1
|
Liu FQ, Qu QY, Lei Y, Chen Q, Chen YX, Li ML, Sun XY, Wu YJ, Huang QS, Fu HX, Kong Y, Li YY, Wang QF, Huang XJ, Zhang XH. High dimensional proteomic mapping of bone marrow immune characteristics in immune thrombocytopenia. SCIENCE CHINA. LIFE SCIENCES 2024; 67:1635-1647. [PMID: 38644444 DOI: 10.1007/s11427-023-2520-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Accepted: 01/09/2024] [Indexed: 04/23/2024]
Abstract
To investigate the role of co-stimulatory and co-inhibitory molecules on immune tolerance in immune thrombocytopenia (ITP), this study mapped the immune cell heterogeneity in the bone marrow of ITP at the single-cell level using Cytometry by Time of Flight (CyTOF). Thirty-six patients with ITP and nine healthy volunteers were enrolled in the study. As soluble immunomodulatory molecules, more sCD25 and sGalectin-9 were detected in ITP patients. On the cell surface, co-stimulatory molecules like ICOS and HVEM were observed to be upregulated in mainly central memory and effector T cells. In contrast, co-inhibitory molecules such as CTLA-4 were significantly reduced in Th1 and Th17 cell subsets. Taking a platelet count of 30×109 L-1 as the cutoff value, ITP patients with high and low platelet counts showed different T cell immune profiles. Antigen-presenting cells such as monocytes and B cells may regulate the activation of T cells through CTLA-4/CD86 and HVEM/BTLA interactions, respectively, and participate in the pathogenesis of ITP. In conclusion, the proteomic and soluble molecular profiles brought insight into the interaction and modulation of immune cells in the bone marrow of ITP. They may offer novel targets to develop personalized immunotherapies.
Collapse
Affiliation(s)
- Feng-Qi Liu
- Peking University People's Hospital, Peking University Institute of Hematology, Beijing, 100044, China
- Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, 100044, China
- National Clinical Research Center for Hematologic Disease, Beijing, 100044, China
- Collaborative Innovation Centre of Hematology, Peking University, Beijing, 100044, China
| | - Qing-Yuan Qu
- Peking University People's Hospital, Peking University Institute of Hematology, Beijing, 100044, China
- Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, 100044, China
- National Clinical Research Center for Hematologic Disease, Beijing, 100044, China
- Collaborative Innovation Centre of Hematology, Peking University, Beijing, 100044, China
| | - Ying Lei
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing, 100101, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Qi Chen
- Peking University People's Hospital, Peking University Institute of Hematology, Beijing, 100044, China
- Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, 100044, China
- National Clinical Research Center for Hematologic Disease, Beijing, 100044, China
- Collaborative Innovation Centre of Hematology, Peking University, Beijing, 100044, China
| | - Yu-Xiu Chen
- Peking University People's Hospital, Peking University Institute of Hematology, Beijing, 100044, China
- Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, 100044, China
- National Clinical Research Center for Hematologic Disease, Beijing, 100044, China
- Collaborative Innovation Centre of Hematology, Peking University, Beijing, 100044, China
| | - Meng-Lin Li
- Peking University People's Hospital, Peking University Institute of Hematology, Beijing, 100044, China
- Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, 100044, China
- National Clinical Research Center for Hematologic Disease, Beijing, 100044, China
- Collaborative Innovation Centre of Hematology, Peking University, Beijing, 100044, China
| | - Xue-Yan Sun
- Peking University People's Hospital, Peking University Institute of Hematology, Beijing, 100044, China
- Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, 100044, China
- National Clinical Research Center for Hematologic Disease, Beijing, 100044, China
- Collaborative Innovation Centre of Hematology, Peking University, Beijing, 100044, China
| | - Ye-Jun Wu
- Peking University People's Hospital, Peking University Institute of Hematology, Beijing, 100044, China
- Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, 100044, China
- National Clinical Research Center for Hematologic Disease, Beijing, 100044, China
- Collaborative Innovation Centre of Hematology, Peking University, Beijing, 100044, China
| | - Qiu-Sha Huang
- Peking University People's Hospital, Peking University Institute of Hematology, Beijing, 100044, China
- Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, 100044, China
- National Clinical Research Center for Hematologic Disease, Beijing, 100044, China
- Collaborative Innovation Centre of Hematology, Peking University, Beijing, 100044, China
| | - Hai-Xia Fu
- Peking University People's Hospital, Peking University Institute of Hematology, Beijing, 100044, China
- Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, 100044, China
- National Clinical Research Center for Hematologic Disease, Beijing, 100044, China
- Collaborative Innovation Centre of Hematology, Peking University, Beijing, 100044, China
| | - Yuan Kong
- Peking University People's Hospital, Peking University Institute of Hematology, Beijing, 100044, China
- Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, 100044, China
- National Clinical Research Center for Hematologic Disease, Beijing, 100044, China
- Collaborative Innovation Centre of Hematology, Peking University, Beijing, 100044, China
| | - Yue-Ying Li
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing, 100101, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Qian-Fei Wang
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing, 100101, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Xiao-Jun Huang
- Peking University People's Hospital, Peking University Institute of Hematology, Beijing, 100044, China
- Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, 100044, China
- National Clinical Research Center for Hematologic Disease, Beijing, 100044, China
- Collaborative Innovation Centre of Hematology, Peking University, Beijing, 100044, China
- Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, 100074, China
- State Key Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing, 100191, China
| | - Xiao-Hui Zhang
- Peking University People's Hospital, Peking University Institute of Hematology, Beijing, 100044, China.
- Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, 100044, China.
- National Clinical Research Center for Hematologic Disease, Beijing, 100044, China.
- Collaborative Innovation Centre of Hematology, Peking University, Beijing, 100044, China.
| |
Collapse
|
2
|
Papachristodoulou E, Kyttaris VC. New and emerging therapies for systemic lupus erythematosus. Clin Immunol 2024; 263:110200. [PMID: 38582250 DOI: 10.1016/j.clim.2024.110200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2024] [Revised: 03/23/2024] [Accepted: 04/01/2024] [Indexed: 04/08/2024]
Abstract
Systemic Lupus Erythematosus (SLE) and lupus nephritis treatment is still based on non-specific immune suppression despite the first biological therapy for the disease having been approved more than a decade ago. Intense basic and translational research has uncovered a multitude of pathways that are actively being evaluated as treatment targets in SLE and lupus nephritis, with two new medications receiving FDA approval in the last 3 years. Herein we provide an overview of targeted therapies for SLE including medications targeting the B lymphocyte compartment, intracellular signaling, co-stimulation, and finally the interferons and other cytokines.
Collapse
Affiliation(s)
- Eleni Papachristodoulou
- Division of Rheumatology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Vasileios C Kyttaris
- Division of Rheumatology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
3
|
da Rosa LC, Scales HE, Benson RA, Brewer JM, McInnes IB, Garside P. The effect of abatacept on T-cell activation is not long-lived in vivo. DISCOVERY IMMUNOLOGY 2024; 3:kyad029. [PMID: 38567291 PMCID: PMC10917171 DOI: 10.1093/discim/kyad029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Revised: 12/06/2023] [Accepted: 01/03/2024] [Indexed: 04/04/2024]
Abstract
Abatacept, a co-stimulatory blocker comprising the extracellular portion of human CTLA-4 linked to the Fc region of IgG1, is approved for the treatment of rheumatoid arthritis. By impairing the interaction between CD28 on T cells and CD80/CD86 on APCs, its mechanisms of action include the suppression of follicular T helper cells (preventing the breach of self-tolerance in B cells), inhibition of cell cycle progression holding T cells in a state described as 'induced naïve' and reduction in DC conditioning. However, less is known about how long these inhibitory effects might last, which is a critical question for therapeutic use in patients. Herein, employing a murine model of OVA-induced DTH, we demonstrate that the effect of abatacept is short-lived in vivo and that the inhibitory effects diminish markedly when treatment is ceased.
Collapse
Affiliation(s)
- Larissa C da Rosa
- School of Infection & Immunity, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8TA, UK
| | - Hannah E Scales
- School of Infection & Immunity, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8TA, UK
| | - Robert A Benson
- School of Infection & Immunity, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8TA, UK
| | - James M Brewer
- School of Infection & Immunity, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8TA, UK
| | - Iain B McInnes
- School of Infection & Immunity, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8TA, UK
| | - Paul Garside
- School of Infection & Immunity, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8TA, UK
| |
Collapse
|
4
|
Guo P, Huang C, Yang Q, Zhong G, Zhang J, Qiu M, Zeng R, Gou K, Zhang C, Qu Y. Advances in Formulations of Microneedle System for Rheumatoid Arthritis Treatment. Int J Nanomedicine 2023; 18:7759-7784. [PMID: 38144510 PMCID: PMC10743780 DOI: 10.2147/ijn.s435251] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Accepted: 12/05/2023] [Indexed: 12/26/2023] Open
Abstract
Rheumatoid arthritis (RA) is a systemic autoimmune disease characterized by chronic joint inflammation, eventually leading to severe disability and premature death. At present, the treatment of RA is mainly to reduce inflammation, swelling, and pain. Commonly used drugs are non-steroidal anti-inflammatory drugs (NSAIDs), glucocorticoids, and disease-modifying anti-rheumatic drugs (DMARDs). These drugs lack specificity and require long-term, high-dose administration, which can cause serious adverse effects. In addition, the oral, intravenous, and intra-articular injections will reduce patient compliance, resulting in high cost and low bioavailability. Due to these limitations, microneedles (MNs) have emerged as a new strategy to efficiently localize the drugs in inflamed joints for the treatment of RA. MNs can overcome the cuticle barrier of the skin without stimulating nerves and blood vessels. Which can increase patient compliance, improve bioavailability, and avoid systemic circulation. This review summarizes and evaluates the application of MNs in RA, especially dissolving MNs (DMNs). We encourage the use of MNs to treat RA, by describing the general properties of MNs, materials, preparation technology, drug release mechanism, and advantages. Furthermore, we discussed the biological safety, development prospects, and future challenges of MNs, hoping to provide a new strategy for the treatment of RA.
Collapse
Affiliation(s)
- Peng Guo
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, People’s Republic of China
| | - Chi Huang
- Department of Pharmacy, Jiang’an Hospital of Traditional Chinese Medicine, Yibin, 644200, People’s Republic of China
| | - Qin Yang
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, People’s Republic of China
| | - Guofeng Zhong
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, People’s Republic of China
| | - Junbo Zhang
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, People’s Republic of China
| | - Mengyu Qiu
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, People’s Republic of China
| | - Rui Zeng
- Institute of Tibetan Plateau, Southwest Minzu University, Chengdu, 610225, People’s Republic of China
| | - Kaijun Gou
- Institute of Tibetan Plateau, Southwest Minzu University, Chengdu, 610225, People’s Republic of China
| | - Chen Zhang
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, People’s Republic of China
| | - Yan Qu
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, People’s Republic of China
| |
Collapse
|
5
|
Kojima M, Suzuki K, Takeshita M, Ohyagi M, Iizuka M, Yamane H, Koga K, Kouro T, Kassai Y, Yoshihara T, Adachi R, Hashikami K, Ota Y, Yoshimoto K, Kaneko Y, Morita R, Yoshimura A, Takeuchi T. Anti-human-TIGIT agonistic antibody ameliorates autoimmune diseases by inhibiting Tfh and Tph cells and enhancing Treg cells. Commun Biol 2023; 6:500. [PMID: 37161050 PMCID: PMC10170076 DOI: 10.1038/s42003-023-04874-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2022] [Accepted: 04/26/2023] [Indexed: 05/11/2023] Open
Abstract
T cells play important roles in autoimmune diseases, but it remains unclear how to optimally manipulate them. We focused on the T cell immunoreceptor with Ig and ITIM domains (TIGIT), a coinhibitory molecule that regulates and is expressed in T cells. In autoimmune diseases, the association between TIGIT-expressing cells and pathogenesis and the function of human-TIGIT (hu-TIGIT) signalling modification have not been fully elucidated. Here we generated anti-hu-TIGIT agonistic monoclonal antibodies (mAbs) and generated hu-TIGIT knock-in mice to accurately evaluate the efficacy of mAb function. Our mAb suppressed the activation of CD4+ T cells, especially follicular helper T and peripheral helper T cells that highly expressed TIGIT, and enhanced the suppressive function of naïve regulatory T cells. These results indicate that our mAb has advantages in restoring the imbalance of T cells that are activated in autoimmune diseases and suggest potential clinical applications for anti-hu-TIGIT agonistic mAbs as therapeutic agents.
Collapse
Affiliation(s)
- Marenori Kojima
- Division of Rheumatology, Department of Internal Medicine, Keio University School of Medicine, Shinjuku-ku, Tokyo, Japan
| | - Katsuya Suzuki
- Division of Rheumatology, Department of Internal Medicine, Keio University School of Medicine, Shinjuku-ku, Tokyo, Japan
| | - Masaru Takeshita
- Division of Rheumatology, Department of Internal Medicine, Keio University School of Medicine, Shinjuku-ku, Tokyo, Japan
| | - Masaki Ohyagi
- Department of Microbiology and Immunology, Keio University School of Medicine, Shinjuku-ku, Tokyo, Japan
| | - Mana Iizuka
- Department of Microbiology and Immunology, Keio University School of Medicine, Shinjuku-ku, Tokyo, Japan
| | - Humitsugu Yamane
- Division of Rheumatology, Department of Internal Medicine, Keio University School of Medicine, Shinjuku-ku, Tokyo, Japan
| | - Keiko Koga
- Immunology Unit, Pharmaceutical Research Division, Takeda Pharmaceutical Company Limited, Fujisawa City, Kanagawa, Japan
| | - Taku Kouro
- Immunology Unit, Pharmaceutical Research Division, Takeda Pharmaceutical Company Limited, Fujisawa City, Kanagawa, Japan
| | - Yoshiaki Kassai
- Immunology Unit, Pharmaceutical Research Division, Takeda Pharmaceutical Company Limited, Fujisawa City, Kanagawa, Japan
| | - Tomoki Yoshihara
- Biomolecular Research Laboratories, Pharmaceutical Research Division, Takeda Pharmaceutical Company Limited, Fujisawa City, Kanagawa, Japan
| | - Ryutaro Adachi
- Biomolecular Research Laboratories, Pharmaceutical Research Division, Takeda Pharmaceutical Company Limited, Fujisawa City, Kanagawa, Japan
| | - Kentarou Hashikami
- Biomolecular Research Laboratories, Pharmaceutical Research Division, Takeda Pharmaceutical Company Limited, Fujisawa City, Kanagawa, Japan
- Axcelead Drug Discovery Partners, Inc., Fujisawa City, Kanagawa, Japan
| | - Yuichiro Ota
- Division of Rheumatology, Department of Internal Medicine, Keio University School of Medicine, Shinjuku-ku, Tokyo, Japan
| | - Keiko Yoshimoto
- Division of Rheumatology, Department of Internal Medicine, Keio University School of Medicine, Shinjuku-ku, Tokyo, Japan
| | - Yuko Kaneko
- Division of Rheumatology, Department of Internal Medicine, Keio University School of Medicine, Shinjuku-ku, Tokyo, Japan
| | - Rimpei Morita
- Department of Microbiology and Immunology, Keio University School of Medicine, Shinjuku-ku, Tokyo, Japan
- Department of Microbiology and Immunology, Nippon Medical School, Bunkyo-ku, Tokyo, Japan
| | - Akihiko Yoshimura
- Department of Microbiology and Immunology, Keio University School of Medicine, Shinjuku-ku, Tokyo, Japan
| | - Tsutomu Takeuchi
- Division of Rheumatology, Department of Internal Medicine, Keio University School of Medicine, Shinjuku-ku, Tokyo, Japan.
| |
Collapse
|
6
|
Park J, Le QV, Wu Y, Lee J, Oh YK. Tolerogenic Nanovaccine for Prevention and Treatment of Autoimmune Encephalomyelitis. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2023; 35:e2202670. [PMID: 36208089 DOI: 10.1002/adma.202202670] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Revised: 09/23/2022] [Indexed: 06/16/2023]
Abstract
Herein, a tolerogenic nanovaccine is developed and tested on an animal model of multiple sclerosis. The nanovaccine is constructed to deliver the self-antigen, myelin oligodendrocyte glycoprotein (MOG) peptide, and dexamethasone on an abatacept-modified polydopamine core nanoparticle (AbaLDPN-MOG). AbaLDPN-MOG can target dendritic cells and undergo endocytosis followed by trafficking to lysosomes. AbaLDPN-MOG blocks the interaction between CD80/CD86 and CD28 in antigen-presenting cells and T cells, leading to decreased interferon gamma secretion. The subcutaneous administration of AbaLDPN-MOG to mice yields significant biodistribution to lymph nodes and, in experimental-autoimmune encephalomyelitis (EAE) model mice, increases the integrity of the myelin basic sheath and minimizes the infiltration of immune cells. EAE mice are treated with AbaLDPN-MOG before or after injection of the autoantigen, MOG. Preimmunization of AbaLDPN-MOG before the injection of MOG completely blocks the development of clinical symptoms. Early treatment with AbaLDPN-MOG at three days after injection of MOG also completely blocks the development of symptoms. Notably, treatment of EAE symptom-developed mice with AbaLDPN-MOG significantly alleviates the symptoms, indicating that the nanovaccine has therapeutic effects. Although AbaLDPN is used for MOG peptide delivery in the EAE model, the concept of AbaLDPN can be widely applied for the prevention and alleviation of other autoimmune diseases.
Collapse
Affiliation(s)
- Jinwon Park
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul, 08826, Republic of Korea
| | - Quoc-Viet Le
- Faculty of Pharmacy, Ton Duc Thang University, Ho Chi Minh City, Vietnam
| | - Yina Wu
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul, 08826, Republic of Korea
| | - Jaiwoo Lee
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul, 08826, Republic of Korea
| | - Yu-Kyoung Oh
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul, 08826, Republic of Korea
| |
Collapse
|
7
|
Yang J, Lin YR, Xiong BJ, Chen ZH, Luo YF, Xu Y, Su YP, Huang HH, Yu CX. Regulation effect of koumine on T-helper cell polarization in rheumatoid arthritis. Eur J Pharmacol 2022; 937:175387. [DOI: 10.1016/j.ejphar.2022.175387] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2022] [Revised: 11/04/2022] [Accepted: 11/07/2022] [Indexed: 11/13/2022]
|
8
|
Cifuentes-Mendiola SE, Solis-Suarez DL, Martínez-Dávalos A, Godínez-Victoria M, García-Hernández AL. CD4 + T-cell activation of bone marrow causes bone fragility and insulin resistance in type 2 diabetes. Bone 2022; 155:116292. [PMID: 34896656 DOI: 10.1016/j.bone.2021.116292] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Revised: 12/02/2021] [Accepted: 12/03/2021] [Indexed: 12/14/2022]
Abstract
Type 2 diabetes mellitus (T2DM) causes an increased risk of bone fractures. However, the pathophysiology of diabetic bone fragility is not completely understood. It has been proposed that an inflammatory microenvironment in bone could be a major mechanism by inducing uncontrolled bone resorption, inadequate bone formation and consequently more porous bones. We propose that activated T-cells in the bone marrow cause a pro-inflammatory microenvironment in bone, and cause bone fragility in T2DM. We induced T2DM in C57BL/6 male mice through a hypercaloric diet rich in carbohydrates and low doses of streptozocin. In T2DM mice we inhibited systemic activation of T-cells with a fusion protein between the extracellular domain of Cytotoxic T-Lymphocyte Antigen 4 and the Fc domain of human immunoglobulin G (CTLA4-Ig). We analysed the effects of T2DM or CTLA4-Ig in lymphocyte cell subsets and antigen-presenting cells in peripheral blood and femoral bone marrow; and their effect on the metabolic phenotype, blood and bone cytokine concentration, femoral bone microarchitecture and biomechanical properties, and the number of osteoblast-like cells in the femoral endosteum. We performed a Pearson multiple correlation analysis between all variables in order to understand the global mechanism. Results demonstrated that CTLA4-Ig decreased the number of activated CD4+ T-cells in the femoral bone marrow and consequently decreased TNF-α and RANK-L concentration in bone, notably improved femoral bone microarchitecture and biomechanical properties, increased the number of osteoblast-like cells, and reduces osteoclastic activity compared to T2DM mice that did not receive the inhibitor. Interestingly, we observed that blood glucose levels and insulin resistance may be related to the increase in activated CD4+ T-cells in the bone marrow. We conclude that bone marrow activated CD4+ T-cells cause poor bone quality and insulin resistance in T2DM.
Collapse
Affiliation(s)
- S E Cifuentes-Mendiola
- Laboratory of Dental Research, Section of Osteoimmunology and Oral Immunology, FES Iztacala, National Autonomous University of Mexico, A. Jiménez Gallardo SN, San Sebastián Xhala, Cuautitlán Izcalli, Estado de México, CP 54714, Mexico; Postgraduate in Biological Sciences, National Autonomous University of Mexico, Mexico, Mexico
| | - D L Solis-Suarez
- Laboratory of Dental Research, Section of Osteoimmunology and Oral Immunology, FES Iztacala, National Autonomous University of Mexico, A. Jiménez Gallardo SN, San Sebastián Xhala, Cuautitlán Izcalli, Estado de México, CP 54714, Mexico
| | - A Martínez-Dávalos
- Physics Institute, National Autonomous University of Mexico, Circuito de la Investigación Científica, Ciudad Universitaria, 04510 México City, Mexico
| | - M Godínez-Victoria
- Sección de Estudios de Posgrado e Investigación, Escuela Superior de Medicina, Instituto Politécnico Nacional, Mexico, Mexico
| | - A L García-Hernández
- Laboratory of Dental Research, Section of Osteoimmunology and Oral Immunology, FES Iztacala, National Autonomous University of Mexico, A. Jiménez Gallardo SN, San Sebastián Xhala, Cuautitlán Izcalli, Estado de México, CP 54714, Mexico.
| |
Collapse
|
9
|
Carvajal Alegria G, Cornec D, Saraux A, Devauchelle-Pensec V, Jamin C, Hillion S, Pers JO, Pochard P. Abatacept Promotes Regulatory B Cell Functions, Enhancing Their Ability to Reduce the Th1 Response in Rheumatoid Arthritis Patients through the Production of IL-10 and TGF-β. THE JOURNAL OF IMMUNOLOGY 2021; 207:470-482. [PMID: 34244295 DOI: 10.4049/jimmunol.2000455] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Accepted: 05/14/2021] [Indexed: 11/19/2022]
Abstract
Abatacept mimics natural CD152 and competes with CD28 for binding to CD80/CD86 on APC, such as B cells, thereby preventing T cell activation. However, its potential impact on B cells has not been identified. The aim of this study was to assess whether abatacept can potentiate the immunoregulatory properties of B cells in vitro and in patients with rheumatoid arthritis (RA). T and B cells from healthy controls were purified. The suppressor properties of B cells in the presence of abatacept or control IgG1 were evaluated based on the ability of these cells to inhibit the polyclonal expansion (anti-CD3/CD28 stimulation) of T cells or their differentiation into Th1 or Th17 cells. Similar analyses were also performed with cells from RA patients before and 3 mo after abatacept initiation. Abatacept significantly potentiated regulatory B cell regulatory functions by enhancing their ability to produce IL-10 and TGF-β, resulting in the increased generation of regulatory T cells and limited T cell proliferation and differentiation into Th1 and Th17 cells. Interestingly, B cells isolated from patients that received a 3-mo treatment with abatacept had an increased ability to reduce T cell functions, confirming the above observations. Abatacept binding to CD80/CD86 induces and promotes regulatory B cell functions by enhancing the ability of these cells to produce IL-10 and TGF-β in vitro and in RA patients.
Collapse
Affiliation(s)
- Guillermo Carvajal Alegria
- UMR 1227 "Lymphocytes B et Autoimmunité," Université de Brest, INSERM, LabEx IGO, Brest, France; and CHU de Brest, Brest, France
| | - Divi Cornec
- UMR 1227 "Lymphocytes B et Autoimmunité," Université de Brest, INSERM, LabEx IGO, Brest, France; and CHU de Brest, Brest, France
| | - Alain Saraux
- UMR 1227 "Lymphocytes B et Autoimmunité," Université de Brest, INSERM, LabEx IGO, Brest, France; and CHU de Brest, Brest, France
| | - Valérie Devauchelle-Pensec
- UMR 1227 "Lymphocytes B et Autoimmunité," Université de Brest, INSERM, LabEx IGO, Brest, France; and CHU de Brest, Brest, France
| | - Christophe Jamin
- UMR 1227 "Lymphocytes B et Autoimmunité," Université de Brest, INSERM, LabEx IGO, Brest, France; and CHU de Brest, Brest, France
| | - Sophie Hillion
- UMR 1227 "Lymphocytes B et Autoimmunité," Université de Brest, INSERM, LabEx IGO, Brest, France; and CHU de Brest, Brest, France
| | - Jacques-Olivier Pers
- UMR 1227 "Lymphocytes B et Autoimmunité," Université de Brest, INSERM, LabEx IGO, Brest, France; and CHU de Brest, Brest, France
| | - Pierre Pochard
- UMR 1227 "Lymphocytes B et Autoimmunité," Université de Brest, INSERM, LabEx IGO, Brest, France; and CHU de Brest, Brest, France
| |
Collapse
|
10
|
Abstract
Therapeutic targeting of immune checkpoints has garnered significant attention in the area of cancer immunotherapy, in which efforts have focused in particular on cytotoxic T lymphocyte antigen 4 (CTLA4) and PD1, both of which are members of the CD28 family. In autoimmunity, these same pathways can be targeted to opposite effect: to curb the over-exuberant immune response. The CTLA4 checkpoint serves as an exemplar, whereby CTLA4 activity is blocked by antibodies in cancer immunotherapy and augmented by the provision of soluble CTLA4 in autoimmunity. Here, we review the targeting of co-stimulatory molecules in autoimmune diseases, focusing in particular on agents directed at members of the CD28 or tumour necrosis factor receptor families. We present the state of the art in co-stimulatory blockade approaches, including rational combinations of immune inhibitory agents, and discuss the future opportunities and challenges in this field.
Collapse
|
11
|
Derambure C, Dzangue-Tchoupou G, D’Agostino MA, Lequerré T, Vittecoq O. Gene expression regulated by abatacept associated with methotrexate and correlation with disease activity in rheumatoid arthritis. PLoS One 2020; 15:e0237143. [PMID: 32760165 PMCID: PMC7410313 DOI: 10.1371/journal.pone.0237143] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2019] [Accepted: 07/21/2020] [Indexed: 11/18/2022] Open
Abstract
Objectives Abatacept acts as a competitive inhibitor of the CD28/(CD80/86) costimulation signal required for T cell activation. Mechanisms of action of abatacept have not been fully investigated. The objective of this study was to provide detailed insight into the mode of action of Abatacept based on gene expression data. Methods In this ancillary study from the APPRAISE trial, we investigated the global molecular effects of Abatacept in whole blood samples collected prospectively in biologic naive rheumatoid arthritis patients (n = 19) at baseline and 6 months after the initiation of Abatacept therapy concomitant with methotrexate. Whole human genome microarrays (4x44K) were performed on both baseline and 6-month samples from responders and non-responders patients categorized according to EULAR criteria. T-test with Benjamini-Hochberg correction was performed to identify significant gene expression changes. Gene Ontology and Single Experiment Analysis tools allowed us to highlight specific biological mechanisms involved in methotrexate/Abatacept. Results In methotrexate/Abatacept responders, 672 genes were significantly (q<0.05) dysregulated at 6 months compared to baseline. Correlation analysis highlighted 19 genes whose dysregulations were significantly associated with disease activity variation (p<0.05) and whose functions were associated with proliferation, apoptosis of cells and mitochondrial metabolism, suggesting a restoration of oxidative signaling. The other 653 gene expression changes were relative to direct or indirect effects of methotrexate/Abatacept treatment and were significantly (p<0.005) involved in pathways relative to mRNA processing, proteasome, angiogenesis, apoptosis and TCR signaling. This study highlights new mechanisms of action of methotrexate/Abatacept and may provide new therapeutic targets to prevent autoimmunity in rheumatoid arthritis.
Collapse
Affiliation(s)
| | | | - Maria Antonietta D’Agostino
- Department of Rheumatology, AP-HP Ambroise Paré Hospital, University of Versailles Saint Quentin en Yvelines, Boulogne-Billancourt, France
| | - Thierry Lequerré
- Normandie Univ, UNIROUEN, Inserm U 1234, Rouen, France
- Department of Rheumatology Inserm CIC/CRB1404, Rouen University Hospital, Rouen Cedex, France
- * E-mail:
| | - Olivier Vittecoq
- Normandie Univ, UNIROUEN, Inserm U 1234, Rouen, France
- Department of Rheumatology Inserm CIC/CRB1404, Rouen University Hospital, Rouen Cedex, France
| |
Collapse
|
12
|
Linsley PS, Greenbaum CJ, Speake C, Long SA, Dufort MJ. B lymphocyte alterations accompany abatacept resistance in new-onset type 1 diabetes. JCI Insight 2019; 4:126136. [PMID: 30830871 PMCID: PMC6478438 DOI: 10.1172/jci.insight.126136] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2018] [Accepted: 01/17/2019] [Indexed: 02/06/2023] Open
Abstract
Costimulatory interactions control T cell activation at sites of activated antigen-presenting cells, including B cells. Blockade of the CD28/CD80/CD86 costimulatory axis with CTLA4Ig (abatacept) is widely used to treat certain autoimmune diseases. While transiently effective in subjects with new-onset type 1 diabetes (T1D), abatacept did not induce long-lasting immune tolerance. To elucidate mechanisms limiting immune tolerance in T1D, we performed unbiased analysis of whole blood transcriptomes and targeted measurements of cell subset levels in subjects from a clinical trial of abatacept in new-onset T1D. We showed that individual subjects displayed age-related immune phenotypes ("immunotypes") at baseline, characterized by elevated levels of B cells or neutrophils, that accompanied rapid or slow progression, respectively, in both abatacept- and placebo-treated groups. A more pronounced immunotype was exhibited by a subset of subjects showing poor response (resistance) to abatacept. This resistance immunotype was characterized by a transient increase in activated B cells (one of the cell types that binds abatacept), reprogrammed costimulatory ligand gene expression, and reduced inhibition of anti-insulin antibodies. Our findings identify immunotypes in T1D subjects that are linked to the rate of disease progression, both in placebo- and abatacept-treated subjects. Furthermore, our results suggest therapeutic approaches to restore immune tolerance in T1D.
Collapse
Affiliation(s)
| | | | | | - S. Alice Long
- Translational Research Program, Benaroya Research Institute at Virginia Mason, Seattle, Washington, USA
| | | |
Collapse
|
13
|
Qin L, Waseem TC, Sahoo A, Bieerkehazhi S, Zhou H, Galkina EV, Nurieva R. Insights Into the Molecular Mechanisms of T Follicular Helper-Mediated Immunity and Pathology. Front Immunol 2018; 9:1884. [PMID: 30158933 PMCID: PMC6104131 DOI: 10.3389/fimmu.2018.01884] [Citation(s) in RCA: 73] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2018] [Accepted: 07/31/2018] [Indexed: 12/18/2022] Open
Abstract
T follicular helper (Tfh) cells play key role in providing help to B cells during germinal center (GC) reactions. Generation of protective antibodies against various infections is an important aspect of Tfh-mediated immune responses and the dysregulation of Tfh cell responses has been implicated in various autoimmune disorders, inflammation, and malignancy. Thus, their differentiation and maintenance must be closely regulated to ensure appropriate help to B cells. The generation and function of Tfh cells is regulated by multiple checkpoints including their early priming stage in T zones and throughout the effector stage of differentiation in GCs. Signaling pathways activated downstream of cytokine and costimulatory receptors as well as consequent activation of subset-specific transcriptional factors are essential steps for Tfh cell generation. Thus, understanding the mechanisms underlying Tfh cell-mediated immunity and pathology will bring into spotlight potential targets for novel therapies. In this review, we discuss the recent findings related to the molecular mechanisms of Tfh cell differentiation and their role in normal immune responses and antibody-mediated diseases.
Collapse
Affiliation(s)
- Lei Qin
- Department of Immunology, University of Texas MD Anderson Cancer Center, Houston, TX, United States.,School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, China
| | - Tayab C Waseem
- Department of Microbiology and Molecular Cell Biology, Eastern Virginia Medical School, Norfolk, VA, United States
| | - Anupama Sahoo
- Department of Immunology, University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Shayahati Bieerkehazhi
- Department of Immunology, University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Hong Zhou
- School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, China
| | - Elena V Galkina
- Department of Microbiology and Molecular Cell Biology, Eastern Virginia Medical School, Norfolk, VA, United States
| | - Roza Nurieva
- Department of Immunology, University of Texas MD Anderson Cancer Center, Houston, TX, United States
| |
Collapse
|
14
|
Prendergast CT, Patakas A, Al-Khabouri S, McIntyre CL, McInnes IB, Brewer JM, Garside P, Benson RA. Visualising the interaction of CD4 T cells and DCs in the evolution of inflammatory arthritis. Ann Rheum Dis 2018; 77:579-588. [PMID: 29358281 DOI: 10.1136/annrheumdis-2017-212279] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2017] [Revised: 12/10/2017] [Accepted: 12/18/2017] [Indexed: 01/08/2023]
Abstract
OBJECTIVES Successful early intervention in rheumatoid arthritis (RA) with the aim of resetting immunological tolerance requires a clearer understanding of how specificity, cellular kinetics and spatial behaviour shape the evolution of articular T cell responses. We aimed to define initial seeding of articular CD4+ T cell responses in early experimental arthritis, evaluating their dynamic behaviour and interactions with dendritic cells (DCs) in the inflamed articular environment. METHODS Antigen-induced arthritis was used to model articular inflammation. Flow cytometry and PCR of T cell receptor (TCR) diversity genes allowed phenotypic analysis of infiltrating T cells. The dynamic interactions of T cells with joint residing DCs were visualised using intravital multiphoton microscopy. RESULTS Initial recruitment of antigen-specific T cells into the joint was paralleled by accumulation of CD4+ T cells with diverse antigen-receptor expression and ability to produce tumour necrosis factor alpha (TNFα) and interferon gamma (IFNγ) on mitogenic restimulation. A proportion of this infiltrate demonstrated slower motility speeds and engaged for longer periods with articular DCs in vivo. Abatacept treatment did not disrupt these interactions but did reduce T cell expression of inducible costimulatory (ICOS) molecule. We also demonstrated that non-specific CD4+ T cells could be recruited during these early articular events. CONCLUSIONS We demonstrate that CD4+ T cells engage with articular DCs supporting antigen specific T cell reactivation. This cellular dialogue can be targeted therapeutically to reduce local T cell activation.
Collapse
Affiliation(s)
- Catriona T Prendergast
- Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - Agapitos Patakas
- Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - Shaima Al-Khabouri
- Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - Claire L McIntyre
- Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - Iain B McInnes
- Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - James M Brewer
- Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - Paul Garside
- Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - Robert A Benson
- Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| |
Collapse
|
15
|
Podojil JR, Hecht I, Chiang MY, Vaknin I, Barbiro I, Novik A, Neria E, Rotman G, Miller SD. ILDR2-Fc Is a Novel Regulator of Immune Homeostasis and Inducer of Antigen-Specific Immune Tolerance. THE JOURNAL OF IMMUNOLOGY 2018; 200:2013-2024. [PMID: 29431690 DOI: 10.4049/jimmunol.1700326] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/03/2017] [Accepted: 12/11/2017] [Indexed: 12/18/2022]
Abstract
ILDR2 is a member of the Ig superfamily, which is implicated in tricellular tight junctions, and has a putative role in pancreatic islet health and survival. We recently found a novel role for ILDR2 in delivering inhibitory signals to T cells. In this article, we show that short-term treatment with ILDR2-Fc results in long-term durable beneficial effects in the relapsing-remitting experimental autoimmune encephalomyelitis and NOD type 1 diabetes models. ILDR2-Fc also promotes transplant engraftment in a minor mismatch bone marrow transplantation model. ILDR2-Fc displays a unique mode of action, combining immunomodulation, regulation of immune homeostasis, and re-establishment of Ag-specific immune tolerance via regulatory T cell induction. These findings support the potential of ILDR-Fc to provide a promising therapeutic approach for the treatment of autoimmune diseases.
Collapse
Affiliation(s)
- Joseph R Podojil
- Department of Microbiology-Immunology, Interdepartmental Immunobiology Center, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611; and
| | | | - Ming-Yi Chiang
- Department of Microbiology-Immunology, Interdepartmental Immunobiology Center, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611; and
| | | | | | | | | | | | - Stephen D Miller
- Department of Microbiology-Immunology, Interdepartmental Immunobiology Center, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611; and
| |
Collapse
|
16
|
Tjärnlund A, Tang Q, Wick C, Dastmalchi M, Mann H, Tomasová Studýnková J, Chura R, Gullick NJ, Salerno R, Rönnelid J, Alexanderson H, Lindroos E, Aggarwal R, Gordon P, Vencovsky J, Lundberg IE. Abatacept in the treatment of adult dermatomyositis and polymyositis: a randomised, phase IIb treatment delayed-start trial. Ann Rheum Dis 2018; 77:55-62. [PMID: 28993346 DOI: 10.1136/annrheumdis-2017-211751] [Citation(s) in RCA: 81] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2017] [Revised: 07/14/2017] [Accepted: 08/05/2017] [Indexed: 11/04/2022]
Abstract
OBJECTIVES To study the effects of abatacept on disease activity and on muscle biopsy features of adult patients with dermatomyositis (DM) or polymyositis (PM). METHODS Twenty patients with DM (n=9) or PM (n=11) with refractory disease were enrolled in a randomised treatment delayed-start trial to receive either immediate active treatment with intravenous abatacept or a 3 month delayed-start. The primary endpoint was number of responders, defined by the International Myositis Assessment and Clinical Studies Group definition of improvement (DOI), after 6 months of treatment. Secondary endpoints included number of responders in the early treatment arm compared with the delayed treatment arm at 3 months. Repeated muscle biopsies were investigated for cellular markers and cytokines. RESULTS 8/19 patients included in the analyses achieved the DOI at 6 months. At 3 months of study, five (50%) patients were responders after active treatment but only one (11%) patient in the delayed treatment arm. Eight adverse events (AEs) were regarded as related to the drug, four mild and four moderate, and three serious AEs, none related to the drug. There was a significant increase in regulatory T cells (Tregs), whereas other markers were unchanged in repeated muscle biopsies. CONCLUSIONS In this pilot study, treatment of patients with DM and PM with abatacept resulted in lower disease activity in nearly half of the patients. In patients with repeat muscle biopsies, an increased frequency of Foxp3+ Tregs suggests a positive effect of treatment in muscle tissue.
Collapse
Affiliation(s)
- Anna Tjärnlund
- Rheumatology Unit, Department of Medicine, Karolinska University Hospital Solna, Karolinska Institutet, Stockholm, Sweden
| | - Quan Tang
- Rheumatology Unit, Department of Medicine, Karolinska University Hospital Solna, Karolinska Institutet, Stockholm, Sweden
| | - Cecilia Wick
- Rheumatology Unit, Department of Medicine, Karolinska University Hospital Solna, Karolinska Institutet, Stockholm, Sweden
| | - Maryam Dastmalchi
- Rheumatology Unit, Department of Medicine, Karolinska University Hospital Solna, Karolinska Institutet, Stockholm, Sweden
| | - Herman Mann
- Department of Rheumatology, Institute of Rheumatology, Prague, Czech Republic
| | | | - Radka Chura
- Department of Rheumatology, King's College Hospital NHS Foundation Trust, London, UK
| | - Nicola J Gullick
- Department of Rheumatology, King's College Hospital NHS Foundation Trust, London, UK
| | - Rosaria Salerno
- Department of Rheumatology, King's College Hospital NHS Foundation Trust, London, UK
| | - Johan Rönnelid
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Helene Alexanderson
- Division of Physiotherapy, Department of Neurobiology, Karolinska Institutet and Physical Therapy Clinic, Karolinska University Hospital, Stockholm, Sweden
| | - Eva Lindroos
- Rheumatology Unit, Department of Medicine, Karolinska University Hospital Solna, Karolinska Institutet, Stockholm, Sweden
| | - Rohit Aggarwal
- Division of Rheumatology and Clinical Rheumatology, University of Pittsburgh School of Medicine, Pittsburgh, USA
| | - Patrick Gordon
- Department of Rheumatology, King's College Hospital NHS Foundation Trust, London, UK
| | - Jiri Vencovsky
- Department of Rheumatology, Institute of Rheumatology, Prague, Czech Republic
| | - Ingrid E Lundberg
- Rheumatology Unit, Department of Medicine, Karolinska University Hospital Solna, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
17
|
Verstappen GM, Meiners PM, Corneth OBJ, Visser A, Arends S, Abdulahad WH, Hendriks RW, Vissink A, Kroese FGM, Bootsma H. Attenuation of Follicular Helper T Cell-Dependent B Cell Hyperactivity by Abatacept Treatment in Primary Sjögren's Syndrome. Arthritis Rheumatol 2017; 69:1850-1861. [DOI: 10.1002/art.40165] [Citation(s) in RCA: 103] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2017] [Accepted: 05/25/2017] [Indexed: 02/07/2023]
Affiliation(s)
- Gwenny M. Verstappen
- University of Groningen, University Medical Center Groningen; Groningen The Netherlands
| | - Petra M. Meiners
- University of Groningen, University Medical Center Groningen; Groningen The Netherlands
| | | | - Annie Visser
- University of Groningen, University Medical Center Groningen; Groningen The Netherlands
| | - Suzanne Arends
- University of Groningen, University Medical Center Groningen; Groningen The Netherlands
| | - Wayel H. Abdulahad
- University of Groningen, University Medical Center Groningen; Groningen The Netherlands
| | | | - Arjan Vissink
- University of Groningen, University Medical Center Groningen; Groningen The Netherlands
| | - Frans G. M. Kroese
- University of Groningen, University Medical Center Groningen; Groningen The Netherlands
| | - Hendrika Bootsma
- University of Groningen, University Medical Center Groningen; Groningen The Netherlands
| |
Collapse
|
18
|
Bandyopadhyay S, Connolly SE, Jabado O, Ye J, Kelly S, Maldonado MA, Westhovens R, Nash P, Merrill JT, Townsend RM. Identification of biomarkers of response to abatacept in patients with SLE using deconvolution of whole blood transcriptomic data from a phase IIb clinical trial. Lupus Sci Med 2017; 4:e000206. [PMID: 29214034 PMCID: PMC5704740 DOI: 10.1136/lupus-2017-000206] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2017] [Revised: 06/05/2017] [Accepted: 06/11/2017] [Indexed: 12/13/2022]
Abstract
Objective To characterise patients with active SLE based on pretreatment gene expression-defined peripheral immune cell patterns and identify clusters enriched for potential responders to abatacept treatment. Methods This post hoc analysis used baseline peripheral whole blood transcriptomic data from patients in a phase IIb trial of intravenous abatacept (~10 mg/kg/month). Cell-specific genes were used with a published deconvolution algorithm to identify immune cell proportions in patient samples, and unsupervised consensus clustering was generated. Efficacy data were re-analysed. Results Patient data (n=144: abatacept: n=98; placebo: n=46) were grouped into four main clusters (C) by predominant characteristic cells: C1—neutrophils; C2—cytotoxic T cells, B-cell receptor-ligated B cells, monocytes, IgG memory B cells, activated T helper cells; C3—plasma cells, activated dendritic cells, activated natural killer cells, neutrophils; C4—activated dendritic cells, cytotoxic T cells. C3 had the highest baseline total British Isles Lupus Assessment Group (BILAG) scores, highest antidouble-stranded DNA autoantibody levels and shortest time to flare (TTF), plus trends in favour of response to abatacept over placebo: adjusted mean difference in BILAG score over 1 year, −4.78 (95% CI −12.49 to 2.92); median TTF, 56 vs 6 days; greater normalisation of complement component 3 and 4 levels. Differential improvements with abatacept were not seen in other clusters, except for median TTF in C1 (201 vs 109 days). Conclusions Immune cell clustering segmented disease severity and responsiveness to abatacept. Definition of immune response cell types may inform design and interpretation of SLE trials and treatment decisions. Trial registration number NCT00119678; results.
Collapse
Affiliation(s)
| | - Sean E Connolly
- US Medical, Bristol-Myers Squibb, Princeton, New Jersey, USA
| | - Omar Jabado
- Translational Bioinformatics, Bristol-Myers Squibb, Princeton, New Jersey, USA
| | - June Ye
- Global Biometric Sciences, Bristol-Myers Squibb, Princeton, New Jersey, USA
| | - Sheila Kelly
- US Medical, Bristol-Myers Squibb, Princeton, New Jersey, USA
| | | | - Rene Westhovens
- Department of Development and Regeneration KU Leuven, Skeletal Biology and Engineering Research Center; Rheumatology, University Hospitals Leuven, Leuven, Belgium
| | - Peter Nash
- Department of Medicine, University of Queensland, Brisbane, Queensland, Australia
| | - Joan T Merrill
- Arthritis and Immunology Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma, USA
| | - Robert M Townsend
- Clinical Biomarkers, Bristol-Myers Squibb, Princeton, New Jersey, USA
| |
Collapse
|
19
|
McInnes IB, Schett G. Pathogenetic insights from the treatment of rheumatoid arthritis. Lancet 2017; 389:2328-2337. [PMID: 28612747 DOI: 10.1016/s0140-6736(17)31472-1] [Citation(s) in RCA: 895] [Impact Index Per Article: 111.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2017] [Revised: 04/25/2017] [Accepted: 04/26/2017] [Indexed: 12/16/2022]
Abstract
Rheumatoid arthritis is a chronic autoimmune disease that causes progressive articular damage, functional loss, and comorbidity. The development of effective biologics and small-molecule kinase inhibitors in the past two decades has substantially improved clinical outcomes. Just as understanding of pathogenesis has led in large part to the development of drugs, so have mode-of-action studies of these specific immune-targeted agents revealed which immune pathways drive articular inflammation and related comorbidities. Cytokine inhibitors have definitively proven a critical role for tumour necrosis factor α and interleukin 6 in disease pathogenesis and possibly also for granulocyte-macrophage colony-stimulating factor. More recently, clinical trials with Janus kinase (JAK) inhibitors have shown that cytokine receptors that signal through the JAK/STAT signalling pathway are important for disease, informing the pathogenetic function of additional cytokines (such as the interferons). Finally, successful use of costimulatory blockade and B-cell depletion in the clinic has revealed that the adaptive immune response and the downstream events initiated by these cells participate directly in synovial inflammation. Taken together, it becomes apparent that understanding the effects of specific immune interventions can elucidate definitive molecular or cellular nodes that are essential to maintain complex inflammatory networks that subserve diseases like rheumatoid arthritis.
Collapse
Affiliation(s)
- Iain B McInnes
- Institute of Infection Immunity and Inflammation, University of Glasgow, Glasgow, UK.
| | - Georg Schett
- Department of Internal Medicine 3-Rheumatology and Immunology, Friedrich-Alexander-University Erlangen-Nürnberg, Erlangen, Germany; Universitätsklinikum Erlangen, Erlangen, Germany
| |
Collapse
|
20
|
Alissafi T, Banos A, Boon L, Sparwasser T, Ghigo A, Wing K, Vassilopoulos D, Boumpas D, Chavakis T, Cadwell K, Verginis P. Tregs restrain dendritic cell autophagy to ameliorate autoimmunity. J Clin Invest 2017; 127:2789-2804. [PMID: 28581446 DOI: 10.1172/jci92079] [Citation(s) in RCA: 91] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2016] [Accepted: 04/07/2017] [Indexed: 12/26/2022] Open
Abstract
Design of efficacious Treg-based therapies and establishment of clinical tolerance in autoimmune diseases have proven to be challenging. The clinical implementation of Treg immunotherapy has been hampered by various impediments related to the stability and isolation procedures of Tregs as well as the specific in vivo targets of Treg modalities. Herein, we have demonstrated that Foxp3+ Tregs potently suppress autoimmune responses in vivo through inhibition of the autophagic machinery in DCs in a cytotoxic T-lymphocyte-associated protein 4-dependent (CTLA4-dependent) manner. Autophagy-deficient DCs exhibited reduced immunogenic potential and failed to prime autoantigen-specific CD4+ T cells to mediate autoimmunity. Mechanistically, CTLA4 binding promoted activation of the PI3K/Akt/mTOR axis and FoxO1 nuclear exclusion in DCs, leading to decreased transcription of the autophagy component microtubule-associated protein 1 light chain 3β (Lc3b). Human DCs treated with CTLA4-Ig, a fusion protein composed of the Fc region of IgG1 and the extracellular domain of CTLA4 (also known as abatacept, marketed as Orencia), demonstrated reduced levels of autophagosome formation, while DCs from CTLA4-Ig-treated rheumatoid arthritis patients displayed diminished LC3B transcripts. Collectively, our data identify the canonical autophagy pathway in DCs as a molecular target of Foxp3+ Treg-mediated suppression that leads to amelioration of autoimmune responses. These findings may pave the way for the development of therapeutic protocols that exploit Tregs for the treatment of autoimmunity as well as diseases in which disturbed tolerance is a common denominator.
Collapse
Affiliation(s)
- Themis Alissafi
- Biomedical Research Foundation of the Academy of Athens, Athens, Greece
| | - Aggelos Banos
- Biomedical Research Foundation of the Academy of Athens, Athens, Greece
| | | | - Tim Sparwasser
- Institute of Infection Immunology, TWINCORE, Centre for Experimental and Clinical Infection Research, Hannover, Germany
| | - Alessandra Ghigo
- Molecular Biotechnology Center, Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy
| | - Kajsa Wing
- Division of Medical Inflammation Research, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Dimitrios Vassilopoulos
- Joint Rheumatology Program, Clinical Immunology-Rheumatology Unit, National and Kapodistrian University of Athens Medical School, Hippokration General Hospital, Athens, Greece
| | - Dimitrios Boumpas
- Biomedical Research Foundation of the Academy of Athens, Athens, Greece.,Joint Rheumatology Program, 4th Department of Medicine, Attikon University Hospital, National and Kapodistrian University of Athens Medical School, Athens, Greece
| | - Triantafyllos Chavakis
- Department of Clinical Pathobiochemistry, Institute for Clinical Chemistry and Laboratory Medicine and Department of Internal Medicine, University of Dresden, Dresden, Germany
| | - Ken Cadwell
- Kimmel Center for Biology and Medicine at the Skirball Institute, New York, New York, USA.,Departments of Microbiology and Medicine, New York University School of Medicine, New York, New York, USA
| | | |
Collapse
|
21
|
Wallberg M, Recino A, Phillips J, Howie D, Vienne M, Paluch C, Azuma M, Wong FS, Waldmann H, Cooke A. Anti-CD3 treatment up-regulates programmed cell death protein-1 expression on activated effector T cells and severely impairs their inflammatory capacity. Immunology 2017; 151:248-260. [PMID: 28211040 PMCID: PMC5418468 DOI: 10.1111/imm.12729] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2016] [Revised: 02/03/2017] [Accepted: 02/13/2017] [Indexed: 01/07/2023] Open
Abstract
T cells play a key role in the pathogenesis of type 1 diabetes, and targeting the CD3 component of the T‐cell receptor complex provides one therapeutic approach. Anti‐CD3 treatment can reverse overt disease in spontaneously diabetic non‐obese diabetic mice, an effect proposed to, at least in part, be caused by a selective depletion of pathogenic cells. We have used a transfer model to further investigate the effects of anti‐CD3 treatment on green fluorescent protein (GFP)+ islet‐specific effector T cells in vivo. The GFP expression allowed us to isolate the known effectors at different time‐points during treatment to assess cell presence in various organs as well as gene expression and cytokine production. We find, in this model, that anti‐CD3 treatment does not preferentially deplete the transferred effector cells, but instead inhibits their metabolic function and their production of interferon‐γ. Programmed cell death protein 1 (PD‐1) expression was up‐regulated on the effector cells from anti‐CD3‐treated mice, and diabetes induced through anti‐PD‐L1 antibody could only be reversed with anti‐CD3 antibody if the anti‐CD3 treatment lasted beyond the point when the anti‐PD‐L1 antibody was washed out of the system. This suggests that PD‐1/PD‐L1 interaction plays an important role in the anti‐CD3 antibody mediated protection. Our data demonstrate an additional mechanism by which anti‐CD3 therapy can reverse diabetogenesis.
Collapse
Affiliation(s)
- Maja Wallberg
- Department of Pathology, University of Cambridge, Cambridge, UK
| | - Asha Recino
- Department of Pathology, University of Cambridge, Cambridge, UK
| | - Jenny Phillips
- Department of Pathology, University of Cambridge, Cambridge, UK
| | - Duncan Howie
- Sir William Dunn School of Pathology, University of Oxford, Oxford, UK
| | - Margaux Vienne
- Department of Pathology, University of Cambridge, Cambridge, UK
| | | | - Miyuki Azuma
- Department of Molecular Immunology Graduate School, Tokyo Medical and Dental University, Tokyo, Japan
| | - F Susan Wong
- Diabetes Research Group, Institute of Molecular and Experimental Medicine, Cardiff School of Medicine, Cardiff University, Cardiff, UK
| | - Herman Waldmann
- Sir William Dunn School of Pathology, University of Oxford, Oxford, UK
| | - Anne Cooke
- Department of Pathology, University of Cambridge, Cambridge, UK
| |
Collapse
|
22
|
Abstract
Rheumatoid arthritis is a chronic inflammatory joint disease, which can cause cartilage and bone damage as well as disability. Early diagnosis is key to optimal therapeutic success, particularly in patients with well-characterised risk factors for poor outcomes such as high disease activity, presence of autoantibodies, and early joint damage. Treatment algorithms involve measuring disease activity with composite indices, applying a treatment-to-target strategy, and use of conventional, biological, and newz non-biological disease-modifying antirheumatic drugs. After the treatment target of stringent remission (or at least low disease activity) is maintained, dose reduction should be attempted. Although the prospects for most patients are now favourable, many still do not respond to current therapies. Accordingly, new therapies are urgently required. In this Seminar, we describe current insights into genetics and aetiology, pathophysiology, epidemiology, assessment, therapeutic agents, and treatment strategies together with unmet needs of patients with rheumatoid arthritis.
Collapse
Affiliation(s)
- Josef S Smolen
- Division of Rheumatology, Department of Medicine 3, Medical University of Vienna, Vienna, Austria; 2nd Department of Medicine, Hietzing Hospital Vienna, Vienna, Austria.
| | - Daniel Aletaha
- Division of Rheumatology, Department of Medicine 3, Medical University of Vienna, Vienna, Austria
| | - Iain B McInnes
- Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, UK
| |
Collapse
|
23
|
Ubiquitous LEA29Y Expression Blocks T Cell Co-Stimulation but Permits Sexual Reproduction in Genetically Modified Pigs. PLoS One 2016; 11:e0155676. [PMID: 27175998 PMCID: PMC4866763 DOI: 10.1371/journal.pone.0155676] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2016] [Accepted: 05/03/2016] [Indexed: 12/15/2022] Open
Abstract
We have successfully established and characterized a genetically modified pig line with ubiquitous expression of LEA29Y, a human CTLA4-Ig derivate. LEA29Y binds human B7.1/CD80 and B7.2/CD86 with high affinity and is thus a potent inhibitor of T cell co-stimulation via this pathway. We have characterized the expression pattern and the biological function of the transgene as well as its impact on the porcine immune system and have evaluated the potential of these transgenic pigs to propagate via assisted breeding methods. The analysis of LEA29Y expression in serum and multiple organs of CAG-LEA transgenic pigs revealed that these animals produce a biologically active transgenic product at a considerable level. They present with an immune system affected by transgene expression, but can be maintained until sexual maturity and propagated by assisted reproduction techniques. Based on previous experience with pancreatic islets expressing LEA29Y, tissues from CAG-LEA29Y transgenic pigs should be protected against rejection by human T cells. Furthermore, their immune-compromised phenotype makes CAG-LEA29Y transgenic pigs an interesting large animal model for testing human cell therapies and will provide an important tool for further clarifying the LEA29Y mode of action.
Collapse
|