1
|
Shen R, Ding Y, Dong Q, Wang Y, Yu J, Pan C, Cai Y, Li Z, Zhang J, Yu K, Zeng Q. IL-4-Induced Gene 1: A Potential Player in Myocardial Infarction. Rev Cardiovasc Med 2024; 25:337. [PMID: 39355609 PMCID: PMC11440439 DOI: 10.31083/j.rcm2509337] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 04/18/2024] [Accepted: 05/06/2024] [Indexed: 10/03/2024] Open
Abstract
Myocardial infarction (MI), a severe outcome of cardiovascular disease, poses a serious threat to human health. Uncontrolled inflammation and excessive cardiomyocyte death, following an infarction event, significantly contribute to both the mortality rate and complications associated with MI. The protein IL-4-induced gene 1 (IL4I1 or FIG1) serves as a natural inhibitor of innate and adaptive immunity, playing a crucial role in CD4+ T cell differentiation, macrophage polarization, and ferroptosis inhibition. Previous studies have linked IL4I1 to acute MI. This review summarizes evidence from both basic and clinical research, highlighting IL4I1 as a critical immunoregulatory enzyme that not only regulates inflammatory responses, but also potentially mitigates MI-induced damage.
Collapse
Affiliation(s)
- Rui Shen
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430022 Wuhan, Hubei, China
- Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430022 Wuhan, Hubei, China
- Hubei Provincial Engineering Research Center of Immunological Diagnosis and Therapy for Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430022 Wuhan, Hubei, China
| | - Yan Ding
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430022 Wuhan, Hubei, China
- Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430022 Wuhan, Hubei, China
- Hubei Provincial Engineering Research Center of Immunological Diagnosis and Therapy for Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430022 Wuhan, Hubei, China
| | - Qian Dong
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430022 Wuhan, Hubei, China
- Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430022 Wuhan, Hubei, China
- Hubei Provincial Engineering Research Center of Immunological Diagnosis and Therapy for Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430022 Wuhan, Hubei, China
| | - Yue Wang
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430022 Wuhan, Hubei, China
- Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430022 Wuhan, Hubei, China
- Hubei Provincial Engineering Research Center of Immunological Diagnosis and Therapy for Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430022 Wuhan, Hubei, China
| | - Jian Yu
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430022 Wuhan, Hubei, China
- Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430022 Wuhan, Hubei, China
- Hubei Provincial Engineering Research Center of Immunological Diagnosis and Therapy for Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430022 Wuhan, Hubei, China
| | - Chengliang Pan
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430022 Wuhan, Hubei, China
- Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430022 Wuhan, Hubei, China
- Hubei Provincial Engineering Research Center of Immunological Diagnosis and Therapy for Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430022 Wuhan, Hubei, China
| | - Yifan Cai
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430022 Wuhan, Hubei, China
- Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430022 Wuhan, Hubei, China
- Hubei Provincial Engineering Research Center of Immunological Diagnosis and Therapy for Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430022 Wuhan, Hubei, China
| | - Zhiyang Li
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430022 Wuhan, Hubei, China
- Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430022 Wuhan, Hubei, China
- Hubei Provincial Engineering Research Center of Immunological Diagnosis and Therapy for Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430022 Wuhan, Hubei, China
| | - Jiangmei Zhang
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430022 Wuhan, Hubei, China
- Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430022 Wuhan, Hubei, China
- Hubei Provincial Engineering Research Center of Immunological Diagnosis and Therapy for Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430022 Wuhan, Hubei, China
| | - Kunwu Yu
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430022 Wuhan, Hubei, China
- Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430022 Wuhan, Hubei, China
- Hubei Provincial Engineering Research Center of Immunological Diagnosis and Therapy for Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430022 Wuhan, Hubei, China
| | - Qiutang Zeng
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430022 Wuhan, Hubei, China
- Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430022 Wuhan, Hubei, China
- Hubei Provincial Engineering Research Center of Immunological Diagnosis and Therapy for Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430022 Wuhan, Hubei, China
| |
Collapse
|
2
|
Grobben Y. Targeting amino acid-metabolizing enzymes for cancer immunotherapy. Front Immunol 2024; 15:1440269. [PMID: 39211039 PMCID: PMC11359565 DOI: 10.3389/fimmu.2024.1440269] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Accepted: 07/23/2024] [Indexed: 09/04/2024] Open
Abstract
Despite the immune system's role in the detection and eradication of abnormal cells, cancer cells often evade elimination by exploitation of various immune escape mechanisms. Among these mechanisms is the ability of cancer cells to upregulate amino acid-metabolizing enzymes, or to induce these enzymes in tumor-infiltrating immunosuppressive cells. Amino acids are fundamental cellular nutrients required for a variety of physiological processes, and their inadequacy can severely impact immune cell function. Amino acid-derived metabolites can additionally dampen the anti-tumor immune response by means of their immunosuppressive activities, whilst some can also promote tumor growth directly. Based on their evident role in tumor immune escape, the amino acid-metabolizing enzymes glutaminase 1 (GLS1), arginase 1 (ARG1), inducible nitric oxide synthase (iNOS), indoleamine 2,3-dioxygenase 1 (IDO1), tryptophan 2,3-dioxygenase (TDO) and interleukin 4 induced 1 (IL4I1) each serve as a promising target for immunotherapeutic intervention. This review summarizes and discusses the involvement of these enzymes in cancer, their effect on the anti-tumor immune response and the recent progress made in the preclinical and clinical evaluation of inhibitors targeting these enzymes.
Collapse
|
3
|
Peng X, Liu C, Zhang L, Chen Y, Mao L, Gao S, Shi X, Zuo L. IL4I1: a novel molecular biomarker represents an inflamed tumor microenvironment and precisely predicts the molecular subtype and immunotherapy response of bladder cancer. Front Pharmacol 2024; 15:1365683. [PMID: 38873416 PMCID: PMC11169701 DOI: 10.3389/fphar.2024.1365683] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Accepted: 05/09/2024] [Indexed: 06/15/2024] Open
Abstract
Introduction: IL4I1, also known as Interleukin-4-induced gene 1, is an enzyme that can modulate the immune system by acting as a L-amino acid oxidase. Nevertheless, a precise understanding of the correlation of IL4I1 with immunological features and immunotherapy efficacy in bladder cancer (BLCA) remains incomplete. Methods: We analyzed RNA sequencing data from the Cancer Genome Atlas (TCGA) to investigate the immune function and prognostic importance of IL4I1 across different cancer types. We further examined the TCGA-BLCA cohort for correlations between IL4I1 and various immunological characteristics of tumor microenvironment (TME), such as cancer immune cycle, immune cell infiltration, immune checkpoint expression and T cell inflamed score. Validation was conducted using two independent cohort, GSE48075 and E-MTAB-4321. Finally, RNA sequencing data from the IMvigor210 cohort and immunohistochemistry assays were employed to validate the predictive value of IL4I1 for the TME and immunotherapy efficacy. Results: In our findings, a positive correlation was observed between IL4I1 expression and immunomodulators expression, immune cell infiltration, the cancer immune cycle, and T cell inflamed score in BLCA, suggesting a significant link to the inflamed TME. In addition, studies have shown that IL4I1 elevated levels of individuals tend to be more performance for basal subtype and exhibit enhanced response rates to diverse treatment modalities, specifically immunotherapy. Clinical data from the IMvigor 210 cohort confirmed a higher rate of response to immunotherapy and better survival benefits in patients with high IL4I1 expression. Discussion: To summarize, our research showed that elevated IL4I1 levels are indicative of an inflamed TME, the basal subtype, and a more favorable response to various treatment methods, especially immune checkpoint blockade therapy in BLCA.
Collapse
Affiliation(s)
- Xiangrong Peng
- Department of Urology, ChangZhou No.2 people’s Hospital, Nanjing Medical University, Changzhou, Jiangsu, China
- Laboratory of Urology, ChangZhou Medical Center, Nanjing Medical University, Changzhou, Jiangsu, China
| | - Chuan Liu
- Department of Urology, ChangZhou No.2 people’s Hospital, Nanjing Medical University, Changzhou, Jiangsu, China
- Laboratory of Urology, ChangZhou Medical Center, Nanjing Medical University, Changzhou, Jiangsu, China
| | - Li Zhang
- Department of Urology, ChangZhou No.2 people’s Hospital, Nanjing Medical University, Changzhou, Jiangsu, China
- Laboratory of Urology, ChangZhou Medical Center, Nanjing Medical University, Changzhou, Jiangsu, China
| | - Yin Chen
- Department of Urology, ChangZhou No.2 people’s Hospital, Nanjing Medical University, Changzhou, Jiangsu, China
- Laboratory of Urology, ChangZhou Medical Center, Nanjing Medical University, Changzhou, Jiangsu, China
| | - Lixin Mao
- Department of Urology, ChangZhou No.2 people’s Hospital, Nanjing Medical University, Changzhou, Jiangsu, China
- Laboratory of Urology, ChangZhou Medical Center, Nanjing Medical University, Changzhou, Jiangsu, China
| | - Shenglin Gao
- Department of Urology, ChangZhou No.2 people’s Hospital, Nanjing Medical University, Changzhou, Jiangsu, China
- Laboratory of Urology, ChangZhou Medical Center, Nanjing Medical University, Changzhou, Jiangsu, China
- Department of Urology, Gonghe County Hospital of Traditional Chinese Medicine, Hainan Tibetan Autonomous Prefecture, Qinghai, China
| | - Xiaokai Shi
- Department of Urology, ChangZhou No.2 people’s Hospital, Nanjing Medical University, Changzhou, Jiangsu, China
- Laboratory of Urology, ChangZhou Medical Center, Nanjing Medical University, Changzhou, Jiangsu, China
| | - Li Zuo
- Department of Urology, ChangZhou No.2 people’s Hospital, Nanjing Medical University, Changzhou, Jiangsu, China
- Laboratory of Urology, ChangZhou Medical Center, Nanjing Medical University, Changzhou, Jiangsu, China
| |
Collapse
|
4
|
Zhang K, Mishra A, Jagannath C. New insight into arginine and tryptophan metabolism in macrophage activation during tuberculosis. Front Immunol 2024; 15:1363938. [PMID: 38605962 PMCID: PMC11008464 DOI: 10.3389/fimmu.2024.1363938] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2023] [Accepted: 03/11/2024] [Indexed: 04/13/2024] Open
Abstract
Arginine and tryptophan are pivotal in orchestrating cytokine-driven macrophage polarization and immune activation. Specifically, interferon-gamma (IFN-γ) stimulates inducible nitric oxide synthase (iNOS) expression), leading to the conversion of arginine into citrulline and nitric oxide (NO), while Interleukin-4 (IL4) promotes arginase activation, shifting arginine metabolism toward ornithine. Concomitantly, IFN-γ triggers indoleamine 2,3-dioxygenase 1 (IDO1) and Interleukin-4 induced 1 (IL4i1), resulting in the conversion of tryptophan into kynurenine and indole-3-pyruvic acid. These metabolic pathways are tightly regulated by NAD+-dependent sirtuin proteins, with Sirt2 and Sirt5 playing integral roles. In this review, we present novel insights that augment our understanding of the metabolic pathways of arginine and tryptophan following Mycobacterium tuberculosis infection, particularly their relevance in macrophage responses. Additionally, we discuss arginine methylation and demethylation and the role of Sirt2 and Sirt5 in regulating tryptophan metabolism and arginine metabolism, potentially driving macrophage polarization.
Collapse
Affiliation(s)
- Kangling Zhang
- Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, TX, United States
| | - Abhishek Mishra
- Department of Pathology and Genomic Medicine, Houston Methodist Research Institute, Weill-Cornell Medicine, Houston, TX, United States
| | - Chinnaswamy Jagannath
- Department of Pathology and Genomic Medicine, Houston Methodist Research Institute, Weill-Cornell Medicine, Houston, TX, United States
| |
Collapse
|
5
|
Bekkat F, Seradj M, Lengagne R, Fiore F, Kato M, Lucas B, Castellano F, Molinier-Frenkel V, Richard Y, Prévost-Blondel A. Upregulation of IL4-induced gene 1 enzyme by B2 cells during melanoma progression impairs their antitumor properties. Eur J Immunol 2024; 54:e2350615. [PMID: 38400692 DOI: 10.1002/eji.202350615] [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: 06/19/2023] [Revised: 01/17/2024] [Accepted: 01/27/2024] [Indexed: 02/25/2024]
Abstract
B cells present in human cutaneous melanoma have been associated with protective or detrimental effects on disease progression according to their phenotype. By using the RET model of spontaneous melanoma and adoptive transfer of B16 melanoma cells, we show that immature and follicular B2 (B2-FO) cells exert a protective effect on melanoma progression by promoting the generation of effector memory T cells and limiting the recruitment of polymorphonuclear myeloid-derived suppressor cells. Unfortunately, this beneficial effect progressively wanes as a consequence of enhanced expression of the IL4-induced gene 1 (IL4I1) enzyme by immature B cells and B2-FO cells. Endogenous IL4I1 selectively decreases CXCR5 expression in splenic immature B cells, subverting their trafficking to primary tumors and enhancing the production of IL-10 by B2 cells, thereby promoting an immunosuppressive microenvironment. Accordingly, B2 cells from RET IL4I1KO mice more efficiently controlled B16 melanoma growth than B2 cells from IL4I1-competent RET mice. Collectively, immature B cells and B2-FO cells are key actors in the control of melanoma growth, but their mobility and functions are differently impaired by IL4I1 overexpression during melanoma progression. Thus, our present data strongly urge us to associate an IL4I1 antagonist with current immunotherapy to improve the treatment of metastatic melanoma.
Collapse
Affiliation(s)
- Fériel Bekkat
- Université Paris Cité, CNRS UMR 8104, INSERM U1016, Institut Cochin, Paris, France
| | - Malvina Seradj
- Université Paris Cité, CNRS UMR 8104, INSERM U1016, Institut Cochin, Paris, France
| | - Renée Lengagne
- Université Paris Cité, CNRS UMR 8104, INSERM U1016, Institut Cochin, Paris, France
| | - Frédéric Fiore
- Centre d'Immunophénomique (CIPHE), Aix Marseille Université, INSERM, CNRS, CELPHEDIA, PHENOMIN, Marseille, France
| | - Masashi Kato
- Department of Occupational and Environmental Health, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan
| | - Bruno Lucas
- Université Paris Cité, CNRS UMR 8104, INSERM U1016, Institut Cochin, Paris, France
| | - Flavia Castellano
- Université Paris Est Créteil, INSERM, IMRB, Créteil, France
- AP-HP, Hôpital Henri Mondor, Département d'Hématologie-Immunologie, Créteil, France
| | - Valérie Molinier-Frenkel
- Université Paris Est Créteil, INSERM, IMRB, Créteil, France
- AP-HP, Hôpital Henri Mondor, Département d'Hématologie-Immunologie, Créteil, France
| | - Yolande Richard
- Université Paris Cité, CNRS UMR 8104, INSERM U1016, Institut Cochin, Paris, France
| | | |
Collapse
|
6
|
Rannikko JH, Bono P, Hynninen J, Hollmén M. Bexmarilimab Activates Human Tumor-Associated Macrophages to Support Adaptive Immune Responses in Interferon-Poor Immune Microenvironments. Cancer Immunol Res 2024; 12:48-59. [PMID: 37922365 PMCID: PMC10762336 DOI: 10.1158/2326-6066.cir-23-0350] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Revised: 09/11/2023] [Accepted: 10/31/2023] [Indexed: 11/05/2023]
Abstract
Immune checkpoint inhibitors (ICI) show substantially greater efficacy in inflamed tumors characterized by preexisting T-cell infiltration and IFN signaling than in noninflamed "cold" tumors, which often remain immunotherapy resistant. The cancer immunotherapy bexmarilimab, which inhibits the scavenger receptor Clever-1 to release macrophage immunosuppression and activate adaptive immunity, has shown treatment benefit in subsets of patients with advanced solid malignancies. However, the mechanisms that determine bexmarilimab therapy outcome in individual patients are unknown. Here we characterized bexmarilimab response in ovarian cancer ascites macrophages ex vivo using single-cell RNA sequencing and demonstrated increased IFN signaling and CXCL10 secretion following bexmarilimab treatment. We further showed that bexmarilimab was most efficacious in macrophages with low baseline IFN signaling, as chronic IFNγ priming abolished bexmarilimab-induced TNFα release. These results highlight an approach to target immunologically cold tumors and to increase the likelihood of their subsequent response to ICIs.
Collapse
Affiliation(s)
- Jenna H. Rannikko
- MediCity Research Laboratory and InFLAMES Flagship, University of Turku, Turku, Finland
| | | | - Johanna Hynninen
- Department of Obstetrics and Gynecology, University of Turku and Turku University Hospital, Turku, Finland
| | - Maija Hollmén
- MediCity Research Laboratory and InFLAMES Flagship, University of Turku, Turku, Finland
| |
Collapse
|
7
|
Wang S, Huang T, Xie Z, Wan L, Ren H, Wu T, Xie L, Luo S, Li M, Xie Z, Fan Q, Huang J, Zeng T, Zhang Y, Zhang M, Wei Y. Transcriptomic and Translatomic Analyses Reveal Insights into the Signaling Pathways of the Innate Immune Response in the Spleens of SPF Chickens Infected with Avian Reovirus. Viruses 2023; 15:2346. [PMID: 38140587 PMCID: PMC10747248 DOI: 10.3390/v15122346] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Revised: 11/23/2023] [Accepted: 11/24/2023] [Indexed: 12/24/2023] Open
Abstract
Avian reovirus (ARV) infection is prevalent in farmed poultry and causes viral arthritis and severe immunosuppression. The spleen plays a very important part in protecting hosts against infectious pathogens. In this research, transcriptome and translatome sequencing technology were combined to investigate the mechanisms of transcriptional and translational regulation in the spleen after ARV infection. On a genome-wide scale, ARV infection can significantly reduce the translation efficiency (TE) of splenic genes. Differentially expressed translational efficiency genes (DTEGs) were identified, including 15 upregulated DTEGs and 396 downregulated DTEGs. These DTEGs were mainly enriched in immune regulation signaling pathways, which indicates that ARV infection reduces the innate immune response in the spleen. In addition, combined analyses revealed that the innate immune response involves the effects of transcriptional and translational regulation. Moreover, we discovered the key gene IL4I1, the most significantly upregulated gene at both the transcriptional and translational levels. Further studies in DF1 cells showed that overexpression of IL4I1 could inhibit the replication of ARV, while inhibiting the expression of endogenous IL4I1 with siRNA promoted the replication of ARV. Overexpression of IL4I1 significantly downregulated the mRNA expression of IFN-β, LGP2, TBK1 and NF-κB; however, the expression of these genes was significantly upregulated after inhibition of IL4I1, suggesting that IL4I1 may be a negative feedback effect of innate immune signaling pathways. In addition, there may be an interaction between IL4I1 and ARV σA protein, and we speculate that the IL4I1 protein plays a regulatory role by interacting with the σA protein. This study not only provides a new perspective on the regulatory mechanisms of the innate immune response after ARV infection but also enriches the knowledge of the host defense mechanisms against ARV invasion and the outcome of ARV evasion of the host's innate immune response.
Collapse
Affiliation(s)
- Sheng Wang
- Guangxi Key Laboratory of Veterinary Biotechnology, Guangxi Veterinary Research Institute, Nanning 530000, China; (S.W.); (L.W.); (H.R.); (L.X.); (S.L.); (M.L.); (Z.X.); (Q.F.); (J.H.); (T.Z.); (Y.Z.); (M.Z.); (Y.W.)
- Key Laboratory of China (Guangxi)-ASEAN Cross-Border Animal Disease Prevention and Control, Ministry of Agriculture and Rural Affairs of China, Nanning 530000, China
| | - Tengda Huang
- Division of Liver Surgery, Department of General Surgery, Laboratory of Liver Surgery, and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China;
| | - Zhixun Xie
- Guangxi Key Laboratory of Veterinary Biotechnology, Guangxi Veterinary Research Institute, Nanning 530000, China; (S.W.); (L.W.); (H.R.); (L.X.); (S.L.); (M.L.); (Z.X.); (Q.F.); (J.H.); (T.Z.); (Y.Z.); (M.Z.); (Y.W.)
- Key Laboratory of China (Guangxi)-ASEAN Cross-Border Animal Disease Prevention and Control, Ministry of Agriculture and Rural Affairs of China, Nanning 530000, China
| | - Lijun Wan
- Guangxi Key Laboratory of Veterinary Biotechnology, Guangxi Veterinary Research Institute, Nanning 530000, China; (S.W.); (L.W.); (H.R.); (L.X.); (S.L.); (M.L.); (Z.X.); (Q.F.); (J.H.); (T.Z.); (Y.Z.); (M.Z.); (Y.W.)
- Key Laboratory of China (Guangxi)-ASEAN Cross-Border Animal Disease Prevention and Control, Ministry of Agriculture and Rural Affairs of China, Nanning 530000, China
| | - Hongyu Ren
- Guangxi Key Laboratory of Veterinary Biotechnology, Guangxi Veterinary Research Institute, Nanning 530000, China; (S.W.); (L.W.); (H.R.); (L.X.); (S.L.); (M.L.); (Z.X.); (Q.F.); (J.H.); (T.Z.); (Y.Z.); (M.Z.); (Y.W.)
- Key Laboratory of China (Guangxi)-ASEAN Cross-Border Animal Disease Prevention and Control, Ministry of Agriculture and Rural Affairs of China, Nanning 530000, China
| | - Tian Wu
- NHC Key Laboratory of Transplant Engineering and Immunology, Regenerative Medicine Research Center, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital of Sichuan University, Chengdu 610041, China;
| | - Liji Xie
- Guangxi Key Laboratory of Veterinary Biotechnology, Guangxi Veterinary Research Institute, Nanning 530000, China; (S.W.); (L.W.); (H.R.); (L.X.); (S.L.); (M.L.); (Z.X.); (Q.F.); (J.H.); (T.Z.); (Y.Z.); (M.Z.); (Y.W.)
- Key Laboratory of China (Guangxi)-ASEAN Cross-Border Animal Disease Prevention and Control, Ministry of Agriculture and Rural Affairs of China, Nanning 530000, China
| | - Sisi Luo
- Guangxi Key Laboratory of Veterinary Biotechnology, Guangxi Veterinary Research Institute, Nanning 530000, China; (S.W.); (L.W.); (H.R.); (L.X.); (S.L.); (M.L.); (Z.X.); (Q.F.); (J.H.); (T.Z.); (Y.Z.); (M.Z.); (Y.W.)
- Key Laboratory of China (Guangxi)-ASEAN Cross-Border Animal Disease Prevention and Control, Ministry of Agriculture and Rural Affairs of China, Nanning 530000, China
| | - Meng Li
- Guangxi Key Laboratory of Veterinary Biotechnology, Guangxi Veterinary Research Institute, Nanning 530000, China; (S.W.); (L.W.); (H.R.); (L.X.); (S.L.); (M.L.); (Z.X.); (Q.F.); (J.H.); (T.Z.); (Y.Z.); (M.Z.); (Y.W.)
- Key Laboratory of China (Guangxi)-ASEAN Cross-Border Animal Disease Prevention and Control, Ministry of Agriculture and Rural Affairs of China, Nanning 530000, China
| | - Zhiqin Xie
- Guangxi Key Laboratory of Veterinary Biotechnology, Guangxi Veterinary Research Institute, Nanning 530000, China; (S.W.); (L.W.); (H.R.); (L.X.); (S.L.); (M.L.); (Z.X.); (Q.F.); (J.H.); (T.Z.); (Y.Z.); (M.Z.); (Y.W.)
- Key Laboratory of China (Guangxi)-ASEAN Cross-Border Animal Disease Prevention and Control, Ministry of Agriculture and Rural Affairs of China, Nanning 530000, China
| | - Qing Fan
- Guangxi Key Laboratory of Veterinary Biotechnology, Guangxi Veterinary Research Institute, Nanning 530000, China; (S.W.); (L.W.); (H.R.); (L.X.); (S.L.); (M.L.); (Z.X.); (Q.F.); (J.H.); (T.Z.); (Y.Z.); (M.Z.); (Y.W.)
- Key Laboratory of China (Guangxi)-ASEAN Cross-Border Animal Disease Prevention and Control, Ministry of Agriculture and Rural Affairs of China, Nanning 530000, China
| | - Jiaoling Huang
- Guangxi Key Laboratory of Veterinary Biotechnology, Guangxi Veterinary Research Institute, Nanning 530000, China; (S.W.); (L.W.); (H.R.); (L.X.); (S.L.); (M.L.); (Z.X.); (Q.F.); (J.H.); (T.Z.); (Y.Z.); (M.Z.); (Y.W.)
- Key Laboratory of China (Guangxi)-ASEAN Cross-Border Animal Disease Prevention and Control, Ministry of Agriculture and Rural Affairs of China, Nanning 530000, China
| | - Tingting Zeng
- Guangxi Key Laboratory of Veterinary Biotechnology, Guangxi Veterinary Research Institute, Nanning 530000, China; (S.W.); (L.W.); (H.R.); (L.X.); (S.L.); (M.L.); (Z.X.); (Q.F.); (J.H.); (T.Z.); (Y.Z.); (M.Z.); (Y.W.)
- Key Laboratory of China (Guangxi)-ASEAN Cross-Border Animal Disease Prevention and Control, Ministry of Agriculture and Rural Affairs of China, Nanning 530000, China
| | - Yanfang Zhang
- Guangxi Key Laboratory of Veterinary Biotechnology, Guangxi Veterinary Research Institute, Nanning 530000, China; (S.W.); (L.W.); (H.R.); (L.X.); (S.L.); (M.L.); (Z.X.); (Q.F.); (J.H.); (T.Z.); (Y.Z.); (M.Z.); (Y.W.)
- Key Laboratory of China (Guangxi)-ASEAN Cross-Border Animal Disease Prevention and Control, Ministry of Agriculture and Rural Affairs of China, Nanning 530000, China
| | - Minxiu Zhang
- Guangxi Key Laboratory of Veterinary Biotechnology, Guangxi Veterinary Research Institute, Nanning 530000, China; (S.W.); (L.W.); (H.R.); (L.X.); (S.L.); (M.L.); (Z.X.); (Q.F.); (J.H.); (T.Z.); (Y.Z.); (M.Z.); (Y.W.)
- Key Laboratory of China (Guangxi)-ASEAN Cross-Border Animal Disease Prevention and Control, Ministry of Agriculture and Rural Affairs of China, Nanning 530000, China
| | - You Wei
- Guangxi Key Laboratory of Veterinary Biotechnology, Guangxi Veterinary Research Institute, Nanning 530000, China; (S.W.); (L.W.); (H.R.); (L.X.); (S.L.); (M.L.); (Z.X.); (Q.F.); (J.H.); (T.Z.); (Y.Z.); (M.Z.); (Y.W.)
- Key Laboratory of China (Guangxi)-ASEAN Cross-Border Animal Disease Prevention and Control, Ministry of Agriculture and Rural Affairs of China, Nanning 530000, China
| |
Collapse
|
8
|
Sutton K, Nash T, Sives S, Borowska D, Mitchell J, Vohra P, Stevens MP, Vervelde L. Disentangling the innate immune responses of intestinal epithelial cells and lamina propria cells to Salmonella Typhimurium infection in chickens. Front Microbiol 2023; 14:1258796. [PMID: 37854334 PMCID: PMC10579587 DOI: 10.3389/fmicb.2023.1258796] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Accepted: 09/19/2023] [Indexed: 10/20/2023] Open
Abstract
Salmonella enterica serovar Typhimurium (STm) is a major foodborne pathogen and poultry are a key reservoir of human infections. To understand the host responses to early stages of Salmonella infection in poultry, we infected 2D and 3D enteroids, the latter of which contains leukocytes, neurons, and mesenchymal cells that are characteristic of the lamina propria. We infected these enteroids with wild-type (WT STm), a non-invasive mutant lacking the prgH gene (ΔprgH STm), or treated them with STm lipopolysaccharide (LPS) and analyzed the expression of innate immune related genes by qPCR at 4 and 8 h. The localization of the tight junction protein, ZO-1, expression was disrupted in WT STm infected enteroids but not ΔprgH STm or LPS treated enteroids, suggesting a loss of epithelial barrier integrity. The innate immune response to LPS was more pronounced in 2D enteroids compared to 3D enteroids and by 8 hpi, the response in 3D enteroids was almost negligible. However, when STm adhered to or invaded the enteroids, both 2D and 3D enteroids exhibited an upregulation of inflammatory responses. The presence of lamina propria cells in 3D enteroids resulted in the unique expression of genes associated with immune functions involved in regulating inflammation. Moreover, 2D and 3D enteroids showed temporal differences in response to bacterial invasion or adherence. At 8 hpi, innate responses in 3D but not 2D enteroids continued to increase after infection with WT STm, whereas the responses to the non-invasive strain decreased at 8 hpi in both 2D and 3D enteroids. In conclusion, STm infection of chicken enteroids recapitulated several observations from in vivo studies of Salmonella-infected chickens, including altered epithelial barrier integrity based on ZO-1 expression and inflammatory responses. Our findings provide evidence that Salmonella-infected enteroids serve as effective models for investigating host-pathogen interactions and exploring the molecular mechanisms of microbial virulence although the 3D model mimics the host more accurately due to the presence of a lamina propria.
Collapse
Affiliation(s)
- Kate Sutton
- Division of Immunology, The Roslin Institute, Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh, United Kingdom
| | - Tessa Nash
- Division of Immunology, The Roslin Institute, Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh, United Kingdom
| | - Samantha Sives
- Division of Immunology, The Roslin Institute, Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh, United Kingdom
| | - Dominika Borowska
- Division of Immunology, The Roslin Institute, Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh, United Kingdom
| | - Jordan Mitchell
- Division of Immunology, The Roslin Institute, Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh, United Kingdom
| | - Prerna Vohra
- Institute for Immunology and Infection Research, School of Biological Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Mark P. Stevens
- Division of Bacteriology, The Roslin Institute, Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh, United Kingdom
| | - Lonneke Vervelde
- Division of Immunology, The Roslin Institute, Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|
9
|
Nabbi A, Beck P, Delaidelli A, Oldridge DA, Sudhaman S, Zhu K, Yang SYC, Mulder DT, Bruce JP, Paulson JN, Raman P, Zhu Y, Resnick AC, Sorensen PH, Sill M, Brabetz S, Lambo S, Malkin D, Johann PD, Kool M, Jones DTW, Pfister SM, Jäger N, Pugh TJ. Transcriptional immunogenomic analysis reveals distinct immunological clusters in paediatric nervous system tumours. Genome Med 2023; 15:67. [PMID: 37679810 PMCID: PMC10486055 DOI: 10.1186/s13073-023-01219-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Accepted: 08/07/2023] [Indexed: 09/09/2023] Open
Abstract
BACKGROUND Cancer immunotherapies including immune checkpoint inhibitors and Chimeric Antigen Receptor (CAR) T-cell therapy have shown variable response rates in paediatric patients highlighting the need to establish robust biomarkers for patient selection. While the tumour microenvironment in adults has been widely studied to delineate determinants of immune response, the immune composition of paediatric solid tumours remains relatively uncharacterized calling for investigations to identify potential immune biomarkers. METHODS To inform immunotherapy approaches in paediatric cancers with embryonal origin, we performed an immunogenomic analysis of RNA-seq data from 925 treatment-naïve paediatric nervous system tumours (pedNST) spanning 12 cancer types from three publicly available data sets. RESULTS Within pedNST, we uncovered four broad immune clusters: Paediatric Inflamed (10%), Myeloid Predominant (30%), Immune Neutral (43%) and Immune Desert (17%). We validated these clusters using immunohistochemistry, methylation immune inference and segmentation analysis of tissue images. We report shared biology of these immune clusters within and across cancer types, and characterization of specific immune cell frequencies as well as T- and B-cell repertoires. We found no associations between immune infiltration levels and tumour mutational burden, although molecular cancer entities were enriched within specific immune clusters. CONCLUSIONS Given the heterogeneity of immune infiltration within pedNST, our findings suggest personalized immunogenomic profiling is needed to guide selection of immunotherapeutic strategies.
Collapse
Affiliation(s)
- Arash Nabbi
- Princess Margaret Cancer Centre, University Health Network, Princess Margaret Cancer Research Tower, Room 9-305, MaRS Centre, 101 College Street, Toronto, M5G 1L7, Canada
| | - Pengbo Beck
- Hopp Children's Cancer Center Heidelberg (KiTZ), Heidelberg, Germany
- Division of Pediatric Neurooncology and German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), B062, Im Neuenheimer Feld 580, 69120, Heidelberg, Germany
| | - Alberto Delaidelli
- Department of Molecular Oncology, British Columbia Cancer Agency, Vancouver, Canada
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, Canada
| | - Derek A Oldridge
- Department of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Center for Computational and Genomic Medicine, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Sumedha Sudhaman
- Division of Hematology/Oncology, Department of Pediatrics, The Hospital for Sick Children, University of Toronto, Toronto, Canada
| | - Kelsey Zhu
- Princess Margaret Cancer Centre, University Health Network, Princess Margaret Cancer Research Tower, Room 9-305, MaRS Centre, 101 College Street, Toronto, M5G 1L7, Canada
| | - S Y Cindy Yang
- Princess Margaret Cancer Centre, University Health Network, Princess Margaret Cancer Research Tower, Room 9-305, MaRS Centre, 101 College Street, Toronto, M5G 1L7, Canada
| | - David T Mulder
- Princess Margaret Cancer Centre, University Health Network, Princess Margaret Cancer Research Tower, Room 9-305, MaRS Centre, 101 College Street, Toronto, M5G 1L7, Canada
| | - Jeffrey P Bruce
- Princess Margaret Cancer Centre, University Health Network, Princess Margaret Cancer Research Tower, Room 9-305, MaRS Centre, 101 College Street, Toronto, M5G 1L7, Canada
| | - Joseph N Paulson
- Department of Biostatistics, Genentech Inc, San Francisco, CA, USA
| | - Pichai Raman
- Division of Neurosurgery, Center for Childhood Cancer Research, Department of Biomedical and Health Informatics and Center for Data-Driven Discovery in Biomedicine, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Yuankun Zhu
- Division of Neurosurgery, Center for Childhood Cancer Research, Department of Biomedical and Health Informatics and Center for Data-Driven Discovery in Biomedicine, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Adam C Resnick
- Division of Neurosurgery, Center for Childhood Cancer Research, Department of Biomedical and Health Informatics and Center for Data-Driven Discovery in Biomedicine, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Poul H Sorensen
- Department of Molecular Oncology, British Columbia Cancer Agency, Vancouver, Canada
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, Canada
| | - Martin Sill
- Hopp Children's Cancer Center Heidelberg (KiTZ), Heidelberg, Germany
- Division of Pediatric Neurooncology and German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), B062, Im Neuenheimer Feld 580, 69120, Heidelberg, Germany
| | - Sebastian Brabetz
- Hopp Children's Cancer Center Heidelberg (KiTZ), Heidelberg, Germany
- Division of Pediatric Neurooncology and German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), B062, Im Neuenheimer Feld 580, 69120, Heidelberg, Germany
| | - Sander Lambo
- Hopp Children's Cancer Center Heidelberg (KiTZ), Heidelberg, Germany
- Division of Pediatric Neurooncology and German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), B062, Im Neuenheimer Feld 580, 69120, Heidelberg, Germany
| | - David Malkin
- Division of Hematology/Oncology, Department of Pediatrics, The Hospital for Sick Children, University of Toronto, Toronto, Canada
| | - Pascal D Johann
- Hopp Children's Cancer Center Heidelberg (KiTZ), Heidelberg, Germany
- Division of Pediatric Neurooncology and German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), B062, Im Neuenheimer Feld 580, 69120, Heidelberg, Germany
- Department of Pediatric Hematology and Oncology, Heidelberg University Hospital, Heidelberg, Germany
| | - Marcel Kool
- Hopp Children's Cancer Center Heidelberg (KiTZ), Heidelberg, Germany
- Division of Pediatric Neurooncology and German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), B062, Im Neuenheimer Feld 580, 69120, Heidelberg, Germany
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands
| | - David T W Jones
- Hopp Children's Cancer Center Heidelberg (KiTZ), Heidelberg, Germany
- Division of Pediatric Glioma Research, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Stefan M Pfister
- Hopp Children's Cancer Center Heidelberg (KiTZ), Heidelberg, Germany
- Division of Pediatric Neurooncology and German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), B062, Im Neuenheimer Feld 580, 69120, Heidelberg, Germany
- Department of Pediatric Hematology and Oncology, Heidelberg University Hospital, Heidelberg, Germany
| | - Natalie Jäger
- Hopp Children's Cancer Center Heidelberg (KiTZ), Heidelberg, Germany.
- Division of Pediatric Neurooncology and German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), B062, Im Neuenheimer Feld 580, 69120, Heidelberg, Germany.
| | - Trevor J Pugh
- Princess Margaret Cancer Centre, University Health Network, Princess Margaret Cancer Research Tower, Room 9-305, MaRS Centre, 101 College Street, Toronto, M5G 1L7, Canada.
- Department of Medical Biophysics, University of Toronto, Toronto, Canada.
- Ontario Institute for Cancer Research, Toronto, Canada.
| |
Collapse
|
10
|
Zuo M, Fang J, Huang P, Liu S, Hou P, Wang S, Liu Z, Feng C, Cao L, Li P, Shi Y, Shao C. IL4I1-catalyzed tryptophan metabolites mediate the anti-inflammatory function of cytokine-primed human muscle stem cells. Cell Death Discov 2023; 9:269. [PMID: 37507432 PMCID: PMC10382538 DOI: 10.1038/s41420-023-01568-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Revised: 07/05/2023] [Accepted: 07/19/2023] [Indexed: 07/30/2023] Open
Abstract
Muscle stem cells (MuSCs) have been demonstrated to exert impressive therapeutic efficacy in disease settings through orchestrating inflammatory microenvironments. Nevertheless, the mechanisms underlying the immunoregulatory property of MuSCs remain largely uncharacterized. Here, we showed that interleukin-4-induced-1 (IL4I1), an essential enzyme that catalyzes indole metabolism in humans, was highly expressed in human MuSCs exposed to IFN-γ and TNF-α. Functionally, the MuSCs were found to inhibit the infiltration of neutrophils into sites of inflammation in a IL4I1-dependent manner and thus ameliorate acute lung injury in mice. Mechanistically, the indole metabolites, including indole-3-pyruvic acid (I3P) and indole-3-aldehyde (I3A), produced by IL4I1, acted as ligands to activate aryl hydrocarbon receptor (AHR), leading to augmented expression of TNF-stimulated gene 6 (TSG-6) in inflammatory cytokine-primed MuSCs. Furthermore, I3P administration alone suppressed neutrophil infiltration into damaged lungs. I3P could also reduce the level of reactive oxygen species in neutrophils. Therefore, our study has uncovered a novel mechanism by which MuSCs acquire their immunoregulatory property and may help to develop or optimize MuSC-based therapies for inflammatory diseases.
Collapse
Affiliation(s)
- Muqiu Zuo
- The Third Affiliated Hospital of Soochow University, Institutes for Translational Medicine, State Key Laboratory of Radiation Medicine and Protection, Suzhou Medical College of Soochow University, Suzhou, Jiangsu, 215123, China
| | - Jiankai Fang
- The Third Affiliated Hospital of Soochow University, Institutes for Translational Medicine, State Key Laboratory of Radiation Medicine and Protection, Suzhou Medical College of Soochow University, Suzhou, Jiangsu, 215123, China
| | - Peiqing Huang
- The Third Affiliated Hospital of Soochow University, Institutes for Translational Medicine, State Key Laboratory of Radiation Medicine and Protection, Suzhou Medical College of Soochow University, Suzhou, Jiangsu, 215123, China
| | - Shisong Liu
- The Third Affiliated Hospital of Soochow University, Institutes for Translational Medicine, State Key Laboratory of Radiation Medicine and Protection, Suzhou Medical College of Soochow University, Suzhou, Jiangsu, 215123, China
| | - Pengbo Hou
- The Third Affiliated Hospital of Soochow University, Institutes for Translational Medicine, State Key Laboratory of Radiation Medicine and Protection, Suzhou Medical College of Soochow University, Suzhou, Jiangsu, 215123, China
- Department of Experimental Medicine and Biochemical Sciences, TOR, University of Rome "Tor Vergata", Rome, 00133, Italy
| | - Shiqing Wang
- The Third Affiliated Hospital of Soochow University, Institutes for Translational Medicine, State Key Laboratory of Radiation Medicine and Protection, Suzhou Medical College of Soochow University, Suzhou, Jiangsu, 215123, China
| | - Zhanhong Liu
- The Third Affiliated Hospital of Soochow University, Institutes for Translational Medicine, State Key Laboratory of Radiation Medicine and Protection, Suzhou Medical College of Soochow University, Suzhou, Jiangsu, 215123, China
- Department of Experimental Medicine and Biochemical Sciences, TOR, University of Rome "Tor Vergata", Rome, 00133, Italy
| | - Chao Feng
- The Third Affiliated Hospital of Soochow University, Institutes for Translational Medicine, State Key Laboratory of Radiation Medicine and Protection, Suzhou Medical College of Soochow University, Suzhou, Jiangsu, 215123, China
- Department of Experimental Medicine and Biochemical Sciences, TOR, University of Rome "Tor Vergata", Rome, 00133, Italy
| | - Lijuan Cao
- The Third Affiliated Hospital of Soochow University, Institutes for Translational Medicine, State Key Laboratory of Radiation Medicine and Protection, Suzhou Medical College of Soochow University, Suzhou, Jiangsu, 215123, China
- Department of Experimental Medicine and Biochemical Sciences, TOR, University of Rome "Tor Vergata", Rome, 00133, Italy
| | - Peishan Li
- The Third Affiliated Hospital of Soochow University, Institutes for Translational Medicine, State Key Laboratory of Radiation Medicine and Protection, Suzhou Medical College of Soochow University, Suzhou, Jiangsu, 215123, China
| | - Yufang Shi
- The Third Affiliated Hospital of Soochow University, Institutes for Translational Medicine, State Key Laboratory of Radiation Medicine and Protection, Suzhou Medical College of Soochow University, Suzhou, Jiangsu, 215123, China.
- Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, Shanghai, 200025, China.
| | - Changshun Shao
- The Third Affiliated Hospital of Soochow University, Institutes for Translational Medicine, State Key Laboratory of Radiation Medicine and Protection, Suzhou Medical College of Soochow University, Suzhou, Jiangsu, 215123, China.
| |
Collapse
|
11
|
Stone TW, Williams RO. Interactions of IDO and the Kynurenine Pathway with Cell Transduction Systems and Metabolism at the Inflammation-Cancer Interface. Cancers (Basel) 2023; 15:cancers15112895. [PMID: 37296860 DOI: 10.3390/cancers15112895] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Revised: 05/10/2023] [Accepted: 05/12/2023] [Indexed: 06/12/2023] Open
Abstract
The mechanisms underlying a relationship between inflammation and cancer are unclear, but much emphasis has been placed on the role of tryptophan metabolism to kynurenine and downstream metabolites, as these make a substantial contribution to the regulation of immune tolerance and susceptibility to cancer. The proposed link is supported by the induction of tryptophan metabolism by indoleamine-2,3-dioxygenase (IDO) or tryptophan-2,3-dioxygenase (TDO), in response to injury, infection or stress. This review will summarize the kynurenine pathway and will then focus on the bi-directional interactions with other transduction pathways and cancer-related factors. The kynurenine pathway can interact with and modify activity in many other transduction systems, potentially generating an extended web of effects other than the direct effects of kynurenine and its metabolites. Conversely, the pharmacological targeting of those other systems could greatly enhance the efficacy of changes in the kynurenine pathway. Indeed, manipulating those interacting pathways could affect inflammatory status and tumor development indirectly via the kynurenine pathway, while pharmacological modulation of the kynurenine pathway could indirectly influence anti-cancer protection. While current efforts are progressing to account for the failure of selective IDO1 inhibitors to inhibit tumor growth and to devise means of circumventing the issue, it is clear that there are wider factors involving the relationship between kynurenines and cancer that merit detailed consideration as alternative drug targets.
Collapse
Affiliation(s)
- Trevor W Stone
- The Kennedy Institute of Rheumatology, NDORMS, University of Oxford, Oxford OX3 7FY, UK
| | - Richard O Williams
- The Kennedy Institute of Rheumatology, NDORMS, University of Oxford, Oxford OX3 7FY, UK
| |
Collapse
|
12
|
Kabat AM, Pearce EL, Pearce EJ. Metabolism in type 2 immune responses. Immunity 2023; 56:723-741. [PMID: 37044062 PMCID: PMC10938369 DOI: 10.1016/j.immuni.2023.03.007] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 03/11/2023] [Accepted: 03/15/2023] [Indexed: 04/14/2023]
Abstract
The immune response is tailored to the environment in which it takes place. Immune cells sense and adapt to changes in their surroundings, and it is now appreciated that in addition to cytokines made by stromal and epithelial cells, metabolic cues provide key adaptation signals. Changes in immune cell activation states are linked to changes in cellular metabolism that support function. Furthermore, metabolites themselves can signal between as well as within cells. Here, we discuss recent progress in our understanding of how metabolic regulation relates to type 2 immunity firstly by considering specifics of metabolism within type 2 immune cells and secondly by stressing how type 2 immune cells are integrated more broadly into the metabolism of the organism as a whole.
Collapse
Affiliation(s)
- Agnieszka M Kabat
- Bloomberg Kimmel Institute, and Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Erika L Pearce
- Bloomberg Kimmel Institute, and Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA; Department of Biochemistry and Molecular Biology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD 21287, USA
| | - Edward J Pearce
- Bloomberg Kimmel Institute, and Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA; Department of Molecular Microbiology and Immunology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD 21287, USA.
| |
Collapse
|
13
|
Human Chorionic Gonadotropin-Stimulated Interleukin-4-Induced-1 (IL4I1) Promotes Human Decidualization via Aryl Hydrocarbon Receptor. Int J Mol Sci 2023; 24:ijms24043163. [PMID: 36834576 PMCID: PMC9959871 DOI: 10.3390/ijms24043163] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Revised: 01/16/2023] [Accepted: 01/20/2023] [Indexed: 02/08/2023] Open
Abstract
Decidualization is necessary for the successful establishment of early pregnancy in rodents and humans. Disturbed decidualization results in recurrent implantation failure, recurrent spontaneous abortion, and preeclampsia. Tryptophan (Trp), one of the essential amino acids in humans, has a positive effect on mammalian pregnancy. Interleukin 4-induced gene 1 (IL4I1) is a recently identified enzyme that can metabolize L-Trp to activate aryl hydrocarbon receptor (AHR). Although IDO1-catalyzed kynurenine (Kyn) from Trp has been shown to enhance human in vitro decidualization via activating AHR, whether IL4I1-catalyzed metabolites of Trp are involved in human decidualization is still unknown. In our study, human chorionic gonadotropin stimulates IL4I1 expression and secretion from human endometrial epithelial cells through ornithine decarboxylase-induced putrescine production. Either IL4I1-catalyzed indole-3-pyruvic acid (I3P) or its metabolite indole-3-aldehyde (I3A) from Trp is able to induce human in vitro decidualization by activating AHR. As a target gene of AHR, Epiregulin induced by I3P and I3A promotes human in vitro decidualization. Our study indicates that IL4I1-catalyzed metabolites from Trp can enhance human in vitro decidualization through AHR-Epiregulin pathway.
Collapse
|
14
|
The exploitation of enzyme-based cancer immunotherapy. Hum Cell 2023; 36:98-120. [PMID: 36334180 DOI: 10.1007/s13577-022-00821-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Accepted: 10/29/2022] [Indexed: 11/06/2022]
Abstract
Cancer immunotherapy utilizes the immune system and its wide-ranging components to deliver anti-tumor responses. In immune escape mechanisms, tumor microenvironment-associated soluble factors and cell surface-bound molecules are mainly accountable for the dysfunctional activity of tumor-specific CD8+ T cells, natural killer (NK) cells, tumor associated macrophages (TAMs) and stromal cells. The myeloid-derived suppressor cells (MDSCs) and Foxp3+ regulatory T cells (Tregs), are also key tumor-promoting immune cells. These potent immunosuppressive networks avert tumor rejection at various stages, affecting immunotherapies' outcomes. Numerous clinical trials have elucidated that disruption of immunosuppression could be achieved via checkpoint inhibitors. Another approach utilizes enzymes that can restore the body's potential to counter cancer by triggering the immune system inhibited by the tumor microenvironment. These immunotherapeutic enzymes can catalyze an immunostimulatory signal and modulate the tumor microenvironment via effector molecules. Herein, we have discussed the immuno-metabolic roles of various enzymes like ATP-dephosphorylating ectoenzymes, inducible Nitric Oxide Synthase, phenylamine, tryptophan, and arginine catabolizing enzymes in cancer immunotherapy. Understanding the detailed molecular mechanisms of the enzymes involved in modulating the tumor microenvironment may help find new opportunities for cancer therapeutics.
Collapse
|
15
|
Gatineau J, Nidercorne C, Dupont A, Puiffe ML, Cohen JL, Molinier-Frenkel V, Niedergang F, Castellano F. IL4I1 binds to TMPRSS13 and competes with SARS-CoV-2 spike. Front Immunol 2022; 13:982839. [PMID: 36131918 PMCID: PMC9483092 DOI: 10.3389/fimmu.2022.982839] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Accepted: 08/09/2022] [Indexed: 11/26/2022] Open
Abstract
The secreted enzyme interleukin four-induced gene 1 (IL4I1) is involved in the negative control of the adaptive immune response. IL4I1 expression in human cancer is frequent and correlates with poor survival and resistance to immunotherapy. Nevertheless, its mechanism of action remains partially unknown. Here, we identified transmembrane serine protease 13 (TMPRSS13) as an immune cell-expressed surface protein that binds IL4I1. TMPRSS13 is a paralog of TMPRSS2, of which the protease activity participates in the cleavage of SARS-CoV-2 spike protein and facilitates virus induced-membrane fusion. We show that TMPRSS13 is expressed by human lymphocytes, monocytes and monocyte-derived macrophages, can cleave the spike protein and allow SARS-CoV-2 spike pseudotyped virus entry into cells. We identify regions of homology between IL4I1 and spike and demonstrate competition between the two proteins for TMPRSS13 binding. These findings may be relevant for both interfering with SARS-CoV-2 infection and limiting IL4I1-dependent immunosuppressive activity in cancer.
Collapse
Affiliation(s)
| | | | | | | | - José L. Cohen
- Univ Paris Est Creteil, INSERM, IMRB, Creteil, France
- AP-HP, Hopital H Mondor, CIC Biotherapies, Créteil, France
| | - Valérie Molinier-Frenkel
- Univ Paris Est Creteil, INSERM, IMRB, Creteil, France
- AP-HP, Hopital Henri Mondor, Departement d’Hematologie-Immunologie, Créteil, France
- *Correspondence: Flavia Castellano, ; Florence Niedergang, ; Valérie Molinier-Frenkel,
| | - Florence Niedergang
- Université Paris Cité, CNRS, INSERM, Institut Cochin, CNRS, Paris, France
- *Correspondence: Flavia Castellano, ; Florence Niedergang, ; Valérie Molinier-Frenkel,
| | - Flavia Castellano
- Univ Paris Est Creteil, INSERM, IMRB, Creteil, France
- AP-HP, Hopital Henri Mondor, Plateforme des Ressources Biologiques, Créteil, France
- *Correspondence: Flavia Castellano, ; Florence Niedergang, ; Valérie Molinier-Frenkel,
| |
Collapse
|
16
|
Kasai K, Nakano M, Ohishi M, Nakamura T, Miura T. Antimicrobial properties of L-amino acid oxidase: biochemical features and biomedical applications. Appl Microbiol Biotechnol 2021; 105:4819-4832. [PMID: 34106313 PMCID: PMC8188536 DOI: 10.1007/s00253-021-11381-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Revised: 05/17/2021] [Accepted: 05/28/2021] [Indexed: 12/19/2022]
Abstract
Abstract Mucus layer that covers the body surface of various animal functions as a defense barrier against microbes, environmental xenobiotics, and predators. Previous studies have reported that L-amino acid oxidase (LAAO), present in several animal fluids, has potent properties against pathogenic bacteria, viruses, and parasites. LAAO catalyzes the oxidative deamination of specific L-amino acids with the generation of hydrogen peroxide and L-amino acid metabolites. Further, the generated hydrogen peroxide is involved in oxidation (direct effect) while the metabolites activate immune responses (indirect effect). Therefore, LAAO exhibits two different mechanisms of bioactivation. Previously, we described the selective, specific, and local oxidative and potent antibacterial actions of various LAAOs as potential therapeutic strategies. In this review, we focus on their biochemical features, enzymatic regulations, and biomedical applications with a view of describing their probable role as biochemical agents and biomarkers for microbial infections, cancer, and autoimmune-mediated diseases. We consider that LAAOs hold implications in biomedicine owing to their antimicrobial activity wherein they can be used in treatment of infectious diseases and as diagnostic biomarkers in the above-mentioned diseased conditions. Key points •Focus on biochemical features, enzymatic regulation, and biomedical applications of LAAOs. •Mechanisms of antimicrobial activity, inflammatory regulation, and immune responses of LAAOs. •Potential biomedical application as an antimicrobial and anti-infection agent, and disease biomarker.
Collapse
Affiliation(s)
- Kosuke Kasai
- Department of Biomedical Sciences, Division of Medical Life Sciences, Graduate School of Health Sciences, Hirosaki University, 66-1, Hon-cho, 036-8564, Hirosaki, Aomori, Japan
| | - Manabu Nakano
- Department of Biomedical Sciences, Division of Medical Life Sciences, Graduate School of Health Sciences, Hirosaki University, 66-1, Hon-cho, 036-8564, Hirosaki, Aomori, Japan
| | | | - Toshiya Nakamura
- Department of Biomedical Sciences, Division of Medical Life Sciences, Graduate School of Health Sciences, Hirosaki University, 66-1, Hon-cho, 036-8564, Hirosaki, Aomori, Japan
| | - Tomisato Miura
- Department of Risk Analysis and Biodosimetry, Institute of Radiation Emergency Medicine, Hirosaki University, 66-1, Hon-cho, 036-8564, Hirosaki, Aomori, Japan.
| |
Collapse
|
17
|
Castellano F, Prevost-Blondel A, Cohen JL, Molinier-Frenkel V. What role for AHR activation in IL4I1-mediated immunosuppression ? Oncoimmunology 2021; 10:1924500. [PMID: 34026337 PMCID: PMC8118450 DOI: 10.1080/2162402x.2021.1924500] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The amino-acid catabolizing enzyme Interleukin-4 induced gene 1 (IL4I1) remains poorly characterized despite it is emerging as a pertinent therapeutic target for cancer. IL4I1 is secreted in the synaptic cleft by antigen-presenting cells. It inhibits TCR signaling, modulates naïve T cell differentiation and limits effector T cell proliferation. IL4I1 expression in tumors shapes the tumor microenvironment and impairs the antitumor cytotoxic T cell response, thereby facilitating cancer immune escape. Several mechanisms participate in these effects. Recent data suggest a role of new IL4I1 metabolites in activation of the aryl-hydrocarbon receptor (AHR). Here, we observe that expression of IL4I1 is poorly correlated with that of validated targets of AHR in human cancers. Moreover, dendritic cells do not upregulate AHR target genes in relation with IL4I1 expression in vivo. Finally, IL4I1 activity toward tryptophan leading to production of AHR-activating products is very low, and should be negligible when tryptophan-degrading enzymes of higher affinity compete for the substrate. We recently showed that IL4I1 expression by dendritic cells directly regulates immune synapse formation and modulates the repertoire and memory differentiation of responding CD8 T cells after viral infection. Thus, IL4I1 may restrain tumor control through regulating the priming of tumor-specific CD8 T cells, independently of AHR activation.
Collapse
Affiliation(s)
- Flavia Castellano
- INSERM, IMRB, Univ Paris Est Creteil, Creteil, France.,Departement d'Hematologie-Immunologie, AP-HP, Hopital Henri Mondor, Créteil, France
| | | | - José L Cohen
- INSERM, IMRB, Univ Paris Est Creteil, Creteil, France.,Centre d'investigation Clinique en Biothérapie, AP-HP, Hopital Henri Mondor, Créteil, France
| | - Valérie Molinier-Frenkel
- INSERM, IMRB, Univ Paris Est Creteil, Creteil, France.,Departement d'Hematologie-Immunologie, AP-HP, Hopital Henri Mondor, Créteil, France
| |
Collapse
|
18
|
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: 4.3] [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
|
19
|
Puiffe ML, Dupont A, Sako N, Gatineau J, Cohen JL, Mestivier D, Lebon A, Prévost-Blondel A, Castellano F, Molinier-Frenkel V. IL4I1 Accelerates the Expansion of Effector CD8 + T Cells at the Expense of Memory Precursors by Increasing the Threshold of T-Cell Activation. Front Immunol 2020; 11:600012. [PMID: 33343572 PMCID: PMC7746639 DOI: 10.3389/fimmu.2020.600012] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Accepted: 11/03/2020] [Indexed: 12/25/2022] Open
Abstract
IL4I1 is an immunoregulatory enzyme that inhibits CD8 T-cell proliferation in vitro and in the tumoral context. Here, we dissected the effect of IL4I1 on CD8 T-cell priming by studying the differentiation of a transgenic CD8 T-cell clone and the endogenous repertoire in a mouse model of acute lymphocytic choriomeningitis virus (LCMV) infection. Unexpectedly, we show that IL4I1 accelerates the expansion of functional effector CD8 T cells during the first several days after infection and increases the average affinity of the elicited repertoire, supporting more efficient LCMV clearance in WT mice than IL4I1-deficient mice. Conversely, IL4I1 restrains the differentiation of CD8 T-cells into long-lived memory precursors and favors the memory response to the most immunodominant peptides. IL4I1 expression does not affect the phenotype or antigen-presenting functions of dendritic cells (DCs), but directly reduces the stability of T-DC immune synapses in vitro, thus dampening T-cell activation. Overall, our results support a model in which IL4I1 increases the threshold of T-cell activation, indirectly promoting the priming of high-affinity clones while limiting memory T-cell differentiation.
Collapse
Affiliation(s)
- Marie-Line Puiffe
- Virus-Immunity-Cancer Department, Institut Mondor de Recherche Biomédicale (IMRB), INSERM U955, Université Paris-Est Créteil, Créteil, France
| | - Aurélie Dupont
- Virus-Immunity-Cancer Department, Institut Mondor de Recherche Biomédicale (IMRB), INSERM U955, Université Paris-Est Créteil, Créteil, France
| | - Nouhoum Sako
- Virus-Immunity-Cancer Department, Institut Mondor de Recherche Biomédicale (IMRB), INSERM U955, Université Paris-Est Créteil, Créteil, France
| | - Jérôme Gatineau
- Virus-Immunity-Cancer Department, Institut Mondor de Recherche Biomédicale (IMRB), INSERM U955, Université Paris-Est Créteil, Créteil, France
| | - José L Cohen
- Virus-Immunity-Cancer Department, Institut Mondor de Recherche Biomédicale (IMRB), INSERM U955, Université Paris-Est Créteil, Créteil, France
| | - Denis Mestivier
- Bioinformatics Core Laboratory, Institut Mondor de Recherche Biomédicale (IMRB), INSERM U955, Université Paris-Est Créteil, Créteil, France
| | - Agnès Lebon
- INSERM U1016, CNRS UMR8104, Institut Cochin, Université de Paris, Paris, France
| | | | - Flavia Castellano
- Virus-Immunity-Cancer Department, Institut Mondor de Recherche Biomédicale (IMRB), INSERM U955, Université Paris-Est Créteil, Créteil, France.,Pathobiology Department, Groupe Hospitalo-Universitaire Chenevier-Mondor, AP-HP, Créteil, France
| | - Valérie Molinier-Frenkel
- Virus-Immunity-Cancer Department, Institut Mondor de Recherche Biomédicale (IMRB), INSERM U955, Université Paris-Est Créteil, Créteil, France.,Pathobiology Department, Groupe Hospitalo-Universitaire Chenevier-Mondor, AP-HP, Créteil, France
| |
Collapse
|
20
|
Kitani Y, Nagashima Y. l-Amino acid oxidase as a fish host-defense molecule. FISH & SHELLFISH IMMUNOLOGY 2020; 106:685-690. [PMID: 32822860 DOI: 10.1016/j.fsi.2020.08.028] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/10/2020] [Revised: 08/15/2020] [Accepted: 08/17/2020] [Indexed: 06/11/2023]
Abstract
An l-amino acid oxidase (LAO) is an amino acid metabolism enzyme that also performs a variety of biological activities. Recently, LAOs have been discovered to be deeply involved in innate immunity in fish because of their antibacterial and antiparasitic activity. The determinant of potent antibacterial/antiparasitic activity is the H2O2 byproduct of LAO enzymatic activity that utilizes the l-amino acid as a substrate. In addition, fish LAOs are upregulated by pathogenic bacteria or parasite infection. Furthermore, some fish LAOs show that the target specificity depends on the virulence of the bacteria. All results reflect that LAOs are new innate immune molecules. This review also describes the potential of the immunomodulatory functions of fish LAOs, not only the innate immune function by a direct oxidation attack of H2O2.
Collapse
Affiliation(s)
- Yoichiro Kitani
- Noto Marine Laboratory, Institute of Nature and Environmental Technology, Kanazawa University, Ogi Mu 4-1 Noto-Cho, Ishikawa, 927-0553, Japan.
| | - Yuji Nagashima
- Department of Agro-Food Science, Niigata Agro-Food University, Hirakidai 2416, Tainai, Niigata, 995-2702, Japan
| |
Collapse
|
21
|
IL4I1 Is a Metabolic Immune Checkpoint that Activates the AHR and Promotes Tumor Progression. Cell 2020; 182:1252-1270.e34. [PMID: 32818467 DOI: 10.1016/j.cell.2020.07.038] [Citation(s) in RCA: 279] [Impact Index Per Article: 69.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Revised: 05/25/2020] [Accepted: 07/28/2020] [Indexed: 01/01/2023]
Abstract
Aryl hydrocarbon receptor (AHR) activation by tryptophan (Trp) catabolites enhances tumor malignancy and suppresses anti-tumor immunity. The context specificity of AHR target genes has so far impeded systematic investigation of AHR activity and its upstream enzymes across human cancers. A pan-tissue AHR signature, derived by natural language processing, revealed that across 32 tumor entities, interleukin-4-induced-1 (IL4I1) associates more frequently with AHR activity than IDO1 or TDO2, hitherto recognized as the main Trp-catabolic enzymes. IL4I1 activates the AHR through the generation of indole metabolites and kynurenic acid. It associates with reduced survival in glioma patients, promotes cancer cell motility, and suppresses adaptive immunity, thereby enhancing the progression of chronic lymphocytic leukemia (CLL) in mice. Immune checkpoint blockade (ICB) induces IDO1 and IL4I1. As IDO1 inhibitors do not block IL4I1, IL4I1 may explain the failure of clinical studies combining ICB with IDO1 inhibition. Taken together, IL4I1 blockade opens new avenues for cancer therapy.
Collapse
|
22
|
Elsheimer-Matulova M, Polansky O, Seidlerova Z, Varmuzova K, Stepanova H, Fedr R, Rychlik I. Interleukin 4 inducible 1 gene (IL4I1) is induced in chicken phagocytes by Salmonella Enteritidis infection. Vet Res 2020; 51:67. [PMID: 32404145 PMCID: PMC7222322 DOI: 10.1186/s13567-020-00792-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2019] [Accepted: 04/29/2020] [Indexed: 12/18/2022] Open
Abstract
In attempt to identify genes that are induced in chickens by Salmonella Enteritidis we identified a new highly inducible gene, interleukin 4 induced 1 gene (IL4I1). IL4I1 reached its peak expression (458× induction) in the cecum of newly hatched chickens 4 days post-infection and remained upregulated for an additional 10 days. IL4I1 was expressed and induced in macrophages and granulocytes, both at the mRNA and protein level. IL4I1 was expressed and induced also in CD4 and γδ T-lymphocytes though at a 50-fold lower level than in phagocytes. Expression of IL4I1 was not detected in CD8 T lymphocytes or B lymphocytes. Mutation of IL4I1 in chicken HD11 macrophages did not affect their bactericidal capacity against S. Enteritidis but negatively affected their oxidative burst after PMA stimulation. We therefore propose that IL4I1 is not directly involved in bactericidal activity of phagocytes and, instead, it is likely involved in the control of inflammatory response and signaling to T and B lymphocytes.
Collapse
Affiliation(s)
| | - Ondrej Polansky
- Veterinary Research Institute, Hudcova 70, 621 00, Brno, Czech Republic
| | - Zuzana Seidlerova
- Veterinary Research Institute, Hudcova 70, 621 00, Brno, Czech Republic
| | | | - Hana Stepanova
- Veterinary Research Institute, Hudcova 70, 621 00, Brno, Czech Republic
| | - Radek Fedr
- Institute of Biophysics, Academy of Sciences of the Czech Republic, Kralovopolska 135, 612 65, Brno, Czech Republic
| | - Ivan Rychlik
- Veterinary Research Institute, Hudcova 70, 621 00, Brno, Czech Republic.
| |
Collapse
|
23
|
Lavalett L, Ortega H, Barrera LF. Human Alveolar and Splenic Macrophage Populations Display a Distinct Transcriptomic Response to Infection With Mycobacterium tuberculosis. Front Immunol 2020; 11:630. [PMID: 32373118 PMCID: PMC7186480 DOI: 10.3389/fimmu.2020.00630] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Accepted: 03/19/2020] [Indexed: 12/14/2022] Open
Abstract
Mycobacterium tuberculosis (Mtb) infects alveolar macrophages (AMs), causing pulmonary tuberculosis (PTB), the most common form of the disease. Less frequently, Mtb is disseminated to many other organs and tissues, resulting in different extrapulmonary forms of TB. Nevertheless, very few studies have addressed the global mRNA response of human AMs, particularly from humans with the active form of the disease. Strikingly, almost no studies have addressed the response of human extrapulmonary macrophages to Mtb infection. In this pilot study, using microarray technology, we examined the transcriptomic ex vivo response of AMs from PTB patients (AMTBs) and AMs from control subjects (AMCTs) infected with two clinical isolates of Mtb. Furthermore, we also studied the infection response of human splenic macrophages (SMs) to Mtb isolates, as a model for extrapulmonary infection, and compared the transcriptomic response between AMs and SMs. Our results showed a striking difference in global mRNA profiles in response to infection between AMs and SMs, implicating a tissue-specific macrophage response to Mtb.
Collapse
Affiliation(s)
- Lelia Lavalett
- Grupo de Inmunología Celular e Inmunogenética, Facultad de Medicina, Instituto de Investigaciones Médicas, Universidad de Antioquia, Medellín, Colombia.,Facultad de Ciencias, Universidad Nacional de Colombia Sede Medellín, Medellín, Colombia
| | - Hector Ortega
- Clínica Cardiovascular Santa María, Medellín, Colombia.,Facultad de Medicina, Universidad de Antioquia, Medellín, Colombia
| | - Luis F Barrera
- Grupo de Inmunología Celular e Inmunogenética, Facultad de Medicina, Instituto de Investigaciones Médicas, Universidad de Antioquia, Medellín, Colombia.,Facultad de Medicina, Universidad de Antioquia, Medellín, Colombia
| |
Collapse
|
24
|
Wu SZ, Wei HX, Jiang D, Li SM, Zou WH, Peng HJ. Genome-Wide CRISPR Screen Identifies Host Factors Required by Toxoplasma gondii Infection. Front Cell Infect Microbiol 2020; 9:460. [PMID: 32039045 PMCID: PMC6987080 DOI: 10.3389/fcimb.2019.00460] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Accepted: 12/16/2019] [Indexed: 11/18/2022] Open
Abstract
Toxoplasma gondii are obligate intracellular protoza, and due to their small genome and limited encoded proteins, they have to exploit host factors for entry, replication, and dissemination. Such host factors can be defined as host dependency factors (HDFs). Though HDFs are inessential for cell viability, they are critical for pathogen infection, and potential ideal targets for therapeutic intervention. However, information about these HDFs required by T. gondii infection is highly deficient. In this study, the genes of human foreskin fibroblast (HFF) cells were comprehensively edited using the lentiviral CRISPR-Cas9-sgRNA library, and then the lentivirus-treated cells were infected with T. gondii at multiplication of infection 1 (MOI = 1) for 10 days to identify HDFs essential for T. gondii infection. The survival cells were harvested and sent for sgRNA sequencing. The sgRNA sequence matched genes or miRNAs were potential HDFs. Some cells in the lentivirus-treated group could survive longer than those in the untreated control group after T. gondii infection. From a pool of 19,050 human genes and 1,864 human pri-miRNAs, 1,193 potential HDFs were identified, including 1,183 genes and 10 pri-miRNAs (corresponding with 17 mature miRNAs). Among them, seven genes and five mature miRNAs were validated with siRNAs, miRNA inhibitors, and mimics, respectively. Bioinformatics analysis revealed that, among the 1,183 genes, 53 potential HDFs were associated with regulation of host actin cytoskeleton and 23 potential HDFs coded immune negative regulators. This result indicated that actin dynamics were indispensable for T. gondii infection, and some host immune negative regulators may be involved in disarming host defenses. Our findings contribute to the current limited knowledge about host factors required by T. gondii infection and provide us with new targets for medication therapy and vaccine exploitation.
Collapse
Affiliation(s)
- Shui-Zhen Wu
- Guangdong Provincial Key Laboratory of Tropical Disease Research, Department of Pathogen Biology, School of Public Health, Southern Medical University, Guangzhou, China
| | - Hai-Xia Wei
- Guangdong Provincial Key Laboratory of Tropical Disease Research, Department of Pathogen Biology, School of Public Health, Southern Medical University, Guangzhou, China
| | - Dan Jiang
- Guangdong Provincial Key Laboratory of Tropical Disease Research, Department of Pathogen Biology, School of Public Health, Southern Medical University, Guangzhou, China
| | - Sheng-Min Li
- Guangdong Provincial Key Laboratory of Tropical Disease Research, Department of Pathogen Biology, School of Public Health, Southern Medical University, Guangzhou, China
| | - Wei-Hao Zou
- Guangdong Provincial Key Laboratory of Tropical Disease Research, Department of Pathogen Biology, School of Public Health, Southern Medical University, Guangzhou, China
| | - Hong-Juan Peng
- Guangdong Provincial Key Laboratory of Tropical Disease Research, Department of Pathogen Biology, School of Public Health, Southern Medical University, Guangzhou, China
| |
Collapse
|
25
|
Presset M, Djordjevic D, Dupont A, Le Gall E, Molinier-Frenkel V, Castellano F. Identification of inhibitors of the immunosuppressive enzyme IL4I1. Bioorg Chem 2019; 94:103463. [PMID: 31812258 DOI: 10.1016/j.bioorg.2019.103463] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2019] [Revised: 11/19/2019] [Accepted: 11/20/2019] [Indexed: 10/25/2022]
Affiliation(s)
- Marc Presset
- Electrochimie et Synthèse Organique, Université Paris Est, ICMPE (UMR 7182), CNRS, UPEC, 2 rue Henri Dunant, F-94320 Thiais, France
| | - Diana Djordjevic
- INSERM, U955, Equipe 09, Créteil, France; Université Paris Est, Faculté de Médecine, Créteil, France
| | - Aurélie Dupont
- INSERM, U955, Equipe 09, Créteil, France; Université Paris Est, Faculté de Médecine, Créteil, France
| | - Erwan Le Gall
- Electrochimie et Synthèse Organique, Université Paris Est, ICMPE (UMR 7182), CNRS, UPEC, 2 rue Henri Dunant, F-94320 Thiais, France
| | - Valérie Molinier-Frenkel
- INSERM, U955, Equipe 09, Créteil, France; Université Paris Est, Faculté de Médecine, Créteil, France; AP-HP, Hôpital H. Mondor - A. Chenevier, Laboratoire d'Immunologie, Créteil, France.
| | - Flavia Castellano
- INSERM, U955, Equipe 09, Créteil, France; Université Paris Est, Faculté de Médecine, Créteil, France.
| |
Collapse
|
26
|
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: 52] [Impact Index Per Article: 10.4] [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
|
27
|
Ramspott JP, Bekkat F, Bod L, Favier M, Terris B, Salomon A, Djerroudi L, Zaenker KS, Richard Y, Molinier-Frenkel V, Castellano F, Avril MF, Prévost-Blondel A. Emerging Role of IL-4–Induced Gene 1 as a Prognostic Biomarker Affecting the Local T-Cell Response in Human Cutaneous Melanoma. J Invest Dermatol 2018; 138:2625-2634. [DOI: 10.1016/j.jid.2018.06.178] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2018] [Revised: 06/04/2018] [Accepted: 06/18/2018] [Indexed: 12/28/2022]
|
28
|
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: 1.7] [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
|
29
|
Szołtysek K, Janus P, Zając G, Stokowy T, Walaszczyk A, Widłak W, Wojtaś B, Gielniewski B, Cockell S, Perkins ND, Kimmel M, Widlak P. RRAD, IL4I1, CDKN1A, and SERPINE1 genes are potentially co-regulated by NF-κB and p53 transcription factors in cells exposed to high doses of ionizing radiation. BMC Genomics 2018; 19:813. [PMID: 30419821 PMCID: PMC6233266 DOI: 10.1186/s12864-018-5211-y] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2018] [Accepted: 10/30/2018] [Indexed: 01/24/2023] Open
Abstract
BACKGROUND The cellular response to ionizing radiation involves activation of p53-dependent pathways and activation of the atypical NF-κB pathway. The crosstalk between these two transcriptional networks include (co)regulation of common gene targets. Here we looked for novel genes potentially (co)regulated by p53 and NF-κB using integrative genomics screening in human osteosarcoma U2-OS cells irradiated with a high dose (4 and 10 Gy). Radiation-induced expression in cells with silenced TP53 or RELA (coding the p65 NF-κB subunit) genes was analyzed by RNA-Seq while radiation-enhanced binding of p53 and RelA in putative regulatory regions was analyzed by ChIP-Seq, then selected candidates were validated by qPCR. RESULTS We identified a subset of radiation-modulated genes whose expression was affected by silencing of both TP53 and RELA, and a subset of radiation-upregulated genes where radiation stimulated binding of both p53 and RelA. For three genes, namely IL4I1, SERPINE1, and CDKN1A, an antagonistic effect of the TP53 and RELA silencing was consistent with radiation-enhanced binding of both p53 and RelA. This suggested the possibility of a direct antagonistic (co)regulation by both factors: activation by NF-κB and inhibition by p53 of IL4I1, and activation by p53 and inhibition by NF-κB of CDKN1A and SERPINE1. On the other hand, radiation-enhanced binding of both p53 and RelA was observed in a putative regulatory region of the RRAD gene whose expression was downregulated both by TP53 and RELA silencing, which suggested a possibility of direct (co)activation by both factors. CONCLUSIONS Four new candidates for genes directly co-regulated by NF-κB and p53 were revealed.
Collapse
Affiliation(s)
- Katarzyna Szołtysek
- Maria Skłodowska-Curie Institute – Oncology Center, Gliwice Branch, Gliwice, Poland
| | - Patryk Janus
- Maria Skłodowska-Curie Institute – Oncology Center, Gliwice Branch, Gliwice, Poland
| | - Gracjana Zając
- Maria Skłodowska-Curie Institute – Oncology Center, Gliwice Branch, Gliwice, Poland
| | - Tomasz Stokowy
- Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Anna Walaszczyk
- Maria Skłodowska-Curie Institute – Oncology Center, Gliwice Branch, Gliwice, Poland
| | - Wiesława Widłak
- Maria Skłodowska-Curie Institute – Oncology Center, Gliwice Branch, Gliwice, Poland
| | - Bartosz Wojtaś
- Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warszawa, Poland
| | | | - Simon Cockell
- Faculty of Medical Sciences, Newcastle University, Newcastle, UK
| | - Neil D. Perkins
- Institute for Cell and Molecular Biosciences, Newcastle University, Newcastle, UK
| | | | - Piotr Widlak
- Maria Skłodowska-Curie Institute – Oncology Center, Gliwice Branch, Gliwice, Poland
| |
Collapse
|
30
|
Janus P, Szołtysek K, Zając G, Stokowy T, Walaszczyk A, Widłak W, Wojtaś B, Gielniewski B, Iwanaszko M, Braun R, Cockell S, Perkins ND, Kimmel M, Widlak P. Pro-inflammatory cytokine and high doses of ionizing radiation have similar effects on the expression of NF-kappaB-dependent genes. Cell Signal 2018; 46:23-31. [PMID: 29476964 DOI: 10.1016/j.cellsig.2018.02.011] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2017] [Revised: 02/19/2018] [Accepted: 02/20/2018] [Indexed: 12/22/2022]
Abstract
The NF-κB transcription factors are activated via diverse molecular mechanisms in response to various types of stimuli. A plethora of functions associated with specific sets of target genes could be regulated differentially by this factor, affecting cellular response to stress including an anticancer treatment. Here we aimed to compare subsets of NF-κB-dependent genes induced in cells stimulated with a pro-inflammatory cytokine and in cells damaged by a high dose of ionizing radiation (4 and 10 Gy). The RelA-containing NF-κB species were activated by the canonical TNFα-induced and the atypical radiation-induced pathways in human osteosarcoma cells. NF-κB-dependent genes were identified using the gene expression profiling (by RNA-Seq) in cells with downregulated RELA combined with the global profiling of RelA binding sites (by ChIP-Seq), with subsequent validation of selected candidates by quantitative PCR. There were 37 NF-κB-dependent protein-coding genes identified: in all cases RelA bound in their regulatory regions upon activation while downregulation of RELA suppressed their stimulus-induced upregulation, which apparently indicated the positive regulation mode. This set of genes included a few "novel" NF-κB-dependent species. Moreover, the evidence for possible negative regulation of ATF3 gene by NF-κB was collected. The kinetics of the NF-κB activation was slower in cells exposed to radiation than in cytokine-stimulated ones. However, subsets of NF-κB-dependent genes upregulated by both types of stimuli were essentially the same. Hence, one should expect that similar cellular processes resulting from activation of the NF-κB pathway could be induced in cells responding to pro-inflammatory cytokines and in cells where so-called "sterile inflammation" response was initiated by radiation-induced damage.
Collapse
Affiliation(s)
- Patryk Janus
- Maria Skłodowska-Curie Institute, Oncology Center, Gliwice Branch, Gliwice, Poland
| | - Katarzyna Szołtysek
- Maria Skłodowska-Curie Institute, Oncology Center, Gliwice Branch, Gliwice, Poland
| | - Gracjana Zając
- Maria Skłodowska-Curie Institute, Oncology Center, Gliwice Branch, Gliwice, Poland
| | - Tomasz Stokowy
- Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Anna Walaszczyk
- Maria Skłodowska-Curie Institute, Oncology Center, Gliwice Branch, Gliwice, Poland
| | - Wiesława Widłak
- Maria Skłodowska-Curie Institute, Oncology Center, Gliwice Branch, Gliwice, Poland
| | - Bartosz Wojtaś
- Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warszawa, Poland
| | | | - Marta Iwanaszko
- Feinberg School of Medicine, Northwestern University, Chicago, USA
| | - Rosemary Braun
- Feinberg School of Medicine, Northwestern University, Chicago, USA
| | - Simon Cockell
- Faculty of Medical Sciences, Newcastle University, Newcastle, UK
| | - Neil D Perkins
- Institute for Cell and Molecular Biosciences, Newcastle University, Newcastle, UK
| | | | - Piotr Widlak
- Maria Skłodowska-Curie Institute, Oncology Center, Gliwice Branch, Gliwice, Poland.
| |
Collapse
|
31
|
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: 3.7] [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
|
32
|
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: 4.0] [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
|
33
|
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: 2.4] [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
|
34
|
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: 2.0] [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
|
35
|
Molinier-Frenkel V, Castellano F. Immunosuppressive enzymes in the tumor microenvironment. FEBS Lett 2017; 591:3135-3157. [DOI: 10.1002/1873-3468.12784] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2017] [Revised: 07/31/2017] [Accepted: 08/03/2017] [Indexed: 12/22/2022]
Affiliation(s)
- Valérie Molinier-Frenkel
- INSERM, U955, Equipe 09; Créteil France
- Faculté de Médecine; Université Paris Est; Créteil France
- Service d'Immunologie Biologique; AP-HP, Hôpital H. Mondor - A. Chenevier; Créteil France
| | - Flavia Castellano
- INSERM, U955, Equipe 09; Créteil France
- Faculté de Médecine; Université Paris Est; Créteil France
- Plateforme de Ressources Biologiques; AP-HP, Hôpital H. Mondor - A. Chenevier; Créteil France
| |
Collapse
|
36
|
Bod L, Lengagne R, Wrobel L, Ramspott JP, Kato M, Avril MF, Castellano F, Molinier-Frenkel V, Prévost-Blondel A. IL4-induced gene 1 promotes tumor growth by shaping the immune microenvironment in melanoma. Oncoimmunology 2017; 6:e1278331. [PMID: 28405502 DOI: 10.1080/2162402x.2016.1278331] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2016] [Revised: 12/21/2016] [Accepted: 12/28/2016] [Indexed: 12/11/2022] Open
Abstract
Amino acid catabolizing enzymes emerged as a crucial mechanism used by tumors to dampen immune responses. The L-phenylalanine oxidase IL-4 induced gene 1 (IL4I1) is expressed by tumor-associated myeloid cells of most solid tumors, including melanoma. We previously provided the only evidence that IL4I1 accelerates tumor growth by limiting the CD8+ T cell mediated immune response, in a mouse model of melanoma cell transplantation. Here, we explored the role of IL4I1 in Ret mice, a spontaneous model of melanoma. We found that IL4I1 was expressed by CD11b+ myeloid cells and that its activity correlated with disease aggressiveness. IL4I1 did not enhance tumor cell proliferation or angiogenesis, but orchestrated the remodeling of the immune compartment within the primary tumor. Indeed, the inactivation of IL4I1 limited the recruitment of polymorphonuclear myeloid-derived suppressor cells and enhanced the infiltration by Th1 and cytotoxic T cells, thus delaying tumor development and metastatic dissemination. Accordingly, human primary melanomas that were poorly infiltrated by IL4I1+ cells exhibited a higher density of CD8+ T cells. Collectively, our findings strengthen the rationale for therapeutic targeting of IL4I1 as one of the key immune regulators.
Collapse
Affiliation(s)
- Lloyd Bod
- Inserm, U1016, Institut Cochin, Paris, France; Cnrs, UMR8104, Paris, France; Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Renée Lengagne
- Inserm, U1016, Institut Cochin, Paris, France; Cnrs, UMR8104, Paris, France; Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | | | - Jan Philipp Ramspott
- Inserm, U1016, Institut Cochin, Paris, France; Cnrs, UMR8104, Paris, France; Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Masashi Kato
- Nagoya University Graduate School of Medicine , Nagoya, Aichi, Japan
| | - Marie-Françoise Avril
- Inserm, U1016, Institut Cochin, Paris, France; Cnrs, UMR8104, Paris, France; Université Paris Descartes, Sorbonne Paris Cité, Paris, France; APHP, Hôpital Cochin, Paris, France
| | - Flavia Castellano
- INSERM, U955, Equipe 09, Créteil, France; Université Paris Est, Faculté de Médecine, Créteil, France; AP-HP, Hôpital H. Mondor - A. Chenevier, Plateforme de Ressources Biologiques, Créteil, France
| | - Valérie Molinier-Frenkel
- INSERM, U955, Equipe 09, Créteil, France; Université Paris Est, Faculté de Médecine, Créteil, France; AP-HP, Hôpital H. Mondor - A. Chenevier, Service d'Immunologie Biologique, Créteil, France
| | - Armelle Prévost-Blondel
- Inserm, U1016, Institut Cochin, Paris, France; Cnrs, UMR8104, Paris, France; Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| |
Collapse
|
37
|
Reales-Calderón JA, Vaz C, Monteoliva L, Molero G, Gil C. Candida albicans Modifies the Protein Composition and Size Distribution of THP-1 Macrophage-Derived Extracellular Vesicles. J Proteome Res 2016; 16:87-105. [PMID: 27740763 DOI: 10.1021/acs.jproteome.6b00605] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The effectiveness of macrophages in the response to systemic candidiasis is crucial to an effective clearance of the pathogen. The secretion of proteins, mRNAs, noncoding RNAs and lipids through extracellular vesicles (EVs) is one of the mechanisms of communication between immune cells. EVs change their cargo to mediate different responses, and may play a role in the response against infections. Thus we have undertaken the first quantitative proteomic analysis on the protein composition of THP-1 macrophage-derived EVs during the interaction with Candida albicans. This study revealed changes in EVs sizes and in protein composition, and allowed the identification and quantification of 717 proteins. Of them, 133 proteins changed their abundance due to the interaction. The differentially abundant proteins were involved in functions relating to immune response, signaling, or cytoskeletal reorganization. THP-1-derived EVs, both from control and from Candida-infected macrophages, had similar effector functions on other THP-1-differenciated macrophages, activating ERK and p38 kinases, and increasing both the secretion of proinflammatory cytokines and the candidacidal activity; while in THP-1 nondifferenciated monocytes, only EVs from infected macrophages increased significantly the TNF-α secretion. Our findings provide new information on the role of macrophage-derived EVs in response to C. albicans infection and in macrophages communication.
Collapse
Affiliation(s)
- Jose Antonio Reales-Calderón
- Departamento de Microbiología II, Facultad de Farmacia, Universidad Complutense de Madrid Plaza de Ramón y Cajal s/n , Madrid 28040, Spain.,Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS) , Madrid 28034, Spain
| | - Catarina Vaz
- Departamento de Microbiología II, Facultad de Farmacia, Universidad Complutense de Madrid Plaza de Ramón y Cajal s/n , Madrid 28040, Spain.,Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS) , Madrid 28034, Spain
| | - Lucía Monteoliva
- Departamento de Microbiología II, Facultad de Farmacia, Universidad Complutense de Madrid Plaza de Ramón y Cajal s/n , Madrid 28040, Spain.,Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS) , Madrid 28034, Spain
| | - Gloria Molero
- Departamento de Microbiología II, Facultad de Farmacia, Universidad Complutense de Madrid Plaza de Ramón y Cajal s/n , Madrid 28040, Spain.,Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS) , Madrid 28034, Spain
| | - Concha Gil
- Departamento de Microbiología II, Facultad de Farmacia, Universidad Complutense de Madrid Plaza de Ramón y Cajal s/n , Madrid 28040, Spain.,Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS) , Madrid 28034, Spain
| |
Collapse
|
38
|
IL4I1 Is a Novel Regulator of M2 Macrophage Polarization That Can Inhibit T Cell Activation via L-Tryptophan and Arginine Depletion and IL-10 Production. PLoS One 2015; 10:e0142979. [PMID: 26599209 PMCID: PMC4658051 DOI: 10.1371/journal.pone.0142979] [Citation(s) in RCA: 83] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2015] [Accepted: 10/29/2015] [Indexed: 11/19/2022] Open
Abstract
Interleukin 4-induced gene-1 (IL4I1) was initially described as an early IL-4-inducible gene in B cells. IL4I1 protein can inhibit T cell proliferation by releasing its enzymatic catabolite, H2O2, and this effect is associated with transient down-regulation of T cell CD3 receptor-zeta (TCRζ) expression. Herein, we show that IL4I1 contributes to the regulation of macrophage programming. We found that expression of IL4I1 increased during bone marrow-derived macrophage (BMDM) differentiation, expression of IL4I1 is much higher in primary macrophages than monocytes, and IL4I1 expression in BMDMs could be induced by Th1 and Th2 cytokines in two different patterns. Gene expression analysis revealed that overexpression of IL4I1 drove the expression of M2 markers (Fizz1, Arg1, YM-1, MR) and inhibited the expression of M1-associated cytokines. Conversely, knockdown of IL4I1 by siRNA resulted in opposite effects, and also attenuated STAT-3 and STAT-6 phosphorylation. Furthermore, IL4I1 produced by macrophages catalyzed L-tryptophan degradation, while levo-1-methyl-tryptophan (L-1-MT), but not dextro-1-methyl-tryptophan, partially rescued IL4I1-dependent inhibition of T cell activation. Other inhibitors, such as diphenylene iodonium (DPI), an anti-IL-10Rα blocking antibody, and a nitric oxide synthase inhibitor, NG-monomethyl-L-arginine, also had this effect. Overall, our findings indicate that IL4I1 promotes an enhanced M2 functional phenotype, which is most likely associated with the phosphorylation of STAT-6 and STAT-3. Moreover, DPI, L-1-MT, NG-monomethyl-L-arginine, and anti-IL-10Rα blocking antibody were all found to be effective IL4I1 inhibitors in vitro.
Collapse
|
39
|
Hare NJ, Chan B, Chan E, Kaufman KL, Britton WJ, Saunders BM. Microparticles released from Mycobacterium tuberculosis-infected human macrophages contain increased levels of the type I interferon inducible proteins including ISG15. Proteomics 2015; 15:3020-9. [PMID: 26036210 DOI: 10.1002/pmic.201400610] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2014] [Revised: 03/26/2015] [Accepted: 05/30/2015] [Indexed: 01/18/2023]
Abstract
Microparticles (MPs) are small membranous particles (100-1000 nm) released under normal steady-state conditions and are thought to provide a communication network between host cells. Previous studies demonstrated that Mycobacterium tuberculosis (M. tb) infection of macrophages increased the release of MPs, and these MPs induced a proinflammatory response from uninfected macrophages in vitro and in vivo following their transfer into uninfected mice. To determine how M. tb infection modulates the protein composition of the MPs, and if this contributes to their proinflammatory properties, we compared the proteomes of MPs derived from M. tb-infected (TBinf-MP) and uninfected human THP-1 monocytic cells. MP proteins were analyzed by GeLC-MS/MS with spectral counting revealing 68 proteins with statistically significant differential abundances. The 42 proteins increased in abundance in TBinf-MPs included proteins associated with immune function (7), lysosomal/endosomal maturation (4), vesicular formation (12), nucleosome proteins (4), and antigen processing (9). Prominent among these were the type I interferon inducible proteins, ISG15, IFIT1, IFIT2, and IFIT3. Exposure of uninfected THP-1 cells to TBinf-MPs induced increased gene expression of isg15, ifit1, ifit2, and ifit3 and the release of proinflammatory cytokines. These proteins may regulate the proinflammatory potential of the MPs and provide candidate biomarkers for M. tb infection.
Collapse
Affiliation(s)
- Nathan J Hare
- Tuberculosis Research Program, Centenary Institute, Disciplines of Medicine, Infectious Diseases and Immunology, Sydney Medical School, The University of Sydney, Newtown, NSW, Australia
| | - Brian Chan
- Tuberculosis Research Program, Centenary Institute, Disciplines of Medicine, Infectious Diseases and Immunology, Sydney Medical School, The University of Sydney, Newtown, NSW, Australia
| | - Edwina Chan
- Tuberculosis Research Program, Centenary Institute, Disciplines of Medicine, Infectious Diseases and Immunology, Sydney Medical School, The University of Sydney, Newtown, NSW, Australia
| | - Kimberley L Kaufman
- School of Molecular Bioscience, The University of Sydney, Sydney, NSW, Australia
| | - Warwick J Britton
- Tuberculosis Research Program, Centenary Institute, Disciplines of Medicine, Infectious Diseases and Immunology, Sydney Medical School, The University of Sydney, Newtown, NSW, Australia
| | - Bernadette M Saunders
- Tuberculosis Research Program, Centenary Institute, Disciplines of Medicine, Infectious Diseases and Immunology, Sydney Medical School, The University of Sydney, Newtown, NSW, Australia.,School of Life Sciences, University of Technology, Sydney, NSW, Australia
| |
Collapse
|
40
|
Cousin C, Aubatin A, Le Gouvello S, Apetoh L, Castellano F, Molinier-Frenkel V. The immunosuppressive enzyme IL4I1 promotes FoxP3(+) regulatory T lymphocyte differentiation. Eur J Immunol 2015; 45:1772-82. [PMID: 25778793 DOI: 10.1002/eji.201445000] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2014] [Revised: 01/30/2015] [Accepted: 03/12/2015] [Indexed: 01/29/2023]
Abstract
IL4I1 (interleukin-4-induced gene 1) is a phenylalanine oxidase produced mainly by APCs of myeloid origin, and converts phenylalanine (Phe) to phenylpyruvate, hydrogen peroxide, and ammonia. We have previously shown that IL4I1 is highly expressed by tumor-associated macrophages from various human cancers and facilitates immune evasion from the cytotoxic response in a murine tumor model. Indeed, IL4I1 inhibits T-cell proliferation via hydrogen peroxide toxicity on effector/memory T cells. Here, we explored the effect of IL4I1 on naïve CD4(+) T-cell differentiation. We show that IL4I1 stimulates the generation of Foxp3(+) regulatory T (Treg) cells in vitro from human and mouse T cells. This effect was observed with IL4I1 from different sources, including the naturally produced enzyme. Conversely, IL4I1 limits Th1 and Th2 polarization while modifying the Th17 phenotype, in particular, by inducing its own production. Analysis of Treg-cell induction under conditions of Phe deprivation and hydrogen peroxide addition suggests that Phe consumption by the enzyme participates in Treg-cell enrichment. In line with this hypothesis, IL4I1 inhibits mTORC1 signaling shortly after T-cell activation. Thus, the IL4I1 enzyme may act on T cells both by direct inhibition of effector cell proliferation and by indirect immunoregulation mediated by Treg-cell induction.
Collapse
Affiliation(s)
- Céline Cousin
- INSERM, U955, Equipe 09 and Equipe 04, Créteil, France.,Université Paris Est, Faculté de médecine, Créteil, France
| | - Aude Aubatin
- INSERM, U955, Equipe 09 and Equipe 04, Créteil, France.,Université Paris Est, Faculté de médecine, Créteil, France
| | - Sabine Le Gouvello
- INSERM, U955, Equipe 09 and Equipe 04, Créteil, France.,Université Paris Est, Faculté de médecine, Créteil, France.,AP-HP, Hôpital H. Mondor - A. Chenevier, Service d'Immunologie Biologique, Créteil, France
| | - Lionel Apetoh
- INSERM, U866, Dijon, France.,Centre Georges François Leclerc, Dijon, France.,Université de Bourgogne, Dijon, France
| | - Flavia Castellano
- INSERM, U955, Equipe 09 and Equipe 04, Créteil, France.,Université Paris Est, Faculté de médecine, Créteil, France.,AP-HP, Hôpital H. Mondor - A. Chenevier, Service d'Immunologie Biologique, Créteil, France
| | - Valérie Molinier-Frenkel
- INSERM, U955, Equipe 09 and Equipe 04, Créteil, France.,Université Paris Est, Faculté de médecine, Créteil, France.,AP-HP, Hôpital H. Mondor - A. Chenevier, Service d'Immunologie Biologique, Créteil, France
| |
Collapse
|
41
|
Dutta NK, Illei PB, Jain SK, Karakousis PC. Characterization of a novel necrotic granuloma model of latent tuberculosis infection and reactivation in mice. THE AMERICAN JOURNAL OF PATHOLOGY 2014; 184:2045-55. [PMID: 24815353 DOI: 10.1016/j.ajpath.2014.03.008] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/07/2014] [Revised: 02/20/2014] [Accepted: 03/18/2014] [Indexed: 01/09/2023]
Abstract
We sought to develop and characterize a novel paucibacillary model in mice, which develops necrotic lung granulomas after infection with Mycobacterium tuberculosis. Six weeks after aerosol immunization with recombinant Mycobacterium bovis bacillus Calmette-Guerin overexpressing the 30-kDa antigen, C3HeB/FeJ mice were aerosol infected with M. tuberculosis H37Rv. Six weeks later, mice were treated with one of three standard regimens for latent tuberculosis infection or tumor necrosis factor (TNF)-neutralizing antibody. Mouse lungs were analyzed by histological features, positron emission tomography/computed tomography, whole-genome microarrays, and RT-PCR. Lungs and sera were studied by multiplex enzyme-linked immunosorbent assays. Paucibacillary infection was established, recapitulating the sterilizing activities of human latent tuberculosis infection regimens. TNF neutralization led to increased lung bacillary load, disrupted granuloma architecture with expanded necrotic foci and reduced tissue hypoxia, and accelerated animal mortality. TNF-neutralized mouse lungs and sera showed significant up-regulation of interferon γ, IL-1β, IL-6, IL-10, chemokine ligands 2 and 3, and matrix metalloproteinase genes. Clinical and microbiological reactivation of paucibacillary infection by TNF neutralization was associated with reduced hypoxia in lung granulomas and induction of matrix metalloproteinases and proinflammatory cytokines. This model may be useful for screening the sterilizing activity of novel anti-tuberculosis drugs, and identifying mycobacterial regulatory and metabolic pathways required for bacillary growth restriction and reactivation.
Collapse
Affiliation(s)
- Noton K Dutta
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Peter B Illei
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Sanjay K Jain
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Petros C Karakousis
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland; Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland.
| |
Collapse
|
42
|
Deeb SJ, Cox J, Schmidt-Supprian M, Mann M. N-linked glycosylation enrichment for in-depth cell surface proteomics of diffuse large B-cell lymphoma subtypes. Mol Cell Proteomics 2013; 13:240-51. [PMID: 24190977 DOI: 10.1074/mcp.m113.033977] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
Global analysis of lymphoma genome integrity and transcriptomes tremendously advanced our understanding of their biology. Technological advances in mass spectrometry-based proteomics promise to complete the picture by allowing the global quantification of proteins and their post-translational modifications. Here we use N-glyco FASP, a recently developed mass spectrometric approach using lectin-enrichment, in conjunction with a super-SILAC approach to quantify N-linked glycoproteins in lymphoma cells. From patient-derived diffuse large B-cell lymphoma cell lines, we mapped 2383 glycosites on 1321 protein groups, which were highly enriched for cell membrane proteins. This N-glyco subproteome alone allowed the segregation of the ABC from the GCB subtypes of diffuse large B-cell lymphoma, which before gene expression studies had been considered one disease entity. Encouragingly, many of the glycopeptides driving the segregation belong to proteins previously characterized as segregators in a deep proteome study of these subtypes (S. J. Deeb et al. MCP 2012 PMID 22442255). This conforms to the high correlation that we observed between the expression level of the glycosites and their corresponding proteins. Detailed examination of glycosites and glycoprotein expression levels uncovered, among other interesting findings, enrichment of transcription factor binding motifs, including known NF-kappa-B related ones. Thus, enrichment of a class of post-translationally modified peptides can classify cancer types as well as reveal cancer specific mechanistic changes.
Collapse
Affiliation(s)
- Sally J Deeb
- Proteomics and Signal Transduction Department, Max-Planck Institute of Biochemistry, D-82152 Martinsried, Germany
| | | | | | | |
Collapse
|
43
|
Puiffe ML, Lachaise I, Molinier-Frenkel V, Castellano F. Antibacterial properties of the mammalian L-amino acid oxidase IL4I1. PLoS One 2013; 8:e54589. [PMID: 23355881 PMCID: PMC3552961 DOI: 10.1371/journal.pone.0054589] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2012] [Accepted: 12/14/2012] [Indexed: 12/20/2022] Open
Abstract
L-amino acid oxidases (LAAO) are flavoproteins that catalyze the oxidative deamination of L-amino acids to a keto-acid along with the production of H2O2 and ammonia. Interleukin 4 induced gene 1 (IL4I1) is a secreted LAAO expressed by macrophages and dendritic cells stimulated by microbial derived products or interferons, which is endowed with immunoregulatory properties. It is the first LAAO described in mammalian innate immune cells. In this work, we show that this enzyme blocks the in vitro and in vivo growth of Gram negative and Gram positive bacteria. This antibiotic effect is primarily mediated by H2O2 production but is amplified by basification of the medium due to the accumulation of ammonia. The depletion of phenylalanine (the primary amino acid catabolized by IL4I1) may also participate in the in vivo inhibition of staphylococci growth. Thus, IL4I1 plays a distinct role compared to other antibacterial enzymes produced by mononuclear phagocytes.
Collapse
Affiliation(s)
- Marie-Line Puiffe
- INSERM, U955, IMRB, Equipe 09, Créteil, France
- Université Paris Est, Faculté de Médecine, Créteil, France
| | - Isabelle Lachaise
- Plateforme Chromatographie Analytique et semi Préparative, ICMPE, Thiais, France
| | - Valérie Molinier-Frenkel
- INSERM, U955, IMRB, Equipe 09, Créteil, France
- Université Paris Est, Faculté de Médecine, Créteil, France
- AP-HP, Hôpital H. Mondor - A. Chenevier, Service d’Immunologie Biologique, Créteil, France
- * E-mail: (FC); (VMF)
| | - Flavia Castellano
- INSERM, U955, IMRB, Equipe 09, Créteil, France
- Université Paris Est, Faculté de Médecine, Créteil, France
- AP-HP, Hôpital H. Mondor - A. Chenevier, Service d’Immunologie Biologique, Créteil, France
- * E-mail: (FC); (VMF)
| |
Collapse
|
44
|
Genes involved in systemic and arterial bed dependent atherosclerosis--Tampere Vascular study. PLoS One 2012; 7:e33787. [PMID: 22509262 PMCID: PMC3324479 DOI: 10.1371/journal.pone.0033787] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2010] [Accepted: 02/19/2012] [Indexed: 12/23/2022] Open
Abstract
Background Atherosclerosis is a complex disease with hundreds of genes influencing its progression. In addition, the phenotype of the disease varies significantly depending on the arterial bed. Methodology/Principal Findings We characterized the genes generally involved in human advanced atherosclerotic (AHA type V–VI) plaques in carotid and femoral arteries as well as aortas from 24 subjects of Tampere Vascular study and compared the results to non-atherosclerotic internal thoracic arteries (n=6) using genome-wide expression array and QRT-PCR. In addition we determined genes that were typical for each arterial plaque studied. To gain a comprehensive insight into the pathologic processes in the plaques we also analyzed pathways and gene sets dysregulated in this disease using gene set enrichment analysis (GSEA). According to the selection criteria used (>3.0 fold change and p-value <0.05), 235 genes were up-regulated and 68 genes down-regulated in the carotid plaques, 242 genes up-regulated and 116 down-regulated in the femoral plaques and 256 genes up-regulated and 49 genes down-regulated in the aortic plaques. Nine genes were found to be specifically induced predominantly in aortic plaques, e.g., lactoferrin, and three genes in femoral plaques, e.g., chondroadherin, whereas no gene was found to be specific for carotid plaques. In pathway analysis, a total of 28 pathways or gene sets were found to be significantly dysregulated in atherosclerotic plaques (false discovery rate [FDR] <0.25). Conclusions This study describes comprehensively the gene expression changes that generally prevail in human atherosclerotic plaques. In addition, site specific genes induced only in femoral or aortic plaques were found, reflecting that atherosclerotic process has unique features in different vascular beds.
Collapse
|
45
|
Santarlasci V, Maggi L, Capone M, Querci V, Beltrame L, Cavalieri D, D'Aiuto E, Cimaz R, Nebbioso A, Liotta F, De Palma R, Maggi E, Cosmi L, Romagnani S, Annunziato F. Rarity of human T helper 17 cells is due to retinoic acid orphan receptor-dependent mechanisms that limit their expansion. Immunity 2012; 36:201-14. [PMID: 22326581 DOI: 10.1016/j.immuni.2011.12.013] [Citation(s) in RCA: 95] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2011] [Revised: 11/07/2011] [Accepted: 12/06/2011] [Indexed: 01/13/2023]
Abstract
The reason why CD4(+) T helper 17 (Th17) cells, despite their well-known pathogenic role in chronic inflammatory disorders, are very rare in the inflammatory sites remains unclear. We demonstrate that human Th17 cells exhibit low ability to proliferate and to produce the T cell growth factor interleukin-2 (IL-2), in response to combined CD3 and CD28 stimulation. This was due to the upregulated expression of IL-4-induced gene 1 (IL4I1) mRNA, a secreted L-phenylalanine oxidase, which associated with a decrease in CD3ζ chain expression and consequent abnormalities in the molecular pathway that allows IL-2 production and cell proliferation. High IL4I1 mRNA expression was detectable in Th17 cell precursors and was strictly dependent on Th17 cell master gene, the retinoid acid related orphan receptor (RORC). Th17 cells also exhibited RORC-dependent CD28 hyperexpression and the ability to produce IL-17A after CD28 stimulation without CD3 triggering. Our findings suggest that the rarity of human Th17 cells in inflamed tissues results from RORC-dependent mechanisms limiting their expansion.
Collapse
Affiliation(s)
- Veronica Santarlasci
- Department of Internal Medicine and DENOTHE Center, University of Florence, 50134 Firenze, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Santer DM, Wiedeman AE, Teal TH, Ghosh P, Elkon KB. Plasmacytoid dendritic cells and C1q differentially regulate inflammatory gene induction by lupus immune complexes. THE JOURNAL OF IMMUNOLOGY 2011; 188:902-15. [PMID: 22147767 DOI: 10.4049/jimmunol.1102797] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Immune complexes (ICs) play a pivotal role in causing inflammation in systemic lupus erythematosus (SLE). Yet, it remains unclear what the dominant blood cell type(s) and inflammation-related gene programs stimulated by lupus ICs are. To address these questions, we exposed normal human PBMCs or CD14(+) isolated monocytes to SLE ICs in the presence or absence of C1q and performed microarray analysis and other tests for cell activation. By microarray analysis, we identified genes and pathways regulated by SLE ICs that are both type I IFN dependent and independent. We also found that C1q-containing ICs markedly reduced expression of the majority of IFN-response genes and also influenced the expression of multiple other genes induced by SLE ICs. Surprisingly, IC activation of isolated CD14(+) monocytes did not upregulate CD40 and CD86 and only modestly stimulated inflammatory gene expression. However, when monocyte subsets were purified and analyzed separately, the low-abundance CD14(dim) ("patrolling") subpopulation was more responsive to ICs. These observations demonstrate the importance of plasmacytoid dendritic cells, CD14(dim) monocytes, and C1q as key regulators of inflammatory properties of ICs and identify many pathways through which they act.
Collapse
Affiliation(s)
- Deanna M Santer
- Department of Immunology, School of Medicine, University of Washington, Seattle, WA 98195, USA
| | | | | | | | | |
Collapse
|
47
|
Lasoudris F, Cousin C, Prevost-Blondel A, Martin-Garcia N, Abd-Alsamad I, Ortonne N, Farcet JP, Castellano F, Molinier-Frenkel V. IL4I1: an inhibitor of the CD8⁺ antitumor T-cell response in vivo. Eur J Immunol 2011; 41:1629-38. [PMID: 21469114 DOI: 10.1002/eji.201041119] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2010] [Revised: 02/04/2011] [Accepted: 03/15/2011] [Indexed: 12/21/2022]
Abstract
The L-phenylalanine oxidase IL4I1 inhibits T-cell proliferation in vitro through H(2) O(2) production, and is highly expressed in tumor-associated macrophages. IL4I1 is also detected by immunohistochemistry in neoplastic cells from several B-cell lymphomas and some non-lymphoid tumors. To evaluate IL4I1's effect on tumor growth, we developed a mouse melanoma model constitutively coexpressing IL4I1 and the GP33 epitope. After GP33 vaccination, tumors developed more frequently in mice injected with IL4I1-expressing cells in comparison with mice receiving control cells. Tumor escape was preceded by a rapid diminution of IFN-γ-producing cytotoxic antitumor CD8(+) T cells. Moreover, tumor incidence was already increased when only 20% of the injected cells expressed IL4I1. The minimal IL4I1 activities leading to tumor escape were close to those detected in human melanoma and mesothelioma. Thus, we demonstrate the immunosuppressive functions of IL4I1 in vivo and suggest that IL4I1 facilitates human tumor growth by inhibiting the CD8(+) antitumor T-cell response.
Collapse
|