1
|
Chu J, Wu Y, Qu Z, Zhuang J, Liu J, Han S, Wu W, Han S. Transcriptional profile and immune infiltration in colorectal cancer reveal the significance of inducible T-cell costimulator as a crucial immune checkpoint molecule. Cancer Med 2024; 13:e7097. [PMID: 38506253 PMCID: PMC10952025 DOI: 10.1002/cam4.7097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Revised: 08/25/2023] [Accepted: 02/17/2024] [Indexed: 03/21/2024] Open
Abstract
BACKGROUND Emergence of novel immuno-therapeutics has shown promising improvement in the clinical outcome of colorectal cancer (CRC). OBJECTIVE To identify robust immune checkpoints based on expression and immune infiltration profiles of clinical CRC samples. METHODS One dataset from The Cancer Genome Atlas database and two from Gene Expression Omnibus were independently employed for the analysis. Genes associated with overall survival were identified, and distribution of each immune checkpoint with respect to different clinical features was determined to explore key immune checkpoints. Multiple staining methods were used to verify the correlation between key immune checkpoint ICOS and clinical pathological features. Differentially expressed mRNA and long non-coding RNA (lncRNA) were then detected for gene set enrichment analysis and gene set variation analysis to investigate the differentially enriched biological processes between low- and high-expression groups. Significant immune-related mRNAs and lncRNA were subjected to competing endogenous RNA (ceRNA) network analysis. Correlation of inducible T-cell costimulator (ICOS) and top 10 genes in ceRNA network were further considered for validation. RESULTS ICOS was identified from 14 immune checkpoints as the most highly correlated gene with survival and clinical features in CRC. The expression of ICOS protein in the poorly differentiated group was lower than that in the moderately differentiated group, and the expression in different pathological stages was significant. In addition, the expressions of ICOS were negatively correlated with Ki67. A conspicuous number of immune-related pathways were enriched in differentially expressed genes in the ICOS high- and low-expression groups. Integration with immune infiltration data revealed a multitude of differentially expressed immune-related genes enriched for ceRNA network. Furthermore, expression of top 10 genes investigated from ceRNA network showed high correlation with ICOS. CONCLUSION ICOS might serve as a robust immune checkpoint for prognosis with several genes being potential targets of ICOS-directed immunotherapy in CRC.
Collapse
Affiliation(s)
- Jian Chu
- Huzhou Central HospitalAffiliated Central Hospital Huzhou UniversityHuzhouChina
- Fifth School of Clinical Medicine of Zhejiang Chinese Medical University (Huzhou Central Hospital)HuzhouChina
- Key Laboratory of Multiomics Research and Clinical Transformation of Digestive Cancer of HuzhouHuzhouChina
| | - Yinghang Wu
- Huzhou Central HospitalAffiliated Central Hospital Huzhou UniversityHuzhouChina
- Fifth School of Clinical Medicine of Zhejiang Chinese Medical University (Huzhou Central Hospital)HuzhouChina
- Key Laboratory of Multiomics Research and Clinical Transformation of Digestive Cancer of HuzhouHuzhouChina
| | - Zhanbo Qu
- Huzhou Central HospitalAffiliated Central Hospital Huzhou UniversityHuzhouChina
- Fifth School of Clinical Medicine of Zhejiang Chinese Medical University (Huzhou Central Hospital)HuzhouChina
- Key Laboratory of Multiomics Research and Clinical Transformation of Digestive Cancer of HuzhouHuzhouChina
| | - Jing Zhuang
- Huzhou Central HospitalAffiliated Central Hospital Huzhou UniversityHuzhouChina
- Fifth School of Clinical Medicine of Zhejiang Chinese Medical University (Huzhou Central Hospital)HuzhouChina
- Key Laboratory of Multiomics Research and Clinical Transformation of Digestive Cancer of HuzhouHuzhouChina
| | - Jiang Liu
- Huzhou Central HospitalAffiliated Central Hospital Huzhou UniversityHuzhouChina
- Fifth School of Clinical Medicine of Zhejiang Chinese Medical University (Huzhou Central Hospital)HuzhouChina
- Key Laboratory of Multiomics Research and Clinical Transformation of Digestive Cancer of HuzhouHuzhouChina
| | - Shugao Han
- Second Affiliated Hospital of School of MedicineZhejiang UniversityHangzhouChina
| | - Wei Wu
- Huzhou Central HospitalAffiliated Central Hospital Huzhou UniversityHuzhouChina
- Fifth School of Clinical Medicine of Zhejiang Chinese Medical University (Huzhou Central Hospital)HuzhouChina
- Key Laboratory of Multiomics Research and Clinical Transformation of Digestive Cancer of HuzhouHuzhouChina
| | - Shuwen Han
- Huzhou Central HospitalAffiliated Central Hospital Huzhou UniversityHuzhouChina
- Fifth School of Clinical Medicine of Zhejiang Chinese Medical University (Huzhou Central Hospital)HuzhouChina
- Key Laboratory of Multiomics Research and Clinical Transformation of Digestive Cancer of HuzhouHuzhouChina
| |
Collapse
|
2
|
Abdel-Rahman SA, Santini BL, Calvo-Barreiro L, Zacharias M, Gabr M. Design of cyclic peptides as novel inhibitors of ICOS/ICOSL interaction. Bioorg Med Chem Lett 2024; 99:129599. [PMID: 38185345 DOI: 10.1016/j.bmcl.2024.129599] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Revised: 12/31/2023] [Accepted: 01/02/2024] [Indexed: 01/09/2024]
Abstract
Compared to small molecules and antibodies, cyclic peptides exhibit unique biochemical and therapeutic attributes in the realm of pharmaceutical applications. The interaction between the inducible costimulator (ICOS) and its ligand (ICOSL) plays a key role in T-cell differentiation and activation. ICOS/ICOSL inhibition results in a reduction in the promotion of immunosuppressive regulatory T cells (Tregs) in both hematologic malignancies and solid tumors. Herein, we implement the computational cPEPmatch approach to design the first examples of cyclic peptides that inhibit ICOS/ICOSL interaction. The top cyclic peptide from our approach possessed an IC50 value of 1.87 ± 0.15 μM as an ICOS/ICOSL inhibitor and exhibited excellent in vitro pharmacokinetic properties as a drug candidate. Our work will lay the groundwork for future endeavors in cancer drug discovery, with the goal of developing cyclic peptides that target the ICOS/ICOSL interaction.
Collapse
Affiliation(s)
- Somaya A Abdel-Rahman
- Department of Radiology, Molecular Imaging Innovations Institute (MI3), Weill Cornell Medicine, New York, NY 10065, USA; Department of Medicinal Chemistry, Faculty of Pharmacy, Mansoura University, Mansoura 35516, Egypt
| | - Brianda L Santini
- Center for Functional Protein Assemblies, Technical University of Munich, Ernst-Otto-Fischer-Straße 8, Garching, Germany
| | - Laura Calvo-Barreiro
- Department of Radiology, Molecular Imaging Innovations Institute (MI3), Weill Cornell Medicine, New York, NY 10065, USA
| | - Martin Zacharias
- Center for Functional Protein Assemblies, Technical University of Munich, Ernst-Otto-Fischer-Straße 8, Garching, Germany
| | - Moustafa Gabr
- Department of Radiology, Molecular Imaging Innovations Institute (MI3), Weill Cornell Medicine, New York, NY 10065, USA.
| |
Collapse
|
3
|
Wang P, Zhang Q, Zhang H, Shao J, Zhang H, Wang Z. Molecular and clinical characterization of ICOS expression in breast cancer through large-scale transcriptome data. PLoS One 2023; 18:e0293469. [PMID: 38127899 PMCID: PMC10734928 DOI: 10.1371/journal.pone.0293469] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Accepted: 10/13/2023] [Indexed: 12/23/2023] Open
Abstract
ICOS (Inducible T Cell Costimulator), one of the co-stimulatory B7 superfamily members, was characterized as a co-stimulatory receptor for T-cell enhancement. However, the role of ICOS in breast cancer remains largely unknown. The present study systematically investigated the expression pattern and its relation to clinical characteristics and immunotherapy by integrating multiple clinical cohorts and large-scale gene expression data. This study included 2994 breast tumor samples with transcriptome data and matched clinical data. To make our findings more reliable, we set the TCGA cohort as the discovery set and the METABRIC cohort as the validation set. The expression of ICOS in breast cancer is strongly associated with major clinical and molecular characteristics. There is an association between higher ICOS expression and malignant subtypes and grades of tumors. In addition, gene ontology analysis based on genes significantly correlated with ICOS expression indicated that the expression of ICOS is mainly associated with immune responses and inflammation. We also observed strong correlations between ICOS and other promising immune-checkpoint molecules, including PD1, PDL1, CTLA4, and IDO1. Furthermore, we found that ICOS expression is associated with the response to anti-PDL1 immunotherapy and may serve as a biomarker for immunotherapy prediction. Our results indicated higher ICOS expression is significantly associated with favorable survival in triple-negative breast cancer (TNBC) patients, but not for all subtypes of breast cancer patients. In summary, ICOS correlates with higher malignant breast cancers, and it contributes to the regulation of the immune microenvironment of breast tumors, making it a potential biomarker and immunotherapy target.
Collapse
Affiliation(s)
- Peng Wang
- Thyroid and Breast Department III, Cangzhou Central Hospital, Cangzhou, Hebei Province, China
| | - Qin Zhang
- Thyroid and Breast Department III, Cangzhou Central Hospital, Cangzhou, Hebei Province, China
| | - Hengle Zhang
- Hebei Medical University, Shijiazhuang, Hebei Province, China
| | - Jianqiang Shao
- Thyroid and Breast Department III, Cangzhou Central Hospital, Cangzhou, Hebei Province, China
| | - Hui Zhang
- Thyroid and Breast Department III, Cangzhou Central Hospital, Cangzhou, Hebei Province, China
| | - Zunyi Wang
- Thyroid and Breast Department III, Cangzhou Central Hospital, Cangzhou, Hebei Province, China
| |
Collapse
|
4
|
Abdel-Rahman SA, Świderek K, Gabr MT. First-in-class small molecule inhibitors of ICOS/ICOSL interaction as a novel class of immunomodulators. RSC Med Chem 2023; 14:1767-1777. [PMID: 37731692 PMCID: PMC10507805 DOI: 10.1039/d3md00150d] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Accepted: 07/28/2023] [Indexed: 09/22/2023] Open
Abstract
The interaction of the inducible co-stimulator (ICOS) with its ligand (ICOSL) plays key roles in T-cell differentiation and activation of T-cell to B-cell functions. The ICOS/ICOSL pathway is a validated target for T-cell lymphomas induced by the proliferation of T-follicular helper (Tfh) cells. Moreover, the inhibition of ICOS/ICOSL interaction can decrease the enhancement of immunosuppressive regulatory T cells (Tregs) in both hematologic malignancies and solid tumors. However, targeting ICOS/ICOSL interaction is currently restricted to monoclonal antibodies (mAbs) and there are no small molecules in existence that can block ICOS/ICOSL. To fill this gap, we report herein the first time-resolved fluorescence resonance energy transfer (TR-FRET) assay to evaluate the ability of small molecules to inhibit ICOS/ICOSL interaction. Implementation of the developed TR-FRET assay in high-throughput screening (HTS) of a focused chemical library resulted in the identification of AG-120 as a first-in-class inhibitor of ICOS/ICOSL interaction. We further employed docking studies and molecular dynamics (MD) simulations to identify the plausible mechanism of blocking ICOS/ICOSL complex formation by AG-120. Using the structure-activity relationship (SAR) by catalog approach, we identified AG-120-X with an IC50 value of 4.68 ± 0.47 μM in the ICOS/ICOSL TR-FRET assay. Remarkably, AG-120-X revealed a dose-dependent ability to block ICOS/ICOSL interaction in a bioluminescent cellular assay based on co-culturing Jurkat T cells expressing ICOS and CHO-K1 cells expressing ICOSL. This work will pave the way for future drug discovery efforts aiming at the development of small molecule inhibitors of ICOS/ICOSL interaction as potential therapeutics for cancer as well as other diseases.
Collapse
Affiliation(s)
- Somaya A Abdel-Rahman
- Department of Radiology, Molecular Imaging Innovations Institute (MI3), Weill Cornell Medicine New York NY 10065 USA
- Department of Medicinal Chemistry, Faculty of Pharmacy, Mansoura University Mansoura 35516 Egypt
| | - Katarzyna Świderek
- BioComp Group, Institute of Advanced Materials (INAM), Universitat Jaume I 12071 Castellon Spain
| | - Moustafa T Gabr
- Department of Radiology, Molecular Imaging Innovations Institute (MI3), Weill Cornell Medicine New York NY 10065 USA
| |
Collapse
|
5
|
Monge C, Stoppa I, Ferraris C, Bozza A, Battaglia L, Cangemi L, Miglio G, Pizzimenti S, Clemente N, Gigliotti CL, Boggio E, Dianzani U, Dianzani C. Parenteral Nanoemulsions Loaded with Combined Immuno- and Chemo-Therapy for Melanoma Treatment. NANOMATERIALS (BASEL, SWITZERLAND) 2022; 12:nano12234233. [PMID: 36500861 PMCID: PMC9740980 DOI: 10.3390/nano12234233] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Revised: 11/22/2022] [Accepted: 11/26/2022] [Indexed: 06/01/2023]
Abstract
High-grade melanoma remains a major life-threatening illness despite the improvement in therapeutic control that has been achieved by means of targeted therapies and immunotherapies in recent years. This work presents a preclinical-level test of a multi-pronged approach that includes the loading of immunotherapeutic (ICOS-Fc), targeted (sorafenib), and chemotherapeutic (temozolomide) agents within Intralipid®, which is a biocompatible nanoemulsion with a long history of safe clinical use for total parenteral nutrition. This drug combination has been shown to inhibit tumor growth and angiogenesis with the involvement of the immune system, and a key role is played by ICOS-Fc. The inhibition of tumor growth in subcutaneous melanoma mouse models has been achieved using sub-therapeutic drug doses, which is most likely the result of the nanoemulsion's targeting properties. If translated to the human setting, this approach should therefore allow therapeutic efficacy to be achieved without increasing the risk of toxic effects.
Collapse
Affiliation(s)
- Chiara Monge
- Dipartimento di Scienza e Tecnologia del Farmaco, Università degli Studi di Torino, via Pietro Giuria 9, 10125 Torino, Italy
| | - Ian Stoppa
- Dipartimento di Scienze della Salute, Università del Piemonte Orientale, via Solaroli 17, 28100 Novara, Italy
| | - Chiara Ferraris
- Dipartimento di Scienza e Tecnologia del Farmaco, Università degli Studi di Torino, via Pietro Giuria 9, 10125 Torino, Italy
- Dipartimento di Scienze Cliniche e Biologiche, Università degli Studi di Torino, Regione Gonzole 10, 10043 Orbassano, Italy
| | - Annalisa Bozza
- Dipartimento di Scienza e Tecnologia del Farmaco, Università degli Studi di Torino, via Pietro Giuria 9, 10125 Torino, Italy
| | - Luigi Battaglia
- Dipartimento di Scienza e Tecnologia del Farmaco, Università degli Studi di Torino, via Pietro Giuria 9, 10125 Torino, Italy
- Nanostructured Interfaces and Surfaces (NIS) Interdepartmental Centre, Università degli Studi di Torino, 10124 Torino, Italy
| | - Luigi Cangemi
- Dipartimento di Scienza e Tecnologia del Farmaco, Università degli Studi di Torino, via Pietro Giuria 9, 10125 Torino, Italy
| | - Gianluca Miglio
- Dipartimento di Scienza e Tecnologia del Farmaco, Università degli Studi di Torino, via Pietro Giuria 9, 10125 Torino, Italy
| | - Stefania Pizzimenti
- Dipartimento di Scienze Cliniche e Biologiche, Università degli Studi di Torino, Corso Raffaello 30, 10124 Torino, Italy
| | - Nausicaa Clemente
- Dipartimento di Scienze della Salute, Università del Piemonte Orientale, via Solaroli 17, 28100 Novara, Italy
| | - Casimiro Luca Gigliotti
- Dipartimento di Scienze della Salute, Università del Piemonte Orientale, via Solaroli 17, 28100 Novara, Italy
| | - Elena Boggio
- Dipartimento di Scienze della Salute, Università del Piemonte Orientale, via Solaroli 17, 28100 Novara, Italy
| | - Umberto Dianzani
- Dipartimento di Scienze della Salute, Università del Piemonte Orientale, via Solaroli 17, 28100 Novara, Italy
- Azienda Ospedaliero-Universitaria Maggiore della Carità, Corso Giuseppe Mazzini 18, 28100 Novara, Italy
| | - Chiara Dianzani
- Dipartimento di Scienza e Tecnologia del Farmaco, Università degli Studi di Torino, via Pietro Giuria 9, 10125 Torino, Italy
| |
Collapse
|
6
|
Gigliotti CL, Boggio E, Favero F, Incarnato D, Santoro C, Oliviero S, Rojo JM, Zucchelli S, Persichetti F, Baldanzi G, Dianzani U, Corà D. Specific transcriptional programs differentiate ICOS from CD28 costimulatory signaling in human Naïve CD4+ T cells. Front Immunol 2022; 13:915963. [PMID: 36131938 PMCID: PMC9484324 DOI: 10.3389/fimmu.2022.915963] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Accepted: 08/11/2022] [Indexed: 11/13/2022] Open
Abstract
Costimulatory molecules of the CD28 family play a crucial role in the activation of immune responses in T lymphocytes, complementing and modulating signals originating from the T-cell receptor (TCR) complex. Although distinct functional roles have been demonstrated for each family member, the specific signaling pathways differentiating ICOS- from CD28-mediated costimulation during early T-cell activation are poorly characterized. In the present study, we have performed RNA-Seq-based global transcriptome profiling of anti-CD3-treated naïve CD4+ T cells upon costimulation through either inducible costimulator (ICOS) or CD28, revealing a set of signaling pathways specifically associated with each signal. In particular, we show that CD3/ICOS costimulation plays a major role in pathways related to STAT3 function and osteoarthritis (OA), whereas the CD3/CD28 axis mainly regulates p38 MAPK signaling. Furthermore, we report the activation of distinct immunometabolic pathways, with CD3/ICOS costimulation preferentially targeting glycosaminoglycans (GAGs) and CD3/CD28 regulating mitochondrial respiratory chain and cholesterol biosynthesis. These data suggest that ICOS and CD28 costimulatory signals play distinct roles during the activation of naïve T cells by modulating distinct sets of immunological and immunometabolic genes.
Collapse
Affiliation(s)
- Casimiro Luca Gigliotti
- Department of Health Sciences, University of Piemonte Orientale, Novara, Italy
- Interdisciplinary Research Center of Autoimmune Diseases (IRCAD), University of Piemonte Orientale, Novara, Italy
| | - Elena Boggio
- Department of Health Sciences, University of Piemonte Orientale, Novara, Italy
- Interdisciplinary Research Center of Autoimmune Diseases (IRCAD), University of Piemonte Orientale, Novara, Italy
| | - Francesco Favero
- Department of Health Sciences, University of Piemonte Orientale, Novara, Italy
- Interdisciplinary Research Center of Autoimmune Diseases (IRCAD), University of Piemonte Orientale, Novara, Italy
- CAAD - Center for Translational Research on Autoimmune and Allergic Disease, Novara, Italy
| | - Danny Incarnato
- Department of Molecular Genetics, Groningen Biomolecular Sciences and Biotechnology Institute (GBB), University of Groningen, Groningen, Netherlands
| | - Claudio Santoro
- Department of Health Sciences, University of Piemonte Orientale, Novara, Italy
- Interdisciplinary Research Center of Autoimmune Diseases (IRCAD), University of Piemonte Orientale, Novara, Italy
- CAAD - Center for Translational Research on Autoimmune and Allergic Disease, Novara, Italy
| | - Salvatore Oliviero
- Dipartimento di Scienze della Vita e Biologia dei Sistemi, Università di Torino, Torino, Italy
- Italian Institute for Genomic Medicine (IIGM), Torino, Italy
| | - Josè Maria Rojo
- Centro de Investigaciones Biológicas Margarita Salas, Consejo Superior de Investigaciones Cientificas (CSIC), Madrid, Spain
| | - Silvia Zucchelli
- Department of Health Sciences, University of Piemonte Orientale, Novara, Italy
- Interdisciplinary Research Center of Autoimmune Diseases (IRCAD), University of Piemonte Orientale, Novara, Italy
- CAAD - Center for Translational Research on Autoimmune and Allergic Disease, Novara, Italy
| | - Francesca Persichetti
- Department of Health Sciences, University of Piemonte Orientale, Novara, Italy
- Interdisciplinary Research Center of Autoimmune Diseases (IRCAD), University of Piemonte Orientale, Novara, Italy
| | - Gianluca Baldanzi
- Interdisciplinary Research Center of Autoimmune Diseases (IRCAD), University of Piemonte Orientale, Novara, Italy
- CAAD - Center for Translational Research on Autoimmune and Allergic Disease, Novara, Italy
- Department of Translational Medicine, University of Piemonte Orientale, Novara, Italy
| | - Umberto Dianzani
- Department of Health Sciences, University of Piemonte Orientale, Novara, Italy
- Interdisciplinary Research Center of Autoimmune Diseases (IRCAD), University of Piemonte Orientale, Novara, Italy
- Biochemical Chemistry, “Maggiore della Carità” University Hospital, Novara, Italy
- *Correspondence: Umberto Dianzani,
| | - Davide Corà
- Interdisciplinary Research Center of Autoimmune Diseases (IRCAD), University of Piemonte Orientale, Novara, Italy
- CAAD - Center for Translational Research on Autoimmune and Allergic Disease, Novara, Italy
- Department of Translational Medicine, University of Piemonte Orientale, Novara, Italy
| |
Collapse
|
7
|
Boggio E, Gigliotti CL, Moia R, Scotta A, Crespi I, Boggione P, De Paoli L, Deambrogi C, Garzaro M, Vidali M, Chiocchetti A, Stoppa I, Rolla R, Dianzani C, Monge C, Clemente N, Gaidano G, Dianzani U. Inducible T-cell co-stimulator (ICOS) and ICOS ligand are novel players in the multiple-myeloma microenvironment. Br J Haematol 2021; 196:1369-1380. [PMID: 34954822 DOI: 10.1111/bjh.17968] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 11/03/2021] [Accepted: 11/09/2021] [Indexed: 12/19/2022]
Abstract
The inducible T-cell co-stimulator (ICOS) is a T-cell receptor that, once bound to ICOS ligand (ICOSL) expressed on several cell types including the B-cell lineage, plays a decisive role in adaptive immunity by regulating the interplay between B and T cells. In addition to its immunomodulatory functions, we have shown that ICOS/ICOSL signalling can inhibit the activity of osteoclasts, unveiling a novel mechanism of lymphocyte-bone cells interactions. ICOS and ICOSL can also be found as soluble forms, namely sICOS and sICOSL. Here we show that: (i) levels of sICOS and sICOSL are increased in multiple myeloma (MM) compared to monoclonal gammopathy of undetermined significance and smouldering MM; (ii) levels of sICOS and sICOSL variably correlate with several markers of tumour burden; and (iii) sICOS levels tend to be higher in Durie-Salmon stage II/III versus stage I MM and correlate with overall survival as an independent variable. Moreover, surface ICOS and ICOSL are expressed in both myeloma cells and normal plasma cells, where they probably regulate different functional stages. Finally, ICOSL triggering inhibits the migration of myeloma cell lines in vitro and the growth of ICOSL+ MOPC-21 myeloma cells in vivo. These results suggest that ICOS and ICOSL represent novel markers and therapeutic targets for MM.
Collapse
Affiliation(s)
- Elena Boggio
- Department of Health Sciences and Interdisciplinary Research Center of Autoimmune Diseases (IRCAD), Università del Piemonte Orientale, Novara, Italy
| | - Casimiro Luca Gigliotti
- Department of Health Sciences and Interdisciplinary Research Center of Autoimmune Diseases (IRCAD), Università del Piemonte Orientale, Novara, Italy
| | - Riccardo Moia
- Division of Hematology, Department of Translational Medicine, Università del Piemonte Orientale, Novara, Italy.,Maggiore della Carità University Hospital, Novara, Italy
| | | | - Ilaria Crespi
- Maggiore della Carità University Hospital, Novara, Italy
| | - Paola Boggione
- Division of Hematology, Department of Translational Medicine, Università del Piemonte Orientale, Novara, Italy.,Maggiore della Carità University Hospital, Novara, Italy
| | - Lorenzo De Paoli
- Division of Hematology, Department of Translational Medicine, Università del Piemonte Orientale, Novara, Italy.,Maggiore della Carità University Hospital, Novara, Italy
| | - Clara Deambrogi
- Division of Hematology, Department of Translational Medicine, Università del Piemonte Orientale, Novara, Italy.,Maggiore della Carità University Hospital, Novara, Italy
| | - Massimiliano Garzaro
- Department of Health Sciences and Interdisciplinary Research Center of Autoimmune Diseases (IRCAD), Università del Piemonte Orientale, Novara, Italy
| | - Matteo Vidali
- Maggiore della Carità University Hospital, Novara, Italy
| | - Annalisa Chiocchetti
- Department of Health Sciences and Interdisciplinary Research Center of Autoimmune Diseases (IRCAD), Università del Piemonte Orientale, Novara, Italy
| | - Ian Stoppa
- Department of Health Sciences and Interdisciplinary Research Center of Autoimmune Diseases (IRCAD), Università del Piemonte Orientale, Novara, Italy
| | - Roberta Rolla
- Department of Health Sciences and Interdisciplinary Research Center of Autoimmune Diseases (IRCAD), Università del Piemonte Orientale, Novara, Italy.,Maggiore della Carità University Hospital, Novara, Italy
| | - Chiara Dianzani
- Department of Scienza e Tecnologia del Farmaco, University of Turin, Turin, Italy
| | - Chiara Monge
- Department of Scienza e Tecnologia del Farmaco, University of Turin, Turin, Italy
| | - Nausicaa Clemente
- Department of Health Sciences and Interdisciplinary Research Center of Autoimmune Diseases (IRCAD), Università del Piemonte Orientale, Novara, Italy
| | - Gianluca Gaidano
- Division of Hematology, Department of Translational Medicine, Università del Piemonte Orientale, Novara, Italy.,Maggiore della Carità University Hospital, Novara, Italy
| | - Umberto Dianzani
- Department of Health Sciences and Interdisciplinary Research Center of Autoimmune Diseases (IRCAD), Università del Piemonte Orientale, Novara, Italy.,Maggiore della Carità University Hospital, Novara, Italy
| |
Collapse
|
8
|
Montes-Casado M, Ojeda G, Criado G, Rojo JM, Portolés P. The PI-3-Kinase P110α Catalytic Subunit of T Lymphocytes Modulates Collagen-Induced Arthritis. Int J Mol Sci 2021; 22:6405. [PMID: 34203838 PMCID: PMC8232790 DOI: 10.3390/ijms22126405] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Accepted: 06/11/2021] [Indexed: 12/21/2022] Open
Abstract
The phosphatidylinositol 3-kinase (PI3K) family of enzymes plays a determinant role in inflammation and autoimmune responses. However, the implication of the different isoforms of catalytic subunits in these processes is not clear. Rheumatoid arthritis (RA) is a chronic, systemic autoimmune inflammatory disease that entails innate and adaptive immune response elements in which PI3K is a potential hub for immune modulation. In a mouse transgenic model with T-cell-specific deletion of p110α catalytic chain (p110α-/-ΔT), we show the modulation of collagen-induced arthritis (CIA) by this isoform of PI3K. In established arthritis, p110α-/-ΔT mice show decreased prevalence of illness than their control siblings, higher IgG1 titers and lower levels of IL-6 in serum, together with decreased ex vivo Collagen II (CII)-induced proliferation, IL-17A secretion and proportion of naive T cells in the lymph nodes. In a pre-arthritis phase, at 13 days post-Ag, T-cell-specific deletion of p110α chain induced an increased, less pathogenic IgG1/IgG2a antibodies ratio; changes in the fraction of naive and effector CD4+ subpopulations; and an increased number of CXCR5+ T cells in the draining lymph nodes of the p110α-/-ΔT mice. Strikingly, T-cell blasts in vitro obtained from non-immunized p110α-/-ΔT mice showed an increased expression of CXCR5, CD44 and ICOS surface markers and defective ICOS-induced signaling towards Akt phosphorylation. These results, plus the accumulation of cells in the lymph nodes in the early phase of the process, could explain the diminished illness incidence and prevalence in the p110α-/-ΔT mice and suggests a modulation of CIA by the p110α catalytic chain of PI3K, opening new avenues of intervention in T-cell-directed therapies to autoimmune diseases.
Collapse
Affiliation(s)
- María Montes-Casado
- Centro Nacional de Microbiología, Instituto de Salud Carlos III (ISCIII), Majadahonda, 28220 Madrid, Spain; (M.M.-C.); (G.O.)
| | - Gloria Ojeda
- Centro Nacional de Microbiología, Instituto de Salud Carlos III (ISCIII), Majadahonda, 28220 Madrid, Spain; (M.M.-C.); (G.O.)
| | - Gabriel Criado
- Grupo de Enfermedades Inflamatorias y Autoinmunes, Instituto de Investigación Hospital 12 de Octubre (i+12), 28041 Madrid, Spain;
| | - José M. Rojo
- Centro de Investigaciones Biológicas Margarita Salas, Departamento de Biomedicina Molecular, Consejo Superior de Investigaciones Científicas (CSIC), 28040 Madrid, Spain
| | - Pilar Portolés
- Centro Nacional de Microbiología, Instituto de Salud Carlos III (ISCIII), Majadahonda, 28220 Madrid, Spain; (M.M.-C.); (G.O.)
- Presidencia, Consejo Superior de Investigaciones Científicas (CSIC), 28006 Madrid, Spain
| |
Collapse
|
9
|
Aragoneses-Fenoll L, Montes-Casado M, Ojeda G, García-Paredes L, Arimura Y, Yagi J, Dianzani U, Portolés P, Rojo JM. Role of endocytosis and trans-endocytosis in ICOS costimulator-induced downmodulation of the ICOS Ligand. J Leukoc Biol 2021; 110:867-884. [PMID: 33527556 PMCID: PMC8597029 DOI: 10.1002/jlb.2a0220-127r] [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: 03/06/2020] [Revised: 12/16/2020] [Accepted: 01/13/2020] [Indexed: 12/20/2022] Open
Abstract
The interaction between the T-lymphocyte costimulatory molecule ICOS and its ligand (ICOS-L) is needed for efficient immune responses, but expression levels are tightly controlled, as altered expression of ICOS or ICOS-L may lead to immunodeficiency, or favor autoimmune diseases and tumor growth. Using cells of mouse B cell lymphoma (M12.C3) and melanoma (B16), or hamster CHO cells transfected with various forms of mouse ICOS-L, and ICOS+ T cell lines, we show that, within minutes, ICOS induces significant downmodulation of surface ICOS-L that is largely mediated by endocytosis and trans-endocytosis. So, after interaction with ICOS+ cells, ICOS-L was found inside permeabilized cells, or in cell lysates, with significant transfer of ICOS from ICOS+ T cells to ICOS-L-expressing cells, and simultaneous loss of surface ICOS by the T cells. Data from cells expressing ICOS-L mutants show that conserved, functionally important residues in the cytoplasmic domain of mouse ICOS-L (Arg300 , Ser307 and Tyr308 ), or removal of ICOS-L cytoplasmic tail have minor effect on its internalization. Internalization was dependent on temperature, and was partially dependent on actin polymerization, the GTPase dynamin, protein kinase C, or the integrity of lipid rafts. In fact, a fraction of ICOS-L was detected in lipid rafts. On the other hand, proteinase inhibitors had negligible effects on early modulation of ICOS-L from the cell surface. Our data add a new mechanism of control of ICOS-L expression to the regulation of ICOS-dependent responses.
Collapse
Affiliation(s)
- Laura Aragoneses-Fenoll
- Unidad de Inmunología Celular, Centro Nacional de Microbiología, Instituto de Salud Carlos III, Majadahonda, Madrid, 28220, Spain
| | - María Montes-Casado
- Unidad de Inmunología Celular, Centro Nacional de Microbiología, Instituto de Salud Carlos III, Majadahonda, Madrid, 28220, Spain
| | - Gloria Ojeda
- Unidad de Inmunología Celular, Centro Nacional de Microbiología, Instituto de Salud Carlos III, Majadahonda, Madrid, 28220, Spain
| | - Lucía García-Paredes
- Departamento de Biomedicina Molecular, Centro de Investigaciones Biológicas Margarita Salas, CSIC, Madrid, 28040, Spain.,Current address: Hospital 12 de Octubre, Departamento de Oncología Médica, Av. de Córdoba, s/n, Madrid, 28041, Spain
| | - Yutaka Arimura
- Host Defense for Animals, Nippon Veterinary and Life Science University, 1-7-1 Kyonan, Musashino, Tokyo, 180-8602, Japan
| | - Junji Yagi
- Department of Microbiology and Immunology, Tokyo Women's Medical University, Tokyo, 108-8639, Japan
| | - Umberto Dianzani
- Interdisciplinary Research Center of Autoimmune Diseases (IRCAD) and Department of Health Sciences, University of Piemonte Orientale (UPO), Novara, 28100, Italy
| | - Pilar Portolés
- Unidad de Inmunología Celular, Centro Nacional de Microbiología, Instituto de Salud Carlos III, Majadahonda, Madrid, 28220, Spain.,Presidencia, Consejo Superior de Investigaciones Científicas, Madrid, 28006, Spain
| | - José M Rojo
- Departamento de Biomedicina Molecular, Centro de Investigaciones Biológicas Margarita Salas, CSIC, Madrid, 28040, Spain
| |
Collapse
|
10
|
Zhao MG, Peng C, Li LM, Chen L, Zhang HF. Circulating Treg cells from patients with cerebral aneurysms displayed deficiency in ICOS expression and function. Clin Exp Pharmacol Physiol 2020; 47:1923-1931. [PMID: 32726458 DOI: 10.1111/1440-1681.13388] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2019] [Revised: 07/12/2020] [Accepted: 07/21/2020] [Indexed: 12/23/2022]
Abstract
Inducible costimulator (ICOS) is a member of the CD28 family. When activated, ICOS signalling promotes FOXP3 CNS2 gene demethylation and stabilizes Treg differentiation. Cerebral aneurysm (CA) is the local ballooning of the cerebral vasculature, characterized by higher levels of inflammation mediators and tissue remodelling. FOXP3+ Treg cell dysfunction may contribute to CA pathogenesis. In this study, the expression and function of ICOS in Treg cells was investigated. Circulating CD4+ CD25hi T cells from CA subjects demonstrated significantly lower levels of ICOS expression than circulating CD4+ CD25hi T cells from healthy subjects. In both healthy subjects and CA subjects, FOXP3+ Treg cells were highly concentrated in the ICOS+ fraction of CD4+ CD25hi T cells. Anti-ICOS costimulation, in combination with anti-CD3 and IL-2, significantly increased FOXP3 expression in CD4+ CD25hi ICOS+ T cells but not in CD4+ CD25hi ICOS- T cells. In addition, anti-CD3/IL-2 and anti-ICOS costimulation significantly elevated the expression of IL-10 and TGF-β, decreased the expression of IL-17, and enhanced CD4+ CD25hi ICOS+ T cell-mediated suppression of autologous CD4+ CD25- Tconv proliferation. Interestingly, CD4+ CD25hi ICOS+ T cells from CA subjects presented lower responsiveness toward anti-ICOS costimulation than CD4+ CD25hi ICOS+ T cells from healthy subjects. Overall, these results demonstrated that ICOS signalling could significantly improve FOXP3 expression and enhance Treg functional potency. However, circulating Treg cells from CA patients displayed reduced ICOS expression and lower responsiveness toward anti-ICOS stimulation.
Collapse
Affiliation(s)
- Ming-Guang Zhao
- Department of Neurosurgery, General Hospital of Northern Theater Command, Shenyang, China
| | - Cheng Peng
- Department of Neurosurgery, General Hospital of Northern Theater Command, Shenyang, China
| | - Li-Ming Li
- Institute of Biotechnology, College of Life and Health Sciences, Northeastern University, Shenyang, China
| | - Lu Chen
- Postgraduate Training Base of General Hospital of Northern Theater Command, Jinzhou Medical University, Shenyang, China
| | - Hai-Feng Zhang
- Department of Neurosurgery, General Hospital of Northern Theater Command, Shenyang, China
| |
Collapse
|
11
|
Savastio S, Cadario F, D'Alfonso S, Stracuzzi M, Pozzi E, Raviolo S, Rizzollo S, Gigliotti L, Boggio E, Bellomo G, Basagni C, Bona G, Rabbone I, Dianzani U, Prodam F. Vitamin D Supplementation Modulates ICOS+ and ICOS- Regulatory T Cell in Siblings of Children With Type 1 Diabetes. J Clin Endocrinol Metab 2020; 105:5897243. [PMID: 32844222 DOI: 10.1210/clinem/dgaa588] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Accepted: 08/21/2020] [Indexed: 02/07/2023]
Abstract
OBJECTIVES Vitamin D plays an immunoregulatory activity. The aim of this study was to assess the correlation between blood serum 25(OH)D levels and Th17 and Treg circulating subsets, mainly Treg/inducible costimulatory-positive (ICOS+), which seems to have a protective role in autoimmunity, in children with type 1 diabetes mellitus (T1D) and their healthy siblings (S). The secondary aim was to evaluate the impact of vitamin D supplementation on these subsets. PATIENTS AND METHODS 22 T1D and 33 S were enrolled. Glucose, hemoglobin A1c, 25 OH vitamin D (25[OH]D), T helper type 17 (Th17; CD4+CCR6+), regulatory T cells (Treg; CD4+CD25+Foxp3+), and Treg/ICOS+ cells were evaluated. According to human leukocyte antigen (HLA) haplotypes, subjects were classified as "at risk" (HLA+), "protective haplotypes" (HLA-; "nested controls"), and "undetermined" (HLAUND). T1D and S subjects were supplemented with cholecalciferol 1000 IU/die and evaluated after 6 months. RESULTS Vitamin D insufficiency (74.4%) and deficiency (43%) were frequent. S subjects with 25(OH)D levels <25 nmol/L had Th17, Treg (p < 0.01), and Treg/ICOS+ (P < 0.05) percentages higher than subjects with 25(OH)D >75 nmol/L. Treg/ICOS+ percentages (P < 0.05) were higher in HLA- S subjects compared to percentages observed in S with T1D. At baseline, in S subjects, a decreasing trend in Th17 and Treg/ICOS+ values (P < 0.05) from vitamin D deficiency to sufficiency was observed; 25(OH)D levels were negative predictors of Treg/ICOS+ (R2 = 0.301) and Th17 percentages (R2 = 0.138). After 6 months, supplemented S subjects showed higher 25(OH)D levels (P < 0.0001), and lower Th17 (P < 0.0001) and Treg/ICOS+ (P < 0.05) percentages than at baseline; supplemented T1D patients only had a decrease in Th17 levels (P < 0.05). CONCLUSION Serum 25(OH)D levels seem to affect Th17 and Treg cell subsets in S subjects, consistent with its immunomodulating role. HLA role should be investigated in a larger population.
Collapse
Affiliation(s)
- Silvia Savastio
- SCDU of Pediatrics, University Hospital Maggiore della Carità, Novara, Italy
| | - Francesco Cadario
- SCDU of Pediatrics, University Hospital Maggiore della Carità, Novara, Italy
- Interdisciplinary Research Center of Autoimmune Diseases, Università del Piemonte Orientale, Novara, Italy
| | - Sandra D'Alfonso
- Department of Health Sciences, Università del Piemonte Orientale, Novara, Italy
| | - Marta Stracuzzi
- SCDU of Pediatrics, University Hospital Maggiore della Carità, Novara, Italy
| | - Erica Pozzi
- SCDU of Pediatrics, University Hospital Maggiore della Carità, Novara, Italy
| | - Silvia Raviolo
- SCDU of Pediatrics, University Hospital Maggiore della Carità, Novara, Italy
| | - Stefano Rizzollo
- SCDU of Pediatrics, University Hospital Maggiore della Carità, Novara, Italy
| | - Luca Gigliotti
- Department of Health Sciences, Università del Piemonte Orientale, Novara, Italy
| | - Elena Boggio
- Department of Health Sciences, Università del Piemonte Orientale, Novara, Italy
| | - Giorgio Bellomo
- Department of Health Sciences, Università del Piemonte Orientale, Novara, Italy
| | - Chiara Basagni
- Department of Health Sciences, Università del Piemonte Orientale, Novara, Italy
| | - Gianni Bona
- Department of Health Sciences, Università del Piemonte Orientale, Novara, Italy
| | - Ivana Rabbone
- SCDU of Pediatrics, University Hospital Maggiore della Carità, Novara, Italy
- Department of Health Sciences, Università del Piemonte Orientale, Novara, Italy
| | - Umberto Dianzani
- Interdisciplinary Research Center of Autoimmune Diseases, Università del Piemonte Orientale, Novara, Italy
- Department of Health Sciences, Università del Piemonte Orientale, Novara, Italy
- SCDU of Clinical Biochemistry, University Hospital Maggiore della Carità, Novara, Italy
| | - Flavia Prodam
- SCDU of Pediatrics, University Hospital Maggiore della Carità, Novara, Italy
- Interdisciplinary Research Center of Autoimmune Diseases, Università del Piemonte Orientale, Novara, Italy
- Department of Health Sciences, Università del Piemonte Orientale, Novara, Italy
| |
Collapse
|
12
|
Abstract
ICOSL/ICOS are costimulatory molecules pertaining to immune checkpoints; their binding transduces signals having anti-tumor activity. Osteopontin (OPN) is here identified as a ligand for ICOSL. OPN binds a different domain from that used by ICOS, and the binding induces a conformational change in OPN, exposing domains that are relevant for its functions. Here we show that in vitro, ICOSL triggering by OPN induces cell migration, while inhibiting anchorage-independent cell growth. The mouse 4T1 breast cancer model confirms these data. In vivo, OPN-triggering of ICOSL increases angiogenesis and tumor metastatization. The findings shed new light on ICOSL function and indicate that another partner beside ICOS may be involved; they also provide a rationale for developing alternative therapeutic approaches targeting this molecular trio. Davide Raineri, Chiara Dianzani et al. show that osteopontin binds ICOSL at a different domain than the one used by ICOS. Activation of ICOSL by osteopontin induces cell migration in vitro and tumor metastatization in a 4T1 breast cancer mouse model; highlighting the functional role of this interaction in cancer progression.
Collapse
|
13
|
Fan X, Wang J, Qin T, Zhang Y, Liu W, Jiang K, Huang D. Exosome miR-27a-3p secreted from adipocytes targets ICOS to promote antitumor immunity in lung adenocarcinoma. Thorac Cancer 2020; 11:1453-1464. [PMID: 32212417 PMCID: PMC7262893 DOI: 10.1111/1759-7714.13411] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Revised: 03/09/2020] [Accepted: 03/10/2020] [Indexed: 01/21/2023] Open
Abstract
Background The clinical benefit of immunotherapy has been limited to a small subset of patients with cancer. Several clinical trials with immune checkpoint inhibitors in multiple cancers have shown some improvement in obese patients. However, how obesity regulates the immune microenvironment remains unclear. Methods Bioinformatic analysis was used to discover immune microenvironmental‐related genes associated with body mass index (BMI). The expression of ICOS in tumor tissues was detected using western blot, immunohistochemistry, quantitative real‐time polymerase chain reaction (RT‐qPCR) and flow cytometry. RT‐qPCR was used to measure the expression of miR‐27a‐3p. The interaction between miR‐27a‐3p and ICOS was confirmed by dual‐luciferase reporter assay. Functional testing of T cells based on proliferation and interferon (IFN)‐gamma secretion was performed using ELISA and flow cytometry. Results ICOS, an immune microenvironment‐related gene, was significantly upregulated in obese patients with lung adenocarcinoma (LUAD). MiR‐27a‐3p showed a negative correlation with ICOS and suppressed the expression of ICOS. We determined that dipocyte‐derived exo‐miR‐27a‐3p could alter the tumor microenvironment by inhibiting ICOS+ T cell proliferation and IFN‐gamma secretion in vitro. Conclusions Adipocyte‐derived exo‐miR‐27a‐3p can inhibit ICOS+ T cell proliferation and IFN‐gamma secretion. The upregulation of ICOS+ T cell functions caused by the downregulation of miR‐27a‐3p in adipose tissue derived exosomes is one of the potential mechanisms for the improved efficacy of immunotherapy in obese LUAD patients.
Collapse
Affiliation(s)
- Xuehan Fan
- Department of Thoracic Oncology, National Clinical Research Center for Cancer, Tianjin Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Jingya Wang
- Department of Thoracic Oncology, National Clinical Research Center for Cancer, Tianjin Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Tingting Qin
- Department of Thoracic Oncology, National Clinical Research Center for Cancer, Tianjin Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Yujia Zhang
- Department of Thoracic Oncology, National Clinical Research Center for Cancer, Tianjin Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Wenting Liu
- Department of Thoracic Oncology, National Clinical Research Center for Cancer, Tianjin Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Kaiting Jiang
- Department of Thoracic Oncology, National Clinical Research Center for Cancer, Tianjin Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Dingzhi Huang
- Department of Thoracic Oncology, National Clinical Research Center for Cancer, Tianjin Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| |
Collapse
|
14
|
Clemente N, Boggio E, Gigliotti LC, Raineri D, Ferrara B, Miglio G, Argenziano M, Chiocchetti A, Cappellano G, Trotta F, Caldera F, Capucchio MT, Yagi J, Rojo MJ, Renò F, Cavalli R, Dianzani C, Dianzani U. Immunotherapy of experimental melanoma with ICOS-Fc loaded in biocompatible and biodegradable nanoparticles. J Control Release 2020; 320:112-124. [PMID: 31962094 DOI: 10.1016/j.jconrel.2020.01.030] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Revised: 01/15/2020] [Accepted: 01/17/2020] [Indexed: 01/18/2023]
Abstract
Inducible T-cell costimulator (ICOS) upon binding to its ligand (ICOSL) mediates adaptive immunity and antitumor response. Thus, antitumor therapies targeting the ICOS/ICOSL pathway hold great promise for cancer treatment. In this regard, ICOSL triggering by a soluble recombinant form of ICOS (ICOS-Fc) hampered adhesiveness and migration of dendritic, endothelial, and tumor cells in vitro. Furthermore, in vivo treatment with ICOS-Fc previously showed the capability to inhibit lung metastatization of ICOSL+ B16-F10 melanoma cells when injected intravenously in mice, but it failed to block the growth of established subcutaneous B16-F10 murine tumors. Thus, we asked whether passive targeting of solid tumors with ICOS-Fc-loaded biocompatible and biodegradable nanoparticles (NPs) could instead prove effectiveness in reducing tumor growth. Here, ICOS-Fc was loaded in two types of polymer nanoparticles, i.e. cross-linked β-cyclodextrin nanosponges (CDNS) and poly(lactic-co-glycolic acid) (PLGA) NPs and in vitro characterized. In vivo experiments showed that treatment of C57BL6/J mice with ICOS-Fc loaded into the two nanoformulations inhibits the growth of established subcutaneous B16-F10 tumors. This anticancer activity appears to involve both anti-angiogenic and immunoregulatory effects, as shown by decreased tumor vascularization and downmodulation of IL-10 and Foxp3, two markers of regulatory T cells (Tregs). Overall, the substantial in vivo anticancer activity of ICOS-Fc-loaded CDNS and PLGA NPs against different components of the tumor microenvironment makes these nanoformulations attractive candidates for future combination cancer therapy.
Collapse
Affiliation(s)
- Nausicaa Clemente
- Department of Health Sciences, Inter Interdisciplinary Research Center of Autoimmune Diseases, UPO, 28100 Novara, Italy
| | - Elena Boggio
- Department of Health Sciences, Inter Interdisciplinary Research Center of Autoimmune Diseases, UPO, 28100 Novara, Italy
| | - Luca Casimiro Gigliotti
- Department of Health Sciences, Inter Interdisciplinary Research Center of Autoimmune Diseases, UPO, 28100 Novara, Italy
| | - Davide Raineri
- Department of Health Sciences, Inter Interdisciplinary Research Center of Autoimmune Diseases, UPO, 28100 Novara, Italy; Center for Translational Research on Autoimmune and Allergic Disease-CAAD, Università del Piemonte Orientale, 28100 Novara, Italy
| | - Benedetta Ferrara
- Department of Scienza e Tecnologia del Farmaco, University of Torino, 10125 Torino, Italy
| | - Gianluca Miglio
- Department of Scienza e Tecnologia del Farmaco, University of Torino, 10125 Torino, Italy
| | - Monica Argenziano
- Department of Scienza e Tecnologia del Farmaco, University of Torino, 10125 Torino, Italy
| | - Annalisa Chiocchetti
- Department of Health Sciences, Inter Interdisciplinary Research Center of Autoimmune Diseases, UPO, 28100 Novara, Italy; Center for Translational Research on Autoimmune and Allergic Disease-CAAD, Università del Piemonte Orientale, 28100 Novara, Italy
| | - Giuseppe Cappellano
- Department of Health Sciences, Inter Interdisciplinary Research Center of Autoimmune Diseases, UPO, 28100 Novara, Italy; Center for Translational Research on Autoimmune and Allergic Disease-CAAD, Università del Piemonte Orientale, 28100 Novara, Italy
| | - Francesco Trotta
- Department of Chemistry, University of Torino, 10125 Torino, Italy
| | - Fabrizio Caldera
- Department of Chemistry, University of Torino, 10125 Torino, Italy
| | | | - Junji Yagi
- Department of Microbiology and Immunology, Tokyo Women's Medical University, Tokyo 108-8639, Japan
| | - Maria Josè Rojo
- Departamento de Medicina Celular y Molecular, Centro de Investigaciones Biologicas, Consejo Superior de Investigaciones Cientıficas, 28040 Madrid, Spain
| | - Filippo Renò
- Department of Health Sciences, Inter Interdisciplinary Research Center of Autoimmune Diseases, UPO, 28100 Novara, Italy
| | - Roberta Cavalli
- Department of Scienza e Tecnologia del Farmaco, University of Torino, 10125 Torino, Italy.
| | - Chiara Dianzani
- Department of Scienza e Tecnologia del Farmaco, University of Torino, 10125 Torino, Italy
| | - Umberto Dianzani
- Department of Health Sciences, Inter Interdisciplinary Research Center of Autoimmune Diseases, UPO, 28100 Novara, Italy; Center for Translational Research on Autoimmune and Allergic Disease-CAAD, Università del Piemonte Orientale, 28100 Novara, Italy
| |
Collapse
|
15
|
Amatore F, Gorvel L, Olive D. Role of Inducible Co-Stimulator (ICOS) in cancer immunotherapy. Expert Opin Biol Ther 2019; 20:141-150. [PMID: 31738626 DOI: 10.1080/14712598.2020.1693540] [Citation(s) in RCA: 63] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Introduction: The promotion of antitumor response by targeting co-stimulatory B7 superfamily members has become evident to create a new wave of cancer immunotherapy. Inducible Co-Stimulator (ICOS), which is expressed on activated T cells, gained interest in the translational medicine community.Areas covered: We performed an extensive literature review using the keywords 'ICOS' and 'cancer', and the Clinicaltrials.gov database for early phase clinical trials targeting ICOS. In this review, we highlight the dual role of ICOS in oncogenesis in different malignancies. We summarize the current state of knowledge about ICOS/ICOSL pathway targeting by immunotherapies.Expert opinion: Due to its multifaceted link with anti-tumor immunity, both antagonist and agonist antibodies might be of interest to target the ICOS/ICOSL pathway for tumor treatment. Indeed, ICOS activation might potentiate the effect of an inhibitory checkpoint blockade, while its neutralization could decrease the function of immunosuppressive Tregs and inhibit lymphoid tumor cells expressing Tfh markers.
Collapse
Affiliation(s)
- Florent Amatore
- Centre de recherche en Cancérologie de Marseille, INSERM U1068, CNRS U7258, Aix Marseille Université, Institut Paoli - Calmettes, Marseille, France
| | - Laurent Gorvel
- Centre de recherche en Cancérologie de Marseille, INSERM U1068, CNRS U7258, Aix Marseille Université, Institut Paoli - Calmettes, Marseille, France
| | - Daniel Olive
- Centre de recherche en Cancérologie de Marseille, INSERM U1068, CNRS U7258, Aix Marseille Université, Institut Paoli - Calmettes, Marseille, France
| |
Collapse
|
16
|
Chiocchetti A, Cappellano G, Dianzani U. To each his own: a personalized vaccine for metastatic melanoma. Gland Surg 2019; 8:329-333. [PMID: 31538056 PMCID: PMC6723019 DOI: 10.21037/gs.2017.11.03] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2017] [Accepted: 10/30/2017] [Indexed: 12/30/2022]
Affiliation(s)
- Annalisa Chiocchetti
- Department of Health Sciences, Interdisciplinary Research Center of Autoimmune Diseases, University of Piemonte Orientale, Novara, Italy
| | - Giuseppe Cappellano
- Department of Dermatology, Venereology and Allergology, Medical University of Innsbruck, Innsbruck, Austria
| | - Umberto Dianzani
- Department of Health Sciences, Interdisciplinary Research Center of Autoimmune Diseases, University of Piemonte Orientale, Novara, Italy
| |
Collapse
|
17
|
Montes-Casado M, Ojeda G, Aragoneses-Fenoll L, López D, de Andrés B, Gaspar ML, Dianzani U, Rojo JM, Portolés P. ICOS deficiency hampers the homeostasis, development and function of NK cells. PLoS One 2019; 14:e0219449. [PMID: 31283790 PMCID: PMC6613708 DOI: 10.1371/journal.pone.0219449] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Accepted: 06/24/2019] [Indexed: 01/02/2023] Open
Abstract
Signaling through the inducible costimulator ICOS is required for the homeostasis and function of various immune cell populations, with an outstanding role in the generation and maintenance of germinal centers. Very recently, it has been suggested that the clinical phenotype of ICOS-deficient patients is much broader than initially anticipated and the innate immune response might be also affected. However, the role of the ICOS/ICOS-Ligand axis in the homeostasis and development of innate NK cells is not known, and reports on its participation in NK cell activation are scarce. NK cells may express low levels of ICOS that are markedly enhanced upon activation. We show here that ICOS-deficient (ICOS-KO) mice present low NK cell numbers and defects in the homeostasis of these cells, with delayed maturation and altered expression of the developmental NK cell markers CD122, NK1.1, CD11b or CD27. Our experiments in mixed bone marrow chimera mice indicate that, both, cell-intrinsic defects of ICOS-KO NK and deficiencies in the milieu of these mice contribute to the altered phenotype. ICOS-deficient NK cells show impaired production of IFN-γ and cytotoxicity, and a final outcome of defects in NK cell-mediated effector function during the response to poly(I:C) or vaccinia virus infection in vivo. Interestingly, we show that murine innate cells like IL-2-cultured NK and bone marrow-derived dendritic cells can simultaneously express ICOS and ICOS-Ligand; both molecules are functional in NK intracellular signaling, enhancing early phosphorylation of Akt and Erk, or IFN-γ secretion in IL-2-activated NK cells. Our study shows the functional importance of the ICOS/ICOS-L pair in NK cell homeostasis, differentiation and activity and suggests novel therapeutic targets for NK manipulation.
Collapse
Affiliation(s)
- María Montes-Casado
- Centro Nacional de Microbiología, Instituto de Salud Carlos III (ISCIII), Majadahonda, Madrid, Spain
| | - Gloria Ojeda
- Centro Nacional de Microbiología, Instituto de Salud Carlos III (ISCIII), Majadahonda, Madrid, Spain
| | - Laura Aragoneses-Fenoll
- Centro Nacional de Microbiología, Instituto de Salud Carlos III (ISCIII), Majadahonda, Madrid, Spain
| | - Daniel López
- Centro Nacional de Microbiología, Instituto de Salud Carlos III (ISCIII), Majadahonda, Madrid, Spain
| | - Belén de Andrés
- Centro Nacional de Microbiología, Instituto de Salud Carlos III (ISCIII), Majadahonda, Madrid, Spain
| | - María Luisa Gaspar
- Centro Nacional de Microbiología, Instituto de Salud Carlos III (ISCIII), Majadahonda, Madrid, Spain
| | - Umberto Dianzani
- Interdisciplinary Research Center of Autoimmune Diseases (IRCAD) and Department of Health Sciences, University of Piemonte Orientale (UPO), Novara, Italy
| | - José M Rojo
- Centro de Investigaciones Biológicas, Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
| | - Pilar Portolés
- Centro Nacional de Microbiología, Instituto de Salud Carlos III (ISCIII), Majadahonda, Madrid, Spain
| |
Collapse
|
18
|
Wan Z, Lin Y, Zhao Y, Qi H. T
FH
cells in bystander and cognate interactions with B cells. Immunol Rev 2019; 288:28-36. [DOI: 10.1111/imr.12747] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2019] [Revised: 01/19/2019] [Accepted: 01/25/2019] [Indexed: 12/15/2022]
Affiliation(s)
- Zurong Wan
- Department of Basic Biomedical Sciences, School of Medicine, Laboratory of Dynamic Immunobiology, Tsinghua‐Peking Center for Life SciencesTsinghua University Beijing China
| | - Yihan Lin
- Department of Basic Biomedical Sciences, School of Medicine, Laboratory of Dynamic Immunobiology, Tsinghua‐Peking Center for Life SciencesTsinghua University Beijing China
| | - Yongshan Zhao
- Department of Basic Biomedical Sciences, School of Medicine, Laboratory of Dynamic Immunobiology, Tsinghua‐Peking Center for Life SciencesTsinghua University Beijing China
| | - Hai Qi
- Department of Basic Biomedical Sciences, School of Medicine, Laboratory of Dynamic Immunobiology, Tsinghua‐Peking Center for Life SciencesTsinghua University Beijing China
| |
Collapse
|
19
|
Amatore F, Gorvel L, Olive D. Inducible Co-Stimulator (ICOS) as a potential therapeutic target for anti-cancer therapy. Expert Opin Ther Targets 2018; 22:343-351. [PMID: 29468927 DOI: 10.1080/14728222.2018.1444753] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
INTRODUCTION The recent success of checkpoint-inhibitors in cancer treatment paved the way for the development of new strategies of agonist and antagonist agents against B7 superfamily members. Inducible Co-Stimulator (ICOS), a co-stimulatory receptor for T-cell enhancement, arouses interest. Areas covered: We performed an extensive literature search with PUBMED using the keywords 'ICOS' and 'cancer' to discuss its involvement in oncogenesis, its expression in different malignancies, and its targeting in relevant preclinical studies. We also searched the Clinicaltrials.gov database for recent updates on early phase clinical trials. Expert opinion: ICOS/ICOSL axis has a dual effect and might participate in anti-tumour T cell response as well as a pro-tumour response due to its connection with regulatory T-cells (Tregs) suppressive activity. Therefore, both antagonist and agonist antibodies might be of interest in the targeting ICOS/ICOSL pathway for cancer treatment. In preclinical studies, ICOS agonist monoclonal antibodies (mAbs) have shown to potentiate the effect of inhibitory checkpoint blockade. In contrast, antagonistic anti-ICOS mAbs could not only inhibit lymphoid tumour cells expressing ICOS, but also dampen immunosuppressive Tregs. Two agonist and one antagonist mAbs are evaluated in phase I/II trials. Efficacy, safety, and combination strategies with anti-ICOS agonist or antagonist have yet to be specified.
Collapse
Affiliation(s)
- Florent Amatore
- a Centre de recherche en Cancérologie de Marseille, INSERM U1068, CNRS U7258 , Aix Marseille Université, Institut Paoli - Calmettes , Marseille , France
| | - Laurent Gorvel
- a Centre de recherche en Cancérologie de Marseille, INSERM U1068, CNRS U7258 , Aix Marseille Université, Institut Paoli - Calmettes , Marseille , France
| | - Daniel Olive
- a Centre de recherche en Cancérologie de Marseille, INSERM U1068, CNRS U7258 , Aix Marseille Université, Institut Paoli - Calmettes , Marseille , France
| |
Collapse
|
20
|
Aragoneses-Fenoll L, Ojeda G, Montes-Casado M, Acosta-Ampudia Y, Dianzani U, Portolés P, Rojo JM. T-Cell-Specific Loss of the PI-3-Kinase p110α Catalytic Subunit Results in Enhanced Cytokine Production and Antitumor Response. Front Immunol 2018. [PMID: 29535720 PMCID: PMC5835342 DOI: 10.3389/fimmu.2018.00332] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Class IA phosphatidylinositol 3-kinase (PI3K) catalytic subunits p110α and p110δ are targets in cancer therapy expressed at high levels in T lymphocytes. The role of p110δ PI3K in normal or pathological immune responses is well established, yet the importance of p110α subunits in T cell-dependent immune responses is not clear. To address this problem, mice with p110α conditionally deleted in CD4+ and CD8+ T lymphocytes (p110α-/-ΔT) were used. p110α-/-ΔT mice show normal development of T cell subsets, but slightly reduced numbers of CD4+ T cells in the spleen. "In vitro," TCR/CD3 plus CD28 activation of naive CD4+ and CD8+ p110α-/-ΔT T cells showed enhanced effector function, particularly IFN-γ secretion, T-bet induction, and Akt, Erk, or P38 activation. Tfh derived from p110α-/-ΔT cells also have enhanced responses when compared to normal mice, and IL-2 expanded p110α-/-ΔT CD8+ T cells had enhanced levels of LAMP-1 and Granzyme B. By contrast, the expansion of p110α-/-ΔT iTreg cells was diminished. Also, p110α-/-ΔT mice had enhanced anti-keyhole limpet hemocyanin (KLH) IFN-γ, or IL-4 responses and IgG1 and IgG2b anti-KLH antibodies, using CFA or Alum as adjuvant, respectively. When compared to WT mice, p110α-/-ΔT mice inoculated with B16.F10 melanoma showed delayed tumor progression. The percentage of CD8+ T lymphocytes was higher and the percentage of Treg cells lower in the spleen of tumor-bearing p110α-/-ΔT mice. Also, IFN-γ production in tumor antigen-activated spleen cells was enhanced. Thus, PI3K p110α plays a significant role in antigen activation and differentiation of CD4+ and CD8+ T lymphocytes modulating antitumor immunity.
Collapse
Affiliation(s)
- Laura Aragoneses-Fenoll
- Unidad de Inmunología Celular, Centro Nacional de Microbiología, Instituto de Salud Carlos III, Majadahonda, Madrid, Spain
| | - Gloria Ojeda
- Unidad de Inmunología Celular, Centro Nacional de Microbiología, Instituto de Salud Carlos III, Majadahonda, Madrid, Spain
| | - María Montes-Casado
- Unidad de Inmunología Celular, Centro Nacional de Microbiología, Instituto de Salud Carlos III, Majadahonda, Madrid, Spain
| | - Yeny Acosta-Ampudia
- Departamento de Medicina Celular y Molecular, Centro de Investigaciones Biológicas, CSIC, Madrid, Spain
| | - Umberto Dianzani
- Interdisciplinary Research Center of Autoimmune Diseases (IRCAD), Department of Health Sciences, University of Piemonte Orientale (UPO), Novara, Italy
| | - Pilar Portolés
- Unidad de Inmunología Celular, Centro Nacional de Microbiología, Instituto de Salud Carlos III, Majadahonda, Madrid, Spain
| | - José M Rojo
- Departamento de Medicina Celular y Molecular, Centro de Investigaciones Biológicas, CSIC, Madrid, Spain
| |
Collapse
|
21
|
Buus TB, Schmidt JD, Bonefeld CM, Geisler C, Lauritsen JPH. Development of interleukin-17-producing Vγ2+ γδ T cells is reduced by ICOS signaling in the thymus. Oncotarget 2017; 7:19341-54. [PMID: 27235509 PMCID: PMC4991387 DOI: 10.18632/oncotarget.8464] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2015] [Accepted: 03/21/2016] [Indexed: 11/25/2022] Open
Abstract
Co-stimulation is an integral part of T cell signaling involved in almost all facets of T cell biology. While much is known about co-stimulation in differentiation and function of conventional αβ T cells, less is known about how co-stimulation affects the development and programming of γδ T cells. In this study, we have investigated the role of inducible T cell co-stimulator (ICOS) on the development of γδ T cells. We show that ICOS is expressed by a population of immature Vγ2+CD45RBlow γδ T cells predisposed to interleukin-17 (IL-17) production. We found that treatment with ICOS specific antibodies drastically reduces fetal development of IL-17-producing γδ T cells by agonistic actions, and that ICOS deficient mice have a significant increase in the population of IL-17-producing Vγ2+ γδ T cells in the thymus, spleen, lymph nodes and skin and exhibit exacerbated sensitization responses to 2,4-dinitrofluorobenzene. In conclusion, this study demonstrates that development of IL-17-producing Vγ2+ γδ T cells is reduced by ICOS signaling in the thymus.
Collapse
Affiliation(s)
- Terkild Brink Buus
- Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Jonas Damgård Schmidt
- Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Charlotte Menné Bonefeld
- Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Carsten Geisler
- Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Jens Peter Holst Lauritsen
- Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
22
|
Osteopontin at the Crossroads of Inflammation and Tumor Progression. Mediators Inflamm 2017; 2017:4049098. [PMID: 28769537 PMCID: PMC5523273 DOI: 10.1155/2017/4049098] [Citation(s) in RCA: 118] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2017] [Accepted: 06/04/2017] [Indexed: 12/13/2022] Open
Abstract
Complex interactions between tumor and host cells regulate systemic tumor dissemination, a process that begins early at the primary tumor site and goes on until tumor cells detach themselves from the tumor mass and start migrating into the blood or lymphatic vessels. Metastatic cells colonize the target organs and are capable of surviving and growing at distant sites. In this context, osteopontin (OPN) appears to be a key determinant of the crosstalk between cancer cells and the host microenvironment, which in turn modulates immune evasion. OPN is overexpressed in several human carcinomas and has been implicated in inflammation, tumor progression, and metastasis. Thus, it represents one of the most attracting targets for cancer therapy. Within the tumor mass, OPN is secreted in various forms either by the tumor itself or by stroma cells, and it can exert either pro- or antitumorigenic effects according to the cell type and tumor microenvironment. Thus, targeting OPN for therapeutic purposes needs to take into account the heterogeneous functions of the multiple OPN forms with regard to cancer formation and progression. In this review, we will describe the role of systemic, tumor-derived, and stroma-derived OPN, highlighting its pivotal role at the crossroads of inflammation and tumor progression.
Collapse
|
23
|
Gigliotti CL, Boggio E, Clemente N, Shivakumar Y, Toth E, Sblattero D, D’Amelio P, Isaia GC, Dianzani C, Yagi J, Rojo JM, Chiocchetti A, Boldorini R, Bosetti M, Dianzani U. ICOS-Ligand Triggering Impairs Osteoclast Differentiation and Function In Vitro and In Vivo. THE JOURNAL OF IMMUNOLOGY 2016; 197:3905-3916. [DOI: 10.4049/jimmunol.1600424] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/14/2016] [Accepted: 09/20/2016] [Indexed: 02/02/2023]
|
24
|
Aragoneses-Fenoll L, Montes-Casado M, Ojeda G, Acosta YY, Herranz J, Martínez S, Blanco-Aparicio C, Criado G, Pastor J, Dianzani U, Portolés P, Rojo JM. ETP-46321, a dual p110α/δ class IA phosphoinositide 3-kinase inhibitor modulates T lymphocyte activation and collagen-induced arthritis. Biochem Pharmacol 2016; 106:56-69. [PMID: 26883061 DOI: 10.1016/j.bcp.2016.02.005] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2015] [Accepted: 02/11/2016] [Indexed: 11/29/2022]
Abstract
Class IA phosphoinositide 3-kinases (PI3Ks) are essential to function of normal and tumor cells, and to modulate immune responses. T lymphocytes express high levels of p110α and p110δ class IA PI3K. Whereas the functioning of PI3K p110δ in immune and autoimmune reactions is well established, the role of p110α is less well understood. Here, a novel dual p110α/δ inhibitor (ETP-46321) and highly specific p110α (A66) or p110δ (IC87114) inhibitors have been compared concerning T cell activation in vitro, as well as the effect on responses to protein antigen and collagen-induced arthritis in vivo. In vitro activation of naive CD4(+) T lymphocytes by anti-CD3 and anti-CD28 was inhibited more effectively by the p110δ inhibitor than by the p110α inhibitor as measured by cytokine secretion (IL-2, IL-10, and IFN-γ), T-bet expression and NFAT activation. In activated CD4(+) T cells re-stimulated through CD3 and ICOS, IC87114 inhibited Akt and Erk activation, and the secretion of IL-2, IL-4, IL-17A, and IFN-γ better than A66. The p110α/δ inhibitor ETP-46321, or p110α plus p110δ inhibitors also inhibited IL-21 secretion by differentiated CD4(+) T follicular (Tfh) or IL-17-producing (Th17) helper cells. In vivo, therapeutic administration of ETP-46321 significantly inhibited responses to protein antigen as well as collagen-induced arthritis, as measured by antigen-specific antibody responses, secretion of IL-10, IL-17A or IFN-γ, or clinical symptoms. Hence, p110α as well as p110δ Class IA PI3Ks are important to immune regulation; inhibition of both subunits may be an effective therapeutic approach in inflammatory autoimmune diseases like rheumatoid arthritis.
Collapse
Affiliation(s)
- L Aragoneses-Fenoll
- Unidad de Inmunología Celular, Centro Nacional de Microbiología, Instituto de Salud Carlos III, Majadahonda, Madrid, Spain
| | - M Montes-Casado
- Unidad de Inmunología Celular, Centro Nacional de Microbiología, Instituto de Salud Carlos III, Majadahonda, Madrid, Spain
| | - G Ojeda
- Unidad de Inmunología Celular, Centro Nacional de Microbiología, Instituto de Salud Carlos III, Majadahonda, Madrid, Spain
| | - Y Y Acosta
- Departamento de Medicina Celular y Molecular, Centro de Investigaciones Biológicas, CSIC, Madrid, Spain
| | - J Herranz
- Departamento de Medicina Celular y Molecular, Centro de Investigaciones Biológicas, CSIC, Madrid, Spain
| | - S Martínez
- Experimental Therapeutics Programme, Spanish National Cancer Research Centre (CNIO), Spain
| | - C Blanco-Aparicio
- Experimental Therapeutics Programme, Spanish National Cancer Research Centre (CNIO), Spain
| | - G Criado
- Hospital 12 de Octubre, Instituto de Investigación Hospital 12 de Octubre (I+12), E-28041 Madrid, Spain
| | - J Pastor
- Experimental Therapeutics Programme, Spanish National Cancer Research Centre (CNIO), Spain
| | - U Dianzani
- Interdisciplinary Research Center of Autoimmune Diseases (IRCAD) and Department of Health Sciences, University of Piemonte Orientale (UPO), Novara, Italy
| | - P Portolés
- Unidad de Inmunología Celular, Centro Nacional de Microbiología, Instituto de Salud Carlos III, Majadahonda, Madrid, Spain.
| | - J M Rojo
- Departamento de Medicina Celular y Molecular, Centro de Investigaciones Biológicas, CSIC, Madrid, Spain.
| |
Collapse
|
25
|
Sakthivel P, Gereke M, Breithaupt A, Fuchs D, Gigliotti L, Gruber AD, Dianzani U, Bruder D. Attenuation of immune-mediated influenza pneumonia by targeting the inducible co-stimulator (ICOS) molecule on T cells. PLoS One 2014; 9:e100970. [PMID: 25029240 PMCID: PMC4100737 DOI: 10.1371/journal.pone.0100970] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2014] [Accepted: 06/01/2014] [Indexed: 11/24/2022] Open
Abstract
Inducible Co-stimulator (ICOS) plays a critical role in mediating T cell differentiation and function and is considered a key player in balancing T effector and T regulatory (Treg) cell responses. Here we show that activation of the ICOS signalling pathway during acute influenza A virus (IAV) infection by application of an agonistic ICOS antibody reduced the frequency of CD8+ T cells in the respiratory tract of IAV infected animals and delayed pathogen elimination. In line with this, immune-mediated influenza pneumonia was significantly ameliorated in mice that received ICOS agonist as indicated by significantly reduced alveolar infiltrations and bronchointerstitial pneumonia, while at the same time virus-related pathology remained unaffected. Importantly, ICOS agonist treatment resulted in expansion of CD4+Foxp3+ Tregs in IAV infected mice, which was associated with elevated levels of the immunosuppressive cytokine IL-10 in the alveolar space. Together, our findings suggest a prominent role of ICOS signaling during acute IAV infection by increasing the Treg/CD8+ T cell ratio with beneficial outcome on immune-mediated pneumonia and underline the suitability of ICOS as potential therapeutic target for immune intervention in those infectious conditions characterized by strong immunopathology rather than virus-mediated cytopathic effects.
Collapse
Affiliation(s)
- Priya Sakthivel
- Immune Regulation Group, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Marcus Gereke
- Immune Regulation Group, Helmholtz Centre for Infection Research, Braunschweig, Germany
- Infection Immunology Group, Institute of Medical Microbiology, Infection Control and Prevention, Otto-von-Guericke University, Magdeburg, Germany
| | - Angele Breithaupt
- Department of Veterinary Medicine, Institute of Veterinary Pathology, Free University, Berlin, Germany
| | - Dietmar Fuchs
- Division of Biological Chemistry, Biocenter, Innsbruck Medical University, Innsbruck, Austria
| | - Luca Gigliotti
- Department of Health Sciences and Interdisciplinary Research Center of Autoimmune Diseases, “A. Avogadro” University of Eastern Piedmont, Novara, Italy
| | - Achim D. Gruber
- Department of Veterinary Medicine, Institute of Veterinary Pathology, Free University, Berlin, Germany
| | - Umberto Dianzani
- Department of Health Sciences and Interdisciplinary Research Center of Autoimmune Diseases, “A. Avogadro” University of Eastern Piedmont, Novara, Italy
| | - Dunja Bruder
- Immune Regulation Group, Helmholtz Centre for Infection Research, Braunschweig, Germany
- Infection Immunology Group, Institute of Medical Microbiology, Infection Control and Prevention, Otto-von-Guericke University, Magdeburg, Germany
- * E-mail:
| |
Collapse
|
26
|
Dianzani C, Minelli R, Gigliotti CL, Occhipinti S, Giovarelli M, Conti L, Boggio E, Shivakumar Y, Baldanzi G, Malacarne V, Orilieri E, Cappellano G, Fantozzi R, Sblattero D, Yagi J, Rojo JM, Chiocchetti A, Dianzani U. B7h Triggering Inhibits the Migration of Tumor Cell Lines. THE JOURNAL OF IMMUNOLOGY 2014; 192:4921-31. [DOI: 10.4049/jimmunol.1300587] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
27
|
Mesturini R, Gigliotti CL, Orilieri E, Cappellano G, Soluri MF, Boggio E, Woldetsadik A, Dianzani C, Sblattero D, Chiocchetti A, Yagi J, Rojo JM, Dianzani U. Differential induction of IL-17, IL-10, and IL-9 in human T helper cells by B7h and B7.1. Cytokine 2013; 64:322-30. [PMID: 23778031 DOI: 10.1016/j.cyto.2013.05.021] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2012] [Revised: 05/07/2013] [Accepted: 05/23/2013] [Indexed: 01/02/2023]
Abstract
ICOS and CD28 are expressed by T cells and are involved in costimulation of cytokine production in T helper (TH) cells. ICOS binds B7h expressed by several cell types, whereas CD28 binds B7.1 and B7.2 expressed by activated antigen presenting cells. This work investigated the role of B7h and B7.1 in TH17 and TH9 cell differentiation by assessing activity of recombinant B7h-Fc and B7.1-Fc on human naïve TH cells activated in the presence of different combinations of exogenous cytokines. In the presence of TGF-β1 and IL-1β (TH17 promoting condition), B7h-Fc was more effective than B7.1-Fc in inducing IL-17A and IL-10 secretion, whereas B7.1-Fc was more effective in inducing IL-17F. Dual costimulation with B7h-Fc and B7.1-Fc displayed an intermediate pattern with predominance of IL-17F over IL-17A, secretion of high levels of IL-10, and secretion of IL-9 levels lower than those induced by B7.1-Fc alone. In the presence of TGF-β1 and IL-4 (TH9 promoting condition), B7h-Fc induced IL-17A only, whereas B7.1-Fc induced also IL-17F, IL-10, and high levels of IL-9. Experiments on memory TH cells showed that B7h-Fc mainly supported secretion of IL-17A and IL-10, whereas B7.1-Fc supported secretion of IL-17A, IL-17F, IL-10, and IL-9. These data indicate that B7h and B7.1 play different roles in modulation of TH17 and TH9 differentiation. This plasticity might be important in the immune response to pathogens and tumors, and in the development of autoimmune diseases, and should be taken in consideration in designing of immunotherapeutic protocols triggering ICOS or CD28.
Collapse
Affiliation(s)
- Riccardo Mesturini
- Interdisciplinary Research Center of Autoimmune Diseases (IRCAD) and Department of Health Sciences, "A. Avogadro" University of Eastern Piedmont, 28100 Novara, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Occhipinti S, Dianzani C, Chiocchetti A, Boggio E, Clemente N, Gigliotti CL, Soluri MF, Minelli R, Fantozzi R, Yagi J, Rojo JM, Sblattero D, Giovarelli M, Dianzani U. Triggering of B7h by the ICOS modulates maturation and migration of monocyte-derived dendritic cells. THE JOURNAL OF IMMUNOLOGY 2012; 190:1125-34. [PMID: 23275603 DOI: 10.4049/jimmunol.1201816] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
B7h, expressed by several cell types, binds ICOS expressed by activated T cells. We have previously shown that B7h triggering by ICOS-Fc inhibits human endothelial cell adhesiveness. This work investigated the effect of ICOS-Fc on human monocyte-derived dendritic cells (DCs). We found that DCs matured with LPS in the presence of ICOS-Fc (mDCs(ICOS)) produced greater amounts of IL-23 and IL-10, and promoted a higher secretion of IL-17A and IL-17F in MLCs than did those DCs matured with LPS alone (mDCs). Moreover, mDCs(ICOS) pulsed with the keyhole limpet hemocyanin Ag during the maturation phase were better stimulators of Ag-specific MHC class I-, but not class II-restricted T cells than mDCs. This was probably due to promotion of cross-presentation because it was not detected when the Flu-MA(58-66) Ag was directly loaded on already matured DCs and mDCs(ICOS). Finally, ICOS-Fc inhibited the adhesion of both immature DCs and mDCs to vascular and lymphoid endothelial cells, their migratory activity, and the expression of the Rac-1 activator β-Pix involved in cell motility. These data suggest that B7h stimulation modulates DC function with effects on their maturation and recruitment into tissues. This opens a novel view on the use of interactors of the ICOS:B7h system as immunomodulatory drugs.
Collapse
Affiliation(s)
- Sergio Occhipinti
- Department of Medicine and Experimental Oncology, University of Torino, 10126 Torino, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Acosta YY, Ojeda G, Zafra MP, Bernardone IS, Sánchez A, Dianzani U, Portolés P, Rojo JM. Dissociation of actin polymerization and lipid raft accumulation by ligation of the Inducible Costimulator (ICOS, CD278). ACTA ACUST UNITED AC 2012. [DOI: 10.1016/j.inmuno.2011.06.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
|
30
|
Acosta YY, Zafra MP, Ojeda G, Bernardone IS, Dianzani U, Portolés P, Rojo JM. Biased binding of class IA phosphatidyl inositol 3-kinase subunits to inducible costimulator (CD278). Cell Mol Life Sci 2011; 68:3065-79. [PMID: 21188463 PMCID: PMC11115116 DOI: 10.1007/s00018-010-0606-1] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2010] [Revised: 11/12/2010] [Accepted: 11/26/2010] [Indexed: 12/01/2022]
Abstract
To better understand T lymphocyte costimulation by inducible costimulator (ICOS; H4; CD278), we analyzed proteins binding to ICOS peptides phosphorylated at the Y(191)MFM motif. Phosphorylated ICOS binds class IA phosphatidyl inositol 3-kinase (PI3-K) p85α, p50-55α and p85β regulatory subunits and p110α, p110δ and p110β catalytic subunits. Intriguingly, T cells expressed high levels of both p110α or p110δ catalytic subunits, yet ICOS peptides, cell surface ICOS or PI3-kinase class IA regulatory subunits preferentially coprecipitated p110α catalytic subunits. Silencing p110α or p110δ partially inhibited Akt/PKB activation induced by anti-CD3 plus anti-ICOS antibodies. However, silencing p110α enhanced and silencing p110δ inhibited Erk activation. Both p110α- and p110δ-specific inhibitors blocked cytokine secretion induced by TCR/CD3 activation with or without ICOS costimulus, but only p110α inhibitors blocked ICOS-induced cell elongation. Thus, p110α and p110δ are essential to optimal T cell activation, but their abundance and activity differentially tune up distinct ICOS signaling pathways.
Collapse
Affiliation(s)
- Yenny Y. Acosta
- Departamento de Medicina Celular y Molecular, Centro de Investigaciones Biológicas, CSIC, Ramiro de Maeztu, 9, 28040 Madrid, Spain
| | - Maria Paz Zafra
- Departamento de Medicina Celular y Molecular, Centro de Investigaciones Biológicas, CSIC, Ramiro de Maeztu, 9, 28040 Madrid, Spain
| | - Gloria Ojeda
- Centro Nacional de Microbiología, Instituto de Salud Carlos III, Majadahonda, 28220 Madrid, Spain
| | - Ilaria Seren Bernardone
- Departamento de Medicina Celular y Molecular, Centro de Investigaciones Biológicas, CSIC, Ramiro de Maeztu, 9, 28040 Madrid, Spain
- Department of Medical Sciences, Interdisciplinary Research Center of Autoimmune Diseases (IRCAD), “A. Avogadro” University of Eastern Piedmont, 28100 Novara, Italy
| | - Umberto Dianzani
- Department of Medical Sciences, Interdisciplinary Research Center of Autoimmune Diseases (IRCAD), “A. Avogadro” University of Eastern Piedmont, 28100 Novara, Italy
| | - Pilar Portolés
- Centro Nacional de Microbiología, Instituto de Salud Carlos III, Majadahonda, 28220 Madrid, Spain
| | - Jose M. Rojo
- Departamento de Medicina Celular y Molecular, Centro de Investigaciones Biológicas, CSIC, Ramiro de Maeztu, 9, 28040 Madrid, Spain
| |
Collapse
|
31
|
Inducible costimulator promotes helper T-cell differentiation through phosphoinositide 3-kinase. Proc Natl Acad Sci U S A 2009; 106:20371-6. [PMID: 19915142 DOI: 10.1073/pnas.0911573106] [Citation(s) in RCA: 191] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The T-cell costimulatory receptors, CD28 and the inducible costimulator (ICOS), are required for the generation of follicular B helper T cells (T(FH)) and germinal center (GC) reaction. A common signal transducer used by CD28 and ICOS is the phosphoinositide 3-kinase (PI3K). Although it is known that CD28-mediated PI3K activation is dispensable for GC reaction, the role of ICOS-driven PI3K signaling has not been defined. We show here that knock-in mice that selectively lost the ability to activate PI3K through ICOS had severe defects in T(FH) generation, GC reaction, antibody class switch, and antibody affinity maturation. In preactivated CD4(+) T cells, ICOS delivered a potent PI3K signal that was critical for the induction of the key T(FH) cytokines, IL-21 and IL-4. Under the same settings, CD28 was unable to activate PI3K but supported a robust secondary expansion of T cells. Thus, our results demonstrate a nonredundant function of ICOS-PI3K pathway in the generation of T(FH) and suggest that CD28 and ICOS play differential roles during a multistep process of T(FH) differentiation.
Collapse
|
32
|
Vu F, Dianzani U, Ware CF, Mak T, Gommerman JL. ICOS, CD40, and lymphotoxin beta receptors signal sequentially and interdependently to initiate a germinal center reaction. THE JOURNAL OF IMMUNOLOGY 2008; 180:2284-93. [PMID: 18250437 DOI: 10.4049/jimmunol.180.4.2284] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Germinal center (GC) responses to T-dependent Ags require effective collaboration between Th cells, activated B cells, and follicular dendritic cells within a highly organized microenvironment. Studies using gene-targeted mice have highlighted nonredundant molecules that are key for initiating and maintaining the GC niche, including the molecules of the ICOS, CD40, and lymphotoxin (LT) pathways. Signaling through ICOS has multiple consequences, including cytokine production, expression of CD40L on Th cells, and differentiation into CXCR5(+) follicular Th cells, all of which are important in the GC reaction. We have therefore taken advantage of ICOS(-/-) mice to dissect which downstream elements are required to initiate the formation of GC. In the context of a T-dependent immune response, we found that GC B cells from ICOS(-/-) mice express lower levels of LTalphabeta compared with wild-type GC B cells in vivo, and stimulation of ICOS on T cells induces LTalphabeta on B cells in vitro. Administration of agonistic anti-LTbeta receptor Ab was unable to restore the GC response in ICOS(-/-) mice, suggesting that additional input from another pathway is required for optimal GC generation. In contrast, treatment with agonistic anti-CD40 Ab in vivo recovered GC networks and restored LTalphabeta expression on GC B cells in ICOS(-/-) mice, and this effect was dependent on LTbeta receptor signaling. Collectively, these data demonstrate that ICOS activation is a prerequisite for the up-regulation of LTalphabeta on GC B cells in vivo and provide a model for cooperation between ICOS, CD40, and LT pathways in the context of the GC response.
Collapse
Affiliation(s)
- Frances Vu
- Department of Immunology, Faculty of Medicine, University of Toronto, 1 King's College Circle, Toronto, Ontario, Canada
| | | | | | | | | |
Collapse
|
33
|
Sakiyama H, Kobayashi S, Dianzani U, Ogiuchi H, Kawashima M, Uchiyama T, Yagi J. Possible involvement of T cell co-stimulation in pustulosis palmaris et plantaris via the induction of inducible co-stimulator in chronic focal infections. J Dermatol Sci 2008; 50:197-207. [PMID: 18308514 DOI: 10.1016/j.jdermsci.2007.12.010] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2007] [Revised: 12/25/2007] [Accepted: 12/27/2007] [Indexed: 10/22/2022]
Abstract
BACKGROUND Inducible co-stimulator (ICOS) is a co-stimulatory receptor on activated T cells that provides the signals needed for Th1 and Th2 responses via its interaction with B7h. Chronic focal infections are closely related to pustulosis palmaris et plantaris (PPP), but the involvement of ICOS in PPP has not been clarified. OBJECTIVE To investigate the effectiveness of treatments for focal infections on PPP skin lesions and the involvement of ICOS-positive T cells at focal infection sites in the tonsils and in PPP lesional skin. METHODS In patients that had undergone a tonsillectomy or dental treatment, the clinical activities of PPP, both the skin lesions and pustulotic arthro-osteitis were followed for over 2 years. The expressions of ICOS and various other activation markers on T cells were examined in tonsil tissue from both PPP patients and non-PPP patients, and the expression levels in peripheral blood were also evaluated in PPP patients and healthy donors. ICOS-positive T cells and B7h expression in PPP and normal skin were examined immunohistochemically. RESULTS The above treatments for focal infections led to a dramatic and persistent improvement in the PPP skin lesions and pustulotic arthro-osteitis. The expression of ICOS, but not of other activation markers, was higher in tonsil tissues from PPP patients than in tonsil tissues from non-PPP patients. B7h was upregulated without numerous ICOS-positive T cell infiltrates in the skin lesions. CONCLUSION The activation of T cells via ICOS co-stimulation in focal infections likely triggers the skin and skeletal inflammation associated with PPP, resulting in tissue damage.
Collapse
Affiliation(s)
- Hiroko Sakiyama
- Department of Oral and [corrected] Maxillofacial Surgery, Tokyo Women's Medical University School of Medicine, Tokyo 162-8666, Japan
| | | | | | | | | | | | | |
Collapse
|
34
|
Rojo JM, Pini E, Ojeda G, Bello R, Dong C, Flavell RA, Dianzani U, Portolés P. CD4+ICOS+ T lymphocytes inhibit T cell activation 'in vitro' and attenuate autoimmune encephalitis 'in vivo'. Int Immunol 2008; 20:577-89. [PMID: 18310064 DOI: 10.1093/intimm/dxn016] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
The inducible co-stimulator (ICOS, CD278) is essential to the efficient development of normal and pathological immune reactions. Since ICOS-deficient mice have enhanced susceptibility to experimental allergic encephalomyelitis (EAE), we have functionally analyzed a CD4+ICOS+ population comprising 6-15% of all CD4+ T cells in secondary lymphoid organs of unmanipulated wild-type mice and checked for their ability to suppress EAE. In C57BL/6 mice, CD4+ICOS+ cells were a major source of cytokines including IFN-gamma, IL-2, IL-4, IL-10 or IL-17A. Upon activation, these cells showed preferentially enhanced production of IL-4 or IL-10 but inhibited IFN-gamma production. In contrast, CD4+ICOS- cells mainly produced IFN-gamma. Interestingly, CD4+ICOS+ cells partially suppressed the proliferation of CD4+ICOS- or CD4+CD25- lymphocytes 'in vitro' by an IL-10-dependent mechanism. Furthermore, CD4+ICOS+ activated and expanded under appropriate conditions yielded a population enriched in cells producing IL-10 and T(h)2 cytokines that also suppressed the proliferation of CD4+CD25- lymphocytes. CD4+ICOS+ cells, before or after expansion in vitro, reduced the severity of EAE when transferred to ICOS-deficient mice. In the same EAE model, lymph node cells from ICOS-deficient mice receiving ICOS+ cells showed reduced IL-17A production and enhanced IL-10 secretion upon antigen activation in vitro. Thus, naturally occurring CD4+ICOS+ cells, expanded or not in vitro, are functionally relevant cells able of protecting ICOS-deficient mice from severe EAE.
Collapse
Affiliation(s)
- Jose M Rojo
- Departamento de Inmunología, Centro de Investigaciones Biológicas, Consejo Superior de Investigaciones Científicas, Ramiro de Maeztu 9, E-28040 Madrid, Spain.
| | | | | | | | | | | | | | | |
Collapse
|
35
|
Castelli L, Comi C, Chiocchetti A, Nicola S, Mesturini R, Giordano M, D'Alfonso S, Cerutti E, Galimberti D, Fenoglio C, Tesser F, Yagi J, Rojo JM, Perla F, Leone M, Scarpini E, Monaco F, Dianzani U. ICOS gene haplotypes correlate with IL10 secretion and multiple sclerosis evolution. J Neuroimmunol 2007; 186:193-8. [PMID: 17481737 DOI: 10.1016/j.jneuroim.2007.03.022] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2006] [Revised: 03/22/2007] [Accepted: 03/28/2007] [Indexed: 12/23/2022]
Abstract
Human ICOS is a T cell costimulatory molecule supporting IL10 secretion. A pilot study investigating variations of the ICOS gene 3'UTR detected 8 polymorphisms forming three haplotypes (A, B, C). Haplotype-A and -C displayed the highest difference. Activated T cells from healthy AA homozygotes expressed significantly less ICOS and secreted more IL10 than AC heterozygotes, whereas AB heterozygotes displayed intermediate levels. Analysis of 441 multiple sclerosis patients and 793 controls showed that frequency of AA homozygosity was significantly lower in MS patients with relapsing-remitting onset (N=416) than in controls (OR=0.70). Moreover, AA patients with relapsing-remitting onset had lower relapse rate and multiple sclerosis severity score than non-AA patients.
Collapse
Affiliation(s)
- Luca Castelli
- Interdisciplinary Research Center of Autoimmune Diseases and Department of Medical Sciences, "A. Avogadro" University of Eastern Piedmont, Novara, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Mesturini R, Nicola S, Chiocchetti A, Bernardone IS, Castelli L, Bensi T, Ferretti M, Comi C, Dong C, Rojo JM, Yagi J, Dianzani U. ICOS cooperates with CD28, IL-2, and IFN-γ and modulates activation of human naïve CD4+ T cells. Eur J Immunol 2006; 36:2601-12. [PMID: 17013990 DOI: 10.1002/eji.200535571] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Several sets of data indicate that ICOS regulates cytokine production in activated T cells, but is less effective on naïve T cells. This work evaluates ICOS function in human naïve CD4+ T cells through an assessment of the effect of soluble forms of the ICOS and CD28 physiological ligands on activation driven by anti-CD3 mAb. ICOS strikingly potentiated secretion of IL-2, IFN-gamma, IL-10, and TNF-alpha, but not IL-4, promoted by optimal stimulation of CD3+CD28, and it was the key switching-factor of activation when cells received suboptimal stimulation of CD3+CD28 or stimulation of CD3 alone in the presence of exogenous IL-2. In these conditions, blockade of IL-2 and IFN-gamma showed that ICOS builds up a positive feedback loop with IFN-gamma, which required IL-2 and was inhibited by IL-4. By contrast, in the absence of CD28 triggering or exogenous IL-2, ICOS-induced costimulation mainly supported expression of TGF-beta1 and FoxP3 and differentiation of regulatory T cells capable to inhibit proliferation of naïve CD4+ T cells driven by allogeneic cells. These data suggest that ICOS favors differentiation of Th effector cells when cooperates with appropriate activation stimuli such as CD3+CD28 or CD3+IL-2, whereas it supports differentiation of regulatory T cells when costimulatory signals are insufficient.
Collapse
Affiliation(s)
- Riccardo Mesturini
- Interdisciplinary Research Center of Autoimmune Diseases and Department of Medical Sciences, "A. Avogadro" University of Eastern Piedmont at Novara, Novara, Italy
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Vidric M, Bladt AT, Dianzani U, Watts TH. Role for inducible costimulator in control of Salmonella enterica serovar Typhimurium infection in mice. Infect Immun 2006; 74:1050-61. [PMID: 16428752 PMCID: PMC1360312 DOI: 10.1128/iai.74.2.1050-1061.2006] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Inducible costimulator (ICOS) is expressed on activated T cells and plays a key role in sustaining and enhancing the effector function of CD4 T cells. Given the function of this molecule in sustaining T-cell responses, we reasoned that ICOS might play an important role in a prolonged infection model, such as Salmonella infection of mice. To test this hypothesis, wild-type (WT) and ICOS-deficient (ICOS-/-) mice were infected systemically with a Salmonella enterica serovar Typhimurium strain expressing the chicken ovalbumin gene (Salmonella-OVA). ICOS-/- mice exhibited greater splenomegaly than WT mice and showed delayed bacterial clearance. The acquired immune response in this model was slow to develop. Maximal T-cell responses to Salmonella-OVA were detected at 3 weeks postinfection in both WT and ICOS-/- mice. CD4 T-cell-dependent gamma interferon production and a class switch to immunoglobulin G2a were severely reduced in ICOS-/- mice. ICOS-/- mice also exhibited a substantial defect in antigen-specific CD8 T-cell responses. In vitro, the effect of anti-ICOS on CD8 T-cell division was greater when CD8 T cells rather than CD4 T cells expressed ICOS, suggesting that the in vivo effects of ICOS on CD8 T cells could be direct. Taken together, these studies show that ICOS plays a critical role in control of Salmonella infection in mice, with effects on antibody, Th1, and CD8 T-cell responses.
Collapse
Affiliation(s)
- Mariana Vidric
- Department of Immunology, University of Toronto, 1 King's College Circle, Toronto, Ontario, Canada M5S 1A8
| | | | | | | |
Collapse
|
38
|
Vidric M, Suh WK, Dianzani U, Mak TW, Watts TH. Cooperation between 4-1BB and ICOS in the immune response to influenza virus revealed by studies of CD28/ICOS-deficient mice. THE JOURNAL OF IMMUNOLOGY 2006; 175:7288-96. [PMID: 16301634 DOI: 10.4049/jimmunol.175.11.7288] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
CD28, ICOS, and 4-1BB each play distinct roles in the CD8 T cell response to influenza virus. CD28-/- mice are severely impaired in primary CD8 T cell expansion and fail to mount a secondary response to influenza. Influenza-specific CD8 T cells expand normally in ICOS-/- mice, with only a small and transient defect late in the primary response and an unimpaired secondary response. Conversely, 4-1BB/4-1BBL interaction is dispensable for the primary CD8 T cell response to influenza, but maintains CD8 T cell survival and controls the size of the secondary response. Previous results showed that a single dose of agonistic anti-4-1BB Ab at priming allowed partial restoration of primary CD8 T cell expansion and full recovery of the secondary CD8 T cell responses to influenza in CD28-/- mice. In this study we show that anti-4-1BB fails to correct the CD8 T cell defect in CD28-/-ICOS-/- mice, suggesting that ICOS partially compensates for CD28 in this model. In support of this hypothesis, we found that anti-4-1BB enhances ICOS expression on both T cell subsets and that anti-4-1BB and anti-ICOS can synergistically activate CD4 and CD8 T cells. Furthermore, ICOS and 4-1BB can cooperate to directly stimulate isolated CD28-/- CD8 T cells. These results reveal a novel interaction between the ICOS and 4-1BB costimulatory pathways as well as unexpected redundancy between CD28 and ICOS in primary CD8 T cell expansion. These findings have implications for costimulation of human T cell responses in diseases such as AIDS or rheumatoid arthritis, in which CD28- T cells accumulate.
Collapse
Affiliation(s)
- Mariana Vidric
- Department of Immunology, University of Toronto, Ontario, Canada
| | | | | | | | | |
Collapse
|
39
|
Yagi J, Arimura Y, Dianzani U, Uede T, Okamoto T, Uchiyama T. Regulatory roles of IL-2 and IL-4 in H4/inducible costimulator expression on activated CD4+ T cells during Th cell development. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2003; 171:783-94. [PMID: 12847246 DOI: 10.4049/jimmunol.171.2.783] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
We found a tight correlation among the levels of H4/inducible costimulator (ICOS) expression, IL-4 production, and GATA-3 induction, using activated CD4(+) T cells obtained from six different murine strains. BALB/c-activated CD4(+) T cells expressed approximately 10-fold more H4/ICOS on their surfaces and produced approximately 10-fold more IL-4 upon restimulation than C57BL/6-activated CD4(+) T cells. BALB/c naive CD4(+) T cells were shown to produce much higher amounts of IL-2 and IL-4 upon primary stimulation than C57BL/6 naive CD4(+) T cells. Neutralization of IL-4 with mAbs in culture of BALB/c naive CD4(+) T cells strongly down-regulated both H4/ICOS expression on activated CD4(+) T cells and IL-4 production upon subsequent restimulation. Conversely, exogenous IL-4 added to the culture of BALB/c or C57BL/6 naive CD4(+) T cells up-regulated H4/ICOS expression and IL-4 production upon restimulation. In addition, retroviral expression of GATA-3 during the stimulation of naive CD4(+) T cells from C57BL/6 or IL-4(-/-) mice increased H4/ICOS expression on activated CD4(+) T cells. A similar effect of IL-2 in the primary culture of BALB/c naive CD4(+) T cells appeared to be mediated by IL-4, the production of which was regulated by IL-2. These data suggest that IL-4 induced by IL-2 is critical to the maintenance of high H4/ICOS expression on BALB/c-activated CD4(+) T cells.
Collapse
Affiliation(s)
- Junji Yagi
- Department of Microbiology and Immunology, Tokyo Women's Medical University School of Medicine, Tokyo, Japan.
| | | | | | | | | | | |
Collapse
|
40
|
Nurieva RI, Duong J, Kishikawa H, Dianzani U, Rojo JM, Ho IC, Flavell RA, Dong C. Transcriptional regulation of th2 differentiation by inducible costimulator. Immunity 2003; 18:801-11. [PMID: 12818161 DOI: 10.1016/s1074-7613(03)00144-4] [Citation(s) in RCA: 117] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Helper T (Th) cell differentiation is accompanied by complex transcriptional changes. Although costimulatory receptors are important in Th differentiation, the underlying mechanisms are poorly understood. Here we examine the transcriptional mechanisms by which ICOS regulates Th2 differentiation and selective IL-4 expression by effector T cells. We found impaired expression of c-Maf transcription factor functionally associated with the IL-4 defect in ICOS(-/-) cells. c-Maf expression in effector cells was regulated by IL-4 levels during Th differentiation. ICOS costimulation potentiated the T cell receptor (TcR)-mediated initial IL-4 production, possibly through the enhancement of NFATc1 expression. These data indicate that ICOS, by enhancing TcR signals at an early stage of T cell activation, regulates IL-4 transcription and T cell function in effector cells.
Collapse
Affiliation(s)
- Roza I Nurieva
- Department of Immunology, University of Washington School of Medicine, Seattle, WA 98195, USA
| | | | | | | | | | | | | | | |
Collapse
|
41
|
Dianzani U, Chiocchetti A, Ramenghi U. Role of inherited defects decreasing Fas function in autoimmunity. Life Sci 2003; 72:2803-24. [PMID: 12697265 DOI: 10.1016/s0024-3205(03)00196-6] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Fas is a death receptor belonging to the TNFR superfamily and induces cell apoptosis by both activating a caspase cascade and altering mitochondria. In the immune system, Fas is involved in the switching-off of the immune responses and cell mediated cytotoxicity. In humans, genetic defects decreasing Fas function cause the Autoimmune Lymphoproliferative Syndrome (ALPS) where autoimmunities are associated with accumulation of polyclonal lymphocytes in the secondary lymphoid tissues and expansion of T cells lacking both CD4 and CD8 (DN cells). Expansion of DN cells is absent in an ALPS variant, named Dianzani's Autoimmune Lymphoproliferative Disease (DALD). The observation that DALD patients' families display increased frequency of autoimmune diseases different from ALPS suggests that defects of Fas function may also play a role in development of "common" autoimmune diseases. This possibility is supported by detection of defective Fas function in substantial proportions of patients with the multiple autoimmune syndrome or aggressive forms of type 1 diabetes or multiple sclerosis. This article reviews data suggesting that development of autoimmune/lymphoproliferative patterns may involve several alterations hitting the Fas system, but might also involve alterations in other systems contributing to the switching-off or proliferation of lymphocytes.
Collapse
Affiliation(s)
- Umberto Dianzani
- Interdisciplinary Research Center of Autoimmune Diseases (IRCAD) and Department of Medical Sciences, "A. Avogadro" University of Eastern Piedmont, Via Solaroli 17, 28100, Novara, Italy.
| | | | | |
Collapse
|
42
|
Nurieva RI, Treuting P, Duong J, Flavell RA, Dong C. Inducible costimulator is essential for collagen-induced arthritis. J Clin Invest 2003; 111:701-6. [PMID: 12618524 PMCID: PMC151904 DOI: 10.1172/jci17321] [Citation(s) in RCA: 88] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
CD4(+) helper Th cells play a major role in the pathogenesis of rheumatoid arthritis. Th cell activation, differentiation, and immune function are regulated by costimulatory molecules. Inducible costimulator (ICOS) is a novel costimulatory receptor expressed on activated T cells. We, as well as others, recently demonstrated its importance in Th2 cytokine expression and Ab class switching by B cells. In this study, we examined the role of ICOS in rheumatoid arthritis using a collagen-induced arthritis model. We found that ICOS knockout mice on the DBA/1 background were completely resistant to collagen-induced arthritis and exhibited absence of joint tissue inflammation. These mice, when immunized with collagen, exhibited reduced anti-collagen IgM Ab's in the initial stage and IgG2a Ab's at the effector phase of collagen-induced arthritis. Furthermore, ICOS regulates the in vitro and in vivo expression of IL-17, a proinflammatory cytokine implicated in rheumatoid arthritis. These data indicate that ICOS is essential for collagen-induced arthritis and may suggest novel means for treating patients with rheumatoid arthritis.
Collapse
Affiliation(s)
- Roza I Nurieva
- Department of Immunology, and. Department of Comparative Medicine, University of Washington School of Medicine, Seattle, Washington, USA
| | | | | | | | | |
Collapse
|
43
|
Arimura Y, Kato H, Dianzani U, Okamoto T, Kamekura S, Buonfiglio D, Miyoshi-Akiyama T, Uchiyama T, Yagi J. A co-stimulatory molecule on activated T cells, H4/ICOS, delivers specific signals in T(h) cells and regulates their responses. Int Immunol 2002; 14:555-66. [PMID: 12039907 DOI: 10.1093/intimm/dxf022] [Citation(s) in RCA: 57] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
We examined the co-stimulatory activity of H4/ICOS on murine activated CD4(+) T cells and found that the cross-linking of H4/ICOS enhanced their proliferation, in addition to raising IFN-gamma, IL-4 and IL-10 production to levels comparable to those induced by CD28. However, IL-2 production was only marginally co-stimulated by H4/ICOS. This distinct pattern of lymphokine production appears to be induced by a specific intracellular signaling event. Compared with CD28, H4/ICOS dominantly elicited the Akt pathway via phosphatidylinositol 3-kinase. In addition, mitogen-activated protein kinase family kinases were activated in different ways by CD28 and H4/ICOS. The strong phosphorylation of p46 c-Jun N-terminal kinase was observed upon CD28 co-stimulation, but was less potently induced by H4/ICOS. The strain diversity in the induction of H4/ICOS was recognized. The expression of H4/ICOS on BALB/c activated CD4(+) T cells was >6-fold higher compared with C57BL/6 activated CD4(+) T cells. Furthermore, BALB/c activated CD4(+) T cells exhibited more T(h)2-deviated lymphokine production as compared with C57BL/6 activated CD4(+) T cells and signaling through H4/ICOS during the primary stimulation of naive CD4(+) T cells promoted the generation of T(h)2 cells. Thus, the difference in H4/ICOS expression on activated CD4(+) T cells, which is regulated among the mouse strains, may also regulate the polarization of T(h) cells.
Collapse
Affiliation(s)
- Yutaka Arimura
- Departments of Microbiology and Immunology, Tokyo Women's Medical University, 8-1 Kawada-cho, Shinjuku-ku, Tokyo 162-8666, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Sakamoto S, Tezuka K, Tsuji T, Hori N, Tamatani T. AILIM/ICOS: its expression and functional analysis with monoclonal antibodies. HYBRIDOMA AND HYBRIDOMICS 2002; 20:293-303. [PMID: 11839247 DOI: 10.1089/15368590152740699] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Activation-inducible lymphocyte immuno-mediatory molecule (AILIM/ICOS) is the third member of the co-stimulatory molecule CD28/CTLA-4 (CD152) family, and an inducible cell surface glycoprotein expressed on lymphocytes following activation. To determine the expression profile of the molecule, we generated monoclonal antibodies (MAbs) against human, rat, and mouse AILIM/ICOS. None of the MAbs bound to AILIM/ICOS of other species. The numbers of AILIM/ICOS-positive cells among human peripheral blood mononuclear cells (PBMC), and rat and mouse splenocytes were very low (0.5, 0.4, and 1.2%, respectively), and the cells included many CD4-positive T cells except in the case of rat. Rat AILIM/ICOS-positive cells among splenocytes included many CD45RA-positive B cells, although the expression on lymph node cells was similar to that on human PBMC and mouse splenocytes. Among rat thymocytes, the AILIM/ICOS expression was mainly localized on CD4- and CD8-double positive T cells. The binding of AILIM/ICOS to B7h-Ig, which is the ligand-Fc chimeric protein, was inhibited by all AILIM/ICOS-specific MAbs except for SG430. The potency of the co-stimulatory activity of CD3 and AILIM/ICOS as to T-cell proliferation was found to be substantial in human. Interestingly, the levels of stimulation with the two types of MAbs were equal to that with CD3 and CD28 despite the different functions of the two MAbs in the AILIM/ICOS-B7h interaction. On the other hand, the potencies in rat and mouse, although two independent MAbs were tested, were relatively lower than that of CD28-mediated co-stimulation.
Collapse
Affiliation(s)
- S Sakamoto
- Pharmaceutical Frontier Research Laboratories, JT Inc., 13-2, Fukuura 1-chome, Kanazawa-ku, Yokohama 236-0004, Japan.
| | | | | | | | | |
Collapse
|
45
|
Buonfiglio D, Bragardo M, Redoglia V, Vaschetto R, Bottarel F, Bonissoni S, Bensi T, Mezzatesta C, Janeway CA, Dianzani U. The T cell activation molecule H4 and the CD28-like molecule ICOS are identical. Eur J Immunol 2000; 30:3463-7. [PMID: 11093165 DOI: 10.1002/1521-4141(2000012)30:12<3463::aid-immu3463>3.0.co;2-5] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
The recently cloned CD28-like molecule ICOS displays striking similarities with H4, characterized some years ago in the mouse and recently in humans. Both molecules are selectively expressed by activated and germinal center T cells, display similar structure, and display co-stimulatory activities. H4 displays lateral association with the CD3/TCR and is expressed by mature thymocytes. In the mouse, H4 is also expressed at high levels by thymic NKT cells that are resistant to negative selection. The aim of this work was to evaluate whether H4 and ICOS are the same molecule using the C398.4A (binding human and mouse H4) and F44 (binding human ICOS) monoclonal antibody (mAb) in parallel experiments on human T cells. ICOS and H4 displayed the same expression pattern in a panel of T cell lines and the same expression kinetics in phytohemagglutinin-activated T cells. C398.4A completely blocked cell staining by F44, whereas F44 partially blocked C398.4A. H4 and ICOS immunoprecipitates displayed identical SDS-PAGE patterns and H4 immunoprecipitation completely removed ICOS from cell lysates. Finally, the C398.4A mAb specifically stained cells transfected with the human or mouse ICOS. These data prove that H4 and ICOS are the same molecule and that F44 and C398.4A bind partially different epitopes.
Collapse
Affiliation(s)
- D Buonfiglio
- Department of Medical Sciences, "A. Avogadro" University of Eastern Piedmont at Novara, Novara, Italy
| | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Lucia MB, Buonfiglio D, Bottarel F, Bensi T, Rutella S, Rumi C, Ortona L, Janeway CA, Cauda R, Dianzani U. Expression of the novel T cell activation molecule hpH4 in HIV-infected patients: correlation with disease status. AIDS Res Hum Retroviruses 2000; 16:549-57. [PMID: 10777145 DOI: 10.1089/088922200308963] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
We have described hpH4, a surface glycoprotein selectively expressed by activated T cells and mature thymocytes and displaying weak lateral association with CD4. The hpH4 expression pattern and biochemical features, together with analysis of its tryptic digest by peptide mass searching using MALDI-MS, suggested that it is a novel molecule. The aim of this work was to evaluate the peripheral blood T cell expression of hpH4 in HIV-infected patients and the interplay between HIV gp120 and hpH4, since both molecules interact with CD4. hpH4 expression during HIV-1 infection was evaluated by assessing 55 patients at various disease stages and following up 3 patients with primary infection and 3 patients with AIDS. hpH4 expression displayed a peak in the early phase of primary infection, dropped to control levels in the asymptomatic phase, and was newly expressed, at low levels, as AIDS developed. The expression kinetics were different than those shown by HLA-DR, CD25, and CD38. The most striking findings were the transient hpH4 expression peak displayed in the earliest stage, which was unique for hpH4. Incubation of T cells from normal donors with HIV gp120 induced transient hpH4 expression in resting CD4+ T cells and potentiated the hpH4 lateral association with CD4 in activated T cells. Moreover, hpH4 triggering inhibited gp120-induced death of CD4+ cells. Therefore, H4 expression may be a response to avoid apoptosis induced by HIV products.
Collapse
Affiliation(s)
- M B Lucia
- Department of Infectious Diseases, Catholic University of Rome, Italy
| | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Hannier S, Triebel F. The MHC class II ligand lymphocyte activation gene-3 is co-distributed with CD8 and CD3-TCR molecules after their engagement by mAb or peptide-MHC class I complexes. Int Immunol 1999; 11:1745-52. [PMID: 10545478 DOI: 10.1093/intimm/11.11.1745] [Citation(s) in RCA: 56] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
Previous studies indicated that signaling through lymphocyte activation gene-3 (LAG-3), a MHC class II ligand, induced by multivalent anti-receptor antibodies led to unresponsiveness to TCR stimulation. Here, lateral distribution of the LAG-3 molecules and its topological relationship (mutual proximity) to the TCR, CD8, CD4, and MHC class I and II molecules were studied in the plasma membrane of activated human T cells in co-capping experiments and conventional fluorescence microscopy. Following TCR engagement by either TCR-specific mAb or MHC-peptide complex recognition in T-B cell conjugates, LAG-3 was found to be specifically associated with the CD3-TCR complex. Similarly, following CD8 engagement LAG-3 and CD8 were co-distributed on the cell surface while only a low percentage of CD4-capped cells displayed LAG-3 co-caps. In addition, LAG-3 was found to be associated with MHC class II (i.e. DR, DP and DQ) and partially with MHC class I molecules. The supramolecular assemblies described here between LAG-3, CD3, CD8 and MHC class II molecules may result from an organization in raft microdomains, a phenomenon known to regulate early events of T cell activation.
Collapse
Affiliation(s)
- S Hannier
- Laboratoire d'Immunologie Cellulaire, Institut Gustave-Roussy, and Laboratoire d'Immunologie des tumeurs, Faculté de Pharmacie, Université Paris XI, 92296 Chatenay-Malabry, France
| | | |
Collapse
|
48
|
Yagi J, Dianzani U, Kato H, Okamoto T, Katsurada T, Buonfiglio D, Miyoshi-Akiyama T, Uchiyama T. Identification of a New Type of Invariant Vα14+ T Cells and Responsiveness to a Superantigen, Yersinia pseudotuberculosis- Derived Mitogen. THE JOURNAL OF IMMUNOLOGY 1999. [DOI: 10.4049/jimmunol.163.6.3083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Abstract
We examined the expression of the H4 T cell activation marker in thymic T cell subpopulations and found that TCR-αβ+ CD4+ thymic T cells are segregated into three subpopulations based upon H4 levels. Thymic T cells with either no or low H4 expression differentiate via the mainstream differentiation pathway in the thymus. H4int thymic T cells, which express a skewed Vβ repertoire of Vβ2, -7, and -8 in their TCRs, show the phenotype of NKT cells: CD44high, Ly6Chigh, NK1.1+, and TCR-αβlow. H4high thymic T cells also show a skewed Vβ repertoire, Vβ2, -7, and -8, and predominantly express an invariant Vα14-Jα281+ α-chain in their TCRs but constitute a distinct population in that they are CD44int, Ly6C−, NK1.1−, and TCR-αβhigh. Thus, invariant Vα14+ thymic T cells consist of ordinary NKT cells and a new type of T cell population. Vβ7+ and Vβ8.1+ invariant Vα14+ thymic T cells are present in DBA/2 mice, which carry mammary tumor virus-7-encoded superantigens, in comparable levels to those in BALB/c mice. Furthermore, Vβ7+ invariant Vα14+ thymic T cells in DBA/2 mice are in the immunologically responsive state, and Yersinia pseudotuberculosis-derived mitogen-induced Vβ7+ invariant Vα14+ thymic T cell blasts from DBA/2 and BALB/c mice exhibited equally enhanced responses upon restimulation with Y. pseudotuberculosis-derived mitogen. Thus, invariant Vα14+ thymic T cells that escape negative selection in DBA/2 mice contain T cells as functionally mature as those in BALB/c mice.
Collapse
Affiliation(s)
| | - Umberto Dianzani
- § Laboratory of Immunology, Department of Medical Science, A. Avogadro University of Eastern Piedmont, Novara, Italy
| | | | | | | | - Donatella Buonfiglio
- § Laboratory of Immunology, Department of Medical Science, A. Avogadro University of Eastern Piedmont, Novara, Italy
| | | | - Takehiko Uchiyama
- *Microbiology and Immunology and
- ‡ Institute of Laboratory Animals, Tokyo Women’s Medical University, Tokyo, Japan; and
| |
Collapse
|
49
|
Buonfiglio D, Bragardo M, Bonissoni S, Redoglia V, Cauda R, Zupo S, Burgio VL, Wolff H, Franssila K, Gaidano G, Carbone A, Janeway CA, Dianzani U. Characterization of a novel human surface molecule selectively expressed by mature thymocytes, activated T cells and subsets of T cell lymphomas. Eur J Immunol 1999; 29:2863-74. [PMID: 10508261 DOI: 10.1002/(sici)1521-4141(199909)29:09<2863::aid-immu2863>3.0.co;2-w] [Citation(s) in RCA: 21] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
We have previously characterized mouse H4 (mH4), a surface glycoprotein recognized by the C398.4A monoclonal antibody. We now show that C398.4A also binds its human putative homolog (hpH4). Both hpH4 and mH4 (1) are selectively expressed by activated T cells and mature thymocytes, (2) are disulfide-linked dimers of two chains (29/37 kDa in humans, 25/29 kDa in mice), whose N-deglycosylation produces a single band at 20 - 21 kDa, and (3) display a low association with CD4 and the TCR. The expression pattern of hpH4 and its biochemical features showed that it is different from other known activation molecules, and this was confirmed when analysis of the tryptic digest of the hpH4 29-kDa band by peptide mass searching using matrix-assisted laser desorption ionization mass spectrometry did not reveal any significant homology with other molecules. In normal lymphoid tissue, hpH4 is expressed by T cells located at the periphery of lymph node germinal centers and paracortical areas. In T cell neoplasia, expression of hpH4 clusters with a subset of peripheral T cell lymphomas with a large-cell component, and with cases of angioimmunoblastic T cell lymphomas. Overall, these data provide evidence for a novel T cell activation molecule that could help in the phenotypic categorization of T cell malignancies.
Collapse
MESH Headings
- Animals
- Antibodies, Monoclonal/metabolism
- Antigens, Differentiation, T-Lymphocyte/biosynthesis
- Antigens, Differentiation, T-Lymphocyte/chemistry
- Antigens, Differentiation, T-Lymphocyte/immunology
- Cell Differentiation/immunology
- Humans
- Lymphocyte Activation/immunology
- Lymphoid Tissue/immunology
- Lymphoid Tissue/metabolism
- Lymphoid Tissue/pathology
- Lymphoma, T-Cell/immunology
- Lymphoma, T-Cell/metabolism
- Mice
- Organ Specificity/immunology
- Sequence Homology, Amino Acid
- T-Lymphocyte Subsets/immunology
- T-Lymphocyte Subsets/metabolism
- T-Lymphocytes/cytology
- T-Lymphocytes/immunology
- T-Lymphocytes/metabolism
- Thymus Gland/cytology
- Thymus Gland/immunology
- Thymus Gland/metabolism
- Tumor Cells, Cultured
Collapse
Affiliation(s)
- D Buonfiglio
- Department of Medical Sciences, A. Avogadro" University of Eastern Piedmont at Novara, Novara, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|