1
|
Run L, Tian Z, Xu L, Du J, Li N, Wang Q, Sun H. Knockdown of IL4I1 Improved High Glucose-evoked Insulin Resistance in HepG2 Cells by Alleviating Inflammation and Lipotoxicity Through AHR Activation. Appl Biochem Biotechnol 2023; 195:6694-6707. [PMID: 36913096 DOI: 10.1007/s12010-023-04399-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/17/2023] [Indexed: 03/14/2023]
Abstract
Insulin resistance (IR) is one of the leading causes of Type 2 diabetes mellitus (T2DM). Inflammation, as a result of the disordered immune response, plays important roles in IR and T2DM. Interleukin-4-induced gene 1 (IL4I1) has been shown to regulate immune response and be involved in inflammation progress. However, there was little known about its roles in T2DM. Here, high glucose (HG)-treated HepG2 cells were used for T2DM investigation in vitro. Our results indicated that the expression of IL4I1 was up-regulated in peripheral blood samples of T2DM-patients and HG-induced HepG2 cells. The silencing of IL4I1 alleviated the HG-evoked IR through elevating the expressions of p-IRS1, p-AKT and GLUT4, and enhancing glucose consumption. Furthermore, IL4I1 knockdown inhibited inflammatory response by reducing the levels of inflammatory mediators, and suppressed the accumulation of lipid metabolites triglyceride (TG) and palmitate (PA) in HG-induced cells. Notably, IL4I1 expression was positively correlated with aryl hydrocarbon receptor (AHR) in peripheral blood samples of T2DM-patients. The silencing of IL4I1 inhibited the AHR signaling by reducing the HG-induced expressions of AHR and CYP1A1. Subsequent experiments confirmed that 2,3,7,8-Tetrachlorodibenzo-p-dioxin (TCDD), an agonist of AHR, reversed the suppressive effects of IL4I1 knockdown on HG-caused inflammation, lipid metabolism and IR in cells. In conclusion, we found that the silencing of IL4I1 attenuated inflammation, lipid metabolism and IR in HG-induced cells via inhibiting AHR signaling, suggesting that IL4I1 might be a potential therapy target for T2DM.
Collapse
Affiliation(s)
- Lin Run
- Department of Pathophysiology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, 710061, Xi'an, Shaanxi, China, NO. 76, Yanta West Road, Yanta District
- Department of Endocrinology, Xi'an Central Hospital Affiliated to Medical College of Xi'an Jiaotong University, 710003, Xi'an, Shaanxi, China
| | - Zhufang Tian
- Department of Endocrinology, Xi'an Central Hospital Affiliated to Medical College of Xi'an Jiaotong University, 710003, Xi'an, Shaanxi, China
| | - Lin Xu
- Department of Endocrinology, The Affiliated Guangren Hospital, Xi'an Jiaotong University College of Medicine, 710004, Xi'an, Shaanxi, China
| | - Junhui Du
- Department of Medicine Interdisciplinary Research, Xi'an Ninth Hospital Affiliated to Medical College of Xi'an Jiaotong University, 710054, Xi'an, Shaanxi, China
| | - Nan Li
- Clinical Laboratory, Xi'an Central Hospital Affiliated to Medical College of Xi'an Jiaotong University, 710003, Xi'an, Shaanxi, China
| | - Qi Wang
- Department of Nuclear Medicine, Xi'an Central Hospital Affiliated to Medical College of Xi'an Jiaotong University, 710003, Xi'an, Shaanxi, China
| | - Hongzhi Sun
- Department of Pathophysiology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, 710061, Xi'an, Shaanxi, China, NO. 76, Yanta West Road, Yanta District.
| |
Collapse
|
2
|
Xu T, Liu J, Xia Y, Wang Z, Li X, Gao Q. Integrated analysis reveals the participation of IL4I1, ITGB7, and FUT7 in reshaping the TNBC immune microenvironment by targeting glycolysis. Ann Med 2021; 53:916-928. [PMID: 34134578 PMCID: PMC8604452 DOI: 10.1080/07853890.2021.1937694] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Accepted: 05/26/2021] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND The overall response rate of immunotherapy in triple-negative breast cancer (TNBC) remains unsatisfactory. Accumulating evidence indicated that glucose metabolic reprogramming could modulate immunotherapy efficacy. However, transcriptomic evidence remains insufficient. METHODS Genes' relationship with glucose metabolism and TNBC-specific immune was demonstrated by weighted gene co-expression network analysis (WGCNA). The glucose metabolic capability was estimated by standardised uptake value (SUV), an indicator of glucose uptake in 18 F-fluorodeoxyglucose positron emission tomography (FDG-PET), and a reflection of cancer metabolic behaviour. PD-(L)1 expression was used to reflect the efficacy of immunotherapy. Additionally, immune infiltration, survival, and gene coexpression profiles were provided. RESULTS Comprehensive analysis revealing that IL4I1, ITGB7, and FUT7 hold the potential to reinforce immunotherapy by reshaping glucose metabolism in TNBC. These results were verified by functional enrichment analysis, which demonstrated their relationships with immune-related signalling pathways and extracellular microenvironment reprogramming. Their expressions have potent positive correlations with Treg and Macrophage cell infiltration and exhausted T cell markers. Meanwhile, their overexpression also lead to poor prognosis. CONCLUSION IL4I1, ITGB7, and FUT7 may be the hub genes that link glucose metabolism, and cancer-specific immunity. They may be potential targets for enhancing ICB treatment by reprogramming the tumour microenvironment and remodelling tumour metabolism.
Collapse
Affiliation(s)
- Tao Xu
- Key Laboratory of the Ministry of Education, Cancer Biology Research Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Department of Thyroid and Breast Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jiahao Liu
- Key Laboratory of the Ministry of Education, Cancer Biology Research Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yu Xia
- Key Laboratory of the Ministry of Education, Cancer Biology Research Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhi Wang
- Key Laboratory of the Ministry of Education, Cancer Biology Research Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xingrui Li
- Department of Thyroid and Breast Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qinglei Gao
- Key Laboratory of the Ministry of Education, Cancer Biology Research Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
3
|
Bonet LFS, Loureiro JP, Pereira GRC, Da Silva ANR, De Mesquita JF. Molecular dynamics and protein frustration analysis of human fused in Sarcoma protein variants in Amyotrophic Lateral Sclerosis type 6: An In Silico approach. PLoS One 2021; 16:e0258061. [PMID: 34587215 PMCID: PMC8480726 DOI: 10.1371/journal.pone.0258061] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Accepted: 09/16/2021] [Indexed: 11/18/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is the most frequent adult-onset motor neuron disorder. The disease is characterized by degeneration of upper and lower motor neurons, leading to death usually within five years after the onset of symptoms. While most cases are sporadic, 5%-10% of cases can be associated with familial inheritance, including ALS type 6, which is associated with mutations in the Fused in Sarcoma (FUS) gene. This work aimed to evaluate how the most frequent ALS-related mutations in FUS, R521C, R521H, and P525L affect the protein structure and function. We used prediction algorithms to analyze the effects of the non-synonymous single nucleotide polymorphisms and performed evolutionary conservation analysis, protein frustration analysis, and molecular dynamics simulations. Most of the prediction algorithms classified the three mutations as deleterious. All three mutations were predicted to reduce protein stability, especially the mutation R521C, which was also predicted to increase chaperone binding tendency. The protein frustration analysis showed an increase in frustration in the interactions involving the mutated residue 521C. Evolutionary conservation analysis showed that residues 521 and 525 of human FUS are highly conserved sites. The molecular dynamics results indicate that protein stability could be compromised in all three mutations. They also affected the exposed surface area and protein compactness. The analyzed mutations also displayed high flexibility in most residues in all variants, most notably in the interaction site with the nuclear import protein of FUS.
Collapse
Affiliation(s)
- L. F. S. Bonet
- Department of Genetics and Molecular Biology, Laboratory of Bioinformatics and Computational Biology, Federal University of the State of Rio de Janeiro, Rio de Janeiro, Brazil
| | - J. P. Loureiro
- Department of Genetics and Molecular Biology, Laboratory of Bioinformatics and Computational Biology, Federal University of the State of Rio de Janeiro, Rio de Janeiro, Brazil
| | - G. R. C. Pereira
- Department of Genetics and Molecular Biology, Laboratory of Bioinformatics and Computational Biology, Federal University of the State of Rio de Janeiro, Rio de Janeiro, Brazil
| | - A. N. R. Da Silva
- Department of Genetics and Molecular Biology, Laboratory of Bioinformatics and Computational Biology, Federal University of the State of Rio de Janeiro, Rio de Janeiro, Brazil
| | - J. F. De Mesquita
- Department of Genetics and Molecular Biology, Laboratory of Bioinformatics and Computational Biology, Federal University of the State of Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
4
|
Castellano F, Molinier-Frenkel V. Control of T-Cell Activation and Signaling by Amino-Acid Catabolizing Enzymes. Front Cell Dev Biol 2020; 8:613416. [PMID: 33392202 PMCID: PMC7773816 DOI: 10.3389/fcell.2020.613416] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Accepted: 11/30/2020] [Indexed: 12/12/2022] Open
Abstract
Amino acids are essential for protein synthesis, epigenetic modification through the methylation of histones, and the maintenance of a controlled balance of oxidoreduction via the production of glutathione and are precursors of certain neurotransmitters. T lymphocytes are particularly sensitive to fluctuations in amino acid levels. During evolution, the production of amino-acid catabolizing enzymes by mainly antigen-presenting cells has become a physiological mechanism to control T-cell activation and polarization. The action of these enzymes interferes with TCR and co-stimulation signaling, allowing tuning of the T-cell response. These capacities can be altered in certain pathological conditions, with relevant consequences for the development of disease.
Collapse
Affiliation(s)
- Flavia Castellano
- Univ Paris Est Creteil, INSERM, IMRB, Creteil, France.,AP-HP, Hopital Henri Mondor, Departement Immunologie-Hématologie, Creteil, France
| | - Valérie Molinier-Frenkel
- Univ Paris Est Creteil, INSERM, IMRB, Creteil, France.,AP-HP, Hopital Henri Mondor, Departement Immunologie-Hématologie, Creteil, France
| |
Collapse
|
5
|
Davidov V, Jensen G, Mai S, Chen SH, Pan PY. Analyzing One Cell at a TIME: Analysis of Myeloid Cell Contributions in the Tumor Immune Microenvironment. Front Immunol 2020; 11:1842. [PMID: 32983100 PMCID: PMC7492293 DOI: 10.3389/fimmu.2020.01842] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Accepted: 07/09/2020] [Indexed: 12/30/2022] Open
Abstract
Tumor-mediated regulation of the host immune system involves an intricate signaling network that results in the tumor's inherent survival benefit. Myeloid cells are central in orchestrating the mechanisms by which tumors escape immune detection and continue their proliferative programming. Myeloid cell activation has historically been classified using a dichotomous system of classical (M1-like) and alternative (M2-like) states, defining general pro- and anti-inflammatory functions, respectively. Explosions in bioinformatics analyses have rapidly expanded the definitions of myeloid cell pro- and anti-inflammatory states with different combinations of tissue- and disease-specific phenotypic and functional markers. These new definitions have allowed researchers to target specific subsets of disease-propagating myeloid cells in order to modify or arrest the natural progression of the associated disease, especially in the context of tumor-immune interactions. Here, we discuss the myeloid cell contribution to solid tumor initiation and maintenance, and strategies to reprogram their phenotypic and functional fate, thereby disabling the network that benefits tumor survival.
Collapse
Affiliation(s)
- Vitaliy Davidov
- Texas A&M College of Medicine, Bryan, TX, United States.,Center for Immunotherapy Research, Cancer Center of Excellence, Houston Methodist Research Institute, Houston, TX, United States
| | - Garrett Jensen
- Texas A&M College of Medicine, Bryan, TX, United States.,Center for Immunotherapy Research, Cancer Center of Excellence, Houston Methodist Research Institute, Houston, TX, United States
| | - Sunny Mai
- Center for Immunotherapy Research, Cancer Center of Excellence, Houston Methodist Research Institute, Houston, TX, United States
| | - Shu-Hsia Chen
- Texas A&M College of Medicine, Bryan, TX, United States.,Center for Immunotherapy Research, Cancer Center of Excellence, Houston Methodist Research Institute, Houston, TX, United States
| | - Ping-Ying Pan
- Texas A&M College of Medicine, Bryan, TX, United States.,Center for Immunotherapy Research, Cancer Center of Excellence, Houston Methodist Research Institute, Houston, TX, United States
| |
Collapse
|
6
|
Molinier-Frenkel V, Prévost-Blondel A, Castellano F. The IL4I1 Enzyme: A New Player in the Immunosuppressive Tumor Microenvironment. Cells 2019; 8:E757. [PMID: 31330829 PMCID: PMC6678094 DOI: 10.3390/cells8070757] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Revised: 07/18/2019] [Accepted: 07/19/2019] [Indexed: 12/14/2022] Open
Abstract
The high metabolic needs of T lymphocytes in response to activation make them particularly vulnerable to modifications of their biochemical milieu. Immunosuppressive enzymes produced in the tumor microenvironment modify nutrient availability by catabolizing essential or semi-essential amino acids and producing toxic catabolites, thus participating in the local sabotage of the antitumor immune response. L-amino-acid oxidases are FAD-bound enzymes found throughout evolution, from bacteria to mammals, and are often endowed with anti-infectious properties. IL4I1 is a secreted L-phenylalanine oxidase mainly produced by inflammatory antigen-presenting cells-in particular, macrophages present in T helper type 1 granulomas and in various types of tumors. In the last decade, it has been shown that IL4I1 is involved in the fine control of B- and T-cell adaptive immune responses. Preclinical models have revealed its role in cancer immune evasion. Recent clinical data highlight IL4I1 as a new potential prognostic marker in human melanoma. As a secreted enzyme, IL4I1 may represent an easily targetable molecule for cancer immunotherapy.
Collapse
Affiliation(s)
- Valérie Molinier-Frenkel
- INSERM, U955, Team 09, 94010 Créteil, France.
- Faculty of Medicine, University Paris Est, 94010 Créteil, France.
- AP-HP, H. Mondor - A. Chenevier Hospital, Biological Immunology Service, 94010 Créteil, France.
| | - Armelle Prévost-Blondel
- INSERM, U1016, Institute Cochin, 75014 Paris, France
- CNRS, UMR8104, 75014 Paris, France
- University Paris Descartes, Sorbonne Paris Cité, 75014 Paris, France
| | - Flavia Castellano
- INSERM, U955, Team 09, 94010 Créteil, France.
- Faculty of Medicine, University Paris Est, 94010 Créteil, France.
| |
Collapse
|
7
|
Pentimalli F, Grelli S, Di Daniele N, Melino G, Amelio I. Cell death pathologies: targeting death pathways and the immune system for cancer therapy. Genes Immun 2018; 20:539-554. [PMID: 30563970 PMCID: PMC6451632 DOI: 10.1038/s41435-018-0052-x] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2018] [Revised: 10/25/2018] [Accepted: 11/13/2018] [Indexed: 12/15/2022]
Abstract
Alterations in the molecular mechanisms of cell death are a common feature of cancer. These alterations enable malignant cells to survive intrinsic death signalling leading to accumulation of genetic aberrations and helping them to cope with adverse conditions. Regulated cell death has historically been exclusively associated with classical apoptosis; however, increasing evidence indicates that several alternative mechanisms orchestrate multiple death pathways, such as ferroptosis, entosis, necroptosis and immunogenic cell death, each with distinct underlying molecular mechanisms. Although pharmacological targeting of cell death pathways has been the subject of intensive efforts in recent decades with a dominant focus on targeting apoptosis, the identification of these novel death pathways has opened additional venues for intervention in cancer cells and the immune system. In this mini-review, we cover some recent progress on major recently emerged cell death modalities, emphasizing their potential clinical and therapeutic implications. We also discuss the interplay between cell death and immune response, highlighting the potential of the combination of traditional anticancer therapy and immunocheckpoint blockade. While attempting to stimulate discussion and draw attention to the possible clinical impact of these more recently emerged cell death modalities, we also cover the major progress achieved in translating strategies for manipulation of apoptotic pathways into the clinic, focusing on the attempts to target the anti-apoptotic protein BCL-2 and the tumour suppressor p53.
Collapse
Affiliation(s)
- Francesca Pentimalli
- Centro Ricerche Oncologiche Mercogliano (CROM), Istituto Nazionale Tumori - IRCCS -Fondazione G. Pascale, Naples, Italy
| | - Sandro Grelli
- Biochemistry Laboratory, Istituto Dermopatico Immacolata (IDI-IRCCS), 00100, Rome, Italy
| | - Nicola Di Daniele
- Biochemistry Laboratory, Istituto Dermopatico Immacolata (IDI-IRCCS), 00100, Rome, Italy
| | - Gerry Melino
- Biochemistry Laboratory, Istituto Dermopatico Immacolata (IDI-IRCCS), 00100, Rome, Italy.,Medical Research Council, Toxicology Unit, University of Cambridge, Hodgkin Building, Lancaster Road, P.O. Box 138, Leicester, LE1 9HN, UK
| | - Ivano Amelio
- Medical Research Council, Toxicology Unit, University of Cambridge, Hodgkin Building, Lancaster Road, P.O. Box 138, Leicester, LE1 9HN, UK.
| |
Collapse
|
8
|
Shigeno Y, Zhang H, Banno T, Usuda K, Nochi T, Inoue R, Watanabe G, Jin W, Benno Y, Nagaoka K. Gut microbiota development in mice is affected by hydrogen peroxide produced from amino acid metabolism during lactation. FASEB J 2018; 33:3343-3352. [PMID: 30433825 DOI: 10.1096/fj.201801462r] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The development of gut microbiota during infancy is an important event that affects the health status of the host; however, the mechanism governing it is not fully understood. l-Amino acid oxidase 1 (LAO1) is a flavoprotein that catalyzes the oxidative deamination of particular l-amino acids and converts them into keto acids, ammonia, and H2O2. Our previous study showed that LAO1 is present in mouse milk and exerts protection against bacteria by its production of H2O2. The data led us to consider whether LAO1, H2O2, or both could impact infant gut microbiota development via mother's milk consumption in mice. Different gut microbiota profiles were observed in the wild-type (WT) and LAO1-knockout mouse pups. The WT pups' microbiota was relatively simple and composed of only a few dominant bacteria, such as Lactobacillus, whereas the lactating knockout pups had high microbiota diversity. Cross-fostering experiments indicated that WT milk (containing LAO1) has the ability to suppress the diversity of microbiota in pups. We observed that the stomach content of pups fed WT milk had LAO1 proteins and the ability to produce H2O2. Moreover, culture experiments showed that Lactobacillus was abundant in the feces of pups fed WT milk and that Lactobacillus was more resistant to H2O2 than Bifidobacterium and Escherichia. Human breast milk produces very little H2O2, which could be the reason for Lactobacillus not being dominant in the feces of breast-fed human infants. In mouse mother's milk, H2O2 is generated from the process of free amino acid metabolism, and H2O2 may be a key player in regulating the initial acquisition and development of gut microbiota, especially growth of Lactobacillus, during infancy.-Shigeno, Y., Zhang, H., Banno, T., Usuda, K., Nochi, T., Inoue, R., Watanabe, G., Jin, W., Benno, Y., Nagaoka, K. Gut microbiota development in mice is affected by hydrogen peroxide produced from amino acid metabolism during lactation.
Collapse
Affiliation(s)
- Yuko Shigeno
- Laboratory of Veterinary Physiology, Department of Veterinary Medicine, Tokyo University of Agriculture and Technology, Tokyo, Japan.,Benno Laboratory, RIKEN Innovation Center, Wako, Japan
| | - Haolin Zhang
- Laboratory of Veterinary Physiology, Department of Veterinary Medicine, Tokyo University of Agriculture and Technology, Tokyo, Japan
| | - Taihei Banno
- Benno Laboratory, RIKEN Innovation Center, Wako, Japan
| | - Kento Usuda
- Laboratory of Veterinary Physiology, Department of Veterinary Medicine, Tokyo University of Agriculture and Technology, Tokyo, Japan
| | - Tomonori Nochi
- Laboratory of Mucosal Immunology, Graduate School of Agricultural Science, Tohoku University, Sendai, Japan
| | - Ryo Inoue
- Laboratory of Animal Science, Department of Agricultural and Life Sciences, Kyoto Prefectural University, Kyoto, Japan; and
| | - Gen Watanabe
- Laboratory of Veterinary Physiology, Department of Veterinary Medicine, Tokyo University of Agriculture and Technology, Tokyo, Japan
| | - Wanzhu Jin
- Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Yoshimi Benno
- Benno Laboratory, RIKEN Innovation Center, Wako, Japan
| | - Kentaro Nagaoka
- Laboratory of Veterinary Physiology, Department of Veterinary Medicine, Tokyo University of Agriculture and Technology, Tokyo, Japan
| |
Collapse
|
9
|
Bod L, Douguet L, Auffray C, Lengagne R, Bekkat F, Rondeau E, Molinier-Frenkel V, Castellano F, Richard Y, Prévost-Blondel A. IL-4-Induced Gene 1: A Negative Immune Checkpoint Controlling B Cell Differentiation and Activation. THE JOURNAL OF IMMUNOLOGY 2017; 200:1027-1038. [PMID: 29288206 DOI: 10.4049/jimmunol.1601609] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/15/2016] [Accepted: 11/26/2017] [Indexed: 11/19/2022]
Abstract
Emerging data highlight the crucial role of enzymes involved in amino acid metabolism in immune cell biology. IL-4-induced gene-1 (IL4I1), a secreted l-phenylalanine oxidase expressed by APCs, has been detected in B cells, yet its immunoregulatory role has only been explored on T cells. In this study, we show that IL4I1 regulates multiple steps in B cell physiology. Indeed, IL4I1 knockout mice exhibit an accelerated B cell egress from the bone marrow, resulting in the accumulation of peripheral follicular B cells. They also present a higher serum level of natural Igs and self-reactive Abs. We also demonstrate that IL4I1 produced by B cells themselves controls the germinal center reaction, plasma cell differentiation, and specific Ab production in response to T dependent Ags, SRBC, and NP-KLH. In vitro, IL4I1-deficient B cells proliferate more efficiently than their wild-type counterparts in response to BCR cross-linking. Moreover, the absence of IL4I1 increases activation of the Syk-Akt-S6kinase signaling pathway and calcium mobilization, and inhibits SHP-1 activity upon BCR engagement, thus supporting that IL4I1 negatively controls BCR-dependent activation. Overall, our study reveals a new perspective on IL4I1 as a key regulator of B cell biology.
Collapse
Affiliation(s)
- Lloyd Bod
- INSERM, U1016, Institut Cochin, 75014 Paris, France.,CNRS, UMR8104, 75014 Paris, France.,Université Paris Descartes, Sorbonne Paris Cité, 75006 Paris, France
| | - Laetitia Douguet
- INSERM, U1016, Institut Cochin, 75014 Paris, France.,CNRS, UMR8104, 75014 Paris, France.,Université Paris Descartes, Sorbonne Paris Cité, 75006 Paris, France
| | - Cédric Auffray
- INSERM, U1016, Institut Cochin, 75014 Paris, France.,CNRS, UMR8104, 75014 Paris, France.,Université Paris Descartes, Sorbonne Paris Cité, 75006 Paris, France
| | - Renée Lengagne
- INSERM, U1016, Institut Cochin, 75014 Paris, France.,CNRS, UMR8104, 75014 Paris, France.,Université Paris Descartes, Sorbonne Paris Cité, 75006 Paris, France
| | - Fériel Bekkat
- INSERM, U1016, Institut Cochin, 75014 Paris, France.,CNRS, UMR8104, 75014 Paris, France.,Université Paris Descartes, Sorbonne Paris Cité, 75006 Paris, France
| | - Elena Rondeau
- INSERM, U1016, Institut Cochin, 75014 Paris, France.,CNRS, UMR8104, 75014 Paris, France.,Université Paris Descartes, Sorbonne Paris Cité, 75006 Paris, France
| | - Valérie Molinier-Frenkel
- INSERM, U955, Equipe 09, 94000 Créteil, France.,Faculté de Médecine, Université Paris Est, 94000 Créteil, France.,Assistance Publique - Hôpitaux de Paris, Hôpital Henri Mondor - Albert Chenevier, Service d'Immunologie Biologique, 94000 Créteil, France; and
| | - Flavia Castellano
- INSERM, U955, Equipe 09, 94000 Créteil, France.,Faculté de Médecine, Université Paris Est, 94000 Créteil, France.,Assistance Publique - Hôpitaux de Paris, Hôpital Henri Mondor - Albert Chenevier, Plateforme de Ressources Biologiques, 94000 Créteil, France
| | - Yolande Richard
- INSERM, U1016, Institut Cochin, 75014 Paris, France; .,CNRS, UMR8104, 75014 Paris, France.,Université Paris Descartes, Sorbonne Paris Cité, 75006 Paris, France
| | - Armelle Prévost-Blondel
- INSERM, U1016, Institut Cochin, 75014 Paris, France; .,CNRS, UMR8104, 75014 Paris, France.,Université Paris Descartes, Sorbonne Paris Cité, 75006 Paris, France
| |
Collapse
|
10
|
An Overview of l-Amino Acid Oxidase Functions from Bacteria to Mammals: Focus on the Immunoregulatory Phenylalanine Oxidase IL4I1. Molecules 2017; 22:molecules22122151. [PMID: 29206151 PMCID: PMC6149928 DOI: 10.3390/molecules22122151] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2017] [Revised: 11/23/2017] [Accepted: 11/29/2017] [Indexed: 01/04/2023] Open
Abstract
l-amino acid oxidases are flavin adenine dinucleotide-dependent enzymes present in all major kingdom of life, from bacteria to mammals. They participate in defense mechanisms by limiting the growth of most bacteria and parasites. A few mammalian LAAOs have been described, of which the enzyme “interleukin-4 induced gene 1” (IL4I1) is the best characterized. IL4I1 mainly oxidizes l-phenylalanine. It is a secreted enzyme physiologically produced by antigen presenting cells of the myeloid and B cell lineages and T helper type (Th) 17 cells. Important roles of IL4I1 in the fine control of the adaptive immune response in mice and humans have emerged during the last few years. Indeed, IL4I1 inhibits T cell proliferation and cytokine production and facilitates naïve CD4+ T-cell differentiation into regulatory T cells in vitro by limiting the capacity of T lymphocytes to respond to clonal receptor stimulation. It may also play a role in controlling the germinal center reaction for antibody production and limiting Th1 and Th17 responses. IL4I1 is expressed in tumor-associated macrophages of most human cancers and in some tumor cell types. Such expression, associated with its capacity to facilitate tumor growth by inhibiting the anti-tumor T-cell response, makes IL4I1 a new potential druggable target in the field of immunomodulation in cancer.
Collapse
|
11
|
Aubatin A, Sako N, Decrouy X, Donnadieu E, Molinier-Frenkel V, Castellano F. IL4-induced gene 1 is secreted at the immune synapse and modulates TCR activation independently of its enzymatic activity. Eur J Immunol 2017; 48:106-119. [PMID: 28891065 DOI: 10.1002/eji.201646769] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2016] [Revised: 08/21/2017] [Accepted: 09/06/2017] [Indexed: 01/11/2023]
Abstract
Amino-acid catabolizing enzymes produced by mononuclear phagocytes play a central role in regulating the immune response. The mammalian phenylalanine-catabolizing enzyme IL4-induced gene 1 (IL4I1) inhibits effector T lymphocyte proliferation and facilitates regulatory T-cell development. IL4I1 expression by macrophages of various human tumors may affect patient prognosis as it facilitates tumor escape from the T-cell response in murine models. Its enzymatic activity appears to participate in its effects, but some actions of IL4I1 remain unclear. Here, we show that the presence of IL4I1 during T-cell activation decreases early signaling events downstream of TCR stimulation, resulting in global T-cell inhibition which is more pronounced when there is CD28 costimulation. Surprisingly, the enzymatic activity of IL4I1 is not involved. Focal secretion of IL4I1 into the immune synaptic cleft and its binding to CD3+ lymphocytes could be important in IL4I1 immunosuppressive mechanism of action.
Collapse
Affiliation(s)
- Aude Aubatin
- INSERM, U955, Equipe 09, Créteil, France.,Faculté de Médecine, Université Paris Est, Créteil, France
| | - Nouhoum Sako
- INSERM, U955, Equipe 09, Créteil, France.,Faculté de Médecine, Université Paris Est, Créteil, France
| | - Xavier Decrouy
- INSERM, U955, Plateforme d'imagerie, Créteil, France.,Faculté de Médecine, Université Paris Est, Créteil, France
| | - Emmanuel Donnadieu
- INSERM, U1016, Institut Cochin, Paris, France.,CNRS, UMR8104, Paris, France.,Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Valérie Molinier-Frenkel
- INSERM, U955, Equipe 09, Créteil, France.,Faculté de Médecine, Université Paris Est, Créteil, France.,AP-HP, Hôpital H. Mondor - A. Chenevier, Service d'Immunologie Biologique, Créteil, France
| | - Flavia Castellano
- INSERM, U955, Equipe 09, Créteil, France.,Faculté de Médecine, Université Paris Est, Créteil, France.,AP-HP, Hôpital H. Mondor - A. Chenevier, Plateforme de Ressources Biologiques, Créteil, France
| |
Collapse
|
12
|
Seddigh P, Bracht T, Molinier-Frenkel V, Castellano F, Kniemeyer O, Schuster M, Weski J, Hasenberg A, Kraus A, Poschet G, Hager T, Theegarten D, Opitz CA, Brakhage AA, Sitek B, Hasenberg M, Gunzer M. Quantitative Analysis of Proteome Modulations in Alveolar Epithelial Type II Cells in Response to Pulmonary Aspergillus fumigatus Infection. Mol Cell Proteomics 2017; 16:2184-2198. [PMID: 28951444 DOI: 10.1074/mcp.ra117.000072] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2017] [Indexed: 12/14/2022] Open
Abstract
The ubiquitous mold Aspergillus fumigatus threatens immunosuppressed patients as inducer of lethal invasive aspergillosis. A. fumigatus conidia are airborne and reach the alveoli, where they encounter alveolar epithelial cells (AEC). Previous studies reported the importance of the surfactant-producing AEC II during A. fumigatus infection via in vitro experiments using cell lines. We established a negative isolation protocol yielding untouched primary murine AEC II with a purity >90%, allowing ex vivo analyses of the cells, which encountered the mold in vivo By label-free proteome analysis of AEC II isolated from mice 24h after A. fumigatus or mock infection we quantified 2256 proteins and found 154 proteins to be significantly differentially abundant between both groups (ANOVA p value ≤ 0.01, ratio of means ≥1.5 or ≤0.67, quantified with ≥2 peptides). Most of these proteins were higher abundant in the infected condition and reflected a comprehensive activation of AEC II on interaction with A. fumigatus This was especially represented by proteins related to oxidative phosphorylation, hence energy production. However, the most strongly induced protein was the l-amino acid oxidase (LAAO) Interleukin 4 induced 1 (IL4I1) with a 42.9 fold higher abundance (ANOVA p value 2.91-10). IL4I1 has previously been found in B cells, macrophages, dendritic cells and rare neurons. Increased IL4I1 abundance in AEC II was confirmed by qPCR, Western blot and immunohistology. Furthermore, A. fumigatus infected lungs showed high levels of IL4I1 metabolic products. Importantly, higher IL4I1 abundance was also confirmed in lung tissue from human aspergilloma. Because LAAO are key enzymes for bactericidal product generation, AEC II might actively participate in pathogen defense. We provide insights into proteome changes of primary AEC II thereby opening new avenues to analyze the molecular changes of this central lung cell on infectious threats. Data are available via ProteomeXchange with identifier PXD005834.
Collapse
Affiliation(s)
- Pegah Seddigh
- From the ‡University Duisburg-Essen, University Hospital, Institute for Experimental Immunology and Imaging, 45147 Essen; Germany
| | - Thilo Bracht
- ¶Ruhr-Universität Bochum, Medizinisches Proteom-Center, 44801 Bochum, Germany
| | | | - Flavia Castellano
- **INSERM U955, Equipe 09, UMR_S955, UPEC, APHP, Hôpital H Mondor, Créteil, France
| | - Olaf Kniemeyer
- ‖Leibniz Institute for Natural Product Research and Infection Biology, Hans-Knöll-Institutes (HKI), Department of Molecular and Applied Microbiology, Jena, 07745 Jena, Germany
| | - Marc Schuster
- From the ‡University Duisburg-Essen, University Hospital, Institute for Experimental Immunology and Imaging, 45147 Essen; Germany
| | - Juliane Weski
- From the ‡University Duisburg-Essen, University Hospital, Institute for Experimental Immunology and Imaging, 45147 Essen; Germany
| | - Anja Hasenberg
- From the ‡University Duisburg-Essen, University Hospital, Institute for Experimental Immunology and Imaging, 45147 Essen; Germany
| | - Andreas Kraus
- From the ‡University Duisburg-Essen, University Hospital, Institute for Experimental Immunology and Imaging, 45147 Essen; Germany
| | - Gernot Poschet
- §§Centre for Organismal Studies (COS), University of Heidelberg, Heidelberg, Germany
| | - Thomas Hager
- ¶¶University Duisburg-Essen, University Hospital, Institute for Pathology, 45147 Essen, Germany
| | - Dirk Theegarten
- ¶¶University Duisburg-Essen, University Hospital, Institute for Pathology, 45147 Essen, Germany
| | - Christiane A Opitz
- ‡‡German Cancer Research Center (DKFZ), Junior Group Brain Cancer Metabolism (G161), 69120 Heidelberg, Germany
| | - Axel A Brakhage
- ‖Leibniz Institute for Natural Product Research and Infection Biology, Hans-Knöll-Institutes (HKI), Department of Molecular and Applied Microbiology, Jena, 07745 Jena, Germany
| | - Barbara Sitek
- ¶Ruhr-Universität Bochum, Medizinisches Proteom-Center, 44801 Bochum, Germany;
| | - Mike Hasenberg
- §University Duisburg-Essen, University Hospital, Imaging Center Essen (IMCES), Electron Microscopy Unit, 45147 Essen, Germany;
| | - Matthias Gunzer
- From the ‡University Duisburg-Essen, University Hospital, Institute for Experimental Immunology and Imaging, 45147 Essen; Germany;
| |
Collapse
|