1
|
Ran R, Trapecar M, Brubaker DK. Systematic analysis of human colorectal cancer scRNA-seq revealed limited pro-tumoral IL-17 production potential in gamma delta T cells. Neoplasia 2024; 58:101072. [PMID: 39454432 PMCID: PMC11539345 DOI: 10.1016/j.neo.2024.101072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Revised: 10/10/2024] [Accepted: 10/10/2024] [Indexed: 10/28/2024]
Abstract
Gamma delta T cells play a crucial role in anti-tumor immunity due to their cytotoxic properties. However, the role and extent of γδ T cells in production of pro-tumorigenic interleukin-17 (IL-17) within the tumor microenvironment of colorectal cancer (CRC) remains controversial. In this study, we re-analyzed nine published human CRC whole-tissue single-cell RNA sequencing datasets, identifying 18,483 γδ T cells out of 951,785 total cells, in the neoplastic or adjacent normal tissue of 165 human CRC patients. Our results confirm that tumor-infiltrating γδ T cells exhibit high cytotoxicity-related transcription in both tumor and adjacent normal tissues, but critically, none of the γδ T cell clusters showed IL-17 production potential. We also identified various γδ T cell subsets, including poised effector-like T cells, tissue-resident memory T cells, progenitor exhausted-like T cells, and exhausted T cells, and noted an increased expression of cytotoxic molecules in tumor-infiltrating γδ T cells compared to their normal area counterparts. We proposed anti-tumor γδ T effector cells may arise from tissue-resident progenitor cells based on the trajectory analysis. Our work demonstrates that γδ T cells in CRC primarily function as cytotoxic effector cells rather than IL-17 producers, mitigating the concerns about their potential pro-tumorigenic roles in CRC, highlighting the importance of accurately characterizing these cells for cancer immunotherapy research and the unneglectable cross-species discrepancy between the mouse and human immune system in the study of cancer immunology.
Collapse
Affiliation(s)
- Ran Ran
- Center for Global Health and Diseases, Department of Pathology, Case Western Reserve University, Cleveland, OH, USA
| | - Martin Trapecar
- Department of Medicine, Johns Hopkins University School of Medicine, Institute for Fundamental Biomedical Research, Johns Hopkins All Children's Hospital, St. Petersburg, FL, USA; Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Douglas K Brubaker
- Center for Global Health and Diseases, Department of Pathology, Case Western Reserve University, Cleveland, OH, USA; The Blood, Heart, Lung, and Immunology Research Center, Case Western Reserve University, University Hospitals of Cleveland, Cleveland, OH, USA.
| |
Collapse
|
2
|
Zu H, Chen X. Epigenetics behind CD8 + T cell activation and exhaustion. Genes Immun 2024:10.1038/s41435-024-00307-1. [PMID: 39543311 DOI: 10.1038/s41435-024-00307-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Revised: 10/29/2024] [Accepted: 10/31/2024] [Indexed: 11/17/2024]
Abstract
CD8+ T cells play a critical role in specific immunity. In recent years, cell therapy has been emerging rapidly. The specific cytotoxic capabilities of these cells enable them to precisely identify and kill cells presenting specific antigens. This has demonstrated promise in the treatment of autoimmune diseases and cancers, with wide-ranging applications and value. However, in some diseases, such as tumors and chronic infections, T cells may adopt an exhausted phenotype, resulting in a loss of cytotoxicity and limiting their further application. Epigenetics plays a significant role in the differentiation and regulation of gene expression in cells. There is extensive evidence indicating that epigenetic remodeling plays an important role in T cell exhaustion. Therefore, further understanding its role in CD8+ T cell function can provide insights into the programmatic regulation of CD8+ T cells from a genetic perspective and overcome these diseases. We attempted to describe the relationship between the activation, function, and exhaustion mechanisms of CD8+ T cells, as well as epigenetics. This understanding makes it possible for us to address the aforementioned issues.
Collapse
Affiliation(s)
- Hao Zu
- Yanjing Medical College, Capital Medical University, 101300, Beijing, China
| | - Xiaoqin Chen
- Yanjing Medical College, Capital Medical University, 101300, Beijing, China.
| |
Collapse
|
3
|
Duan W, Zhou Z, Huang Y, Cui Y, Jin X, Liu R, Chen L. Euphorbia helioscopia L. inhibits lung tumorigenesis through alleviating exhausted T cell induced by chronic inflammation. JOURNAL OF ETHNOPHARMACOLOGY 2024; 338:119097. [PMID: 39537116 DOI: 10.1016/j.jep.2024.119097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Revised: 11/06/2024] [Accepted: 11/09/2024] [Indexed: 11/16/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Euphorbia helioscopia L. (ZQ) is a very effective traditional Chinese medicine for treating pneumonia and lung cancer. However, the effects and mechanisms by which ZQ prevents lung tumorigenesis in the presence of chronic inflammation remain unexplored. AIM To examine the effects and mechanisms of ZQ in alleviating chronic inflammation-induced T cell exhaustion and inhibiting lung tumorigenesis. METHODS A mice model of lung tumorigenesis under chronic inflammation conditions was established by repeated administration of lipopolysaccharide (LPS) and exposure to the tobacco carcinogen nitrosamine 4-(methylnitrosamino)-1-(3-pyridyl)-1-butanone (NNK). Mice were treated with ZQ ( 0.9, 1.8, and 3.6 g/kg/day) for 25 weeks. Lung pathology and tumor incidence were assessed, and inflammatory cytokine levels in bronchoalveolar lavage fluid (BALF) and serum were measured. Additionally, the proportions of CD3+ T, CD4+ T, and CD8+ T cells and their inhibitory receptors expression were evaluated. In vitro, T cell exhaustion models were induced using inflammatory-conditioned media, followed by treatment with ZQ (0.5, 2, 8 μg/mL). T cell exhaustion markers and characteristics were analyzed, and molecular mechanisms were explored using RNA sequencing and Immunoblotting analysis. RESULTS In vivo, ZQ significantly reduced inflammatory infiltration and lung damage, tumor incidence, number, size, and lung and spleen indices in mice. It also markedly lowered the levels of pro-inflammatory cytokines and immunosuppressive cytokines in BALF and serum. Additionally, ZQ improved the proportions of CD3+ T, CD4+ T, and CD8+ T cells and significantly decreased the expression of inhibitory receptors on CD4+ T and CD8+ T cells in the lung tissues and spleen. In vitro, ZQ effectively alleviated T cell exhaustion induced by the inflammatory environment, marked by reduced expression of inhibitory receptors, increased cytokine secretion, improved proliferation, and enhanced tumoricidal activity. RNA sequencing revealed that ZQ significantly downregulated the JAK-STAT signaling and upregulated stemness-associated pathways. Immunoblotting results indicated that treatment with ZQ markedly reduced the phosphorylation of Signal transducer and activator of transcription 3 (STAT3) and increased the expression of T cell factor -1/7 (TCF1/7). CONCLUSION ZQ inhibits lung tumorigenesis in LPS/NNK-treated mice through alleviating exhausted T cells induced by chronic inflammation, which is attributed to the suppression of STAT3 activation and the maintenance of stemness characteristics in T cells. These findings provide experimental evidence for the potential use of ZQ in preventing and treating lung tumourigenesis in patients with chronic inflammation and the clinical management of lung cancer patients with concomitant chronic inflammation.
Collapse
Affiliation(s)
- Wenbin Duan
- School of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang, 330004, China; National Pharmaceutical Engineering Center for Solid Preparation in Chinese Herbal Medicine, Jiangxi University of Chinese Medicine, Nanchang 330006, China; Key Laboratory for Evaluation on Anti-Tumor Effect of Chinese Medicine by Strengthening Body Resistance to Eliminate Pathogenic Factors, Nanchang 330006, China; Key Laboratory of Effective Material Basis of TCM, Jiangxi Province, Jiangxi University of Chinese Medicine, Nanchang 330006, China.
| | - Ziye Zhou
- School of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang, 330004, China.
| | - Yuqing Huang
- School of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang, 330004, China.
| | - Yaru Cui
- School of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang, 330004, China; National Pharmaceutical Engineering Center for Solid Preparation in Chinese Herbal Medicine, Jiangxi University of Chinese Medicine, Nanchang 330006, China; Key Laboratory of Effective Material Basis of TCM, Jiangxi Province, Jiangxi University of Chinese Medicine, Nanchang 330006, China.
| | - Xuhui Jin
- School of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang, 330004, China.
| | - Ronghua Liu
- School of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang, 330004, China.
| | - Lanying Chen
- School of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang, 330004, China; National Pharmaceutical Engineering Center for Solid Preparation in Chinese Herbal Medicine, Jiangxi University of Chinese Medicine, Nanchang 330006, China; Key Laboratory for Evaluation on Anti-Tumor Effect of Chinese Medicine by Strengthening Body Resistance to Eliminate Pathogenic Factors, Nanchang 330006, China; Key Laboratory of Effective Material Basis of TCM, Jiangxi Province, Jiangxi University of Chinese Medicine, Nanchang 330006, China.
| |
Collapse
|
4
|
Strobl K, Klufa J, Jin R, Artner-Gent L, Krauß D, Novoszel P, Strobl J, Stary G, Vujic I, Griss J, Holcmann M, Farlik M, Homey B, Sibilia M, Bauer T. JAK-STAT1 as therapeutic target for EGFR deficiency-associated inflammation and scarring alopecia. EMBO Mol Med 2024:10.1038/s44321-024-00166-3. [PMID: 39521937 DOI: 10.1038/s44321-024-00166-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Revised: 10/18/2024] [Accepted: 10/24/2024] [Indexed: 11/16/2024] Open
Abstract
The hair follicle stem cell niche is an immune-privileged microenvironment, characterized by reduced antigen presentation, thus shielding against permanent immune-mediated tissue damage. In this study, we demonstrated the protective role of hair follicle-specific epidermal growth factor receptor (EGFR) against scarring hair follicle destruction. Mechanistically, disruption of EGFR signaling generated a cell-intrinsic hypersensitivity within the JAK-STAT1 pathway, which, synergistically with interferon gamma expressing CD8 T-cell and NK-cell-mediated inflammation, compromised the stem cell niche. Hair follicle-specific genetic depletion of either JAK1/2 or STAT1 or therapeutic inhibition of JAK1/2 ameliorated the inflammation, restored skin barrier function and activated the residual stem cells to resume hair growth in mouse models of epidermal and hair follicle-specific EGFR deletion. Skin biopsies from EGFR inhibitor-treated and cicatricial alopecia patients revealed an active JAK-STAT1 signaling signature along with upregulation of antigen presentation and downregulation of key components of the EGFR pathway. Our findings offer molecular insights and highlight a mechanism-based therapeutic strategy for addressing chronic folliculitis associated with EGFR-inhibitor anti-cancer therapy and cicatricial alopecia.
Collapse
Affiliation(s)
- Karoline Strobl
- Center for Cancer Research, Medical University of Vienna and Comprehensive Cancer Center, Vienna, 1090, Austria
| | - Jörg Klufa
- Center for Cancer Research, Medical University of Vienna and Comprehensive Cancer Center, Vienna, 1090, Austria
| | - Regina Jin
- Center for Cancer Research, Medical University of Vienna and Comprehensive Cancer Center, Vienna, 1090, Austria
| | - Lena Artner-Gent
- Center for Cancer Research, Medical University of Vienna and Comprehensive Cancer Center, Vienna, 1090, Austria
| | - Dana Krauß
- Center for Cancer Research, Medical University of Vienna and Comprehensive Cancer Center, Vienna, 1090, Austria
| | - Philipp Novoszel
- Center for Cancer Research, Medical University of Vienna and Comprehensive Cancer Center, Vienna, 1090, Austria
| | - Johanna Strobl
- Department of Dermatology, Medical University of Vienna, Vienna, 1090, Austria
| | - Georg Stary
- Department of Dermatology, Medical University of Vienna, Vienna, 1090, Austria
| | - Igor Vujic
- Department of Dermatology, Venereology and Allergy, Clinical Center Landstrasse, Vienna, 1030, Austria
| | - Johannes Griss
- Department of Dermatology, Medical University of Vienna, Vienna, 1090, Austria
| | - Martin Holcmann
- Center for Cancer Research, Medical University of Vienna and Comprehensive Cancer Center, Vienna, 1090, Austria
| | - Matthias Farlik
- Department of Dermatology, Medical University of Vienna, Vienna, 1090, Austria
| | - Bernhard Homey
- Department of Dermatology, University Hospital Düsseldorf, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| | - Maria Sibilia
- Center for Cancer Research, Medical University of Vienna and Comprehensive Cancer Center, Vienna, 1090, Austria.
| | - Thomas Bauer
- Center for Cancer Research, Medical University of Vienna and Comprehensive Cancer Center, Vienna, 1090, Austria.
| |
Collapse
|
5
|
Li X, Liu Y, Gui J, Gan L, Xue J. Cell Identity and Spatial Distribution of PD-1/PD-L1 Blockade Responders. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2400702. [PMID: 39248327 PMCID: PMC11538707 DOI: 10.1002/advs.202400702] [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: 01/19/2024] [Revised: 07/08/2024] [Indexed: 09/10/2024]
Abstract
The programmed death 1 (PD-1)/programmed death ligand 1 (PD-L1) axis inhibits T cell activity, impairing anti-tumor immunity. Blocking this axis with therapeutic antibodies is one of the most promising anti-tumor immunotherapies. It has long been recognized that PD-1/PD-L1 blockade reinvigorates exhausted T (TEX) cells already present in the tumor microenvironment (TME). However, recent advancements in high-throughput gene sequencing and bioinformatic tools have provided researchers with a more granular and dynamic insight into PD-1/PD-L1 blockade-responding cells, extending beyond the TME and TEX populations. This review provides an update on the cell identity, spatial distribution, and treatment-induced spatiotemporal dynamics of PD-1/PD-L1 blockade responders. It also provides a synopsis of preliminary reports of potential PD-1/PD-L1 blockade responders other than T cells to depict a panoramic picture. Important questions to answer in further studies and the translational and clinical potential of the evolving understandings are also discussed.
Collapse
Affiliation(s)
- Xintong Li
- Division of Thoracic Tumor Multimodality TreatmentState Key Laboratory of Biotherapy and Cancer CenterNational Clinical Research Center for GeriatricsWest China HospitalSichuan UniversityChengdu610041China
| | - Yuanxin Liu
- Division of Thoracic Tumor Multimodality TreatmentState Key Laboratory of Biotherapy and Cancer CenterNational Clinical Research Center for GeriatricsWest China HospitalSichuan UniversityChengdu610041China
| | - Jun Gui
- State Key Laboratory of Systems Medicine for CancerRenji‐Med X Clinical Stem Cell Research CenterRen Ji HospitalShanghai Jiao Tong University School of MedicineShanghai200127China
| | - Lu Gan
- Research Laboratory of Emergency MedicineDepartment of Emergency MedicineNational Clinical Research Center for GeriatricsWest China HospitalSichuan UniversityChengdu610041China
| | - Jianxin Xue
- Division of Thoracic Tumor Multimodality TreatmentState Key Laboratory of Biotherapy and Cancer CenterNational Clinical Research Center for GeriatricsLaboratory of Clinical Cell TherapyWest China HospitalSichuan UniversityChengdu610041China
| |
Collapse
|
6
|
Tang Y, Chen Z, Zuo Q, Kang Y. Regulation of CD8+ T cells by lipid metabolism in cancer progression. Cell Mol Immunol 2024; 21:1215-1230. [PMID: 39402302 PMCID: PMC11527989 DOI: 10.1038/s41423-024-01224-z] [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: 07/25/2024] [Accepted: 09/22/2024] [Indexed: 11/02/2024] Open
Abstract
Dysregulation of lipid metabolism is a key characteristic of the tumor microenvironment, where tumor cells utilize lipids for proliferation, survival, metastasis, and evasion of immune surveillance. Lipid metabolism has become a critical regulator of CD8+ T-cell-mediated antitumor immunity, with excess lipids in the tumor microenvironment impeding CD8+ T-cell activities. Considering the limited efficacy of immunotherapy in many solid tumors, targeting lipid metabolism to enhance CD8+ T-cell effector functions could significantly improve immunotherapy outcomes. In this review, we examine recent findings on how lipid metabolic processes, including lipid uptake, synthesis, and oxidation, regulate CD8+ T cells within tumors. We also assessed the impact of different lipids on CD8+ T-cell-mediated antitumor immunity, with a particular focus on how lipid metabolism affects mitochondrial function in tumor-infiltrating CD8+ T cells. Furthermore, as cancer is a systemic disease, we examined systemic factors linking lipid metabolism to CD8+ T-cell effector function. Finally, we summarize current therapeutic approaches that target lipid metabolism to increase antitumor immunity and enhance immunotherapy. Understanding the molecular and functional interplay between lipid metabolism and CD8+ T cells offers promising therapeutic opportunities for cancer treatment.
Collapse
Affiliation(s)
- Yong Tang
- Department of Molecular Biology, Princeton University, Princeton, NJ, 08544, USA
- Ludwig Institute for Cancer Research Princeton Branch, Princeton, NJ, 08544, USA
| | - Ziqing Chen
- Department of Molecular Biology, Princeton University, Princeton, NJ, 08544, USA
- Ludwig Institute for Cancer Research Princeton Branch, Princeton, NJ, 08544, USA
| | - Qianying Zuo
- Department of Molecular Biology, Princeton University, Princeton, NJ, 08544, USA
- Ludwig Institute for Cancer Research Princeton Branch, Princeton, NJ, 08544, USA
| | - Yibin Kang
- Department of Molecular Biology, Princeton University, Princeton, NJ, 08544, USA.
- Ludwig Institute for Cancer Research Princeton Branch, Princeton, NJ, 08544, USA.
- Cancer Metabolism and Growth Program, Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, 08903, USA.
| |
Collapse
|
7
|
Wang Q, Yin X, Huang X, Zhang L, Lu H. Impact of mitochondrial dysfunction on the antitumor effects of immune cells. Front Immunol 2024; 15:1428596. [PMID: 39464876 PMCID: PMC11502362 DOI: 10.3389/fimmu.2024.1428596] [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: 05/08/2024] [Accepted: 09/24/2024] [Indexed: 10/29/2024] Open
Abstract
Mitochondrial dysfunction, a hallmark of immune cell failure, affects the antitumor effects of immune cells through metabolic reprogramming, fission, fusion, biogenesis, and immune checkpoint signal transduction of mitochondria. According to researchers, restoring damaged mitochondrial function can enhance the efficacy of immune cells. Nevertheless, the mechanism of mitochondrial dysfunction in immune cells in patients with cancer is unclear. In this review, we recapitulate the impact of mitochondrial dysfunction on the antitumor effects of T cells, natural killer cells, dendritic cells, and tumor-associated macrophage and propose that targeting mitochondria can provide new strategies for antitumor therapy.
Collapse
Affiliation(s)
- Quan Wang
- Department of Radiation Oncology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Xiangzhi Yin
- Department of Orthopaedics, Affiliated Hospital of Qingdao University, Qingdao, China
| | - Xiaotong Huang
- Department of Radiation Oncology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Lu Zhang
- Department of Radiation Oncology, The Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Haijun Lu
- Department of Radiation Oncology, The Affiliated Hospital of Qingdao University, Qingdao, China
| |
Collapse
|
8
|
Wang Y, Jia J, Wang F, Fang Y, Yang Y, Zhou Q, Yuan W, Gu X, Hu J, Yang S. Pre-metastatic niche: formation, characteristics and therapeutic implication. Signal Transduct Target Ther 2024; 9:236. [PMID: 39317708 PMCID: PMC11422510 DOI: 10.1038/s41392-024-01937-7] [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: 02/28/2024] [Revised: 06/29/2024] [Accepted: 07/23/2024] [Indexed: 09/26/2024] Open
Abstract
Distant metastasis is a primary cause of mortality and contributes to poor surgical outcomes in cancer patients. Before the development of organ-specific metastasis, the formation of a pre-metastatic niche is pivotal in promoting the spread of cancer cells. This review delves into the intricate landscape of the pre-metastatic niche, focusing on the roles of tumor-derived secreted factors, extracellular vesicles, and circulating tumor cells in shaping the metastatic niche. The discussion encompasses cellular elements such as macrophages, neutrophils, bone marrow-derived suppressive cells, and T/B cells, in addition to molecular factors like secreted substances from tumors and extracellular vesicles, within the framework of pre-metastatic niche formation. Insights into the temporal mechanisms of pre-metastatic niche formation such as epithelial-mesenchymal transition, immunosuppression, extracellular matrix remodeling, metabolic reprogramming, vascular permeability and angiogenesis are provided. Furthermore, the landscape of pre-metastatic niche in different metastatic organs like lymph nodes, lungs, liver, brain, and bones is elucidated. Therapeutic approaches targeting the cellular and molecular components of pre-metastatic niche, as well as interventions targeting signaling pathways such as the TGF-β, VEGF, and MET pathways, are highlighted. This review aims to enhance our understanding of pre-metastatic niche dynamics and provide insights for developing effective therapeutic strategies to combat tumor metastasis.
Collapse
Affiliation(s)
- Yuhang Wang
- Department of Colorectal Surgery, The First Affiliated Hospital of Zhengzhou University, 1 East Jianshe Road, Zhengzhou, 450000, China
| | - Jiachi Jia
- College of Medicine, Zhengzhou University, Zhengzhou, 450001, China
| | - Fuqi Wang
- Department of Colorectal Surgery, The First Affiliated Hospital of Zhengzhou University, 1 East Jianshe Road, Zhengzhou, 450000, China
| | - Yingshuai Fang
- College of Medicine, Zhengzhou University, Zhengzhou, 450001, China
| | - Yabing Yang
- College of Medicine, Zhengzhou University, Zhengzhou, 450001, China
| | - Quanbo Zhou
- Department of Colorectal Surgery, The First Affiliated Hospital of Zhengzhou University, 1 East Jianshe Road, Zhengzhou, 450000, China
| | - Weitang Yuan
- Department of Colorectal Surgery, The First Affiliated Hospital of Zhengzhou University, 1 East Jianshe Road, Zhengzhou, 450000, China
| | - Xiaoming Gu
- Department of Colorectal Surgery, The First Affiliated Hospital of Zhengzhou University, 1 East Jianshe Road, Zhengzhou, 450000, China.
| | - Junhong Hu
- Department of Colorectal Surgery, The First Affiliated Hospital of Zhengzhou University, 1 East Jianshe Road, Zhengzhou, 450000, China.
| | - Shuaixi Yang
- Department of Colorectal Surgery, The First Affiliated Hospital of Zhengzhou University, 1 East Jianshe Road, Zhengzhou, 450000, China.
| |
Collapse
|
9
|
Zhang G, Chen X, Yu C, Cui L, Chen N, Yi G, Wang S, Wei H, Liang Y, Ye S, Zhou Y. FNIP1 suppresses colorectal cancer progression through inhibiting STAT3 phosphorylation and nuclear translocation. iScience 2024; 27:110730. [PMID: 39262790 PMCID: PMC11389550 DOI: 10.1016/j.isci.2024.110730] [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: 12/09/2023] [Revised: 06/09/2024] [Accepted: 08/12/2024] [Indexed: 09/13/2024] Open
Abstract
Folliculin interacting protein 1 (FNIP1) primarily participates in regulating cellular energy metabolism and is associated with Birt-Hogg-Dubé (BHD) syndrome. Although FNIP1 has been demonstrated to function as both a tumor suppressor and promoter, its role in colorectal cancer (CRC) remains unclear. Our study demonstrated a significant downregulation of FNIP1 in CRC, correlating with shorter overall and disease-specific survival. FNIP1 may potentially serve as an independent prognostic factor in CRC. Moreover, FNIP1 inhibited CRC progression in vitro and in vivo. Mechanistically, FNIP1 bound to phosphorylated signal transducer and activator of transcription-3 (p-STAT3) and downregulated its expression. FNIP1 deletion increased STAT3 phosphorylation and nuclear localization, thereby promoting CRC progression. The use of p-STAT3-specific chemical inhibitors successfully mitigated excessive tumorigenesis resulting from FNIP1 absence. Thus, our results suggest that FNIP1 hinders CRC progression by suppressing STAT3 phosphorylation and nuclear translocation. FNIP1 may be a candidate prognostic indicator and a therapeutic target for intervention in CRC.
Collapse
Affiliation(s)
- Guixia Zhang
- Department of Gastroenterology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524001, China
| | - Xintian Chen
- Department of Gastroenterology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524001, China
| | - Caiyuan Yu
- Department of Gastroenterology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524001, China
| | - Lijiao Cui
- Department of Gastroenterology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524001, China
| | - Ningning Chen
- Department of Gastroenterology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524001, China
| | - Guanrong Yi
- Department of Gastroenterology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524001, China
| | - Shan Wang
- Department of Gastroenterology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524001, China
| | - Haiyun Wei
- Department of Gastroenterology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524001, China
| | - Youxin Liang
- Department of Gastroenterology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524001, China
| | - Shicai Ye
- Department of Gastroenterology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524001, China
| | - Yu Zhou
- Department of Gastroenterology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524001, China
| |
Collapse
|
10
|
Li W, Hua S, Yang J, Cao Y, Gao R, Sun H, Yang K, Wang Y, Peng P. Investigating immune dysregulation and hub genes in septic cardiomyopathy development. Sci Rep 2024; 14:21608. [PMID: 39294340 PMCID: PMC11411067 DOI: 10.1038/s41598-024-72724-1] [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: 03/21/2023] [Accepted: 09/10/2024] [Indexed: 09/20/2024] Open
Abstract
Septic cardiomyopathy is a life-threatening heart dysfunction caused by severe infection. Considering the complexity of pathogenesis and high mortality, the identification of efficient biomarkers are needed to guide clinical practice. Based on multimicroarray analysis, this study aimed to explore the pathogenesis of septic cardiomyopathy and the related immune landscape. The results showed that septic cardiomyopathy resulted in organ dysfunction due to extreme pro- and anti-inflammatory effects. In this process, KLRG1, PRF1, BCL6, GAB2, MMP9, IL1R1, JAK3, IL6ST, and SERPINE1 were identified as the hub genes regulating the immune landscape of septic cardiomyopathy. Nine transcription factors regulated the expression of these genes: SRF, STAT1, SP1, RELA, PPARG, NFKB1, PPARA, SMAD3, and STAT3. The hub genes activated the Th17 cell differentiation pathway, JAK-STAT signaling pathway, and cytokine‒cytokine receptor interaction pathway. These pathways were mainly involved in regulating the inflammatory response, adaptive immune response, leukocyte-mediated immunity, cytokine-mediated immunity, immune effector processes, myeloid cell differentiation, and T-helper cell differentiation. These nine hub genes could be considered biomarkers for the early prediction of septic cardiomyopathy.
Collapse
Affiliation(s)
- Wenli Li
- Emergency Trauma Center, The First Affiliated Hospital of Xinjiang Medical University, No. 137, Liyushan South Road, Urumqi, 830011, Xinjiang, People's Republic of China
| | - Shi Hua
- Department of Neurosurgery, Linyi People's Hospital, Linyi, People's Republic of China
| | - Jianzhong Yang
- Emergency Trauma Center, The First Affiliated Hospital of Xinjiang Medical University, No. 137, Liyushan South Road, Urumqi, 830011, Xinjiang, People's Republic of China
| | - Yang Cao
- Emergency Trauma Center, The First Affiliated Hospital of Xinjiang Medical University, No. 137, Liyushan South Road, Urumqi, 830011, Xinjiang, People's Republic of China
| | - Ranran Gao
- Emergency Trauma Center, The First Affiliated Hospital of Xinjiang Medical University, No. 137, Liyushan South Road, Urumqi, 830011, Xinjiang, People's Republic of China
| | - Hu Sun
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xinjiang Medical University, Ürümqi, People's Republic of China
| | - Kai Yang
- Emergency Trauma Center, The First Affiliated Hospital of Xinjiang Medical University, No. 137, Liyushan South Road, Urumqi, 830011, Xinjiang, People's Republic of China
| | - Ying Wang
- Medical Department, The First Affiliated Hospital of Xinjiang Medical University, Ürümqi, People's Republic of China
| | - Peng Peng
- Emergency Trauma Center, The First Affiliated Hospital of Xinjiang Medical University, No. 137, Liyushan South Road, Urumqi, 830011, Xinjiang, People's Republic of China.
| |
Collapse
|
11
|
Lopresti L, Tatangelo V, Baldari CT, Patrussi L. Rewiring the T cell-suppressive cytokine landscape of the tumor microenvironment: a new frontier for precision anti-cancer therapy. Front Immunol 2024; 15:1418527. [PMID: 39281678 PMCID: PMC11392891 DOI: 10.3389/fimmu.2024.1418527] [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: 04/16/2024] [Accepted: 08/06/2024] [Indexed: 09/18/2024] Open
Abstract
T lymphocytes that infiltrate the tumor microenvironment (TME) often fail to function as effective anti-cancer agents. Within the TME, cell-to-cell inhibitory interactions play significant roles in dampening their anti-tumor activities. Recent studies have revealed that soluble factors released in the TME by immune and non-immune cells, as well as by tumor cells themselves, contribute to the exacerbation of T cell exhaustion. Our understanding of the cytokine landscape of the TME, their interrelationships, and their impact on cancer development is still at its early stages. In this review, we aim to shed light on Interleukin (IL) -6, IL-9, and IL-10, a small group of JAK/STAT signaling-dependent cytokines harboring T cell-suppressive effects in the TME and summarize their mechanisms of action. Additionally, we will explore how advancements in scientific research can help us overcoming the obstacles posed by cytokines that suppress T cells in tumors, with the ultimate objective of stimulating further investigations for the development of novel therapeutic strategies to counteract their tumor-promoting activities.
Collapse
Affiliation(s)
| | | | | | - Laura Patrussi
- Department of Life Sciences, University of Siena, Siena, Italy
| |
Collapse
|
12
|
Yuan L, Meng Y, Xiang J. KLF4 Induces Colorectal Cancer by Promoting EMT via STAT3 Activation. Dig Dis Sci 2024; 69:2841-2855. [PMID: 38816600 DOI: 10.1007/s10620-024-08473-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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2024] [Accepted: 05/01/2024] [Indexed: 06/01/2024]
Abstract
OBJECTIVE Krüppel-like factor 4 (KLF4) has been demonstrated to exert a pro-carcinogenic effect in solid tissues. However, the precise biological function and underlying mechanisms in colorectal cancer (CRC) remains elucidated. AIMS To investigate whether KLF4 participates in the proliferation and invasion of CRC. METHODS The expression of KLF4 was investigated using immunohistochemistry and immunoblotting. The clinical significance of KLF4 was evaluated. Furthermore, the effect of inhibiting or overexpressing KLF4 on tumor was examined. Immunoblotting and qPCR were used to detect Epithelial-mesenchymal transition-related proteins levels. Additionally, the molecular function of KLF4 is related to the STAT3 signaling pathway and was determined through JASPAR, GSEA analysis, and in vitro experiments. RESULTS KLF4 exhibits down-regulated expression in CRC and is part of the vessel invasion, TNM stage, and worse prognosis. In vitro studies have shown that KLF4 promotes cellular proliferation and invasion, as well as EMT processes. Xenograft tumor models confirmed the oncogenic role of KLF4 in nude mice. Furthermore, GSEA and JASPAR databases analysis reveal that the binding of KLF4 to the signal transducer and activator of transcription 3 (STAT3) promoter site induces activation of p-STAT3 signaling. Subsequent targeting of STAT3 confirmed its pivotal role in mediating the oncogenic effects exerted by KLF4. CONCLUSION The study suggests that KLF4 activates STAT3 signaling, inducing epithelial-mesenchymal transition, thereby promoting CRC progression.
Collapse
Affiliation(s)
- Lebin Yuan
- Department of Nail and Breast Surgery, Affiliated Xiangyang Central Hospital of Hubei University of Arts and Science, Xiangyang Center Hospital, Xiangyang, Hubei, China
| | - Yanqiu Meng
- Oncology Department, First Affiliated Hospital of Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Jiajia Xiang
- Molecular Centre Laboratory, The Second Affiliated Hospital of Nanchang University, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China.
| |
Collapse
|
13
|
Ran R, Trapecar M, Brubaker DK. Systematic Analysis of Human Colorectal Cancer scRNA-seq Revealed Limited Pro-tumoral IL-17 Production Potential in Gamma Delta T Cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.18.604156. [PMID: 39071278 PMCID: PMC11275756 DOI: 10.1101/2024.07.18.604156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/30/2024]
Abstract
Gamma delta (γδ) T cells play a crucial role in anti-tumor immunity due to their cytotoxic properties. However, the role and extent of γδ T cells in production of pro-tumorigenic interleukin- 17 (IL-17) within the tumor microenvironment (TME) of colorectal cancer (CRC) remains controversial. In this study, we re-analyzed nine published human CRC whole-tissue single-cell RNA sequencing (scRNA-seq) datasets, identifying 18,483 γδ T cells out of 951,785 total cells, in the neoplastic or adjacent normal tissue of 165 human CRC patients. Our results confirm that tumor-infiltrating γδ T cells exhibit high cytotoxicity-related transcription in both tumor and adjacent normal tissues, but critically, none of the γδ T cell clusters showed IL-17 production potential. We also identified various γδ T cell subsets, including Teff, TRM, Tpex, and Tex, and noted an increased expression of cytotoxic molecules in tumor-infiltrating γδ T cells compared to their normal area counterparts. Our work demonstrates that γδ T cells in CRC primarily function as cytotoxic effector cells rather than IL-17 producers, mitigating the concerns about their potential pro-tumorigenic roles in CRC, highlighting the importance of accurately characterizing these cells for cancer immunotherapy research and the unneglectable cross-species discrepancy between the mouse and human immune system in the study of cancer immunology.
Collapse
Affiliation(s)
- Ran Ran
- Center for Global Health and Diseases, Department of Pathology, Case Western Reserve University, Cleveland, OH
| | - Martin Trapecar
- Department of Medicine, Johns Hopkins University School of Medicine, Institute for Fundamental Biomedical Research, Johns Hopkins All Children’s Hospital, St. Petersburg, FL, USA
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Douglas K. Brubaker
- Center for Global Health and Diseases, Department of Pathology, Case Western Reserve University, Cleveland, OH
- The Blood, Heart, Lung, and Immunology Research Center, Case Western Reserve University, University Hospitals of Cleveland, Cleveland, OH
| |
Collapse
|
14
|
van der Meijs NL, Travecedo MA, Marcelo F, van Vliet SJ. The pleiotropic CLEC10A: implications for harnessing this receptor in the tumor microenvironment. Expert Opin Ther Targets 2024; 28:601-612. [PMID: 38946482 DOI: 10.1080/14728222.2024.2374743] [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: 02/06/2024] [Accepted: 06/27/2024] [Indexed: 07/02/2024]
Abstract
INTRODUCTION CLEC10A is a C-type lectin receptor that specifically marks the conventional dendritic cell subsets two and three (cDC2 and DC3). It has a unique recognition profile of glycan antigens, with terminal N-Acetylgalactosamine residues that are frequently present in the tumor microenvironment. Even though CLEC10A expression allows for precise targeting of cDC2 and DC3 for the treatment of cancer, CLEC10A signaling has also been associated with anti-inflammatory responses that would promote tumor growth. AREAS COVERED Here, we review the potential benefits and drawbacks of CLEC10A engagement in the tumor microenvironment. We discuss the CLEC10A-mediated effects in different cell types and incorporate the pleiotropic effects of IL-10, the main anti-inflammatory response upon CLEC10A binding. EXPERT OPINION To translate this to a successful CLEC10A-mediated immunotherapy with limited tumor-promoting capacities, finding the right ligand presentation and adjuvant combination will be key.
Collapse
Affiliation(s)
- Nadia L van der Meijs
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC Location Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Amsterdam Institute for Infection and Immunology, Inflammatory Diseases, Amsterdam, The Netherlands
| | - Maria Alejandra Travecedo
- UCIBIO - Applied Molecular Biosciences Unit, Department of Chemistry, NOVA School of Science and Technology, NOVA University Lisbon, Caparica, Portugal
- Associate Laboratory i4HB - Institute for Health and Bioeconomy, NOVA School of Science and Technology, NOVA University Lisbon, Caparica, Portugal
| | - Filipa Marcelo
- UCIBIO - Applied Molecular Biosciences Unit, Department of Chemistry, NOVA School of Science and Technology, NOVA University Lisbon, Caparica, Portugal
- Associate Laboratory i4HB - Institute for Health and Bioeconomy, NOVA School of Science and Technology, NOVA University Lisbon, Caparica, Portugal
| | - Sandra J van Vliet
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC Location Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Amsterdam Institute for Infection and Immunology, Inflammatory Diseases, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Cancer Biology and Immunology, Amsterdam, The Netherlands
| |
Collapse
|
15
|
Shi H, Chen S, Chi H. Immunometabolism of CD8 + T cell differentiation in cancer. Trends Cancer 2024; 10:610-626. [PMID: 38693002 PMCID: PMC11342304 DOI: 10.1016/j.trecan.2024.03.010] [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: 02/04/2024] [Revised: 03/19/2024] [Accepted: 03/21/2024] [Indexed: 05/03/2024]
Abstract
CD8+ cytotoxic T lymphocytes (CTLs) are central mediators of tumor immunity and immunotherapies. Upon tumor antigen recognition, CTLs differentiate from naive/memory-like toward terminally exhausted populations with more limited function against tumors. Such differentiation is regulated by both immune signals, including T cell receptors (TCRs), co-stimulation, and cytokines, and metabolism-associated processes. These immune signals shape the metabolic landscape via signaling, transcriptional and post-transcriptional mechanisms, while metabolic processes in turn exert spatiotemporal effects to modulate the strength and duration of immune signaling. Here, we review the bidirectional regulation between immune signals and metabolic processes, including nutrient uptake and intracellular metabolic pathways, in shaping CTL differentiation and exhaustion. We also discuss the mechanisms underlying how specific nutrient sources and metabolite-mediated signaling events orchestrate CTL biology. Understanding how metabolic programs and their interplay with immune signals instruct CTL differentiation and exhaustion is crucial to uncover tumor-immune interactions and design novel immunotherapies.
Collapse
Affiliation(s)
- Hao Shi
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Sidi Chen
- Department of Genetics, Yale University School of Medicine, New Haven, CT, USA; System Biology Institute, Integrated Science & Technology Center, West Haven, CT, USA.
| | - Hongbo Chi
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, USA.
| |
Collapse
|
16
|
Ma Z, Zhou F, Jin H, Wu X. Crosstalk between CXCL12/CXCR4/ACKR3 and the STAT3 Pathway. Cells 2024; 13:1027. [PMID: 38920657 PMCID: PMC11201928 DOI: 10.3390/cells13121027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Revised: 06/09/2024] [Accepted: 06/10/2024] [Indexed: 06/27/2024] Open
Abstract
The reciprocal modulation between the CXCL12/CXCR4/ACKR3 axis and the STAT3 signaling pathway plays a crucial role in the progression of various diseases and neoplasms. Activation of the CXCL12/CXCR4/ACKR3 axis triggers the STAT3 pathway through multiple mechanisms, while the STAT3 pathway also regulates the expression of CXCL12. This review offers a thorough and systematic analysis of the reciprocal regulatory mechanisms between the CXCL12/CXCR4/ACKR3 signaling axis and the STAT3 signaling pathway in the context of diseases, particularly tumors. It explores the potential clinical applications in tumor treatment, highlighting possible therapeutic targets and novel strategies for targeted tumor therapy.
Collapse
Affiliation(s)
| | | | | | - Xiaoming Wu
- Laboratory of Molecular Genetics of Aging & Tumor, Medical School, Kunming University of Science and Technology, Chenggong Campus, 727 South Jingming Road, Kunming 650500, China; (Z.M.); (F.Z.); (H.J.)
| |
Collapse
|
17
|
Ahn T, Bae EA, Seo H. Decoding and overcoming T cell exhaustion: Epigenetic and transcriptional dynamics in CAR-T cells against solid tumors. Mol Ther 2024; 32:1617-1627. [PMID: 38582965 PMCID: PMC11184340 DOI: 10.1016/j.ymthe.2024.04.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2023] [Revised: 02/14/2024] [Accepted: 04/03/2024] [Indexed: 04/08/2024] Open
Abstract
T cell exhaustion, which is observed in various chronic infections and malignancies, is characterized by elevated expression of multiple inhibitory receptors, impaired effector functions, decreased proliferation, and reduced cytokine production. Notably, while adoptive T cell therapies, such as chimeric antigen receptor (CAR)-T therapy, have shown promise in treating cancer and other diseases, the efficacy of these therapies is often compromised by T cell exhaustion. It is imperative, therefore, to understand the mechanisms underlying this exhaustion to promote advances in T cell-related therapies. Here, we divided exhausted T cells into three distinct subsets according to their developmental and functional profiles: stem-like progenitor cells, intermediately exhausted cells, and terminally exhausted cells. These subsets are carefully regulated by synergistic mechanisms that involve transcriptional and epigenetic modulators. Key transcription factors, such as TCF1, BACH2, and TOX, are crucial for defining and sustaining exhaustion phenotypes. Concurrently, epigenetic regulators, such as TET2 and DNMT3A, shape the chromatin dynamics that direct T cell fate. The interplay of these molecular drivers has recently been highlighted in CAR-T research, revealing promising therapeutic directions. Thus, a profound understanding of exhausted T cell hierarchies and their molecular complexities may reveal innovative and improved tumor treatment strategies.
Collapse
Affiliation(s)
- Taeyoung Ahn
- Laboratory of Cell & Gene Therapy, Institute of Pharmaceutical Sciences, Seoul National University, Seoul 08826, Republic of Korea
| | - Eun-Ah Bae
- Laboratory of Immunology, Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, and College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea
| | - Hyungseok Seo
- Laboratory of Cell & Gene Therapy, Institute of Pharmaceutical Sciences, Seoul National University, Seoul 08826, Republic of Korea.
| |
Collapse
|
18
|
Zhang Y, Huang X, Yu M, Zhang M, Zhao L, Yan Y, Zhang L, Wang X. The integrate profiling of single-cell and spatial transcriptome RNA-seq reveals tumor heterogeneity, therapeutic targets, and prognostic subtypes in ccRCC. Cancer Gene Ther 2024; 31:917-932. [PMID: 38480978 DOI: 10.1038/s41417-024-00755-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Revised: 02/20/2024] [Accepted: 02/27/2024] [Indexed: 06/23/2024]
Abstract
Clear-cell renal cell carcinoma (ccRCC) is the most common type of RCC; however, the intratumoral heterogeneity in ccRCC remains unclear. We first identified markers and biological features of each cell cluster using bioinformatics analysis based on single-cell and spatial transcriptome RNA-sequencing data. We found that gene copy number loss on chromosome 3p and amplification on chromosome 5q were common features in ccRCC cells. Meanwhile, NNMT and HILPDA, which are associated with the response to hypoxia and metabolism, are potential therapeutic targets for ccRCC. In addition, CD8+ exhausted T cells (LAG3+ HAVCR2+), CD8+ proliferated T cells (STMN+), and M2-like macrophages (CD68+ CD163+ APOC1+), which are closely associated with immunosuppression, played vital roles in ccRCC occurrence and development. These results were further verified by whole exome sequencing, cell line and xenograft experiments, and immunofluorescence staining. Finally, we divide patients with ccRCC into three subtypes using unsupervised cluster analysis. and generated a classifier to reproduce these subtypes using the eXtreme Gradient Boosting algorithm. Our classifier can help clinicians evaluate prognosis and design personalized treatment strategies for ccRCC. In summary, our work provides a new perspective for understanding tumor heterogeneity and will aid in the design of antitumor therapeutic strategies for ccRCC.
Collapse
Affiliation(s)
- Yanlong Zhang
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, 100015, China
- Shanxi Medical University, Shanxi Bethune Hospital, Taiyuan, Shanxi, China
- Beijing Key Laboratory of Emerging Infectious Diseases, Institute of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, 100015, China
- Beijing Institute of Infectious Diseases, Beijing, 100015, China
- National Center for Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, 100015, China
- Department of Urology, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Xuefeng Huang
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, 100015, China
- Beijing Key Laboratory of Emerging Infectious Diseases, Institute of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, 100015, China
- Beijing Institute of Infectious Diseases, Beijing, 100015, China
- National Center for Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, 100015, China
| | - Minghang Yu
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, 100015, China
- Beijing Key Laboratory of Emerging Infectious Diseases, Institute of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, 100015, China
- Beijing Institute of Infectious Diseases, Beijing, 100015, China
- National Center for Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, 100015, China
| | - Menghan Zhang
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, 100015, China
| | - Li Zhao
- Shanxi Medical University, Shanxi Bethune Hospital, Taiyuan, Shanxi, China
| | - Yong Yan
- Department of Urology, Beijing Shijitan Hospital, Capital Medical University, Beijing, China.
| | - Liyun Zhang
- Shanxi Medical University, Shanxi Bethune Hospital, Taiyuan, Shanxi, China.
| | - Xi Wang
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, 100015, China.
- Beijing Key Laboratory of Emerging Infectious Diseases, Institute of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, 100015, China.
- Beijing Institute of Infectious Diseases, Beijing, 100015, China.
- National Center for Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, 100015, China.
| |
Collapse
|
19
|
Smith AL, Skupa SA, Eiken AP, Reznicek TE, Schmitz E, Williams N, Moore DY, D’Angelo CR, Kallam A, Lunning MA, Bociek RG, Vose JM, Mohamed E, Mahr AR, Denton PW, Powell B, Bollag G, Rowley MJ, El-Gamal D. BET inhibition reforms the immune microenvironment and alleviates T cell dysfunction in chronic lymphocytic leukemia. JCI Insight 2024; 9:e177054. [PMID: 38775157 PMCID: PMC11141939 DOI: 10.1172/jci.insight.177054] [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: 11/01/2023] [Accepted: 04/12/2024] [Indexed: 06/02/2024] Open
Abstract
Redundant tumor microenvironment (TME) immunosuppressive mechanisms and epigenetic maintenance of terminal T cell exhaustion greatly hinder functional antitumor immune responses in chronic lymphocytic leukemia (CLL). Bromodomain and extraterminal (BET) proteins regulate key pathways contributing to CLL pathogenesis and TME interactions, including T cell function and differentiation. Herein, we report that blocking BET protein function alleviates immunosuppressive networks in the CLL TME and repairs inherent CLL T cell defects. The pan-BET inhibitor OPN-51107 reduced exhaustion-associated cell signatures resulting in improved T cell proliferation and effector function in the Eμ-TCL1 splenic TME. Following BET inhibition (BET-i), TME T cells coexpressed significantly fewer inhibitory receptors (IRs) (e.g., PD-1, CD160, CD244, LAG3, VISTA). Complementary results were witnessed in primary CLL cultures, wherein OPN-51107 exerted proinflammatory effects on T cells, regardless of leukemic cell burden. BET-i additionally promotes a progenitor T cell phenotype through reduced expression of transcription factors that maintain terminal differentiation and increased expression of TCF-1, at least in part through altered chromatin accessibility. Moreover, direct T cell effects of BET-i were unmatched by common targeted therapies in CLL. This study demonstrates the immunomodulatory action of BET-i on CLL T cells and supports the inclusion of BET inhibitors in the management of CLL to alleviate terminal T cell dysfunction and potentially enhance tumoricidal T cell activity.
Collapse
Affiliation(s)
| | | | | | | | | | - Nolan Williams
- Eppley Institute for Research in Cancer and Allied Diseases
| | - Dalia Y. Moore
- Eppley Institute for Research in Cancer and Allied Diseases
| | - Christopher R. D’Angelo
- Division of Hematology and Oncology, Department of Internal Medicine, and
- Fred & Pamela Buffett Cancer Center (FPBCC), University of Nebraska Medical Center (UNMC), Omaha, Nebraska, USA
| | - Avyakta Kallam
- Division of Hematology and Oncology, Department of Internal Medicine, and
- Fred & Pamela Buffett Cancer Center (FPBCC), University of Nebraska Medical Center (UNMC), Omaha, Nebraska, USA
| | - Matthew A. Lunning
- Division of Hematology and Oncology, Department of Internal Medicine, and
- Fred & Pamela Buffett Cancer Center (FPBCC), University of Nebraska Medical Center (UNMC), Omaha, Nebraska, USA
| | - R. Gregory Bociek
- Division of Hematology and Oncology, Department of Internal Medicine, and
- Fred & Pamela Buffett Cancer Center (FPBCC), University of Nebraska Medical Center (UNMC), Omaha, Nebraska, USA
| | - Julie M. Vose
- Division of Hematology and Oncology, Department of Internal Medicine, and
- Fred & Pamela Buffett Cancer Center (FPBCC), University of Nebraska Medical Center (UNMC), Omaha, Nebraska, USA
| | - Eslam Mohamed
- College of Medicine and College of Graduate Studies, California Northstate University, Elk Grove, California, USA
| | - Anna R. Mahr
- Department of Biology, University of Nebraska at Omaha, Omaha, Nebraska, USA
| | - Paul W. Denton
- Department of Biology, University of Nebraska at Omaha, Omaha, Nebraska, USA
| | - Ben Powell
- Plexxikon Inc., South San Francisco, California, USA
| | | | | | - Dalia El-Gamal
- Eppley Institute for Research in Cancer and Allied Diseases
- Fred & Pamela Buffett Cancer Center (FPBCC), University of Nebraska Medical Center (UNMC), Omaha, Nebraska, USA
| |
Collapse
|
20
|
Zheng JM, Lou CX, Huang YL, Song WT, Luo YC, Mo GY, Tan LY, Chen SW, Li BJ. Associations between immune cell phenotypes and lung cancer subtypes: insights from mendelian randomization analysis. BMC Pulm Med 2024; 24:242. [PMID: 38755605 PMCID: PMC11100125 DOI: 10.1186/s12890-024-03059-w] [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: 01/02/2024] [Accepted: 05/10/2024] [Indexed: 05/18/2024] Open
Abstract
INTRODUCTION Lung cancer is a common malignant tumor, and different types of immune cells may have different effects on the occurrence and development of lung cancer subtypes, including lung squamous cell carcinoma (LUSC) and lung adenocarcinoma (LUAD). However, the causal relationship between immune phenotype and lung cancer is still unclear. METHODS This study utilized a comprehensive dataset containing 731 immune phenotypes from the European Bioinformatics Institute (EBI) to evaluate the potential causal relationship between immune phenotypes and LUSC and LUAD using the inverse variance weighted (IVW) method in Mendelian randomization (MR). Sensitivity analyses, including MR-Egger intercept, Cochran Q test, and others, were conducted for the robustness of the results. The study results were further validated through meta-analysis using data from the Transdisciplinary Research Into Cancer of the Lung (TRICL) data. Additionally, confounding factors were excluded to ensure the robustness of the findings. RESULTS Among the final selection of 729 immune cell phenotypes, three immune phenotypes exhibited statistically significant effects with LUSC. CD28 expression on resting CD4 regulatory T cells (OR 1.0980, 95% CI: 1.0627-1.1344, p < 0.0001) and CD45RA + CD28- CD8 + T cell %T cell (OR 1.0011, 95% CI: 1.0007; 1.0015, p < 0.0001) were associated with increased susceptibility to LUSC. Conversely, CCR2 expression on monocytes (OR 0.9399, 95% CI: 0.9177-0.9625, p < 0.0001) was correlated with a decreased risk of LUSC. However, no significant causal relationships were established between any immune cell phenotypes and LUAD. CONCLUSION This study demonstrates that specific immune cell types are associated with the risk of LUSC but not with LUAD. While these findings are derived solely from European populations, they still provide clues for a deeper understanding of the immunological mechanisms underlying lung cancer and may offer new directions for future therapeutic strategies and preventive measures.
Collapse
Affiliation(s)
- Jin-Min Zheng
- Department of Surgery, Guangxi Medical University, Nanning, Guangxi, China
| | - Chen-Xi Lou
- Department of Surgery, Guangxi University of Chinese Medicine, Nanning, Guangxi, China
| | - Yu-Liang Huang
- Department of Surgery, Guangxi Medical University, Nanning, Guangxi, China
| | - Wen-Tao Song
- Department of Surgery, Youjiang Medical University For Nationalities, Baise, Guangxi, China
| | - Yi-Chen Luo
- Department of thoracic surgery, Guangxi Academy of Medical Sciences and the People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi, China
| | - Guan-Yong Mo
- Department of thoracic surgery, Guilin Medical University, Guilin, Guangxi, China
| | - Lin-Yuan Tan
- Department of Surgery, Guangxi Medical University, Nanning, Guangxi, China
| | - Shang-Wei Chen
- Department of thoracic surgery, Guangxi Academy of Medical Sciences and the People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi, China.
| | - Bai-Jun Li
- Department of thoracic surgery, Tumor Hospital of Guangxi Medical University, Nanning, Guangxi, China.
| |
Collapse
|
21
|
Sharma P, Guo A, Poudel S, Boada-Romero E, Verbist KC, Palacios G, Immadisetty K, Chen MJ, Haydar D, Mishra A, Peng J, Madan Babu M, Krenciute G, Glazer ES, Green DR. Rapid metabolic regulation of a novel arginine methylation of KCa3.1 attenuates T cell exhaustion. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.09.593421. [PMID: 38798680 PMCID: PMC11118966 DOI: 10.1101/2024.05.09.593421] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
T cell exhaustion is linked to persistent antigen exposure and perturbed activation events, correlating with poor disease prognosis. Tumor-mediated T cell exhaustion is well documented; however, how the nutrient-deprived tumor niche affects T cell receptor (TCR) activation is largely unclear. We show that methionine metabolism licenses optimal TCR signaling by regulating the protein arginine methylome, and limiting methionine availability during early TCR signaling promotes subsequent T cell exhaustion. We discovered a novel arginine methylation of a Ca 2+ -activated potassium transporter, KCa3.1, prevention of which results in increased Ca 2+ -mediated NFAT1 activation, NFAT1 promoter occupancy, and T cell exhaustion. Furthermore, methionine supplementation reduces nuclear NFAT1 in tumor-infiltrating T cells and augments their anti-tumor activity. These findings demonstrate metabolic regulation of T cell exhaustion determined during TCR engagement.
Collapse
|
22
|
Zhu W, Zhang Z, Chen J, Chen X, Huang L, Zhang X, Huang X, Ma N, Xu W, Yi X, Lu X, Fu X, Li S, Mo G, Wang Y, Yuan G, Zang M, Li Q, Jiang X, He Y, Wu S, He Y, Li Y, Hou J. A novel engineered IL-21 receptor arms T-cell receptor-engineered T cells (TCR-T cells) against hepatocellular carcinoma. Signal Transduct Target Ther 2024; 9:101. [PMID: 38643203 PMCID: PMC11032311 DOI: 10.1038/s41392-024-01792-6] [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: 07/04/2023] [Revised: 01/30/2024] [Accepted: 03/07/2024] [Indexed: 04/22/2024] Open
Abstract
Strategies to improve T cell therapy efficacy in solid tumors such as hepatocellular carcinoma (HCC) are urgently needed. The common cytokine receptor γ chain (γc) family cytokines such as IL-2, IL-7, IL-15 and IL-21 play fundamental roles in T cell development, differentiation and effector phases. This study aims to determine the combination effects of IL-21 in T cell therapy against HCC and investigate optimized strategies to utilize the effect of IL-21 signal in T cell therapy. The antitumor function of AFP-specific T cell receptor-engineered T cells (TCR-T) was augmented by exogenous IL-21 in vitro and in vivo. IL-21 enhanced proliferation capacity, promoted memory differentiation, downregulated PD-1 expression and alleviated apoptosis in TCR-T after activation. A novel engineered IL-21 receptor was established, and TCR-T armed with the novel engineered IL-21 receptors (IL-21R-TCR-T) showed upregulated phosphorylated STAT3 expression without exogenous IL-21 ligand. IL-21R-TCR-T showed better proliferation upon activation and superior antitumor function in vitro and in vivo. IL-21R-TCR-T exhibited a less differentiated, exhausted and apoptotic phenotype than conventional TCR-T upon repetitive tumor antigen stimulation. The novel IL-21 receptor in our study programs powerful TCR-T and can avoid side effects induced by IL-21 systemic utilization. The novel IL-21 receptor creates new opportunities for next-generation TCR-T against HCC.
Collapse
Affiliation(s)
- Wei Zhu
- State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Zhiming Zhang
- State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Jinzhang Chen
- State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Xiaolan Chen
- State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Lei Huang
- Institute of Cellular Medicine, Newcastle University Medical School, Newcastle, UK
| | - Xiaoyong Zhang
- State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Xuan Huang
- State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Na Ma
- Department of Pathology, The First People's Hospital of Foshan, Foshan, China
| | - Weikang Xu
- State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Xuan Yi
- State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Dermatology Hospital, Southern Medical University, Guangzhou, China
| | - Xinyu Lu
- State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Xin Fu
- State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Siwei Li
- State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Guoheng Mo
- State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yiyue Wang
- State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Guosheng Yuan
- State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Mengya Zang
- State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Qi Li
- State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Xiaotao Jiang
- Department of Immunology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Yajing He
- State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Sha Wu
- Department of Immunology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
- Microbiome Medicine Center, Department of Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China
- Key Laboratory of Proteomics of Guangdong Province, Demonstration Center for Experimental Education of Basic Medical Sciences of China, Guangzhou, China
| | - Yukai He
- Medical College of Georgia, Augusta University, 1120 15th Street, Augusta, GA, USA
| | - Yongyin Li
- State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, China.
| | - Jinlin Hou
- State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, China.
| |
Collapse
|
23
|
Cui Z, Li H, Liu C, Wang J, Chen C, Hu S, Zhao X, Li G. Single-cell data revealed exhaustion of characteristic NK cell subpopulations and T cell subpopulations in hepatocellular carcinoma. Aging (Albany NY) 2024; 16:6550-6565. [PMID: 38604154 PMCID: PMC11042964 DOI: 10.18632/aging.205723] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Accepted: 03/13/2024] [Indexed: 04/13/2024]
Abstract
BACKGROUND The treatment and prognosis of patients with advanced hepatocellular carcinoma (HCC) have been a major medical challenge. Unraveling the landscape of tumor immune infiltrating cells (TIICs) in the immune microenvironment of HCC is of great significance to probe the molecular mechanisms. METHODS Based on single-cell data of HCC, the cell landscape was revealed from the perspective of TIICs. Special cell subpopulations were determined by the expression levels of marker genes. Differential expression analysis was conducted. The activity of each subpopulation was determined based on the highly expressed genes. CTLA4+ T-cell subpopulations affecting the prognosis of HCC were determined based on survival analysis. A single-cell regulatory network inference and clustering analysis was also performed to determine the transcription factor regulatory networks in the CTLA4+ T cell subpopulations. RESULTS 10 cell types were identified and NK cells and T cells showed high abundance in tumor tissues. Two NK cells subpopulations were present, FGFBP2+ NK cells, B3GNT7+ NK cells. Four T cells subpopulations were present, LAG3+ T cells, CTLA4+ T cells, RCAN3+ T cells, and HPGDS+ Th2 cells. FGFBP2+ NK cells, and CTLA4+ T cells were the exhaustive subpopulation. High CTLA4+ T cells contributed to poor prognostic outcomes and promoted tumor progression. Finally, a network of transcription factors regulated by NR3C1, STAT1, and STAT3, which were activated, was present in CTLA4+ T cells. CONCLUSION CTLA4+ T cell subsets in HCC exhibited functional exhaustion characteristics that probably inhibited T cell function through a transcription factor network dominated by NR3C1, STAT1, and STAT3.
Collapse
Affiliation(s)
- Zhongfeng Cui
- Department of Clinical Laboratory, Henan Provincial Infectious Disease Hospital, Zhengzhou 450000, China
| | - Hongzhi Li
- Department of Tuberculosis, Henan Provincial Infectious Disease Hospital, Zhengzhou 450000, China
| | - Chunli Liu
- Department of Infectious Diseases and Hepatology, Henan Provincial Infectious Disease Hospital, Zhengzhou 450000, China
| | - Juan Wang
- Department of Infectious Diseases and Hepatology, Henan Provincial Infectious Disease Hospital, Zhengzhou 450000, China
| | - Chunguang Chen
- Department of Clinical Laboratory, Henan Provincial Infectious Disease Hospital, Zhengzhou 450000, China
| | - Shanlei Hu
- Department of Infectious Diseases and Hepatology, Henan Provincial Infectious Disease Hospital, Zhengzhou 450000, China
| | - Xiaoli Zhao
- Department of Infectious Diseases and Hepatology, Henan Provincial Infectious Disease Hospital, Zhengzhou 450000, China
| | - Guangming Li
- Department of Infectious Diseases and Hepatology, Henan Provincial Infectious Disease Hospital, Zhengzhou 450000, China
| |
Collapse
|
24
|
Feng Y, An Q, Zhao Z, Wu M, Yang C, Liang W, Xu X, Jiang T, Zhang G. Beta-elemene: A phytochemical with promise as a drug candidate for tumor therapy and adjuvant tumor therapy. Biomed Pharmacother 2024; 172:116266. [PMID: 38350368 DOI: 10.1016/j.biopha.2024.116266] [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: 11/22/2023] [Revised: 02/06/2024] [Accepted: 02/06/2024] [Indexed: 02/15/2024] Open
Abstract
BACKGROUND β-Elemene (IUPAC name: (1 S,2 S,4 R)-1-ethenyl-1-methyl-2,4-bis(prop-1-en-2-yl) cyclohexane), is a natural compound found in turmeric root. Studies have demonstrated its diverse biological functions, including its anti-tumor properties, which have been extensively investigated. However, these have not yet been reviewed. The aim of this review was to provide a comprehensive summary of β-elemene research, with respect to disease treatment. METHODS β-Elemene-related articles were found in PubMed, ScienceDirect, and Google Scholar databases to systematically summarize its structure, pharmacokinetics, metabolism, and pharmacological activity. We also searched the Traditional Chinese Medicine System Pharmacology database for therapeutic targets of β-elemene. We further combined these targets with the relevant literature for KEGG and GO analyses. RESULTS Studies on the molecular mechanisms underlying β-elemene activity indicate that it regulates multiple pathways, including STAT3, MAPKs, Cyclin-dependent kinase 1/cyclin B, Notch, PI3K/AKT, reactive oxygen species, METTL3, PTEN, p53, FAK, MMP, TGF-β/Smad signaling. Through these molecular pathways, β-elemene has been implicated in tumor cell proliferation, apoptosis, migration, and invasion and improving the immune microenvironment. Additionally, β-elemene increases chemotherapeutic drug sensitivity and reverses resistance by inhibiting DNA damage repair and regulating pathways including CTR1, pak1, ERK1/2, ABC transporter protein, Prx-1 and ERCC-1. Nonetheless, owing to its lipophilicity and low bioavailability, additional structural modifications could improve the efficacy of this drug. CONCLUSION β-Elemene exhibits low toxicity with good safety, inhibiting various tumor types via diverse mechanisms in vivo and in vitro. When combined with chemotherapeutic drugs, it enhances efficacy, reduces toxicity, and improves tumor killing. Thus, β-elemene has vast potential for research and development.
Collapse
Affiliation(s)
- Yewen Feng
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Zhejiang 310053, China; Key Laboratory of Blood-stasis-toxin Syndrome of Zhejiang Province, Zhejiang 310053, China; Traditional Chinese Medicine "Preventing Disease" Wisdom Health Project Research Center of Zhejiang, Zhejiang 310053, China
| | - Qingwen An
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Zhejiang 310053, China; Key Laboratory of Blood-stasis-toxin Syndrome of Zhejiang Province, Zhejiang 310053, China; Traditional Chinese Medicine "Preventing Disease" Wisdom Health Project Research Center of Zhejiang, Zhejiang 310053, China
| | - Zhengqi Zhao
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Zhejiang 310053, China; Key Laboratory of Blood-stasis-toxin Syndrome of Zhejiang Province, Zhejiang 310053, China; Traditional Chinese Medicine "Preventing Disease" Wisdom Health Project Research Center of Zhejiang, Zhejiang 310053, China
| | - Mengting Wu
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Zhejiang 310053, China; Key Laboratory of Blood-stasis-toxin Syndrome of Zhejiang Province, Zhejiang 310053, China; Traditional Chinese Medicine "Preventing Disease" Wisdom Health Project Research Center of Zhejiang, Zhejiang 310053, China
| | - Chuqi Yang
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Zhejiang 310053, China; Key Laboratory of Blood-stasis-toxin Syndrome of Zhejiang Province, Zhejiang 310053, China; Traditional Chinese Medicine "Preventing Disease" Wisdom Health Project Research Center of Zhejiang, Zhejiang 310053, China
| | - WeiYu Liang
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Zhejiang 310053, China; Key Laboratory of Blood-stasis-toxin Syndrome of Zhejiang Province, Zhejiang 310053, China; Traditional Chinese Medicine "Preventing Disease" Wisdom Health Project Research Center of Zhejiang, Zhejiang 310053, China
| | - Xuefei Xu
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Zhejiang 310053, China; Key Laboratory of Blood-stasis-toxin Syndrome of Zhejiang Province, Zhejiang 310053, China; Traditional Chinese Medicine "Preventing Disease" Wisdom Health Project Research Center of Zhejiang, Zhejiang 310053, China
| | - Tao Jiang
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Zhejiang 310053, China; Key Laboratory of Blood-stasis-toxin Syndrome of Zhejiang Province, Zhejiang 310053, China; Traditional Chinese Medicine "Preventing Disease" Wisdom Health Project Research Center of Zhejiang, Zhejiang 310053, China.
| | - Guangji Zhang
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Zhejiang 310053, China; Key Laboratory of Blood-stasis-toxin Syndrome of Zhejiang Province, Zhejiang 310053, China; Traditional Chinese Medicine "Preventing Disease" Wisdom Health Project Research Center of Zhejiang, Zhejiang 310053, China.
| |
Collapse
|
25
|
Kagoya Y. Cytokine signaling in chimeric antigen receptor T-cell therapy. Int Immunol 2024; 36:49-56. [PMID: 37591521 PMCID: PMC10872714 DOI: 10.1093/intimm/dxad033] [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: 05/12/2023] [Accepted: 08/15/2023] [Indexed: 08/19/2023] Open
Abstract
Adoptive immunotherapy using chimeric antigen-receptor (CAR)-engineered T cells can induce robust antitumor responses against hematologic malignancies. However, its efficacy is not durable in the majority of the patients, warranting further improvement of T-cell functions. Cytokine signaling is one of the key cascades regulating T-cell survival and effector functions. In addition to cytokines that use the common γ chain as a receptor subunit, multiple cytokines regulate T-cell functions directly or indirectly. Modulating cytokine signaling in CAR-T cells by genetic engineering is one promising strategy to augment their therapeutic efficacy. These strategies include ectopic expression of cytokines, cytokine receptors, and synthetic molecules that mimic endogenous cytokine signaling. Alternatively, autocrine IL-2 signaling can be augmented through reprogramming of CAR-T cell properties through transcriptional and epigenetic modification. On the other hand, cytokine production by CAR-T cells triggers systemic inflammatory responses, which mainly manifest as adverse events such as cytokine-release syndrome (CRS) and neurotoxicity. In addition to inhibiting direct inflammatory mediators such as IL-6 and IL-1 released from activated macrophages, suppression of T-cell-derived cytokines associated with the priming of macrophages can be accomplished through genetic modification of CAR-T cells. In this review, I will outline recently developed synthetic biology approaches to exploit cytokine signaling to enhance CAR-T cell functions. I will also discuss therapeutic target molecules to prevent or alleviate CAR-T cell-related toxicities.
Collapse
Affiliation(s)
- Yuki Kagoya
- Division of Tumor Immunology, Institute for Advanced Medical Research, Keio University School of Medicine, 35 Shinanomachi, Shinjuku, Tokyo 160-8582, Japan
| |
Collapse
|
26
|
Díaz-Basabe A, Lattanzi G, Perillo F, Amoroso C, Baeri A, Farini A, Torrente Y, Penna G, Rescigno M, Ghidini M, Cassinotti E, Baldari L, Boni L, Vecchi M, Caprioli F, Facciotti F, Strati F. Porphyromonas gingivalis fuels colorectal cancer through CHI3L1-mediated iNKT cell-driven immune evasion. Gut Microbes 2024; 16:2388801. [PMID: 39132842 PMCID: PMC11321422 DOI: 10.1080/19490976.2024.2388801] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Revised: 07/19/2024] [Accepted: 07/31/2024] [Indexed: 08/13/2024] Open
Abstract
The interaction between the gut microbiota and invariant Natural Killer T (iNKT) cells plays a pivotal role in colorectal cancer (CRC). The pathobiont Fusobacterium nucleatum influences the anti-tumor functions of CRC-infiltrating iNKT cells. However, the impact of other bacteria associated with CRC, like Porphyromonas gingivalis, on their activation status remains unexplored. In this study, we demonstrate that mucosa-associated P. gingivalis induces a protumour phenotype in iNKT cells, subsequently influencing the composition of mononuclear-phagocyte cells within the tumor microenvironment. Mechanistically, in vivo and in vitro experiments showed that P. gingivalis reduces the cytotoxic functions of iNKT cells, hampering the iNKT cell lytic machinery through increased expression of chitinase 3-like-1 protein (CHI3L1). Neutralization of CHI3L1 effectively restores iNKT cell cytotoxic functions suggesting a therapeutic potential to reactivate iNKT cell-mediated antitumour immunity. In conclusion, our data demonstrate how P. gingivalis accelerates CRC progression by inducing the upregulation of CHI3L1 in iNKT cells, thus impairing their cytotoxic functions and promoting host tumor immune evasion.
Collapse
Affiliation(s)
- Angélica Díaz-Basabe
- Department of Experimental Oncology, European Institute of Oncology IRCCS, Milan, Italy
- Department of Oncology and Hemato-Oncology, Università degli Studi di Milano, Milan, Italy
| | - Georgia Lattanzi
- Department of Experimental Oncology, European Institute of Oncology IRCCS, Milan, Italy
- Gastroenterology and Endoscopy Unit, Fondazione IRCCS Cà Granda, Ospedale Maggiore Policlinico, Milan, Italy
| | - Federica Perillo
- Department of Experimental Oncology, European Institute of Oncology IRCCS, Milan, Italy
- Department of Oncology and Hemato-Oncology, Università degli Studi di Milano, Milan, Italy
| | - Chiara Amoroso
- Gastroenterology and Endoscopy Unit, Fondazione IRCCS Cà Granda, Ospedale Maggiore Policlinico, Milan, Italy
| | - Alberto Baeri
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Milan, Italy
| | - Andrea Farini
- Neurology Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Yvan Torrente
- Neurology Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Milan, Italy
- Centro Dino Ferrari, Department of Pathophysiology and Transplantation, Università degli Studi di Milano, Milano, Italy
| | - Giuseppe Penna
- IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| | - Maria Rescigno
- IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
- Department of Biomedical Sciences, Humanitas University, Milan, Italy
| | - Michele Ghidini
- Medical Oncology, Fondazione IRCCS Ca’ Granda, Ospedale Maggiore Policlinico, Milan, Italy
| | - Elisa Cassinotti
- Department of General and Minimally Invasive Surgery, Fondazione IRCCS Ca’ Granda, Ospedale Maggiore Policlinico, Milan, Italy
| | - Ludovica Baldari
- Department of General and Minimally Invasive Surgery, Fondazione IRCCS Ca’ Granda, Ospedale Maggiore Policlinico, Milan, Italy
| | - Luigi Boni
- Department of General and Minimally Invasive Surgery, Fondazione IRCCS Ca’ Granda, Ospedale Maggiore Policlinico, Milan, Italy
| | - Maurizio Vecchi
- Gastroenterology and Endoscopy Unit, Fondazione IRCCS Cà Granda, Ospedale Maggiore Policlinico, Milan, Italy
- Department of Pathophysiology and Transplantation, Università degli Studi di Milano, Milan, Italy
| | - Flavio Caprioli
- Gastroenterology and Endoscopy Unit, Fondazione IRCCS Cà Granda, Ospedale Maggiore Policlinico, Milan, Italy
- Department of Pathophysiology and Transplantation, Università degli Studi di Milano, Milan, Italy
| | - Federica Facciotti
- Department of Experimental Oncology, European Institute of Oncology IRCCS, Milan, Italy
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Milan, Italy
| | - Francesco Strati
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Milan, Italy
| |
Collapse
|
27
|
Cui Y, Wang K, Jiang D, Jiang Y, Shi D, DeGregori J, Waxman S, Ren R. Promoting longevity with less cancer: The 2022 International Conference on Aging and Cancer. AGING AND CANCER 2023; 4:111-120. [DOI: 10.1002/aac2.12068] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Accepted: 10/05/2023] [Indexed: 11/04/2024]
Abstract
AbstractAging and cancer are increasingly becoming big challenges for public health worldwide due to increased human life expectancy. Meanwhile, aging is one of the major risk factors for cancer. In December 2019, the first International Conference on Aging and Cancer was held in Haikou, Hainan province (island), China, preluding the establishment of the International Center for Aging and Cancer (ICAC) at Hainan, an institute dedicated to the research at the intersection of aging and cancer. Since then, the ICAC has hosted the annual conference each December in Hainan. The 2022 ICAC conference, with the theme of “promoting longevity with less cancer,” invited 17 internationally renowned scientists to share their new research and insights. Topics included DNA methylation in rejuvenation, development, and cellular senescence; lifespan regulation and longevity manipulation; metabolism and aging; cellular senescence and diseases; and novel therapeutics for cancer and antiaging/anticancer drug discovery. The forum highlighted the interconnectedness of aging and senescence with cancer evolution and risk. Although there is hope for preventing diseases like cancer by modulating systems that also control lifespan, attention has to be paid to the conflicting needs and competing demands in human biology.
Collapse
Affiliation(s)
- Yan Cui
- International Center for Aging and Cancer Hainan Medical University Haikou Hainan China
| | - Kai Wang
- International Center for Aging and Cancer Hainan Medical University Haikou Hainan China
| | - Danli Jiang
- International Center for Aging and Cancer Hainan Medical University Haikou Hainan China
| | - Yizhou Jiang
- International Center for Aging and Cancer Hainan Medical University Haikou Hainan China
| | - Dawei Shi
- International Center for Aging and Cancer Hainan Medical University Haikou Hainan China
| | - James DeGregori
- Department of Biochemistry and Molecular Genetics University of Colorado Anschutz Medical Campus Aurora Colorado USA
| | - Samuel Waxman
- Department of Hematology/Oncology Icahn School of Medicine at Mount Sinai New York City New York USA
| | - Ruibao Ren
- International Center for Aging and Cancer Hainan Medical University Haikou Hainan China
| |
Collapse
|
28
|
Nowicki TS, Peters CW, Quiros C, Kidd CK, Kawakami M, Klomhaus AM, Baselga-Carretero I, Kaplan-Lefko P, Macabali MH, Perez Garcilazo I, Berent-Maoz B, Comin-Anduix B, Ribas A. Infusion Product TNFα, Th2, and STAT3 Activities Are Associated with Clinical Responses to Transgenic T-cell Receptor Cell Therapy. Cancer Immunol Res 2023; 11:1589-1597. [PMID: 37871333 PMCID: PMC10702871 DOI: 10.1158/2326-6066.cir-23-0577] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Revised: 08/31/2023] [Accepted: 10/16/2023] [Indexed: 10/25/2023]
Abstract
Transgenic T-cell receptor (TCR) T cell-based adoptive cell therapies for solid tumors are associated with dramatic initial response rates, but there remain many instances of treatment failure and disease relapse. The association of infusion product cytokine profiles with clinical response has not been explored in the context of TCR T-cell therapy products. Single-cell antigen-dependent secretomic and proteomic analysis of preinfusion clinical TCR T-cell therapy products revealed that TNFα cytokine functionality of CD8+ T cells and phospho-STAT3 signaling in these cells were both associated with superior clinical responsiveness to therapy. By contrast, CD4+ T-helper 2 cell cytokine profiles were associated with inferior clinical responses. In parallel, preinfusion levels of IL15, Flt3-L, and CX3CL1 were all found to be associated with clinical response to therapy. These results have implications for the development of therapeutic biomarkers and identify potential targets for enrichment in the design of transgenic TCR T-cell therapies for solid tumors.
Collapse
Affiliation(s)
- Theodore S. Nowicki
- Division of Pediatric Hematology-Oncology, Department of Pediatrics, University of California Los Angeles, Los Angeles, California
- Department of Microbiology, Immunology, and Molecular Genetics, University of California Los Angeles, Los Angeles, California
- Jonsson Comprehensive Cancer Center, University of California Los Angeles, Los Angeles, California
- Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, University of California Los Angeles, Los Angeles, California
- Molecular Biology Institute, University of California Los Angeles, Los Angeles, California
| | - Cole W. Peters
- Division of Pediatric Hematology-Oncology, Department of Pediatrics, University of California Los Angeles, Los Angeles, California
| | - Crystal Quiros
- Division of Pediatric Hematology-Oncology, Department of Pediatrics, University of California Los Angeles, Los Angeles, California
| | - Conner K. Kidd
- Division of Pediatric Hematology-Oncology, Department of Pediatrics, University of California Los Angeles, Los Angeles, California
| | - Moe Kawakami
- Division of Pediatric Hematology-Oncology, Department of Pediatrics, University of California Los Angeles, Los Angeles, California
| | - Alexandra M. Klomhaus
- Department of General Internal Medicine and Health Services Research, University of California, Los Angeles, California
| | - Ignacio Baselga-Carretero
- Division of Hematology-Oncology, Department of Medicine, University of California Los Angeles, Los Angeles, California
| | - Paula Kaplan-Lefko
- Division of Hematology-Oncology, Department of Medicine, University of California Los Angeles, Los Angeles, California
| | - Mignonette H. Macabali
- Division of Hematology-Oncology, Department of Medicine, University of California Los Angeles, Los Angeles, California
| | - Ivan Perez Garcilazo
- Division of Hematology-Oncology, Department of Medicine, University of California Los Angeles, Los Angeles, California
| | - Beata Berent-Maoz
- Division of Hematology-Oncology, Department of Medicine, University of California Los Angeles, Los Angeles, California
| | - Begoña Comin-Anduix
- Jonsson Comprehensive Cancer Center, University of California Los Angeles, Los Angeles, California
- Division of Surgical Oncology, Department of Surgery, University of California Los Angeles, Los Angeles, California
| | - Antoni Ribas
- Jonsson Comprehensive Cancer Center, University of California Los Angeles, Los Angeles, California
- Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, University of California Los Angeles, Los Angeles, California
- Division of Hematology-Oncology, Department of Medicine, University of California Los Angeles, Los Angeles, California
- Division of Surgical Oncology, Department of Surgery, University of California Los Angeles, Los Angeles, California
- Department of Molecular and Medical Pharmacology, University of California Los Angeles, Los Angeles, California
| |
Collapse
|
29
|
A Heieis G, Everts B. O-GlcNAcylation at the center of antitumor immunity. Curr Opin Biotechnol 2023; 84:103009. [PMID: 37863017 DOI: 10.1016/j.copbio.2023.103009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Revised: 09/18/2023] [Accepted: 09/21/2023] [Indexed: 10/22/2023]
Abstract
The post-translational modification known as O-GlcNAcylation is a highly dysregulated process in tumors, and a key contributor to malignant transformation. In contrast, after three decades since its discovery, very little has been revealed about this process in the immune system. With the prospect of targeting O-GlcNAcylation as tumor therapy, greater understanding of how it regulates immune responses in the context of the tumor microenvironment will be needed. Here, we discuss recent discoveries from which a picture is emerging that O-GlcNAcylation, in either tumors or in immune cells, could negatively impact overall antitumor immune responses. We propose that interference with O-GlcNAcylation thus holds promise for cancer treatment from both perspectives.
Collapse
Affiliation(s)
- Graham A Heieis
- Department of Parasitology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, the Netherlands
| | - Bart Everts
- Department of Parasitology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, the Netherlands.
| |
Collapse
|
30
|
Sun Q, Cai D, Liu D, Zhao X, Li R, Xu W, Xie B, Gou M, Wei K, Li Y, Huang J, Chi X, Wei P, Hao J, Guo X, Pan B, Fu Y, Ni L, Dong C. BCL6 promotes a stem-like CD8 + T cell program in cancer via antagonizing BLIMP1. Sci Immunol 2023; 8:eadh1306. [PMID: 37862431 DOI: 10.1126/sciimmunol.adh1306] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2023] [Accepted: 08/30/2023] [Indexed: 10/22/2023]
Abstract
Overcoming CD8+ T cell exhaustion is critical in cancer immunotherapy. Recently, an intratumor stem/progenitor-like CD8+ T cell (Tprog cell) population that mediates the persistence of antitumor responses has been defined, which can further develop into a terminally differentiated CD8+ T cell (Tterm cell) subpopulation with potent cytotoxic functions. Tprog cells are the main responders to immune checkpoint blockade therapies, yet how extrinsic signals via transcription factors control Tprog cell generation and persistence in tumors is unclear. Here, we found that BCL6 inhibits tumor-specific Tterm cell generation from Tprog cell downstream of TCF1. We show that Bcl6 deficiency reduced the persistence of Tprog cells, without affecting their generation, thus abrogating long-term tumor control. High-level BCL6 expression was observed in tumor-specific T cells in draining lymph nodes (LNs) and was associated with T cell exhaustion. This was observed in TOX+TCF1+ Tprog cells in both LNs and tumors. BCL6 expression in CD8+ T cells was up-regulated by TGF-β-SMAD2 signaling but down-regulated by the IL-2-STAT5 pathway. Mechanistically, BCL6 transcriptionally repressed the expression of Tterm cell-associated genes and induced those of Tprog cell-related genes, in a manner antagonistic to BLIMP1. Prdm1 deficiency also promoted the Tprog cell program and greatly improved the efficacy of anti-PD-1 therapy. Thus, we identified the TGF-β-BCL6 and IL-2-BLIMP1 antagonistic pathways in regulation of antitumor CD8+ T cells, which may benefit the development of long-lasting and effective cancer immunotherapy.
Collapse
Affiliation(s)
- Qinli Sun
- Institute for Immunology and School of Medicine, Tsinghua University, Beijing 100084, China
| | - Dongli Cai
- Shanghai Immune Therapy Institute, New Cornerstone Science Laboratory, Shanghai Jiao Tong University School of Medicine-Affiliated Renji Hospital, Shanghai 200127, China
- Department of Gynaecology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai 201204, China
| | - Dingfeng Liu
- Shanghai Immune Therapy Institute, New Cornerstone Science Laboratory, Shanghai Jiao Tong University School of Medicine-Affiliated Renji Hospital, Shanghai 200127, China
- Department of Gynaecology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai 201204, China
| | - Xiaohong Zhao
- Institute for Immunology and School of Medicine, Tsinghua University, Beijing 100084, China
| | - Ruifeng Li
- Institute for Immunology and School of Medicine, Tsinghua University, Beijing 100084, China
- Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing 100084, China
| | - Wei Xu
- Institute for Immunology and School of Medicine, Tsinghua University, Beijing 100084, China
- Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing 100084, China
| | - Bowen Xie
- Institute for Immunology and School of Medicine, Tsinghua University, Beijing 100084, China
- Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing 100084, China
| | - Mengting Gou
- Shanghai Immune Therapy Institute, New Cornerstone Science Laboratory, Shanghai Jiao Tong University School of Medicine-Affiliated Renji Hospital, Shanghai 200127, China
| | - Kun Wei
- Institute for Immunology and School of Medicine, Tsinghua University, Beijing 100084, China
| | - Yuling Li
- Institute for Immunology and School of Medicine, Tsinghua University, Beijing 100084, China
- College of Life Science and Peking University-Tsinghua University-National Institute of Biological Sciences Joint Graduate Program, Tsinghua University, Beijing 100084, China
| | - Jinling Huang
- Institute for Immunology and School of Medicine, Tsinghua University, Beijing 100084, China
| | - Xinxin Chi
- Institute for Immunology and School of Medicine, Tsinghua University, Beijing 100084, China
| | - Peng Wei
- Institute for Immunology and School of Medicine, Tsinghua University, Beijing 100084, China
- Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing 100084, China
| | - Jing Hao
- Shanghai Immune Therapy Institute, New Cornerstone Science Laboratory, Shanghai Jiao Tong University School of Medicine-Affiliated Renji Hospital, Shanghai 200127, China
| | - Xinyi Guo
- Shanghai Immune Therapy Institute, New Cornerstone Science Laboratory, Shanghai Jiao Tong University School of Medicine-Affiliated Renji Hospital, Shanghai 200127, China
| | - Birui Pan
- Institute for Immunology and School of Medicine, Tsinghua University, Beijing 100084, China
| | - Yujie Fu
- Institute for Immunology and School of Medicine, Tsinghua University, Beijing 100084, China
- Shanghai Immune Therapy Institute, New Cornerstone Science Laboratory, Shanghai Jiao Tong University School of Medicine-Affiliated Renji Hospital, Shanghai 200127, China
| | - Ling Ni
- Institute for Immunology and School of Medicine, Tsinghua University, Beijing 100084, China
| | - Chen Dong
- Institute for Immunology and School of Medicine, Tsinghua University, Beijing 100084, China
- Shanghai Immune Therapy Institute, New Cornerstone Science Laboratory, Shanghai Jiao Tong University School of Medicine-Affiliated Renji Hospital, Shanghai 200127, China
- Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing 100084, China
- Research Unit of Immune Regulation and Immune Diseases of Chinese Academy of Medical Sciences, Shanghai Jiao Tong University School of Medicine-Affiliated Renji Hospital, Shanghai 200127, China
| |
Collapse
|
31
|
Chang Y, Jiang M, Wang Y, Fu Q, Lin S, Wu J, Di W. Erucic acid improves the progress of pregnancy complicated with systemic lupus erythematosus by inhibiting the effector function of CD8 + T cells. MedComm (Beijing) 2023; 4:e382. [PMID: 37771913 PMCID: PMC10522964 DOI: 10.1002/mco2.382] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Revised: 08/19/2023] [Accepted: 08/20/2023] [Indexed: 09/30/2023] Open
Abstract
Pathogenic CD8+ T cells are pivotal contributors to the onset of systemic lupus erythematosus (SLE). Erucic acid (EA) has been proven to have anti-inflammatory activity. However, the capacity of EA to regulate pathogenic CD8+ T cells in the context of pregnancy complicated with SLE (pSLE) remains unclear. In our investigation, we observed augmented CD8+ T cell effector function juxtaposed with diminished EA levels in pSLE patients relative to healthy pregnant controls. Significantly, plasma EA levels exhibited a negative correlation with the severity of pSLE-associated complications. In blood from patients with pSLE, EA inhibited the effector function of CD8+ T cells, concurrently dampening the maintenance of stem cell-like memory CD8+ T cells. Mechanistically, EA orchestrated the inhibition of CD8+ T cell effector function by impeding signal transducer and activator of transcription 3 phosphorylation and promoting ferroptosis. Moreover, EA supplementation in pregnant MRL/lpr mice manifested as the attenuation of uterine CD8+ T cell effector function, culminating in the mitigation of placental pathological damage. Our findings uncover the immune response modulatory effects of EA upon pathogenic CD8+ cells, thereby unveiling new perspectives for therapeutic strategies targeting pSLE patients.
Collapse
Affiliation(s)
- Yanling Chang
- Department of Obstetrics and GynecologyRenji HospitalSchool of MedicineShanghai Jiao Tong UniversityShanghaiChina
- Department of Obstetrics and GynecologyShanghai Key Laboratory of Gynecologic OncologyShanghaiChina
| | - Meng Jiang
- Department of Obstetrics and GynecologyRenji HospitalSchool of MedicineShanghai Jiao Tong UniversityShanghaiChina
- Department of Obstetrics and GynecologyShanghai Key Laboratory of Gynecologic OncologyShanghaiChina
| | - You Wang
- Department of Obstetrics and GynecologyRenji HospitalSchool of MedicineShanghai Jiao Tong UniversityShanghaiChina
- Department of Obstetrics and GynecologyShanghai Key Laboratory of Gynecologic OncologyShanghaiChina
| | - Qiong Fu
- Department of RheumatologyRenji HospitalSchool of MedicineShanghai Jiao Tong UniversityShanghaiChina
- Shanghai Institute of RheumatologyShanghaiChina
| | - Sihan Lin
- Department of Obstetrics and GynecologyRenji HospitalSchool of MedicineShanghai Jiao Tong UniversityShanghaiChina
- Department of Obstetrics and GynecologyShanghai Key Laboratory of Gynecologic OncologyShanghaiChina
| | - Jiayue Wu
- Department of Obstetrics and GynecologyRenji HospitalSchool of MedicineShanghai Jiao Tong UniversityShanghaiChina
- Department of Obstetrics and GynecologyShanghai Key Laboratory of Gynecologic OncologyShanghaiChina
| | - Wen Di
- Department of Obstetrics and GynecologyRenji HospitalSchool of MedicineShanghai Jiao Tong UniversityShanghaiChina
- Department of Obstetrics and GynecologyShanghai Key Laboratory of Gynecologic OncologyShanghaiChina
- Department of Obstetrics and Gynecology, State Key Laboratory of Oncogenes and Related GenesShanghai Cancer InstituteRenji HospitalSchool of MedicineShanghai Jiao Tong UniversityShanghaiChina
| |
Collapse
|
32
|
Jiang H, Ren Y, Yu J, Hu S, Zhang J. Analysis of lactate metabolism-related genes and their association with immune infiltration in septic shock via bioinformatics method. Front Genet 2023; 14:1223243. [PMID: 37564869 PMCID: PMC10410269 DOI: 10.3389/fgene.2023.1223243] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Accepted: 07/17/2023] [Indexed: 08/12/2023] Open
Abstract
Background: Lactate, as an essential clinical evaluation index of septic shock, is crucial in the incidence and progression of septic shock. This study aims to investigate the differential expression, regulatory relationship, clinical diagnostic efficacy, and immune infiltration of lactate metabolism-related genes (LMGs) in septic shock. Methods: Two sepsis shock datasets (GSE26440 and GSE131761) were screened from the GEO database, and the common differentially expressed genes (DEGs) of the two datasets were screened out. LMGs were selected from the GeneCards database, and lactate metabolism-related DEGs (LMDEGs) were determined by integrating DEGs and LMGs. Protein-protein interaction networks, mRNA-miRNA, mRNA-RBP, and mRNA-TF interaction networks were constructed using STRING, miRDB, ENCORI, and CHIPBase databases, respectively. Receiver operating characteristic (ROC) curves were constructed for each of the LMDEGs to evaluate the diagnostic efficacy of the expression changes in relation to septic shock. Finally, immune infiltration analysis was performed using ssGSEA and CIBERSORT. Results: This study identified 10 LMDEGs, including LDHB, STAT3, LDHA, GSR, FOXM1, PDP1, GCDH, GCKR, ABCC1, and CDKN3. Enrichment analysis revealed that DEGs were significantly enriched in pathways such as pyruvate metabolism, hypoxia pathway, and immune-inflammatory pathways. PPI networks based on LMDEGs, as well as 148 pairs of mRNA-miRNA interactions, 243 pairs of mRNA-RBP interactions, and 119 pairs of mRNA-TF interactions were established. ROC curves of eight LMDEGs (LDHA, GSR, STAT3, CDKN3, FOXM1, GCKR, PDP1, and LDHB) with consistent expression patterns in two datasets had an area under the curve (AUC) ranging from 0.662 to 0.889. The results of ssGSEA and CIBERSORT both showed significant differences in the infiltration of various immune cells, including CD8 T cells, T regulatory cells, and natural killer cells, and LMDEGs such as STAT3, LDHB, LDHA, PDP1, GSR, FOXM1, and CDKN3 were significantly associated with various immune cells. Conclusion: The LMDEGs are significantly associated with the immune-inflammatory response in septic shock and have a certain diagnostic accuracy for septic shock.
Collapse
Affiliation(s)
- Huimin Jiang
- Emergency Intensive Care Unit, Ningxiang People’s Hospital Affiliated to Hunan University of Chinese Medicine, Changsha, China
| | - Yun Ren
- Emergency Department, Ningxiang People’s Hospital Affiliated to Hunan University of Chinese Medicine, Changsha, China
| | - Jiale Yu
- Emergency Department, Ningxiang People’s Hospital Affiliated to Hunan University of Chinese Medicine, Changsha, China
| | - Sheng Hu
- Emergency Department, Ningxiang People’s Hospital Affiliated to Hunan University of Chinese Medicine, Changsha, China
| | - Jihui Zhang
- Emergency Intensive Care Unit, Ningxiang People’s Hospital Affiliated to Hunan University of Chinese Medicine, Changsha, China
- Emergency Department, Ningxiang People’s Hospital Affiliated to Hunan University of Chinese Medicine, Changsha, China
| |
Collapse
|
33
|
Zhang T, Shi Q, Gu H, Yu B, Yin S, Ge Q, Mo X, Liu X, Huang J. CCDC134 facilitates T cell activation through the regulation of early T cell receptor signaling. Front Immunol 2023; 14:1133111. [PMID: 37234153 PMCID: PMC10206301 DOI: 10.3389/fimmu.2023.1133111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Accepted: 04/28/2023] [Indexed: 05/27/2023] Open
Abstract
Modulation of surface T cell antigen receptor (TCR) expression is crucial for proper T cell development and maintenance of mature T cell function at steady state and upon stimulation. We previously determined that CCDC134 (coiled-coil domain containing 134), a cytokine-like molecule that served as a potential member of the γc cytokine family, contributes to antitumor responses by augmenting CD8+ T cell-mediated immunity. Here we show that T cell-specific deletion of Ccdc134 decreased peripheral mature CD4+ and CD8+ T cells, which resulted in impaired T cell homeostasis. Moreover, Ccdc134-deficient T cells exhibited an attenuated response to TCR stimulation in vitro, showing lower activation and proliferative capacity. This was further reflected in vivo, rendering mice refractory to T cell-mediated inflammatory and antitumor responses. More importantly, CCDC134 is associated with TCR signaling components, including CD3ϵ, and attenuated TCR signaling in Ccdc134-deficient T cells via altered CD3ϵ ubiquitination and degradation. Taken together, these findings suggest a role for CCDC134 as a positive regulator of TCR-proximal signaling and provide insight into the cell-intrinsic functional consequences of Ccdc134 deficiency in the attenuation of T cell-mediated inflammatory and antitumor responses.
Collapse
Affiliation(s)
- Tianzhuo Zhang
- Department of Immunology, School of Basic Medical Sciences, Peking University, and National Health Commission (NHC) Key Laboratory of Medical Immunology, Peking University, Beijing, China
| | - Qianwen Shi
- Department of Immunology, School of Basic Medical Sciences, Peking University, and National Health Commission (NHC) Key Laboratory of Medical Immunology, Peking University, Beijing, China
| | - Huining Gu
- Department of Immunology, School of Basic Medical Sciences, Peking University, and National Health Commission (NHC) Key Laboratory of Medical Immunology, Peking University, Beijing, China
| | - Biaoyi Yu
- Department of Immunology, School of Basic Medical Sciences, Peking University, and National Health Commission (NHC) Key Laboratory of Medical Immunology, Peking University, Beijing, China
| | - Sha Yin
- Department of Immunology, School of Basic Medical Sciences, Peking University, and National Health Commission (NHC) Key Laboratory of Medical Immunology, Peking University, Beijing, China
- Shaanxi Institute for Pediatric Diseases, Xi’an Key Laboratory of Children’s Health and Diseases, Xi’an Children’s Hospital, The Affiliated Children’s Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
| | - Qing Ge
- Department of Immunology, School of Basic Medical Sciences, Peking University, and National Health Commission (NHC) Key Laboratory of Medical Immunology, Peking University, Beijing, China
| | - Xiaoning Mo
- Department of Immunology, School of Basic Medical Sciences, Peking University, and National Health Commission (NHC) Key Laboratory of Medical Immunology, Peking University, Beijing, China
| | - Xiaofeng Liu
- Hepatopancreatobiliary Surgery Department I, Key laboratory of Carcinogenesis and Translational Research, Ministry of Education/Beijing, Peking University Cancer Hospital & Institute, Beijing, China
| | - Jing Huang
- Department of Immunology, School of Basic Medical Sciences, Peking University, and National Health Commission (NHC) Key Laboratory of Medical Immunology, Peking University, Beijing, China
| |
Collapse
|
34
|
Zhang S, Tu Q, Qian X, Wang J, Ma C, Yang L, Liu Y, Wu R, Liu M. Deficiency of Kif15 gene inhibits tumor growth due to host CD8 +T lymphocytes increase. Biochem Biophys Res Commun 2023; 655:110-117. [PMID: 36934586 DOI: 10.1016/j.bbrc.2023.03.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Revised: 03/02/2023] [Accepted: 03/02/2023] [Indexed: 03/11/2023]
Abstract
Kif15, also name kinesin-12, is a microtubule (MT) associate protein, which functions as a regulator of MT-dependent transport or spindle organization. Previous studies reported Kif15 increases in many tumors, however the effect of host Kif15 gene lack on tumor growth is not investigated. In this study, CRISPR/Cas9 mediated Kif15 gene knockout (Kif15-/-) mice were established and HE (Hematoxylin-Eosin) assay revealed no significant differences of morphology in most adult tissues (heart, liver, lung, kidney, and brain) except a retarded development of spleen in adult Kif15-/- mice. RNA sequence analysis of adult spleen tissues of Kif15-/- and Kif15+/+ mice was performed, and the results revealed that a total of 438 mRNAs were significantly differentially expressed in Kif15 knockout spleen, showing the top biological process was immune system process. FCM (Flow Cytometry) assay showed the percentage of CD8+ T lymphocytes notably increased in spleens of 9 w and 12 w old Kif15-/- mice. The CD8+ T lymphocytes are cytotoxic effector cells fighting against tumor. We thus detected the tumor growth in Kif15-/- mice using the melanoma cells inoculated subcutaneously. The tumor size significantly reduced in Kif15-/- mice. We finally detected whether Kif15 dysfunction affects the phagocytic function of macrophages on tumor cells, and the result showed Kif15 inhibitor treated macrophages significantly promoted the phagocytosis in vitro. In summary, this study revealed that the tumor-bearing mice of Kif15 gene deficiency notably inhibited tumor growth due to innate immune activation, which was the first report of the relation of Kif15 on the immunoreactivity.
Collapse
Affiliation(s)
- Siming Zhang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, China; Cancer Research Center Nantong, Nantong Tumor Hospital & Affiliated Tumor Hospital of Nantong University, Nantong, China
| | - Qifeng Tu
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, China
| | - Xiaowei Qian
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, China
| | - Junpei Wang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, China
| | - Chao Ma
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, China
| | - Liu Yang
- Department of Neurosurgery, Affiliated Hospital of Nantong University, Nantong, China
| | - Yan Liu
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, China
| | - Ronghua Wu
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, China.
| | - Mei Liu
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, China.
| |
Collapse
|