1
|
Christopoulos G, Christopoulou V, Stamatiou K, Babionitakis A, Routsias JG. Association Between Soluble Cell Adhesion Molecules (sP-Selectin, sE-Selectin, and sICAM-1) and Antibodies Against the Antigens of Proteus mirabilis in Rheumatoid Arthritis Patients. Cureus 2024; 16:e64942. [PMID: 39156359 PMCID: PMC11330672 DOI: 10.7759/cureus.64942] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/19/2024] [Indexed: 08/20/2024] Open
Abstract
Objective The purpose of this study was to examine the association between the serum concentration of soluble cell adhesion molecules (CAMs) and antibodies against antigens of Proteus mirabilis (P. mirabilis) in rheumatoid arthritis (RA) patients, taking into consideration the implication of P. mirabilis in the etiopathogenesis of RA. Methods The serum levels of soluble P-selectin (sP-selectin), soluble E-selectin (sE-selectin), and soluble intercellular adhesion molecule-1 (sICAM-1) were determined by sandwich enzyme-linked immunosorbent assay (ELISA) in 59 RA patients and 36 healthy controls. Using the same ELISA method, the serum levels of class-specific antibodies against hemolysin (HpmB), urease C (UreC), and urease F (UreF) enzymes of P. mirabilis were also measured. Results In this study, increased levels of sP-selectin and sICAM-1 were observed in RA patients, while the levels of sE-selectin were increased in comparison with healthy controls but did not present a statistically significant difference. Moreover, increased levels of antibodies against HpmB, UreC, and UreF of P. mirabilis were found. Additionally, it was observed that the sE-selectin levels presented a significant correlation with IgG antibodies against the UreF antigen (there is no corresponding antigen in human tissue) in all the RA patients. A statistically significant correlation was observed between levels of soluble CAMs and antibodies against P. mirabilis in the different subgroups. Conclusion The observed correlation between soluble CAMs and antibodies against antigens of P. mirabilis, specifically in the subgroup of biologic therapy, indicates that P. mirabilis exists and provokes refractory in the treatment of RA.
Collapse
Affiliation(s)
| | - Vasiliki Christopoulou
- Department of Internal Medicine - Propaedeutic, Attikon University Hospital, Athens, GRC
| | | | - Andreas Babionitakis
- Department of Pathophysiology, Medical School National & Kapodistrian University of Athens, Athens, GRC
| | - John G Routsias
- Department of Microbiology, Medical School National & Kapodistrian University of Athens, Athens, GRC
| |
Collapse
|
2
|
Liu C, Yan Z, Zhang X, Xia T, Ashaolu JO, Olatunji OJ, Ashaolu TJ. Food-derived bioactive peptides potentiating therapeutic intervention in rheumatoid arthritis. Heliyon 2024; 10:e31104. [PMID: 38778960 PMCID: PMC11109807 DOI: 10.1016/j.heliyon.2024.e31104] [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: 04/05/2024] [Revised: 05/09/2024] [Accepted: 05/09/2024] [Indexed: 05/25/2024] Open
Abstract
Rheumatoid arthritis (RA) is a chronic inflammatory autoimmune disease that affects the joints of the human body and is projected to have a prevalence age-standardized rate of 1.5 million new cases worldwide by 2030. Several conventional and non-conventional preventive and therapeutic interventions have been suggested but they have their side effects including nausea, abdominal pain, liver damage, ulcers, heightened blood pressure, coagulation, and bleeding. Interestingly, several food-derived peptides (FDPs) from both plant and animal sources are increasingly gaining a reputation for their potential in the management or therapy of RA with little or no side effects. In this review, the concept of inflammation, its major types (acute and chronic), and RA identified as a chronic type were discussed based on its pathogenesis and pathophysiology. The conventional treatment options for RA were briefly outlined as the backdrop of introducing the FDPs that potentiate therapeutic effects in the management of RA.
Collapse
Affiliation(s)
- Chunhong Liu
- Second People's Hospital of Wuhu City, 241001, Anhui, China
| | - Zheng Yan
- Second People's Hospital of Wuhu City, 241001, Anhui, China
| | - Xiaohai Zhang
- Second People's Hospital of Wuhu City, 241001, Anhui, China
| | - Taibao Xia
- Second People's Hospital of Wuhu City, 241001, Anhui, China
| | - Joseph Opeoluwa Ashaolu
- Department of Public Health, Faculty of Basic Medical Sciences, Redeemers University, PMB 230, Ede, Osun State, Nigeria
| | | | - Tolulope Joshua Ashaolu
- Institute for Global Health Innovations, Duy Tan University, Da Nang, 550000, Viet Nam
- Faculty of Medicine, Duy Tan University, Da Nang, 550000, Viet Nam
| |
Collapse
|
3
|
Qu Y, Li D, Liu W, Shi D. Molecular consideration relevant to the mechanism of the comorbidity between psoriasis and systemic lupus erythematosus (Review). Exp Ther Med 2023; 26:482. [PMID: 37745036 PMCID: PMC10515117 DOI: 10.3892/etm.2023.12181] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Accepted: 08/03/2023] [Indexed: 09/26/2023] Open
Abstract
Systemic lupus erythematosus (SLE), a common autoimmune disease with a global incidence and newly diagnosed population estimated at 5.14 (range, 1.4-15.13) per 100,000 person-years and 0.40 million people annually, respectively, affects multiple tissues and organs; for example, skin, blood system, heart and kidneys. Accumulating data has also demonstrated that psoriasis (PS) can be a systemic inflammatory disease, which can affect organs other than the skin and occur alongside other autoimmune diseases, such as inflammatory bowel disease, multiple sclerosis, rheumatoid arthritis and SLE. The current explanations for the possible comorbidity of PS and SLE include: i) The two diseases share susceptible gene loci; ii) they share a common IL-23/T helper 17 (Th17) axis inflammatory pathway; and iii) the immunopathogenesis of the two conditions is a consequence of the interactions between IL-17 cytokines with effector Th17 cells, T regulatory cells, as well as B cells. In addition, the therapeutic efficacy of IL-17 or TNF-α inhibitors has been demonstrated in PS, and has also become evident in SLE. However, the mechanisms have not been investigated. To the best of our knowledge, there remains a lack of substantial studies on the correlation between PS and SLE. In the present review, the literature, with regards to the epidemiology, genetic predisposition, inflammatory mechanisms and treatment of the patients with both PS and SLE, has been reviewed. Further investigations into the molecular pathogenic mechanism may provide drug targets that could benefit the patients with concomitant PS and SLE.
Collapse
Affiliation(s)
- Yuying Qu
- Department of Dermatology, College of Clinical Medicine, Jining Medical University, Jining, Shandong 272067, P.R. China
| | - Dongmei Li
- Department of Microbiology and Immunology, Georgetown University Medical Center, Washington, DC 20057, USA
| | - Weida Liu
- Department of Medical Mycology, Chinese Academy of Medical Sciences Institute of Dermatology, Nanjing, Jiangsu 272002, P.R. China
| | - Dongmei Shi
- Department of Dermatology, Jining No. 1 People's Hospital, Jining, Shandong 272011, P.R. China
| |
Collapse
|
4
|
Alamino VA, Onofrio LI, Acosta CDV, Ferrero PV, Zacca ER, Cadile II, Mussano ED, Onetti LB, Montes CL, Gruppi A, Acosta Rodriguez EV. Tofacitinib treatment of rheumatoid arthritis increases senescent T cell frequency in patients and limits T cell function in vitro. Eur J Immunol 2023; 53:e2250353. [PMID: 37179252 PMCID: PMC10524217 DOI: 10.1002/eji.202250353] [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: 12/26/2022] [Revised: 04/11/2023] [Accepted: 05/11/2023] [Indexed: 05/15/2023]
Abstract
Unraveling the immune signatures in rheumatoid arthritis (RA) patients receiving various treatment regimens can aid in comprehending the immune mechanisms' role in treatment efficacy and side effects. Given the critical role of cellular immunity in RA pathogenesis, we sought to identify T-cell profiles characterizing RA patients under specific treatments. We compared 75 immunophenotypic and biochemical variables in healthy donors (HD) and RA patients, including those receiving different treatments as well as treatment-free patients. Additionally, we conducted in vitro experiments to evaluate the direct effect of tofacitinib on purified naïve and memory CD4+ and CD8+ T cells. Multivariate analysis revealed that tofacitinib-treated patients segregated from HD at the expense of T-cell activation, differentiation, and effector function-related variables. Additionally, tofacitinib led to an accumulation of peripheral senescent memory CD4+ and CD8+ T cells. In vitro, tofacitinib impaired the activation, proliferation, and effector molecules expression and triggered senescence pathways in T-cell subsets upon TCR-engagement, with the most significant impact on memory CD8+ T cells. Our findings suggest that tofacitinib may activate immunosenescence pathways while simultaneously inhibiting effector functions in T cells, both effects likely contributing to the high clinical success and reported side effects of this JAK inhibitor in RA.
Collapse
Affiliation(s)
- Vanina A Alamino
- Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI-CONICET), Facultad de Ciencias Químicas, Universidad Nacional de Córdoba (UNC), Córdoba, Argentina
- Laboratorio de Inmunología, Hospital Nacional de Clínicas, UNC, Córdoba, Argentina
| | - Luisina I Onofrio
- Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI-CONICET), Facultad de Ciencias Químicas, Universidad Nacional de Córdoba (UNC), Córdoba, Argentina
- Laboratorio de Inmunología, Hospital Nacional de Clínicas, UNC, Córdoba, Argentina
| | | | - Paola V Ferrero
- Laboratorio de Inmunología, Hospital Nacional de Clínicas, UNC, Córdoba, Argentina
| | - Estefanía R Zacca
- Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI-CONICET), Facultad de Ciencias Químicas, Universidad Nacional de Córdoba (UNC), Córdoba, Argentina
- Laboratorio de Inmunología, Hospital Nacional de Clínicas, UNC, Córdoba, Argentina
| | - Isaac I Cadile
- Servicio de Reumatología, Hospital Nacional de Clínicas, UNC, Córdoba, Argentina
| | - Eduardo D Mussano
- Servicio de Reumatología, Hospital Nacional de Clínicas, UNC, Córdoba, Argentina
| | - Laura B Onetti
- Servicio de Reumatología, Hospital Nacional de Clínicas, UNC, Córdoba, Argentina
| | - Carolina L Montes
- Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI-CONICET), Facultad de Ciencias Químicas, Universidad Nacional de Córdoba (UNC), Córdoba, Argentina
| | - Adriana Gruppi
- Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI-CONICET), Facultad de Ciencias Químicas, Universidad Nacional de Córdoba (UNC), Córdoba, Argentina
| | - Eva V Acosta Rodriguez
- Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI-CONICET), Facultad de Ciencias Químicas, Universidad Nacional de Córdoba (UNC), Córdoba, Argentina
| |
Collapse
|
5
|
Lama M, Sarkar R, Ghosh B. Serum Cytokine Profiles in Patients with Rheumatoid Arthritis Before and After Treatment with Methotrexate. J Interferon Cytokine Res 2023; 43:344-350. [PMID: 37566477 DOI: 10.1089/jir.2023.0078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/13/2023] Open
Abstract
Rheumatoid arthritis (RA) is an inflammatory autoimmune illness affecting around 1% of the population globally. Cytokines have a crucial role in the pathogenesis of RA. The objectives of the present study were to compare the serum cytokine profiles between methotrexate (MTX)-treated and MTX-naive RA patient groups, MTX-treated RA patient group and healthy controls, and MTX-naive RA patient group and healthy controls. Enzyme linked immunosorbent assay (ELISA) kits were used to quantify the serum concentrations of tumor necrosis factor-alpha (TNF-α), interleukin (IL)-1β, IL-17, IL-6, interferon-gamma (IFN-γ), and IL-10 in 80 RA patients (48 MTX treated and 32 MTX naive) and 80 healthy controls. For all cytokine assays, absorbance was measured at 450 nm using a microplate reader (Bio-Rad). Independent sample t-test was used to compare the serum cytokine concentrations between the study groups using SPSS version 25. MTX-treated RA patient group had significantly reduced serum levels of TNF-α (36.13 ± 17.64 versus 45.82 ± 23.07, *P = 0.037), IL-17 (307.85 ± 151.74 versus 435.42 ± 241.19, **P = 0.006), and IFN-γ (414.93 ± 212.13 versus 527.15 ± 269.61, *P = 0.041) compared to MTX-naive RA patients. Both MTX-treated and MTX-naive RA patient groups had significantly high serum levels of TNF-α, IL-1β, IL-17, IL-6, IFN-γ, and IL-10 when compared to healthy controls (***P < 0.001). Downregulation of the serum concentrations of certain key cytokines, viz. TNF-α, IL-17, and IFN-γ, demonstrates the anti-inflammatory effect of MTX in RA patients.
Collapse
Affiliation(s)
- Manoj Lama
- Molecular Immunology Laboratory, Department of Zoology, University of Gour Banga, Malda, India
| | - Rajat Sarkar
- Molecular Immunology Laboratory, Department of Zoology, University of Gour Banga, Malda, India
| | - Bappaditya Ghosh
- Department of Orthopaedics, Malda Medical College and Hospital, Malda, India
| |
Collapse
|
6
|
Peng X, Wang Q, Li W, Ge G, Peng J, Xu Y, Yang H, Bai J, Geng D. Comprehensive overview of microRNA function in rheumatoid arthritis. Bone Res 2023; 11:8. [PMID: 36690624 PMCID: PMC9870909 DOI: 10.1038/s41413-023-00244-1] [Citation(s) in RCA: 21] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2022] [Revised: 11/15/2022] [Accepted: 12/04/2022] [Indexed: 01/25/2023] Open
Abstract
MicroRNAs (miRNAs), a class of endogenous single-stranded short noncoding RNAs, have emerged as vital epigenetic regulators of both pathological and physiological processes in animals. They direct fundamental cellular pathways and processes by fine-tuning the expression of multiple genes at the posttranscriptional level. Growing evidence suggests that miRNAs are implicated in the onset and development of rheumatoid arthritis (RA). RA is a chronic inflammatory disease that mainly affects synovial joints. This common autoimmune disorder is characterized by a complex and multifaceted pathogenesis, and its morbidity, disability and mortality rates remain consistently high. More in-depth insights into the underlying mechanisms of RA are required to address unmet clinical needs and optimize treatment. Herein, we comprehensively review the deregulated miRNAs and impaired cellular functions in RA to shed light on several aspects of RA pathogenesis, with a focus on excessive inflammation, synovial hyperplasia and progressive joint damage. This review also provides promising targets for innovative therapies of RA. In addition, we discuss the regulatory roles and clinical potential of extracellular miRNAs in RA, highlighting their prospective applications as diagnostic and predictive biomarkers.
Collapse
Affiliation(s)
- Xiaole Peng
- grid.429222.d0000 0004 1798 0228Department of Orthopedics, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, 215006 Jiangsu P. R. China
| | - Qing Wang
- grid.429222.d0000 0004 1798 0228Department of Orthopedics, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, 215006 Jiangsu P. R. China
| | - Wenming Li
- grid.429222.d0000 0004 1798 0228Department of Orthopedics, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, 215006 Jiangsu P. R. China
| | - Gaoran Ge
- grid.429222.d0000 0004 1798 0228Department of Orthopedics, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, 215006 Jiangsu P. R. China
| | - Jiachen Peng
- grid.413390.c0000 0004 1757 6938Department of Orthopedics, Affiliated Hospital of Zunyi Medical University, 563000 Zunyi, P. R. China
| | - Yaozeng Xu
- grid.429222.d0000 0004 1798 0228Department of Orthopedics, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, 215006 Jiangsu P. R. China
| | - Huilin Yang
- grid.429222.d0000 0004 1798 0228Department of Orthopedics, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, 215006 Jiangsu P. R. China
| | - Jiaxiang Bai
- grid.429222.d0000 0004 1798 0228Department of Orthopedics, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, 215006 Jiangsu P. R. China
| | - Dechun Geng
- grid.429222.d0000 0004 1798 0228Department of Orthopedics, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, 215006 Jiangsu P. R. China
| |
Collapse
|
7
|
Mijnheer G, Servaas NH, Leong JY, Boltjes A, Spierings E, Chen P, Lai L, Petrelli A, Vastert S, de Boer RJ, Albani S, Pandit A, van Wijk F. Compartmentalization and persistence of dominant (regulatory) T cell clones indicates antigen skewing in juvenile idiopathic arthritis. eLife 2023; 12:79016. [PMID: 36688525 PMCID: PMC9995115 DOI: 10.7554/elife.79016] [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: 03/29/2022] [Accepted: 01/20/2023] [Indexed: 01/24/2023] Open
Abstract
Autoimmune inflammation is characterized by tissue infiltration and expansion of antigen-specific T cells. Although this inflammation is often limited to specific target tissues, it remains yet to be explored whether distinct affected sites are infiltrated with the same, persistent T cell clones. Here, we performed CyTOF analysis and T cell receptor (TCR) sequencing to study immune cell composition and (hyper-)expansion of circulating and joint-derived Tregs and non-Tregs in juvenile idiopathic arthritis (JIA). We studied different joints affected at the same time, as well as over the course of relapsing-remitting disease. We found that the composition and functional characteristics of immune infiltrates are strikingly similar between joints within one patient, and observed a strong overlap between dominant T cell clones, especially Treg, of which some could also be detected in circulation and persisted over the course of relapsing-remitting disease. Moreover, these T cell clones were characterized by a high degree of sequence similarity, indicating the presence of TCR clusters responding to the same antigens. These data suggest that in localized autoimmune disease, there is autoantigen-driven expansion of both Teffector and Treg clones that are highly persistent and are (re)circulating. These dominant clones might represent interesting therapeutic targets.
Collapse
Affiliation(s)
- Gerdien Mijnheer
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht UniversityUtrechtNetherlands
| | - Nila Hendrika Servaas
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht UniversityUtrechtNetherlands
| | - Jing Yao Leong
- Translational Immunology Institute, Singhealth/Duke-NUS Academic Medical Centre, the AcademiaSingaporeSingapore
| | - Arjan Boltjes
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht UniversityUtrechtNetherlands
| | - Eric Spierings
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht UniversityUtrechtNetherlands
| | - Phyllis Chen
- Translational Immunology Institute, Singhealth/Duke-NUS Academic Medical Centre, the AcademiaSingaporeSingapore
| | - Liyun Lai
- Translational Immunology Institute, Singhealth/Duke-NUS Academic Medical Centre, the AcademiaSingaporeSingapore
| | - Alessandra Petrelli
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht UniversityUtrechtNetherlands
| | - Sebastiaan Vastert
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht UniversityUtrechtNetherlands
- Pediatric Immunology & Rheumatology, Wilhelmina Children’s Hospital, University Medical Center Utrecht, Utrecht UniversityUtrechtNetherlands
| | - Rob J de Boer
- Theoretical Biology, Utrecht UniversityUtrechtNetherlands
| | - Salvatore Albani
- Translational Immunology Institute, Singhealth/Duke-NUS Academic Medical Centre, the AcademiaSingaporeSingapore
| | - Aridaman Pandit
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht UniversityUtrechtNetherlands
| | - Femke van Wijk
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht UniversityUtrechtNetherlands
| |
Collapse
|
8
|
Fernández-Ruiz JC, Ochoa-González FDL, Zapata-Zúñiga M, Mondragon-Marín E, Lara-Ramírez EE, Ruíz-Carrillo JL, DelaCruz-Flores PA, Layseca-Espinosa E, Enciso-Moreno JA, Castañeda-Delgado JE. GPR15 expressed in T lymphocytes from RA patients is involved in leukocyte chemotaxis to the synovium. J Leukoc Biol 2022; 112:1209-1221. [PMID: 36164808 DOI: 10.1002/jlb.3ma0822-263rr] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 08/26/2022] [Accepted: 09/02/2022] [Indexed: 12/24/2022] Open
Abstract
The rheumatoid arthritis (RA) inflammatory process occurs in the joints where immune cells are attracted into the synovium to promote remodeling and tissue damage. GPR15 is a G protein-coupled receptor (GPCR) located on chromosome 3 and has similarity in its sequence with chemokine receptors. Recent evidence indicates that GPR15 may be associated with modulation of the chronic inflammatory response. We evaluated the expression of GPR15 and GPR15L in blood and synovial tissue samples from RA patients, as well as to perform a functional migration assay in response to GPR15L. The expression of GPR15 and c10orf99/gpr15l mRNA was analyzed by RT-qPCR. Samples of synovial fluid and peripheral blood were analyzed for CD45+CD3+CD4+GPR15+ and CD45+CD3+CD8+GPR15+ T cell frequency comparing RA patients versus control subjects by flow cytometry. Migration assays were performed using PBMCs isolated from these individuals in response to the synthetic GPR15 ligand. Statistical analysis included Kruskal-Wallis test, T-test, or Mann-Whitney U test, according to data distribution. A higher expression in the mRNA for GPR15 was identified in early RA subjects. The frequencies of CD4+/CD8+ GPR15+ T lymphocytes are higher in RA patients comparing with healthy subjects. Also, the frequency CD4+/CD8+ GPR15+ T lymphocytes are higher in synovial fluid of established RA patients comparing with OA patients. GPR15 and GPR15L are present in the synovial tissue of RA patients and GPR15L promotes migration of PBMCs from RA patients and healthy subjects. Our results suggest that GPR15/GPR15L have a pathogenic role in RA and their antagonizing could be a therapeutic approach in RA.
Collapse
Affiliation(s)
- Julio Cesar Fernández-Ruiz
- Unidad de Investigación Biomédica de Zacatecas, Instituto Mexicano del Seguro Social (IMSS), Zacatecas, Zacatecas, México.,Centro de Investigación en Ciencias de la Salud y Biomedicina, Univerisidad Autónoma de San Luis Potosí, San Luis Potosí, San Luis Potosí, México
| | - Fátima de Lourdes Ochoa-González
- Unidad de Investigación Biomédica de Zacatecas, Instituto Mexicano del Seguro Social (IMSS), Zacatecas, Zacatecas, México.,Doctorado en ciencias básicas, Universidad Autónoma de Zacatecas, Zacatecas, Zacatecas, México.,Área de Ciencias de la Salud, Universidad Autónoma de Zacatecas, Zacatecas, Zacatecas, México
| | - Martín Zapata-Zúñiga
- Hospital Rural No. 51 IMSS Bienestar, Villanueva, Zacatecas, México.,Unidad Académica de Medicina Humana y Ciencias de la Salud, Universidad Autónoma de Zacatecas, Zacatecas, Zacatecas, México
| | - Eduardo Mondragon-Marín
- Unidad de traumatología y ortopedia, Hospital general del Instituto Mexicano del Seguro Social Zacatecas "Emilio Varela Luján", Zacatecas, Zacatecas, México
| | - Edgar E Lara-Ramírez
- Unidad de Investigación Biomédica de Zacatecas, Instituto Mexicano del Seguro Social (IMSS), Zacatecas, Zacatecas, México
| | - Jose Luis Ruíz-Carrillo
- Unidad de Investigación Biomédica de Zacatecas, Instituto Mexicano del Seguro Social (IMSS), Zacatecas, Zacatecas, México.,Centro de Investigación en Ciencias de la Salud y Biomedicina, Univerisidad Autónoma de San Luis Potosí, San Luis Potosí, San Luis Potosí, México
| | - Paola Amayrani DelaCruz-Flores
- Unidad de Investigación Biomédica de Zacatecas, Instituto Mexicano del Seguro Social (IMSS), Zacatecas, Zacatecas, México
| | - Esther Layseca-Espinosa
- Centro de Investigación en Ciencias de la Salud y Biomedicina, Univerisidad Autónoma de San Luis Potosí, San Luis Potosí, San Luis Potosí, México
| | - José Antonio Enciso-Moreno
- Unidad de Investigación Biomédica de Zacatecas, Instituto Mexicano del Seguro Social (IMSS), Zacatecas, Zacatecas, México.,Maestría en química clínica diagnóstica, Facultad de Química, Universidad Autónoma de Querétaro, Santiago de Queretáro, Querétaro, México
| | - Julio Enrique Castañeda-Delgado
- Unidad de Investigación Biomédica de Zacatecas, Instituto Mexicano del Seguro Social (IMSS), Zacatecas, Zacatecas, México.,Cátedras CONACYT, Consejo Nacional de Ciencia y Tecnología, Ciudad de México, México
| |
Collapse
|
9
|
How does age determine the development of human immune-mediated arthritis? Nat Rev Rheumatol 2022; 18:501-512. [PMID: 35948692 PMCID: PMC9363867 DOI: 10.1038/s41584-022-00814-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/01/2022] [Indexed: 11/08/2022]
Abstract
Does age substantially affect the emergence of human immune-mediated arthritis? Children do not usually develop immune-mediated articular inflammation during their first year of life. In patients with juvenile idiopathic arthritis, this apparent ‘immune privilege’ disintegrates, and chronic inflammation is associated with variable autoantibody signatures and patterns of disease that resemble adult arthritis phenotypes. Numerous mechanisms might be involved in this shift, including genetic and epigenetic predisposing factors, maturation of the immune system with a progressive modulation of putative tolerogenic controls, parallel development of microbial dysbiosis, accumulation of a pro-inflammatory burden driven by environmental exposures (the exposome) and comorbidity-related drivers. By exploring these mechanisms, we expand the discussion of three (not mutually exclusive) hypotheses on how these factors can contribute to the differences and similarities between the loss of immune tolerance in children and the development of established immune-mediated arthritis in adults. These three hypotheses relate to a critical window in genetics and epigenetics, immune maturation, and the accumulation of burden. The varied manifestation of the underlying mechanisms among individuals is only beginning to be clarified, but the establishment of a framework can facilitate the development of an integrated understanding of the pathogenesis of arthritis across all ages. In this Review, the authors discuss age-related arthropathy and the similarities and differences between childhood loss of immune tolerance and adult development of immune-mediated arthritis, and develop three hypotheses describing age-related mechanisms that contribute to the onset of arthritis. The arthritis-free ‘immune privilege’ of early childhood is overridden by multiple mechanisms, progressively and age-dependently, generating recognizable patterns of chronic inflammatory arthritis. The emergence of arthritis involves interconnected mechanisms related to immune priming, to a situational susceptibility and to the accumulation of an inflammatory burden. The accumulation of epigenetic drift may contribute to differences across ages. The exposome is expected to contribute to arthritis emergence in adults as well as in children.
Collapse
|
10
|
West HC, Davies J, Henderson S, Adegun OK, Ward S, Ferrer IR, Tye CA, Vallejo AF, Jardine L, Collin M, Polak ME, Bennett CL. Loss of T cell tolerance in the skin following immunopathology is linked to failed restoration of the dermal niche by recruited macrophages. Cell Rep 2022; 39:110819. [PMID: 35584681 DOI: 10.1016/j.celrep.2022.110819] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 03/17/2022] [Accepted: 04/22/2022] [Indexed: 11/03/2022] Open
Abstract
T cell pathology in the skin leads to monocyte influx, but we have little understanding of the fate of recruited cells within the diseased niche, or the long-term impact on cutaneous immune homeostasis. By combining a murine model of acute graft-versus-host disease (aGVHD) with analysis of patient samples, we demonstrate that pathology initiates dermis-specific macrophage differentiation and show that aGVHD-primed macrophages continue to dominate the dermal compartment at the relative expense of quiescent MHCIIint cells. Exposure of the altered dermal niche to topical haptens after disease resolution results in hyper-activation of regulatory T cells (Treg), but local breakdown in tolerance. Disease-imprinted macrophages express increased IL-1β and are predicted to elicit altered TNF superfamily interactions with cutaneous Treg, and we demonstrate the direct loss of T cell regulation within the resolved skin. Thus, T cell pathology leaves an immunological scar in the skin marked by failure to re-set immune homeostasis.
Collapse
Affiliation(s)
- Heather C West
- Department of Haematology, University College London (UCL) Cancer Institute, London WC1E 6DD, UK; Institute for Immunity and Transplantation, Division of Infection and Immunity, University College London, London NW3 2PF, UK
| | - James Davies
- Department of Haematology, University College London (UCL) Cancer Institute, London WC1E 6DD, UK; Institute for Immunity and Transplantation, Division of Infection and Immunity, University College London, London NW3 2PF, UK
| | - Stephen Henderson
- Bill Lyons Informatics Centre, Cancer Institute, University College London, London WC1E 6DD, UK
| | - Oluyori K Adegun
- Department of Cellular Pathology, University College London Hospitals NHS Foundation Trust, London, UK
| | - Sophie Ward
- Department of Haematology, University College London (UCL) Cancer Institute, London WC1E 6DD, UK; Institute for Immunity and Transplantation, Division of Infection and Immunity, University College London, London NW3 2PF, UK
| | - Ivana R Ferrer
- Department of Haematology, University College London (UCL) Cancer Institute, London WC1E 6DD, UK; Institute for Immunity and Transplantation, Division of Infection and Immunity, University College London, London NW3 2PF, UK
| | - Chanidapa A Tye
- Department of Haematology, University College London (UCL) Cancer Institute, London WC1E 6DD, UK; Institute for Immunity and Transplantation, Division of Infection and Immunity, University College London, London NW3 2PF, UK
| | - Andres F Vallejo
- Clinical and Experimental Sciences (Sir Henry Wellcome Laboratories, Faculty of Medicine) and Institute for Life Sciences, University of Southampton, Southampton, UK
| | - Laura Jardine
- Biosciences Institute, Newcastle University, Newcastle Upon Tyne, UK
| | - Matthew Collin
- Newcastle University Translational and Clinical Research Institute and NIHR Newcastle Biomedical Research Centre, Newcastle University, Newcastle Upon Tyne, UK
| | - Marta E Polak
- Clinical and Experimental Sciences (Sir Henry Wellcome Laboratories, Faculty of Medicine) and Institute for Life Sciences, University of Southampton, Southampton, UK
| | - Clare L Bennett
- Department of Haematology, University College London (UCL) Cancer Institute, London WC1E 6DD, UK; Institute for Immunity and Transplantation, Division of Infection and Immunity, University College London, London NW3 2PF, UK.
| |
Collapse
|
11
|
Gottwick C, Carambia A, Herkel J. Harnessing the liver to induce antigen-specific immune tolerance. Semin Immunopathol 2022; 44:475-484. [PMID: 35513495 PMCID: PMC9256566 DOI: 10.1007/s00281-022-00942-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Accepted: 04/20/2022] [Indexed: 12/17/2022]
Abstract
Autoimmune diseases develop when the adaptive immune system attacks the body’s own antigens leading to tissue damage. At least 80 different conditions are believed to have an autoimmune aetiology, including rheumatoid arthritis, type I diabetes, multiple sclerosis or systemic lupus erythematosus. Collectively, autoimmune diseases are a leading cause of severe health impairment along with substantial socioeconomic costs. Current treatments are mostly symptomatic and non-specific, and it is typically not possible to cure these diseases. Thus, the development of more causative treatments that suppress only the pathogenic immune responses, but spare general immunity is of great biomedical interest. The liver offers considerable potential for development of such antigen-specific immunotherapies, as it has a distinct physiological capacity to induce immune tolerance. Indeed, the liver has been shown to specifically suppress autoimmune responses to organ allografts co-transplanted with the liver or to autoantigens that were transferred to the liver. Liver tolerance is established by a unique microenvironment that facilitates interactions between liver-resident antigen-presenting cells and lymphocytes passing by in the low blood flow within the hepatic sinusoids. Here, we summarise current concepts and mechanisms of liver immune tolerance, and review present approaches to harness liver tolerance for antigen-specific immunotherapy.
Collapse
Affiliation(s)
- Cornelia Gottwick
- First Department of Medicine, University Medical Centre Hamburg-Eppendorf, Hamburg, Germany.
| | - Antonella Carambia
- First Department of Medicine, University Medical Centre Hamburg-Eppendorf, Hamburg, Germany.
| | - Johannes Herkel
- First Department of Medicine, University Medical Centre Hamburg-Eppendorf, Hamburg, Germany.
| |
Collapse
|
12
|
Jang S, Kwon EJ, Lee JJ. Rheumatoid Arthritis: Pathogenic Roles of Diverse Immune Cells. Int J Mol Sci 2022; 23:ijms23020905. [PMID: 35055087 PMCID: PMC8780115 DOI: 10.3390/ijms23020905] [Citation(s) in RCA: 148] [Impact Index Per Article: 74.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 01/11/2022] [Accepted: 01/12/2022] [Indexed: 02/06/2023] Open
Abstract
Rheumatoid arthritis (RA) is a chronic, systemic autoimmune disease associated with synovial tissue proliferation, pannus formation, cartilage destruction, and systemic complications. Currently, advanced understandings of the pathologic mechanisms of autoreactive CD4+ T cells, B cells, macrophages, inflammatory cytokines, chemokines, and autoantibodies that cause RA have been achieved, despite the fact that much remains to be elucidated. This review provides an updated pathogenesis of RA which will unveil novel therapeutic targets.
Collapse
Affiliation(s)
- Sunhee Jang
- Division of Rheumatology, Department of Internal Medicine, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea; (S.J.); (E.-J.K.)
- Yonsei Hangang Hospital, 25 Mapodaero, Mapogu, Seoul 04167, Korea
| | - Eui-Jong Kwon
- Division of Rheumatology, Department of Internal Medicine, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea; (S.J.); (E.-J.K.)
- Chemical, Biological, Radiological, and Nuclear (CBRN) Defense Research Institute, Armed Forces CBRN Defense Command, Seoul 06591, Korea
| | - Jennifer Jooha Lee
- Division of Rheumatology, Department of Internal Medicine, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea; (S.J.); (E.-J.K.)
- Correspondence: ; Tel.: +82-2-2258-6010; Fax: +82-2-2258-2022
| |
Collapse
|
13
|
Lee HR, Yoo SJ, Kim J, Park CK, Kang SW. Reduction of Oxidative Stress in Peripheral Blood Mononuclear Cells Attenuates the Inflammatory Response of Fibroblast-like Synoviocytes in Rheumatoid Arthritis. Int J Mol Sci 2021; 22:ijms222212411. [PMID: 34830290 PMCID: PMC8624216 DOI: 10.3390/ijms222212411] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Revised: 11/12/2021] [Accepted: 11/15/2021] [Indexed: 12/29/2022] Open
Abstract
The production and oxidation mechanism of reactive oxygen species (ROS) are out of balance in rheumatoid arthritis (RA). However, the correlation between ROS and T cell subsets in RA remains unclear. Peripheral blood mononuclear cells (PBMCs) from patients with RA (n = 40) and healthy controls (n = 10) were isolated from whole blood samples. Synovial tissues (n = 3) and synovial fluid (n = 10) were obtained from patients with RA. The repartition of T cell subsets and expression of ROS and cytokines were examined according to RA severity. Fibroblast-like synoviocytes (FLSs) from patients with RA were stimulated with PBMCs and the expression of inflammation-related molecules were measured by RT-PCR and cytokine array. Regulatory T cells from patients with moderate (5.1 > DAS28 ≥ 3.2) RA showed the highest expression of mitochondrial ROS among the groups based on disease severity. Although ROS levels steadily increased with RA severity, there was a slight decline in severe RA (DAS28 ≥ 5.1) compared with moderate RA. The expression of inflammatory cytokines in RA FLSs were significantly inhibited when FLSs were co-cultured with PBMCs treated with ROS inhibitor. These findings provide a novel approach to suppress inflammatory response of FLSs through ROS regulation in PBMCs.
Collapse
Affiliation(s)
- Ha-Reum Lee
- Division of Rheumatology, Department of Internal Medicine, Chungnam National University Hospital, 282 Munhwaro, Daejeon 35015, Korea; (H.-R.L.); (S.-J.Y.); (J.K.)
- Research Institute for Medical Sciences, School of Medicine, Chungnam National University, 266 Munhwaro, Daejeon 35015, Korea
| | - Su-Jin Yoo
- Division of Rheumatology, Department of Internal Medicine, Chungnam National University Hospital, 282 Munhwaro, Daejeon 35015, Korea; (H.-R.L.); (S.-J.Y.); (J.K.)
| | - Jinhyun Kim
- Division of Rheumatology, Department of Internal Medicine, Chungnam National University Hospital, 282 Munhwaro, Daejeon 35015, Korea; (H.-R.L.); (S.-J.Y.); (J.K.)
| | - Chan Keol Park
- Division of Rheumatology, Department of Internal Medicine, Chungnam National University Sejong Hospital, 20 Bodeum-7-ro, Sejong 30099, Korea;
| | - Seong Wook Kang
- Division of Rheumatology, Department of Internal Medicine, Chungnam National University Hospital, 282 Munhwaro, Daejeon 35015, Korea; (H.-R.L.); (S.-J.Y.); (J.K.)
- Research Institute for Medical Sciences, School of Medicine, Chungnam National University, 266 Munhwaro, Daejeon 35015, Korea
- Correspondence: ; Tel.: +82-42-338-2428
| |
Collapse
|
14
|
Samat AAK, van der Geest J, Vastert SJ, van Loosdregt J, van Wijk F. Tissue-Resident Memory T Cells in Chronic Inflammation-Local Cells with Systemic Effects? Cells 2021; 10:409. [PMID: 33669367 PMCID: PMC7920248 DOI: 10.3390/cells10020409] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Revised: 02/09/2021] [Accepted: 02/10/2021] [Indexed: 02/06/2023] Open
Abstract
Chronic inflammatory diseases such as rheumatoid arthritis (RA), Juvenile Idiopathic Arthritis (JIA), psoriasis, and inflammatory bowel disease (IBD) are characterized by systemic as well as local tissue inflammation, often with a relapsing-remitting course. Tissue-resident memory T cells (TRM) enter non-lymphoid tissue (NLT) as part of the anamnestic immune response, especially in barrier tissues, and have been proposed to fuel chronic inflammation. TRM display a distinct gene expression profile, including upregulation of CD69 and downregulation of CD62L, CCR7, and S1PR1. However, not all TRM are consistent with this profile, and it is now more evident that the TRM compartment comprises a heterogeneous population, with differences in their function and activation state. Interestingly, the paradigm of TRM remaining resident in NLT has also been challenged. T cells with TRM characteristics were identified in both lymph and circulation in murine and human studies, displaying similarities with circulating memory T cells. This suggests that re-activated TRM are capable of retrograde migration from NLT via differential gene expression, mediating tissue egress and circulation. Circulating 'ex-TRM' retain a propensity for return to NLT, especially to their tissue of origin. Additionally, memory T cells with TRM characteristics have been identified in blood from patients with chronic inflammatory disease, leading to the hypothesis that TRM egress from inflamed tissue as well. The presence of TRM in both tissue and circulation has important implications for the development of novel therapies targeting chronic inflammation, and circulating 'ex-TRM' may provide a vital diagnostic tool in the form of biomarkers. This review elaborates on the recent developments in the field of TRM in the context of chronic inflammatory diseases.
Collapse
Affiliation(s)
- Anoushka Ashok Kumar Samat
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht University, 3584 CX Utrecht, The Netherlands; (A.A.K.S.); (J.v.d.G.); (S.J.V.); (J.v.L.)
| | - Jolijn van der Geest
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht University, 3584 CX Utrecht, The Netherlands; (A.A.K.S.); (J.v.d.G.); (S.J.V.); (J.v.L.)
| | - Sebastiaan J. Vastert
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht University, 3584 CX Utrecht, The Netherlands; (A.A.K.S.); (J.v.d.G.); (S.J.V.); (J.v.L.)
- Paediatric Rheumatology and Immunology, Wilhelmina Children’s Hospital, Utrecht University, 3584 EA Utrecht, The Netherlands
| | - Jorg van Loosdregt
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht University, 3584 CX Utrecht, The Netherlands; (A.A.K.S.); (J.v.d.G.); (S.J.V.); (J.v.L.)
| | - Femke van Wijk
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht University, 3584 CX Utrecht, The Netherlands; (A.A.K.S.); (J.v.d.G.); (S.J.V.); (J.v.L.)
| |
Collapse
|
15
|
Present Status and Future Trends of Natural-Derived Compounds Targeting T Helper (Th) 17 and Microsomal Prostaglandin E Synthase-1 (mPGES-1) as Alternative Therapies for Autoimmune and Inflammatory-Based Diseases. Molecules 2020; 25:molecules25246016. [PMID: 33353211 PMCID: PMC7766998 DOI: 10.3390/molecules25246016] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2020] [Revised: 12/07/2020] [Accepted: 12/17/2020] [Indexed: 12/19/2022] Open
Abstract
Several natural-based compounds and products are reported to possess anti-inflammatory and immunomodulatory activity both in vitro and in vivo. The primary target for these activities is the inhibition of eicosanoid-generating enzymes, including phospholipase A2, cyclooxygenases (COXs), and lipoxygenases, leading to reduced prostanoids and leukotrienes. Other mechanisms include modulation of protein kinases and activation of transcriptases. However, only a limited number of studies and reviews highlight the potential modulation of the coupling enzymatic pathway COX-2/mPGES-1 and Th17/Treg circulating cells. Here, we provide a brief overview of natural products/compounds, currently included in the Italian list of botanicals and the BELFRIT, in different fields of interest such as inflammation and immunity. In this context, we focus our opinion on novel therapeutic targets such as COX-2/mPGES-1 coupling enzymes and Th17/Treg circulating repertoire. This paper is dedicated to the scientific career of Professor Nicola Mascolo for his profound dedication to the study of natural compounds.
Collapse
|
16
|
Methrotexate Treatment Inmunomodulates Abnormal Cytokine Expression by T CD4 Lymphocytes Present in DMARD-Naïve Rheumatoid Arthritis Patients. Int J Mol Sci 2020; 21:ijms21186847. [PMID: 32961930 PMCID: PMC7555887 DOI: 10.3390/ijms21186847] [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: 07/27/2020] [Revised: 09/11/2020] [Accepted: 09/14/2020] [Indexed: 01/18/2023] Open
Abstract
CD4+T-lymphocytes are relevant in the pathogenesis of rheumatoid arthritis (RA), however, their potential involvement in early RA remains elusive. Methotrexate (MTX) is a commonly used disease-modifying antirheumatic drug (DMARD), but its mechanism has not been fully established. In 47 new-onset DMARD-naïve RA patients, we investigated the pattern of IFNγ, IL-4 and IL-17A expression by naïve (TN), central (TCM), effector memory (TEM) and effector (TE) CD4+ subsets; their STAT-1, STAT-6 and STAT-3 transcription factors phosphorylation, and the circulating levels of IFNγ, IL-4 and IL-17. We also studied the RA patients after 3 and 6 months of MTX treatment and according their clinical response. CD4+T-lymphocyte subsets and cytokine expression were measured using flow cytometry. New-onset DMARD-naïve RA patients showed a significant expansion of IL-17A+, IFNγ+ and IL-17A+IFNγ+ CD4+T-lymphocyte subsets and increased intracellular STAT-1 and STAT-3 phosphorylation. Under basal conditions, nonresponder patients showed increased numbers of circulating IL-17A producing TN and TMC CD4+T-lymphocytes and IFNγ producing TN, TCM, TEM CD4+T-lymphocytes with respect to responders. After 6 months, the numbers of CD4+IL-17A+TN remained significantly increased in nonresponders. In conclusion, CD4+T-lymphocytes in new-onset DMARD-naïve RA patients show IL-17A and IFNγ abnormalities in TN, indicating their relevant role in early disease pathogenesis. Different patterns of CD4+ modulation are identified in MTX responders and nonresponders.
Collapse
|
17
|
Wu R, Li N, Zhao X, Ding T, Xue H, Gao C, Li X, Wang C. Low-dose Interleukin-2: Biology and therapeutic prospects in rheumatoid arthritis. Autoimmun Rev 2020; 19:102645. [PMID: 32801037 DOI: 10.1016/j.autrev.2020.102645] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Accepted: 03/16/2020] [Indexed: 02/07/2023]
Abstract
Rheumatoid arthritis (RA) is a chronic aggressive arthritis that is characterized with systemic inflammation response, the production of abnormal antibodies, and persistent synovitis. One of the key mechanisms underlying the pathogenesis of RA is the imbalance of CD4 + T lymphocyte subsets, from T helper (Th) 17 cells and regulatory T (Treg) cells to T follicular helper (Tfh) cells and T follicular regulatory (Tfr) cells, which can mediate autoimmune inflammatory response to promote the overproduction of cytokines and abnormal antibodies. Although the treatment of RA has greatly changed due to the discovery of biological agents such as anti-TNF, the remission of it is still not satisfactory, thus, it is urgently required new treatment to realize the sustained remission of RA via restoring the immune tolerance. Interleukin-2 (IL-2) has been discovered to be a pleiotropic cytokine to promote inflammatory response and maintain immune tolerance. Low-dose IL-2 therapy is a driver of the imbalance between autoimmunity and immune tolerance towards immune tolerance, which has been tried to treat various autoimmune diseases. Recent researches show that low-dose IL-2 is a promising treatment for RA. In this review, we summarize the advances understandings in the biology of IL-2 and highlight the impact of the IL-2 pathway on the balance of Th17/Treg and Tfh/Tfr aiming to investigate the role of IL-2-mediated immune tolerance in RA and discuss the application and the therapeutic prospect of low-dose IL-2 in the treatment of RA.
Collapse
Affiliation(s)
- Ruihe Wu
- Department of Rheumatology, the Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Na Li
- Department of Rheumatology, the Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Xiangcong Zhao
- Department of Rheumatology, the Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Tingting Ding
- Department of Rheumatology, the Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Hongwei Xue
- Department of Rheumatology, the Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Chong Gao
- Pathology, Joint Program in Transfusion Medicine, Brigham and Women's Hospital/Children's Hospital, Harvard Medical School, Boston, MA, USA.
| | - Xiaofeng Li
- Department of Rheumatology, the Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Caihong Wang
- Department of Rheumatology, the Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China.
| |
Collapse
|
18
|
Jiang C, Wang H, Xue M, Lin L, Wang J, Cai G, Shen Q. Reprograming of peripheral Foxp3+ regulatory T cell towards Th17-like cell in patients with active systemic lupus erythematosus. Clin Immunol 2019; 209:108267. [DOI: 10.1016/j.clim.2019.108267] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2019] [Revised: 08/30/2019] [Accepted: 10/04/2019] [Indexed: 12/29/2022]
|
19
|
Fibrosis-4 index at diagnosis is associated with all-cause mortality in patients with microscopic polyangiitis and granulomatosis with polyangiitis. BMC Gastroenterol 2019; 19:90. [PMID: 31196158 PMCID: PMC6567497 DOI: 10.1186/s12876-019-1007-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2018] [Accepted: 05/31/2019] [Indexed: 12/28/2022] Open
Abstract
Background The fibrosis-4 index (FIB-4) has been reported to be associated with all-cause mortality in several chronic diseases. In this study, we investigated whether at diagnosis could be associated with all-cause mortality in patients with microscopic polyangiitis (MPA) and granulomatosis with polyangiitis (GPA). Methods We retrospectively reviewed the medical records of 132 MPA and GPA patients without chronic liver diseases. Conventional risk factors included old age (≥ 65 years), male gender, diabetes mellitus (DM) and hypertension (HTN) at diagnosis, and disease-related risk factor included GPA, antineutrophil cytoplasmic antibody, Birmingham vasculitis activity score (BVAS) and five factor score (FFS (2009)). The cut-off of FIB-4 for significant liver fibrosis (S2–4) was set at 1.45. Results The mean age was 57.2 years and 27 patients (20.5%) had significant liver fibrosis (FIB-4 ≥ 1.45). Fifteen patients (11.4%) died during follow-up. In the univariable Cox Hazards model, age ≥ 65 years (Hazard ratio (HR) 5.055), DM (HR 3.446), HTN (HR 4.611), FFS (2009) ≥ 2 (HR 4.849) and FIB-4 ≥ 1.45 (HR 9.958) at diagnosis were significantly associated with all-cause mortality. In the multivariable Cox Hazards model, only FIB-4 at diagnosis ≥1.45 (HR 6.253, 95% confidence interval 1.398, 27.963) was associated with all-cause mortality during the follow-up in patients with MPA and GPA. Conclusions FIB-4 at diagnosis ≥1.45 is an independent predictor of all-cause mortality during follow-up in patients with MPA and GPA, and furthermore its predictive potential is higher than those of conventional and AAV-related risk factors for all-cause mortality. Electronic supplementary material The online version of this article (10.1186/s12876-019-1007-z) contains supplementary material, which is available to authorized users.
Collapse
|
20
|
Mijnheer G, van Wijk F. T-Cell Compartmentalization and Functional Adaptation in Autoimmune Inflammation: Lessons From Pediatric Rheumatic Diseases. Front Immunol 2019; 10:940. [PMID: 31143175 PMCID: PMC6520654 DOI: 10.3389/fimmu.2019.00940] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Accepted: 04/11/2019] [Indexed: 12/30/2022] Open
Abstract
Chronic inflammatory diseases are characterized by a disturbed immune balance leading to recurring episodes of inflammation in specific target tissues, such as the joints in juvenile idiopathic arthritis. The tissue becomes infiltrated by multiple types of immune cell, including high numbers of CD4 and CD8 T-cells, which are mostly effector memory cells. Locally, these T-cells display an environment-adapted phenotype, induced by inflammation- and tissue-specific instructions. Some of the infiltrated T-cells may become tissue resident and play a role in relapses of inflammation. Adaptation to the environment may lead to functional (re)programming of cells and altered cellular interactions and responses. For example, specifically at the site of inflammation both CD4 and CD8 T-cells can become resistant to regulatory T-cell-mediated regulation. In addition, CD8 and CD4 T-cells show a unique profile with pro- and anti-inflammatory features coexisting in the same compartment. Also regulatory T-cells are neither homogeneous nor static in nature and show features of functional differentiation, and plasticity in inflammatory environments. Here we will discuss the recent insights in T-cell functional specialization, regulation, and clonal expansion in local (tissue) inflammation.
Collapse
Affiliation(s)
- Gerdien Mijnheer
- Laboratory of Translational Immunology, Wilhelmina Children's Hospital, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
| | - Femke van Wijk
- Laboratory of Translational Immunology, Wilhelmina Children's Hospital, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
| |
Collapse
|
21
|
Joshi RN, Fernandes SJ, Shang MM, Kiani NA, Gomez-Cabrero D, Tegnér J, Schmidt A. Phosphatase inhibitor PPP1R11 modulates resistance of human T cells toward Treg-mediated suppression of cytokine expression. J Leukoc Biol 2019; 106:413-430. [PMID: 30882958 PMCID: PMC6850362 DOI: 10.1002/jlb.2a0618-228r] [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: 06/18/2018] [Revised: 01/15/2019] [Accepted: 03/07/2019] [Indexed: 12/17/2022] Open
Abstract
Regulatory T cells (Tregs) act as indispensable unit for maintaining peripheral immune tolerance mainly by regulating effector T cells. T cells resistant to suppression by Tregs pose therapeutic challenges in the treatment of autoimmune diseases, while augmenting susceptibility to suppression may be desirable for cancer therapy. To understand the cell intrinsic signals in T cells during suppression by Tregs, we have previously performed a global phosphoproteomic characterization. We revealed altered phosphorylation of protein phosphatase 1 regulatory subunit 11 (PPP1R11; Inhibitor‐3) in conventional T cells upon suppression by Tregs. Here, we show that silencing of PPP1R11 renders T cells resistant toward Treg‐mediated suppression of TCR‐induced cytokine expression. Furthermore, whole‐transcriptome sequencing revealed that PPP1R11 differentially regulates not only the expression of specific T cell stimulation‐induced cytokines but also other molecules and pathways in T cells. We further confirmed the target of PPP1R11, PP1, to augment TCR‐induced cytokine expression. In conclusion, we present PPP1R11 as a novel negative regulator of T cell activation‐induced cytokine expression. Targeting PPP1R11 may have therapeutic potential to regulate the T cell activation status including modulating the susceptibility of T cells toward Treg‐mediated suppression, specifically altering the stimulation‐induced T cell cytokine milieu.
Collapse
Affiliation(s)
- Rubin N Joshi
- Unit of Computational Medicine, Center for Molecular Medicine, Department of Medicine Solna, Karolinska University Hospital and Science for Life Laboratory, Karolinska Institutet, Stockholm, Sweden
| | - Sunjay Jude Fernandes
- Unit of Computational Medicine, Center for Molecular Medicine, Department of Medicine Solna, Karolinska University Hospital and Science for Life Laboratory, Karolinska Institutet, Stockholm, Sweden
| | - Ming-Mei Shang
- Unit of Computational Medicine, Center for Molecular Medicine, Department of Medicine Solna, Karolinska University Hospital and Science for Life Laboratory, Karolinska Institutet, Stockholm, Sweden.,Division of Rheumatology, Department of Medicine Solna, Karolinska University Hospital, Karolinska Institutet, Stockholm, Sweden
| | - Narsis A Kiani
- Unit of Computational Medicine, Center for Molecular Medicine, Department of Medicine Solna, Karolinska University Hospital and Science for Life Laboratory, Karolinska Institutet, Stockholm, Sweden
| | - David Gomez-Cabrero
- Unit of Computational Medicine, Center for Molecular Medicine, Department of Medicine Solna, Karolinska University Hospital and Science for Life Laboratory, Karolinska Institutet, Stockholm, Sweden.,Mucosal and Salivary Biology Division, King's College London Dental Institute, London, United Kingdom.,Translational Bioinformatics Unit, NavarraBiomed, Departamento de Salud-Universidad Pública de Navarra, Pamplona, Navarra, Spain
| | - Jesper Tegnér
- Unit of Computational Medicine, Center for Molecular Medicine, Department of Medicine Solna, Karolinska University Hospital and Science for Life Laboratory, Karolinska Institutet, Stockholm, Sweden.,Biological and Environmental Sciences and Engineering Division, Computer, Electrical and Mathematical Sciences and Engineering Division, King Abdullah University of Science and Technology (KAUST), Thuwal, Saudi Arabia
| | - Angelika Schmidt
- Unit of Computational Medicine, Center for Molecular Medicine, Department of Medicine Solna, Karolinska University Hospital and Science for Life Laboratory, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
22
|
Nijhuis L, Peeters JGC, Vastert SJ, van Loosdregt J. Restoring T Cell Tolerance, Exploring the Potential of Histone Deacetylase Inhibitors for the Treatment of Juvenile Idiopathic Arthritis. Front Immunol 2019; 10:151. [PMID: 30792714 PMCID: PMC6374297 DOI: 10.3389/fimmu.2019.00151] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2018] [Accepted: 01/17/2019] [Indexed: 12/24/2022] Open
Abstract
Juvenile Idiopathic Arthritis (JIA) is characterized by a loss of immune tolerance. Here, the balance between the activity of effector T (Teff) cells and regulatory T (Treg) cells is disturbed resulting in chronic inflammation in the joints. Presently, therapeutic strategies are predominantly aimed at suppressing immune activation and pro-inflammatory effector mechanisms, ignoring the opportunity to also promote tolerance by boosting the regulatory side of the immune balance. Histone deacetylases (HDACs) can deacetylate both histone and non-histone proteins and have been demonstrated to modulate epigenetic regulation as well as cellular signaling in various cell types. Importantly, HDACs are potent regulators of both Teff cell and Treg cell function and can thus be regarded as attractive therapeutic targets in chronic inflammatory arthritis. HDAC inhibitors (HDACi) have proven therapeutic potential in the cancer field, and are presently being explored for their potential in the treatment of autoimmune diseases. Specific HDACi have already been demonstrated to reduce the secretion of pro-inflammatory cytokines by Teff cells, and promote Treg numbers and suppressive capacity in vitro and in vivo. In this review, we outline the role of the different classes of HDACs in both Teff cell and Treg cell function. Furthermore, we will review the effect of different HDACi on T cell tolerance and explore their potential as a therapeutic strategy for the treatment of oligoarticular and polyarticular JIA.
Collapse
Affiliation(s)
- Lotte Nijhuis
- Laboratory of Translational Immunology, Department of Pediatric Immunology & Rheumatology, University Medical Center Utrecht, University of Utrecht, Utrecht, Netherlands
| | - Janneke G C Peeters
- Laboratory of Translational Immunology, Department of Pediatric Immunology & Rheumatology, University Medical Center Utrecht, University of Utrecht, Utrecht, Netherlands
| | - Sebastiaan J Vastert
- Laboratory of Translational Immunology, Department of Pediatric Immunology & Rheumatology, University Medical Center Utrecht, University of Utrecht, Utrecht, Netherlands
| | - Jorg van Loosdregt
- Laboratory of Translational Immunology, Department of Pediatric Immunology & Rheumatology, University Medical Center Utrecht, University of Utrecht, Utrecht, Netherlands
| |
Collapse
|
23
|
Hassouna A, M. Abd Elgwad M, Fahmy H. Stromal Stem Cells: Nature, Biology and Potential Therapeutic Applications. STROMAL CELLS - STRUCTURE, FUNCTION, AND THERAPEUTIC IMPLICATIONS 2019. [DOI: 10.5772/intechopen.77346] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/01/2023]
|
24
|
Kim SU, Kim BK, Park JY, Kim DY, Ahn SH, Park YB, Han KH, Lee SW. Fibrosis-4 index at diagnosis can predict all-cause mortality in patients with rheumatoid arthritis: A retrospective monocentric study. Mod Rheumatol 2019; 30:70-77. [PMID: 30557057 DOI: 10.1080/14397595.2018.1558760] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Affiliation(s)
- Seung Up Kim
- Department of Internal Medicine, Institute of Gastroenterology, Yonsei University College of Medicine, Seoul, Republic of Korea
- Yonsei Liver Center, Severance Hospital, Seoul, Republic of Korea
| | - Beom Kyung Kim
- Department of Internal Medicine, Institute of Gastroenterology, Yonsei University College of Medicine, Seoul, Republic of Korea
- Yonsei Liver Center, Severance Hospital, Seoul, Republic of Korea
| | - Jun Yong Park
- Department of Internal Medicine, Institute of Gastroenterology, Yonsei University College of Medicine, Seoul, Republic of Korea
- Yonsei Liver Center, Severance Hospital, Seoul, Republic of Korea
| | - Do Young Kim
- Department of Internal Medicine, Institute of Gastroenterology, Yonsei University College of Medicine, Seoul, Republic of Korea
- Yonsei Liver Center, Severance Hospital, Seoul, Republic of Korea
| | - Sang Hoon Ahn
- Department of Internal Medicine, Institute of Gastroenterology, Yonsei University College of Medicine, Seoul, Republic of Korea
- Yonsei Liver Center, Severance Hospital, Seoul, Republic of Korea
| | - Yong-Beom Park
- Division of Rheumatology, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
- Yonsei University College of Medicine, Institute for Immunology and Immunological Diseases, Seoul, Republic of Korea
| | - Kwang-Hyub Han
- Department of Internal Medicine, Institute of Gastroenterology, Yonsei University College of Medicine, Seoul, Republic of Korea
- Yonsei Liver Center, Severance Hospital, Seoul, Republic of Korea
| | - Sang-Won Lee
- Division of Rheumatology, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
- Yonsei University College of Medicine, Institute for Immunology and Immunological Diseases, Seoul, Republic of Korea
| |
Collapse
|
25
|
Li Q, Wang B, Mu K, Zhang J. The pathogenesis of thyroid autoimmune diseases: New T lymphocytes – Cytokines circuits beyond the Th1−Th2 paradigm. J Cell Physiol 2018; 234:2204-2216. [PMID: 30246383 DOI: 10.1002/jcp.27180] [Citation(s) in RCA: 73] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2018] [Revised: 05/22/2018] [Accepted: 07/17/2018] [Indexed: 12/24/2022]
Affiliation(s)
- Qian Li
- Department of EndocrinologyJinshan Hospital of Fudan UniversityShanghai China
| | - Bin Wang
- Department of EndocrinologyJinshan Hospital of Fudan UniversityShanghai China
| | - Kaida Mu
- Department of EndocrinologyShanghai University of Medicine & Health Sciences Affiliated Zhoupu HospitalShanghai China
| | - Jin‐An Zhang
- Department of EndocrinologyShanghai University of Medicine & Health Sciences Affiliated Zhoupu HospitalShanghai China
| |
Collapse
|
26
|
Stefanov S, Ganeva M, Stoilov N, Boyadzhieva V, Telcharova-Mihaylovska A. The role of gut microbiota in juvenile idiopathic arthritis. BIOTECHNOL BIOTEC EQ 2018. [DOI: 10.1080/13102818.2018.1512376] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022] Open
Affiliation(s)
- Stefan Stefanov
- Department of Pediatric Rheumatology, University Children’s Hospital, Medical University of Sofia, Sofia, Bulgaria
| | - Margarita Ganeva
- Department of Pediatric Rheumatology, University Children’s Hospital, Medical University of Sofia, Sofia, Bulgaria
| | - Nikolay Stoilov
- Clinic of Rheumatology, University Hospital ‘St. I. Rilski’, Medical University of Sofia, Sofia, Bulgaria
| | - Vladimira Boyadzhieva
- Clinic of Rheumatology, University Hospital ‘St. I. Rilski’, Medical University of Sofia, Sofia, Bulgaria
| | | |
Collapse
|
27
|
Song L, Wang Y, Sui Y, Sun J, Li D, Li G, Liu J, Li T, Shu Q. High Interleukin-37 (IL-37) Expression and Increased Mucin-Domain Containing-3 (TIM-3) on Peripheral T Cells in Patients with Rheumatoid Arthritis. Med Sci Monit 2018; 24:5660-5667. [PMID: 30106887 PMCID: PMC6104553 DOI: 10.12659/msm.909254] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Background Anti-inflammatory mediators such as mucin-domain containing-3 (TIM-3) and IL-37 play an important role in the regulation of Th1-mediated immunity. This study was designed to investigate the proportions of various T cell subsets and monocytes in the peripheral blood of rheumatoid arthritis (RA) patients, as well as the level of TIM-3 on these cells and serum cytokine levels. Material/Methods We enrolled 59 RA patients and 46 age- and sex-matched healthy controls in this study. The proportion of T cells and TIM-3 expression on these T cells were determined by flow cytometry. Cytokine levels in serum were determined by ELISA. Results Compared with the healthy controls, the proportions of CD3+CD4+ T cells and CD3+CD4+CD25+CD127low T cells in the peripheral blood were significantly higher in RA patients. However, RA patients had significantly lower proportions of CD3+CD8+ T cells and CD3+CD4−CD8− T cells. TIM-3 was highly expressed on CD3+CD4+, CD3+CD8+, CD3+CD4+CD25+CD127low, and CD3+CD4−CD8− T cells, as well as CD14+ monocytes, in RA patients. Nevertheless, no correlation between TIM-3 level and an RA disease activity score of 28 was found. The elevated serum levels of IL-6 and IL-37 were positively correlated with tumor necrosis factor-α (TNF-α). Conclusions Both pro-inflammatory cytokines (TNF-α and IL-6) and anti-inflammatory mediators (TIM-3 and IL-37) simultaneously contribute to the pathogenesis of RA. TIM-3 and IL-37 may be used as potential biomarkers of active RA.
Collapse
Affiliation(s)
- Lijun Song
- Shenzhen Research Institute of Shandong University, Shenzhen, Guangdong, China (mainland).,Department of Rheumatology, Qilu Hospital of Shandong University, Ji'nan, Shandong, China (mainland)
| | - Yunqing Wang
- Department of Rheumatology, Qilu Hospital of Shandong University, Ji'nan, Shandong, China (mainland).,Department of Rheumatology and Immunology, Guangdong Second Provincial General Hospital, Guangzhou, Guangdong, China (mainland)
| | - Yameng Sui
- Department of Rheumatology, Qilu Hospital of Shandong University, Ji'nan, Shandong, China (mainland).,School of Medicine, Shandong University, Ji'nan, Shandong, China (mainland)
| | - Jiao Sun
- Department of Rheumatology, Qilu Hospital of Shandong University, , China (mainland).,School of Medicine, Shandong University, , China (mainland)
| | - Dong Li
- Shenzhen Research Institute of Shandong University, Shenzhen, Guangdong, China (mainland).,Cryomedicine Laboratory, Qilu Hospital of Shandong University, Ji'nan, Shandong, China (mainland)
| | - Guosheng Li
- Shenzhen Research Institute of Shandong University, Shenzhen, Guangdong, China (mainland).,Department of Hematology, Qilu Hospital of Shandong University, Ji'nan, Shandong, China (mainland)
| | - Jianwei Liu
- Department of Rheumatology, Qilu Hospital of Shandong University, Ji'nan, Shandong, China (mainland)
| | - Tianwang Li
- Department of Rheumatology and Immunology, Guangdong Second Provincial General Hospital, Guangzhou, Guangdong, China (mainland)
| | - Qiang Shu
- Shenzhen Research Institute of Shandong University, Shenzhen, Guangdong, China (mainland).,Department of Rheumatology, Qilu Hospital of Shandong University, Ji'nan, Shandong, China (mainland)
| |
Collapse
|
28
|
Gaber T, Schönbeck K, Hoff H, Tran CL, Strehl C, Lang A, Ohrndorf S, Pfeiffenberger M, Röhner E, Matziolis G, Burmester GR, Buttgereit F, Hoff P. CTLA-4 Mediates Inhibitory Function of Mesenchymal Stem/Stromal Cells. Int J Mol Sci 2018; 19:ijms19082312. [PMID: 30087255 PMCID: PMC6121442 DOI: 10.3390/ijms19082312] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2018] [Revised: 08/01/2018] [Accepted: 08/02/2018] [Indexed: 02/07/2023] Open
Abstract
Mesenchymal stem/stromal cells (MSCs) are stem cells of the connective tissue, possess a plastic phenotype, and are able to differentiate into various tissues. Besides their role in tissue regeneration, MSCs perform additional functions as a modulator or inhibitor of immune responses. Due to their pleiotropic function, MSCs have also gained therapeutic importance for the treatment of autoimmune diseases and for improving fracture healing and cartilage regeneration. However, the therapeutic/immunomodulatory mode of action of MSCs is largely unknown. Here, we describe that MSCs express the inhibitory receptor CTLA-4 (cytotoxic T lymphocyte antigen 4). We show that depending on the environmental conditions, MSCs express different isoforms of CTLA-4 with the secreted isoform (sCTLA-4) being the most abundant under hypoxic conditions. Furthermore, we demonstrate that the immunosuppressive function of MSCs is mediated mainly by the secretion of CTLA-4. These findings open new ways for treatment when tissue regeneration/fracture healing is difficult.
Collapse
Affiliation(s)
- Timo Gaber
- Department of Rheumatology and Clinical Immunology, Charité⁻Universitätsmedizin Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 10117 Berlin, Germany.
- German Rheumatism Research Centre (DRFZ) Berlin, A Leibniz Institute, 10117 Berlin, Germany.
| | - Kerstin Schönbeck
- Department of Rheumatology and Clinical Immunology, Charité⁻Universitätsmedizin Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 10117 Berlin, Germany.
- German Rheumatism Research Centre (DRFZ) Berlin, A Leibniz Institute, 10117 Berlin, Germany.
| | - Holger Hoff
- German Rheumatism Research Centre (DRFZ) Berlin, A Leibniz Institute, 10117 Berlin, Germany.
| | - Cam Loan Tran
- Department of Rheumatology and Clinical Immunology, Charité⁻Universitätsmedizin Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 10117 Berlin, Germany.
- German Rheumatism Research Centre (DRFZ) Berlin, A Leibniz Institute, 10117 Berlin, Germany.
| | - Cindy Strehl
- Department of Rheumatology and Clinical Immunology, Charité⁻Universitätsmedizin Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 10117 Berlin, Germany.
- German Rheumatism Research Centre (DRFZ) Berlin, A Leibniz Institute, 10117 Berlin, Germany.
| | - Annemarie Lang
- Department of Rheumatology and Clinical Immunology, Charité⁻Universitätsmedizin Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 10117 Berlin, Germany.
- German Rheumatism Research Centre (DRFZ) Berlin, A Leibniz Institute, 10117 Berlin, Germany.
| | - Sarah Ohrndorf
- Department of Rheumatology and Clinical Immunology, Charité⁻Universitätsmedizin Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 10117 Berlin, Germany.
| | - Moritz Pfeiffenberger
- Department of Rheumatology and Clinical Immunology, Charité⁻Universitätsmedizin Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 10117 Berlin, Germany.
- German Rheumatism Research Centre (DRFZ) Berlin, A Leibniz Institute, 10117 Berlin, Germany.
| | - Eric Röhner
- Department of Orthopedics, Campus Eisenberg, Jena University Hospital, Klosterlausnitzer Straße 81, 07607 Eisenberg, Germany.
| | - Georg Matziolis
- Department of Orthopedics, Campus Eisenberg, Jena University Hospital, Klosterlausnitzer Straße 81, 07607 Eisenberg, Germany.
| | - Gerd-R Burmester
- Department of Rheumatology and Clinical Immunology, Charité⁻Universitätsmedizin Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 10117 Berlin, Germany.
- German Rheumatism Research Centre (DRFZ) Berlin, A Leibniz Institute, 10117 Berlin, Germany.
| | - Frank Buttgereit
- Department of Rheumatology and Clinical Immunology, Charité⁻Universitätsmedizin Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 10117 Berlin, Germany.
- German Rheumatism Research Centre (DRFZ) Berlin, A Leibniz Institute, 10117 Berlin, Germany.
| | - Paula Hoff
- Department of Rheumatology and Clinical Immunology, Charité⁻Universitätsmedizin Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 10117 Berlin, Germany.
- German Rheumatism Research Centre (DRFZ) Berlin, A Leibniz Institute, 10117 Berlin, Germany.
- Department of Orthopedics, Campus Eisenberg, Jena University Hospital, Klosterlausnitzer Straße 81, 07607 Eisenberg, Germany.
- Endokrinologikum Berlin, 10117 Berlin, Germany.
| |
Collapse
|
29
|
Ohradanova-Repic A, Machacek C, Charvet C, Lager F, Le Roux D, Platzer R, Leksa V, Mitulovic G, Burkard TR, Zlabinger GJ, Fischer MB, Feuillet V, Renault G, Blüml S, Benko M, Suchanek M, Huppa JB, Matsuyama T, Cavaco-Paulo A, Bismuth G, Stockinger H. Extracellular Purine Metabolism Is the Switchboard of Immunosuppressive Macrophages and a Novel Target to Treat Diseases With Macrophage Imbalances. Front Immunol 2018; 9:852. [PMID: 29780382 PMCID: PMC5946032 DOI: 10.3389/fimmu.2018.00852] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2018] [Accepted: 04/06/2018] [Indexed: 11/13/2022] Open
Abstract
If misregulated, macrophage (Mϕ)-T cell interactions can drive chronic inflammation thereby causing diseases, such as rheumatoid arthritis (RA). We report that in a proinflammatory environment, granulocyte-Mϕ (GM-CSF)- and Mϕ colony-stimulating factor (M-CSF)-dependent Mϕs have dichotomous effects on T cell activity. While GM-CSF-dependent Mϕs show a highly stimulatory activity typical for M1 Mϕs, M-CSF-dependent Mϕs, marked by folate receptor β (FRβ), adopt an immunosuppressive M2 phenotype. We find the latter to be caused by the purinergic pathway that directs release of extracellular ATP and its conversion to immunosuppressive adenosine by co-expressed CD39 and CD73. Since we observed a misbalance between immunosuppressive and immunostimulatory Mϕs in human and murine arthritic joints, we devised a new strategy for RA treatment based on targeted delivery of a novel methotrexate (MTX) formulation to the immunosuppressive FRβ+CD39+CD73+ Mϕs, which boosts adenosine production and curtails the dominance of proinflammatory Mϕs. In contrast to untargeted MTX, this approach leads to potent alleviation of inflammation in the murine arthritis model. In conclusion, we define the Mϕ extracellular purine metabolism as a novel checkpoint in Mϕ cell fate decision-making and an attractive target to control pathological Mϕs in immune-mediated diseases.
Collapse
Affiliation(s)
- Anna Ohradanova-Repic
- Molecular Immunology Unit, Institute for Hygiene and Applied Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Christian Machacek
- Molecular Immunology Unit, Institute for Hygiene and Applied Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Celine Charvet
- Institut National de la Santé et de la Recherche Médicale, INSERM U1016, Institut Cochin, Paris, France.,Université Paris Descartes, Paris, France.,Centre National de la Recherche Scientifique (CNRS), UMR 8104, Paris, France
| | - Franck Lager
- Institut National de la Santé et de la Recherche Médicale, INSERM U1016, Institut Cochin, Paris, France.,Université Paris Descartes, Paris, France.,Centre National de la Recherche Scientifique (CNRS), UMR 8104, Paris, France
| | - Delphine Le Roux
- Institut National de la Santé et de la Recherche Médicale, INSERM U1016, Institut Cochin, Paris, France.,Université Paris Descartes, Paris, France.,Centre National de la Recherche Scientifique (CNRS), UMR 8104, Paris, France
| | - René Platzer
- Molecular Immunology Unit, Institute for Hygiene and Applied Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Vladimir Leksa
- Molecular Immunology Unit, Institute for Hygiene and Applied Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria.,Laboratory of Molecular Immunology, Institute of Molecular Biology, Slovak Academy of Sciences, Bratislava, Slovakia
| | - Goran Mitulovic
- Clinical Department of Medical and Chemical Laboratory Diagnostics, Medical University of Vienna, Vienna, Austria
| | - Thomas R Burkard
- Bioinformatics Department of the Research Institute of Molecular Pathology and the Institute of Molecular Biotechnology of the Austrian Academy of Sciences, Vienna, Austria
| | - Gerhard J Zlabinger
- Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Michael B Fischer
- Department of Transfusion Medicine, Medical University of Vienna, Vienna, Austria.,Center for Biomedical Technology, Danube University Krems, Krems, Austria
| | - Vincent Feuillet
- Institut National de la Santé et de la Recherche Médicale, INSERM U1016, Institut Cochin, Paris, France.,Université Paris Descartes, Paris, France.,Centre National de la Recherche Scientifique (CNRS), UMR 8104, Paris, France
| | - Gilles Renault
- Institut National de la Santé et de la Recherche Médicale, INSERM U1016, Institut Cochin, Paris, France.,Université Paris Descartes, Paris, France.,Centre National de la Recherche Scientifique (CNRS), UMR 8104, Paris, France
| | - Stephan Blüml
- Division of Rheumatology, Internal Medicine III, Medical University of Vienna, Vienna, Austria
| | | | | | - Johannes B Huppa
- Molecular Immunology Unit, Institute for Hygiene and Applied Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Takami Matsuyama
- The Center for Advanced Biomedical Sciences and Swine Research, Kagoshima University, Kagoshima, Japan
| | - Artur Cavaco-Paulo
- Centre of Biological Engineering, University of Minho, Campus of Gualtar, Braga, Portugal
| | - Georges Bismuth
- Institut National de la Santé et de la Recherche Médicale, INSERM U1016, Institut Cochin, Paris, France.,Université Paris Descartes, Paris, France.,Centre National de la Recherche Scientifique (CNRS), UMR 8104, Paris, France
| | - Hannes Stockinger
- Molecular Immunology Unit, Institute for Hygiene and Applied Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
30
|
van Loosdregt J, van Wijk F, Prakken B, Vastert B. Update on research and clinical translation on specific clinical areas from biology to bedside: Unpacking the mysteries of juvenile idiopathic arthritis pathogenesis. Best Pract Res Clin Rheumatol 2018; 31:460-475. [PMID: 29773267 DOI: 10.1016/j.berh.2018.02.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2017] [Revised: 02/01/2018] [Accepted: 02/01/2018] [Indexed: 02/08/2023]
Abstract
In the past decades, we have gained important insights into the mechanisms of disease and therapy underlying chronic inflammation in juvenile idiopathic arthritis (JIA). These insights have resulted in several game-changing therapeutic modalities for many patients. However, additional progress still has to be made with regard to efficacy, cost reduction, minimization of side effects, and dose-tapering and stop strategies of maintenance drugs. Moreover, to really transform the current therapeutic strategies into personalized medicine, we need validated biomarkers to translate increased insights into clinical practice. In this article, we describe recent developments in JIA research and outline how clinical innovations need to go hand in hand with basic discoveries to really effect care for patients. Facilitating the transition from bench to bedside is crucial for addressing the major current challenges in JIA management. When successful, it will set new standards for a safe, targeted, and personalized medicine in JIA.
Collapse
Affiliation(s)
- Jorg van Loosdregt
- Department of Pediatric Immunology & Rheumatology, Laboratory for Translational Medicine, University Medical Centre Utrecht, University of Utrecht, Lundlaan 6, P.O. Box 85090, 3584 EA/3508 AB, Utrecht, The Netherlands
| | - Femke van Wijk
- Department of Pediatric Immunology & Rheumatology, Laboratory for Translational Medicine, University Medical Centre Utrecht, University of Utrecht, Lundlaan 6, P.O. Box 85090, 3584 EA/3508 AB, Utrecht, The Netherlands
| | - Berent Prakken
- Department of Pediatric Immunology & Rheumatology, Laboratory for Translational Medicine, University Medical Centre Utrecht, University of Utrecht, Lundlaan 6, P.O. Box 85090, 3584 EA/3508 AB, Utrecht, The Netherlands
| | - Bas Vastert
- Department of Pediatric Immunology & Rheumatology, Laboratory for Translational Medicine, University Medical Centre Utrecht, University of Utrecht, Lundlaan 6, P.O. Box 85090, 3584 EA/3508 AB, Utrecht, The Netherlands.
| |
Collapse
|
31
|
Sudres M, Verdier J, Truffault F, Le Panse R, Berrih-Aknin S. Pathophysiological mechanisms of autoimmunity. Ann N Y Acad Sci 2018; 1413:59-68. [PMID: 29377165 DOI: 10.1111/nyas.13560] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2017] [Revised: 10/29/2017] [Accepted: 10/30/2017] [Indexed: 12/12/2022]
Abstract
Autoimmune diseases (AIDs) are chronic disorders characterized by inflammatory reactions against self-antigens that can be either systemic or organ specific. AIDs can differ in their epidemiologic features and clinical presentations, yet all share a remarkable complexity. AIDs result from an interplay of genetic and epigenetic factors with environmental components that are associated with imbalances in the immune system. Many of the pathogenic mechanisms of AIDs are also implicated in myasthenia gravis (MG), an AID in which inflammation of the thymus leads to a neuromuscular disorder. Our goal here is to highlight the similarities and differences between MG and other AIDs by reviewing the common transcriptome signatures and the development of germinal centers and by discussing some unresolved questions about autoimmune mechanisms. This review will propose hypotheses to explain the origin of regulatory T (Treg ) cell defects and the causes of chronicity and specificity of AIDs.
Collapse
Affiliation(s)
- Muriel Sudres
- INSERM U974, Paris, France.,UPMC Sorbonne Universités, Paris, France.,AIM, Institut de Myologie, Paris, France
| | - Julien Verdier
- INSERM U974, Paris, France.,UPMC Sorbonne Universités, Paris, France.,AIM, Institut de Myologie, Paris, France
| | - Frédérique Truffault
- INSERM U974, Paris, France.,UPMC Sorbonne Universités, Paris, France.,AIM, Institut de Myologie, Paris, France
| | - Rozen Le Panse
- INSERM U974, Paris, France.,UPMC Sorbonne Universités, Paris, France.,AIM, Institut de Myologie, Paris, France
| | - Sonia Berrih-Aknin
- INSERM U974, Paris, France.,UPMC Sorbonne Universités, Paris, France.,AIM, Institut de Myologie, Paris, France
| |
Collapse
|
32
|
Alivernini S, Gremese E, McSharry C, Tolusso B, Ferraccioli G, McInnes IB, Kurowska-Stolarska M. MicroRNA-155-at the Critical Interface of Innate and Adaptive Immunity in Arthritis. Front Immunol 2018; 8:1932. [PMID: 29354135 PMCID: PMC5760508 DOI: 10.3389/fimmu.2017.01932] [Citation(s) in RCA: 131] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2017] [Accepted: 12/15/2017] [Indexed: 12/17/2022] Open
Abstract
MicroRNAs (miRNAs) are small non-coding RNAs that fine-tune the cell response to a changing environment by modulating the cell transcriptome. miR-155 is a multifunctional miRNA enriched in cells of the immune system and is indispensable for the immune response. However, when deregulated, miR-155 contributes to the development of chronic inflammation, autoimmunity, cancer, and fibrosis. Herein, we review the evidence for the pathogenic role of miR-155 in driving aberrant activation of the immune system in rheumatoid arthritis, and its potential as a disease biomarker and therapeutic target.
Collapse
Affiliation(s)
- Stefano Alivernini
- Institute of Rheumatology - Fondazione Policlinico Universitario A. Gemelli, Catholic University of the Sacred Heart, Rome, Italy
| | - Elisa Gremese
- Institute of Rheumatology - Fondazione Policlinico Universitario A. Gemelli, Catholic University of the Sacred Heart, Rome, Italy
| | - Charles McSharry
- Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, United Kingdom
| | - Barbara Tolusso
- Institute of Rheumatology - Fondazione Policlinico Universitario A. Gemelli, Catholic University of the Sacred Heart, Rome, Italy
| | - Gianfranco Ferraccioli
- Institute of Rheumatology - Fondazione Policlinico Universitario A. Gemelli, Catholic University of the Sacred Heart, Rome, Italy
| | - Iain B McInnes
- Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, United Kingdom.,Rheumatoid Arthritis Pathogenesis Centre of Excellence (RACE), Glasgow, United Kingdom
| | - Mariola Kurowska-Stolarska
- Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, United Kingdom.,Rheumatoid Arthritis Pathogenesis Centre of Excellence (RACE), Glasgow, United Kingdom
| |
Collapse
|
33
|
Joshi RN, Binai NA, Marabita F, Sui Z, Altman A, Heck AJR, Tegnér J, Schmidt A. Phosphoproteomics Reveals Regulatory T Cell-Mediated DEF6 Dephosphorylation That Affects Cytokine Expression in Human Conventional T Cells. Front Immunol 2017; 8:1163. [PMID: 28993769 PMCID: PMC5622166 DOI: 10.3389/fimmu.2017.01163] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2017] [Accepted: 09/01/2017] [Indexed: 12/25/2022] Open
Abstract
Regulatory T cells (Tregs) control key events of immune tolerance, primarily by suppression of effector T cells. We previously revealed that Tregs rapidly suppress T cell receptor (TCR)-induced calcium store depletion in conventional CD4+CD25− T cells (Tcons) independently of IP3 levels, consequently inhibiting NFAT signaling and effector cytokine expression. Here, we study Treg suppression mechanisms through unbiased phosphoproteomics of primary human Tcons upon TCR stimulation and Treg-mediated suppression, respectively. Tregs induced a state of overall decreased phosphorylation as opposed to TCR stimulation. We discovered novel phosphosites (T595_S597) in the DEF6 (SLAT) protein that were phosphorylated upon TCR stimulation and conversely dephosphorylated upon coculture with Tregs. Mutation of these DEF6 phosphosites abrogated interaction of DEF6 with the IP3 receptor and affected NFAT activation and cytokine transcription in primary Tcons. This novel mechanism and phosphoproteomics data resource may aid in modifying sensitivity of Tcons to Treg-mediated suppression in autoimmune disease or cancer.
Collapse
Affiliation(s)
- Rubin N Joshi
- Unit of Computational Medicine, Center for Molecular Medicine, Department of Medicine Solna, Karolinska University Hospital, Science for Life Laboratory, Karolinska Institutet, Stockholm, Sweden
| | - Nadine A Binai
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, Netherlands.,Netherlands Proteomics Centre, Utrecht, Netherlands
| | - Francesco Marabita
- Unit of Computational Medicine, Center for Molecular Medicine, Department of Medicine Solna, Karolinska University Hospital, Science for Life Laboratory, Karolinska Institutet, Stockholm, Sweden
| | - Zhenhua Sui
- Division of Cell Biology, La Jolla Institute for Allergy and Immunology, La Jolla, CA, United States
| | - Amnon Altman
- Division of Cell Biology, La Jolla Institute for Allergy and Immunology, La Jolla, CA, United States
| | - Albert J R Heck
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, Netherlands.,Netherlands Proteomics Centre, Utrecht, Netherlands
| | - Jesper Tegnér
- Unit of Computational Medicine, Center for Molecular Medicine, Department of Medicine Solna, Karolinska University Hospital, Science for Life Laboratory, Karolinska Institutet, Stockholm, Sweden.,Biological and Environmental Sciences and Engineering Division, King Abdullah University of Science and Technology (KAUST), Thuwal, Saudi Arabia.,Computer, Electrical and Mathematical Sciences and Engineering Division, King Abdullah University of Science and Technology (KAUST), Thuwal, Saudi Arabia
| | - Angelika Schmidt
- Unit of Computational Medicine, Center for Molecular Medicine, Department of Medicine Solna, Karolinska University Hospital, Science for Life Laboratory, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
34
|
Th17 in Animal Models of Rheumatoid Arthritis. J Clin Med 2017; 6:jcm6070073. [PMID: 28753982 PMCID: PMC5532581 DOI: 10.3390/jcm6070073] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2017] [Revised: 07/13/2017] [Accepted: 07/15/2017] [Indexed: 01/04/2023] Open
Abstract
IL-17-secreting helper CD4 T cells (Th17 cells) constitute a newly identified subset of helper CD4 T cells that play a key role in the development of rheumatoid arthritis (RA) in its animal models. Recently, several models of spontaneous RA, which elucidate the mechanism of RA onset, have been discovered. These animal models shed new light on the role of Th17 in the development of autoimmune arthritis. Th17 cells coordinate inflammation and promote joint destruction, acting on various cells, including neutrophils, macrophages, synovial fibroblasts, and osteoclasts. Regulatory T cells cannot control Th17 cells under conditions of inflammation. In this review, the pathogenic role of Th17 cells in arthritis development, which was revealed by the recent animal models of RA, is discussed.
Collapse
|
35
|
Pozsgay J, Szekanecz Z, Sármay G. Antigen-specific immunotherapies in rheumatic diseases. Nat Rev Rheumatol 2017; 13:525-537. [DOI: 10.1038/nrrheum.2017.107] [Citation(s) in RCA: 63] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
36
|
Shu X, Chen F, Peng Q, Lu X, Tian X, Wang Y, Wang G. Potential role of autophagy in T‑cell survival in polymyositis and dermatomyositis. Mol Med Rep 2017; 16:1180-1188. [PMID: 28586060 PMCID: PMC5562052 DOI: 10.3892/mmr.2017.6693] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2016] [Accepted: 02/10/2017] [Indexed: 12/23/2022] Open
Abstract
Peripheral blood T lymphocytopenia has previously been identified in polymyositis/dermatomyositis (PM/DM) patients. Therefore, the present study aimed to examine the potential role of autophagy in peripheral blood T cell survival in PM/DM patients. Transmission electron microscopy was used to detect the formation of autophagosomes of peripheral blood cluster of differentiation (CD)3+ T cells obtained from 24 patients with PM/DM and 21 healthy controls. Protein and mRNA expression levels of autophagy‑related molecules were examined by western blot analysis and reverse transcription‑quantitative polymerase chain reaction, respectively. The number of peripheral blood CD3+ T cells decreased significantly in PM/DM patients. The median percentage of apoptosis of CD3+ T cells in PM/DM patients was significantly increased compared with healthy controls. Furthermore, the number of autophagosomes and the expression of the autophagy markers microtubule‑associated protein 1A/1B‑light chain 3 (LC3) and Beclin‑1 were significantly reduced in the circulating CD3+ T cells of PM/DM patients compared with those of healthy controls. LC3 and Beclin‑1 protein levels correlated negatively with apoptosis rates in circulating CD3+ T cells in patients with PM/DM. CD3+ T cells from PM/DM patients treated with rapamycin increased autophagy and decreased apoptosis compared with untreated cells (P<0.05). In conclusion, these results suggested that autophagy may serve a potential protective role in the peripheral blood T cells of patients with PM/DM.
Collapse
Affiliation(s)
- Xiaoming Shu
- Department of Rheumatology, China‑Japan Friendship Hospital, Chaoyang, Beijing 100029, P.R. China
| | - Fang Chen
- Department of Rheumatology, China‑Japan Friendship Hospital, Chaoyang, Beijing 100029, P.R. China
| | - Qinglin Peng
- Department of Rheumatology, China‑Japan Friendship Hospital, Chaoyang, Beijing 100029, P.R. China
| | - Xin Lu
- Department of Rheumatology, China‑Japan Friendship Hospital, Chaoyang, Beijing 100029, P.R. China
| | - Xiaolan Tian
- Department of Rheumatology, China‑Japan Friendship Hospital, Chaoyang, Beijing 100029, P.R. China
| | - Yan Wang
- Department of Rheumatology, China‑Japan Friendship Hospital, Chaoyang, Beijing 100029, P.R. China
| | - Guochun Wang
- Department of Rheumatology, China‑Japan Friendship Hospital, Chaoyang, Beijing 100029, P.R. China
| |
Collapse
|
37
|
Xianfanghuomingyin, a Chinese Compound Medicine, Modulates the Proliferation and Differentiation of T Lymphocyte in a Collagen-Induced Arthritis Mouse Model. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2016; 2016:6356871. [PMID: 27656238 PMCID: PMC5021507 DOI: 10.1155/2016/6356871] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/05/2016] [Revised: 07/17/2016] [Accepted: 07/26/2016] [Indexed: 12/29/2022]
Abstract
In traditional Chinese medicine (TCM), xianfanghuomingyin (XFHM) is used to treat autoimmune diseases, including rheumatoid arthritis (RA). Here, we studied the mechanisms underlying its treatment effects, especially its anti-inflammatory effects in a collagen-induced arthritis (CIA) mouse model. We found that cartilage destruction and pannus formation were alleviated by treatment with XFHM. The abnormal differentiation of Th1 and Th17 cells was downregulated significantly by XFHM, and Th2 and Treg cells were upregulated. Moreover, the expression levels of specific cytokines and transcription factors related to Th1 cells (interferon γ [IFNγ], T-bet) and Th17 cells (interleukin- [IL-] 17) and the nuclear receptor retinoic acid receptor-related orphan receptor-gamma (RORγ) were downregulated. Serum IL-4 and GATA-3, which contribute to Th2 cells differentiation, increased significantly after XFHM administration. These results indicate that XFHM can restore the balance of T lymphocytes and reestablish the immunological tolerance to inhibit autoinflammatory disorder of RA. Taken together, XFHM can be used as a complementary or alternative traditional medicine to treat RA.
Collapse
|
38
|
Affiliation(s)
- Sonia Berrih-Aknin
- INSERM U974; Paris France
- CNRS FRE3617; Paris France
- Sorbonne University; UPMC Univ Paris 06; Paris France
- AIM; Institute of Myology; Paris France
| |
Collapse
|
39
|
Way EE, Trevejo-Nunez G, Kane LP, Steiner BH, Puri KD, Kolls JK, Chen K. Dose-Dependent Suppression of Cytokine production from T cells by a Novel Phosphoinositide 3-Kinase Delta Inhibitor. Sci Rep 2016; 6:30384. [PMID: 27461849 PMCID: PMC4961957 DOI: 10.1038/srep30384] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2016] [Accepted: 07/04/2016] [Indexed: 12/20/2022] Open
Abstract
There remains a significant need for development of effective small molecules that can inhibit cytokine-mediated inflammation. Phosphoinositide 3 kinase (PI3K) is a direct upstream activator of AKT, and plays a critical role in multiple cell signaling pathways, cell cycle progression, and cell growth, and PI3K inhibitors have been approved or are in clinical development. We examined novel PI3Kdelta inhibitors, which are highly selective for the p110delta isoform of in CD3/CD28 stimulated T-cell cytokine production. In vitro generated CD4+ T effector cells stimulated in the presence of a PI3Kdelta inhibitor demonstrated a dose-dependent suppression of cytokines produced by Th1, Th2, and Th17 cells. This effect was T-cell intrinsic, and we observed similar effects on human PBMCs. Th17 cells expressing a constitutively activated form of AKT were resistant to PI3Kdelta inhibition, suggesting that the inhibitor is acting through AKT signaling pathways. Additionally, PI3Kdelta inhibition decreased IL-17 production in vivo and decreased neutrophil recruitment to the lung in a murine model of acute pulmonary inflammation. These experiments show that targeting PI3Kdelta activity can modulate T-cell cytokine production and reduce inflammation in vivo, suggesting that PI3Kdelta inhibition could have therapeutic potential in treating inflammatory diseases.
Collapse
Affiliation(s)
- Emily E Way
- Richard King Mellon Foundation Institute for Pediatric Research, Children's Hospital of Pittsburgh, Pittsburgh, PA, USA
| | - Giraldina Trevejo-Nunez
- Richard King Mellon Foundation Institute for Pediatric Research, Children's Hospital of Pittsburgh, Pittsburgh, PA, USA
| | - Lawrence P Kane
- Department of Immunology, University of Pittsburgh, Pittsburgh, PA, USA
| | | | | | - Jay K Kolls
- Richard King Mellon Foundation Institute for Pediatric Research, Children's Hospital of Pittsburgh, Pittsburgh, PA, USA
| | - Kong Chen
- Richard King Mellon Foundation Institute for Pediatric Research, Children's Hospital of Pittsburgh, Pittsburgh, PA, USA
| |
Collapse
|
40
|
Barberá A, Lorenzo N, van Kooten P, van Roon J, de Jager W, Prada D, Gómez J, Padrón G, van Eden W, Broere F, Del Carmen Domínguez M. APL1, an altered peptide ligand derived from human heat-shock protein 60, increases the frequency of Tregs and its suppressive capacity against antigen responding effector CD4 + T cells from rheumatoid arthritis patients. Cell Stress Chaperones 2016; 21:735-44. [PMID: 27241313 PMCID: PMC4908004 DOI: 10.1007/s12192-016-0698-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2016] [Revised: 04/29/2016] [Accepted: 05/04/2016] [Indexed: 12/29/2022] Open
Abstract
Rheumatoid arthritis (RA) is a systemic autoimmune disease characterized by a chronic relapsing-remitting joint inflammation. Perturbations in the balance between CD4 + T cells producing IL-17 and CD4 + CD25(high)FoxP3 + Tregs correlate with irreversible bone and cartilage destruction in RA. APL1 is an altered peptide ligand derived from a CD4+ T-cell epitope of human HSP60, an autoantigen expressed in the inflamed synovium, which increases the frequency of CD4 + CD25(high)FoxP3+ Tregs in peripheral blood mononuclear cells from RA patients. The aim of this study was to evaluate the suppressive capacity of Tregs induced by APL1 on proliferation of effector CD4+ T cells using co-culture experiments. Enhanced Treg-mediated suppression was observed in APL1-treated cultures compared with cells cultured only with media. Subsequent analyses using autologous cross-over experiments showed that the enhanced Treg suppression in APL1-treated cultures could reflect increased suppressive function of Tregs against APL1-responsive T cells. On the other hand, APL1-treatment had a significant effect reducing IL-17 levels produced by effector CD4+ T cells. Hence, this peptide has the ability to increase the frequency of Tregs and their suppressive properties whereas effector T cells produce less IL-17. Thus, we propose that APL1 therapy could help to ameliorate the pathogenic Th17/Treg balance in RA patients.
Collapse
Affiliation(s)
- Ariana Barberá
- Biomedical Research Department, Center for Genetic Engineering and Biotechnology, P.O. Box 6162, Havana, 11300, Cuba
- Department of Medicine, Cambridge Institute for Medical Research, University of Cambridge, Cambridge, UK
| | - Noraylis Lorenzo
- Biomedical Research Department, Center for Genetic Engineering and Biotechnology, P.O. Box 6162, Havana, 11300, Cuba
| | - Peter van Kooten
- Infectious Diseases and Immunology Department, Faculty of Veterinary Medicine, University of Utrecht, P.O. Box 80165, NL-3508 TD, Utrecht, The Netherlands
| | - Joel van Roon
- Department of Rheumatology and Clinical and Laboratory of Translational Immunology, University Medical Centrum Utrecht, PO Box 85500, 3508 GA, Utrecht, The Netherlands
| | - Wilco de Jager
- Department of Pediatric Immunology and Laboratory of Translational Immunology, University Medical Center Utrecht, PO Box 85500, 3508 GA, Utrecht, The Netherlands
| | - Dinorah Prada
- Institute of Rheumatology, Ave 10 of October, No. 130, Havana, Cuba
| | - Jorge Gómez
- Institute of Rheumatology, Ave 10 of October, No. 130, Havana, Cuba
| | - Gabriel Padrón
- Biomedical Research Department, Center for Genetic Engineering and Biotechnology, P.O. Box 6162, Havana, 11300, Cuba
| | - Willem van Eden
- Infectious Diseases and Immunology Department, Faculty of Veterinary Medicine, University of Utrecht, P.O. Box 80165, NL-3508 TD, Utrecht, The Netherlands
| | - Femke Broere
- Infectious Diseases and Immunology Department, Faculty of Veterinary Medicine, University of Utrecht, P.O. Box 80165, NL-3508 TD, Utrecht, The Netherlands
| | - María Del Carmen Domínguez
- Biomedical Research Department, Center for Genetic Engineering and Biotechnology, P.O. Box 6162, Havana, 11300, Cuba.
| |
Collapse
|
41
|
Rossetti M, Spreafico R, Consolaro A, Leong JY, Chua C, Massa M, Saidin S, Magni-Manzoni S, Arkachaisri T, Wallace CA, Gattorno M, Martini A, Lovell DJ, Albani S. TCR repertoire sequencing identifies synovial Treg cell clonotypes in the bloodstream during active inflammation in human arthritis. Ann Rheum Dis 2016; 76:435-441. [PMID: 27311837 PMCID: PMC5284348 DOI: 10.1136/annrheumdis-2015-208992] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2015] [Revised: 04/30/2016] [Accepted: 05/20/2016] [Indexed: 01/07/2023]
Abstract
OBJECTIVES The imbalance between effector and regulatory T (Treg) cells is crucial in the pathogenesis of autoimmune arthritis. Immune responses are often investigated in the blood because of its accessibility, but circulating lymphocytes are not representative of those found in inflamed tissues. This disconnect hinders our understanding of the mechanisms underlying disease. Our goal was to identify Treg cells implicated in autoimmunity at the inflamed joints, and also readily detectable in the blood upon recirculation. METHODS We compared Treg cells of patients with juvenile idiopathic arthritis responding or not to therapy by using: (i) T cell receptor (TCR) sequencing, to identify clonotypes shared between blood and synovial fluid; (ii) FOXP3 Treg cell-specific demethylated region DNA methylation assays, to investigate their stability and (iii) flow cytometry and suppression assays to probe their tolerogenic functions. RESULTS We found a subset of synovial Treg cells that recirculated into the bloodstream of patients with juvenile idiopathic and adult rheumatoid arthritis. These inflammation-associated (ia)Treg cells, but not other blood Treg cells, expanded during active disease and proliferated in response to their cognate antigens. Despite the typical inflammatory-skewed balance of immune mechanisms in arthritis, iaTreg cells were stably committed to the regulatory lineage and fully suppressive. A fraction of iaTreg clonotypes were in common with pathogenic effector T cells. CONCLUSIONS Using an innovative antigen-agnostic approach, we uncovered a population of bona fide synovial Treg cells readily accessible from the blood and selectively expanding during active disease, paving the way to non-invasive diagnostics and better understanding of the pathogenesis of autoimmunity.
Collapse
Affiliation(s)
- Maura Rossetti
- SingHealth Translational Immunology and Inflammation Centre, SingHealth and Duke-NUS Graduate Medical School, Singapore, Singapore.,Translational Research Unit, Sanford-Burnham Medical Research Institute, San Diego, California, USA.,Department of Pathology and Laboratory Medicine, University of California Los Angeles, Los Angeles, California, USA
| | - Roberto Spreafico
- SingHealth Translational Immunology and Inflammation Centre, SingHealth and Duke-NUS Graduate Medical School, Singapore, Singapore.,Translational Research Unit, Sanford-Burnham Medical Research Institute, San Diego, California, USA.,Department of Microbiology, Immunology and Molecular Genetics, University of California Los Angeles, Los Angeles, California, USA
| | - Alessandro Consolaro
- Second Pediatrics Division, University of Genoa and G Gaslini Institute, Genova, Italy
| | - Jing Yao Leong
- SingHealth Translational Immunology and Inflammation Centre, SingHealth and Duke-NUS Graduate Medical School, Singapore, Singapore
| | - Camillus Chua
- SingHealth Translational Immunology and Inflammation Centre, SingHealth and Duke-NUS Graduate Medical School, Singapore, Singapore
| | - Margherita Massa
- Lab Biotecnologie, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Suzan Saidin
- SingHealth Translational Immunology and Inflammation Centre, SingHealth and Duke-NUS Graduate Medical School, Singapore, Singapore
| | - Silvia Magni-Manzoni
- Pediatric Rheumatology Unit, IRCCS Ospedale Pediatrico Bambino Gesù, Rome, Italy
| | - Thaschawee Arkachaisri
- Duke-NUS Graduate Medical School and Rheumatology and Immunology Service, KK Women's and Children's Hospital, Singapore, Singapore
| | - Carol A Wallace
- Seattle Children's Hospital and Research Institute, Seattle, Washington, USA
| | - Marco Gattorno
- Second Pediatrics Division, University of Genoa and G Gaslini Institute, Genova, Italy
| | - Alberto Martini
- Second Pediatrics Division, University of Genoa and G Gaslini Institute, Genova, Italy
| | - Daniel J Lovell
- Division of Rheumatology, Cincinnati Children's Hospital Medical Center, and Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Salvatore Albani
- SingHealth Translational Immunology and Inflammation Centre, SingHealth and Duke-NUS Graduate Medical School, Singapore, Singapore
| |
Collapse
|
42
|
Transcriptome Signatures Reveal Rapid Induction of Immune-Responsive Genes in Human Memory CD8(+) T Cells. Sci Rep 2016; 6:27005. [PMID: 27243788 PMCID: PMC4886650 DOI: 10.1038/srep27005] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2016] [Accepted: 05/12/2016] [Indexed: 01/05/2023] Open
Abstract
Memory T cells (TM) play a prominent role in protection and auto-immunity due to their ability to mount a more effective response than naïve T cells (TN). However, the molecular mechanisms underlying enhanced functionality of TM are not well defined, particularly in human TM. We examined the global gene expression profiles of human CD8+ TN and TM before and after stimulation. There were 1,284, 1,373 and 1,629 differentially expressed genes between TN and TM at 0 hr, 4 hr and 24 hr after stimulation, respectively, with more genes expressed to higher levels in TM. Genes rapidly up-regulated in TN cells were largely involved in nitrogen, nucleoside and amino acid metabolisms. In contrast, those in CD8+ TM were significantly enriched for immune-response-associated processes, including cytokine production, lymphocyte activation and chemotaxis. Multiple cytokines were rapidly up-regulated in TM cells, including effector cytokines known to be produced by CD8+ T cells and important for their functions, as well as regulatory cytokines, both pro- and anti-inflammatory, that are not typically produced by CD8+ T cells. These results provide new insights into molecular mechanisms that contribute to the enhanced functionality of human CD8+ TM and their prominent role in protection and auto-immunity.
Collapse
|
43
|
Yang L, Qiao G, Hassan Y, Li Z, Zhang X, Kong H, Zeng W, Yin F, Zhang J. Program Death-1 Suppresses Autoimmune Arthritis by Inhibiting Th17 Response. Arch Immunol Ther Exp (Warsz) 2016; 64:417-23. [PMID: 27197661 DOI: 10.1007/s00005-016-0404-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2015] [Accepted: 03/14/2016] [Indexed: 12/16/2022]
Abstract
Program death-1 (PD-1) is a co-inhibitory receptor inducibly expressed on activated T cells. PD-1 has been reported to be associated with the development of several autoimmune diseases including rheumatoid arthritis, but the precise cellular and molecular mechanisms have not been fully elucidated. To study the role of PD-1 in the pathogenesis of rheumatoid arthritis and the possible underlying mechanisms, we performed collagen-induced arthritis (CIA) in C57BL/6 mice. Here, we show that PD-1 deficiency leads to the development of severe CIA in mice. When analyzing T cells from CIA mice ex vivo, we noticed aberrant antigen-specific Th17 responses in mice lacking PD-1. This is possibly due to deregulated activation of PKC-θ and Akt. In support of this notion, treating Pdcd1 (-/-) mice with an inhibitor of PI3-kinase that is upstream of PKC-θ and Akt significantly suppressed the disease severity. Therefore, our data indicate that PD-1 dampens antigen-specific Th17 response, thus inhibiting the disease.
Collapse
Affiliation(s)
- Lifen Yang
- Department of Pediatrics, Xiangya Hospital, Central South University, No. 87, Xiangya Road, Changsha, 410008, Hunan, People's Republic of China.,Section of Nephrology, Department of Medicine, The Committees on Immunology and Molecular Medicine, The University of Chicago, Chicago, IL, 60637, USA
| | - Guilin Qiao
- Section of Nephrology, Department of Medicine, The Committees on Immunology and Molecular Medicine, The University of Chicago, Chicago, IL, 60637, USA
| | - Yassir Hassan
- Section of Nephrology, Department of Medicine, The Committees on Immunology and Molecular Medicine, The University of Chicago, Chicago, IL, 60637, USA
| | - Zhenping Li
- Section of Nephrology, Department of Medicine, The Committees on Immunology and Molecular Medicine, The University of Chicago, Chicago, IL, 60637, USA
| | - Xiaoqing Zhang
- Department of Pediatrics, Xiangya Hospital, Central South University, No. 87, Xiangya Road, Changsha, 410008, Hunan, People's Republic of China
| | - Huimin Kong
- Department of Pediatrics, Xiangya Hospital, Central South University, No. 87, Xiangya Road, Changsha, 410008, Hunan, People's Republic of China
| | - Weimin Zeng
- Department of Physiology, Xiangya Medical School, Central South University, Hunan, 410008, People's Republic of China
| | - Fei Yin
- Department of Pediatrics, Xiangya Hospital, Central South University, No. 87, Xiangya Road, Changsha, 410008, Hunan, People's Republic of China.
| | - Jian Zhang
- Section of Nephrology, Department of Medicine, The Committees on Immunology and Molecular Medicine, The University of Chicago, Chicago, IL, 60637, USA. .,Department of Microbial Infection and Immunity, The Ohio State University, 784 Biomedical Research Tower, 460 West 12th Avenue, Columbus, OH, 43210, USA.
| |
Collapse
|
44
|
Petrelli A, Wehrens EJ, Scholman RC, Prakken BJ, Vastert SJ, van Wijk F. Self-Sustained Resistance to Suppression of CD8+ Teff Cells at the Site of Autoimmune Inflammation Can Be Reversed by Tumor Necrosis Factor and Interferon-γ Blockade. Arthritis Rheumatol 2016; 68:229-36. [PMID: 26360332 DOI: 10.1002/art.39418] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2015] [Accepted: 08/27/2015] [Indexed: 12/31/2022]
Abstract
OBJECTIVE Resistance of Teff cells to Treg cell-mediated suppression contributes to the breakdown of peripheral tolerance in the inflamed joints of patients with juvenile idiopathic arthritis (JIA). However, unanswered questions are whether this resistant phenotype is self-sustained and whether CD8+ and CD4+ Teff cells share the same mechanism of resistance to suppression. We undertook this study to investigate intrinsic resistance of CD8+ Teff cells to suppression and to determine how this can be targeted therapeutically. METHODS CD8+ or CD4+ Teff cells were cultured with or without antigen-presenting cells (APCs) in Treg cell-dependent and -independent suppression assays. Synovial fluid (SF)-derived Teff cells were crosscultured with peripheral blood (PB) Treg cells from JIA patients or healthy controls. Tumor necrosis factor (TNF) or interferon-γ (IFNγ) blocking agents were used to restore Teff cell responsiveness to suppression. RESULTS Suppression of cell proliferation and cytokine production in CD8+ Teff cells from the SF of JIA patients was severely impaired compared to that in CD8+ Teff cells from the PB of JIA patients, regardless of the presence of APCs and CD4+ Teff cells. Similar to CD4+ Teff cells, impaired suppression of CD8+ Teff cells was shown to be an intrinsic feature of this cell population. While TNF blockade restored both CD8+ and CD4+ Teff cell susceptibility to suppression, autocrine release of IFNγ selectively sustained CD8+ Teff cell resistance, which could be relieved by IFNγ blockade. CONCLUSION Unlike CD4+ Teff cells, resistance of CD8+ Teff cells to suppression at the site of autoimmune inflammation is maintained by autocrine release of IFNγ, and blockade of IFNγ restores CD8+ Teff cell responsiveness to suppression. These findings indicate a potential therapeutic value of blocking IFNγ to restore immune regulation in JIA.
Collapse
Affiliation(s)
| | | | | | | | | | - Femke van Wijk
- University Medical Center Utrecht, Utrecht, The Netherlands
| |
Collapse
|
45
|
|
46
|
Chen SY, Hsu WT, Chen YL, Chien CH, Chiang BL. Lymphocyte-activation gene 3(+) (LAG3(+)) forkhead box protein 3(-) (FOXP3(-)) regulatory T cells induced by B cells alleviates joint inflammation in collagen-induced arthritis. J Autoimmun 2016; 68:75-85. [PMID: 26908164 DOI: 10.1016/j.jaut.2016.02.002] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2015] [Revised: 02/01/2016] [Accepted: 02/01/2016] [Indexed: 11/18/2022]
Abstract
Rheumatoid arthritis (RA) is an autoimmune disease in which dysregulated immune cells primarily target synovial joints. Despite recent advances in the treatment of RA, including the introduction of biologic therapies and employment of combination disease-modifying antirheumatic drug strategies, remission rates remain suboptimal. Previous studies have demonstrated that the adoptive transfer of induced regulatory T cells (iTregs) was effective in treating a murine model of collagen-induced arthritis (CIA). The objective of this study was to develop optimal potential iTreg-based therapy for CIA by adoptively transferring LAG3(+) Treg-of-B cells. B-cell-induced Treg-of-B cells expressed LAG3 but not Foxp3 (designated LAG3(+) Treg-of-B), and secreted IL-4, IL-10, and TGF-β. Furthermore, LAG3(+) Treg-of-B cells suppressed the proliferation of CD4(+)CD25(-) responder T cells through both LAG3 and IL-10 production. In the murine CIA model, adoptive transfer of LAG3(+) Treg-of-B cells alleviated the joint severity as well as local and systemic inflammation. Treatment with LAG3(+) Treg-of-B cells also promoted IL-10 production in lymphocytes isolated from the spleen and draining lymph nodes. Moreover, mice receiving LAG3(+) Treg-of-B cell treatment showed significantly less pronounced osteolysis in the hind footpads, which correlated with the downregulation of tartrate-resistant acid phosphatase expression. In conclusion, we identified a novel subset of Tregs for CIA treatment. This insight may facilitate exploring novel regulatory T-cell-based therapies for human autoimmune diseases.
Collapse
Affiliation(s)
- Szu-Ying Chen
- Graduate Institute of Clinical Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Wan-Tseng Hsu
- Graduate Institute of Clinical Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Yi-Lien Chen
- Graduate Institute of Clinical Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Chien-Hui Chien
- Graduate Institute of Immunology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Bor-Luen Chiang
- Graduate Institute of Clinical Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan; Graduate Institute of Immunology, College of Medicine, National Taiwan University, Taipei, Taiwan.
| |
Collapse
|
47
|
Win SJ, Kühl AA, Sparwasser T, Hünig T, Kamradt T. In vivo activation of Treg cells with a CD28 superagonist prevents and ameliorates chronic destructive arthritis in mice. Eur J Immunol 2016; 46:1193-202. [PMID: 26711629 DOI: 10.1002/eji.201546104] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2015] [Revised: 12/01/2015] [Accepted: 12/22/2015] [Indexed: 01/29/2023]
Abstract
Although regulatory T (Treg) cells are necessary to prevent autoimmune diseases, including arthritis, whether Treg cells can ameliorate established inflammatory disease is controversial. Using the glucose-6-phosphate isomerase (G6PI)-induced arthritis model in mice, we aimed to determine the therapeutic efficacy of increasing Treg cell number and function during chronic destructive arthritis. Chronic destructive arthritis was induced by transient depletion of Treg cells prior to immunization with G6PI. At different time points after disease induction, mice were treated with a CD28 superagonistic antibody (CD28SA). CD28SA treatment during the induction phase of arthritis ameliorated the acute signs of arthritis and completely prevented the development of chronic destructive arthritis. CD28SA treatment of mice with fully developed arthritis induced a significant reduction in clinical and histological signs of arthritis. When given during the chronic destructive phase of arthritis, 56 days after disease induction, CD28SA treatment resulted in a modest reduction of clinical signs of arthritis and a reduction in histopathological signs of joint inflammation. Our data show that increasing the number and activation of Treg cells by a CD28SA is therapeutically effective in experimental arthritis.
Collapse
Affiliation(s)
- Stephanie J Win
- Institute of Immunology, Universitätsklinikum Jena, Jena, Germany
| | - Anja A Kühl
- Department of Medicine 1-Gastroenterology, Infectious Diseases and Rheumatology and Research Centre ImmunoSciences, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Tim Sparwasser
- Institute for Infection Immunology, Centre for Experimental and Clinical Infection Research, TWINCORE, Hannover, Germany
| | - Thomas Hünig
- Institute of Virology and Immunobiology, University of Würzburg, Würzburg, Germany
| | - Thomas Kamradt
- Institute of Immunology, Universitätsklinikum Jena, Jena, Germany
| |
Collapse
|
48
|
Roberts CA, Dickinson AK, Taams LS. The Interplay Between Monocytes/Macrophages and CD4(+) T Cell Subsets in Rheumatoid Arthritis. Front Immunol 2015; 6:571. [PMID: 26635790 PMCID: PMC4652039 DOI: 10.3389/fimmu.2015.00571] [Citation(s) in RCA: 150] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2015] [Accepted: 10/26/2015] [Indexed: 12/24/2022] Open
Abstract
Rheumatoid arthritis (RA) is a chronic inflammatory disease characterized by inflammation of the synovial lining (synovitis). The inflammation in the RA joint is associated with and driven by immune cell infiltration, synovial hyperproliferation, and excessive production of proinflammatory mediators, such as tumor necrosis factor α (TNFα), interferon γ (IFNγ), interleukin (IL)-1β, IL-6, and IL-17, eventually resulting in damage to the cartilage and underlying bone. The RA joint harbors a wide range of immune cell types, including monocytes, macrophages, and CD4(+) T cells (both proinflammatory and regulatory). The interplay between CD14(+) myeloid cells and CD4(+) T cells can significantly influence CD4(+) T cell function, and conversely, effector vs. regulatory CD4(+) T cell subsets can exert profound effects on monocyte/macrophage function. In this review, we will discuss how the interplay between CD4(+) T cells and monocytes/macrophages may contribute to the immunopathology of RA.
Collapse
Affiliation(s)
- Ceri A Roberts
- Centre for Molecular and Cellular Biology of Inflammation (CMCBI), Division of Immunology, Infection and Inflammatory Disease, King's College London , London , UK
| | - Abigail K Dickinson
- Centre for Molecular and Cellular Biology of Inflammation (CMCBI), Division of Immunology, Infection and Inflammatory Disease, King's College London , London , UK
| | - Leonie S Taams
- Centre for Molecular and Cellular Biology of Inflammation (CMCBI), Division of Immunology, Infection and Inflammatory Disease, King's College London , London , UK
| |
Collapse
|
49
|
Nacka-Aleksić M, Pilipović I, Stojić-Vukanić Z, Kosec D, Bufan B, Vujnović I, Arsenović-Ranin N, Dimitrijević M, Leposavić G. Sexual dimorphism in the aged rat CD4+ T lymphocyte-mediated immune response elicited by inoculation with spinal cord homogenate. Mech Ageing Dev 2015; 152:15-31. [PMID: 26408399 DOI: 10.1016/j.mad.2015.09.004] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2015] [Accepted: 09/20/2015] [Indexed: 01/13/2023]
Abstract
Considering the crucial pathogenic role of CD4+ T cells in experimental autoimmune encephalomyelitis (EAE) and the opposite direction of the sexual dimorphism in the severity of the disease in 22-24-and 3-month-old dark agouti rats, sex differences in CD4+ T-cell-mediated immune response in aged rats immunized for EAE were examined and compared with those in young animals. In the inductive phase of EAE, fewer activated CD4+ lymphocytes were retrieved from draining lymph nodes of male (developing less severe disease) compared with female rats, due, at least partly, to their lesser expansion. The former reflected a greater suppressive capacity of CD4+CD25+Foxp3+ cells. Consequently, CD4+ lymphocyte infiltration into the spinal cord of aged male rats was diminished. At the peak of EAE, the frequency of reactivated cells was lower, whereas that of the regulatory CD4+ cells was higher in male rat spinal cord. Consistently, microglial activation and the expression of proinflammatory/damaging cytokines in male rat spinal cord mononuclear cells were diminished. Additionally, the frequency of the highly pathogenic IL-17+IFN-γ+ T lymphocytes infiltrating their spinal cord was lower. Together, these results point to (i) an age-specificity in CD4+ cell-mediated immune response and (ii) mechanisms underlying the sex differences in this response in aged rats.
Collapse
Affiliation(s)
- Mirjana Nacka-Aleksić
- Department of Physiology, Faculty of Pharmacy, University of Belgrade, 450 Vojvode Stepe, 11221 Belgrade, Serbia
| | - Ivan Pilipović
- Immunology Research Centre "Branislav Janković", Institute of Virology, Vaccines and Sera "Torlak", 458 Vojvode Stepe, 11221 Belgrade, Serbia
| | - Zorica Stojić-Vukanić
- Department of Microbiology and Immunology, Faculty of Pharmacy, University of Belgrade, 450 Vojvode Stepe, 11221 Belgrade, Serbia
| | - Duško Kosec
- Immunology Research Centre "Branislav Janković", Institute of Virology, Vaccines and Sera "Torlak", 458 Vojvode Stepe, 11221 Belgrade, Serbia
| | - Biljana Bufan
- Department of Microbiology and Immunology, Faculty of Pharmacy, University of Belgrade, 450 Vojvode Stepe, 11221 Belgrade, Serbia
| | - Ivana Vujnović
- Immunology Research Centre "Branislav Janković", Institute of Virology, Vaccines and Sera "Torlak", 458 Vojvode Stepe, 11221 Belgrade, Serbia
| | - Nevena Arsenović-Ranin
- Department of Microbiology and Immunology, Faculty of Pharmacy, University of Belgrade, 450 Vojvode Stepe, 11221 Belgrade, Serbia
| | - Mirjana Dimitrijević
- Immunology Research Centre "Branislav Janković", Institute of Virology, Vaccines and Sera "Torlak", 458 Vojvode Stepe, 11221 Belgrade, Serbia
| | - Gordana Leposavić
- Department of Physiology, Faculty of Pharmacy, University of Belgrade, 450 Vojvode Stepe, 11221 Belgrade, Serbia.
| |
Collapse
|
50
|
Ozen S. Decade in review-paediatric rheumatology: A field on the move. Nat Rev Rheumatol 2015; 11:625-6. [PMID: 26390845 DOI: 10.1038/nrrheum.2015.130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Seza Ozen
- Department of Pediatric Rheumatology, Hacettepe University, Sihhiye, 06100 Ankara, Turkey
| |
Collapse
|