1
|
Li X, Li J, Zheng Y, Lee SJ, Zhou J, Giobbie-Hurder A, Butterfield LH, Dranoff G, Hodi FS. Granulocyte-Macrophage Colony-Stimulating Factor Influence on Soluble and Membrane-Bound ICOS in Combination with Immune Checkpoint Blockade. Cancer Immunol Res 2023; 11:1100-1113. [PMID: 37262321 PMCID: PMC10398357 DOI: 10.1158/2326-6066.cir-22-0702] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Revised: 01/03/2023] [Accepted: 05/23/2023] [Indexed: 06/03/2023]
Abstract
With the successful development of immune checkpoint blockade, there remains the continued need to improve efficacy and decrease toxicities. The addition of granulocyte-macrophage colony-stimulating factor (GM-CSF) to ipilimumab has previously demonstrated both an improvement in efficacy and decrease in the incidence of high-grade adverse events. ICOS+CD4+ or ICOS+CD8+ peripheral blood T cells are significantly greater in the patients treated with ipilimumab plus GM-CSF than in the patients treated with ipilimumab alone. To better understand the effects of GM-CSF on inducible T-cell costimulator (ICOS) and clinical outcomes, the relative roles of identified soluble ICOS and membrane-bound ICOS were evaluated. The ICOS splice variant was secreted and found to have immunologic suppressive effects. Changes in soluble ICOS splice variant levels in treated patients correlated with clinical outcomes. GM-CSF enhanced membrane-bound ICOS in an IL12-dependent manner but did not increase soluble ICOS levels. Whereas soluble ICOS plays a role in immune suppression, GM-CSF efficacy involves increasing membrane-bound ICOS and induction of dendritic cell development. Thus, soluble ICOS splice variants may be used as a biomarker for GM-CSF and immune checkpoint blockade-based therapies.
Collapse
Affiliation(s)
- Xiaoyu Li
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
- Melanoma Division, Dana-Farber Cancer Institute, Boston, Massachusetts
- Center for Immuno-Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Jingjing Li
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
- Melanoma Division, Dana-Farber Cancer Institute, Boston, Massachusetts
- Center for Immuno-Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Yue Zheng
- Department of Data Sciences, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Sandra J. Lee
- Department of Data Sciences, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Jun Zhou
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
- Melanoma Division, Dana-Farber Cancer Institute, Boston, Massachusetts
- Center for Immuno-Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Anita Giobbie-Hurder
- Melanoma Division, Dana-Farber Cancer Institute, Boston, Massachusetts
- Center for Immuno-Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
- Department of Data Sciences, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Lisa H. Butterfield
- Parker Institute for Cancer Immunotherapy and University of California San Francisco, San Francisco, California
| | - Glenn Dranoff
- Immuno-Oncology, Novartis Institutes for Biomedical Research, Cambridge, Massachusetts
| | - F. Stephen Hodi
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
- Melanoma Division, Dana-Farber Cancer Institute, Boston, Massachusetts
- Center for Immuno-Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| |
Collapse
|
2
|
Kobayashi S, Nagafuchi Y, Shoda H, Fujio K. The Pathophysiological Roles of Regulatory T Cells in the Early Phase of Systemic Sclerosis. Front Immunol 2022; 13:900638. [PMID: 35686127 PMCID: PMC9172592 DOI: 10.3389/fimmu.2022.900638] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Accepted: 04/26/2022] [Indexed: 11/13/2022] Open
Abstract
Systemic sclerosis (SSc) is an autoimmune disease that is characterized by vascular damage and fibrosis. Both clinical manifestations and immunological disturbances are diverse according to the disease duration. Particularly, changes in immunological processes are prominent in the early phase of SSc. The orchestration of several subsets of immune cells promotes autoimmune responses and inflammation, and eventually stimulates pro-fibrotic processes. Many reports have indicated that CD4+ T cells play pivotal roles in pathogenesis in the early phase of SSc. In particular, the pathogenic roles of regulatory T (Treg) cells have been investigated. Although the results were controversial, recent reports suggested an increase of Treg cells in the early phase of SSc patients. Treg cells secrete transforming growth factor-β (TGF-β), which promotes myofibroblast activation and fibrosis. In addition, the dysfunction of Treg cells in the early phase of SSc was reported, which results in the development of autoimmunity and inflammation. Notably, Treg cells have the plasticity to convert to T-helper17 (Th17) cells under pro-inflammatory conditions. Th17 cells secrete IL-17A, which could also promote myofibroblast transformation and fibrosis and contributes to vasculopathy, although the issue is still controversial. Our recent transcriptomic comparison between the early and late phases of SSc revealed a clear difference of gene expression patterns only in Treg cells. The gene signature of an activated Treg cell subpopulation was expanded in the early phase of SSc and the oxidative phosphorylation pathway was enhanced, which can promote Th17 differentiation. And this result was accompanied by the increase in Th17 cells frequency. Therefore, an imbalance between Treg and Th17 cells could also have an important role in the pathogenesis of the early phase of SSc. In this review, we outlined the roles of Treg cells in the early phase of SSc, summarizing the data of both human and mouse models. The contributions of Treg cells to autoimmunity, vasculopathy, and fibrosis were revealed, based on the dysfunction and imbalance of Treg cells. We also referred to the potential development in treatment strategies in SSc.
Collapse
Affiliation(s)
- Satomi Kobayashi
- Department of Allergy and Rheumatology, Graduate School of Medicine, The University of Tokyo, Bunkyo-ku, Japan
- Department of Medicine and Rheumatology, Tokyo Metropolitan Geriatric Hospital, Itabashi-ku, Japan
| | - Yasuo Nagafuchi
- Department of Allergy and Rheumatology, Graduate School of Medicine, The University of Tokyo, Bunkyo-ku, Japan
- Department of Functional Genomics and Immunological Diseases, Graduate School of Medicine, The University of Tokyo, Bunkyo-ku, Japan
| | - Hirofumi Shoda
- Department of Allergy and Rheumatology, Graduate School of Medicine, The University of Tokyo, Bunkyo-ku, Japan
| | - Keishi Fujio
- Department of Allergy and Rheumatology, Graduate School of Medicine, The University of Tokyo, Bunkyo-ku, Japan
| |
Collapse
|
3
|
A Mechanistic Insight into the Pathogenic Role of Interleukin 17A in Systemic Autoimmune Diseases. Mediators Inflamm 2022; 2022:6600264. [PMID: 35620115 PMCID: PMC9129985 DOI: 10.1155/2022/6600264] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 01/04/2022] [Accepted: 04/25/2022] [Indexed: 12/13/2022] Open
Abstract
Interleukin 17A (IL-17A) has been put forward as a strong ally in our fight against invading pathogens across exposed epithelial surfaces by serving an antimicrobial immunosurveillance role in these tissues to protect the barrier integrity. Amongst other mechanisms that prevent tissue injury mediated by potential microbial threats and promote restoration of epithelial homeostasis, IL-17A attracts effector cells to the site of inflammation and support the host response by driving the development of ectopic lymphoid structures. Accumulating evidence now underscores an integral role of IL-17A in driving the pathophysiology and clinical manifestations in three potentially life-threatening autoimmune diseases, namely, systemic lupus erythematosus, Sjögren’s syndrome, and systemic sclerosis. Available studies provide convincing evidence that the abundance of IL-17A in target tissues and its prime source, which is T helper 17 cells (Th17) and double negative T cells (DNT), is not an innocent bystander but in fact seems to be prerequisite for organ pathology. In this regard, IL-17A has been directly implicated in critical steps of autoimmunity. This review reports on the synergistic interactions of IL-17A with other critical determinants such as B cells, neutrophils, stromal cells, and the vasculature that promote the characteristic immunopathology of these autoimmune diseases. The summary of observations provided by this review may have empowering implications for IL-17A-based strategies to prevent clinical manifestations in a broad spectrum of autoimmune conditions.
Collapse
|
4
|
Bellan M, Murano F, Ceruti F, Piccinino C, Tonello S, Minisini R, Giubertoni A, Sola D, Pedrazzoli R, Maglione V, Manfredi GF, Acquaviva A, Piffero R, Patti G, Pirisi M, Sainaghi PP. Increased Levels of ICOS and ICOSL Are Associated to Pulmonary Arterial Hypertension in Patients Affected by Connective Tissue Diseases. Diagnostics (Basel) 2022; 12:diagnostics12030704. [PMID: 35328257 PMCID: PMC8947069 DOI: 10.3390/diagnostics12030704] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Revised: 03/04/2022] [Accepted: 03/09/2022] [Indexed: 12/07/2022] Open
Abstract
Background: Pulmonary hypertension (PH) is a life-threatening complication of connective tissue diseases (CTD); in this study, we aimed at investigating the potential role of inducible co-stimulator (ICOS) and its ligand (ICOS-L) as biomarkers of PH in CTD. Materials and Methods: We recruited 109 patients: 84 CTD patients, 13 patients with CTD complicated by pulmonary arterial hypertension (PAH), and 12 subjects with PAH alone. All recruited patients underwent a complete clinical and instrumental assessment along with quantitative measurement of serum ICOS and ICOS-L. Results: Independently of the underlying cause, patients with PAH were older and had a lower glomerular filtration rate. Interestingly, patients with both CTD-related and CTD-unrelated PAH had higher ICOS and ICOS-L serum concentrations than CTD patients (0.0001 for both). When compared to CTD patients, those affected by CTD-PAH showed higher ICOS (440 (240–600) vs. 170 (105–275) pg/mL, p = 0.0001) and ICOS-L serum concentrations (6000 (4300–7000) vs. 2450 (1500–4100) pg/mL; p = 0.0001). In a logistic regression, ICOS and ICOS-L were associated with a diagnosis of PAH, independently from age, gender, and renal function. The corresponding receiver operating characteristic (ROC) curves demonstrated a good diagnostic performance for both ICOS and ICOS-L. Conclusions: ICOS and ICOS-L are increased in patients with PAH, irrespectively from the underlying cause, and represent promising candidate biomarkers for the diagnostic screening for PAH among CTDs patients.
Collapse
Affiliation(s)
- Mattia Bellan
- Department of Translational Medicine (DiMeT), Università del Piemonte Orientale (UPO), 28100 Novara, Italy; (F.M.); (F.C.); (S.T.); (R.M.); (A.G.); (V.M.); (G.F.M.); (A.A.); (R.P.); (G.P.); (M.P.); (P.P.S.)
- Rheumatology Unit, Department of Internal Medicine, AOU Maggiore della Carità, 28100 Novara, Italy; (D.S.); (R.P.)
- Center for Translational Research on Autoimmune and Allergic Disease (CAAD), Università del Piemonte Orientale (UPO), 28100 Novara, Italy
- Correspondence: ; Tel.: +39-0321-3737512
| | - Francesco Murano
- Department of Translational Medicine (DiMeT), Università del Piemonte Orientale (UPO), 28100 Novara, Italy; (F.M.); (F.C.); (S.T.); (R.M.); (A.G.); (V.M.); (G.F.M.); (A.A.); (R.P.); (G.P.); (M.P.); (P.P.S.)
- Rheumatology Unit, Department of Internal Medicine, AOU Maggiore della Carità, 28100 Novara, Italy; (D.S.); (R.P.)
| | - Federico Ceruti
- Department of Translational Medicine (DiMeT), Università del Piemonte Orientale (UPO), 28100 Novara, Italy; (F.M.); (F.C.); (S.T.); (R.M.); (A.G.); (V.M.); (G.F.M.); (A.A.); (R.P.); (G.P.); (M.P.); (P.P.S.)
- Rheumatology Unit, Department of Internal Medicine, AOU Maggiore della Carità, 28100 Novara, Italy; (D.S.); (R.P.)
| | | | - Stelvio Tonello
- Department of Translational Medicine (DiMeT), Università del Piemonte Orientale (UPO), 28100 Novara, Italy; (F.M.); (F.C.); (S.T.); (R.M.); (A.G.); (V.M.); (G.F.M.); (A.A.); (R.P.); (G.P.); (M.P.); (P.P.S.)
| | - Rosalba Minisini
- Department of Translational Medicine (DiMeT), Università del Piemonte Orientale (UPO), 28100 Novara, Italy; (F.M.); (F.C.); (S.T.); (R.M.); (A.G.); (V.M.); (G.F.M.); (A.A.); (R.P.); (G.P.); (M.P.); (P.P.S.)
| | - Ailia Giubertoni
- Department of Translational Medicine (DiMeT), Università del Piemonte Orientale (UPO), 28100 Novara, Italy; (F.M.); (F.C.); (S.T.); (R.M.); (A.G.); (V.M.); (G.F.M.); (A.A.); (R.P.); (G.P.); (M.P.); (P.P.S.)
- Division of Cardiology, AOU Maggiore della Carità, 28100 Novara, Italy;
| | - Daniele Sola
- Rheumatology Unit, Department of Internal Medicine, AOU Maggiore della Carità, 28100 Novara, Italy; (D.S.); (R.P.)
| | - Roberta Pedrazzoli
- Rheumatology Unit, Department of Internal Medicine, AOU Maggiore della Carità, 28100 Novara, Italy; (D.S.); (R.P.)
| | - Veronica Maglione
- Department of Translational Medicine (DiMeT), Università del Piemonte Orientale (UPO), 28100 Novara, Italy; (F.M.); (F.C.); (S.T.); (R.M.); (A.G.); (V.M.); (G.F.M.); (A.A.); (R.P.); (G.P.); (M.P.); (P.P.S.)
| | - Giulia Francesca Manfredi
- Department of Translational Medicine (DiMeT), Università del Piemonte Orientale (UPO), 28100 Novara, Italy; (F.M.); (F.C.); (S.T.); (R.M.); (A.G.); (V.M.); (G.F.M.); (A.A.); (R.P.); (G.P.); (M.P.); (P.P.S.)
- Rheumatology Unit, Department of Internal Medicine, AOU Maggiore della Carità, 28100 Novara, Italy; (D.S.); (R.P.)
| | - Antonio Acquaviva
- Department of Translational Medicine (DiMeT), Università del Piemonte Orientale (UPO), 28100 Novara, Italy; (F.M.); (F.C.); (S.T.); (R.M.); (A.G.); (V.M.); (G.F.M.); (A.A.); (R.P.); (G.P.); (M.P.); (P.P.S.)
- Rheumatology Unit, Department of Internal Medicine, AOU Maggiore della Carità, 28100 Novara, Italy; (D.S.); (R.P.)
| | - Roberto Piffero
- Department of Translational Medicine (DiMeT), Università del Piemonte Orientale (UPO), 28100 Novara, Italy; (F.M.); (F.C.); (S.T.); (R.M.); (A.G.); (V.M.); (G.F.M.); (A.A.); (R.P.); (G.P.); (M.P.); (P.P.S.)
- Rheumatology Unit, Department of Internal Medicine, AOU Maggiore della Carità, 28100 Novara, Italy; (D.S.); (R.P.)
| | - Giuseppe Patti
- Department of Translational Medicine (DiMeT), Università del Piemonte Orientale (UPO), 28100 Novara, Italy; (F.M.); (F.C.); (S.T.); (R.M.); (A.G.); (V.M.); (G.F.M.); (A.A.); (R.P.); (G.P.); (M.P.); (P.P.S.)
- Division of Cardiology, AOU Maggiore della Carità, 28100 Novara, Italy;
| | - Mario Pirisi
- Department of Translational Medicine (DiMeT), Università del Piemonte Orientale (UPO), 28100 Novara, Italy; (F.M.); (F.C.); (S.T.); (R.M.); (A.G.); (V.M.); (G.F.M.); (A.A.); (R.P.); (G.P.); (M.P.); (P.P.S.)
- Rheumatology Unit, Department of Internal Medicine, AOU Maggiore della Carità, 28100 Novara, Italy; (D.S.); (R.P.)
- Center for Translational Research on Autoimmune and Allergic Disease (CAAD), Università del Piemonte Orientale (UPO), 28100 Novara, Italy
| | - Pier Paolo Sainaghi
- Department of Translational Medicine (DiMeT), Università del Piemonte Orientale (UPO), 28100 Novara, Italy; (F.M.); (F.C.); (S.T.); (R.M.); (A.G.); (V.M.); (G.F.M.); (A.A.); (R.P.); (G.P.); (M.P.); (P.P.S.)
- Rheumatology Unit, Department of Internal Medicine, AOU Maggiore della Carità, 28100 Novara, Italy; (D.S.); (R.P.)
- Center for Translational Research on Autoimmune and Allergic Disease (CAAD), Università del Piemonte Orientale (UPO), 28100 Novara, Italy
| |
Collapse
|
5
|
García-Espinoza JA, Muñoz-Valle JF, García-Chagollán M, Hernández-Bello J, Palafox-Sánchez CA, López-Villalobos EF, Sánchez-Zuno GA, Martínez-Bonilla GE, Cerpa-Cruz S, Carrillo-Ballesteros FJ, Oregon-Romero E. ICOS Gene Polymorphisms (IVS1 + 173 T/C and c. 1624 C/T) in Primary Sjögren's Syndrome Patients: Analysis of ICOS Expression. Curr Issues Mol Biol 2022; 44:764-776. [PMID: 35723338 PMCID: PMC8929044 DOI: 10.3390/cimb44020053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Revised: 01/26/2022] [Accepted: 01/31/2022] [Indexed: 11/16/2022] Open
Abstract
Background: Primary Sjögren’s syndrome (pSS) is a systemic autoimmune disease, which affects exocrine glands. T cell activation is a trigger mechanism in the immune response. Hyperreactivity of T cells and antibody production are features in pSS. ICOS can be critical in the pathogenesis of pSS. Methods: A total of 134 pSS patients and 134 control subjects (CS) were included. Genotyping was performed by PCR-RFLP. ICOS mRNA expression was quantified by real-time PCR, and CD4+ ICOS+ T cells were determined by flow cytometry. Results: The ICOS IVS1 + 173 T>C polymorphisms were not associated with susceptibility to pSS (p = 0.393, CI = 0.503−1.311). However, the c.1624 C>T polymorphism was associated with a reduction in the risk of development of pSS (p = 0.015, CI = 0.294−0.884). An increase in ICOS mRNA expression in patients was observed (3.7-fold). Furthermore, pSS patients showed an increase in membranal-ICOS expression (mICOS). High expression of mICOS (MFI) was associated with lymphocytic infiltration. Conclusions: The IVS1 + 173 polymorphism is not a genetic marker for the development of pSS, while c.1624 T allele was associated with a low risk. However, elevated mICOS expression in pSS patients with high lymphocytic infiltration was found. ICOS may have an important role in the immunopathogenesis of pSS and should be analyzed in T cell subsets in pSS patients as a possible disease marker.
Collapse
Affiliation(s)
- José Antonio García-Espinoza
- Instituto de Investigación en Ciencias Biomédicas, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara 44340, Mexico; (J.A.G.-E.); (J.F.M.-V.); (M.G.-C.); (J.H.-B.); (C.A.P.-S.); (E.F.L.-V.); (G.A.S.-Z.)
| | - José Francisco Muñoz-Valle
- Instituto de Investigación en Ciencias Biomédicas, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara 44340, Mexico; (J.A.G.-E.); (J.F.M.-V.); (M.G.-C.); (J.H.-B.); (C.A.P.-S.); (E.F.L.-V.); (G.A.S.-Z.)
| | - Mariel García-Chagollán
- Instituto de Investigación en Ciencias Biomédicas, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara 44340, Mexico; (J.A.G.-E.); (J.F.M.-V.); (M.G.-C.); (J.H.-B.); (C.A.P.-S.); (E.F.L.-V.); (G.A.S.-Z.)
| | - Jorge Hernández-Bello
- Instituto de Investigación en Ciencias Biomédicas, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara 44340, Mexico; (J.A.G.-E.); (J.F.M.-V.); (M.G.-C.); (J.H.-B.); (C.A.P.-S.); (E.F.L.-V.); (G.A.S.-Z.)
| | - Claudia Azucena Palafox-Sánchez
- Instituto de Investigación en Ciencias Biomédicas, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara 44340, Mexico; (J.A.G.-E.); (J.F.M.-V.); (M.G.-C.); (J.H.-B.); (C.A.P.-S.); (E.F.L.-V.); (G.A.S.-Z.)
| | - Erika Fabiola López-Villalobos
- Instituto de Investigación en Ciencias Biomédicas, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara 44340, Mexico; (J.A.G.-E.); (J.F.M.-V.); (M.G.-C.); (J.H.-B.); (C.A.P.-S.); (E.F.L.-V.); (G.A.S.-Z.)
| | - Gabriela Athziri Sánchez-Zuno
- Instituto de Investigación en Ciencias Biomédicas, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara 44340, Mexico; (J.A.G.-E.); (J.F.M.-V.); (M.G.-C.); (J.H.-B.); (C.A.P.-S.); (E.F.L.-V.); (G.A.S.-Z.)
| | - Gloria Esther Martínez-Bonilla
- Servicio de Reumatología, O.P.D. Hospital Civil de Guadalajara “Fray Antonio Alcalde”, Guadalajara 44280, Mexico; (G.E.M.-B.); (S.C.-C.)
| | - Sergio Cerpa-Cruz
- Servicio de Reumatología, O.P.D. Hospital Civil de Guadalajara “Fray Antonio Alcalde”, Guadalajara 44280, Mexico; (G.E.M.-B.); (S.C.-C.)
| | - Francisco Josue Carrillo-Ballesteros
- Departamento de Farmacobiología, Centro Universitarios de Ciencias Exactas e Ingenierias, Universidad de Guadalajara, Guadalajara 44430, Mexico;
| | - Edith Oregon-Romero
- Instituto de Investigación en Ciencias Biomédicas, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara 44340, Mexico; (J.A.G.-E.); (J.F.M.-V.); (M.G.-C.); (J.H.-B.); (C.A.P.-S.); (E.F.L.-V.); (G.A.S.-Z.)
- Correspondence: ; Tel.: +52-1-3310585200 (ext. 34200)
| |
Collapse
|
6
|
Orvain C, Cauvet A, Prudent A, Guignabert C, Thuillet R, Ottaviani M, Tu L, Duhalde F, Nicco C, Batteux F, Avouac J, Wang N, Seaberg MA, Dillon SR, Allanore Y. Acazicolcept (ALPN-101), a dual ICOS/CD28 antagonist, demonstrates efficacy in systemic sclerosis preclinical mouse models. Arthritis Res Ther 2022; 24:13. [PMID: 34986869 PMCID: PMC8728910 DOI: 10.1186/s13075-021-02709-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Accepted: 12/20/2021] [Indexed: 11/24/2022] Open
Abstract
Background Uncontrolled immune response with T cell activation has a key role in the pathogenesis of systemic sclerosis (SSc), a disorder that is characterized by generalized fibrosis affecting particularly the lungs and skin. Costimulatory molecules are key players during immune activation, and recent evidence supports a role of CD28 and ICOS in the development of fibrosis. We herein investigated the efficacy of acazicolcept (ALPN-101), a dual ICOS/CD28 antagonist, in two complementary SSc-related mouse models recapitulating skin fibrosis, interstitial lung disease, and pulmonary hypertension. Methods Expression of circulating soluble ICOS and skin-expressed ICOS was investigated in SSc patients. Thereafter, acazicolcept was evaluated in the hypochlorous acid (HOCL)-induced dermal fibrosis mouse model and in the Fra-2 transgenic (Tg) mouse model. In each model, mice received 400 μg of acazicolcept or a molar-matched dose of an Fc control protein twice a week for 6 weeks. After 6 weeks, skin and lung were evaluated. Results ICOS was significantly increased in the sera from SSc patients and in SSc skin biopsies as compared to samples from healthy controls. Similar body weight changes were observed between Fc control and acazicolcept groups in both HOCL and Fra-2 Tg mice suggesting a good tolerance of acazicolcept treatment. In mice challenged with HOCL, acazicolcept induced a significant decrease in dermal thickness, collagen content, myofibroblast number, and inflammatory infiltrates characterized by B cells, T cells, neutrophils, and macrophages. In the Fra-2 Tg mouse model, acazicolcept treatment reduced lung collagen content, fibrillar collagen, histological fibrosis score, and right ventricular systolic pressure (RVSP). A reduction in frequency of CD4+ and T effector memory cells and an increase in the percentage of CD4+ T naïve cells in spleen and lung of acazicolcept-treated Fra-2 Tg mice was observed as compared to Fc control-treated Fra-2 Tg mice. Moreover, acazicolcept reduced CD69 and PD-1 expression on CD4+ T cells from the spleen and the lung. Target engagement by acazicolcept was demonstrated by blockade of CD28 and ICOS detection by flow cytometry in treated mice. Conclusions Our results confirm the importance of costimulatory molecules in inflammatory-driven fibrosis. Our data highlight a key role of ICOS and CD28 in SSc. Using complementary models, we demonstrated that dual ICOS/CD28 blockade by acazicolcept decreased dermal and pulmonary fibrosis and alleviated pulmonary hypertension. These results pave the way for subsequent research on ICOS/CD28-targeted therapies. Supplementary Information The online version contains supplementary material available at 10.1186/s13075-021-02709-2.
Collapse
Affiliation(s)
- Cindy Orvain
- Université Paris Descartes, Sorbonne Paris Cité, INSERM U1016, Institut Cochin, CNRS UMR8104, Paris, France
| | - Anne Cauvet
- Université Paris Descartes, Sorbonne Paris Cité, INSERM U1016, Institut Cochin, CNRS UMR8104, Paris, France
| | - Alexis Prudent
- Université Paris Descartes, Sorbonne Paris Cité, INSERM U1016, Institut Cochin, CNRS UMR8104, Paris, France
| | - Christophe Guignabert
- INSERM UMR_S 999, Le Plessis-Robinson, France.,Université Paris-Sud, Université Paris-Saclay, Le Kremlin-Bicêtre, France
| | - Raphaël Thuillet
- INSERM UMR_S 999, Le Plessis-Robinson, France.,Université Paris-Sud, Université Paris-Saclay, Le Kremlin-Bicêtre, France
| | - Mina Ottaviani
- INSERM UMR_S 999, Le Plessis-Robinson, France.,Université Paris-Sud, Université Paris-Saclay, Le Kremlin-Bicêtre, France
| | - Ly Tu
- INSERM UMR_S 999, Le Plessis-Robinson, France.,Université Paris-Sud, Université Paris-Saclay, Le Kremlin-Bicêtre, France
| | - Fanny Duhalde
- Université Paris Descartes, Sorbonne Paris Cité, INSERM U1016, Institut Cochin, CNRS UMR8104, Paris, France
| | - Carole Nicco
- Université Paris Descartes, Sorbonne Paris Cité, INSERM U1016, Institut Cochin, CNRS UMR8104, Paris, France
| | - Frédéric Batteux
- Université Paris Descartes, Sorbonne Paris Cité, INSERM U1016, Institut Cochin, CNRS UMR8104, Paris, France.,Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpital Universitaire Paris Centre (HUPC), Centre Hospitalier Universitaire (CHU) Cochin, Service d'immunologie biologique (Professeur Batteux), Paris, France
| | - Jérôme Avouac
- Université Paris Descartes, Sorbonne Paris Cité, INSERM U1016, Institut Cochin, CNRS UMR8104, Paris, France.,Université Paris Descartes, Sorbonne Paris Cité, Service de Rhumatologie A, Hôpital 27 rue du Faubourg Saint-Jacques, Cochin, 75014, Paris, France
| | | | | | | | - Yannick Allanore
- Université Paris Descartes, Sorbonne Paris Cité, INSERM U1016, Institut Cochin, CNRS UMR8104, Paris, France. .,Université Paris Descartes, Sorbonne Paris Cité, Service de Rhumatologie A, Hôpital 27 rue du Faubourg Saint-Jacques, Cochin, 75014, Paris, France.
| |
Collapse
|
7
|
Gao Y, Dunlap G, Elahee M, Rao DA. Patterns of T-Cell Phenotypes in Rheumatic Diseases From Single-Cell Studies of Tissue. ACR Open Rheumatol 2021; 3:601-613. [PMID: 34255929 PMCID: PMC8449042 DOI: 10.1002/acr2.11296] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Accepted: 05/19/2021] [Indexed: 02/06/2023] Open
Abstract
High-dimensional analyses of tissue samples from patients with rheumatic diseases are providing increasingly detailed descriptions of the immune cell populations that infiltrate tissues in different rheumatic diseases. Here we review key observations emerging from high-dimensional analyses of T cells within tissues in different rheumatic diseases, highlighting common themes across diseases as well as distinguishing features. Single-cell RNA sequencing analyses capture several dimensions of T-cell states, yet surprisingly, these analyses generally have not demonstrated distinct clusters of paradigmatic T-cell effector subsets, such as T helper (Th) 1, Th2, and Th17 cells. Rather, global transcriptomics robustly identify both proliferating T cells and regulatory T cells and have also helped to reveal new effector subsets in inflamed tissues, including T peripheral helper cells and granzyme K+ T cells. Further characterization of the T-cell populations that accumulate within target tissues should enable more precise targeting of biologic therapies and accelerate development of more specific biomarkers to track activity of relevant immune pathways in patients with rheumatic diseases.
Collapse
Affiliation(s)
- Yidan Gao
- Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Garrett Dunlap
- Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Mehreen Elahee
- Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Deepak A Rao
- Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
8
|
Worrell JC, O'Reilly S. Bi-directional communication: Conversations between fibroblasts and immune cells in systemic sclerosis. J Autoimmun 2020; 113:102526. [PMID: 32713676 DOI: 10.1016/j.jaut.2020.102526] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Revised: 07/13/2020] [Accepted: 07/14/2020] [Indexed: 02/09/2023]
Abstract
Systemic Sclerosis (SSc) is an autoimmune idiopathic connective tissue disease, characterized by aberrant fibro-proliferative and inflammatory responses, causing fibrosis of multiple organs. In recent years the interactions between innate and adaptive immune cells with resident fibroblasts have been uncovered. Cross-talk between immune and stromal cells mediates activation of stromal cells to myofibroblasts; key cells in the pathophysiology of fibrosis. These cells and their cytokines appear to mediate their effects in both a paracrine and autocrine fashion. This review examines the role of innate and adaptive immune cells in SSc, focusing on recent advances that have illuminated our understanding of ongoing bi-directional communication between immune and stromal cells. Finally, we appraise current and future therapies and how these may be useful in a disease that currently has no specific disease modifying treatment.
Collapse
Affiliation(s)
- Julie C Worrell
- Insititute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, UK
| | - Steven O'Reilly
- Durham University, Biosciences, Faculty of Science, Durham, UK. steven.o'
| |
Collapse
|
9
|
Regulatory T cells in patients with early untreated rheumatoid arthritis: Phenotypic changes in the course of methotrexate treatment. Biochimie 2020; 174:9-17. [PMID: 32275944 DOI: 10.1016/j.biochi.2020.03.014] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2019] [Revised: 02/25/2020] [Accepted: 03/20/2020] [Indexed: 12/15/2022]
Abstract
Rheumatoid arthritis (RA) is frequent systemic autoimmune disease characterized by excessive activation of collagen-specific T helper cells, and elevated level of autoantibodies in the serum. Development of RA is associated with defect in compartment of regulatory CD4+Foxp3+ T cells (Treg), but data concerning suppressive potential of Treg population in RA patients are contradictory and depend on the stage of disease. In this study we aimed to characterize abundance and phenotypic markers of CD4+Foxp3+ Treg in peripheral blood of healthy donors compared to untreated early RA patients to find potential correlations with the disease activity, antibody level, and absolute numbers and proportion of different subpopulations of T cells. Moreover, we assessed the influence of methotrexate (MT) treatment on percentage and absolute numbers of CD4+Foxp3+ Treg from the peripheral blood of untreated early RA patients. We demonstrate that increase and phenotypic changes in Treg population correlate well with response to MT. Analysis of the cohorts of matched RA patients (n = 45) and healthy controls (n = 20) revealed that patients with untreated early RA demonstrate substantial decrease in blood Treg percentage and absolute number, as well as low level of activated Treg surface markers in comparison to healthy control. The defect in Treg compartment negatively correlates with both RA activity and antibody level. MT treatment of patients with early untreated RA increases both proportion and absolute number of Treg with high level of activation markers, suggesting an increase of their functional capacity. Here we speculate the role of Tregs as specific cellular marker of successful RA treatment.
Collapse
|
10
|
Yan X, Gu Y, Wang C, Sun S, Wang X, Tian J, Wang M, Ji X, Duan X, Gao H, Fang Q, Dong W, Zhang X, Xue Q. Unbalanced expression of membrane-bound and soluble inducible costimulator and programmed cell death 1 in patients with myasthenia gravis. Clin Immunol 2019; 207:68-78. [PMID: 31374257 DOI: 10.1016/j.clim.2019.07.011] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Revised: 07/23/2019] [Accepted: 07/23/2019] [Indexed: 02/01/2023]
Abstract
This study aimed to investigate the possible functions and mechanisms of positive and negative costimulatory molecules in the pathological process of myasthenia gravis (MG). The expression levels of membrane-bound inducible costimulator (ICOS) and programmed cell death 1 (PD-1) in peripheral blood T cells, their corresponding ligands ICOSL and PDL-1 on B cells, and their soluble forms (sICOS, sPD-1, sICOSL, and sPDL-1) in plasma were detected in patients with untreated-stage MG (USMG) and remission-stage MG (RSMG). The results showed that the expression levels of membrane-bound ICOS and PD-1 in the peripheral blood T cells of the USMG group and their corresponding ligands ICOSL and PD-L1 on B cells were significantly increased compared to those in the RSMG group and healthy controls (HCs). The levels of sICOSL and sPD-1 were significantly upregulated in USMG patients compared to those in the RSMG and HC groups, while the levels of sICOS and sPD-L1 were not different. The expression of PD-L1 on CD19+ B cells was positively correlated with the concentrations of AchR Ab in the USMG group. The expression of ICOS and PD-1 in CD4+ T cells and the expression of ICOSL and PD-L1 on CD19+ B cells were positively correlated with the quantitative myasthenia gravis (QMG) scores in the USMG group. Also, in the USMG group, the plasma levels of sICOSL and sPD-1 were positively correlated with the QMG scores. In addition, the percentage of peripheral blood follicular helper T (Tfh) cells in the USMG group was positively correlated with ICOS and PD-1 expression on CD4+ T cells and ICOSL and PD-L1 expression on CD19+ B cells. There were positive correlations between sICOSL and sPD-1 levels and the percentage of peripheral blood Tfh cells and plasma interleukin-21 (IL-21) levels in the USMG group. The results suggest that the positive ICOS/ICOSL and negative PD-1/PD-L1 costimulatory molecule pairs participate in the pathological process of MG. Abnormal sICOSL and sPD-1 expression might interfere with the normal signal transduction of ICOS and PD-1 on Tfh cells, causing excessive activation of Tfh cells and promotion of disease progression. sICOSL and sPD-1 have potential value in monitoring MG disease states.
Collapse
Affiliation(s)
- Xiaoming Yan
- Neurology Department, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, China
| | - Yanzheng Gu
- Institute of Clinical Immunology, Jiangsu Key Laboratory of Clinical Immunology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, China; Suzhou Clinical Medical Center of Neurology, Suzhou, Jiangsu 215004, China
| | - Caiqin Wang
- Neurology Department, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, China
| | - Simao Sun
- Neurology Department, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, China
| | - Xiaozhu Wang
- Neurology Department, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, China
| | - Jingluan Tian
- Neurology Department, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, China
| | - Mingyuan Wang
- Suzhou Red Cross Central Blood Station, Suzhou, Jiangsu 215006, China
| | - Xiaopei Ji
- Neurology Department, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, China
| | - Xiaoyu Duan
- Neurology Department, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, China
| | - Hanqing Gao
- Neurology Department, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, China
| | - Qi Fang
- Neurology Department, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, China; Suzhou Clinical Medical Center of Neurology, Suzhou, Jiangsu 215004, China
| | - Wanli Dong
- Neurology Department, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, China
| | - Xueguang Zhang
- Institute of Clinical Immunology, Jiangsu Key Laboratory of Clinical Immunology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, China
| | - Qun Xue
- Neurology Department, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, China; Institute of Clinical Immunology, Jiangsu Key Laboratory of Clinical Immunology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, China; Suzhou Clinical Medical Center of Neurology, Suzhou, Jiangsu 215004, China.
| |
Collapse
|
11
|
Ricard L, Jachiet V, Malard F, Ye Y, Stocker N, Rivière S, Senet P, Monfort JB, Fain O, Mohty M, Gaugler B, Mekinian A. Circulating follicular helper T cells are increased in systemic sclerosis and promote plasmablast differentiation through the IL-21 pathway which can be inhibited by ruxolitinib. Ann Rheum Dis 2019; 78:539-550. [PMID: 30760472 DOI: 10.1136/annrheumdis-2018-214382] [Citation(s) in RCA: 64] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Revised: 12/18/2018] [Accepted: 12/19/2018] [Indexed: 12/29/2022]
Abstract
OBJECTIVES Systemic sclerosis (SSc) is an autoimmune disease characterised by widespread fibrosis, microangiopathy and autoantibodies. Follicular helper T (Tfh) cells CD4+CXCR5+PD-1+ cooperate with B lymphocytes to induce the differentiation of plasmocytes secreting immunoglobulins (Ig). Circulating Tfh (cTfh) cells are increased in several autoimmune diseases. However, there are no data about cTfh cells and their interaction with B cells in SSc. The aim of this study was to perform a quantitative and functional analysis of cTfh cells in SSc. METHODS Using flow cytometry, we analysed cTfh cells from 50 patients with SSc and 32 healthy controls (HC). In vitro coculture experiments of sorted cTfh and B cells were performed for functional analysis. IgG and IgM production were measured by ELISA. RESULTS We observed that cTfh cell numbers are increased in patients with SSc compared with HC. Furthermore, the increase in cTfh cells was more potent in patients with severe forms of SSc such as diffuse SSc and in the presence of arterial pulmonary hypertension. cTfh cells from patients with SSc present an activated Tfh phenotype, with high expression of BCL-6, increased capacity to produce IL-21 in comparison with healthy controls. In vitro, cTfh cells from patients with SSc had higher capacity to stimulate the differentiation of CD19+CD27+CD38hi B cells and their secretion of IgG and IgM through the IL-21 pathway than Tfh cells from healthy controls. Blocking IL-21R or using the JAK1/2 inhibitor ruxolitinib reduced the Tfh cells' capacity to stimulate the plasmablasts and decreased the Ig production. CONCLUSIONS Circulating Tfh cells are increased in SSc and correlate with SSc severity. The IL-21 pathway or JAK1/2 blockade by ruxolitinib could be a promising strategy in the treatment of SSc.
Collapse
Affiliation(s)
- Laure Ricard
- Centre de Recherche Saint-Antoine (CRSA), Sorbonne Université, INSERM U938, Paris, France.,Service de Médecine Interne et de l'Inflammation (DHU i2B), AP-HP, Hôpital Saint-Antoine, Paris, France
| | - Vincent Jachiet
- Centre de Recherche Saint-Antoine (CRSA), Sorbonne Université, INSERM U938, Paris, France.,Service de Médecine Interne et de l'Inflammation (DHU i2B), AP-HP, Hôpital Saint-Antoine, Paris, France
| | - Florent Malard
- Centre de Recherche Saint-Antoine (CRSA), Sorbonne Université, INSERM U938, Paris, France.,Service d'Hématologie Clinique et Thérapie Cellulaire, AP-HP, Hôpital Saint-Antoine, Paris, France
| | - Yishan Ye
- Centre de Recherche Saint-Antoine (CRSA), Sorbonne Université, INSERM U938, Paris, France
| | - Nicolas Stocker
- Centre de Recherche Saint-Antoine (CRSA), Sorbonne Université, INSERM U938, Paris, France
| | - Sébastien Rivière
- Service de Médecine Interne et de l'Inflammation (DHU i2B), AP-HP, Hôpital Saint-Antoine, Paris, France
| | - Patricia Senet
- Service de Dermatologie, AP-HP, Hôpital Tenon, Paris, France
| | | | - Olivier Fain
- Service de Médecine Interne et de l'Inflammation (DHU i2B), AP-HP, Hôpital Saint-Antoine, Paris, France
| | - Mohamad Mohty
- Centre de Recherche Saint-Antoine (CRSA), Sorbonne Université, INSERM U938, Paris, France.,Service d'Hématologie Clinique et Thérapie Cellulaire, AP-HP, Hôpital Saint-Antoine, Paris, France
| | - Béatrice Gaugler
- Centre de Recherche Saint-Antoine (CRSA), Sorbonne Université, INSERM U938, Paris, France.,Service d'Hématologie Clinique et Thérapie Cellulaire, AP-HP, Hôpital Saint-Antoine, Paris, France
| | - Arsène Mekinian
- Centre de Recherche Saint-Antoine (CRSA), Sorbonne Université, INSERM U938, Paris, France .,Service de Médecine Interne et de l'Inflammation (DHU i2B), AP-HP, Hôpital Saint-Antoine, Paris, France
| |
Collapse
|
12
|
Boleto G, Allanore Y, Avouac J. Targeting Costimulatory Pathways in Systemic Sclerosis. Front Immunol 2018; 9:2998. [PMID: 30619351 PMCID: PMC6305435 DOI: 10.3389/fimmu.2018.02998] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2018] [Accepted: 12/04/2018] [Indexed: 01/04/2023] Open
Abstract
Systemic sclerosis (SSc) is an autoimmune T-cell disease that is characterized by pathological fibrosis of the skin and internal organs. SSc is considered a prototype condition for studying the links between autoimmunity and fibrosis. Costimulatory pathways such as CD28/CTLA-4, ICOS-B7RP1, CD70-CD27, CD40-CD154, or OX40-OX40L play an essential role in the modulation of T-cell and inflammatory immune responses. A growing body of evidence suggests that T-cell costimulation signals might be implicated in the pathogenesis of SSc. CD28, CTLA-4, ICOS, and OX40L are overexpressed in patients with SSc, particularly in patients with cutaneous diffuse forms. In pre-clinical models of SSc, T-cell costimulation blockade with abatacept (CTLA-4-Ig) prevented and induced the regression of inflammation-driven dermal fibrosis, improved digestive involvement, prevented lung fibrosis, and attenuated pulmonary hypertension in complementary models of SSc. Likewise, potent anti-fibrotic effects were seen with the blockade of OX40L by reducing the infiltration of inflammatory cells into lesional tissues leading to decreased fibroblast activation. Concerning clinical effects, a preliminary observational study suggested some effectiveness of abatacept on inflammatory joint involvement, whereas clinical improvement of skin fibrosis was observed in a small placebo-controlled randomized trial. Currently there is one ongoing phase II clinical trial assessing the efficacy of abatacept in SSc (ASSET trial, NCT02161406). Overall, given the lack of available effective agents and the known toxic effects of immunosuppressive agents approved for use in SSc, costimulatory pathways offer the advantage of a targeted approach to costimulatory signals and potentially a better safety profile.
Collapse
Affiliation(s)
- Gonçalo Boleto
- Université Paris Descartes, Sorbonne Paris Cité, INSERM U1016, Institut Cochin, CNRS UMR8104, Paris, France.,Université Paris Descartes, Sorbonne Paris Cité, Service de Rhumatologie A, Hôpital Cochin, Paris, France
| | - Yannick Allanore
- Université Paris Descartes, Sorbonne Paris Cité, INSERM U1016, Institut Cochin, CNRS UMR8104, Paris, France.,Université Paris Descartes, Sorbonne Paris Cité, Service de Rhumatologie A, Hôpital Cochin, Paris, France
| | - Jérôme Avouac
- Université Paris Descartes, Sorbonne Paris Cité, INSERM U1016, Institut Cochin, CNRS UMR8104, Paris, France.,Université Paris Descartes, Sorbonne Paris Cité, Service de Rhumatologie A, Hôpital Cochin, Paris, France
| |
Collapse
|
13
|
Taylor DK, Mittereder N, Kuta E, Delaney T, Burwell T, Dacosta K, Zhao W, Cheng LI, Brown C, Boutrin A, Guo X, White WI, Zhu J, Dong H, Bowen MA, Lin J, Gao C, Yu L, Ramaswamy M, Gaudreau MC, Woods R, Herbst R, Carlesso G. T follicular helper–like cells contribute to skin fibrosis. Sci Transl Med 2018. [DOI: 10.1126/scitranslmed.aaf5307] [Citation(s) in RCA: 65] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
|
14
|
D'Amico F, Fiorito G, Skarmoutsou E, Granata M, Rossi GA, Trovato C, Bellocchi C, Marchini M, Beretta L, Mazzarino MC. FOXP3, ICOS and ICOSL gene polymorphisms in systemic sclerosis: FOXP3 rs2294020 is associated with disease progression in a female Italian population. Immunobiology 2017; 223:112-117. [PMID: 29030005 DOI: 10.1016/j.imbio.2017.10.004] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2017] [Revised: 07/13/2017] [Accepted: 10/03/2017] [Indexed: 11/18/2022]
Abstract
Systemic sclerosis (SSc), an autoimmune disorder, is characterized by vasculopathy, inflammation, progressive perivascular and interstitial fibrosis. Its pathogenesis is largely unknown, however strong evidences suggest that genetic predisposition may contribute to SSc development. Several gene polymorphisms involved in regulatory T cell function have been identified in many autoimmune diseases, including SSc. Moreover, dysregulation of co-stimulatory and/or co-inhibitory signals, including ICOS signalling, can lead to autoimmunity. The aim of the present study was to investigate the association of the FOXP3 rs2294020, ICOS rs6726035 and ICOSL rs378299 SNPs with both the susceptibility and the progression to SSc in an Italian case-series of patients. SNP genotyping results were successfully obtained from a total of 350 subjects including 166 individuals with SSc and 184 healthy controls. Although analysis tests did not show any significant associations between the SNPs under study and susceptibility to SSc, the occurrence of FOXP3 rs2294020 in female patients was associated with decreased time to progression from early to definite SSc (allelic model: HR=1.43; CI=1.03-1.99; p=0.03; dominant model: HR=1.54; CI=1.04-2.28; p=0.03). The inclusion of presence of ACA autoantibodies in the model did not significantly change the estimates. No conclusions can be drawn for the susceptibility to the disease or the time to progression in men due to the low statistical power. This study provides evidence of the association of rs2294020 with SSc evolution in female patients, modulating the time of progression from the diagnosis of early SSc to the diagnosis of definite SSc, while no effect on SSc susceptibility per se was found. rs2294020 may be considered a disease-modifying gene-variant rather than a disease-susceptibility SNP in SSc.
Collapse
Affiliation(s)
- Fabio D'Amico
- Department of Biomedical and Biotechnological Sciences, University of Catania, Via S. Sofia 97, 95123 Catania, Italy.
| | - Giovanni Fiorito
- Department of Medical Sciences, University of Turin, via Nizza 52, 10126 Turin, Italy; Italian Istitute for Genomic Medicine (IIGM), via Nizza 52, 10126 Turin, Italy
| | - Evangelia Skarmoutsou
- Department of Biomedical and Biotechnological Sciences, University of Catania, Via S. Sofia 97, 95123 Catania, Italy
| | - Mariagrazia Granata
- Department of Biomedical and Biotechnological Sciences, University of Catania, Via S. Sofia 97, 95123 Catania, Italy
| | - Giulio A Rossi
- Department of Biomedical and Biotechnological Sciences, University of Catania, Via S. Sofia 97, 95123 Catania, Italy
| | - Chiara Trovato
- Department of Biomedical and Biotechnological Sciences, University of Catania, Via S. Sofia 97, 95123 Catania, Italy
| | - Chiara Bellocchi
- Referral Center for Systemic Autoimmune Diseases, University of Milan and Fondazione IRCCS Ospedale Maggiore Policlinico, Mangiagalli e Regina Elena, via Pace 9, I-20122 Milan, Italy
| | - Maurizio Marchini
- Referral Center for Systemic Autoimmune Diseases, University of Milan and Fondazione IRCCS Ospedale Maggiore Policlinico, Mangiagalli e Regina Elena, via Pace 9, I-20122 Milan, Italy
| | - Lorenzo Beretta
- Referral Center for Systemic Autoimmune Diseases, University of Milan and Fondazione IRCCS Ospedale Maggiore Policlinico, Mangiagalli e Regina Elena, via Pace 9, I-20122 Milan, Italy
| | - Maria Clorinda Mazzarino
- Department of Biomedical and Biotechnological Sciences, University of Catania, Via S. Sofia 97, 95123 Catania, Italy
| |
Collapse
|
15
|
Effects of ICOS+ T cell depletion via afucosylated monoclonal antibody MEDI-570 on pregnant cynomolgus monkeys and the developing offspring. Reprod Toxicol 2017; 74:116-133. [PMID: 28916434 DOI: 10.1016/j.reprotox.2017.08.018] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2017] [Revised: 08/15/2017] [Accepted: 08/29/2017] [Indexed: 01/11/2023]
Abstract
MEDI-570 is a fully human afucosylated monoclonal antibody (MAb) against Inducible T-cell costimulator (ICOS), highly expressed on CD4+ T follicular helper (TFH) cells. Effects of MEDI-570 were evaluated in an enhanced pre-postnatal development toxicity (ePPND) study in cynomolgus monkeys. Administration to pregnant monkeys did not cause any abortifacient effects. Changes in hematology and peripheral blood T lymphocyte subsets in maternal animals and infants and the attenuated infant IgG immune response to keyhole limpet hemocyanin (KLH) were attributed to MEDI-570 pharmacology. Adverse findings included aggressive fibromatosis in one dam and two infant losses in the high dose group with anatomic pathology findings suggestive of atypical lymphoid hyperplasia. The margin of safety relative to the no observed adverse effect level (NOAEL) for the highest planned clinical dose in the Phase 1a study was 7. This study suggests that women of child bearing potential employ effective methods of contraception while being treated with MEDI-570.
Collapse
|
16
|
Mesquita D, Cruvinel WM, Resende LS, Mesquita FV, Silva NP, Câmara NOS, Andrade LEC. Follicular helper T cell in immunity and autoimmunity. ACTA ACUST UNITED AC 2016; 49:e5209. [PMID: 27096200 PMCID: PMC4843212 DOI: 10.1590/1414-431x20165209] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2015] [Accepted: 01/27/2016] [Indexed: 11/22/2022]
Abstract
The traditional concept that effector T helper (Th) responses are mediated by Th1/Th2 cell subtypes has been broadened by the recent demonstration of two new effector T helper cells, the IL-17 producing cells (Th17) and the follicular helper T cells (Tfh). These new subsets have many features in common, such as the ability to produce IL-21 and to express the IL-23 receptor (IL23R), the inducible co-stimulatory molecule ICOS, and the transcription factor c-Maf, all of them essential for expansion and establishment of the final pool of both subsets. Tfh cells differ from Th17 by their ability to home to B cell areas in secondary lymphoid tissue through interactions mediated by the chemokine receptor CXCR5 and its ligand CXCL13. These CXCR5+ CD4+ T cells are considered an effector T cell type specialized in B cell help, with a transcriptional profile distinct from Th1 and Th2 cells. The role of Tfh cells and its primary product, IL-21, on B-cell activation and differentiation is essential for humoral immunity against infectious agents. However, when deregulated, Tfh cells could represent an important mechanism contributing to exacerbated humoral response and autoantibody production in autoimmune diseases. This review highlights the importance of Tfh cells by focusing on their biology and differentiation processes in the context of normal immune response to infectious microorganisms and their role in the pathogenesis of autoimmune diseases.
Collapse
Affiliation(s)
- D Mesquita
- Divisão de Reumatologia, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, SP, Brasil
| | - W M Cruvinel
- Divisão de Reumatologia, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, SP, Brasil
| | - L S Resende
- Divisão de Farmacologia, Instituto de Ciências Biológicas, Universidade de São Paulo, São Paulo, SP, Brasil
| | - F V Mesquita
- Divisão de Reumatologia, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, SP, Brasil
| | - N P Silva
- Divisão de Reumatologia, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, SP, Brasil
| | - N O S Câmara
- Divisão de Imunologia, Faculdade de Medicina, Universidade de São Paulo, São Paulo, SP, Brasil
| | - L E C Andrade
- Divisão de Reumatologia, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, SP, Brasil
| |
Collapse
|
17
|
Sanges S, Guerrier T, Launay D, Lefèvre G, Labalette M, Forestier A, Sobanski V, Corli J, Hauspie C, Jendoubi M, Yakoub-Agha I, Hatron PY, Hachulla E, Dubucquoi S. Role of B cells in the pathogenesis of systemic sclerosis. Rev Med Interne 2016; 38:113-124. [PMID: 27020403 DOI: 10.1016/j.revmed.2016.02.016] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2015] [Revised: 02/17/2016] [Accepted: 02/27/2016] [Indexed: 12/28/2022]
Abstract
Systemic sclerosis (SSc) is an orphan disease characterized by progressive fibrosis of the skin and internal organs. Aside from vasculopathy and fibrotic processes, its pathogenesis involves an aberrant activation of immune cells, among which B cells seem to play a significant role. Indeed, B cell homeostasis is disturbed during SSc: the memory subset is activated and displays an increased susceptibility to apoptosis, which is responsible for their decreased number. This chronic loss of B cells enhances bone marrow production of the naïve subset that accounts for their increased number in peripheral blood. This permanent activation state can be explained mainly by two mechanisms: a dysregulation of B cell receptor (BCR) signaling, and an overproduction of B cell survival signals, B cell activating factor (BAFF) and a proliferation-inducing ligand (APRIL). These disturbances of B cell homeostasis induce several functional anomalies that participate in the inflammatory and fibrotic events observed during SSc: autoantibody production (some being directly pathogenic); secretion of pro-inflammatory and pro-fibrotic cytokines (interleukin-6); direct cooperation with other SSc-involved cells [fibroblasts, through transforming growth factor-β (TGF-β) signaling, and T cells]. These data justify the evaluation of anti-B cell strategies as therapeutic options for SSc, such as B cell depletion or blockage of B cell survival signaling.
Collapse
Affiliation(s)
- S Sanges
- Université de Lille, U995, Lille Inflammation Research International Center (LIRIC), 59000 Lille, France; Inserm, U995, 59000 Lille, France; CHU de Lille, département de médecine interne et immunologie clinique, 59000 Lille, France; Centre national de référence maladies systémiques et auto-immunes rares (sclérodermie systémique), 59000 Lille, France; FHU Immune-Mediated Inflammatory Diseases and Targeted Therapies, 59000 Lille, France
| | - T Guerrier
- Université de Lille, U995, Lille Inflammation Research International Center (LIRIC), 59000 Lille, France; Inserm, U995, 59000 Lille, France; CHU de Lille, Centre de biologie-pathologie-génétique, institut d'Immunologie, 59000 Lille, France
| | - D Launay
- Université de Lille, U995, Lille Inflammation Research International Center (LIRIC), 59000 Lille, France; Inserm, U995, 59000 Lille, France; CHU de Lille, département de médecine interne et immunologie clinique, 59000 Lille, France; Centre national de référence maladies systémiques et auto-immunes rares (sclérodermie systémique), 59000 Lille, France; FHU Immune-Mediated Inflammatory Diseases and Targeted Therapies, 59000 Lille, France.
| | - G Lefèvre
- Université de Lille, U995, Lille Inflammation Research International Center (LIRIC), 59000 Lille, France; Inserm, U995, 59000 Lille, France; CHU de Lille, département de médecine interne et immunologie clinique, 59000 Lille, France; Centre national de référence maladies systémiques et auto-immunes rares (sclérodermie systémique), 59000 Lille, France; FHU Immune-Mediated Inflammatory Diseases and Targeted Therapies, 59000 Lille, France; CHU de Lille, Centre de biologie-pathologie-génétique, institut d'Immunologie, 59000 Lille, France
| | - M Labalette
- Université de Lille, U995, Lille Inflammation Research International Center (LIRIC), 59000 Lille, France; Inserm, U995, 59000 Lille, France; FHU Immune-Mediated Inflammatory Diseases and Targeted Therapies, 59000 Lille, France; CHU de Lille, Centre de biologie-pathologie-génétique, institut d'Immunologie, 59000 Lille, France
| | - A Forestier
- Université de Lille, U995, Lille Inflammation Research International Center (LIRIC), 59000 Lille, France; Inserm, U995, 59000 Lille, France; CHU de Lille, département de médecine interne et immunologie clinique, 59000 Lille, France; Centre national de référence maladies systémiques et auto-immunes rares (sclérodermie systémique), 59000 Lille, France; FHU Immune-Mediated Inflammatory Diseases and Targeted Therapies, 59000 Lille, France
| | - V Sobanski
- Université de Lille, U995, Lille Inflammation Research International Center (LIRIC), 59000 Lille, France; Inserm, U995, 59000 Lille, France; CHU de Lille, département de médecine interne et immunologie clinique, 59000 Lille, France; Centre national de référence maladies systémiques et auto-immunes rares (sclérodermie systémique), 59000 Lille, France; FHU Immune-Mediated Inflammatory Diseases and Targeted Therapies, 59000 Lille, France
| | - J Corli
- Université de Lille, U995, Lille Inflammation Research International Center (LIRIC), 59000 Lille, France; Inserm, U995, 59000 Lille, France; FHU Immune-Mediated Inflammatory Diseases and Targeted Therapies, 59000 Lille, France; CHU de Lille, département de rhumatologie, 59000 Lille, France
| | - C Hauspie
- Université de Lille, U995, Lille Inflammation Research International Center (LIRIC), 59000 Lille, France; Inserm, U995, 59000 Lille, France; CHU de Lille, Centre de biologie-pathologie-génétique, institut d'Immunologie, 59000 Lille, France
| | - M Jendoubi
- Université de Lille, U995, Lille Inflammation Research International Center (LIRIC), 59000 Lille, France; Inserm, U995, 59000 Lille, France
| | - I Yakoub-Agha
- Université de Lille, U995, Lille Inflammation Research International Center (LIRIC), 59000 Lille, France; Inserm, U995, 59000 Lille, France; FHU Immune-Mediated Inflammatory Diseases and Targeted Therapies, 59000 Lille, France; CHU de Lille, département des maladies du sang, 59000 Lille, France
| | - P-Y Hatron
- Université de Lille, U995, Lille Inflammation Research International Center (LIRIC), 59000 Lille, France; CHU de Lille, département de médecine interne et immunologie clinique, 59000 Lille, France; Centre national de référence maladies systémiques et auto-immunes rares (sclérodermie systémique), 59000 Lille, France; FHU Immune-Mediated Inflammatory Diseases and Targeted Therapies, 59000 Lille, France
| | - E Hachulla
- Université de Lille, U995, Lille Inflammation Research International Center (LIRIC), 59000 Lille, France; Inserm, U995, 59000 Lille, France; CHU de Lille, département de médecine interne et immunologie clinique, 59000 Lille, France; Centre national de référence maladies systémiques et auto-immunes rares (sclérodermie systémique), 59000 Lille, France; FHU Immune-Mediated Inflammatory Diseases and Targeted Therapies, 59000 Lille, France
| | - S Dubucquoi
- Université de Lille, U995, Lille Inflammation Research International Center (LIRIC), 59000 Lille, France; Inserm, U995, 59000 Lille, France; FHU Immune-Mediated Inflammatory Diseases and Targeted Therapies, 59000 Lille, France; CHU de Lille, Centre de biologie-pathologie-génétique, institut d'Immunologie, 59000 Lille, France
| |
Collapse
|
18
|
Yan JW, Wang YJ, Peng WJ, Tao JH, Wan YN, Li BZ, Mei B, Chen B, Yao H, Yang GJ, Li XP, Ye DQ, Wang J. Therapeutic potential of interleukin-17 in inflammation and autoimmune diseases. Expert Opin Ther Targets 2013; 18:29-41. [PMID: 24147601 DOI: 10.1517/14728222.2013.843669] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
INTRODUCTION Interleukin-17 (IL-17) is a proinflammatory cytokine that mainly produced by T helper 17 (Th17) cells. In this article, we discussed the role of IL-17 in inflammation and autoimmune diseases, and the therapeutic strategies targeting IL-17. AREAS COVERED In this article, we discussed the proinflammatory cytokine IL-17 and IL-17 receptors signals, and their regulation. IL-17 expression was abnormal in the bacterium, virus and fungus infection, and its higher level caused the tissue inflammation. IL-17 was involved in the pathological process of autoimmune diseases, such as systemic sclerosis, rheumatoid arthritis, ankylosing spondylitis and systemic lupus erythematosus, and IL-17 has been put as a therapeutic target in the clinic. EXPERT OPINION IL-17/IL-17R signals and their application in inflammation process still need to be explored. Therapeutic strategies targeting IL-17 in autoimmune diseases ameliorated the inadequate response to anti-TNF-α therapy.
Collapse
Affiliation(s)
- Jun-Wei Yan
- Anhui Medical University, School of Public Health, Department of Epidemiology and Biostatistics , NO.81, Meishan Road, Hefei, Anhui, 230032, PR , China +86 551 65161175 ; +86 551 65161126 ;
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|