1
|
Liu J, Wu M, Yang Y, Mei X, Wang L, Wang J, Wang Z, He S, Liu H, Jiang H, Qu S, Zhang Y, Chen Y, Tian X, Huang Y, Wang H. BTN3A1 expressed in cervical cancer cells promotes Vγ9Vδ2 T cells exhaustion through upregulating transcription factors NR4A2/3 downstream of TCR signaling. Cell Commun Signal 2024; 22:459. [PMID: 39342337 PMCID: PMC11439235 DOI: 10.1186/s12964-024-01834-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Accepted: 09/17/2024] [Indexed: 10/01/2024] Open
Abstract
BACKGROUND Clinical trials have shown that immunotherapy based on Vγ9Vδ2 T cells (Vδ2 T cells) is safe and well-tolerated for various cancers including cervical cancer (CC), but its overall treatment efficacy remains limited. Therefore, exploring the mechanisms underlying the suboptimal efficacy of Vδ2 T cell-based cancer immunotherapy is crucial for enabling its successful clinical translation. METHODS Tumor samples from CC patients and CC cell line-derived xenograft (CDX) mice were analyzed using flow cytometry to examine the exhausted phenotype of tumor-infiltrating Vδ2 T cells. The interrelationship between BTN3A1 expression and Vδ2 T cells in CC, along with their correlation with patient prognosis, was analyzed using data from The Cancer Genome Atlas (TCGA) database. CC cell lines with BTN3A1 knockout (KO) and overexpression (OE) were constructed through lentivirus transduction, which were then co-cultured with expanded Vδ2 T cells, followed by detecting the function of Vδ2 T cells using flow cytometry. The pathways and transcription factors (TFs) related to BTN3A1-induced Vδ2 T cells exhaustion and the factors affecting BTN3A1 expression were identified by RNA-seq analysis, which was confirmed by flow cytometry, Western Blot, and gene manipulation. RESULTS Tumor-infiltrating Vδ2 T cells exhibited an exhausted phenotype in both CC patients and CDX mice. BTN3A1 expressed in CC is highly enhancing exhaustion markers, while reducing the secretion of effector molecules in Vδ2 T cells. Blocking TCR or knocking down nuclear receptor subfamily 4 group A (NR4A) 2/3 can reverse BTN3A1-induced exhaustion in Vδ2 T cells. On the other hand, IFN-γ secreted by Vδ2 T cells promoted the expression of BTN3A1 and PD-L1. CONCLUSIONS Through binding γδ TCRs, BTN3A1 expressed on tumor cells, which is induced by IFN-γ, can promote Vδ2 T cells to upregulate the expression of TFs NR4A2/3, thereby affecting their activation and expression of exhaustion-related molecules in the tumor microenvironment (TME). Therefore, targeting BTN3A1 might overcome the immunosuppressive effect of the TME on Vδ2 T cells in CC.
Collapse
MESH Headings
- Humans
- Uterine Cervical Neoplasms/genetics
- Uterine Cervical Neoplasms/pathology
- Uterine Cervical Neoplasms/immunology
- Uterine Cervical Neoplasms/metabolism
- Female
- Animals
- Up-Regulation
- Signal Transduction
- Mice
- Cell Line, Tumor
- Butyrophilins/genetics
- Butyrophilins/metabolism
- DNA-Binding Proteins/genetics
- DNA-Binding Proteins/metabolism
- Receptors, Thyroid Hormone/genetics
- Receptors, Thyroid Hormone/metabolism
- T-Lymphocytes/immunology
- T-Lymphocytes/metabolism
- Receptors, Antigen, T-Cell, gamma-delta/genetics
- Receptors, Antigen, T-Cell, gamma-delta/metabolism
- Gene Expression Regulation, Neoplastic
- Receptors, Steroid
Collapse
Affiliation(s)
- Jian Liu
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Min Wu
- Department of Gynecologic Oncology, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Yifan Yang
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xinyu Mei
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Liming Wang
- School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jingyu Wang
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zixuan Wang
- School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shan He
- School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hangyu Liu
- School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Han Jiang
- School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shen Qu
- School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yuwei Zhang
- School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ying Chen
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xun Tian
- Department of Obstetrics and Gynecology, Academician Expert Workstation, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430014, China.
| | - Yafei Huang
- Department of Pathogen Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan, China.
| | - Hui Wang
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
- Department of Gynecologic Oncology, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.
- Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
- Zhejiang Key Laboratory of Precision Diagnosis and Therapy for Major Gynecological Diseases, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.
| |
Collapse
|
2
|
Jiang Y, Wang Y, Chen G, Sun F, Wu Q, Huang Q, Zeng D, Qiu W, Wang J, Yao Z, Liang B, Li S, Wu J, Huang N, Wang Y, Chen J, Zhai X, Huang L, Xu B, Yamamoto M, Tsukamoto T, Nomura S, Liao W, Shi M. Nicotinamide metabolism face-off between macrophages and fibroblasts manipulates the microenvironment in gastric cancer. Cell Metab 2024; 36:1806-1822.e11. [PMID: 38897198 DOI: 10.1016/j.cmet.2024.05.013] [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: 10/12/2023] [Revised: 03/06/2024] [Accepted: 05/22/2024] [Indexed: 06/21/2024]
Abstract
Immune checkpoint blockade has led to breakthroughs in the treatment of advanced gastric cancer. However, the prominent heterogeneity in gastric cancer, notably the heterogeneity of the tumor microenvironment, highlights the idea that the antitumor response is a reflection of multifactorial interactions. Through transcriptomic analysis and dynamic plasma sample analysis, we identified a metabolic "face-off" mechanism within the tumor microenvironment, as shown by the dual prognostic significance of nicotinamide metabolism. Specifically, macrophages and fibroblasts expressing the rate-limiting enzymes nicotinamide phosphoribosyltransferase and nicotinamide N-methyltransferase, respectively, regulate the nicotinamide/1-methylnicotinamide ratio and CD8+ T cell function. Mechanistically, nicotinamide N-methyltransferase is transcriptionally activated by the NOTCH pathway transcription factor RBP-J and is further inhibited by macrophage-derived extracellular vesicles containing nicotinamide phosphoribosyltransferase via the SIRT1/NICD axis. Manipulating nicotinamide metabolism through autologous injection of extracellular vesicles restored CD8+ T cell cytotoxicity and the anti-PD-1 response in gastric cancer.
Collapse
Affiliation(s)
- Yu Jiang
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Yawen Wang
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Guofeng Chen
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Fei Sun
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Qijing Wu
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Qiong Huang
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Dongqiang Zeng
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Wenjun Qiu
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Jiao Wang
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Zhiqi Yao
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Bishan Liang
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Shaowei Li
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Jianhua Wu
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Na Huang
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Yuanyuan Wang
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Jingsong Chen
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Xiaohui Zhai
- Department of Medical Oncology, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Li Huang
- Department of Oncology, First Affiliated Hospital, Gannan Medical University, Ganzhou, China; Jiangxi Clinical Medical Research Center for Cancer, Ganzhou, China
| | - Beibei Xu
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Masami Yamamoto
- Laboratory of Physiological Pathology, School of Veterinary Nursing and Technology, Nippon Veterinary and Life Science University, Tokyo, Japan
| | - Tetsuya Tsukamoto
- Department of Diagnostic Pathology, Fujita Health University School of Medicine, Toyoake, Aichi, Japan
| | - Sachiyo Nomura
- Department of Gastrointestinal Surgery, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Wangjun Liao
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China; Cancer Center, the Sixth Affiliated Hospital, School of Medicine, South China University of Technology, Foshan, China.
| | - Min Shi
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China.
| |
Collapse
|
3
|
Wang Y, Zeng Y, Yang W, Wang X, Jiang J. Targeting CD8 + T cells with natural products for tumor therapy: Revealing insights into the mechanisms. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 129:155608. [PMID: 38642413 DOI: 10.1016/j.phymed.2024.155608] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/24/2024] [Revised: 03/27/2024] [Accepted: 04/07/2024] [Indexed: 04/22/2024]
Abstract
BACKGROUND Despite significant advances in cancer immunotherapy over the past decades, such as T cell-engaging chimeric antigen receptor (CAR)-T cell therapy and immune checkpoint blockade (ICB), therapeutic failure resulting from various factors remains prevalent. Therefore, developing combinational immunotherapeutic strategies is of great significance for improving the clinical outcome of cancer immunotherapy. Natural products are substances that naturally exist in various living organisms with multiple pharmacological or biological activities, and some of them have been found to have anti-tumor potential. Notably, emerging evidences have suggested that several natural compounds may boost the anti-tumor effects through activating immune response of hosts, in which CD8+ T cells play a pivotal role. METHODS The data of this review come from PubMed, Web of Science, Google Scholar, and ClinicalTrials (https://clinicaltrials.gov/) with the keywords "CD8+ T cell", "anti-tumor", "immunity", "signal 1", "signal 2", "signal 3", "natural products", "T cell receptor (TCR)", "co-stimulation", "co-inhibition", "immune checkpoint", "inflammatory cytokine", "hesperidin", "ginsenoside", "quercetin", "curcumin", "apigenin", "dendrobium officinale polysaccharides (DOPS)", "luteolin", "shikonin", "licochalcone A", "erianin", "resveratrol", "procyanidin", "berberine", "usnic acid", "naringenin", "6-gingerol", "ganoderma lucidum polysaccharide (GL-PS)", "neem leaf glycoprotein (NLGP)", "paclitaxel", "source", "pharmacological activities", and "toxicity". These literatures were published between 1993 and 2023. RESULTS Natural products have considerable advantages as anti-tumor drugs based on the various species, wide distribution, low price, and few side effects. This review summarized the effects and mechanisms of some natural products that exhibit anti-tumor effects via targeting CD8+ T cells, mainly focused on the three signals that activate CD8+ T cells: TCR, co-stimulation, and inflammatory cytokines. CONCLUSION Clarifying the role and underlying mechanism of natural products in cancer immunotherapy may provide more options for combinational treatment strategies and benefit cancer therapy, to shed light on identifying potential natural compounds for improving the clinical outcome in cancer immunotherapy.
Collapse
Affiliation(s)
- Yuke Wang
- West China School of Public Health and West China Fourth Hospital, West China School of Basic Medical Sciences & Forensic Medicine and State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, China; Department of Neurosurgery, Medical Research Center, The Third People's Hospital of Chengdu, The Affiliated Hospital of Southwest Jiaotong University, The Second Chengdu Hospital Affiliated to Chongqing Medical University, Chengdu, China
| | - Yan Zeng
- West China School of Public Health and West China Fourth Hospital, West China School of Basic Medical Sciences & Forensic Medicine and State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Wenyong Yang
- Department of Neurosurgery, Medical Research Center, The Third People's Hospital of Chengdu, The Affiliated Hospital of Southwest Jiaotong University, The Second Chengdu Hospital Affiliated to Chongqing Medical University, Chengdu, China
| | - Xiuxuan Wang
- Research and Development Department, Beijing DCTY Biotech Co., Ltd., Beijing, China
| | - Jingwen Jiang
- West China School of Public Health and West China Fourth Hospital, West China School of Basic Medical Sciences & Forensic Medicine and State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
4
|
Nguyen Ky M, Duran A, Hasantari I, Bru A, Deloire M, Brochet B, Ruet A, Schmitt N. Natalizumab Treatment Induces Proinflammatory CD4 T Cells Preferentially in the Integrin β7+ Compartment. NEUROLOGY(R) NEUROIMMUNOLOGY & NEUROINFLAMMATION 2023; 10:e200166. [PMID: 37739811 PMCID: PMC10519437 DOI: 10.1212/nxi.0000000000200166] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Accepted: 07/19/2023] [Indexed: 09/24/2023]
Abstract
BACKGROUND AND OBJECTIVES Natalizumab, a monoclonal humanized antibody targeting integrin α4, inhibits the transmigration of lymphocytes into the CNS by preventing the interaction of integrin α4β1 with V-CAM expressed on brain vascular endothelial cells. Although natalizumab treatment reduces the clinical relapse rate in patients with relapsing-remitting MS, its discontinuation after reactivation of the JC virus is associated with a rebound of the disease in 20% of patients. The mechanisms of this rebound are not elucidated, but natalizumab increases the frequencies of circulating CD4 T cells expressing proinflammatory cytokines as well as the proportion of circulating Th17/Th1 cells (Th1-like Th17 cells). Gut-derived memory CD4 T cells are a population of growing interest in the pathogenesis of MS, but whether and how their properties are affected by natalizumab is not known. Here, we studied the phenotype and cytokine expression profile of circulating gut-derived memory CD4 T cells in patients with relapsing-remitting MS under natalizumab. METHODS We identified gut-derived memory CD4 T cells by their expression of integrin β7 and compared their properties and those of integrin β7- memory CD4 T cells across healthy donors and patients with relapsing-remitting MS treated or not with natalizumab. We also compared the capacity of integrin β7- and integrin β7+ CD4 T-cell subsets to transmigrate in vitro across a model of blood-brain barrier. RESULTS The proportions of proinflammatory Th17/Th1 cells as well as of IL-17A+IFNγ+ and IL-17A+GM-CSF+ cells were higher in memory CD4 T cells expressing integrin β7 in patients receiving natalizumab compared with healthy donors and patients with relapsing-remitting MS not receiving natalizumab. By contrast, integrin β7 negative memory CD4 T cells only presented a modest increased in their proportion of Th17/Th1 cells under natalizumab. We further observed that integrin β7+ Th17/Th1 cells migrated as efficiently as integrin β7- Th17/Th1 across a monolayer of brain microvascular endothelial cells. DISCUSSION Our study shows that circulating integrin β7+ memory CD4 T cells of patients with relapsing-remitting MS under natalizumab are enriched in proinflammatory cells supporting the hypothesis that integrin β7+ memory CD4 T cells could play a pathogenic role in the disease rebound observed at natalizumab discontinuation.
Collapse
Affiliation(s)
- Mélanie Nguyen Ky
- From the Immunoconcept (M.N.K., A.D., I.H., A.B., N.S.), CNRS UMR 5164, University of Bordeaux; Service de Neurologie (M.D., A.R.), CRC SEP, Centre Hospitalier Universitaire (CHU) de Bordeaux; and INSERM U 1215 (B.B., A.R.), Neurocentre Magendie, University of Bordeaux, France
| | - Adrien Duran
- From the Immunoconcept (M.N.K., A.D., I.H., A.B., N.S.), CNRS UMR 5164, University of Bordeaux; Service de Neurologie (M.D., A.R.), CRC SEP, Centre Hospitalier Universitaire (CHU) de Bordeaux; and INSERM U 1215 (B.B., A.R.), Neurocentre Magendie, University of Bordeaux, France
| | - Iris Hasantari
- From the Immunoconcept (M.N.K., A.D., I.H., A.B., N.S.), CNRS UMR 5164, University of Bordeaux; Service de Neurologie (M.D., A.R.), CRC SEP, Centre Hospitalier Universitaire (CHU) de Bordeaux; and INSERM U 1215 (B.B., A.R.), Neurocentre Magendie, University of Bordeaux, France
| | - Agnès Bru
- From the Immunoconcept (M.N.K., A.D., I.H., A.B., N.S.), CNRS UMR 5164, University of Bordeaux; Service de Neurologie (M.D., A.R.), CRC SEP, Centre Hospitalier Universitaire (CHU) de Bordeaux; and INSERM U 1215 (B.B., A.R.), Neurocentre Magendie, University of Bordeaux, France
| | - Mathilde Deloire
- From the Immunoconcept (M.N.K., A.D., I.H., A.B., N.S.), CNRS UMR 5164, University of Bordeaux; Service de Neurologie (M.D., A.R.), CRC SEP, Centre Hospitalier Universitaire (CHU) de Bordeaux; and INSERM U 1215 (B.B., A.R.), Neurocentre Magendie, University of Bordeaux, France
| | - Bruno Brochet
- From the Immunoconcept (M.N.K., A.D., I.H., A.B., N.S.), CNRS UMR 5164, University of Bordeaux; Service de Neurologie (M.D., A.R.), CRC SEP, Centre Hospitalier Universitaire (CHU) de Bordeaux; and INSERM U 1215 (B.B., A.R.), Neurocentre Magendie, University of Bordeaux, France
| | - Aurélie Ruet
- From the Immunoconcept (M.N.K., A.D., I.H., A.B., N.S.), CNRS UMR 5164, University of Bordeaux; Service de Neurologie (M.D., A.R.), CRC SEP, Centre Hospitalier Universitaire (CHU) de Bordeaux; and INSERM U 1215 (B.B., A.R.), Neurocentre Magendie, University of Bordeaux, France
| | - Nathalie Schmitt
- From the Immunoconcept (M.N.K., A.D., I.H., A.B., N.S.), CNRS UMR 5164, University of Bordeaux; Service de Neurologie (M.D., A.R.), CRC SEP, Centre Hospitalier Universitaire (CHU) de Bordeaux; and INSERM U 1215 (B.B., A.R.), Neurocentre Magendie, University of Bordeaux, France.
| |
Collapse
|
5
|
Zhu X, Liu B, Ruan Z, Chen M, Li C, Shi H, Huang X, Yu H, Zhou Y, Zhu H, Sun J, Wei Y, Xu W, Dong J. TMT-Based Quantitative Proteomic Analysis Reveals Downregulation of ITGAL and Syk by the Effects of Cycloastragenol in OVA-Induced Asthmatic Mice. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:6842530. [PMID: 36329800 PMCID: PMC9626231 DOI: 10.1155/2022/6842530] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Revised: 08/28/2022] [Accepted: 09/29/2022] [Indexed: 12/08/2023]
Abstract
BACKGROUND Cycloastragenol (CAG) has been reported to alleviate airway inflammation in ovalbumin- (OVA-) induced asthmatic mice. However, its specific mechanisms remain unclear. OBJECTIVE This study is aimed at investigating the effects of CAG on asthma, comparing its efficacy with dexamethasone (DEX), and elucidating the mechanism of CAG's regulation. METHODS The asthma mouse model was induced by OVA. CAG at the optimal dose of 125 mg/kg was given every day from day 0 for 20-day prevention or from day 14 for a 7-day treatment. We observed the preventive and therapeutic effects of CAG in asthmatic mice by evaluating the airway inflammation, AHR, and mucus secretion. Lung proteins were used for TMT-based quantitative proteomic analysis to enunciate its regulatory mechanisms. RESULTS The early administration of 125 mg/kg CAG before asthma happened prevented asthmatic mice from AHR, airway inflammation, and mucus hypersecretion, returning to nearly the original baseline. Alternatively, the administration of CAG during asthma also had the same therapeutic effects as DEX. The proteomic analysis revealed that the therapeutical effects of CAG were associated with 248 differentially expressed proteins and 3 enriched KEGG pathways. We then focused on 3 differentially expressed proteins (ITGAL, Syk, and Vav1) and demonstrated that CAG treatment downregulated ITGAL, Syk, and Vav1 by quantitative real-time PCR, western blot analysis, and immunohistochemical staining. CONCLUSION These findings suggest that CAG exerts preventive and protective effects on asthma by inhibiting ITGAL, Syk, and the downstream target Vav1.
Collapse
Affiliation(s)
- Xueyi Zhu
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Baojun Liu
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, China
- Institutes of Integrative Medicine, Fudan University, Shanghai, China
| | - Zhenhui Ruan
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Mengmeng Chen
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Congcong Li
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Hanlin Shi
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Xi Huang
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Hang Yu
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Yaolong Zhou
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Hehua Zhu
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Jing Sun
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, China
- Institutes of Integrative Medicine, Fudan University, Shanghai, China
| | - Ying Wei
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, China
- Institutes of Integrative Medicine, Fudan University, Shanghai, China
| | - Weifang Xu
- Shenzhen Hospital of Guangzhou University of Chinese Medicine (Futian), Shenzhen, China
| | - Jingcheng Dong
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, China
- Institutes of Integrative Medicine, Fudan University, Shanghai, China
| |
Collapse
|
6
|
Sun D, Cai X, Shen F, Fan L, Yang H, Zheng S, Zhou L, Chen K, Wang Z. Transcriptome-Wide m6A Methylome and m6A-Modified Gene Analysis in Asthma. Front Cell Dev Biol 2022; 10:799459. [PMID: 35712670 PMCID: PMC9197130 DOI: 10.3389/fcell.2022.799459] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Accepted: 04/19/2022] [Indexed: 11/25/2022] Open
Abstract
N6-methyladenosine (m6A) modification is one of the most prevalent RNA modification forms and is an important posttranscriptional mechanism for regulating genes. In previous research, we found that m6A regulator–mediated RNA methylation modification was involved in asthma; however, the specific modified genes are not clear. In this study, we systematically evaluated the transcriptome-wide m6A methylome and m6A-modified genes in asthma. Here, we performed two high-throughput sequencing methods, methylated RNA immunoprecipitation sequencing (MeRIP-seq), and RNA sequencing (RNA-seq) to identify key genes with m6A modification in asthma. Through difference analysis, we found that 416 methylation peaks were significantly upregulated and 152 methylation peaks were significantly downregulated, and it was mainly distributed in 3′ UTR. Furthermore, compared with the control group, there were 2,505 significantly upregulated genes and 4,715 significantly downregulated genes in the asthma group. Next, through a combined analysis of transcriptome and differential peaks, 14 differentially expressed genes related to RNA methylation modification were screened. Finally, through 87 health controls and 411 asthma cases from the U-BIOPRED (Unbiased Biomarkers for the Prediction of Respiratory Disease Outcomes) program, we verified three m6A-modified key genes (BCL11A, MATK, and CD300A) and found that they were mainly distributed in exons and enriched in 3' UTR. Our findings suggested that intervening in m6A-modified genes may provide a new idea for the treatment of asthma.
Collapse
Affiliation(s)
- Deyang Sun
- The First Clinical College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Xiaolu Cai
- The First Clinical College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Fenglin Shen
- The First Clinical College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Liming Fan
- The First Clinical College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Huan Yang
- The First Clinical College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Suqun Zheng
- Department of Respiration, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| | - Linshui Zhou
- Department of Respiration, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| | - Ke Chen
- The First Clinical College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Zhen Wang
- The First Clinical College, Zhejiang Chinese Medical University, Hangzhou, China
- Department of Respiration, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
- *Correspondence: Zhen Wang,
| |
Collapse
|
7
|
Understanding the Role of LFA-1 in Leukocyte Adhesion Deficiency Type I (LAD I): Moving towards Inflammation? Int J Mol Sci 2022; 23:ijms23073578. [PMID: 35408940 PMCID: PMC8998723 DOI: 10.3390/ijms23073578] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 03/18/2022] [Accepted: 03/21/2022] [Indexed: 02/04/2023] Open
Abstract
LFA-1 (Lymphocyte function-associated antigen-1) is a heterodimeric integrin (CD11a/CD18) present on the surface of all leukocytes; it is essential for leukocyte recruitment to the site of tissue inflammation, but also for other immunological processes such as T cell activation and formation of the immunological synapse. Absent or dysfunctional expression of LFA-1, caused by mutations in the ITGB2 (integrin subunit beta 2) gene, results in a rare immunodeficiency syndrome known as Leukocyte adhesion deficiency type I (LAD I). Patients suffering from severe LAD I present with recurrent infections of the skin and mucosa, as well as inflammatory symptoms complicating the clinical course of the disease before and after allogeneic hematopoietic stem cell transplantation (alloHSCT); alloHSCT is currently the only established curative treatment option. With this review, we aim to provide an overview of the intrinsic role of inflammation in LAD I.
Collapse
|
8
|
Johansen KH, Golec DP, Thomsen JH, Schwartzberg PL, Okkenhaug K. PI3K in T Cell Adhesion and Trafficking. Front Immunol 2021; 12:708908. [PMID: 34421914 PMCID: PMC8377255 DOI: 10.3389/fimmu.2021.708908] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Accepted: 07/19/2021] [Indexed: 12/12/2022] Open
Abstract
PI3K signalling is required for activation, differentiation, and trafficking of T cells. PI3Kδ, the dominant PI3K isoform in T cells, has been extensively characterised using PI3Kδ mutant mouse models and PI3K inhibitors. Furthermore, characterisation of patients with Activated PI3K Delta Syndrome (APDS) and mouse models with hyperactive PI3Kδ have shed light on how increased PI3Kδ activity affects T cell functions. An important function of PI3Kδ is that it acts downstream of TCR stimulation to activate the major T cell integrin, LFA-1, which controls transendothelial migration of T cells as well as their interaction with antigen-presenting cells. PI3Kδ also suppresses the cell surface expression of CD62L and CCR7 which controls the migration of T cells across high endothelial venules in the lymph nodes and S1PR1 which controls lymph node egress. Therefore, PI3Kδ can control both entry and exit of T cells from lymph nodes as well as the recruitment to and retention of T cells within inflamed tissues. This review will focus on the regulation of adhesion receptors by PI3Kδ and how this contributes to T cell trafficking and localisation. These findings are relevant for our understanding of how PI3Kδ inhibitors may affect T cell redistribution and function.
Collapse
Affiliation(s)
- Kristoffer H Johansen
- Department of Pathology, University of Cambridge, Cambridge, United Kingdom.,Laboratory of Immune System Biology, NIAID, NIH, Bethesda, MD, United States
| | - Dominic P Golec
- Laboratory of Immune System Biology, NIAID, NIH, Bethesda, MD, United States
| | - Julie H Thomsen
- Department of Pathology, University of Cambridge, Cambridge, United Kingdom
| | | | - Klaus Okkenhaug
- Department of Pathology, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
9
|
Tiwari A, Wang AL, Li J, Lutz SM, Kho AT, Weiss ST, Tantisira KG, McGeachie MJ. Seasonal Variation in miR-328-3p and let-7d-3p Are Associated With Seasonal Allergies and Asthma Symptoms in Children. ALLERGY, ASTHMA & IMMUNOLOGY RESEARCH 2021; 13:576-588. [PMID: 34212545 PMCID: PMC8255344 DOI: 10.4168/aair.2021.13.4.576] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Revised: 09/26/2020] [Accepted: 10/08/2020] [Indexed: 12/26/2022]
Abstract
OBJECTIVE MicroRNAs (miRs) are small non-coding RNA molecules of around 18-22 nucleotides that are key regulators of many biologic processes, particularly inflammation. The purpose of this study was to determine the association of circulating miRs from asthmatic children with seasonal variation in allergic inflammation and asthma symptoms. METHODS We used available small RNA sequencing on blood serum from 398 children with mild-to-moderate asthma from the Childhood Asthma Management Program. We used seasonal asthma symptom data at the study baseline and allergen affection status from baseline skin prick tests as primary outcomes. We identified differentially expressed (DE) miRs between pairs of seasons using DESeq2. Regression analysis was used to identify associations between allergy status to specific seasonal allergens and DE miRs in 4 seasons and between seasonal asthma symptom data and DE miRs. We performed pathway enrichment analysis for target genes of the DE miRs using DAVID. RESULTS After quality control, 398 samples underwent differential analysis between the 4 seasons. We found 52 unique miRs from a total of 81 DE miRs across seasons. Further investigation of the association between these miRs and sensitization to seasonal allergens using skin prick tests revealed that 26 unique miRs from a total of 38 miRs were significantly associated with a same-season allergen. Comparison between seasonal asthma symptom data revealed that 2 of these 26 miRs also had significant associations with asthma symptoms in the same seasons: miR-328-3p (P < 0.03) and let-7d-3p (P < 0.05). Enrichment analysis showed that the most enriched pathway clusters were Rap1, Ras, and MAPK signaling pathways. CONCLUSION Our results show seasonal variation in miR-328-3p and let-7d-3p are significantly associated with seasonal asthma symptoms and seasonal allergies. These indicate a potentially protective role for let-7d-3p and a deleterious role for miR-328-3p in asthmatics sensitized to mulberry. Further work will determine whether these miRs are drivers or results of the allergic response.
Collapse
Affiliation(s)
- Anshul Tiwari
- Channing Division of Network Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Alberta L Wang
- Channing Division of Network Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Jiang Li
- Channing Division of Network Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Sharon M Lutz
- PRecisiOn Medicine Translational Research (PROMoTeR) Center, Department of Population Medicine, Harvard Pilgrim Health Care Institute and Harvard Medical School, Boston, MA, USA
| | - Alvin T Kho
- Channing Division of Network Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Computational Health Informatics Program, Boston Children's Hospital, Boston, MA, USA
| | - Scott T Weiss
- Channing Division of Network Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Kelan G Tantisira
- Channing Division of Network Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Michael J McGeachie
- Channing Division of Network Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
10
|
Wang S, Sun J, Chen K, Ma P, Lei Q, Xing S, Cao Z, Sun S, Yu Z, Liu Y, Li N. Perspectives of tumor-infiltrating lymphocyte treatment in solid tumors. BMC Med 2021; 19:140. [PMID: 34112147 PMCID: PMC8194199 DOI: 10.1186/s12916-021-02006-4] [Citation(s) in RCA: 114] [Impact Index Per Article: 28.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Accepted: 05/13/2021] [Indexed: 12/26/2022] Open
Abstract
Tumor-infiltrating lymphocyte (TIL) therapy is a type of adoptive cellular therapy by harvesting infiltrated lymphocytes from tumors, culturing and amplifying them in vitro and then infusing back to treat patients. Its diverse TCR clonality, superior tumor-homing ability, and low off-target toxicity endow TIL therapy unique advantages in treating solid tumors compared with other adoptive cellular therapies. Nevertheless, the successful application of TIL therapy currently is still limited to several types of tumors. Herein in this review, we summarize the fundamental work in the field of TIL therapy and the current landscape and advances of TIL clinical trials worldwide. Moreover, the limitations of the current TIL regimen have been discussed and the opportunities and challenges in the development of next-generation TIL are highlighted. Finally, the future directions of TIL therapy towards a broader clinical application have been proposed.
Collapse
Affiliation(s)
- Shuhang Wang
- Clinical Cancer Center, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | | | - Kun Chen
- NHC Key Laboratory of Pulmonary Immunological Diseases, Guizhou Provincial People's Hospital, Guiyang, China
| | - Peiwen Ma
- Clinical Cancer Center, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Qi Lei
- Clinical Cancer Center, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Shujun Xing
- Clinical Cancer Center, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | | | - Shujun Sun
- Queen Mary School, Nanchang University, Nanchang, 330006, China
| | | | - Yarong Liu
- Grit Biotechnology Ltd., Shanghai, China.
| | - Ning Li
- Clinical Cancer Center, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| |
Collapse
|
11
|
Emo K, Reilly EC, Sportiello M, Yang H, Topham DJ. T cell and chemokine receptors differentially control CD8 T cell motility behavior in the infected airways immediately before and after virus clearance in a primary infection. PLoS One 2020; 15:e0227157. [PMID: 32817719 PMCID: PMC7444504 DOI: 10.1371/journal.pone.0227157] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Accepted: 04/21/2020] [Indexed: 11/19/2022] Open
Abstract
In mice, experimental influenza virus infection stimulates CD8 T cell infiltration of the airways. Virus is cleared by day 9, and between days 8 and 9 there is an abrupt change in CD8 T cell motility behavior transitioning from low velocity and high confinement on day 8, to high velocity with continued high confinement on day 9. We hypothesized that loss of virus and/or antigen signals in the context of high chemokine levels drives the T cells into a rapid surveillance mode. Virus infection induces chemokine production, which may change when the virus is cleared. We therefore sought to examine this period of rapid changes to the T cell environment in the tissue and seek evidence on the roles of peptide-MHC and chemokine receptor interactions. Experiments were performed to block G protein coupled receptor (GPCR) signaling with Pertussis toxin (Ptx). Ptx treatment generally reduced cell velocities and mildly increased confinement suggesting chemokine mediated arrest (velocity <2 μm/min) (Friedman RS, 2005), except on day 8 when velocity increased and confinement was relieved. Blocking specific peptide-MHC with monoclonal antibody unexpectedly decreased velocities on days 7 through 9, suggesting TCR/peptide-MHC interactions promote cell mobility in the tissue. Together, these results suggest the T cells are engaged with antigen bearing and chemokine producing cells that affect motility in ways that vary with the day after infection. The increase in velocities on day 9 were reversed by addition of specific peptide, consistent with the idea that antigen signals become limiting on day 9 compared to earlier time points. Thus, antigen and chemokine signals act to alternately promote and restrict CD8 T cell motility until the point of virus clearance, suggesting the switch in motility behavior on day 9 may be due to a combination of limiting antigen in the presence of high chemokine signals as the virus is cleared.
Collapse
Affiliation(s)
- Kris Emo
- David H. Smith Center for Vaccine Biology and Immunology, Department of Microbiology and Immunology, Rochester, NY, United States of America
| | - Emma C. Reilly
- David H. Smith Center for Vaccine Biology and Immunology, Department of Microbiology and Immunology, Rochester, NY, United States of America
| | - Mike Sportiello
- David H. Smith Center for Vaccine Biology and Immunology, Department of Microbiology and Immunology, Rochester, NY, United States of America
| | - Hongmei Yang
- Department of Biostatistics and Computational Biology, University of Rochester Medical Center, Rochester, NY, United States of America
| | - David J. Topham
- David H. Smith Center for Vaccine Biology and Immunology, Department of Microbiology and Immunology, Rochester, NY, United States of America
- * E-mail:
| |
Collapse
|
12
|
Zhang L, Peng TL, Wang L, Meng XH, Zhu W, Zeng Y, Zhu JQ, Zhou Y, Xiao HM, Deng HW. Network-based Transcriptome-wide Expression Study for Postmenopausal Osteoporosis. J Clin Endocrinol Metab 2020; 105:5850085. [PMID: 32483604 PMCID: PMC7320836 DOI: 10.1210/clinem/dgaa319] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Accepted: 05/27/2020] [Indexed: 01/08/2023]
Abstract
PURPOSE Menopause is a crucial physiological transition during a woman's life, and it occurs with growing risks of health issues like osteoporosis. To identify postmenopausal osteoporosis-related genes, we performed transcriptome-wide expression analyses for human peripheral blood monocytes (PBMs) using Affymetrix 1.0 ST arrays in 40 Caucasian postmenopausal women with discordant bone mineral density (BMD) levels. METHODS We performed multiscale embedded gene coexpression network analysis (MEGENA) to study functionally orchestrating clusters of differentially expressed genes in the form of functional networks. Gene sets net correlations analysis (GSNCA) was applied to assess how the coexpression structure of a predefined gene set differs in high and low BMD groups. Bayesian network (BN) analysis was used to identify important regulation patterns between potential risk genes for osteoporosis. A small interfering ribonucleic acid (siRNA)-based gene silencing in vitro experiment was performed to validate the findings from BN analysis. RESULT MEGENA showed that the "T cell receptor signaling pathway" and the "osteoclast differentiation pathway" were significantly enriched in the identified compact network, which is significantly correlated with BMD variation. GSNCA revealed that the coexpression structure of the "Signaling by TGF-beta receptor complex pathway" is significantly different between the 2 BMD discordant groups; the hub genes in the postmenopausal low and high BMD group are FURIN and SMAD3 respectively. With siRNA in vitro experiments, we confirmed the regulation relationship of TGFBR2-SMAD7 and TGFBR1-SMURF2. MAIN CONCLUSION The present study suggests that biological signals involved in monocyte recruitment, monocyte/macrophage lineage development, osteoclast formation, and osteoclast differentiation might function together in PBMs that contribute to the pathogenesis of postmenopausal osteoporosis.
Collapse
Affiliation(s)
- Lan Zhang
- Center for Biomedical informatics and Genomics, Department of Medicine, Tulane University, New Orleans, Louisiana
| | - Tian-Liu Peng
- Institute of Reproduction and Stem Cell Engineering, School of Basic Medical Science, Central South University, Changsha, Hunan, China
| | - Le Wang
- Institute of Reproduction and Stem Cell Engineering, School of Basic Medical Science, Central South University, Changsha, Hunan, China
| | - Xiang-He Meng
- Laboratory of Molecular and Statistical Genetics, College of Life Sciences, Hunan Normal University, Changsha, Hunan, China
| | - Wei Zhu
- Center for Biomedical informatics and Genomics, Department of Medicine, Tulane University, New Orleans, Louisiana
| | - Yong Zeng
- Center for Biomedical informatics and Genomics, Department of Medicine, Tulane University, New Orleans, Louisiana
| | - Jia-Qiang Zhu
- Department of Biostatistics, University of Michigan, Ann Arbor, Michigan
| | - Yu Zhou
- Center for Biomedical informatics and Genomics, Department of Medicine, Tulane University, New Orleans, Louisiana
| | - Hong-Mei Xiao
- Institute of Reproduction and Stem Cell Engineering, School of Basic Medical Science, Central South University, Changsha, Hunan, China
| | - Hong-Wen Deng
- Center for Biomedical informatics and Genomics, Department of Medicine, Tulane University, New Orleans, Louisiana
- Correspondence and Reprint Requests: Hong-Wen Deng, Center for Biomedical Informatics and Genomics, Department of Medicine, School of Medicine, Tulane University, New Orleans, LA 70112, USA. E-mail:
| |
Collapse
|
13
|
Network Pharmacology Approach to Investigate the Preventive Mechanism of Hunan Expert Group Recommended Chinese Medicine Prevention No. 2 Prescription Against COVID-19. DIGITAL CHINESE MEDICINE 2020. [PMCID: PMC7366109 DOI: 10.1016/j.dcmed.2020.06.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Objective To explore the possible preventive mechanism of Hunan expert group recommended Chinese medicine prescription of No. 2 (Pre-No. 2) against coronavirus disease 2019 (COVID-19) by network pharmacology method. Methods The target proteins of effective components and active compounds in Pre-No. 2 were screened by searching the Tradi-tional Chinese Medicine Systems Pharmacology Database and Analysis Platform (TCMSP). A component-target-disease interac-tion network of Pre-No. 2 was constructed by Cytoscape 3.7.2, gene ontology (GO) analysis, and Kyoto encyclopedia of genes and genomes (KEGG) analysis of target protein pathway by DAVID. Results A total of 163 compounds and 278 target protein targets in Pre-No. 2 were collected from the TCMSP database. Kaempferol, wogonin, 7-methoxy-2-methyl isoflavone, formononetin, isorhamnetin, and licochalcone A were the most frequent targets in the regulatory network. GO enrichment analysis showed that Pre-No. 2 regulated response to virus, viral processes, humoral immune responses, defense responses to virus and viral entry into host cells. KEGG enrichment analysis showed that the formula regulated the NF-κB signaling pathway, B cell receptor signaling pathway, viral carcinogenesis, T cell signaling pathway and FcγR-mediated phagocytosis signaling pathway. Conclusions Pre-No. 2 may play a preventive role against COVID-19 through regulation of the Toll-like signaling, T cell signaling, B cell signaling and other signaling pathways. It may re-gulate the immune system to protect against anti-influenza virus.
Collapse
|
14
|
Perez LG, Kempski J, McGee HM, Pelzcar P, Agalioti T, Giannou A, Konczalla L, Brockmann L, Wahib R, Xu H, Vesely MCA, Soukou S, Steglich B, Bedke T, Manthey C, Seiz O, Diercks BP, Gnafakis S, Guse AH, Perez D, Izbicki JR, Gagliani N, Flavell RA, Huber S. TGF-β signaling in Th17 cells promotes IL-22 production and colitis-associated colon cancer. Nat Commun 2020; 11:2608. [PMID: 32451418 PMCID: PMC7248087 DOI: 10.1038/s41467-020-16363-w] [Citation(s) in RCA: 94] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Accepted: 04/24/2020] [Indexed: 12/18/2022] Open
Abstract
IL-22 has dual functions during tumorigenesis. Short term IL-22 production protects against genotoxic stress, whereas uncontrolled IL-22 activity promotes tumor growth; therefore, tight regulation of IL-22 is essential. TGF-β1 promotes the differentiation of Th17 cells, which are known to be a major source of IL-22, but the effect of TGF-β signaling on the production of IL-22 in CD4+ T cells is controversial. Here we show an increased presence of IL-17+IL-22+ cells and TGF-β1 in colorectal cancer compared to normal adjacent tissue, whereas the frequency of IL-22 single producing cells is not changed. Accordingly, TGF-β signaling in CD4+ T cells (specifically Th17 cells) promotes the emergence of IL-22-producing Th17 cells and thereby tumorigenesis in mice. IL-22 single producing T cells, however, are not dependent on TGF-β signaling. We show that TGF-β, via AhR induction, and PI3K signaling promotes IL-22 production in Th17 cells.
Collapse
Affiliation(s)
- Laura Garcia Perez
- I. Department of Medicine, University Medical Center Hamburg-Eppendorf, 20246, Hamburg, Germany
| | - Jan Kempski
- I. Department of Medicine, University Medical Center Hamburg-Eppendorf, 20246, Hamburg, Germany
| | - Heather M McGee
- Center for Immunobiology and Microbial Pathogenesis, Salk Institute for Biological Studies, 10010, La Jolla, CA, USA
| | - Penelope Pelzcar
- I. Department of Medicine, University Medical Center Hamburg-Eppendorf, 20246, Hamburg, Germany
| | - Theodora Agalioti
- Department of General, Visceral and Thoracic Surgery, University Medical Center Hamburg-Eppendorf, 20246, Hamburg, Germany
| | - Anastasios Giannou
- I. Department of Medicine, University Medical Center Hamburg-Eppendorf, 20246, Hamburg, Germany
| | - Leonie Konczalla
- Department of General, Visceral and Thoracic Surgery, University Medical Center Hamburg-Eppendorf, 20246, Hamburg, Germany
| | - Leonie Brockmann
- Department of Microbiology and Immunology, Columbia University, New York, NY, USA
| | - Ramez Wahib
- Department of General, Visceral and Thoracic Surgery, University Medical Center Hamburg-Eppendorf, 20246, Hamburg, Germany
| | - Hao Xu
- Department of Immunobiology, School of Medicine, Yale University, New Haven, CT, 06520, USA
| | | | - Shiwa Soukou
- I. Department of Medicine, University Medical Center Hamburg-Eppendorf, 20246, Hamburg, Germany
| | - Babett Steglich
- I. Department of Medicine, University Medical Center Hamburg-Eppendorf, 20246, Hamburg, Germany
- Department of General, Visceral and Thoracic Surgery, University Medical Center Hamburg-Eppendorf, 20246, Hamburg, Germany
| | - Tanja Bedke
- I. Department of Medicine, University Medical Center Hamburg-Eppendorf, 20246, Hamburg, Germany
| | - Carolin Manthey
- I. Department of Medicine, University Medical Center Hamburg-Eppendorf, 20246, Hamburg, Germany
| | - Oliver Seiz
- I. Department of Medicine, University Medical Center Hamburg-Eppendorf, 20246, Hamburg, Germany
| | - Björn-Philipp Diercks
- Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, 20246, Hamburg, Germany
| | - Stylianos Gnafakis
- Laboratory of Innate Immunity, Department of Microbiology, Infectious Diseases and Immunology, Charité - Universitätsmedizin Berlin, Hindenburgdamm 30, 12203, Berlin, Germany
- Berlin Institute of Health (BIH), Anna-Louisa-Karsch Strasse 2, 10117, Berlin, Germany
- Mucosal and Developmental Immunology, Deutsches Rheuma-Forschungszentrum, Charitéplatz 1, 10117, Berlin, Germany
| | - Andreas H Guse
- Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, 20246, Hamburg, Germany
| | - Daniel Perez
- Department of General, Visceral and Thoracic Surgery, University Medical Center Hamburg-Eppendorf, 20246, Hamburg, Germany
| | - Jakob R Izbicki
- Department of General, Visceral and Thoracic Surgery, University Medical Center Hamburg-Eppendorf, 20246, Hamburg, Germany
| | - Nicola Gagliani
- I. Department of Medicine, University Medical Center Hamburg-Eppendorf, 20246, Hamburg, Germany
- Department of General, Visceral and Thoracic Surgery, University Medical Center Hamburg-Eppendorf, 20246, Hamburg, Germany
- Immunology and Allergy Unit, Department of Medicine, Solna, Karolinska Institute and University Hospital, Stockholm, Sweden
| | - Richard A Flavell
- Department of Immunobiology, School of Medicine, Yale University, New Haven, CT, 06520, USA.
- Howard Hughes Medical Institute, School of Medicine, Yale University, New Haven, CT, 06520, USA.
| | - Samuel Huber
- I. Department of Medicine, University Medical Center Hamburg-Eppendorf, 20246, Hamburg, Germany.
| |
Collapse
|
15
|
Zhang W, Yang M, Wang Y, Wu X, Zhang X, Ding Y, Yin Z. Genomic analysis reveals selection signatures of the Wannan Black pig during domestication and breeding. ASIAN-AUSTRALASIAN JOURNAL OF ANIMAL SCIENCES 2019; 33:712-721. [PMID: 31480149 PMCID: PMC7206397 DOI: 10.5713/ajas.19.0289] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/07/2019] [Accepted: 08/01/2019] [Indexed: 12/31/2022]
Abstract
Objective The Wannan Black pig is a typical Chinese indigenous, disease-resistant pig breed with high fertility, and a crude-feed tolerance that has been bred by artificial selection in the south of Anhui province for a long time. However, genome variation, genetic relationships with other pig breeds, and domestication, remain poorly understood. Here, we focus on elucidating the genetic characteristics of the Wannan Black pig and identifying selection signatures during domestication and breeding. Methods We identified the whole-genome variation in the Wannan Black pig and performed population admixture analyses to determine genetic relationships with other domesticated pig breeds and wild boars. Then, we identified the selection signatures between the Wannan Black pig and Asian wild boars in 100-kb windows sliding in 10 kb steps by using two approaches: the fixation index (FST) and π ratios. Results Resequencing the Wannan Black pig genome yielded 501.52 G of raw data. After calling single-nucleotide variants (SNVs) and insertions/deletions (InDels), we identified 21,316,754 SNVs and 5,067,206 InDels (2,898,582 inserts and 2,168,624 deletions). Additionally, we found genes associated with growth, immunity, and digestive functions. Conclusion Our findings help in explaining the unique genetic and phenotypic characteristics of Wannan Black pigs, which in turn can be informative for future breeding programs of Wannan Black pigs.
Collapse
Affiliation(s)
- Wei Zhang
- College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, China
| | - Min Yang
- College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, China
| | - Yuanlang Wang
- College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, China
| | - Xudong Wu
- College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, China
| | - Xiaodong Zhang
- College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, China
| | - Yueyun Ding
- College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, China
| | - Zongjun Yin
- College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, China
| |
Collapse
|
16
|
Jiang Y, Xie M, Fan W, Xue J, Zhou Z, Tang J, Chen G, Hou S. Transcriptome Analysis Reveals Differential Expression of Genes Regulating Hepatic Triglyceride Metabolism in Pekin Ducks During Dietary Threonine Deficiency. Front Genet 2019; 10:710. [PMID: 31428138 PMCID: PMC6688585 DOI: 10.3389/fgene.2019.00710] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2019] [Accepted: 07/04/2019] [Indexed: 12/12/2022] Open
Abstract
Dietary threonine (Thr) deficiency increases hepatic triglyceride accumulation in Pekin ducks, which results in fatty liver disease and impairs hepatic function. However, the underlying molecular mechanisms altered by dietary Thr deficiency are still unknown. To identify the underlying molecular changes, 180 one-day-old ducklings were divided into three groups, including Thr deficiency group (Thr-D), Thr sufficiency group (Thr-S), and pair-fed group (Pair-F) that was fed with a Thr-sufficient diet but with reduced daily feed intake. The results showed that feed intake was similar between Thr-D and Pair-F groups, but weight gain rate and final body weight in the Thr-D group were lower than those in the Pair-F group. Feed intake, weight gain, and body weight in Thr-D and Pair-F groups were lower than those in the Thr-S group. The Thr-D diet reduced abdominal fat percentage but increased hepatic triglyceride content when compared with that of the Thr-S and Pair-F groups. The Pair-F reduced hepatic levels of C15:0, C17:0, C18:0, C20:0, C20:4n6, and C22:0 and also reduced total fatty acid, saturated fatty acid, and unsaturated fatty acid content when compared with those of the Thr-D and Thr-S groups. The Thr-D diet increased hepatic content of C6:0, C17:1, C18:3n6, C20:0, C20:1n9, and C22:2, as well as reduced the content of C18:2n6t and C23:0 when compared with those of the Thr-S group. Transcriptome analysis in the liver indicated that the Thr-D diet upregulated genes related to fatty acid and triglyceride synthesis and downregulated genes related to fatty acid oxidation and triglyceride transport. Gene ontology analysis showed that more genes related to lipid metabolism processes and molecular function were differentially expressed in the Thr-D group relative to Thr-S and Pair-F groups than in the Pair-F group relative to the Thr-S group. KEGG pathway analysis showed that differentially expressed genes were enriched in signal transduction, immune, hormone, lipid, and amino acid metabolism pathways. Our findings indicated that the Thr-D diet increased hepatic triglyceride and fatty acid accumulation via increasing fatty acid and triglyceride synthesis and reducing fatty acid oxidation and triglyceride transport. These findings provide novel insights into our understanding of the molecular mechanisms underlying fat accumulation in the liver caused by dietary threonine deficiency.
Collapse
Affiliation(s)
- Yong Jiang
- Key Laboratory of Animal (Poultry) Genetics Breeding and Reproduction, Ministry of Agriculture and Rural Affairs; Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, China.,College of Animal Science and Technology, Yangzhou University, Yangzhou, China
| | - Ming Xie
- Key Laboratory of Animal (Poultry) Genetics Breeding and Reproduction, Ministry of Agriculture and Rural Affairs; Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Wenlei Fan
- Key Laboratory of Animal (Poultry) Genetics Breeding and Reproduction, Ministry of Agriculture and Rural Affairs; Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, China.,College of Food Science and Engineering, Qingdao Agricultural University, Qingdao, China
| | - Jiajia Xue
- Key Laboratory of Animal (Poultry) Genetics Breeding and Reproduction, Ministry of Agriculture and Rural Affairs; Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Zhengkui Zhou
- Key Laboratory of Animal (Poultry) Genetics Breeding and Reproduction, Ministry of Agriculture and Rural Affairs; Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Jing Tang
- Key Laboratory of Animal (Poultry) Genetics Breeding and Reproduction, Ministry of Agriculture and Rural Affairs; Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Guohong Chen
- College of Animal Science and Technology, Yangzhou University, Yangzhou, China
| | - Shuisheng Hou
- Key Laboratory of Animal (Poultry) Genetics Breeding and Reproduction, Ministry of Agriculture and Rural Affairs; Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, China
| |
Collapse
|
17
|
Palm AKE, Henry C. Remembrance of Things Past: Long-Term B Cell Memory After Infection and Vaccination. Front Immunol 2019; 10:1787. [PMID: 31417562 PMCID: PMC6685390 DOI: 10.3389/fimmu.2019.01787] [Citation(s) in RCA: 175] [Impact Index Per Article: 29.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Accepted: 07/16/2019] [Indexed: 02/03/2023] Open
Abstract
The success of vaccines is dependent on the generation and maintenance of immunological memory. The immune system can remember previously encountered pathogens, and memory B and T cells are critical in secondary responses to infection. Studies in mice have helped to understand how different memory B cell populations are generated following antigen exposure and how affinity for the antigen is determinant to B cell fate. Additionally, such studies were fundamental in defining memory B cell niches and how B cells respond following subsequent exposure with the same antigen. On the other hand, human studies are essential to the development of better, newer vaccines but sometimes limited by the difficulty to access primary and secondary lymphoid organs. However, work using human influenza and HIV virus infection and/or immunization in particular has significantly advanced today's understanding of memory B cells. This review will focus on the generation, function, and longevity of B-cell mediated immunological memory (memory B cells and plasma cells) in response to infection and vaccination both in mice and in humans.
Collapse
Affiliation(s)
- Anna-Karin E Palm
- Section of Rheumatology, Department of Medicine, University of Chicago, Chicago, IL, United States
| | - Carole Henry
- Section of Rheumatology, Department of Medicine, University of Chicago, Chicago, IL, United States
| |
Collapse
|
18
|
Nakagawa R, Muroyama R, Saeki C, Oikawa T, Kaise Y, Koike K, Arai J, Nakano M, Matsubara Y, Takano K, Hirata Y, Saruta M, Zeniya M, Kato N. CD4 + T cells from patients with primary biliary cholangitis show T cell activation and differentially expressed T-cell receptor repertoires. Hepatol Res 2019; 49:653-662. [PMID: 30690835 DOI: 10.1111/hepr.13318] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2018] [Revised: 01/08/2019] [Accepted: 01/22/2019] [Indexed: 02/08/2023]
Abstract
AIM Primary biliary cholangitis (PBC) is an autoimmune liver disease with unknown pathogenesis. In PBC, activation of T-cell receptor (TCR) signaling is associated with inflammatory cytokine production through N-Ras upregulation. Although the CD4+ T cell TCR repertoire could be associated with PBC pathogenesis, it has not been evaluated. Thus, we analyzed the PBC-CD4+ T cell TCR repertoire using next generation sequencing (NGS). METHODS Four PBC patients (one treatment-naïve and three receiving ursodeoxycholic acid) and three healthy individuals were enrolled. NRAS expression in CD4+ T cells was assessed by quantitative reverse transcription-polymerase chain reaction (qRT-PCR). N-Ras dynamics in CD4+ T cells were assessed by qRT-PCR and GTP-N-Ras activation assay. The TCR α- (TRA) and β-chain (TRB) repertoires on CD4+ T cells were analyzed by NGS and profiled using hierarchical analysis. Motif analysis was undertaken to elucidate the structure of PBC-specific TCRs. RESULTS NRAS was upregulated in PBC relative to control CD4+ T cells (P < 0.05), and N-Ras enhanced T cell activation in CD4+ T cells. Among 2668 TRAs and 841 TRBs, 20 and 11, respectively, were differentially expressed in PBC compared to that in controls (P < 0.05, fold-change >2). Among them, TRAV29/J22, TRBV6-5/J2-6, and TRBV10-1/J2-1 were expressed in PBC but the expression was negligible in the controls, with more mature and longer forms observed in PBC-CD4+ T cells. CONCLUSIONS N-Ras was upregulated in PBC-CD4+ T cells, and it enhanced TCR activation, indicating that PBC-CD4+ T cells were activated by N-Ras upregulation with differentially expressed TCR repertoires on their surfaces.
Collapse
Affiliation(s)
- Ryo Nakagawa
- Division of Advanced Genome Medicine, Institute of Medical Science, University of Tokyo, Tokyo, Japan.,Department of Gastroenterology and Hepatology, Jikei University School of Medicine, Tokyo, Japan.,Department of Gastroenterology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Ryosuke Muroyama
- Division of Advanced Genome Medicine, Institute of Medical Science, University of Tokyo, Tokyo, Japan
| | - Chisato Saeki
- Department of Gastroenterology and Hepatology, Jikei University School of Medicine, Tokyo, Japan
| | - Tsunekazu Oikawa
- Department of Gastroenterology and Hepatology, Jikei University School of Medicine, Tokyo, Japan
| | - Yoshimi Kaise
- Division of Advanced Genome Medicine, Institute of Medical Science, University of Tokyo, Tokyo, Japan
| | - Kazuhiko Koike
- Department of Gastroenterology and Hepatology, Jikei University School of Medicine, Tokyo, Japan
| | - Jun Arai
- Division of Advanced Genome Medicine, Institute of Medical Science, University of Tokyo, Tokyo, Japan
| | - Masanori Nakano
- Department of Gastroenterology and Hepatology, Jikei University School of Medicine, Tokyo, Japan
| | - Yasuo Matsubara
- Division of Advanced Genome Medicine, Institute of Medical Science, University of Tokyo, Tokyo, Japan
| | - Keiko Takano
- Department of Gastroenterology and Hepatology, Jikei University School of Medicine, Tokyo, Japan
| | - Yoshihiro Hirata
- Division of Advanced Genome Medicine, Institute of Medical Science, University of Tokyo, Tokyo, Japan
| | - Masayuki Saruta
- Department of Gastroenterology and Hepatology, Jikei University School of Medicine, Tokyo, Japan
| | - Mikio Zeniya
- Sanno Medical Center, International University of Health and Welfare, Tokyo, Japan
| | - Naoya Kato
- Division of Advanced Genome Medicine, Institute of Medical Science, University of Tokyo, Tokyo, Japan.,Department of Gastroenterology, Graduate School of Medicine, Chiba University, Chiba, Japan
| |
Collapse
|
19
|
Zaiss DM, Minutti CM, Knipper JA. Immune- and non-immune-mediated roles of regulatory T-cells during wound healing. Immunology 2019; 157:190-197. [PMID: 30866049 DOI: 10.1111/imm.13057] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2019] [Accepted: 01/29/2019] [Indexed: 12/16/2022] Open
Abstract
The immune system has a well-established contribution to tissue homeostasis and wound healing. However, in many cases immune responses themselves can cause severe tissue damage. Thus, the question arose to which extent cells of the immune system directly contribute to the process of wound healing and to which extent the resolution of excessive immune responses may indirectly contribute to wound healing. FoxP3-expressing CD4 T-cells, so-called regulatory T-cells (Tregs ), have an important contribution in the regulation of immune responses; and, in recent years, it has been suggested that Tregs next to an immune-regulatory, 'damage-limiting' function may also have an immune-independent 'damage-resolving' direct role in wound healing. In particular, the release of the epidermal growth factor-like growth factor Amphiregulin by tissue-resident Tregs during wound repair suggested such a function. Our recent findings have now revealed that Amphiregulin induces the local release of bio-active transforming growth factor (TGF)β, a cytokine involved both in immune regulation as well as in the process of wound repair. In light of these findings, we discuss whether, by locally activating TGFβ, Treg -derived Amphiregulin may contribute to both wound repair and immune suppression. Furthermore, we propose that Treg -derived Amphiregulin in an autocrine way may enable an IL-33-mediated survival and expansion of tissue-resident Tregs upon injury. Furthermore, Treg -derived Amphiregulin may contribute to a constitutive, low-level release of bio-active TGFβ within tissues, leading to continuous tissue regeneration and to an immune-suppressive environment, which may keep inflammation-prone tissues in an homeostatic state.
Collapse
Affiliation(s)
- Dietmar M Zaiss
- Institute of Immunology and Infection Research, University of Edinburgh, Edinburgh, UK
| | - Carlos M Minutti
- Institute of Immunology and Infection Research, University of Edinburgh, Edinburgh, UK.,Immunobiology Laboratory, The Francis Crick Institute, London, UK
| | - Johanna A Knipper
- Institute of Immunology and Infection Research, University of Edinburgh, Edinburgh, UK
| |
Collapse
|
20
|
|
21
|
Jeevaratnam K, Salvage SC, Li M, Huang CLH. Regulatory actions of 3',5'-cyclic adenosine monophosphate on osteoclast function: possible roles of Epac-mediated signaling. Ann N Y Acad Sci 2018; 1433:18-28. [PMID: 29846007 DOI: 10.1111/nyas.13861] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2018] [Revised: 04/20/2018] [Accepted: 04/26/2018] [Indexed: 02/02/2023]
Abstract
Alterations in cellular levels of the second messenger 3',5'-cyclic adenosine monophosphate ([cAMP]i ) regulate a wide range of physiologically important cellular signaling processes in numerous cell types. Osteoclasts are terminally differentiated, multinucleated cells specialized for bone resorption. Their systemic regulator, calcitonin, triggers morphometrically and pharmacologically distinct retraction (R) and quiescence (Q) effects on cell-spread area and protrusion-retraction motility, respectively, paralleling its inhibition of bone resorption. Q effects were reproduced by cholera toxin-mediated Gs -protein activation known to increase [cAMP]i , unaccompanied by the [Ca2+ ]i changes contrastingly associated with R effects. We explore a hypothesis implicating cAMP signaling involving guanine nucleotide-exchange activation of the small GTPase Ras-proximate-1 (Rap1) by exchange proteins directly activated by cAMP (Epac). Rap1 activates integrin clustering, cell adhesion to bone matrix, associated cytoskeletal modifications and signaling processes, and transmembrane transduction functions. Epac activation enhanced, whereas Epac inhibition or shRNA-mediated knockdown compromised, the appearance of markers for osteoclast differentiation and motility following stimulation by receptor activator of nuclear factor kappa-Β ligand (RANKL). Deficiencies in talin and Rap1 compromised in vivo bone resorption, producing osteopetrotic phenotypes in genetically modified murine models. Translational implications of an Epac-Rap1 signaling hypothesis in relationship to N-bisphosphonate actions on prenylation and membrane localization of small GTPases are discussed.
Collapse
Affiliation(s)
- Kamalan Jeevaratnam
- Faculty of Health and Medical Sciences, University of Surrey, Guildford, UK.,School of Medicine, Perdana University - Royal College of Surgeons Ireland, Selangor, Malaysia
| | - Samantha C Salvage
- Department of Biochemistry, University of Cambridge, Cambridge, UK.,Physiological Laboratory, University of Cambridge, Cambridge, UK
| | - Mengye Li
- Physiological Laboratory, University of Cambridge, Cambridge, UK
| | - Christopher L-H Huang
- Department of Biochemistry, University of Cambridge, Cambridge, UK.,Physiological Laboratory, University of Cambridge, Cambridge, UK
| |
Collapse
|
22
|
Li H, Liu L, Liu C, Zhuang J, Zhou C, Yang J, Gao C, Liu G, Lv Q, Sun C. Deciphering Key Pharmacological Pathways of Qingdai Acting on Chronic Myeloid Leukemia Using a Network Pharmacology-Based Strategy. Med Sci Monit 2018; 24:5668-5688. [PMID: 30108199 PMCID: PMC6106618 DOI: 10.12659/msm.908756] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Qingdai, a traditional Chinese medicine (TCM) used for the treatment of chronic myeloid leukemia (CML) with good efficacy, has been used in China for decades. However, due to the complexity of traditional Chinese medicinal compounds, the pharmacological mechanism of Qingdai needs further research. In this study, we investigated the pharmacological mechanisms of Qingdai in the treatment of CML using network pharmacology approaches. First, components in Qingdai that were selected by pharmacokinetic profiles and biological activity predicted putative targets based on a combination of 2D and 3D similarity measures with known ligands. Then, an interaction network of Qingdai putative targets and known therapeutic targets for the treatment of chronic myeloid leukemia was constructed. By calculating the 4 topological features (degree, betweenness, closeness, and coreness) of each node in the network, we identified the candidate Qingdai targets according to their network topological importance. The composite compounds of Qingdai and the corresponding candidate major targets were further validated by a molecular docking simulation. Seven components in Qingdai were selected and 32 candidate Qingdai targets were identified; these were more frequently involved in cytokine-cytokine receptor interaction, cell cycle, p53 signaling pathway, MAPK signaling pathway, and immune system-related pathways, which all play important roles in the progression of CML. Finally, the molecular docking simulation showed that 23 pairs of chemical components and candidate Qingdai targets had effective binding. This network-based pharmacology study suggests that Qingdai acts through the regulation of candidate targets to interfere with CML and thus regulates the occurrence and development of CML.
Collapse
Affiliation(s)
- Huayao Li
- College of First Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China (mainland)
| | - Lijuan Liu
- College of First Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China (mainland).,Department of Oncology, Affilited Hospital of Weifang Medical University, Weifang, Shandong, China (mainland)
| | - Cun Liu
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China (mainland)
| | - Jing Zhuang
- Departmen of Oncology, Weifang Traditional Chinese Hospital, Weifang, Shandong, China (mainland)
| | - Chao Zhou
- Departmen of Oncology, Weifang Traditional Chinese Hospital, Weifang, Shandong, China (mainland)
| | - Jing Yang
- Departmen of Oncology, Weifang Traditional Chinese Hospital, Weifang, Shandong, China (mainland)
| | - Chundi Gao
- College of First Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China (mainland)
| | - Gongxi Liu
- Departmen of Oncology, Weifang Traditional Chinese Hospital, Weifang, Shandong, China (mainland)
| | - Qingliang Lv
- Department of Interventional Radiology, Weifang People's Hospital, Weifang, Shandong, China (mainland)
| | - Changgang Sun
- Department of Oncology, Affilited Hospital of Weifang Medical University, Weifang, Shandong, China (mainland)
| |
Collapse
|
23
|
Finetti F, Baldari CT. The immunological synapse as a pharmacological target. Pharmacol Res 2018; 134:118-133. [PMID: 29898412 DOI: 10.1016/j.phrs.2018.06.009] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Revised: 05/25/2018] [Accepted: 06/07/2018] [Indexed: 12/29/2022]
Abstract
The development of T cell mediated immunity relies on the assembly of a highly specialized interface between T cell and antigen presenting cell (APC), known as the immunological synapse (IS). IS assembly is triggered when the T cell receptor (TCR) binds to specific peptide antigen presented in association to the major histocompatibility complex (MHC) by the APC, and is followed by the spatiotemporal dynamic redistribution of TCR, integrins, co-stimulatory receptors and signaling molecules, allowing for the fine-tuning and integration of the signals that lead to T cell activation. The knowledge acquired to date about the mechanisms of IS assembly underscores this structure as a robust pharmacological target. The activity of molecules involved in IS assembly and function can be targeted by specific compounds to modulate the immune response in a number of disorders, including cancers and autoimmune diseases, or in transplanted patients. Here, we will review the state-of-the art of the current therapies which exploit the IS to modulate the immune response.
Collapse
Affiliation(s)
- Francesca Finetti
- Department of Life Sciences, University of Siena, via A. Moro 2, Siena, 53100, Italy.
| | - Cosima T Baldari
- Department of Life Sciences, University of Siena, via A. Moro 2, Siena, 53100, Italy
| |
Collapse
|
24
|
GWAS analysis using interspecific backcross progenies reveals superior blue catfish alleles responsible for strong resistance against enteric septicemia of catfish. Mol Genet Genomics 2018; 293:1107-1120. [PMID: 29737402 DOI: 10.1007/s00438-018-1443-4] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2017] [Accepted: 05/02/2018] [Indexed: 12/11/2022]
Abstract
Infectious diseases pose significant threats to the catfish industry. Enteric septicemia of catfish (ESC) caused by Edwardsiella ictaluri is the most devastating disease for catfish aquaculture, causing huge economic losses annually. Channel catfish and blue catfish exhibit great contrast in resistance against ESC, with channel catfish being highly susceptible and blue catfish being highly resistant. As such, the interspecific backcross progenies provide an ideal system for the identification of quantitative trait locus (QTL). We previously reported one significant QTL on linkage group (LG) 1 using the third-generation backcrosses, but the number of founders used to make the second- and third-generation backcross progenies was very small. Although the third-generation backcross progenies provided a greater power for fine mapping than the first-generation backcrosses, some major QTL for disease resistance may have been missing due to the small numbers of founders used to produce the higher generation backcrosses. In this study, we performed a genome-wide association study using first-generation backcrosses with the catfish 690 K SNP arrays to identify additional ESC disease resistance QTL, especially those at the species level. Two genomic regions on LG1 and LG23 were determined to be significantly associated with ESC resistance as revealed by a mixed linear model and family-based association test. Examination of the resistance alleles indicated their origin from blue catfish, indicating that at least two major disease resistance loci exist among blue catfish populations. Upon further validation, markers linked with major ESC disease resistance QTL should be useful for marker-assisted introgression, allowing development of highly ESC resistant breeds of catfish.
Collapse
|
25
|
T Follicular Helper Cell-Germinal Center B Cell Interaction Strength Regulates Entry into Plasma Cell or Recycling Germinal Center Cell Fate. Immunity 2018; 48:702-715.e4. [DOI: 10.1016/j.immuni.2018.03.027] [Citation(s) in RCA: 166] [Impact Index Per Article: 23.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2017] [Revised: 01/15/2018] [Accepted: 03/23/2018] [Indexed: 12/17/2022]
|
26
|
Yuan J, Wang Z, Zou D, Peng Q, Peng R, Zou F. Expression profiling of planarians shed light on a dual role of programmed cell death during the regeneration. J Cell Biochem 2018; 119:5875-5884. [PMID: 29575081 DOI: 10.1002/jcb.26779] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2017] [Accepted: 02/02/2018] [Indexed: 01/20/2023]
Abstract
Most animals hold the ability to regenerate damaged cells, tissues, and even any lost part of their bodies. To date, there is little known about the precise regulatory mechanism of regeneration and many fundamental questions remain unanswered. To further understand the precise regulatory mechanism of regeneration, we used planarian Dugesia japonica as a model and sequenced the transcriptomes of their regenerated tissues at different regeneration stages. Through de novo assembly and expression profiling, we found that Heat shock protein and MAPK pathway were involved into early response of regeneration in D. japonica. In addition, immune response, cell proliferation, and migration were activated during regeneration. Of notes, our results revealed a specific functional role of programmed cell death (PCD) in regeneration of D. japonica. PCD may not only remove the damaged and superfluous tissues for further patterning with regenerated tissues, but also provide signals to trigger neoblasts proliferation and differentiation directly. Together, our results revealed Heat shock protein and MAPK pathway mediated early response of regeneration and found a dual role of PCD in regeneration D. japonica. Meanwhile, we constructed regulatory networks of apoptosis, autophagy, and related signaling pathways and proposed a schematic model, which provided a global landscape of regeneration.
Collapse
Affiliation(s)
- Junsong Yuan
- College of Life Sciences, Sichuan University, Chengdu, Sichuan, P.R. China
| | - Zhihong Wang
- College of Life Sciences, Sichuan University, Chengdu, Sichuan, P.R. China
| | - Di Zou
- College of Life Sciences, Sichuan University, Chengdu, Sichuan, P.R. China
| | - Quekun Peng
- Department of Biomedical Science, Chengdu Medical College, Chengdu, Sichuan, China
| | - Rui Peng
- College of Life Sciences, Sichuan University, Chengdu, Sichuan, P.R. China
| | - Fangdong Zou
- College of Life Sciences, Sichuan University, Chengdu, Sichuan, P.R. China
| |
Collapse
|
27
|
Artificial Methods for T Cell Activation: Critical Tools in T Cell Biology and T Cell Immunotherapy. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1064:207-219. [PMID: 30471035 DOI: 10.1007/978-981-13-0445-3_13] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Antigen-specific immunity conferred by T lymphocytes is a result of complex molecular interactions at the immunological synapse. A variety of biomimetic approaches have been devised to artificially induce T cell activation either to study the T cell biology or to expand and prime the therapeutic T cell populations. Here we first briefly review the molecular and cellular, structural and phenotypical bases that are involved in T cell activation. The artificial methods for T cell activation are then discussed in two grand categories, the soluble (3D) and the surface-anchored (2D) platforms with their design parameters. With the growing number of successful adoptive T cell therapies, the spurring demands for effective and safe T cell expansion as well as precise control over resulting T cell functions and phenotypes warrant the extensions of engineering parameters in the development of novel methodologies for T cell activation.
Collapse
|
28
|
Xiong Y, Ye C, Yang N, Li M, Liu H. Ubc9 Binds to ADAP and Is Required for Rap1 Membrane Recruitment, Rac1 Activation, and Integrin-Mediated T Cell Adhesion. THE JOURNAL OF IMMUNOLOGY 2017; 199:4142-4154. [PMID: 29127148 DOI: 10.4049/jimmunol.1700572] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/21/2017] [Accepted: 10/16/2017] [Indexed: 11/19/2022]
Abstract
Although the immune adaptor adhesion and degranulation-promoting adaptor protein (ADAP) acts as a key mediator of integrin inside-out signaling leading to T cell adhesion, the regulation of this adaptor during integrin activation and clustering remains unclear. We now identify Ubc9, the sole small ubiquitin-related modifier E2 conjugase, as an essential regulator of ADAP where it is required for TCR-induced membrane recruitment of the small GTPase Rap1 and its effector protein RapL and for activation of the small GTPase Rac1 in T cell adhesion. We show that Ubc9 interacted directly with ADAP in vitro and in vivo, and the association was increased in response to anti-CD3 stimulation. The Ubc9-binding domain on ADAP was mapped to a nuclear localization sequence (aa 674-700) within ADAP. Knockdown of Ubc9 by short hairpin RNA or expression of the Ubc9-binding-deficient ADAP mutant significantly decreased TCR-induced integrin adhesion to ICAM-1 and fibronectin, as well as LFA-1 clustering, although it had little effect on the TCR proximal signaling responses and TCR-induced IL-2 transcription. Furthermore, downregulation of Ubc9 impaired TCR-mediated Rac1 activation and attenuated the membrane targeting of Rap1 and RapL, but not Rap1-interacting adaptor molecule. Taken together, our data demonstrate for the first time, to our knowledge, that Ubc9 acts as a functional binding partner of ADAP and plays a selective role in integrin-mediated T cell adhesion via modulation of Rap1-RapL membrane recruitment and Rac1 activation.
Collapse
Affiliation(s)
- Yiwei Xiong
- Department of Biological Sciences, Xi'an Jiaotong-Liverpool University, Suzhou, Jiangsu 215123, China; and
| | - Chengjin Ye
- Department of Veterinary Medicine, Zhejiang A & F University, Lin'an, Hangzhou, Zhejiang 311300, China
| | - Naiqi Yang
- Department of Biological Sciences, Xi'an Jiaotong-Liverpool University, Suzhou, Jiangsu 215123, China; and
| | - Madanqi Li
- Department of Biological Sciences, Xi'an Jiaotong-Liverpool University, Suzhou, Jiangsu 215123, China; and
| | - Hebin Liu
- Department of Biological Sciences, Xi'an Jiaotong-Liverpool University, Suzhou, Jiangsu 215123, China; and .,Department of Veterinary Medicine, Zhejiang A & F University, Lin'an, Hangzhou, Zhejiang 311300, China
| |
Collapse
|
29
|
Xiao Y, Zhao S, Li B. Aplastic anemia is related to alterations in T cell receptor signaling. Stem Cell Investig 2017; 4:85. [PMID: 29167806 DOI: 10.21037/sci.2017.09.07] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2017] [Accepted: 09/15/2017] [Indexed: 01/05/2023]
Abstract
Aplastic anemia (AA) is a disease characterized by bone marrow hematopoietic dysfunction and peripheral blood pancytopenia, which is thought to be mediated by an abnormal T cell-induced immune response. T cell receptor (TCR) signaling is pivotal for T cell development and function. An aberrant TCR signaling leads to an unbalanced immune system that can result in a range of immune-related disorders, including autoimmune diseases, chronic infections, and tumors. In this article, we briefly review the T cell immune pathophysiology of AA, the physiology of normal TCR signaling and its regulatory factors, and clinical and laboratory findings of TCR signaling molecules and their regulatory factors in AA.
Collapse
Affiliation(s)
- Yankai Xiao
- Key Laboratory for Regenerative Medicine of Ministry of Education, Jinan University, Guangzhou 510632, China.,Institute of Hematology, Medical College, Jinan University, Guangzhou 510632, China
| | - Suwen Zhao
- Key Laboratory for Regenerative Medicine of Ministry of Education, Jinan University, Guangzhou 510632, China.,Institute of Hematology, Medical College, Jinan University, Guangzhou 510632, China
| | - Bo Li
- Institute of Hematology, Medical College, Jinan University, Guangzhou 510632, China
| |
Collapse
|
30
|
Sackstein R, Schatton T, Barthel SR. T-lymphocyte homing: an underappreciated yet critical hurdle for successful cancer immunotherapy. J Transl Med 2017; 97:669-697. [PMID: 28346400 PMCID: PMC5446300 DOI: 10.1038/labinvest.2017.25] [Citation(s) in RCA: 144] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2016] [Revised: 01/17/2017] [Accepted: 01/22/2017] [Indexed: 12/13/2022] Open
Abstract
Advances in cancer immunotherapy have offered new hope for patients with metastatic disease. This unfolding success story has been exemplified by a growing arsenal of novel immunotherapeutics, including blocking antibodies targeting immune checkpoint pathways, cancer vaccines, and adoptive cell therapy (ACT). Nonetheless, clinical benefit remains highly variable and patient-specific, in part, because all immunotherapeutic regimens vitally hinge on the capacity of endogenous and/or adoptively transferred T-effector (Teff) cells, including chimeric antigen receptor (CAR) T cells, to home efficiently into tumor target tissue. Thus, defects intrinsic to the multi-step T-cell homing cascade have become an obvious, though significantly underappreciated contributor to immunotherapy resistance. Conspicuous have been low intralesional frequencies of tumor-infiltrating T-lymphocytes (TILs) below clinically beneficial threshold levels, and peripheral rather than deep lesional TIL infiltration. Therefore, a Teff cell 'homing deficit' may arguably represent a dominant factor responsible for ineffective immunotherapeutic outcomes, as tumors resistant to immune-targeted killing thrive in such permissive, immune-vacuous microenvironments. Fortunately, emerging data is shedding light into the diverse mechanisms of immune escape by which tumors restrict Teff cell trafficking and lesional penetrance. In this review, we scrutinize evolving knowledge on the molecular determinants of Teff cell navigation into tumors. By integrating recently described, though sporadic information of pivotal adhesive and chemokine homing signatures within the tumor microenvironment with better established paradigms of T-cell trafficking under homeostatic or infectious disease scenarios, we seek to refine currently incomplete models of Teff cell entry into tumor tissue. We further summarize how cancers thwart homing to escape immune-mediated destruction and raise awareness of the potential impact of immune checkpoint blockers on Teff cell homing. Finally, we speculate on innovative therapeutic opportunities for augmenting Teff cell homing capabilities to improve immunotherapy-based tumor eradication in cancer patients, with special focus on malignant melanoma.
Collapse
Affiliation(s)
- Robert Sackstein
- Department of Dermatology, Brigham & Women’s Hospital, Harvard Medical School, Boston, MA, 02115, USA,Department of Medicine, Brigham & Women’s Hospital, Harvard Medical School, Boston, MA, 02115, USA,Harvard Skin Disease Research Center, Brigham & Women’s Hospital, Harvard Medical School, Boston, MA, 02115, USA,Program of Excellence in Glycosciences, Harvard Medical School, 77 Avenue Louis Pasteur, Rm 671, Boston, MA 02115, USA
| | - Tobias Schatton
- Department of Dermatology, Brigham & Women’s Hospital, Harvard Medical School, Boston, MA, 02115, USA,Harvard Skin Disease Research Center, Brigham & Women’s Hospital, Harvard Medical School, Boston, MA, 02115, USA,Harvard Stem Cell Institute, Harvard Medical School, Boston, MA 02115, USA,Department of Medicine, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Steven R. Barthel
- Department of Dermatology, Brigham & Women’s Hospital, Harvard Medical School, Boston, MA, 02115, USA,Harvard Skin Disease Research Center, Brigham & Women’s Hospital, Harvard Medical School, Boston, MA, 02115, USA,Harvard Stem Cell Institute, Harvard Medical School, Boston, MA 02115, USA,Correspondence to: Dr. Steven R. Barthel, Harvard Institutes of Medicine, Rm. 673B, 77 Avenue Louis Pasteur, Boston, MA 02115;
| |
Collapse
|
31
|
Recruitment of calcineurin to the TCR positively regulates T cell activation. Nat Immunol 2016; 18:196-204. [PMID: 27941787 DOI: 10.1038/ni.3640] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2016] [Accepted: 11/21/2016] [Indexed: 12/17/2022]
Abstract
Calcineurin is a phosphatase whose primary targets in T cells are NFAT transcription factors, and inhibition of calcineurin activity by treatment with cyclosporin A (CsA) or FK506 is a cornerstone of immunosuppressive therapies. Here we found that calcineurin was recruited to the T cell antigen receptor (TCR) signaling complex, where it reversed inhibitory phosphorylation of the tyrosine kinase Lck on Ser59 (LckS59). Loss of calcineurin activity impaired phosphorylation of Tyr493 of the tyrosine kinase ZAP-70 (ZAP-70Y493), as well as some downstream pathways in a manner consistent with signaling in cells expressing LckS59A (Lck that cannot be phosphorylated) or LckS59E (a phosphomimetic mutant). Notably, CsA inhibited integrin-LFA-1-dependent and NFAT-independent adhesion of T cells to the intercellular adhesion molecule ICAM-1, with little effect on cells expressing mutant Lck. These results provide new understanding of how widely used immunosuppressive drugs interfere with essential processes in the immune response.
Collapse
|
32
|
Moura Rosa P, Gopalakrishnan N, Ibrahim H, Haug M, Halaas Ø. The intercell dynamics of T cells and dendritic cells in a lymph node-on-a-chip flow device. LAB ON A CHIP 2016; 16:3728-40. [PMID: 27560793 DOI: 10.1039/c6lc00702c] [Citation(s) in RCA: 66] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/12/2023]
Abstract
T cells play a central role in immunity towards cancer and infectious diseases. T cell responses are initiated in the T cell zone of the lymph node (LN), where resident antigen-bearing dendritic cells (DCs) prime and activate antigen-specific T cells passing by. In the present study, we investigated the T cell : DC interaction in a microfluidic device to understand the intercellular dynamics and physiological conditions in the LN. We show random migration of antigen-specific T cells onto the antigen-presenting DC monolayer independent of the flow direction with a mean T cell : DC dwell time of 12.8 min and a mean velocity of 6 μm min(-1). Furthermore, we investigated the antigen specific vs. unspecific attachment and detachment of CD8(+) and CD4(+) T cells to DCs under varying shear stress. In our system, CD4(+) T cells showed long stable contacts with APCs, whereas CD8(+) T cells presented transient interactions with DCs. By varying the shear stress from 0.01 to 100 Dyn cm(-2), it was also evident that there was a much stronger attachment of antigen-specific than unspecific T cells to stationary DCs up to 1-12 Dyn cm(-2). The mechanical force of the cell : cell interaction associated with the pMHC-TCR match under controlled tangential shear force was estimated to be in the range of 0.25-4.8 nN. Finally, upon performing attachment & detachment tests, there was a steady accumulation of antigen specific CD8(+) T cells and CD4(+) T cells on DCs at low shear stresses, which were released at a stress of 12 Dyn cm(-2). This microphysiological model provides new possibilities to recreate a controlled mechanical force threshold of pMHC-TCR binding, allowing the investigation of intercellular signalling of immune synapses and therapeutic targets for immunotherapy.
Collapse
Affiliation(s)
- Patrícia Moura Rosa
- Department of Cancer Research and Molecular Medicine, Norwegian University of Science and Technology, 7489 Trondheim, Norway.
| | | | | | | | | |
Collapse
|
33
|
Fiege JK, Beura LK, Burbach BJ, Shimizu Y. Adhesion- and Degranulation-Promoting Adapter Protein Promotes CD8 T Cell Differentiation and Resident Memory Formation and Function during an Acute Infection. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2016; 197:2079-89. [PMID: 27521337 PMCID: PMC5010998 DOI: 10.4049/jimmunol.1501805] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/10/2015] [Accepted: 07/13/2016] [Indexed: 11/19/2022]
Abstract
During acute infections, naive Ag-specific CD8 T cells are activated and differentiate into effector T cells, most of which undergo contraction after pathogen clearance. A small population of CD8 T cells persists as memory to protect against future infections. We investigated the role of adhesion- and degranulation-promoting adapter protein (ADAP) in promoting CD8 T cell responses to a systemic infection. Naive Ag-specific CD8 T cells lacking ADAP exhibited a modest expansion defect early after Listeria monocytogenes or vesicular stomatitis virus infection but comparable cytolytic function at the peak of response. However, reduced numbers of ADAP-deficient CD8 T cells were present in the spleen after the peak of the response. ADAP deficiency resulted in a greater frequency of CD127(+) CD8 memory precursors in secondary lymphoid organs during the contraction phase. Reduced numbers of ADAP-deficient killer cell lectin-like receptor G1(-) CD8 resident memory T (TRM) cell precursors were present in a variety of nonlymphoid tissues at the peak of the immune response, and consequently the total numbers of ADAP-deficient TRM cells were reduced at memory time points. TRM cells that did form in the absence of ADAP were defective in effector molecule expression. ADAP-deficient TRM cells exhibited impaired effector function after Ag rechallenge, correlating with defects in their ability to form T cell-APC conjugates. However, ADAP-deficient TRM cells responded to TGF-β signals and recruited circulating memory CD8 T cells. Thus, ADAP regulates CD8 T cell differentiation events following acute pathogen challenge that are critical for the formation and selected functions of TRM cells in nonlymphoid tissues.
Collapse
Affiliation(s)
- Jessica K Fiege
- Department of Laboratory Medicine and Pathology, Center for Immunology, Masonic Cancer Center, University of Minnesota Medical School, Minneapolis, MN 55455; and
| | - Lalit K Beura
- Department of Microbiology and Immunology, Center for Immunology, University of Minnesota Medical School, Minneapolis, MN 55455
| | - Brandon J Burbach
- Department of Laboratory Medicine and Pathology, Center for Immunology, Masonic Cancer Center, University of Minnesota Medical School, Minneapolis, MN 55455; and
| | - Yoji Shimizu
- Department of Laboratory Medicine and Pathology, Center for Immunology, Masonic Cancer Center, University of Minnesota Medical School, Minneapolis, MN 55455; and
| |
Collapse
|
34
|
Köchl R, Thelen F, Vanes L, Brazao TF, Fountain K, Xie J, Huang CL, Lyck R, Stein JV, Tybulewicz VLJ. WNK1 kinase balances T cell adhesion versus migration in vivo. Nat Immunol 2016; 17:1075-83. [PMID: 27400149 PMCID: PMC4994873 DOI: 10.1038/ni.3495] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2016] [Accepted: 05/25/2016] [Indexed: 12/15/2022]
Abstract
Adhesion and migration of T cells are controlled by chemokines and by adhesion molecules, especially integrins, and have critical roles in the normal physiological function of T lymphocytes. Using an RNA-mediated interference screen, we identified the WNK1 kinase as a regulator of both integrin-mediated adhesion and T cell migration. We found that WNK1 is a negative regulator of integrin-mediated adhesion, whereas it acts as a positive regulator of migration via the kinases OXSR1 and STK39 and the ion co-transporter SLC12A2. WNK1-deficient T cells home less efficiently to lymphoid organs and migrate more slowly through them. Our results reveal that a pathway previously known only to regulate salt homeostasis in the kidney functions to balance T cell adhesion and migration.
Collapse
Affiliation(s)
| | - Flavian Thelen
- Theodor Kocher Institute, University of Bern, Bern, Switzerland
| | | | | | | | - Jian Xie
- University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Chou-Long Huang
- University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Ruth Lyck
- Theodor Kocher Institute, University of Bern, Bern, Switzerland
| | - Jens V. Stein
- Theodor Kocher Institute, University of Bern, Bern, Switzerland
| | | |
Collapse
|
35
|
Fu H, Ward EJ, Marelli-Berg FM. Mechanisms of T cell organotropism. Cell Mol Life Sci 2016; 73:3009-33. [PMID: 27038487 PMCID: PMC4951510 DOI: 10.1007/s00018-016-2211-4] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2016] [Revised: 03/21/2016] [Accepted: 03/22/2016] [Indexed: 02/06/2023]
Abstract
Protective immunity relies upon T cell differentiation and subsequent migration to target tissues. Similarly, immune homeostasis requires the localization of regulatory T cells (Tregs) to the sites where immunity takes place. While naïve T lymphocytes recirculate predominantly in secondary lymphoid tissue, primed T cells and activated Tregs must traffic to the antigen rich non-lymphoid tissue to exert effector and regulatory responses, respectively. Following priming in draining lymph nodes, T cells acquire the 'homing receptors' to facilitate their access to specific tissues and organs. An additional level of topographic specificity is provided by T cells receptor recognition of antigen displayed by the endothelium. Furthermore, co-stimulatory signals (such as those induced by CD28) have been shown not only to regulate T cell activation and differentiation, but also to orchestrate the anatomy of the ensuing T cell response. We here review the molecular mechanisms supporting trafficking of both effector and regulatory T cells to specific antigen-rich tissues.
Collapse
Affiliation(s)
- Hongmei Fu
- William Harvey Research Institute, Heart Centre, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, Charterhouse Square, London, EC1M 6BQ, UK
| | - Eleanor Jayne Ward
- William Harvey Research Institute, Heart Centre, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, Charterhouse Square, London, EC1M 6BQ, UK
| | - Federica M Marelli-Berg
- William Harvey Research Institute, Heart Centre, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, Charterhouse Square, London, EC1M 6BQ, UK.
| |
Collapse
|
36
|
Lin F, Xue D, Xie T, Pan Z. HMGB1 promotes cellular chemokine synthesis and potentiates mesenchymal stromal cell migration via Rap1 activation. Mol Med Rep 2016; 14:1283-9. [PMID: 27314424 DOI: 10.3892/mmr.2016.5398] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2015] [Accepted: 04/08/2016] [Indexed: 11/06/2022] Open
Abstract
The migration of mesenchymal stem cells (MSCs) and osteogenic differentiation occupy an important role in fracture healing. High mobility group box 1 (HMGB1), a widely distributed inflammatory factor in fractures, has been confirmed to act as a chemoattractant to MSCs. To investigate the effect of HMGB1 on MSC migration and the underlying mechanism, the synthesis of MSC chemokines, and the consequent activation of signaling pathways following HMGB1 stimulation, were evaluated. A Quantibody® array was performed to determine which chemokines were secreted from MSCs with or without treatment with HMGB1. The results indicated differential chemokine synthesis by MSCs following treatment with HMGB1, including that of CCL4 and CCL13. In addition, the Ras‑associated protein‑1 (Rap1) signaling pathway was markedly activated in the HMGB1‑treated groups, suggesting that HMGB1 may enhance the migrational ability of MSCs via Rap1 activation. Furthermore, HMGB1 was able to promote the secretion of various chemokines derived from MSCs, which would, in turn, increase the mobility of MSCs. Taken together, these results provide a mechanistic basis for developing novel approaches to promote fracture healing.
Collapse
Affiliation(s)
- Feng Lin
- Department of Orthopedics, The First People's Hospital of Xiaoshan, Hangzhou, Zhejiang 311200, P.R. China
| | - Deting Xue
- Department of Orthopedics, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310009, P.R. China
| | - Tao Xie
- Department of Orthopedics, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310009, P.R. China
| | - Zhijun Pan
- Department of Orthopedics, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310009, P.R. China
| |
Collapse
|
37
|
Rajasekaran K, Riese MJ, Rao S, Wang L, Thakar MS, Sentman CL, Malarkannan S. Signaling in Effector Lymphocytes: Insights toward Safer Immunotherapy. Front Immunol 2016; 7:176. [PMID: 27242783 PMCID: PMC4863891 DOI: 10.3389/fimmu.2016.00176] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2016] [Accepted: 04/20/2016] [Indexed: 12/15/2022] Open
Abstract
Receptors on T and NK cells systematically propagate highly complex signaling cascades that direct immune effector functions, leading to protective immunity. While extensive studies have delineated hundreds of signaling events that take place upon receptor engagement, the precise molecular mechanism that differentially regulates the induction or repression of a unique effector function is yet to be fully defined. Such knowledge can potentiate the tailoring of signal transductions and transform cancer immunotherapies. Targeted manipulations of signaling cascades can augment one effector function such as antitumor cytotoxicity while contain the overt generation of pro-inflammatory cytokines that contribute to treatment-related toxicity such as “cytokine storm” and “cytokine-release syndrome” or lead to autoimmune diseases. Here, we summarize how individual signaling molecules or nodes may be optimally targeted to permit selective ablation of toxic immune side effects.
Collapse
Affiliation(s)
- Kamalakannan Rajasekaran
- Laboratory of Molecular Immunology and Immunotherapy, Blood Research Institute , Milwaukee, WI , USA
| | - Matthew J Riese
- Laboratory of Lymphocyte Biology, Blood Research Institute, Milwaukee, WI, USA; Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, USA; Department of Microbiology and Molecular Genetics, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Sridhar Rao
- Laboratory of Stem Cell Transcriptional Regulation, Blood Research Institute, Milwaukee, WI, USA; Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Li Wang
- Department of Medicine, Medical College of Wisconsin , Milwaukee, WI , USA
| | - Monica S Thakar
- Laboratory of Molecular Immunology and Immunotherapy, Blood Research Institute, Milwaukee, WI, USA; Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Charles L Sentman
- Department of Microbiology and Immunology, Center for Synthetic Immunity at the Geisel School of Medicine at Dartmouth , Lebanon, NH , USA
| | - Subramaniam Malarkannan
- Laboratory of Molecular Immunology and Immunotherapy, Blood Research Institute, Milwaukee, WI, USA; Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, USA; Department of Microbiology and Molecular Genetics, Medical College of Wisconsin, Milwaukee, WI, USA; Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI, USA
| |
Collapse
|
38
|
Martos-Sitcha JA, Mancera JM, Calduch-Giner JA, Yúfera M, Martínez-Rodríguez G, Pérez-Sánchez J. Unraveling the Tissue-Specific Gene Signatures of Gilthead Sea Bream (Sparus aurata L.) after Hyper- and Hypo-Osmotic Challenges. PLoS One 2016; 11:e0148113. [PMID: 26828928 PMCID: PMC4734831 DOI: 10.1371/journal.pone.0148113] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2015] [Accepted: 01/13/2016] [Indexed: 11/19/2022] Open
Abstract
A custom microarray was used for the transcriptomic profiling of liver, gills and hypothalamus in response to hypo- (38‰ → 5‰) or hyper- (38‰ → 55‰) osmotic challenges (7 days after salinity transfer) in gilthead sea bream (Sparus aurata) juveniles. The total number of differentially expressed genes was 777. Among them, 341 and 310 were differentially expressed in liver after hypo- and hyper-osmotic challenges, respectively. The magnitude of changes was lower in gills and hypothalamus with around 131 and 160 responsive genes in at least one osmotic stress condition, respectively. Regardless of tissue, a number of genes were equally regulated in either hypo- and hyper-osmotic challenges: 127 out of 524 in liver, 11 out of 131 in gills and 19 out of 160 in hypothalamus. In liver and gills, functional analysis of differentially expressed genes recognized two major clusters of overlapping canonical pathways that were mostly related to “Energy Metabolism” and “Oxidative Stress”. The later cluster was represented in all the analyzed tissues, including the hypothalamus, where differentially expressed genes related to “Cell and tissue architecture” were also over-represented. Overall the response for “Energy Metabolism” was the up-regulation, whereas for oxidative stress-related genes the type of response was highly dependent of tissue. These results support common and different osmoregulatory responses in the three analyzed tissues, helping to load new allostatic conditions or even to return to basal levels after hypo- or hyper-osmotic challenges according to the different physiological role of each tissue.
Collapse
Affiliation(s)
- Juan Antonio Martos-Sitcha
- Instituto de Ciencias Marinas de Andalucía, Consejo Superior de Investigaciones Científicas (ICMAN-CSIC), E-11519, Puerto Real (Cádiz), Spain
- Department of Biology, Faculty of Marine and Environmental Sciences, Campus de Excelencia Internacional del Mar (CEI-MAR), University of Cádiz, E-11519, Puerto Real (Cádiz), Spain
- * E-mail:
| | - Juan Miguel Mancera
- Department of Biology, Faculty of Marine and Environmental Sciences, Campus de Excelencia Internacional del Mar (CEI-MAR), University of Cádiz, E-11519, Puerto Real (Cádiz), Spain
| | - Josep Alvar Calduch-Giner
- Nutrigenomics and Fish Growth Endocrinology Group, Institute of Aquaculture Torre de la Sal, Consejo Superior de Investigaciones Científicas (IATS-CSIC), Ribera de Cabanes, E-12595, Castellón, Spain
| | - Manuel Yúfera
- Instituto de Ciencias Marinas de Andalucía, Consejo Superior de Investigaciones Científicas (ICMAN-CSIC), E-11519, Puerto Real (Cádiz), Spain
| | - Gonzalo Martínez-Rodríguez
- Instituto de Ciencias Marinas de Andalucía, Consejo Superior de Investigaciones Científicas (ICMAN-CSIC), E-11519, Puerto Real (Cádiz), Spain
| | - Jaume Pérez-Sánchez
- Nutrigenomics and Fish Growth Endocrinology Group, Institute of Aquaculture Torre de la Sal, Consejo Superior de Investigaciones Científicas (IATS-CSIC), Ribera de Cabanes, E-12595, Castellón, Spain
| |
Collapse
|
39
|
Niedergang F, Di Bartolo V, Alcover A. Comparative Anatomy of Phagocytic and Immunological Synapses. Front Immunol 2016; 7:18. [PMID: 26858721 PMCID: PMC4729869 DOI: 10.3389/fimmu.2016.00018] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2015] [Accepted: 01/14/2016] [Indexed: 11/17/2022] Open
Abstract
The generation of phagocytic cups and immunological synapses are crucial events of the innate and adaptive immune responses, respectively. They are triggered by distinct immune receptors and performed by different cell types. However, growing experimental evidence shows that a very close series of molecular and cellular events control these two processes. Thus, the tight and dynamic interplay between receptor signaling, actin and microtubule cytoskeleton, and targeted vesicle traffic are all critical features to build functional phagosomes and immunological synapses. Interestingly, both phagocytic cups and immunological synapses display particular spatial and temporal patterns of receptors and signaling molecules, leading to the notion of “phagocytic synapse.” Here, we discuss both types of structures, their organization, and the mechanisms by which they are generated and regulated.
Collapse
Affiliation(s)
- Florence Niedergang
- U1016, Institut Cochin, INSERM, Paris, France; UMR 8104, CNRS, Paris, France; Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Vincenzo Di Bartolo
- Lymphocyte Cell Biology Unit, Department of Immunology, Institut Pasteur, Paris, France; U1221, INSERM, Paris, France
| | - Andrés Alcover
- Lymphocyte Cell Biology Unit, Department of Immunology, Institut Pasteur, Paris, France; U1221, INSERM, Paris, France
| |
Collapse
|
40
|
Martinelli R, Carman CV. An Endothelial Planar Cell Model for Imaging Immunological Synapse Dynamics. J Vis Exp 2015:e53288. [PMID: 26780561 DOI: 10.3791/53288] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022] Open
Abstract
Adaptive immunity is regulated by dynamic interactions between T cells and antigen presenting cells ('APCs') referred to as 'immunological synapses'. Within these intimate cell-cell interfaces discrete sub-cellular clusters of MHC/Ag-TCR, F-actin, adhesion and signaling molecules form and remodel rapidly. These dynamics are thought to be critical determinants of both the efficiency and quality of the immune responses that develop and therefore of protective versus pathologic immunity. Current understanding of immunological synapses with physiologic APCs is limited by the inadequacy of the obtainable imaging resolution. Though artificial substrate models (e.g., planar lipid bilayers) offer excellent resolution and have been extremely valuable tools, they are inherently non-physiologic and oversimplified. Vascular and lymphatic endothelial cells have emerged as an important peripheral tissue (or stromal) compartment of 'semi-professional APCs'. These APCs (which express most of the molecular machinery of professional APCs) have the unique feature of forming virtually planar cell surface and are readily transfectable (e.g., with fluorescent protein reporters). Herein a basic approach to implement endothelial cells as a novel and physiologic 'planar cellular APC model' for improved imaging and interrogation of fundamental antigenic signaling processes will be described.
Collapse
|
41
|
Carman CV, Martinelli R. T Lymphocyte-Endothelial Interactions: Emerging Understanding of Trafficking and Antigen-Specific Immunity. Front Immunol 2015; 6:603. [PMID: 26635815 PMCID: PMC4657048 DOI: 10.3389/fimmu.2015.00603] [Citation(s) in RCA: 143] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2015] [Accepted: 11/10/2015] [Indexed: 12/26/2022] Open
Abstract
Antigen-specific immunity requires regulated trafficking of T cells in and out of diverse tissues in order to orchestrate lymphocyte development, immune surveillance, responses, and memory. The endothelium serves as a unique barrier, as well as a sentinel, between the blood and the tissues, and as such it plays an essential locally tuned role in regulating T cell migration and information exchange. While it is well established that chemoattractants and adhesion molecules are major determinants of T cell trafficking, emerging studies have now enumerated a large number of molecular players as well as a range of discrete cellular remodeling activities (e.g., transmigratory cups and invadosome-like protrusions) that participate in directed migration and pathfinding by T cells. In addition to providing trafficking cues, intimate cell-cell interaction between lymphocytes and endothelial cells provide instruction to T cells that influence their activation and differentiation states. Perhaps the most intriguing and underappreciated of these "sentinel" roles is the ability of the endothelium to act as a non-hematopoietic "semiprofessional" antigen-presenting cell. Close contacts between circulating T cells and antigen-presenting endothelium may play unique non-redundant roles in shaping adaptive immune responses within the periphery. A better understanding of the mechanisms directing T cell trafficking and the antigen-presenting role of the endothelium may not only increase our knowledge of the adaptive immune response but also empower the utility of emerging immunomodulatory therapeutics.
Collapse
Affiliation(s)
- Christopher V Carman
- Center for Vascular Biology Research, Department of Medicine and Emergency Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School , Boston, MA , USA
| | - Roberta Martinelli
- Center for Vascular Biology Research, Department of Medicine and Emergency Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School , Boston, MA , USA
| |
Collapse
|
42
|
Abstract
Immunological memory is a key feature of adaptive immunity. It provides the organism with long-lived and robust protection against infection. In organ transplantation, memory T cells pose a significant threat by causing allograft rejection that is generally resistant to immunosuppressive therapy. Therefore, a more thorough understanding of memory T cell biology is needed to improve the survival of transplanted organs without compromising the host’s ability to fight infections. This review will focus on the mechanisms by which memory T cells migrate to the site where their target antigen is present, with particular emphasis on their migration to transplanted organs. First, we will define the known subsets of memory T cells (central, effector, and tissue resident) and their circulation patterns. Second, we will review the cellular and molecular mechanisms by which memory T cells migrate to inflamed and non-inflamed tissues and highlight the emerging paradigm of antigen-driven, trans-endothelial migration. Third, we will discuss the relevance of this knowledge to organ transplantation and the prevention or treatment of allograft rejection.
Collapse
Affiliation(s)
- Qianqian Zhang
- Tsinghua University School of Medicine , Beijing , China ; University of Pittsburgh School of Medicine , Pittsburgh, PA , USA
| | - Fadi G Lakkis
- University of Pittsburgh School of Medicine , Pittsburgh, PA , USA
| |
Collapse
|
43
|
Involvement of the Rac1-IRSp53-Wave2-Arp2/3 Signaling Pathway in HIV-1 Gag Particle Release in CD4 T Cells. J Virol 2015; 89:8162-81. [PMID: 26018170 DOI: 10.1128/jvi.00469-15] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2015] [Accepted: 05/14/2015] [Indexed: 12/18/2022] Open
Abstract
UNLABELLED During HIV-1 assembly, the Gag viral proteins are targeted and assemble at the inner leaflet of the cell plasma membrane. This process could modulate the cortical actin cytoskeleton, located underneath the plasma membrane, since actin dynamics are able to promote localized membrane reorganization. In addition, activated small Rho GTPases are known for regulating actin dynamics and membrane remodeling. Therefore, the modulation of such Rho GTPase activity and of F-actin by the Gag protein during virus particle formation was considered. Here, we studied the implication of the main Rac1, Cdc42, and RhoA small GTPases, and some of their effectors, in this process. The effect of small interfering RNA (siRNA)-mediated Rho GTPases and silencing of their effectors on Gag localization, Gag membrane attachment, and virus-like particle production was analyzed by immunofluorescence coupled to confocal microscopy, membrane flotation assays, and immunoblot assays, respectively. In parallel, the effect of Gag expression on the Rac1 activation level was monitored by G-LISA, and the intracellular F-actin content in T cells was monitored by flow cytometry and fluorescence microscopy. Our results revealed the involvement of activated Rac1 and of the IRSp53-Wave2-Arp2/3 signaling pathway in HIV-1 Gag membrane localization and particle release in T cells as well as a role for actin branching and polymerization, and this was solely dependent on the Gag viral protein. In conclusion, our results highlight a new role for the Rac1-IRSp53-Wave2-Arp2/3 signaling pathway in the late steps of HIV-1 replication in CD4 T lymphocytes. IMPORTANCE During HIV-1 assembly, the Gag proteins are targeted and assembled at the inner leaflet of the host cell plasma membrane. Gag interacts with specific membrane phospholipids that can also modulate the regulation of cortical actin cytoskeleton dynamics. Actin dynamics can promote localized membrane reorganization and thus can be involved in facilitating Gag assembly and particle formation. Activated small Rho GTPases and effectors are regulators of actin dynamics and membrane remodeling. We thus studied the effects of the Rac1, Cdc42, and RhoA GTPases and their specific effectors on HIV-1 Gag membrane localization and viral particle release in T cells. Our results show that activated Rac1 and the IRSp53-Wave2-Arp2/3 signaling pathway are involved in Gag plasma membrane localization and viral particle production. This work uncovers a role for cortical actin through the activation of Rac1 and the IRSp53/Wave2 signaling pathway in HIV-1 particle formation in CD4 T lymphocytes.
Collapse
|
44
|
Dura B, Dougan SK, Barisa M, Hoehl MM, Lo CT, Ploegh HL, Voldman J. Profiling lymphocyte interactions at the single-cell level by microfluidic cell pairing. Nat Commun 2015; 6:5940. [DOI: 10.1038/ncomms6940] [Citation(s) in RCA: 123] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2014] [Accepted: 11/24/2014] [Indexed: 02/07/2023] Open
|
45
|
Chen W, Liu Y, Zhu S, Green CD, Wei G, Han JDJ. Improved nucleosome-positioning algorithm iNPS for accurate nucleosome positioning from sequencing data. Nat Commun 2014; 5:4909. [DOI: 10.1038/ncomms5909] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2013] [Accepted: 08/04/2014] [Indexed: 11/09/2022] Open
|
46
|
Rocha-Perugini V, González-Granado JM, Tejera E, López-Martín S, Yañez-Mó M, Sánchez-Madrid F. Tetraspanins CD9 and CD151 at the immune synapse support T-cell integrin signaling. Eur J Immunol 2014; 44:1967-75. [PMID: 24723389 DOI: 10.1002/eji.201344235] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2013] [Revised: 02/25/2014] [Accepted: 04/02/2014] [Indexed: 11/06/2022]
Abstract
Understanding how the immune response is activated and amplified requires detailed knowledge of the stages in the formation of the immunological synapse (IS) between T lymphocytes and antigen-presenting cells (APCs). We show that tetraspanins CD9 and CD151 congregate at the T-cell side of the IS. Silencing of CD9 or CD151 blunts the IL-2 secretion and expression of the activation marker CD69 by APC-conjugated T lymphocytes, but does not affect the accumulation of CD3 or actin to the IS, or the translocation of the microtubule-organizing center toward the T-B contact area. CD9 or CD151 silencing diminishes the relocalization of α4β1 integrin to the IS and reduces the accumulation of high-affinity β1 integrins at the cell-cell contact. These changes are accompanied by diminished phosphorylation of the integrin downstream targets FAK and ERK1/2. Our results suggest that CD9 and CD151 support integrin-mediated signaling at the IS.
Collapse
Affiliation(s)
- Vera Rocha-Perugini
- Servicio de Inmunología, Hospital de la Princesa, Instituto de Investigación Sanitaria La Princesa, Madrid, Spain; Vascular Biology and Inflammation Department, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | | | | | | | | | | |
Collapse
|
47
|
Tong Q, He S, Xie F, Mochizuki K, Liu Y, Mochizuki I, Meng L, Sun H, Zhang Y, Guo Y, Hexner E, Zhang Y. Ezh2 regulates transcriptional and posttranslational expression of T-bet and promotes Th1 cell responses mediating aplastic anemia in mice. THE JOURNAL OF IMMUNOLOGY 2014; 192:5012-22. [PMID: 24760151 DOI: 10.4049/jimmunol.1302943] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Acquired aplastic anemia (AA) is a potentially fatal bone marrow (BM) failure syndrome. IFN-γ-producing Th1 CD4(+) T cells mediate the immune destruction of hematopoietic cells, and they are central to the pathogenesis. However, the molecular events that control the development of BM-destructive Th1 cells remain largely unknown. Ezh2 is a chromatin-modifying enzyme that regulates multiple cellular processes primarily by silencing gene expression. We recently reported that Ezh2 is crucial for inflammatory T cell responses after allogeneic BM transplantation. To elucidate whether Ezh2 mediates pathogenic Th1 responses in AA and the mechanism of Ezh2 action in regulating Th1 cells, we studied the effects of Ezh2 inhibition in CD4(+) T cells using a mouse model of human AA. Conditionally deleting Ezh2 in mature T cells dramatically reduced the production of BM-destructive Th1 cells in vivo, decreased BM-infiltrating Th1 cells, and rescued mice from BM failure. Ezh2 inhibition resulted in significant decrease in the expression of Tbx21 and Stat4, which encode transcription factors T-bet and STAT4, respectively. Introduction of T-bet but not STAT4 into Ezh2-deficient T cells fully rescued their differentiation into Th1 cells mediating AA. Ezh2 bound to the Tbx21 promoter in Th1 cells and directly activated Tbx21 transcription. Unexpectedly, Ezh2 was also required to prevent proteasome-mediated degradation of T-bet protein in Th1 cells. Our results demonstrate that Ezh2 promotes the generation of BM-destructive Th1 cells through a mechanism of transcriptional and posttranscriptional regulation of T-bet. These results also highlight the therapeutic potential of Ezh2 inhibition in reducing AA and other autoimmune diseases.
Collapse
Affiliation(s)
- Qing Tong
- International Joint Cancer Institute, Second Military Medical University, Shanghai 200433, China; Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, 48109
| | - Shan He
- Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, 48109; Department of Microbiology and Immunology, Fels Institute for Cancer Research and Molecular Biology, Temple University, Philadelphia, PA 19140
| | - Fang Xie
- Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, 48109
| | - Kazuhiro Mochizuki
- Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, 48109
| | - Yongnian Liu
- Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, 48109; Department of Microbiology and Immunology, Fels Institute for Cancer Research and Molecular Biology, Temple University, Philadelphia, PA 19140
| | - Izumi Mochizuki
- Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, 48109
| | - Lijun Meng
- Department of Microbiology and Immunology, Fels Institute for Cancer Research and Molecular Biology, Temple University, Philadelphia, PA 19140; Institute of Health Sciences, Shanghai Institutes for Biological Sciences Chinese Academy of Sciences, Shanghai 200433, China; and
| | - Hongxing Sun
- Institute of Health Sciences, Shanghai Institutes for Biological Sciences Chinese Academy of Sciences, Shanghai 200433, China; and
| | - Yanyun Zhang
- Institute of Health Sciences, Shanghai Institutes for Biological Sciences Chinese Academy of Sciences, Shanghai 200433, China; and
| | - Yajun Guo
- International Joint Cancer Institute, Second Military Medical University, Shanghai 200433, China
| | - Elizabeth Hexner
- Department of Medicine and Abramson Cancer Center, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104
| | - Yi Zhang
- Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, 48109; Department of Microbiology and Immunology, Fels Institute for Cancer Research and Molecular Biology, Temple University, Philadelphia, PA 19140;
| |
Collapse
|
48
|
Jenkins MR, Stinchcombe JC, Au-Yeung BB, Asano Y, Ritter AT, Weiss A, Griffiths GM. Distinct structural and catalytic roles for Zap70 in formation of the immunological synapse in CTL. eLife 2014; 3:e01310. [PMID: 24596147 PMCID: PMC3936284 DOI: 10.7554/elife.01310] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2013] [Accepted: 01/22/2014] [Indexed: 11/13/2022] Open
Abstract
T cell receptor (TCR) activation leads to a dramatic reorganisation of both membranes and receptors as the immunological synapse forms. Using a genetic model to rapidly inhibit Zap70 catalytic activity we examined synapse formation between cytotoxic T lymphocytes and their targets. In the absence of Zap70 catalytic activity Vav-1 activation occurs and synapse formation is arrested at a stage with actin and integrin rich interdigitations forming the interface between the two cells. The membranes at the synapse are unable to flatten to provide extended contact, and Lck does not cluster to form the central supramolecular activation cluster (cSMAC). Centrosome polarisation is initiated but aborts before reaching the synapse and the granules do not polarise. Our findings reveal distinct roles for Zap70 as a structural protein regulating integrin-mediated control of actin vs its catalytic activity that regulates TCR-mediated control of actin and membrane remodelling during formation of the immunological synapse. DOI: http://dx.doi.org/10.7554/eLife.01310.001.
Collapse
Affiliation(s)
- Misty R Jenkins
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge, United Kingdom
| | - Jane C Stinchcombe
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge, United Kingdom
| | - Byron B Au-Yeung
- Department of Medicine, University of California, San Francisco, San Francisco, United States
- Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, United States
- Howard Hughes Medical Institue, University of California, San Francisco, San Francisco, United States
| | - Yukako Asano
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge, United Kingdom
| | - Alex T Ritter
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge, United Kingdom
- Cell Biology and Metabolism Branch, National Institutes of Health, Bethesda, United States
| | - Arthur Weiss
- Department of Medicine, University of California, San Francisco, San Francisco, United States
- Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, United States
- Howard Hughes Medical Institue, University of California, San Francisco, San Francisco, United States
| | - Gillian M Griffiths
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
49
|
Genome-wide association study combining pathway analysis for typical sporadic amyotrophic lateral sclerosis in Chinese Han populations. Neurobiol Aging 2014; 35:1778.e9-1778.e23. [PMID: 24529757 DOI: 10.1016/j.neurobiolaging.2014.01.014] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2013] [Revised: 12/20/2013] [Accepted: 01/12/2014] [Indexed: 01/06/2023]
Abstract
Sporadic amyotrophic lateral sclerosis (sALS) is a severe neurodegenerative disease that causes progressive motor neuron death. Although the etiology of sALS remains unknown, genetic variants are thought to predispose individuals to the disease. Several recent genome-wide association studies have identified a number of loci that increase sALS susceptibility, but these only explain a small proportion of the disease. To extend the current genetic evidence and to identify novel candidates of sALS, we performed a pooling genome-wide association study by 859,311 autosomal single-nucleotide polymorphisms of IlluminaHumanOmniZhongHua-8 combining pathway analysis in 250 typical sALS cases precluding age, clinical course, and phenotype interference and 250 control subjects from Chinese Han populations (CHP). The results revealed that 8 novel loci of 1p34.3, 3p21.1, 3p22.2, 10p15.2, 22q12.1, 3q13.11, 11q25, 12q24.33, and 5 previously reported loci of CNTN4 (kgp11325216), ATXN1 (kgp8327591), C9orf72 (kgp6016770), ITPR2 (kgp3041552), and SOD1 (kgp10760302) were associated with sALS from CHP. Furthermore, the pathway analysis based on the Gene Set Analysis Toolkit V2 showed that 10 top pathways were strongly associated with sALS from CHP, and among them, the 7 most potentially candidate pathways were phosphatidylinositol signaling system, Wnt signaling pathway, axon guidance, MAPK signaling pathway, neurotrophin signaling pathway, arachidonic acid metabolism, and T-cell receptor signaling pathway, a total of 39 significantly associate genes in 7 candidate pathways was suggested to involve in the pathogenesis of sALS from CHP. In conclusion, our results revealed several new loci and pathways related to sALS from CHP and extend the association evidence for partial loci, genes, and pathways, which were previously identified in other populations. Thus, our data provided new clues for exploring the pathogenesis of sALS.
Collapse
|
50
|
Abstract
At least 468 individual genes have been manipulated by molecular methods to study their effects on the initiation, promotion, and progression of atherosclerosis. Most clinicians and many investigators, even in related disciplines, find many of these genes and the related pathways entirely foreign. Medical schools generally do not attempt to incorporate the relevant molecular biology into their curriculum. A number of key signaling pathways are highly relevant to atherogenesis and are presented to provide a context for the gene manipulations summarized herein. The pathways include the following: the insulin receptor (and other receptor tyrosine kinases); Ras and MAPK activation; TNF-α and related family members leading to activation of NF-κB; effects of reactive oxygen species (ROS) on signaling; endothelial adaptations to flow including G protein-coupled receptor (GPCR) and integrin-related signaling; activation of endothelial and other cells by modified lipoproteins; purinergic signaling; control of leukocyte adhesion to endothelium, migration, and further activation; foam cell formation; and macrophage and vascular smooth muscle cell signaling related to proliferation, efferocytosis, and apoptosis. This review is intended primarily as an introduction to these key signaling pathways. They have become the focus of modern atherosclerosis research and will undoubtedly provide a rich resource for future innovation toward intervention and prevention of the number one cause of death in the modern world.
Collapse
Affiliation(s)
- Paul N Hopkins
- Cardiovascular Genetics, Department of Internal Medicine, University of Utah, Salt Lake City, Utah, USA.
| |
Collapse
|