen, University Duisburg-Essen, Essen, Germany
| | | | | | | | - Wolfgang Sommergruber
- Boehringer Ingelheim, Rcv GmbH & Co Kg, Vienna, Austria
- Department of Biotechnology, University of Applied Sciences, Vienna, Austria
| | - Greg J. Kocher
- Division of General Thoracic Surgery, Bern University Hospital, Bern, Switzerland
| | - Sean R.R. Hall
- Division of General Thoracic Surgery, Bern University Hospital, Bern, Switzerland
- Department of BioMedical Research, University of Bern, Bern, Switzerland
| |
Collapse
|
22
|
Abstract
The cytokine, transforming growth factor beta (TGF-β), has a history of more than 40 years. TGF-β is secreted by many tumor cells and is associated with tumor growth and cancer immunity. The canonical TGF-β signaling pathway, SMAD, controls both tumor metastasis and immune regulation, thereby regulating cancer immunity. TGF-β regulates multiple types of immune cells in tumor microenvironment, including T cells, natural killer (NK) cells, and macrophages. One of the main roles of TGF-β in the tumor microenvironment is the generation of regulatory T cells, which contribute to the suppression of anti-tumor immunity. Because cancer is one of the highest causes of death globally, the discovery of immune checkpoint inhibitors by Honjo and Allison in cancer immunotherapy earned a Nobel Prize in 2018. TGF-β also regulates the levels of immune checkpoints inhibitory receptors on immune cells. Immune checkpoints inhibitors are now being developed along with anti-TGF-β antibody and/or TGF-β inhibitors. More recently, chimeric antigen receptors (CARs) were applied to cancer immunity and tried to combine with TGF-β blockers.
Collapse
Affiliation(s)
| | - WanJun Chen
- Mucosal Immunology Section, NIDCR, National Institute of Health
| | - Hiroyuki Shibata
- Department of Clinical Oncology, Akita University Graduate School of Medicine
| |
Collapse
|
23
|
Musiol S, Alessandrini F, Jakwerth CA, Chaker AM, Schneider E, Guerth F, Schnautz B, Grosch J, Ghiordanescu I, Ullmann JT, Kau J, Plaschke M, Haak S, Buch T, Schmidt-Weber CB, Zissler UM. TGF-β1 Drives Inflammatory Th Cell But Not Treg Cell Compartment Upon Allergen Exposure. Front Immunol 2022; 12:763243. [PMID: 35069535 PMCID: PMC8777012 DOI: 10.3389/fimmu.2021.763243] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Accepted: 11/29/2021] [Indexed: 12/22/2022] Open
Abstract
TGF-β1 is known to have a pro-inflammatory impact by inducing Th9 and Th17 cells, while it also induces anti-inflammatory Treg cells (Tregs). In the context of allergic airway inflammation (AAI) its dual role can be of critical importance in influencing the outcome of the disease. Here we demonstrate that TGF-β is a major player in AAI by driving effector T cells, while Tregs differentiate independently. Induction of experimental AAI and airway hyperreactivity in a mouse model with inducible genetic ablation of the gene encoding for TGFβ-receptor 2 (Tgfbr2) on CD4+T cells significantly reduced the disease phenotype. Further, it blocked the induction of pro-inflammatory T cell frequencies (Th2, Th9, Th17), but increased Treg cells. To translate these findings into a human clinically relevant context, Th2, Th9 and Treg cells were quantified both locally in induced sputum and systemically in blood of allergic rhinitis and asthma patients with or without allergen-specific immunotherapy (AIT). Natural allergen exposure induced local and systemic Th2, Th9, and reduced Tregs cells, while therapeutic allergen exposure by AIT suppressed Th2 and Th9 cell frequencies along with TGF-β and IL-9 secretion. Altogether, these findings support that neutralization of TGF-β represents a viable therapeutic option in allergy and asthma, not posing the risk of immune dysregulation by impacting Tregs cells.
Collapse
Affiliation(s)
- Stephanie Musiol
- Center of Allergy & Environment (ZAUM), Technical University of Munich (TUM) and Helmholtz Center Munich, German Research Center for Environmental Health, Members of the German Center of Lung Research (DZL), Munich, Germany
| | - Francesca Alessandrini
- Center of Allergy & Environment (ZAUM), Technical University of Munich (TUM) and Helmholtz Center Munich, German Research Center for Environmental Health, Members of the German Center of Lung Research (DZL), Munich, Germany
| | - Constanze A Jakwerth
- Center of Allergy & Environment (ZAUM), Technical University of Munich (TUM) and Helmholtz Center Munich, German Research Center for Environmental Health, Members of the German Center of Lung Research (DZL), Munich, Germany
| | - Adam M Chaker
- Center of Allergy & Environment (ZAUM), Technical University of Munich (TUM) and Helmholtz Center Munich, German Research Center for Environmental Health, Members of the German Center of Lung Research (DZL), Munich, Germany.,Department of Otorhinolaryngology, Klinikum rechts der Isar, TUM School of Medicine, Technical University Munich, Munich, Germany
| | - Evelyn Schneider
- Center of Allergy & Environment (ZAUM), Technical University of Munich (TUM) and Helmholtz Center Munich, German Research Center for Environmental Health, Members of the German Center of Lung Research (DZL), Munich, Germany
| | - Ferdinand Guerth
- Center of Allergy & Environment (ZAUM), Technical University of Munich (TUM) and Helmholtz Center Munich, German Research Center for Environmental Health, Members of the German Center of Lung Research (DZL), Munich, Germany
| | - Benjamin Schnautz
- Center of Allergy & Environment (ZAUM), Technical University of Munich (TUM) and Helmholtz Center Munich, German Research Center for Environmental Health, Members of the German Center of Lung Research (DZL), Munich, Germany
| | - Johanna Grosch
- Center of Allergy & Environment (ZAUM), Technical University of Munich (TUM) and Helmholtz Center Munich, German Research Center for Environmental Health, Members of the German Center of Lung Research (DZL), Munich, Germany
| | - Ileana Ghiordanescu
- Center of Allergy & Environment (ZAUM), Technical University of Munich (TUM) and Helmholtz Center Munich, German Research Center for Environmental Health, Members of the German Center of Lung Research (DZL), Munich, Germany
| | - Julia T Ullmann
- Center of Allergy & Environment (ZAUM), Technical University of Munich (TUM) and Helmholtz Center Munich, German Research Center for Environmental Health, Members of the German Center of Lung Research (DZL), Munich, Germany
| | - Josephine Kau
- Center of Allergy & Environment (ZAUM), Technical University of Munich (TUM) and Helmholtz Center Munich, German Research Center for Environmental Health, Members of the German Center of Lung Research (DZL), Munich, Germany
| | - Mirjam Plaschke
- Center of Allergy & Environment (ZAUM), Technical University of Munich (TUM) and Helmholtz Center Munich, German Research Center for Environmental Health, Members of the German Center of Lung Research (DZL), Munich, Germany
| | - Stefan Haak
- Institute of Laboratory Animal Science, University of Zurich, Zurich, Switzerland
| | - Thorsten Buch
- Institute of Laboratory Animal Science, University of Zurich, Zurich, Switzerland
| | - Carsten B Schmidt-Weber
- Center of Allergy & Environment (ZAUM), Technical University of Munich (TUM) and Helmholtz Center Munich, German Research Center for Environmental Health, Members of the German Center of Lung Research (DZL), Munich, Germany
| | - Ulrich M Zissler
- Center of Allergy & Environment (ZAUM), Technical University of Munich (TUM) and Helmholtz Center Munich, German Research Center for Environmental Health, Members of the German Center of Lung Research (DZL), Munich, Germany
| |
Collapse
|
24
|
Pei S, Huang M, Huang J, Zhu X, Wang H, Romano S, Deng X, Wang Y, Luo Y, Hao S, Xu J, Yu T, Zhu Q, Yuan J, Shen K, Liu Z, Hu G, Peng C, Luo Q, Wen Z, Dai D, Xiao Y. BFAR coordinates TGFβ signaling to modulate Th9-mediated cancer immunotherapy. J Exp Med 2021; 218:212036. [PMID: 33914044 PMCID: PMC8091105 DOI: 10.1084/jem.20202144] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Revised: 01/25/2021] [Accepted: 03/04/2021] [Indexed: 01/05/2023] Open
Abstract
TGFβ is essential for the generation of anti-tumor Th9 cells; on the other hand, it causes resistance against anti-tumor immunity. Despite recent progress, the underlying mechanism reconciling the double-edged effect of TGFβ signaling in Th9-mediated cancer immunotherapy remains elusive. Here, we find that TGFβ-induced down-regulation of bifunctional apoptosis regulator (BFAR) represents the key mechanism preventing the sustained activation of TGFβ signaling and thus impairing Th9 inducibility. Mechanistically, BFAR mediates K63-linked ubiquitination of TGFβR1 at K268, which is critical to activate TGFβ signaling. Thus, BFAR deficiency or K268R knock-in mutation suppresses TGFβR1 ubiquitination and Th9 differentiation, thereby inhibiting Th9-mediated cancer immunotherapy. More interestingly, BFAR-overexpressed Th9 cells exhibit promising therapeutic efficacy to curtail tumor growth and metastasis and promote the sensitivity of anti–PD-1–mediated checkpoint immunotherapy. Thus, our findings establish BFAR as a key TGFβ-regulated gene to fine-tune TGFβ signaling that causes Th9 induction insensitivity, and they highlight the translational potential of BFAR in promoting Th9-mediated cancer immunotherapy.
Collapse
Affiliation(s)
- Siyu Pei
- Chinese Academy of Sciences Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Mingzhu Huang
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Jia Huang
- Department of Thoracic Surgical Oncology, Shanghai Lung Cancer Center, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaodong Zhu
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Hui Wang
- Department of Thoracic Surgical Oncology, Shanghai Lung Cancer Center, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Simona Romano
- Department of Molecular Medicine and Medical Biotechnology, University of Naples, Federico II, Naples, Italy
| | - Xiuyu Deng
- Chinese Academy of Sciences Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Yan Wang
- Chinese Academy of Sciences Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Yixiao Luo
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Shumeng Hao
- Chinese Academy of Sciences Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Jing Xu
- Chinese Academy of Sciences Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Tao Yu
- Chinese Academy of Sciences Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Qingchen Zhu
- Chinese Academy of Sciences Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Jia Yuan
- Chinese Academy of Sciences Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Kunwei Shen
- Comprehensive Breast Health Center, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Zhiqiang Liu
- Tianjin Key Laboratory of Cellular Homeostasis and Human Diseases, School of Basic Medical Science, Tianjin Medical University, Tianjin, China
| | - Guohong Hu
- Chinese Academy of Sciences Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Chao Peng
- National Facility for Protein Science in Shanghai, Zhangjiang Lab, Shanghai, China
| | - Qingquan Luo
- Department of Thoracic Surgical Oncology, Shanghai Lung Cancer Center, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhenzhen Wen
- Department of Gastroenterology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Dongfang Dai
- The Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, Nanjing, China
| | - Yichuan Xiao
- Chinese Academy of Sciences Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| |
Collapse
|
25
|
Abstract
CD4 T cell effector subsets not only profoundly affect cancer progression, but recent evidence also underscores their critical contribution to the anticancer efficacy of immune checkpoint inhibitors. In 2012, the two seminal studies suggested the superior antimelanoma activity of TH9 cells over other T cell subsets upon adoptive T cell transfer. While these findings provided great impetus to investigate further the unique functions of TH9 cells and explore their relevance in cancer immunotherapy, the following questions still remain outstanding: are TH9 cell anticancer functions restricted to melanoma? What are the factors favouring TH9 cell effector functions? What is the contribution of TH9 cells to cancer immunotherapy treatments? Can TH9 cells be identified in humans and, if so, what is their clinical relevance? By reviewing the studies addressing these questions, we will discuss how TH9 cells could be therapeutically harnessed for cancer immunotherapy strategies.
Collapse
Affiliation(s)
- Isis Benoit-Lizon
- INSERM, U1231, Dijon, France; Faculté de Médecine, Université de Bourgogne Franche Comté, Dijon, France
| | - Lionel Apetoh
- INSERM, U1231, Dijon, France; Faculté de Médecine, Université de Bourgogne Franche Comté, Dijon, France; Evergrande Center for Immunologic Diseases and Ann Romney Center for Neurologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA, USA.
| |
Collapse
|
26
|
Zhang Z, Zhou Q, Ouyang J, Pu J, Hou J, Zhang J. Expression and clinical significance of interleukin-9 in renal tumors. Transl Androl Urol 2021; 9:2657-2664. [PMID: 33457237 PMCID: PMC7807335 DOI: 10.21037/tau-20-761] [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] [Indexed: 11/18/2022] Open
Abstract
Background To measure expression levels of interleukin-9 (IL-9) in renal tumors and to determine the clinical significance of those levels. Methods Using TCGA database analysis, we found that the expression of IL-9 in renal clear cell carcinoma was significantly down-regulated, and was significantly related to survival. We then verified this using experiments. We enrolled 66 patients who underwent surgical resection of renal tumors between January and December 2018 at the First Affiliated Hospital of Soochow University. Their tumor tissues were paired with adjacent normal tissues and IL-9 expression levels were measured using immunohistochemistry. We determined the correlation of IL-9 expression with clinicopathological features and progression-free survival (PFS). Results Expression of IL-9 in renal tumors was significantly lower than in adjacent normal tissues (P<0.0001). There was a significant negative correlation between IL-9 expression levels and R.E.N.A.L. scores (P=0.0277) as well as with differentiation (P=0.0041). However, no significant correlation was found between IL-9 levels and clinicopathological features, including gender (P=0.0716), age (P=0.2566), body mass index (BMI) (P=0.7941), tumor size (P=0.4193) or TNM staging (P=0.5402). PFS time in renal tumor patients with positive IL-9 expression was similar to that of patients with negative IL-9 expression. Conclusions IL-9 expression was higher in adjacent normal tissues than in renal tumors. Low expression of IL-9 was detected when R.E.N.A.L. score was high or cell differentiation was poor, suggesting that IL-9 may may play a protective role in renal tumor microenvironments.
Collapse
Affiliation(s)
- Zhiyu Zhang
- Department of Urology, the First Affiliated Hospital of Soochow University, Suzhou, China
| | - Qi Zhou
- Department of Urology, the First Affiliated Hospital of Soochow University, Suzhou, China
| | - Jun Ouyang
- Department of Urology, the First Affiliated Hospital of Soochow University, Suzhou, China
| | - Jinxian Pu
- Department of Urology, the First Affiliated Hospital of Soochow University, Suzhou, China
| | - Jianquan Hou
- Department of Urology, the First Affiliated Hospital of Soochow University, Suzhou, China
| | - Jianglei Zhang
- Department of Urology, the First Affiliated Hospital of Soochow University, Suzhou, China
| |
Collapse
|
27
|
Li T, Wu B, Yang T, Zhang L, Jin K. The outstanding antitumor capacity of CD4 + T helper lymphocytes. Biochim Biophys Acta Rev Cancer 2020; 1874:188439. [PMID: 32980465 DOI: 10.1016/j.bbcan.2020.188439] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Revised: 09/10/2020] [Accepted: 09/21/2020] [Indexed: 02/05/2023]
Abstract
Over the past decades, tumor-resident immune cells have been extensively studied to dissect their biological functions and clinical roles. Tumor-infiltrating CD8+ T cells, because of their cytotoxic and killing ability, have been under the spotlight for a long time, whereas CD4+ T cells are considered just a supporting actor in the field of cancer immunotherapy. Until recently, accumulating evidence has demonstrated the ability of CD4+ T cells in eradicating solid tumors, and their functions in mediating antitumor immunity have been investigated in various orientations. In this review, we highlight the pivotal role of CD4+ T cells in eliciting vigorous antitumor immune responses, summarize key signaling axes and molecular networks behind these antitumor functions, and also propose possible targets and promising strategies which might translate into more efficient immunotherapies against human cancers.
Collapse
Affiliation(s)
- Tong Li
- Laboratory of Human Diseases and Immunotherapies, West China Hospital, Sichuan University, Chengdu 610041, China; State Key Laboratory of Biotherapy, Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu 610041, China
| | - Bowen Wu
- School of Medicine, Stanford University, Stanford, CA 94304, USA
| | - Tao Yang
- Laboratory of Human Diseases and Immunotherapies, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Long Zhang
- MOE Laboratory of Biosystems Homeostasis and Protection and Innovation Center for Cell Signaling Network, Life Sciences Institute, Zhejiang University, Hangzhou 310058, China
| | - Ke Jin
- Laboratory of Human Diseases and Immunotherapies, West China Hospital, Sichuan University, Chengdu 610041, China.
| |
Collapse
|
28
|
Chen T, Guo J, Cai Z, Li B, Sun L, Shen Y, Wang S, Wang Z, Wang Z, Wang Y, Zhou H, Cai Z, Ye Z. Th9 Cell Differentiation and Its Dual Effects in Tumor Development. Front Immunol 2020; 11:1026. [PMID: 32508847 PMCID: PMC7251969 DOI: 10.3389/fimmu.2020.01026] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Accepted: 04/28/2020] [Indexed: 12/17/2022] Open
Abstract
With the improved understanding of the molecular pathogenesis and characteristics of cancers, the critical role of the immune system in preventing tumor development has been widely accepted. The understanding of the relationship between the immune system and cancer progression is constantly evolving, from the cancer immunosurveillance hypothesis to immunoediting theory and the delicate balance in the tumor microenvironment. Currently, immunotherapy is regarded as a promising strategy against cancers. Although adoptive cell therapy (ACT) has shown some exciting results regarding the rejection of tumors, the effect is not always satisfactory. Cellular therapy with CD4+ T cells remains to be further explored since the current ACT is mainly focused on CD8+ cytotoxic T lymphocytes (CTLs). Recently, Th9 cells, a subgroup of CD4+ T helper cells characterized by the secretion of IL-9 and IL-10, have been reported to be effective in the elimination of solid tumors and to exhibit superior antitumor properties to Th1 and Th17 cells. In this review, we summarize the most recent advances in the understanding of Th9 cell differentiation and the dual role, both anti-tumor and pro-tumor effects, of Th9 cells in tumor progression.
Collapse
Affiliation(s)
- Tao Chen
- Department of Orthopedics, Musculoskeletal Tumor Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
- Institute of Orthopedic Research, Zhejiang University, Hangzhou, China
| | - Jufeng Guo
- Department of Breast Surgery, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Zhenhai Cai
- Department of Orthopedics Surgery, The Second Affiliated Hospital of Jiaxing University, Jiaxing, China
| | - Binghao Li
- Department of Orthopedics, Musculoskeletal Tumor Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
- Institute of Orthopedic Research, Zhejiang University, Hangzhou, China
| | - Lingling Sun
- Department of Orthopedics, Musculoskeletal Tumor Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
- Institute of Orthopedic Research, Zhejiang University, Hangzhou, China
| | - Yingying Shen
- Institute of Immunology, Zhejiang University School of Medicine, Hangzhou, China
| | - Shengdong Wang
- Department of Orthopedics, Musculoskeletal Tumor Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
- Institute of Orthopedic Research, Zhejiang University, Hangzhou, China
| | - Zhan Wang
- Department of Orthopedics, Musculoskeletal Tumor Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
- Institute of Orthopedic Research, Zhejiang University, Hangzhou, China
| | - Zenan Wang
- Department of Orthopedics, Musculoskeletal Tumor Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
- Institute of Orthopedic Research, Zhejiang University, Hangzhou, China
| | - Yucheng Wang
- Department of Orthopedics, Musculoskeletal Tumor Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
- Institute of Orthopedic Research, Zhejiang University, Hangzhou, China
| | - Hao Zhou
- Department of Orthopedics, Musculoskeletal Tumor Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
- Institute of Orthopedic Research, Zhejiang University, Hangzhou, China
| | - Zhijian Cai
- Department of Orthopedics, Musculoskeletal Tumor Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
- Institute of Orthopedic Research, Zhejiang University, Hangzhou, China
- Institute of Immunology, Zhejiang University School of Medicine, Hangzhou, China
| | - Zhaoming Ye
- Department of Orthopedics, Musculoskeletal Tumor Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
- Institute of Orthopedic Research, Zhejiang University, Hangzhou, China
| |
Collapse
|
29
|
Li W, Ma N, Yuwen T, Yu B, Zhou Y, Yao Y, Li Q, Chen X, Wan J, Zhang Y, Zhang W. Comprehensive analysis of circRNA expression profiles and circRNA-associated competing endogenous RNA networks in the development of mouse thymus. J Cell Mol Med 2020; 24:6340-6349. [PMID: 32307889 PMCID: PMC7294154 DOI: 10.1111/jcmm.15276] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Revised: 03/19/2020] [Accepted: 03/28/2020] [Indexed: 12/13/2022] Open
Abstract
The thymus plays an irreplaceable role as a primary lymphoid organ. However, the complicate processes of its development and involution are incompletely understood. Accumulating evidence indicates that non-coding RNAs play key roles in the regulation of biological development. At present, the studies of the circRNA profiles and of circRNA-associated competing endogenous RNAs (ceRNAs) in the thymus are still scarce. Here, deep-RNA sequencing was used to study the biological mechanisms underlying the development process (from 2-week-old to 6-week-old) and the recession process (from 6-week-old to 3-month-old) of the mouse thymus. It was found that 196 circRNAs, 233 miRNAs and 3807 mRNAs were significantly dysregulated. The circRNA-associated ceRNA networks were constructed in the mouse thymus, which were mainly involved in early embryonic development and the proliferation and division of T cells. Taken together, these results elucidated the regulatory roles of ceRNAs in the development and involution processes of the mouse thymus.
Collapse
Affiliation(s)
- Wenting Li
- Biomedical Research Institute, Shenzhen Peking University - The Hong Kong University of Science and Technology Medical Center, Shenzhen, China
| | - Nana Ma
- Biomedical Research Institute, Shenzhen Peking University - The Hong Kong University of Science and Technology Medical Center, Shenzhen, China
| | - Ting Yuwen
- Biomedical Research Institute, Shenzhen Peking University - The Hong Kong University of Science and Technology Medical Center, Shenzhen, China
| | - Bo Yu
- Department of Dermatology, Peking University Shenzhen Hospital, Shenzhen, China
| | - Yao Zhou
- Hainan Provincial Key Laboratory for human reproductive medicine and Genetic Research, The First Affiliated Hospital of Hainan Medical University, Hainan Medical University, Hainan, China
| | - Yufei Yao
- Hainan Provincial Key Laboratory for human reproductive medicine and Genetic Research, The First Affiliated Hospital of Hainan Medical University, Hainan Medical University, Hainan, China
| | - Qi Li
- Hainan Provincial Key Laboratory for human reproductive medicine and Genetic Research, The First Affiliated Hospital of Hainan Medical University, Hainan Medical University, Hainan, China
| | - Xiaofan Chen
- Biomedical Research Institute, Shenzhen Peking University - The Hong Kong University of Science and Technology Medical Center, Shenzhen, China
| | - Jun Wan
- Biomedical Research Institute, Shenzhen Peking University - The Hong Kong University of Science and Technology Medical Center, Shenzhen, China.,Division of Life Science, The Hong Kong University of Science and Technology, Kowloon, Hong Kong, China.,Greater Bay Biomedical Innocenter, Shenzhen Bay Laboratory, Shenzhen, China
| | - Yu Zhang
- Hainan Provincial Key Laboratory for human reproductive medicine and Genetic Research, The First Affiliated Hospital of Hainan Medical University, Hainan Medical University, Hainan, China.,Department of Reproductive Medicine, The First Affiliated Hospital of Hainan Medical University, Hainan Medical University, Hainan, China.,Hainan Provincial Clinical Research Center for Thalassemia, The First Affiliated Hospital of Hainan Medical University, Hainan Medical University, Hainan, China.,Key Laboratory of Tropical Translational Medicine of Ministry of Education, Hainan Medical University, Hainan, China
| | - Wei Zhang
- Biomedical Research Institute, Shenzhen Peking University - The Hong Kong University of Science and Technology Medical Center, Shenzhen, China.,Greater Bay Biomedical Innocenter, Shenzhen Bay Laboratory, Shenzhen, China
| |
Collapse
|
30
|
Wan J, Wu Y, Ji X, Huang L, Cai W, Su Z, Wang S, Xu H. IL-9 and IL-9-producing cells in tumor immunity. Cell Commun Signal 2020; 18:50. [PMID: 32228589 PMCID: PMC7104514 DOI: 10.1186/s12964-020-00538-5] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Accepted: 02/19/2020] [Indexed: 12/11/2022] Open
Abstract
Abstract Interleukin (IL)-9 belongs to the IL-2Rγc chain family and is a multifunctional cytokine that can regulate the function of many kinds of cells. It was originally identified as a growth factor of T cells and mast cells. In previous studies, IL-9 was mainly involved in the development of allergic diseases, autoimmune diseases and parasite infections. Recently, IL-9, as a double-edged sword in the development of cancers, has attracted extensive attention. Since T-helper 9 (Th9) cell-derived IL-9 was verified to play a powerful antitumor role in solid tumors, an increasing number of researchers have started to pay attention to the role of IL-9-skewed CD8+ T (Tc9) cells, mast cells and Vδ2 T cell-derived IL-9 in tumor immunity. Here, we review recent studies on IL-9 and several kinds of IL-9-producing cells in tumor immunity to provide useful insight into tumorigenesis and treatment. Video Abstract
Graphical abstract ![]()
Collapse
Affiliation(s)
- Jie Wan
- Department of Immunology, Jiangsu University, Zhenjiang, 212013, China
| | - Yinqiu Wu
- Department of Immunology, Jiangsu University, Zhenjiang, 212013, China
| | - Xiaoyun Ji
- Department of Immunology, Jiangsu University, Zhenjiang, 212013, China
| | - Lan Huang
- Department of Immunology, Jiangsu University, Zhenjiang, 212013, China
| | - Wei Cai
- Department of Immunology, Jiangsu University, Zhenjiang, 212013, China
| | - Zhaoliang Su
- Department of Immunology, Jiangsu University, Zhenjiang, 212013, China.,China International Genomics Research Center (IGRC), Jiangsu University, Zhenjiang, 212013, China
| | - Shengjun Wang
- Department of Immunology, Jiangsu University, Zhenjiang, 212013, China.,Department of Laboratory Medicine, The Affiliated People's Hospital, Jiangsu University, Zhenjiang, 212001, China
| | - Huaxi Xu
- Department of Immunology, Jiangsu University, Zhenjiang, 212013, China.
| |
Collapse
|
31
|
Hu L, Xu J, Wu T, Fan Z, Sun L, Liu Y, Li Y, Zhang C, Wang J, Ding Y, Wang S. Depletion of ID3 enhances mesenchymal stem cells therapy by targeting BMP4 in Sjögren's syndrome. Cell Death Dis 2020; 11:172. [PMID: 32139667 PMCID: PMC7058624 DOI: 10.1038/s41419-020-2359-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Revised: 02/15/2020] [Accepted: 02/18/2020] [Indexed: 12/11/2022]
Abstract
Mesenchymal stem cell (MSCs) transplantation has been used to treat Sjögren’s syndrome (SS) based on the immunoregulatory properties of MSCs. However, the effectiveness need improving and its underlying intrinsic mechanisms remain largely unknown. Here, we show that Id3 is upregulated in bone marrow-derived MSCs (BMMSCs) isolated from NOD/ShiLtJ mice, a widely used SS model, compared with ICR mice as control, suggesting that it functions in SS development and therapy. Transplantation of Id3-deficient BMMSCs rescues salivary gland function more effective than wild-type BMMSCs in NOD/ShiLtJ mice. Mechanistically, we show that ID3 negatively regulated BMP4 expression by preventing binding of basic helix–loop–helix protein E2A to the promoter of the Bmp4 gene. BMP4 in turn promoted PGE2 production in MSCs, and exhibited enhanced suppressive activities of T-cell proliferation and Th1 differentiation. Importantly, BMMSCs from SS patients showed significantly lower BMP4 and PGE2 expression than those from healthy individuals. Taken together, our findings revealed the targeting Id3 may be therapeutically useful for improving MSC immunoregulation and effectiveness of MSCs therapy for SS.
Collapse
Affiliation(s)
- Lei Hu
- Salivary Gland Disease Center and Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Capital Medical University School of Stomatology, Beijing, 100050, China.
| | - Junji Xu
- Salivary Gland Disease Center and Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Capital Medical University School of Stomatology, Beijing, 100050, China.,National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Tingting Wu
- Salivary Gland Disease Center and Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Capital Medical University School of Stomatology, Beijing, 100050, China
| | - Zhipeng Fan
- Salivary Gland Disease Center and Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Capital Medical University School of Stomatology, Beijing, 100050, China
| | - Lingyun Sun
- Department of Rheumatology and Immunology, Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China
| | - Yi Liu
- Salivary Gland Disease Center and Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Capital Medical University School of Stomatology, Beijing, 100050, China
| | - Yan Li
- Health Management Center, The Third Xiangya Hospital of Central South University, Changsha, China.,Fortune Link Triones (Beijing) Scitech Co., Ltd., Beijing, China
| | - Chunmei Zhang
- Salivary Gland Disease Center and Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Capital Medical University School of Stomatology, Beijing, 100050, China
| | - Jingsong Wang
- Salivary Gland Disease Center and Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Capital Medical University School of Stomatology, Beijing, 100050, China.,Department of Biochemistry and Molecular Biology, Capital Medical University School of Basic Medical Sciences, Beijing, 100069, China
| | - Yaozhong Ding
- Department of Immunology, Capital Medical University, Beijing, 100069, China
| | - Songlin Wang
- Salivary Gland Disease Center and Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Capital Medical University School of Stomatology, Beijing, 100050, China. .,Department of Biochemistry and Molecular Biology, Capital Medical University School of Basic Medical Sciences, Beijing, 100069, China.
| |
Collapse
|
32
|
Chen Z, Bozec A, Ramming A, Schett G. Anti-inflammatory and immune-regulatory cytokines in rheumatoid arthritis. Nat Rev Rheumatol 2020; 15:9-17. [PMID: 30341437 DOI: 10.1038/s41584-018-0109-2] [Citation(s) in RCA: 462] [Impact Index Per Article: 92.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Rheumatoid arthritis (RA) is a chronic autoimmune disease characterized by a failure of spontaneous resolution of inflammation. Although the pro-inflammatory cytokines and mediators that trigger RA have been the focus of intense investigations, the regulatory and anti-inflammatory cytokines responsible for the suppression and resolution of disease in a context-dependent manner have been less well characterized. However, knowledge of the pathways that control the suppression and resolution of inflammation in RA is clinically relevant and conceptually important for understanding the pathophysiology of the disease and for the development of treatments that enable long-term remission. Cytokine-mediated processes such as the activation of T helper 2 cells by IL-4 and IL-13, the resolution of inflammation by IL-9, IL-5-induced eosinophil expansion, IL-33-mediated macrophage polarization, the production of IL-10 by regulatory B cells and IL-27-mediated suppression of lymphoid follicle formation are all involved in governing the regulation and resolution of inflammation in RA. By better understanding these immune-regulatory signalling pathways, new therapeutic strategies for RA can be envisioned that aim to balance and resolve, rather than suppress, inflammation.
Collapse
Affiliation(s)
- Zhu Chen
- Department of Rheumatology and Immunology, The First Affiliated Hospital of the University of Science and Technology of China, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Aline Bozec
- Department of Internal Medicine 3, Friedrich Alexander University Erlangen-Nuremberg and Universitatsklinikum Erlangen, Erlangen, Germany
| | - Andreas Ramming
- Department of Internal Medicine 3, Friedrich Alexander University Erlangen-Nuremberg and Universitatsklinikum Erlangen, Erlangen, Germany
| | - Georg Schett
- Department of Internal Medicine 3, Friedrich Alexander University Erlangen-Nuremberg and Universitatsklinikum Erlangen, Erlangen, Germany.
| |
Collapse
|
33
|
Abstract
Interleukin (IL)-9 is a pleiotropic cytokine, which can function as a positive or negative regulator of immune responses on multiple types of cells. The role of IL-9 was originally known in allergic disease and parasite infections. Interestingly, recent studies demonstrate its presence in the tumor tissues of mice and humans, and the association between IL-9 and tumor progression has been revisited following the discovery of T helper (Th) 9 cells. Tumor-specific Th9 cells are considered to be the main subset of CD4+ T cells that produce high level of IL-9 and exhibit an IL-9-dependent robust anti-cancer function in solid tumors. IL-9 exerts an unprecedented anti-tumor immunity not only by inducing innate and adaptive immune responses but also directly promoting apoptosis of tumor cells. The objective of this review is to summarize the latest advances regarding the anti-tumor mechanisms of IL-9 and Th9 cells.
Collapse
Affiliation(s)
- Ningbo Zheng
- Department of Microbiology & Immunology, Wake Forest School of Medicine , Winston-Salem, NC, USA
| | - Yong Lu
- Department of Microbiology & Immunology, Wake Forest School of Medicine , Winston-Salem, NC, USA
| |
Collapse
|
34
|
IL-9 and Th9 Cells in Tumor Immunity. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1240:35-46. [DOI: 10.1007/978-3-030-38315-2_3] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
35
|
Li Y, Liu Z, Wang J, Yu J, Li Z, Yang H, Tang J, Chen Z. Receptor-Interacting Protein Kinase 3 Deficiency Recruits Myeloid-Derived Suppressor Cells to Hepatocellular Carcinoma Through the Chemokine (C-X-C Motif) Ligand 1-Chemokine (C-X-C Motif) Receptor 2 Axis. Hepatology 2019; 70:1564-1581. [PMID: 31021443 PMCID: PMC6900048 DOI: 10.1002/hep.30676] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/02/2019] [Accepted: 04/16/2019] [Indexed: 12/15/2022]
Abstract
Receptor-interacting protein kinase 3 (RIP3) is the core regulator that switches cell death from apoptosis to necrosis. However, its role in tumor immunity is unknown. In this study, decreased RIP3 expression was observed in patients with hepatocellular carcinoma (HCC), which correlates with myeloid-derived suppressor cell (MDSC) accumulation. Moreover, RIP3 is a prognosis factor for patients with HCC. We further found that RIP3 knockdown results in an increase of MDSCs and a decrease of interferon gamma-positive (IFN-γ+ ) cluster of differentiation 8-positive (CD8+ ) tumor-infiltrating lymphocytes (IFN-γ+ CD8+ T cells) in hepatoma tissues, thus promoting immune escape and HCC growth in immunocompetent mice. By phosphorylating P65Ser536 and promoting phosphorylated P65Ser536 nuclear translocation, RIP3 knockdown increases the expression of chemokine (C-X-C motif) ligand 1 (CXCL1) in HCC cells. RIP3 knockdown induces MDSC recruitment through the CXCL1-chemokine (C-X-C motif) receptor 2 (CXCR2) axis. Furthermore, a CXCR2 antagonist substantially suppresses MDSC chemotaxis and HCC growth in RIP3 knockout mice. Conclusion: RIP3 deficiency is an essential factor directing MDSC homing to HCC and promoting CXCL1/CXCR2-induced MDSC chemotaxis to facilitate HCC immune escape and HCC progression; blocking the CXCL1-CXCR2 chemokine axis may provide an immunological therapeutic approach to suppress progression of RIP3 deficiency HCC.
Collapse
Affiliation(s)
- Yi‐Ming Li
- National Translational Science Center for Molecular Medicine & Department of Cell BiologyThe Fourth Military Medical UniversityXi'anChina
| | - Zhen‐Yu Liu
- National Translational Science Center for Molecular Medicine & Department of Cell BiologyThe Fourth Military Medical UniversityXi'anChina
| | - Jian‐Chao Wang
- National Translational Science Center for Molecular Medicine & Department of Cell BiologyThe Fourth Military Medical UniversityXi'anChina
| | - Jing‐Min Yu
- National Translational Science Center for Molecular Medicine & Department of Cell BiologyThe Fourth Military Medical UniversityXi'anChina
| | - Zhu‐Chun Li
- National Translational Science Center for Molecular Medicine & Department of Cell BiologyThe Fourth Military Medical UniversityXi'anChina
| | - Hai‐Jiao Yang
- National Translational Science Center for Molecular Medicine & Department of Cell BiologyThe Fourth Military Medical UniversityXi'anChina
| | - Juan Tang
- National Translational Science Center for Molecular Medicine & Department of Cell BiologyThe Fourth Military Medical UniversityXi'anChina
| | - Zhi‐Nan Chen
- National Translational Science Center for Molecular Medicine & Department of Cell BiologyThe Fourth Military Medical UniversityXi'anChina
| |
Collapse
|
36
|
Xiao D, Li X, Rouchka EC, Waigel S, Zacharias W, McMasters KM, Hao H. Comparative gene expression analysis in melanocytes driven by tumor cell-derived exosomes. Exp Cell Res 2019; 386:111690. [PMID: 31678172 DOI: 10.1016/j.yexcr.2019.111690] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Revised: 10/25/2019] [Accepted: 10/27/2019] [Indexed: 12/31/2022]
Abstract
Abundant with organelle-like membranous structures, the tumor microenvironment is composed of cancer cells that secrete exosomes. Studies have shown that these secreted exosomes transport RNA and active molecules to other cells to reshape the tumor microenvironment and promote tumor growth. In fact, we found that exosomes derived from melanoma cells drive pre-malignant transition in primary melanocytes. However, there is little available in the scientific literature on how exosomes modulate melanocytes in the microenvironment to optimize conditions for tumor progression and metastasis. We therefore focused this current study on identifying these conditions genetically. Through RNA sequencing, we analyzed gene expression levels of melanocytes driven by exosomes derived from melanoma and lung cancer cells compared with those without exosome controls. Significant differences were found in gene expression patterns of melanocytes driven by exosomes derived from melanoma and lung cancer cells. In the melanocytes responding to exosomes derived from melanoma cells, genes of lipopolysaccharide and regulation of leukocyte chemotaxis were predominant. In the melanocytes responding to exosomes derived from lung cancer cells, genes of DNA replication and mitotic nuclear division played an important role. These results provide further mechanistic understanding of tumor progression promoted by tumor-derived exosomes. This will also help identify potential therapeutic targets for melanoma progression.
Collapse
Affiliation(s)
- Deyi Xiao
- Hiram C. Polk, Jr., MD Department of Surgery, University of Louisville School of Medicine, Louisville, KY, 40292, USA
| | - Xiaohong Li
- Bioinformatics Laboratory, University of Louisville School of Medicine, Louisville, KY, 40292, USA
| | - Eric C Rouchka
- Bioinformatics Laboratory, University of Louisville School of Medicine, Louisville, KY, 40292, USA
| | - Sabine Waigel
- Genomics Facility, University of Louisville School of Medicine, Louisville, KY, 40292, USA
| | - Wolfgang Zacharias
- Genomics Facility, University of Louisville School of Medicine, Louisville, KY, 40292, USA; Department of Medicine and Department of Pharmacology and Toxicology, James Graham Brown Cancer Center, University of Louisville School of Medicine, Louisville, KY, 40292, USA
| | - Kelly M McMasters
- Hiram C. Polk, Jr., MD Department of Surgery, University of Louisville School of Medicine, Louisville, KY, 40292, USA
| | - Hongying Hao
- Hiram C. Polk, Jr., MD Department of Surgery, University of Louisville School of Medicine, Louisville, KY, 40292, USA.
| |
Collapse
|
37
|
Zhang D, Jin W, Wu R, Li J, Park SA, Tu E, Zanvit P, Xu J, Liu O, Cain A, Chen W. High Glucose Intake Exacerbates Autoimmunity through Reactive-Oxygen-Species-Mediated TGF-β Cytokine Activation. Immunity 2019; 51:671-681.e5. [PMID: 31451397 PMCID: PMC9811990 DOI: 10.1016/j.immuni.2019.08.001] [Citation(s) in RCA: 191] [Impact Index Per Article: 31.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Revised: 05/08/2019] [Accepted: 07/30/2019] [Indexed: 01/07/2023]
Abstract
Diet has been suggested to be a potential environmental risk factor for the increasing incidence of autoimmune diseases, yet the underlying mechanisms remain elusive. Here, we show that high glucose intake exacerbated autoimmunity in mouse models of colitis and experimental autoimmune encephalomyelitis (EAE). We elucidated that high amounts of glucose specifically promoted T helper-17 (Th17) cell differentiation by activating transforming growth factor-β (TGF-β) from its latent form through upregulation of reactive oxygen species (ROS) in T cells. We further determined that mitochondrial ROS (mtROS) are key for high glucose-induced TGF-β activation and Th17 cell generation. We have thus revealed a previously unrecognized mechanism underlying the adverse effects of high glucose intake in the pathogenesis of autoimmunity and inflammation.
Collapse
Affiliation(s)
- Dunfang Zhang
- Mucosal Immunology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD 20892, USA
| | - Wenwen Jin
- Mucosal Immunology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD 20892, USA
| | - Ruiqing Wu
- Mucosal Immunology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD 20892, USA
| | - Jia Li
- Mucosal Immunology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD 20892, USA
| | - Sang-A Park
- Mucosal Immunology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD 20892, USA
| | - Eric Tu
- Mucosal Immunology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD 20892, USA
| | - Peter Zanvit
- Mucosal Immunology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD 20892, USA
| | - Junji Xu
- Mucosal Immunology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD 20892, USA
| | - Ousheng Liu
- Mucosal Immunology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD 20892, USA
| | - Alexander Cain
- Mucosal Immunology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD 20892, USA
| | - WanJun Chen
- Mucosal Immunology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD 20892, USA.
| |
Collapse
|
38
|
Romano O, Miccio A. GATA factor transcriptional activity: Insights from genome-wide binding profiles. IUBMB Life 2019; 72:10-26. [PMID: 31574210 DOI: 10.1002/iub.2169] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Accepted: 09/05/2019] [Indexed: 01/07/2023]
Abstract
The members of the GATA family of transcription factors have homologous zinc fingers and bind to similar sequence motifs. Recent advances in genome-wide technologies and the integration of bioinformatics data have led to a better understanding of how GATA factors regulate gene expression; GATA-factor-induced transcriptional and epigenetic changes have now been analyzed at unprecedented levels of detail. Here, we review the results of genome-wide studies of GATA factor occupancy in human and murine cell lines and primary cells (as determined by chromatin immunoprecipitation sequencing), and then discuss the molecular mechanisms underlying the mediation of transcriptional and epigenetic regulation by GATA factors.
Collapse
Affiliation(s)
- Oriana Romano
- Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Annarita Miccio
- Laboratory of chromatin and gene regulation during development, Imagine Institute, INSERM UMR, Paris, France.,Paris Descartes, Sorbonne Paris Cité University, Imagine Institute, Paris, France
| |
Collapse
|
39
|
The dichotomous function of interleukin-9 in cancer diseases. J Mol Med (Berl) 2019; 97:1377-1383. [PMID: 31396657 DOI: 10.1007/s00109-019-01826-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Revised: 07/09/2019] [Accepted: 07/25/2019] [Indexed: 12/20/2022]
Abstract
The pleiotropic function of the cytokine IL-9 is so far described in many inflammation processes and autoimmune diseases. But its role in cancer immunology is rather diverse as it can have a pro-tumorigenic function as well as anti-tumorigenic characteristics. In various disease models of cancer, this cytokine is involved in different signaling pathways triggering the expression of proteins involved in cell growth, migration, and transformation or repressing cells from the adaptive immune system to reject tumor growth. Additionally, there are even therapeutic approaches for IL-9 in cancer development. This review will give an overview of the various roles of IL-9 in different immune organs and cells and provide an insight in the current state of research in the IL-9-dependent cancer area.
Collapse
|
40
|
Hu B, Li G, Ye Z, Gustafson CE, Tian L, Weyand CM, Goronzy JJ. Transcription factor networks in aged naïve CD4 T cells bias lineage differentiation. Aging Cell 2019; 18:e12957. [PMID: 31264370 PMCID: PMC6612640 DOI: 10.1111/acel.12957] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2018] [Revised: 02/17/2019] [Accepted: 03/18/2019] [Indexed: 12/13/2022] Open
Abstract
With reduced thymic activity, the population of naïve T cells in humans is maintained by homeostatic proliferation throughout adult life. In young adults, naïve CD4 T cells have enormous proliferative potential and plasticity to differentiate into different lineages. Here, we explored whether naïve CD4 T-cell aging is associated with a partial loss of this unbiased multipotency. We find that naïve CD4 T cells from older individuals have developed a propensity to develop into TH9 cells. Two major mechanisms contribute to this predisposition. First, responsiveness to transforming growth factor β (TGFβ) stimulation is enhanced with age due to an upregulation of the TGFβR3 receptor that results in increased expression of the transcription factor PU.1. Secondly, aged naïve CD4 T cells display altered transcription factor profiles in response to T-cell receptor stimulation, including enhanced expression of BATF and IRF4 and reduced expression of ID3 and BCL6. These transcription factors are involved in TH9 differentiation as well as IL9 transcription suggesting that the aging-associated changes in the transcription factor profile favor TH9 commitment.
Collapse
Affiliation(s)
- Bin Hu
- Department of Medicine, Division of Immunology and RheumatologyStanford UniversityStanfordCaliforniaUSA
- Department of MedicinePalo Alto Veterans Administration Healthcare SystemPalo AltoCaliforniaUSA
| | - Guangjin Li
- Department of MedicinePalo Alto Veterans Administration Healthcare SystemPalo AltoCaliforniaUSA
| | - Zhongde Ye
- Department of MedicinePalo Alto Veterans Administration Healthcare SystemPalo AltoCaliforniaUSA
| | - Claire E. Gustafson
- Department of Medicine, Division of Immunology and RheumatologyStanford UniversityStanfordCaliforniaUSA
- Department of MedicinePalo Alto Veterans Administration Healthcare SystemPalo AltoCaliforniaUSA
| | - Lu Tian
- Department of Biomedical Data ScienceStanford University School of MedicineStanfordCaliforniaUSA
| | - Cornelia M. Weyand
- Department of Medicine, Division of Immunology and RheumatologyStanford UniversityStanfordCaliforniaUSA
- Department of MedicinePalo Alto Veterans Administration Healthcare SystemPalo AltoCaliforniaUSA
| | - Jörg J. Goronzy
- Department of Medicine, Division of Immunology and RheumatologyStanford UniversityStanfordCaliforniaUSA
- Department of MedicinePalo Alto Veterans Administration Healthcare SystemPalo AltoCaliforniaUSA
| |
Collapse
|
41
|
Dahmani A, Janelle V, Carli C, Richaud M, Lamarche C, Khalili M, Goupil M, Bezverbnaya K, Bramson JL, Delisle JS. TGFβ Programs Central Memory Differentiation in Ex Vivo-Stimulated Human T Cells. Cancer Immunol Res 2019; 7:1426-1439. [PMID: 31308016 DOI: 10.1158/2326-6066.cir-18-0691] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2018] [Revised: 03/27/2019] [Accepted: 07/09/2019] [Indexed: 11/16/2022]
Abstract
The adoptive transfer of ex vivo-expanded T cells is a promising approach to treat several malignancies. Several lines of evidence support that the infusion of T cells with early memory features, capable of expanding and persisting after transfer, are associated with better outcomes. We report herein that exposure to exogenous TGFβ during human T-cell stimulation ex vivo leads to the accumulation of early/central memory (Tcm) cells. Exposure to TGFβ suppressed the expression of BLIMP-1, a key orchestrator of effector T-cell differentiation, and led to the upregulation of the memory-associated transcription factor ID3. Accordingly, this was associated with an early memory transcriptional signature in both CD4+ and CD8+ T-cell subsets. The T cells stimulated in the presence of TGFβ expanded normally, and displayed polyfunctional features and no suppressive activity. The adoptive transfer of ex vivo-stimulated T cells into immunodeficient mice confirmed that TGFβ-conditioned cells had an enhanced capacity to persist and mediate xenogeneic graft-versus-host disease, as predicted by their early T-cell memory phenotype. Chimeric antigen receptor-expressing T cells generated in the presence of exogenous TGFβ were cytotoxic and more effective at controlling tumor growth in immunodeficient animals. This work unveils a new role for TGFβ in memory T-cell differentiation and indicates that TGFβ signaling may be harnessed to program Tcm differentiation in the context of ex vivo T-cell stimulation for adoptive immunotherapy in humans.
Collapse
Affiliation(s)
- Amina Dahmani
- Centre de Recherche de l'Hôpital Maisonneuve-Rosemont (CRHMR), Montreal, Quebec, Canada
| | - Valérie Janelle
- Centre de Recherche de l'Hôpital Maisonneuve-Rosemont (CRHMR), Montreal, Quebec, Canada
| | - Cédric Carli
- Centre de Recherche de l'Hôpital Maisonneuve-Rosemont (CRHMR), Montreal, Quebec, Canada
| | - Manon Richaud
- Centre de Recherche de l'Hôpital Maisonneuve-Rosemont (CRHMR), Montreal, Quebec, Canada
| | - Caroline Lamarche
- Centre de Recherche de l'Hôpital Maisonneuve-Rosemont (CRHMR), Montreal, Quebec, Canada
- Department of Surgery, University of British Columbia, Vancouver, British Columbia, Canada
- BC Children's Hospital Research Institute, Vancouver, British Columbia, Canada
| | - Myriam Khalili
- Centre de Recherche de l'Hôpital Maisonneuve-Rosemont (CRHMR), Montreal, Quebec, Canada
| | - Mathieu Goupil
- Centre de Recherche de l'Hôpital Maisonneuve-Rosemont (CRHMR), Montreal, Quebec, Canada
| | - Ksenia Bezverbnaya
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Jonathan L Bramson
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Jean-Sébastien Delisle
- Centre de Recherche de l'Hôpital Maisonneuve-Rosemont (CRHMR), Montreal, Quebec, Canada.
- Département de Médecine, Université de Montréal, Montreal, Quebec, Canada
- Hematology-Oncology Division, Hôpital Maisonneuve-Rosemont, Montreal, Quebec, Canada
| |
Collapse
|
42
|
Shen Y, Song Z, Lu X, Ma Z, Lu C, Zhang B, Chen Y, Duan M, Apetoh L, Li X, Guo J, Miao Y, Zhang G, Yang D, Cai Z, Wang J. Fas signaling-mediated T H9 cell differentiation favors bowel inflammation and antitumor functions. Nat Commun 2019; 10:2924. [PMID: 31266950 PMCID: PMC6606754 DOI: 10.1038/s41467-019-10889-4] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2018] [Accepted: 06/07/2019] [Indexed: 11/17/2022] Open
Abstract
Fas induces apoptosis in activated T cell to maintain immune homeostasis, but the effects of non-apoptotic Fas signaling on T cells remain unclear. Here we show that Fas promotes TH9 cell differentiation by activating NF-κB via Ca2+-dependent PKC-β activation. In addition, PKC-β also phosphorylates p38 to inactivate NFAT1 and reduce NFAT1-NF-κB synergy to promote the Fas-induced TH9 transcription program. Fas ligation exacerbates inflammatory bowel disease by increasing TH9 cell differentiation, and promotes antitumor activity in p38 inhibitor-treated TH9 cells. Furthermore, low-dose p38 inhibitor suppresses tumor growth without inducing systemic adverse effects. In patients with tumor, relatively high TH9 cell numbers are associated with good prognosis. Our study thus implicates Fas in CD4+ T cells as a target for inflammatory bowel disease therapy. Furthermore, simultaneous Fas ligation and low-dose p38 inhibition may be an effective approach for TH9 cell induction and cancer therapy.
Collapse
Affiliation(s)
- Yingying Shen
- Institute of Immunology and Bone Marrow Transplantation Center of the First Affiliated Hospital, Zhejiang University School of Medicine, 310058, Hangzhou, China
- Institute of Hematology, Zhejiang University and Zhejiang Engineering Laboratory for Stem Cell and Immunotherapy, 310003, Hangzhou, China
- Institute of Immunology and Department of Orthopaedics of the Second Affiliated Hospital, Zhejiang University School of Medicine, 310058, Hangzhou, China
| | - Zhengbo Song
- Department of Medical Oncology, Zhejiang Cancer Hospital, 310022, Hangzhou, China
| | - Xinliang Lu
- Institute of Immunology and Bone Marrow Transplantation Center of the First Affiliated Hospital, Zhejiang University School of Medicine, 310058, Hangzhou, China
| | - Zeyu Ma
- Institute of Immunology and Bone Marrow Transplantation Center of the First Affiliated Hospital, Zhejiang University School of Medicine, 310058, Hangzhou, China
| | - Chaojie Lu
- Institute of Immunology and Bone Marrow Transplantation Center of the First Affiliated Hospital, Zhejiang University School of Medicine, 310058, Hangzhou, China
| | - Bei Zhang
- Institute of Immunology and Bone Marrow Transplantation Center of the First Affiliated Hospital, Zhejiang University School of Medicine, 310058, Hangzhou, China
| | - Yinghu Chen
- Division of Infection Disease, Zhejiang Key Laboratory for Neonatal Diseases, Children's Hospital, Zhejiang University School of Medicine, 310006, Hangzhou, China
| | - Meng Duan
- Chronic Disease Research Institute, School of Public Health, School of Medicine, Zhejiang University, 310058, Hangzhou, China
| | - Lionel Apetoh
- INSERM, U866, Dijon, France
- Faculté de Médecine, Université de Bourgogne, Dijon, 21000, France
| | - Xu Li
- School of Life Science, Westlake University, 310024, Hangzhou, China
| | - Jufeng Guo
- Department of Breast Surgery, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, 310006, Hangzhou, China
| | - Ying Miao
- Clinical Trial Center, Qingdao Municipal Hospital, 266011, Qingdao, China
| | - Gensheng Zhang
- Department of Critical Care Medicine, Second Affiliated Hospital, Zhejiang University School of Medicine, 310009, Hangzhou, China
| | - Diya Yang
- Xinyuan Institute of Medicine and Biotechnology, School of Life Sciences, Zhejiang Sci-Tech University, 310018, Hangzhou, China
| | - Zhijian Cai
- Institute of Immunology and Department of Orthopaedics of the Second Affiliated Hospital, Zhejiang University School of Medicine, 310058, Hangzhou, China.
| | - Jianli Wang
- Institute of Immunology and Bone Marrow Transplantation Center of the First Affiliated Hospital, Zhejiang University School of Medicine, 310058, Hangzhou, China.
- Institute of Hematology, Zhejiang University and Zhejiang Engineering Laboratory for Stem Cell and Immunotherapy, 310003, Hangzhou, China.
| |
Collapse
|
43
|
Inhibitor of DNA binding in heart development and cardiovascular diseases. Cell Commun Signal 2019; 17:51. [PMID: 31126344 PMCID: PMC6534900 DOI: 10.1186/s12964-019-0365-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Accepted: 05/14/2019] [Indexed: 02/05/2023] Open
Abstract
Id proteins, inhibitors of DNA binding, are transcription regulators containing a highly conserved helix-loop-helix domain. During multiple stages of normal cardiogenesis, Id proteins play major roles in early development and participate in the differentiation and proliferation of cardiac progenitor cells and mature cardiomyocytes. The fact that a depletion of Ids can cause a variety of defects in cardiac structure and conduction function is further evidence of their involvement in heart development. Multiple signalling pathways and growth factors are involved in the regulation of Ids in a cell- and tissue- specific manner to affect heart development. Recent studies have demonstrated that Ids are related to multiple aspects of cardiovascular diseases, including congenital structural, coronary heart disease, and arrhythmia. Although a growing body of research has elucidated the important role of Ids, no comprehensive review has previously compiled these scattered findings. Here, we introduce and summarize the roles of Id proteins in heart development, with the hope that this overview of key findings might shed light on the molecular basis of consequential cardiovascular diseases. Furthermore, we described the future prospective researches needed to enable advancement in the maintainance of the proliferative capacity of cardiomyocytes. Additionally, research focusing on increasing embryonic stem cell culture adaptability will help to improve the future therapeutic application of cardiac regeneration.
Collapse
|
44
|
Lee JE, Zhu Z, Bai Q, Brady TJ, Xiao H, Wakefield MR, Fang Y. The Role of Interleukin-9 in Cancer. Pathol Oncol Res 2019; 26:2017-2022. [PMID: 31016637 DOI: 10.1007/s12253-019-00665-6] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2019] [Accepted: 04/12/2019] [Indexed: 10/27/2022]
Abstract
Interluekin-9 (IL-9) is produced predominantly by helper T cells such as Th2 and Th9 cells. It normally functions through the activation of a JAK/STAT pathway and plays a critical role in immunity and the pathogenesis of cancer. In cancer, it yields different responses depending on the cancer cell line involved. This review is a summary of what is known about the involvement of IL-9 in various cancer cell lines as well as its role in immunity with a focus on allergic responses.
Collapse
Affiliation(s)
- Jacob E Lee
- Department of Microbiology, Immunology & Pathology, Des Moines University College of Osteopathic Medicine, Des Moines, Iowa, 50312, USA
| | - Ziwen Zhu
- Department of Surgery, University of Missouri School of Medicine, Columbia, MO, USA
| | - Qian Bai
- Department of Surgery, University of Missouri School of Medicine, Columbia, MO, USA
| | - Tucker J Brady
- Department of Microbiology, Immunology & Pathology, Des Moines University College of Osteopathic Medicine, Des Moines, Iowa, 50312, USA
| | - Huaping Xiao
- Department of Microbiology, Immunology & Pathology, Des Moines University College of Osteopathic Medicine, Des Moines, Iowa, 50312, USA.,The Affiliated Hospital of Xiangnan University, Chenzhou, Hunan, China
| | - Mark R Wakefield
- Department of Surgery, University of Missouri School of Medicine, Columbia, MO, USA
| | - Yujiang Fang
- Department of Microbiology, Immunology & Pathology, Des Moines University College of Osteopathic Medicine, Des Moines, Iowa, 50312, USA. .,Department of Surgery, University of Missouri School of Medicine, Columbia, MO, USA.
| |
Collapse
|
45
|
IL-4 together with IL-1β induces antitumor Th9 cell differentiation in the absence of TGF-β signaling. Nat Commun 2019; 10:1376. [PMID: 30914642 PMCID: PMC6435687 DOI: 10.1038/s41467-019-09401-9] [Citation(s) in RCA: 80] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2018] [Accepted: 02/28/2019] [Indexed: 12/21/2022] Open
Abstract
IL-9-producing CD4+ (Th9) cells are a subset of CD4+ T-helper cells that are endowed with powerful antitumor capacity. Both IL-4 and TGF-β have been reported to be indispensable for Th9 cell-priming and differentiation. Here we show, by contrast, that Th9 cell development can occur in the absence of TGF-β signaling. When TGF-β was replaced by IL-1β, the combination of IL-1β and IL-4 efficiently promoted IL-9-producing T cells (Th9IL-4+IL-1β). Th9IL-4+ IL-1β cells are phenotypically distinct T cells compared to classic Th9 cells (Th9IL-4+TGF-β) and other Th cells, and are enriched for IL-1 and NF-κB gene signatures. Inhibition of NF-κB but not TGF-β-signaling negates IL-9 production by Th9IL-4+IL-1β cells. Furthermore, when compared with classic Th9IL-4+TGF-β cells, Th9IL-4+IL-1β cells are less exhausted, exhibit cytotoxic T effector gene signature and tumor killing function, and exert a superior antitumor response in a mouse melanoma model. Our study thus describes an alternative pathway for Th9 cell differentiation and provides a potential avenue for antitumor therapies. CD4+ helper T cells producing IL-9 (Th9) have been implicated in anti-tumor immunity, with Th9 differentiation inducible in vitro via IL-4 and TGFβ treatment. Here the authors show that replacing TGFβ with IL-1β induces a distinct IL-9+ CD4+ population that have strong cytotoxic and anti-tumor activity in preclinical mouse models.
Collapse
|
46
|
Tu E, Chia CPZ, Chen W, Zhang D, Park SA, Jin W, Wang D, Alegre ML, Zhang YE, Sun L, Chen W. T Cell Receptor-Regulated TGF-β Type I Receptor Expression Determines T Cell Quiescence and Activation. Immunity 2019; 48:745-759.e6. [PMID: 29669252 DOI: 10.1016/j.immuni.2018.03.025] [Citation(s) in RCA: 65] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2017] [Revised: 01/16/2018] [Accepted: 03/22/2018] [Indexed: 12/12/2022]
Abstract
It is unclear how quiescence is enforced in naive T cells, but activation by foreign antigens and self-antigens is allowed, despite the presence of inhibitory signals. We showed that active transforming growth factor β (TGF-β) signaling was present in naive T cells, and T cell receptor (TCR) engagement reduced TGF-β signaling during T cell activation by downregulating TGF-β type 1 receptor (TβRI) through activation of caspase recruitment domain-containing protein 11 (CARD11) and nuclear factor κB (NF-κB). TGF-β prevented TCR-mediated TβRI downregulation, but this was abrogated by interleukin-6 (IL-6). Mitigation of TCR-mediated TβRI downregulation through overexpression of TβRI in naive and activated T cells rendered T cells less responsive and suppressed autoimmunity. Naive T cells in autoimmune patients exhibited reduced TβRI expression and increased TCR-driven proliferation compared to healthy subjects. Thus, TCR-mediated regulation of TβRI-TGF-β signaling acts as a crucial criterion to determine T cell quiescence and activation.
Collapse
Affiliation(s)
- Eric Tu
- Mucosal Immunology Section, NIDCR, NIH, Bethesda, MD 20892, USA
| | - Cheryl P Z Chia
- Mucosal Immunology Section, NIDCR, NIH, Bethesda, MD 20892, USA
| | - Weiwei Chen
- Department of Rheumatology and Immunology, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China
| | - Dunfang Zhang
- Mucosal Immunology Section, NIDCR, NIH, Bethesda, MD 20892, USA
| | - Sang A Park
- Mucosal Immunology Section, NIDCR, NIH, Bethesda, MD 20892, USA
| | - Wenwen Jin
- Mucosal Immunology Section, NIDCR, NIH, Bethesda, MD 20892, USA
| | - Dandan Wang
- Department of Rheumatology and Immunology, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China
| | | | - Ying E Zhang
- Laboratory of Cellular and Molecular Biology, Center for Cancer Research, NCI, NIH, Bethesda, MD 20892, USA
| | - Lingyun Sun
- Department of Rheumatology and Immunology, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China.
| | - WanJun Chen
- Mucosal Immunology Section, NIDCR, NIH, Bethesda, MD 20892, USA.
| |
Collapse
|
47
|
Yazdani R, Shapoori S, Rezaeepoor M, Sanaei R, Ganjalikhani-Hakemi M, Azizi G, Rae W, Aghamohammadi A, Rezaei N. Features and roles of T helper 9 cells and interleukin 9 in immunological diseases. Allergol Immunopathol (Madr) 2019; 47:90-104. [PMID: 29703631 DOI: 10.1016/j.aller.2018.02.003] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2017] [Revised: 01/28/2018] [Accepted: 02/09/2018] [Indexed: 02/08/2023]
Abstract
T helper 9 (TH9) cells are considered as newly classified helper T cells that have an important role in the regulation of immune responses. Since these cells preferentially produce IL-9, these cells are termed TH9 cells. Recently, the role of TH9 and its signature cytokine (IL-9) has been investigated in a wide range of diseases, including autoimmunity, allergy, infections, cancer and immunodeficiency. Herein, we review the most recent data concerning TH9 cells and IL-9 as well as their roles in disease. These insights suggest that TH9 cells are a future target for therapeutic intervention.
Collapse
Affiliation(s)
- R Yazdani
- Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Science, Tehran, Iran
| | - S Shapoori
- Department of Immunology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - M Rezaeepoor
- Department of Immunology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - R Sanaei
- Department of Immunology, School of Medicine, Iran University of Medical Science, Tehran, Iran
| | - M Ganjalikhani-Hakemi
- Department of Immunology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran.
| | - G Azizi
- Non-Communicable Diseases Research Center, Alborz University of Medical Sciences, Karaj, Iran; Department of Laboratory Medicine, Imam Hassan Mojtaba Hospital, Alborz University of Medical Sciences, Karaj, Iran
| | - W Rae
- Department of Immunology, MP8, University Hospital Southampton NHS Foundation Trust, Tremona Road, Southampton, Hampshire SO16 6YD, UK
| | - A Aghamohammadi
- Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Science, Tehran, Iran
| | - N Rezaei
- Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Science, Tehran, Iran; Department of Immunology, School of Medicine, Tehran University of Medical Science, Tehran, Iran; Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, Iran.
| |
Collapse
|
48
|
王 进, 董 晓, 朱 新, 赵 华, 毛 德, 赵 鑫. [Expression of interleukin-9 in colon cancer tissues and its clinical significance]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2018; 38:943-948. [PMID: 30187869 PMCID: PMC6744043 DOI: 10.3969/j.issn.1673-4254.2018.08.07] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 04/01/2018] [Indexed: 12/12/2022]
Abstract
OBJECTIVE To investigate the expression of interleukin-9 (IL-9) in colon cancer tissues and its clinical significance. METHODS Immunohistochenmistry and qRT-PCR were used to detect the expressions of IL-9 protein and mRNA in 92 colon cancer tissues and paired adjacent normal tissues. The correlation of IL-9 expressions with the clinicopathological features and prognosis of the patients was analyzed. RESULTS IL-9 protein and mRNA expressions were significantly higher in adjacent normal tissues than in the colon cancer tissues (P < 0.001). In colon cancer patients, IL-9 expression was significantly correlated with TNM stage (P=0.013), Ducks stage (P=0.025) and lymph node metastasis (P=0.004) but not with gender, age, tumor size, differentiation or hepatic metastasis (P > 0.05). The survival time of colon cancer patients with positive IL-9 expression was significantly longer than that of patients negative for IL-9 expression (P=0.015). CONCLUSIONS IL-9 expression is lowered in colon cancer tissues compoved with in the adjacent normal tissues. IL-9 expression is negatively correlated with TNM staging, Ducks staging and lymph node metastasis but positively with good prognosis, suggesting its important role in the tumor microenvironment of colon cancer.
Collapse
Affiliation(s)
- 进 王
- />苏州大学附属第一医院普外科,江苏 苏州 215006Department of General Surgery, First Affiliated Hospital of Soochow University, Suzhou 215006, China
| | - 晓强 董
- />苏州大学附属第一医院普外科,江苏 苏州 215006Department of General Surgery, First Affiliated Hospital of Soochow University, Suzhou 215006, China
| | - 新国 朱
- />苏州大学附属第一医院普外科,江苏 苏州 215006Department of General Surgery, First Affiliated Hospital of Soochow University, Suzhou 215006, China
| | - 华 赵
- />苏州大学附属第一医院普外科,江苏 苏州 215006Department of General Surgery, First Affiliated Hospital of Soochow University, Suzhou 215006, China
| | - 德利 毛
- />苏州大学附属第一医院普外科,江苏 苏州 215006Department of General Surgery, First Affiliated Hospital of Soochow University, Suzhou 215006, China
| | - 鑫 赵
- />苏州大学附属第一医院普外科,江苏 苏州 215006Department of General Surgery, First Affiliated Hospital of Soochow University, Suzhou 215006, China
| |
Collapse
|
49
|
Li XF, Ling K, Yan XJ. Significance of expression of ID-1, ID-3, and NF-κB in colorectal adenocarcinoma. Shijie Huaren Xiaohua Zazhi 2018; 26:1307-1312. [DOI: 10.11569/wcjd.v26.i21.1307] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM To detect the expression of inhibitor of differentiation/DNA binding (ID)-1, ID-3, and nuclear factor-kappa B (NF-κB) in colorectal adenocarcinoma and to analyze their clinical significance.
METHODS Eighty-eight colorectal adenocarcinoma tissues, 43 colorectal high-grade intraepithelial neoplasia tissues, and 34 normal colonic mucosal tissues (>5 cm away from the edge of tumor) were collected. Expression of ID-1, ID-3, and NF-κB in these tissue samples was detected by immunochemistry.
RESULTS Expression of ID-1, ID-3, and NF-κB differed significantly between colorectal adenocarcinoma tissues and control tissues. Expression of ID-1, ID-3, and NF-κB was correlated with proliferation index and lesion depth. Expression of ID-1 and ID-3 was correlated with tumor differentiation. Expression of NF-κB was correlated with metastasis. There was a positive correlation between ID-1 and ID-3 expression in colorectal adenocarcinoma tissues.
CONCLUSION High expression of ID-1, ID-3 and NF-κB can promote the formation and progression of colorectal adenocarcinoma. ID-1 and ID-3 may have a synergistic effect.
Collapse
Affiliation(s)
- Xue-Feng Li
- Department of Internal Medicine, the Second People Hospital of Fuyang District, Hangzhou 311404, Zhejiang Province, China
| | - Kai Ling
- Department of Gastroenterology, the Second People Hospital of Fuyang District, Hangzhou 311404, Zhejiang Province, China
| | - Xiao-Jun Yan
- Department of Gastroenterology, the Second People Hospital of Fuyang District, Hangzhou 311404, Zhejiang Province, China
| |
Collapse
|
50
|
Shaw TN, Houston SA, Wemyss K, Bridgeman HM, Barbera TA, Zangerle-Murray T, Strangward P, Ridley AJL, Wang P, Tamoutounour S, Allen JE, Konkel JE, Grainger JR. Tissue-resident macrophages in the intestine are long lived and defined by Tim-4 and CD4 expression. J Exp Med 2018; 215:1507-1518. [PMID: 29789388 PMCID: PMC5987925 DOI: 10.1084/jem.20180019] [Citation(s) in RCA: 278] [Impact Index Per Article: 39.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2018] [Revised: 03/29/2018] [Accepted: 05/02/2018] [Indexed: 12/23/2022] Open
Abstract
Intestinal macrophages represent the last tissue macrophages thought to entirely adhere to van Furth's decades-old continuous monocyte replenishment model. In this study, Shaw et al. identify a population of intestinal macrophages that are long lived and maintained independently of monocyte replenishment over long periods of time. A defining feature of resident gut macrophages is their high replenishment rate from blood monocytes attributed to tonic commensal stimulation of this site. In contrast, almost all other tissues contain locally maintained macrophage populations, which coexist with monocyte-replenished cells at homeostasis. In this study, we identified three transcriptionally distinct mouse gut macrophage subsets that segregate based on expression of Tim-4 and CD4. Challenging current understanding, Tim-4+CD4+ gut macrophages were found to be locally maintained, while Tim-4–CD4+ macrophages had a slow turnover from blood monocytes; indeed, Tim-4–CD4– macrophages were the only subset with the high monocyte-replenishment rate currently attributed to gut macrophages. Moreover, all macrophage subpopulations required live microbiota to sustain their numbers, not only those derived from blood monocytes. These findings oppose the prevailing paradigm that all macrophages in the adult mouse gut rapidly turn over from monocytes in a microbiome-dependent manner; instead, these findings supplant it with a model of ontogenetic diversity where locally maintained subsets coexist with rapidly replaced monocyte-derived populations.
Collapse
Affiliation(s)
- Tovah N Shaw
- Manchester Collaborative Centre for Inflammation Research, University of Manchester, Manchester, England, UK.,School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, England, UK
| | - Stephanie A Houston
- Manchester Collaborative Centre for Inflammation Research, University of Manchester, Manchester, England, UK.,School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, England, UK
| | - Kelly Wemyss
- Manchester Collaborative Centre for Inflammation Research, University of Manchester, Manchester, England, UK.,School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, England, UK
| | - Hayley M Bridgeman
- Manchester Collaborative Centre for Inflammation Research, University of Manchester, Manchester, England, UK.,School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, England, UK
| | - Thomas A Barbera
- Manchester Collaborative Centre for Inflammation Research, University of Manchester, Manchester, England, UK.,School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, England, UK
| | - Tamsin Zangerle-Murray
- Manchester Collaborative Centre for Inflammation Research, University of Manchester, Manchester, England, UK.,School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, England, UK
| | - Patrick Strangward
- School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, England, UK
| | - Amanda J L Ridley
- Manchester Collaborative Centre for Inflammation Research, University of Manchester, Manchester, England, UK.,School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, England, UK
| | - Ping Wang
- School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, England, UK
| | - Samira Tamoutounour
- Mucosal Immunology Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
| | - Judith E Allen
- School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, England, UK.,Wellcome Trust Centre for Cell-Matrix Research, University of Manchester, Manchester, England, UK
| | - Joanne E Konkel
- Manchester Collaborative Centre for Inflammation Research, University of Manchester, Manchester, England, UK.,School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, England, UK
| | - John R Grainger
- Manchester Collaborative Centre for Inflammation Research, University of Manchester, Manchester, England, UK .,School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, England, UK
| |
Collapse
|