1
|
Miura S, Habibabady ZA, Pollok F, Ma M, Rosales IA, Kinoshita K, Pratts S, McGrath G, Chaban R, Fogarty S, Meibohm B, Daugherty B, Lederman S, Pierson RN. TNX-1500, a crystallizable fragment-modified anti-CD154 antibody, prolongs nonhuman primate cardiac allograft survival. Am J Transplant 2023; 23:1182-1193. [PMID: 37030662 PMCID: PMC10524282 DOI: 10.1016/j.ajt.2023.03.025] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Revised: 03/16/2023] [Accepted: 03/29/2023] [Indexed: 04/10/2023]
Abstract
Blockade of the CD40/CD154 T cell costimulation pathway is a promising approach to supplement or replace current clinical immunosuppression in solid organ transplantation. We evaluated the tolerability and activity of a novel humanized anti-CD154 monoclonal antibody, TNX-1500 (TNX), in a nonhuman primate heterotopic cardiac allogeneic (allo) transplant model. TNX-1500 contains a rupluzimab fragment antigen-binding region and an immunoglobin G4 crystallizable fragment region engineered to reduce binding to the crystallizable fragment gamma receptor IIa and associated risks of thrombosis. Recipients were treated for 6 months with standard-dose TNX (sTNX) monotherapy, low-dose TNX monotherapy (loTNX), or loTNX with mycophenolate mofetil (MMF) (loTNX + MMF). Results were compared with historical data using chimeric humanized 5c8 monotherapy dosed as for loTNX but discontinued at 3 months. Median survival time was similar for humanized 5c8 and both loTNX groups, but significantly longer with sTNX (>265 days) than with loTNX (99 days) or loTNX + MMF (88 days) (P < 0.05 for both comparisons against sTNX). Standard-dose TNX prevented antidonor alloantibody elaboration, inhibited chronic rejection, and was associated with a significantly reduced effector T cells/regulatory T cells ratio relative to loTNX with MMF. No thrombotic complications were observed. This study demonstrated that TNX was well tolerated, prolongs allograft survival, and prevents alloantibody production and cardiac allograft vasculopathy in a stringent preclinical nonhuman primate heart allotransplant model.
Collapse
Affiliation(s)
- Shuhei Miura
- Center for Transplantation Sciences, Department of Surgery, Massachusetts General Hospital, Boston, Massachusetts, USA; Department of Cardiovascular Surgery, Sapporo Medical University, Sapporo, Japan; Department of Cardiovascular Surgery, Teine Keijinkai Hospital, Sapporo, Japan.
| | - Zahra A Habibabady
- Center for Transplantation Sciences, Department of Surgery, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Franziska Pollok
- Center for Transplantation Sciences, Department of Surgery, Massachusetts General Hospital, Boston, Massachusetts, USA; Department of Anesthesiology, University Hospital Hamburg-Eppendorf, Hamburg, Germany
| | - Madelyn Ma
- Center for Transplantation Sciences, Department of Surgery, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Ivy A Rosales
- Department of Pathology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Kohei Kinoshita
- Center for Transplantation Sciences, Department of Surgery, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Shannon Pratts
- Center for Transplantation Sciences, Department of Surgery, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Gannon McGrath
- Center for Transplantation Sciences, Department of Surgery, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Ryan Chaban
- Center for Transplantation Sciences, Department of Surgery, Massachusetts General Hospital, Boston, Massachusetts, USA
| | | | - Bernd Meibohm
- College of Pharmacy, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | | | | | - Richard N Pierson
- Center for Transplantation Sciences, Department of Surgery, Massachusetts General Hospital, Boston, Massachusetts, USA.
| |
Collapse
|
2
|
Ramos-Martínez I, Ramos-Martínez E, Cerbón M, Pérez-Torres A, Pérez-Campos Mayoral L, Hernández-Huerta MT, Martínez-Cruz M, Pérez-Santiago AD, Sánchez-Medina MA, García-Montalvo IA, Zenteno E, Matias-Cervantes CA, Ojeda-Meixueiro V, Pérez-Campos E. The Role of B Cell and T Cell Glycosylation in Systemic Lupus Erythematosus. Int J Mol Sci 2023; 24:863. [PMID: 36614306 PMCID: PMC9820943 DOI: 10.3390/ijms24010863] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Revised: 12/05/2022] [Accepted: 12/06/2022] [Indexed: 01/05/2023] Open
Abstract
Glycosylation is a post-translational modification that affects the stability, structure, antigenicity and charge of proteins. In the immune system, glycosylation is involved in the regulation of ligand-receptor interactions, such as in B-cell and T-cell activating receptors. Alterations in glycosylation have been described in several autoimmune diseases, such as systemic lupus erythematosus (SLE), in which alterations have been found mainly in the glycosylation of B lymphocytes, T lymphocytes and immunoglobulins. In immunoglobulin G of lupus patients, a decrease in galactosylation, sialylation, and nucleotide fucose, as well as an increase in the N-acetylglucosamine bisector, are observed. These changes in glycoisolation affect the interactions of immunoglobulins with Fc receptors and are associated with pericarditis, proteinuria, nephritis, and the presence of antinuclear antibodies. In T cells, alterations have been described in the glycosylation of receptors involved in activation, such as the T cell receptor; these changes affect the affinity with their ligands and modulate the binding to endogenous lectins such as galectins. In T cells from lupus patients, a decrease in galectin 1 binding is observed, which could favor activation and reduce apoptosis. Furthermore, these alterations in glycosylation correlate with disease activity and clinical manifestations, and thus have potential use as biomarkers. In this review, we summarize findings on glycosylation alterations in SLE and how they relate to immune system defects and their clinical manifestations.
Collapse
Affiliation(s)
- Ivan Ramos-Martínez
- Departamento de Medicina y Zootecnia de Cerdos, Facultad de Medicina Veterinaria y Zootecnia, Universidad Nacional Autónoma de México, Ciudad de México 04510, Mexico
| | - Edgar Ramos-Martínez
- Facultad de Química, Universidad Nacional Autónoma de México, Ciudad de México 04510, Mexico
- Escuela de Ciencias, Universidad Autónoma Benito Juárez de Oaxaca, Oaxaca 68120, Mexico
| | - Marco Cerbón
- Unidad de Investigación en Reproducción Humana, Instituto Nacional de Perinatología “Isidro Espinosa de los Reyes”—Facultad de Química, Universidad Nacional Autónoma de México, Ciudad de México 04510, Mexico
| | - Armando Pérez-Torres
- Departamento de Biología Celular y Tisular, Facultad de Medicina, Universidad Nacional Autónoma de México, Ciudad de México 04510, Mexico
| | | | - María Teresa Hernández-Huerta
- CONACyT, Facultad de Medicina y Cirugía, Universidad Autónoma “Benito Juárez” de Oaxaca (UABJO), Oaxaca 68020, Mexico
| | | | | | | | | | - Edgar Zenteno
- Departamento de Bioquímica, Facultad de Medicina, Universidad Nacional Autónoma de México, Ciudad de México 04510, Mexico
| | | | | | | |
Collapse
|
3
|
Immune-relevant genes of systemic lupus erythematosus by transcriptome profiling analysis. Cytokine 2022; 158:155975. [PMID: 35964416 DOI: 10.1016/j.cyto.2022.155975] [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: 04/11/2022] [Revised: 07/18/2022] [Accepted: 07/19/2022] [Indexed: 11/23/2022]
Abstract
OBJECTIVE Our study aimed to reveal the roles of long noncoding RNA (lncRNA) and immune-relevant genes in systemic lupus erythematosus (SLE). METHODS Expression profiling dataset GSE65391 consisted of 72 healthy blood samples and 924 SLE blood samples was downloaded from the Gene Expression Omnibus. Differentially expressed RNAs (DERs) in SLE samples were identified using the Limma package. Immune-relevant DERs were uncovered after assessing the immune cell infiltration types of different samples. Modules significantly related to the disease were extracted via weighted gene co-expression network analysis (WGCNA), followed by module-trait analysis. LncRNA-mRNA co-expression network was constructed for DERs in immune-relevant modules. Genes related to SLE were further revealed via the Comparative Toxicogenomics Database (CTD). Validation assay was conducted by another independent expression profiling dataset, GSE46907 (consisted of 5 healthy blood samples and 5 SLE blood samples) and quantitative reverse transcription-polymerase chain reaction (qRT-PCR) analysis of 15 healthy blood samples and 15 SLE blood samples. RESULTS A total of 606 DERs, including 19 lncRNAs and 587 mRNAs, were identified. Obvious differences were observed in the proportions of 20 immune cell types, and 378 immune-relevant DERs were revealed. Genes in the lncRNA-mRNA co-expression network were significantly enriched in primary immunodeficiency, hematopoietic cell lineage, B cell receptor signaling pathway, SLE (up-regulated FCGR3A, down-regulated CD40LG, up-regulated HIST1H2BE, down-regulated human leukocyte antigen [HLA]-DOA, down-regulated HLA-DOB, up-regulated HIST1H3D, and up-regulated HIST1H2BC), and intestinal immune network for IgA production. SLE-related gene, CD40LG, retrieved from CTD has co-expression interactions with seven differentially expressed lncRNAs (HCG27, LINC02555, LINC02210, DHRS4-AS1, MIR600HG, DANCR, and LINC01278). CD40LG and HLA-DOA, were observed down-regulated, and FCGR3A, and HIST1H2BE were up-regulated in validation dataset GES46907. Moreover, CD40LG was validated to be down-regulated using qRT-PCR. CONCLUSIONS Our results provide new insight in understanding the pathogenesis of SLE and might be helpful for developing new therapeutic approaches of SLE by modulating immune response.
Collapse
|
4
|
Cognasse F, Duchez AC, Audoux E, Ebermeyer T, Arthaud CA, Prier A, Eyraud MA, Mismetti P, Garraud O, Bertoletti L, Hamzeh-Cognasse H. Platelets as Key Factors in Inflammation: Focus on CD40L/CD40. Front Immunol 2022; 13:825892. [PMID: 35185916 PMCID: PMC8850464 DOI: 10.3389/fimmu.2022.825892] [Citation(s) in RCA: 71] [Impact Index Per Article: 23.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Accepted: 01/14/2022] [Indexed: 12/16/2022] Open
Abstract
Platelets are anucleate cytoplasmic fragments derived from the fragmentation of medullary megakaryocytes. Activated platelets adhere to the damaged endothelium by means of glycoproteins on their surface, forming the platelet plug. Activated platelets can also secrete the contents of their granules, notably the growth factors contained in the α-granules, which are involved in platelet aggregation and maintain endothelial activation, but also contribute to vascular repair and angiogenesis. Platelets also have a major inflammatory and immune function in antibacterial defence, essentially through their Toll-like Receptors (TLRs) and Sialic acid-binding immunoglobulin-type lectin (SIGLEC). Platelet activation also contributes to the extensive release of anti- or pro-inflammatory mediators such as IL-1β, RANTES (Regulated on Activation, Normal T Expressed and Secreted) or CD154, also known as the CD40-ligand. Platelets are involved in the direct activation of immune cells, polynuclear neutrophils (PNNs) and dendritic cells via the CD40L/CD40 complex. As a general rule, all of the studies presented in this review show that platelets are capable of covering most of the stages of inflammation, primarily through the CD40L/CD40 interaction, thus confirming their own role in this pathophysiological condition.
Collapse
Affiliation(s)
- Fabrice Cognasse
- Etablissement Français du Sang Auvergne-Rhône-Alpes, Saint-Etienne, France.,SAINBIOSE, INSERM, U1059, University of Lyon, Saint-Etienne, France
| | - Anne Claire Duchez
- Etablissement Français du Sang Auvergne-Rhône-Alpes, Saint-Etienne, France.,SAINBIOSE, INSERM, U1059, University of Lyon, Saint-Etienne, France
| | - Estelle Audoux
- Etablissement Français du Sang Auvergne-Rhône-Alpes, Saint-Etienne, France.,SAINBIOSE, INSERM, U1059, University of Lyon, Saint-Etienne, France
| | - Theo Ebermeyer
- Etablissement Français du Sang Auvergne-Rhône-Alpes, Saint-Etienne, France.,SAINBIOSE, INSERM, U1059, University of Lyon, Saint-Etienne, France
| | - Charles Antoine Arthaud
- Etablissement Français du Sang Auvergne-Rhône-Alpes, Saint-Etienne, France.,SAINBIOSE, INSERM, U1059, University of Lyon, Saint-Etienne, France
| | - Amelie Prier
- Etablissement Français du Sang Auvergne-Rhône-Alpes, Saint-Etienne, France.,SAINBIOSE, INSERM, U1059, University of Lyon, Saint-Etienne, France
| | - Marie Ange Eyraud
- Etablissement Français du Sang Auvergne-Rhône-Alpes, Saint-Etienne, France.,SAINBIOSE, INSERM, U1059, University of Lyon, Saint-Etienne, France
| | - Patrick Mismetti
- SAINBIOSE, INSERM, U1059, University of Lyon, Saint-Etienne, France.,Vascular and Therapeutic Medicine Department, Saint-Etienne University Hospital Center, Saint-Etienne, France
| | - Olivier Garraud
- SAINBIOSE, INSERM, U1059, University of Lyon, Saint-Etienne, France
| | - Laurent Bertoletti
- SAINBIOSE, INSERM, U1059, University of Lyon, Saint-Etienne, France.,Vascular and Therapeutic Medicine Department, Saint-Etienne University Hospital Center, Saint-Etienne, France
| | | |
Collapse
|
5
|
Piantoni S, Regola F, Masneri S, Merletti M, Lowin T, Airò P, Tincani A, Franceschini F, Andreoli L, Pongratz G. Characterization of B- and T-Cell Compartment and B-Cell Related Factors Belonging to the TNF/TNFR Superfamily in Patients With Clinically Active Systemic Lupus Erythematosus: Baseline BAFF Serum Levels Are the Strongest Predictor of Response to Belimumab after Twelve Months of Therapy. Front Pharmacol 2021; 12:666971. [PMID: 34093196 PMCID: PMC8176088 DOI: 10.3389/fphar.2021.666971] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Accepted: 04/30/2021] [Indexed: 12/14/2022] Open
Abstract
Background: Patients with systemic lupus erythematosus (SLE) show increased serum levels of tumor necrosis factor (TNF)/TNF receptor (R) superfamily member, e.g. BAFF (B lymphocyte stimulator). Belimumab, a monoclonal antibody against soluble BAFF, is used for treatment of SLE. Although B cells are the main target, a BAFF-dependent T-cell activation pathway also plays a role. High levels of anti-DNA antibodies and low complement at baseline are known predictors of response to Belimumab. Objectives: To explore the association of circulating lymphocytes and serum levels of B- cell related TNF/TNFR superfamily members with response to Belimumab in SLE patients. Methods: Twenty-one SLE patients received Belimumab. Clinical evaluation and laboratory tests were performed at baseline, at 6 and 12 months. TNF super-family members (BAFF, APRIL, sBCMA, sCD40L, sTACI, TWEAK) were tested by high-sensitivity ELISA in all patients, and lymphocyte immunophenotyping was performed by flow cytometry in ten subjects. SLE-disease activity was assessed by SLEDAI-2K score. Linear regression modeling was used to investigate parameters influencing SLEDAI-2K and anti-dsDNA antibody titers over time and for predictive models. Results: Clinical improvement was observed in all patients. A global reduction of circulating B cells, especially naïve, was detected, without variation in the T-cell compartment. All TNF family members decreased, whereas APRIL remained constant. The increase in serum levels of C3 (p = 0.0004) and sTACI (p = 0.0285) was associated with a decrease of SLEDAI-2K. The increase of C4 (p = 0.027) and sBCMA (p = 0.0015) and the increase of CD8+ T cells (p = 0.0160) were associated with a decrease, whereas an increase of sCD40L in serum (p = 0.0018) and increased number of CD4+ T cells (p = 0.0029) were associated with an increase, in anti-dsDNA antibody titers, respectively. Using stepwise forward inclusion, the minimal model to predict SLEDAI-2K response at 12 months included BAFF (p = 3.0e − 07) and SLEDAI-2K (p = 7.0e − 04) at baseline. Baseline APRIL levels also showed an association, although the overall model fit was weaker. Conclusion: In our real-life cohort, baseline serum levels of BAFF were the best predictor of response to Belimumab, confirming post-hoc results of the BLISS study and suggesting the utility of this particular biomarker for the identification of patients who are more likely to respond.
Collapse
Affiliation(s)
- Silvia Piantoni
- Rheumatology and Clinical Immunology Unit, ASST Spedali Civili of Brescia, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - Francesca Regola
- Rheumatology and Clinical Immunology Unit, ASST Spedali Civili of Brescia, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - Stefania Masneri
- Rheumatology and Clinical Immunology Unit, ASST Spedali Civili of Brescia, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - Michele Merletti
- Rheumatology and Clinical Immunology Unit, ASST Spedali Civili of Brescia, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - Torsten Lowin
- Department of Rheumatology and Hiller Research Center for Rheumatology, University Hospital Düsseldorf, Düsseldorf, Germany
| | - Paolo Airò
- Rheumatology and Clinical Immunology Unit, ASST Spedali Civili of Brescia, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - Angela Tincani
- Rheumatology and Clinical Immunology Unit, ASST Spedali Civili of Brescia, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - Franco Franceschini
- Rheumatology and Clinical Immunology Unit, ASST Spedali Civili of Brescia, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - Laura Andreoli
- Rheumatology and Clinical Immunology Unit, ASST Spedali Civili of Brescia, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - Georg Pongratz
- Department of Rheumatology and Hiller Research Center for Rheumatology, University Hospital Düsseldorf, Düsseldorf, Germany
| |
Collapse
|
6
|
Furie RA, Bruce IN, Dörner T, Leon MG, Leszczyński P, Urowitz M, Haier B, Jimenez T, Brittain C, Liu J, Barbey C, Stach C. Phase 2, Randomized, Placebo-Controlled Trial of Dapirolizumab Pegol in Patients with Moderate-to-Severe Active Systemic Lupus Erythematosus. Rheumatology (Oxford) 2021; 60:5397-5407. [PMID: 33956056 PMCID: PMC9194804 DOI: 10.1093/rheumatology/keab381] [Citation(s) in RCA: 61] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Revised: 04/14/2021] [Indexed: 11/18/2022] Open
Abstract
Objective To evaluate the dose–response, efficacy and safety of dapirolizumab pegol (DZP) in
patients with SLE. Methods Adults with moderately to severely active SLE (SLEDAI-2K score ≥6 and ≥1 BILAG A or ≥2
BILAG B domain scores), receiving stable CS (≤40 mg/day prednisone-equivalent),
antimalarial or immunosuppressant drugs were included. Patients with stable LN
(proteinuria ≤2 g/day) not receiving high-dose CS or CYC were permitted entry.
Randomized patients received placebo or i.v. DZP (6/24/45 mg/kg) and standard-of-care
(SOC) treatment every 4 weeks to week 24, after which patients received only SOC to week
48. The primary objective was to establish a dose–response relationship based on week 24
BILAG-Based Composite Lupus Assessment (BICLA) responder rates. Results All DZP groups exhibited improvements in clinical and immunological outcomes
vs placebo at week 24; however, BICLA responder rates did not fit
pre-specified dose–response models [best-fitting model
(Emax): P = 0.07]. Incidences of serious
treatment-emergent adverse events across DZP groups were low and similar to placebo.
Following DZP withdrawal, SLEDAI-2K, physician’s global assessment (PGA), BILAG, and
Cutaneous Lupus Erythematosus Disease Area and Severity Index (CLASI) scores stabilized;
BICLA and SLE Responder Index (SRI-4) responder rates declined (likely due to
interventions with disallowed escape medications), BILAG flares increased, and
immunologic parameters returned towards baseline. Conclusions Although the primary objective was not met, DZP appeared to be well tolerated, and
patients exhibited improvements across multiple clinical and immunological measures of
disease activity after 24 weeks relative to placebo. The potential clinical benefit of
DZP warrants further investigation.
Collapse
Affiliation(s)
| | - Ian N Bruce
- NIHR Manchester Biomedical Research Centre, Manchester University Hospitals NHS Trust, Manchester, UK.,Centre for Musculoskeletal Research, University of Manchester, Manchester Academic Health Science Centre, Manchester, UK
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
7
|
Cognasse F, Hally K, Fauteux-Daniel S, Eyraud MA, Arthaud CA, Fagan J, Mismetti P, Hamzeh-Cognasse H, Laradi S, Garraud O, Larsen P. Effects and Side Effects of Platelet Transfusion. Hamostaseologie 2021; 41:128-135. [PMID: 33711849 DOI: 10.1055/a-1347-6551] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Aside from their canonical role in hemostasis, it is increasingly recognized that platelets have inflammatory functions and can regulate both adaptive and innate immune responses. The main topic this review aims to cover is the proinflammatory effects and side effects of platelet transfusion. Platelets prepared for transfusion are subject to stress injury upon collection, preparation, and storage. With these types of stress, they undergo morphologic, metabolic, and functional modulations which are likely to induce platelet activation and the release of biological response modifiers (BRMs). As a consequence, platelet concentrates (PCs) accumulate BRMs during processing and storage, and these BRMs are ultimately transfused alongside platelets. It has been shown that BRMs present in PCs can induce immune responses and posttransfusion reactions in the transfusion recipient. Several recent reports within the transfusion literature have investigated the concept of platelets as immune cells. Nevertheless, current and future investigations will face the challenge of encompassing the immunological role of platelets in the scope of transfusion.
Collapse
Affiliation(s)
- Fabrice Cognasse
- Etablissement Français du Sang Auvergne-Rhône-Alpes, Saint-Étienne, France.,SAINBIOSE, INSERM U1059, University of Lyon, Université Jean-Monnet-Saint-Etienne, France, France
| | - Kathryn Hally
- Department of Surgery and Anaesthesia, University of Otago, Wellington, New Zealand.,Wellington Cardiovascular Research Group, Wellington, New Zealand.,School of Biological Sciences, Victoria University of Wellington, Wellington, New Zealand
| | - Sebastien Fauteux-Daniel
- Etablissement Français du Sang Auvergne-Rhône-Alpes, Saint-Étienne, France.,SAINBIOSE, INSERM U1059, University of Lyon, Université Jean-Monnet-Saint-Etienne, France, France
| | - Marie-Ange Eyraud
- Etablissement Français du Sang Auvergne-Rhône-Alpes, Saint-Étienne, France.,SAINBIOSE, INSERM U1059, University of Lyon, Université Jean-Monnet-Saint-Etienne, France, France
| | - Charles-Antoine Arthaud
- Etablissement Français du Sang Auvergne-Rhône-Alpes, Saint-Étienne, France.,SAINBIOSE, INSERM U1059, University of Lyon, Université Jean-Monnet-Saint-Etienne, France, France
| | - Jocelyne Fagan
- Etablissement Français du Sang Auvergne-Rhône-Alpes, Saint-Étienne, France.,SAINBIOSE, INSERM U1059, University of Lyon, Université Jean-Monnet-Saint-Etienne, France, France
| | - Patrick Mismetti
- SAINBIOSE, INSERM U1059, University of Lyon, Université Jean-Monnet-Saint-Etienne, France, France
| | - Hind Hamzeh-Cognasse
- SAINBIOSE, INSERM U1059, University of Lyon, Université Jean-Monnet-Saint-Etienne, France, France
| | - Sandrine Laradi
- Etablissement Français du Sang Auvergne-Rhône-Alpes, Saint-Étienne, France.,SAINBIOSE, INSERM U1059, University of Lyon, Université Jean-Monnet-Saint-Etienne, France, France
| | - Olivier Garraud
- SAINBIOSE, INSERM U1059, University of Lyon, Université Jean-Monnet-Saint-Etienne, France, France
| | - Peter Larsen
- Department of Surgery and Anaesthesia, University of Otago, Wellington, New Zealand.,Wellington Cardiovascular Research Group, Wellington, New Zealand.,School of Biological Sciences, Victoria University of Wellington, Wellington, New Zealand
| |
Collapse
|
8
|
Transcriptional Regulation of CD40 Expression by 4 Ribosomal Proteins via a Functional SNP on a Disease-Associated CD40 Locus. Genes (Basel) 2020; 11:genes11121526. [PMID: 33371207 PMCID: PMC7767238 DOI: 10.3390/genes11121526] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Revised: 12/13/2020] [Accepted: 12/14/2020] [Indexed: 01/02/2023] Open
Abstract
Previously, using FREP-MS, we identified a protein complex including eight proteins that specifically bind to the functional SNP (fSNP) rs6032664 at a CD40 locus associated with autoimmune diseases. Among these eight proteins, four are ribosomal proteins RPL26, RPL4, RPL8, and RPS9 that normally make up the ribosomal subunits involved in the cellular process of protein translation. So far, no publication has shown these ribosomal proteins function as transcriptional regulators. In this work, we demonstrate that four ribosomal proteins: RPL26, RPL4, RPL8, and RPS9 are bona fide CD40 transcriptional regulators via binding to rs6032664. In addition, we show that suppression of CD40 expression by RPL26 RNAi knockdown inactivates NF-κB p65 by dephosphorylation via NF-κB signaling pathway in fibroblast-like synoviocytes (FLS), which further reduces the transcription of disease-associated risk genes such as STAT4, CD86, TRAF1 and ICAM1 as the direct targets of NF-κB p65. Based on these findings, a disease-associated risk gene transcriptional regulation network (TRN) is generated, in which decreased expression of, at least, RPL26 results in the downregulation of risk genes: STAT4, CD86, TRAF1 and ICAM1, as well as the two proinflammatory cytokines: IL1β and IL6 via CD40-induced NF-κB signaling. We believe that further characterization of this disease-associated TRN in the CD40-induced NF-κB signaling by identifying both the upstream and downstream regulators will potentially enable us to identify the best targets for drug development.
Collapse
|
9
|
Mousavi MJ, Mahmoudi M, Ghotloo S. Escape from X chromosome inactivation and female bias of autoimmune diseases. Mol Med 2020; 26:127. [PMID: 33297945 PMCID: PMC7727198 DOI: 10.1186/s10020-020-00256-1] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Accepted: 12/02/2020] [Indexed: 12/13/2022] Open
Abstract
Generally, autoimmune diseases are more prevalent in females than males. Various predisposing factors, including female sex hormones, X chromosome genes, and the microbiome have been implicated in the female bias of autoimmune diseases. During embryogenesis, one of the X chromosomes in the females is transcriptionally inactivated, in a process called X chromosome inactivation (XCI). This equalizes the impact of two X chromosomes in the females. However, some genes escape from XCI, providing a basis for the dual expression dosage of the given gene in the females. In the present review, the contribution of the escape genes to the female bias of autoimmune diseases will be discussed.
Collapse
Affiliation(s)
- Mohammad Javad Mousavi
- Department of Hematology, Faculty of Allied Medicine, Bushehr University of Medical Sciences, Bushehr, Iran
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Mahdi Mahmoudi
- Rheumatology Research Center, Tehran University of Medical Sciences, Tehran, Iran.
| | - Somayeh Ghotloo
- Department of Medical Laboratory Sciences, School of Allied Medical Sciences, Kashan University of Medical Sciences, Kashan, Iran.
| |
Collapse
|
10
|
Bachsais M, Salti S, Zaoui K, Hassan GS, Aoudjit F, Mourad W. CD154 inhibits death of T cells via a Cis interaction with the α5β1 integrin. PLoS One 2020; 15:e0235753. [PMID: 32745080 PMCID: PMC7398495 DOI: 10.1371/journal.pone.0235753] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Accepted: 06/22/2020] [Indexed: 12/30/2022] Open
Abstract
CD154 plays a major role in the pathogenesis of several autoimmune and inflammatory diseases. In addition to CD40, soluble CD154 (sCD154) binds to other receptors namely αIIbβ3, αMβ2, α5β1 and αvβ3 integrins. We have previously reported that binding of sCD154 to α5β1 integrin expressed on several human T cell lines is capable of inhibiting Fas-induced cell death. In the current study, we show that such effect of the sCD154/α5β1 interaction is not restricted to the cell death response induced by Fas but could also be exhibited toward other death signals such as TRAIL and TNF- α. We also demonstrate that sCD154 is capable of inhibiting Fas-mediated death of human activated T cells, more importantly of CD4+ than CD8+ T ones. Our data also show that membrane-bound CD154 and α5β1 integrin expressed on the surface of distinct cells failed to influence cell death responses. However, when membrane-bound CD154 and α5β1 are expressed on the surface of same cell, their interaction was capable of down regulating cell death. CD154 was shown to co-localize with the α5β1 integrin on the surface of these cells. These data strongly suggest a cis-type of interaction between CD154 and α5β1 when both are expressed on the same cell surface, rather than a trans-interaction which usually implicates the ligand and its receptor each expressed on the surface of a distinct cell. Taken together, these findings add to the list of roles through which CD154 is contributing to the pathogenesis of autoimmune-inflammatory diseases, i.e. by protecting T cells from death and enhancing their survival.
Collapse
Affiliation(s)
- Meriem Bachsais
- Laboratoire d’Immunologie Cellulaire et Moléculaire, Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CR-CHUM), Montréal, Québec, Canada
| | - Suzanne Salti
- Laboratoire d’Immunologie Cellulaire et Moléculaire, Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CR-CHUM), Montréal, Québec, Canada
| | - Kossay Zaoui
- Laboratoire d’Immunologie Cellulaire et Moléculaire, Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CR-CHUM), Montréal, Québec, Canada
| | - Ghada S. Hassan
- Laboratoire d’Immunologie Cellulaire et Moléculaire, Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CR-CHUM), Montréal, Québec, Canada
| | - Fawzi Aoudjit
- Centre de recherche du CHU de Québec-Université Laval, Québec, Québec, Canada
| | - Walid Mourad
- Laboratoire d’Immunologie Cellulaire et Moléculaire, Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CR-CHUM), Montréal, Québec, Canada
- * E-mail:
| |
Collapse
|
11
|
Immune checkpoint molecules. Possible future therapeutic implications in autoimmune diseases. J Autoimmun 2019; 104:102333. [DOI: 10.1016/j.jaut.2019.102333] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Accepted: 08/29/2019] [Indexed: 02/07/2023]
|
12
|
Sivakumar R, Abboud G, Mathews CE, Atkinson MA, Morel L. Protective Role of Myeloid Cells Expressing a G-CSF Receptor Polymorphism in an Induced Model of Lupus. Front Immunol 2018; 9:1053. [PMID: 29868014 PMCID: PMC5954343 DOI: 10.3389/fimmu.2018.01053] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2017] [Accepted: 04/27/2018] [Indexed: 01/11/2023] Open
Abstract
The genetic analysis of the lupus-prone NZM2410 mouse has identified a suppressor locus, Sle2c2, which confers resistance to spontaneous lupus in combination with NZM2410 susceptibility loci, or in the chronic graft-versus-host disease (cGVHD) induced model of lupus in the B6.Sle2c2 congenic strain. The candidate gene for Sle2c2, the Csf3r gene encoding the granulocyte colony-stimulating factor receptor (G-CSF-R/CD114), was validated when cGVHD was restored in B6.Sle2c2 mice after treatment with G-CSF. The goal of the project reported herein was to investigate the myeloid cells that confer resistance to cGVHD and to ascertain if the mechanism behind their suppression involves the G-CSF pathway. We showed that despite expressing the highest levels of G-CSF-R, neutrophils play only a modest role in the autoimmune activation induced by cGVHD. We also found reduced expression levels of G-CSF-R on the surface of dendritic cells (DCs) and a differential distribution of DC subsets in response to cGVHD in B6.Sle2c2 versus B6 mice. The CD8α+ DC subset, known for its tolerogenic phenotype, was expanded upon induction of cGVHD in B6.Sle2c2 mice. In addition, the deficiency of CD8α+ DC subset enhanced the severity of cGVHD in B6.Batf3-/- and B6.Sle2c2 mice, confirming their role in suppression of cGVHD. B6.Sle2c2DCs presented lowered activation and antigen presentation abilities and expressed lower levels of genes associated with DC activation and maturation. Exposure to exogenous G-CSF reversed the majority of these phenotypes, suggesting that tolerogenic DCs maintained through a defective G-CSF-R pathway mediated the resistance to cGVHD in B6.Sle2c2 mice.
Collapse
Affiliation(s)
- Ramya Sivakumar
- Department of Pathology, Immunology, Laboratory Medicine, University of Florida Diabetes Institute, University of Florida, Gainesville, FL, United States
| | - Georges Abboud
- Department of Pathology, Immunology, Laboratory Medicine, University of Florida Diabetes Institute, University of Florida, Gainesville, FL, United States
| | - Clayton E Mathews
- Department of Pathology, Immunology, Laboratory Medicine, University of Florida Diabetes Institute, University of Florida, Gainesville, FL, United States
| | - Mark A Atkinson
- Department of Pathology, Immunology, Laboratory Medicine, University of Florida Diabetes Institute, University of Florida, Gainesville, FL, United States
| | - Laurence Morel
- Department of Pathology, Immunology, Laboratory Medicine, University of Florida Diabetes Institute, University of Florida, Gainesville, FL, United States
| |
Collapse
|
13
|
Lee JI, Choi YJ, Park HJ, Jung KC, Park SH. RD-05, a novel anti-CD154 antibody, efficiently inhibits generation of anti-drug antibody without the risk of thrombus formation in non-human primates. Biochem Biophys Res Commun 2018; 498:996-1001. [PMID: 29550477 DOI: 10.1016/j.bbrc.2018.03.099] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2018] [Accepted: 03/13/2018] [Indexed: 11/28/2022]
Abstract
Antibody formation against therapeutic agents, such as tumor necrosis factor inhibitors and Factor VIII, that leads to treatment failure has become a major challenge in the treatment of rheumatoid arthritis and hemophilia. It is well known that anti-CD154 antibodies have the highest potential to inhibit these types of adverse immune responses. Nevertheless, the formation of thromboemboli is the major hurdle in the clinical application of these anti-CD154 blocking antibodies. For this, we attempted to derive an idea as to how this major complication can be eliminated. Consequently, we developed a novel anti-CD154 chimeric antibody, which was made by genetic modification of a portion of human IgG4 Fc. This antibody has an almost comparable antigen binding affinity to a previously developed 5C8 clone and near completely inhibited CD40-CD154 interaction and T cell-dependent B cell activation in vitro. Even under the condition, where we injected immune complexes comprised of RD-05 and CD154 antigen, the formation of thromboembolism was not seen in human FcγRIIA-transgenic mice, whereas the converse was exactly true in the case of 5C8 antibody. Notably, just two injections of RD-05 antibody was sufficient to inhibit the antibody formation against adalimumab during 3-4 months in cynomolgus macaques, in which adalimumab was repeatedly injected for 12 weeks. Based on these findings, we suggest that this RD-05 antibody can be applied to antibody-mediated autoimmune diseases, including systemic lupus erythematosus and immune thrombocytopenic purpura.
Collapse
Affiliation(s)
- Jae-Il Lee
- Transplantation Research Institute, Seoul National University Medical Research Center, Seoul, 03080, Republic of Korea; Department of Medicine, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea; Graduate Course of Translational Medicine, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea
| | - Yun-Jung Choi
- Graduate Course of Translational Medicine, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea
| | - Hi-Jung Park
- Graduate Course of Translational Medicine, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea
| | - Kyeong Cheon Jung
- Transplantation Research Institute, Seoul National University Medical Research Center, Seoul, 03080, Republic of Korea; Graduate Course of Translational Medicine, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea; Department of Pathology, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea.
| | - Seong Hoe Park
- Transplantation Research Institute, Seoul National University Medical Research Center, Seoul, 03080, Republic of Korea; Department of Medicine, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea.
| |
Collapse
|
14
|
Cassia M, Alberici F, Gallieni M, Jayne D. Lupus nephritis and B-cell targeting therapy. Expert Rev Clin Immunol 2017; 13:951-962. [PMID: 28800401 DOI: 10.1080/1744666x.2017.1366855] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
INTRODUCTION Lupus Nephritis (LN) is a severe manifestation of Systemic Lupus Erythematosus (SLE) with a significant prognostic impact. Over a prolonged course, an exhaustion of treatment alternatives may occur and further therapeutic options are needed. B cells play a pivotal role in disease pathogenesis and represent an attractive therapeutic target. Areas covered: This review provides an update regarding targeting B cells in LN. The rational for this approach, as well as currently available and future targets are discussed. Expert commentary: Despite its wide clinical use and the encouraging results from retrospective studies, a role of rituximab in LN has not been prospectively confirmed. Trial design methodologies as well as intrinsic limitations of this approach may be responsible and rituximab use is currently limited as a rescue treatment or in settings where a strong steroid sparing effect is warranted. Despite belimumab now being licensed for use in SLE, the evidence in LN is weak although prospective trials are on-going. The combination of different targeted approaches as well as a focus on new clinical end-points may be strategies to identify new therapeutic options.
Collapse
Affiliation(s)
- Matthias Cassia
- a Nephrology and Immunology Unit, ASST Santi Paolo e Carlo , San Carlo Borromeo Hospital , Milano , Italy.,b Department of Biomedical and Clinical Sciences "L.Sacco" , University of Milan , Milano , Italy
| | - Federico Alberici
- a Nephrology and Immunology Unit, ASST Santi Paolo e Carlo , San Carlo Borromeo Hospital , Milano , Italy
| | - Maurizio Gallieni
- a Nephrology and Immunology Unit, ASST Santi Paolo e Carlo , San Carlo Borromeo Hospital , Milano , Italy.,b Department of Biomedical and Clinical Sciences "L.Sacco" , University of Milan , Milano , Italy
| | - David Jayne
- c Department of Medicine , University of Cambridge , Cambridge , UK
| |
Collapse
|
15
|
Li G, Cunin P, Wu D, Diogo D, Yang Y, Okada Y, Plenge RM, Nigrovic PA. The Rheumatoid Arthritis Risk Variant CCR6DNP Regulates CCR6 via PARP-1. PLoS Genet 2016; 12:e1006292. [PMID: 27626929 PMCID: PMC5023119 DOI: 10.1371/journal.pgen.1006292] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2016] [Accepted: 08/10/2016] [Indexed: 12/29/2022] Open
Abstract
Understanding the implications of genome-wide association studies (GWAS) for disease biology requires both identification of causal variants and definition of how these variants alter gene function. The non-coding triallelic dinucleotide polymorphism CCR6DNP is associated with risk for rheumatoid arthritis, and is considered likely causal because allelic variation correlates with expression of the chemokine receptor CCR6. Using transcription activator-like effector nuclease (TALEN) gene editing, we confirmed that CCR6DNP regulates CCR6. To identify the associated transcription factor, we applied a novel assay, Flanking Restriction Enhanced Pulldown (FREP), to identify specific association of poly (ADP-ribose) polymerase 1 (PARP-1) with CCR6DNP consistent with the established allelic risk hierarchy. Correspondingly, manipulation of PARP-1 expression or activity impaired CCR6 expression in several lineages. These findings show that CCR6DNP is a causal variant through which PARP-1 regulates CCR6, and introduce a highly efficient approach to interrogate non-coding genetic polymorphisms associated with human disease. Genome-wide association studies (GWAS) identify loci associated with human disease risk, but bridging the gap between locus and mechanism has proven particularly difficult in cases where associated variants do not alter coding. We aimed to develop a generalizable approach to this problem. Previously, a dual nucleotide polymorphism within the first intron of CCR6 (termed the CCR6DNP) had been associated with risk for rheumatoid arthritis, but the pathway by which this variant altered gene expression could not be determined. Here, we employed sequence perturbation to confirm a regulatory role for the CCR6DNP. Next, using a new technique termed Flanking Restriction Enhanced Pulldown (FREP), we identified PARP-1 as the protein that regulates CCR6 expression through allelic association with the CCR6DNP, a finding confirmed by chromatin immunoprecipitation and functional assays. These findings reveal an unexpected regulatory pathway for CCR6 implicated in rheumatoid arthritis and other disease by human genetics, and more generally introduce a novel approach to identifying regulatory protein-DNA interactions.
Collapse
Affiliation(s)
- Gang Li
- Division of Rheumatology, Immunology and Allergy, Brigham and Women’s Hospital, Boston, Massachusetts, United States of America
- * E-mail: (GL); (PAN)
| | - Pierre Cunin
- Division of Rheumatology, Immunology and Allergy, Brigham and Women’s Hospital, Boston, Massachusetts, United States of America
| | - Di Wu
- Division of Rheumatology, Immunology and Allergy, Brigham and Women’s Hospital, Boston, Massachusetts, United States of America
- Department of Statistics, Harvard University, Cambridge, Massachusetts, United States of America
- Centre for Cancer Research, Monash Institute of Medical Research, Monash University, Clayton, Victoria, Australia
| | - Dorothée Diogo
- Division of Rheumatology, Immunology and Allergy, Brigham and Women’s Hospital, Boston, Massachusetts, United States of America
- Division of Genetics, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
- Program in Medical and Population Genetics, Broad Institute, Cambridge, Massachusetts, United States of America
| | - Yu Yang
- Division of Rheumatology, Immunology and Allergy, Brigham and Women’s Hospital, Boston, Massachusetts, United States of America
| | - Yukinori Okada
- Department of Human Genetics and Disease Diversity, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
- Laboratory for Statistical Analysis, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
- Department of Statistical Genetics, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Robert M. Plenge
- Division of Rheumatology, Immunology and Allergy, Brigham and Women’s Hospital, Boston, Massachusetts, United States of America
| | - Peter A. Nigrovic
- Division of Rheumatology, Immunology and Allergy, Brigham and Women’s Hospital, Boston, Massachusetts, United States of America
- Division of Immunology, Boston Children’s Hospital, Boston, Massachusetts, United States of America
- * E-mail: (GL); (PAN)
| |
Collapse
|
16
|
Abstract
Atherosclerosis is a chronic inflammatory disease that is initiated by the retention and accumulation of cholesterol-containing lipoproteins, particularly low-density lipoprotein, in the artery wall. In the arterial intima, lipoprotein components that are generated through oxidative, lipolytic, and proteolytic activities lead to the formation of several danger-associated molecular patterns, which can activate innate immune cells as well as vascular cells. Moreover, self- and non-self-antigens, such as apolipoprotein B-100 and heat shock proteins, can contribute to vascular inflammation by triggering the response of T and B cells locally. This process can influence the initiation, progression, and stability of plaques. Substantial clinical and experimental data support that the modulation of adaptive immune system may be used for treating and preventing atherosclerosis. This may lead to the development of more selective and less harmful interventions, while keeping host defense mechanisms against infections and tumors intact. Approaches such as vaccination might become a realistic option for cardiovascular disease, especially if they can elicit regulatory T and B cells and the secretion of atheroprotective antibodies. Nevertheless, difficulties in translating certain experimental data into new clinical therapies remain a challenge. In this review, we discuss important studies on the function of T- and B-cell immunity in atherosclerosis and their manipulation to develop novel therapeutic strategies against cardiovascular disease.
Collapse
Affiliation(s)
- Daniel F J Ketelhuth
- From the Department of Medicine, Center for Molecular Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden.
| | - Göran K Hansson
- From the Department of Medicine, Center for Molecular Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
17
|
Chimote AA, Hajdu P, Kottyan LC, Harley JB, Yun Y, Conforti L. Nanovesicle-targeted Kv1.3 knockdown in memory T cells suppresses CD40L expression and memory phenotype. J Autoimmun 2016; 69:86-93. [PMID: 26994905 DOI: 10.1016/j.jaut.2016.03.004] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2016] [Revised: 03/07/2016] [Accepted: 03/08/2016] [Indexed: 02/06/2023]
Abstract
Ca(2+) signaling controls activation and effector functions of T lymphocytes. Ca(2+) levels also regulate NFAT activation and CD40 ligand (CD40L) expression in T cells. CD40L in activated memory T cells binds to its cognate receptor, CD40, on other cell types resulting in the production of antibodies and pro-inflammatory mediators. The CD40L/CD40 interaction is implicated in the pathogenesis of autoimmune disorders and CD40L is widely recognized as a therapeutic target. Ca(2+) signaling in T cells is regulated by Kv1.3 channels. We have developed lipid nanoparticles that deliver Kv1.3 siRNAs (Kv1.3-NPs) selectively to CD45RO(+) memory T cells and reduce the activation-induced Ca(2+) influx. Herein we report that Kv1.3-NPs reduced NFAT activation and CD40L expression exclusively in CD45RO(+) T cells. Furthermore, Kv1.3-NPs suppressed cytokine release and induced a phenotype switch of T cells from predominantly memory to naïve. These findings indicate that Kv1.3-NPs operate as targeted immune suppressive agents with promising therapeutic potentials.
Collapse
Affiliation(s)
- Ameet A Chimote
- Department of Internal Medicine, Division of Nephrology, University of Cincinnati, Cincinnati, OH, USA
| | - Peter Hajdu
- Department of Internal Medicine, Division of Nephrology, University of Cincinnati, Cincinnati, OH, USA
| | - Leah C Kottyan
- Center for Autoimmune Genomics and Etiology, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - John B Harley
- Center for Autoimmune Genomics and Etiology, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, OH, USA; US Department of Veterans Affairs Medical Center, Cincinnati, OH, USA
| | - Yeoheung Yun
- North Carolina A & T State University, Chemical, Biological and Bioengineering Department, Greensboro, NC, USA
| | - Laura Conforti
- Department of Internal Medicine, Division of Nephrology, University of Cincinnati, Cincinnati, OH, USA.
| |
Collapse
|
18
|
Arman M, Krauel K. Human platelet IgG Fc receptor FcγRIIA in immunity and thrombosis. J Thromb Haemost 2015; 13:893-908. [PMID: 25900780 DOI: 10.1111/jth.12905] [Citation(s) in RCA: 100] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2014] [Accepted: 03/10/2015] [Indexed: 01/23/2023]
Abstract
Beyond their prominent role in hemostasis and thrombosis, platelets are increasingly recognized as having immunologic functions. Supporting this, human platelets express FcγRIIA (CD32a), a low-affinity Fc receptor (FcR) for the constant region of IgG that recognizes immune complexes (ICs) and IgG-opsonized cells with high avidity. In leukocytes, FcγRIIA engagement initiates strong effector functions that are key for immune and inflammatory responses, including cytokine release, antibody-dependent cell-mediated killing of pathogens, and internalization of ICs. However, the physiologic relevance of platelet-expressed FcγRIIA has received little attention in previous reviews on FcRs. This article summarizes and discusses the available information on human platelet FcγRIIA. The importance of this receptor in heparin-induced thrombocytopenia, a prothrombotic adverse drug effect, is well documented. However, studies demonstrating platelet activation by IgG-opsonized bacteria point to the physiologic relevance of platelet FcγRIIA in immunity. In this context, platelet activation and secretion may facilitate both a direct antimicrobial function of platelets and crosstalk with other immune cells. Additionally, a role for platelet FcγRIIA in IgG-independent hemostasis and physiologic thrombosis, by means of amplifying integrin αII b β3 outside-in signaling, has also been proposed. Nonetheless, the thrombotic complications found in some infective and autoimmune diseases may result from unbalanced FcγRIIA-mediated platelet aggregation. Moreover, FcγRIIA is not expressed in mice, and thrombocytopenia and/or thrombotic events found after drug administration can only be recapitulated by the use of human FcγRIIA-transgenic mice. Altogether, the available data support a functional role for platelet FcγRIIA in health and disease, and emphasize the need for further investigation of this receptor.
Collapse
Affiliation(s)
- M Arman
- Centre for Cardiovascular Sciences, Institute of Biomedical Research, School of Clinical and Experimental Medicine, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - K Krauel
- Institut für Immunologie und Transfusionsmedizin, Universitätsmedizin Greifswald, Greifswald, Germany
| |
Collapse
|
19
|
Ketelhuth DFJ, Hansson GK. Modulation of autoimmunity and atherosclerosis - common targets and promising translational approaches against disease. Circ J 2015; 79:924-33. [PMID: 25766275 DOI: 10.1253/circj.cj-15-0167] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Atherosclerosis is a chronic inflammatory disease of the arterial wall that is influenced by several risk factors, including hyperlipidemia and hypertension. Autoimmune diseases substantially increase the risk for cardiovascular disease (CVD). Although atherosclerotic CVD, such as myocardial and stroke, is much more prevalent than classical autoimmune conditions such as rheumatoid arthritis, psoriasis, and systemic lupus erythematosus, these types of pathology have many similarities, raising the possibility that therapies against autoimmune disease can have beneficial effects on CVD. Substantial clinical and experimental data support the potential for immunomodulatory approaches to combating both autoimmune and cardiovascular diseases, including classical immunosuppressants, anticytokine therapy, the targeting of T and B cells and their responses, and vaccination. In this review, we discuss experimental and clinical studies that have used immunomodulatory approaches to mitigate autoimmune reactions and examine their potential to prevent and treat atherosclerotic CVD.
Collapse
Affiliation(s)
- Daniel F J Ketelhuth
- Center for Molecular Medicine, Department of Medicine, Karolinska Institutet, Karolinska University Hospital
| | | |
Collapse
|
20
|
Crist SA, Elzey BD, Ahmann MT, Ratliff TL. Early growth response-1 (EGR-1) and nuclear factor of activated T cells (NFAT) cooperate to mediate CD40L expression in megakaryocytes and platelets. J Biol Chem 2013; 288:33985-33996. [PMID: 24106272 DOI: 10.1074/jbc.m113.511881] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Increasing evidence implicates circulating platelets as mediators of chronic inflammatory and autoimmune diseases via the expression and release of CD40L, an important modulator of inflammation and adaptive immune responses traditionally associated with activated T cells. Emerging evidence suggests that platelet CD40L is dynamically regulated in several chronic inflammatory and autoimmune diseases and may mediate progression and secondary pathology associated with those disease states. The present study identifies NFATc2 as a key transcriptional modulator of CD40L expression in megakaryocytes and inflammatory activity of platelets. Furthermore, the current data show that EGR-1, a member of the early growth response family of zinc finger transcription factors, modulates NFATc2-dependent regulation of CD40L expression in megakaryocytes. Our novel demonstration that in vivo biochemical or genetic inhibition of NFATc2 activity in megakaryocyte diminishes platelet CD40L implicates the NFATc2/EGR-1 axis as a key regulatory pathway of inflammatory and immunomodulatory activity in platelets and represents a target for the development of therapeutics for the potential treatment of chronic inflammatory and autoimmune diseases.
Collapse
Affiliation(s)
- Scott A Crist
- Department of Comparative Pathobiology, Purdue University School of Veterinary Medicine, West Lafayette, Indiana 47907; Purdue Center for Cancer Research, Purdue University, West Lafayette, Indiana 47907
| | - Bennett D Elzey
- Department of Comparative Pathobiology, Purdue University School of Veterinary Medicine, West Lafayette, Indiana 47907; Purdue Center for Cancer Research, Purdue University, West Lafayette, Indiana 47907
| | - Michelle T Ahmann
- Department of Comparative Pathobiology, Purdue University School of Veterinary Medicine, West Lafayette, Indiana 47907
| | - Timothy L Ratliff
- Department of Comparative Pathobiology, Purdue University School of Veterinary Medicine, West Lafayette, Indiana 47907; Purdue Center for Cancer Research, Purdue University, West Lafayette, Indiana 47907.
| |
Collapse
|
21
|
Abstract
Systemic lupus erythematosus (SLE) is a chronic, multisystem disorder characterised by loss of tolerance to endogenous nuclear antigens and autoantibody formation. Recent insight into the immunopathogenesis of lupus has provided the foundation for a novel class of agents which target specific, dysregulated components of the immune system. Efforts have focused predominantly on B-cell depleting therapies, of which belimumab was the first to demonstrate success in phase III studies and thus receive marketing authorisation. Off-label prescribing of rituximab in refractory cases is common and supported by uncontrolled studies, which suggest a favourable risk:benefit profile. However, two placebo-controlled trials failed to show benefit, possibly because of inappropriate patient selection and other aspects of trial methodology. Inhibition of dysregulated co-stimulatory signals and cytokines are other therapeutic strategies currently under investigation. Some candidate drugs failed to meet primary endpoints in early-phase clinical trials, yet demonstrated clinical benefit when alternative assessment criteria were applied or specific patient sub-groups analysed. Well-designed studies of greater size and duration are needed to clarify the therapeutic utility of these agents. Future immunomodulatory strategies targeting interferon-alpha, T cells, oxidative stress and epigenetic abnormalities may reduce multisystem disease activity and prolong survival in this complex and heterogeneic disease.
Collapse
Affiliation(s)
- P Grech
- King's College London, School of Medicine, London, UK; and 2King’s College London, The Rayne Institute, St Thomas’ Hospital, London, UK
| | - MA Khamashta
- King's College London, School of Medicine, London, UK; and 2King’s College London, The Rayne Institute, St Thomas’ Hospital, London, UK
| |
Collapse
|
22
|
Elzey BD, Ratliff TL, Sowa JM, Crist SA. Platelet CD40L at the interface of adaptive immunity. Thromb Res 2011; 127:180-3. [PMID: 21075431 PMCID: PMC3073541 DOI: 10.1016/j.thromres.2010.10.011] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2010] [Revised: 10/15/2010] [Accepted: 10/17/2010] [Indexed: 12/13/2022]
Abstract
Initiated by the finding that platelets express functional CD40 ligand (CD40L, CD154), many new roles for platelets have been discovered in unanticipated areas, including the immune response. When current literature is considered as a whole, the picture that is emerging begins to show that platelets are able to significantly affect, for better or worse, the overall health and condition of the mammalian host. Animal models have made significant contributions to our expanding knowledge of platelet function, much of which is anticipated to be clinically relevant. While still mostly circumstantial, the evidence supports a critical role for CD40L in many normal and disease processes.
Collapse
Affiliation(s)
- Bennett D Elzey
- Department of Comparative Pathobiology, Purdue University, West Lafayette, IN, USA.
| | | | | | | |
Collapse
|
23
|
Accelerated atherosclerosis in systemic lupus erythematosus: role of proinflammatory cytokines and therapeutic approaches. J Biomed Biotechnol 2010; 2010. [PMID: 20936125 PMCID: PMC2948929 DOI: 10.1155/2010/607084] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2010] [Revised: 06/21/2010] [Accepted: 07/26/2010] [Indexed: 12/02/2022] Open
Abstract
Systemic lupus erythematosus (SLE), a chronic multisystem autoimmune disease with a broad range of clinical manifestations, is associated with accelerated atherosclerosis (AT) and increased risk of cardiovascular complications. Relevant factors directly influencing the development of AT comprise immune complex generation, complement activation, and changes in the production and activity of a complex network of cytokines, including type I and II interferons, B lymphocyte stimulator (BLyS), TNFα, IL-6, IL-17 and migration macrophage inhibitor (MIF). Autoantibodies, also responsible for cytokine expression and activation, play a supplementary key role in the development of AT. Genomic and proteomic studies have contributed to the discovery of genes and proteins involved in AT, including some that may be suitable to be used as biomarkers. All that data has allowed the development of new drugs, most of them evaluated in clinical trials: inhibitors of IFN and TNFα, B cell directed therapies, synthetic oligodeoxynucleotides, intravenous immunoglobulin, or statins. The focus of the present paper is to summarize recent evidence showing the role of cytokines in the development of AT in SLE and the rationale, and safety concerns, in the use of combined therapy to prevent AT and cardiovascular disease.
Collapse
|
24
|
Robles-Carrillo L, Meyer T, Hatfield M, Desai H, Dávila M, Langer F, Amaya M, Garber E, Francis JL, Hsu YM, Amirkhosravi A. Anti-CD40L Immune Complexes Potently Activate Platelets In Vitro and Cause Thrombosis in FCGR2A Transgenic Mice. THE JOURNAL OF IMMUNOLOGY 2010; 185:1577-83. [DOI: 10.4049/jimmunol.0903888] [Citation(s) in RCA: 103] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
25
|
Torres A, Askari AD, Malemud CJ. Cardiovascular disease complications in systemic lupus erythematosus. Biomark Med 2010; 3:239-52. [PMID: 20477476 DOI: 10.2217/bmm.09.14] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Systemic lupus erythematosus (SLE) is a highly variable autoimmune disease characterized by aberrant host-immune responses and chronic inflammation. Recently, a strong association between cardiovascular (CV) disease and SLE has emerged. Thus, low serum, high-density lipoprotein strongly correlated with elevated erythrocyte sedimentation rate, IL-6, TNF-alpha and the SLE disease activity index after adjusting for age, gender, race, BMI, insulin sensitivity and any concurrent drug use. In SLE, CV disease is characterized by increased VEGF, which may alter vascular hemostasis and promote neoangiogenesis. Increased low-density lipoprotein-cholesterol and proinflammatory high-density lipoprotein-cholesterol uptake by monocytes together with enhanced low-density lipoprotein-cholesterol oxidation results in the deposition of altered cholesterol forms into the vascular wall. This contributes to precocious and accelerated development of coronary artery plaques. Cholesterol-reducing drugs should be considered in the standard of care of SLE patients, especially in those with an unfavorable CV disease risk profile, which could reduce the probability of atherosclerosis progressing to CV disease or stroke in these patients.
Collapse
Affiliation(s)
- Alexander Torres
- Department of Medicine, Division of Rheumatic Diseases, University Hospitals Case Medical Center, 2061 Cornell Road, Cleveland, OH 44106-5076, USA
| | | | | |
Collapse
|
26
|
Estrogen receptor signaling and its relationship to cytokines in systemic lupus erythematosus. J Biomed Biotechnol 2010; 2010:317452. [PMID: 20617147 PMCID: PMC2896666 DOI: 10.1155/2010/317452] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2010] [Revised: 03/18/2010] [Accepted: 03/31/2010] [Indexed: 01/19/2023] Open
Abstract
Dysregulation of cytokines is among the main abnormalities in Systemic Lupus Erythematosus (SLE). However, although, estrogens, which are known to be involved in lupus disease,
influence cytokine production, the underlying molecular mechanisms remain poorly defined.
Recent evidence demonstrates the presence of estrogen receptor in various cell types of the
immune system, while divergent effects of estrogens on the cytokine regulation are thought to be
implicated. In this paper, we provide an overview of the current knowledge as to how estrogen-induced
modulation of cytokine production in SLE is mediated by the estrogen receptor while
simultaneously clarifying various aspects of estrogen receptor signaling in this disease. The
estrogen receptor subtypes, their structure, and the mode of action of estrogens by gene activation
and via extranuclear effects are briefly presented. Results regarding the possible correlation
between estrogen receptor gene polymorphisms and quantitative changes in the receptor protein
to SLE pathology and cytokine production are reviewed.
Collapse
|
27
|
Bartoloni Bocci E, Luccioli F, Angrisani C, Moscatelli S, Alunno A, Gerli R. Accelerated atherosclerosis in systemic lupus erythematosus and other connective tissue diseases. Expert Rev Clin Immunol 2010; 3:531-41. [PMID: 20477158 DOI: 10.1586/1744666x.3.4.531] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Connective tissue diseases are associated with increased morbidity and mortality related to a higher rate of cardiovascular events and higher prevalence of subclinical atherosclerosis. Atherosclerosis is now considered a multifactorial process where autoimmunity and chronic inflammation play an important pathogenic role. In systemic autoimmune rheumatic diseases in general, and in systemic lupus erythematosus in particular, atherosclerosis cannot be explained by traditional cardiovascular risk factors alone. Cellular and humoral mechanisms, together with specific factors associated with the disease itself and/or its treatments, have been advocated to explain the acceleration of arterial wall organic damage in these patients. Endothelial dysfunction, carotid intima-media thickness and plaque evaluations provide accurate detection of atherosclerotic process at a preclinical stage, before appearance of clinical disease, allowing preventive measure introduction with the aim to modify the cardiovascular risk in subjects with systemic autoimmune rheumatic diseases.
Collapse
Affiliation(s)
- Elena Bartoloni Bocci
- University of Perugia, Rheumatology Unit, Department of Clinical & Experimental Medicine, Policlinico Monteluce, I-06122 Perugia, Italy.
| | | | | | | | | | | |
Collapse
|
28
|
LOSS OF CD40 ENDOGENOUS S-NITROSYLATION DURING INFLAMMATORY RESPONSE IN ENDOTOXEMIC MICE AND PATIENTS WITH SEPSIS. Shock 2010; 33:626-33. [DOI: 10.1097/shk.0b013e3181cb88e6] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
|
29
|
Zhou Y, Yuan J, Pan Y, Fei Y, Qiu X, Hu N, Luo Y, Lei W, Li Y, Long H, Sawalha AH, Richardson B, Lu Q. T cell CD40LG gene expression and the production of IgG by autologous B cells in systemic lupus erythematosus. Clin Immunol 2009; 132:362-70. [PMID: 19520616 PMCID: PMC2810511 DOI: 10.1016/j.clim.2009.05.011] [Citation(s) in RCA: 79] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2008] [Revised: 05/05/2009] [Accepted: 05/12/2009] [Indexed: 02/05/2023]
Abstract
CD40 ligand (CD40LG), encoded on the X chromosome, has been reported to be overexpressed on lupus T cells. Herein, we investigated the effect of DNA demethylation on T cell CD40LG expression and the production of IgG by autologous B cells in lupus. We found normal human T cells transfected with CD40LG induced autologous B cell activation and plasma cell differentiation. Both female lupus CD4+ T cells and demethylating agents treated CD4+ T cells overexpressed CD40LG mRNA. Further, lupus T cells from both genders or demethylated CD4+ T cells from healthy women overstimulated autologous B cells, and this could be reversed with anti-CD40LG Ab in only females. We demonstrated that female lupus CD4+ T cells and demethylated CD4+ T cells express high level of CD40LG and overstimulate B cells to produce IgG. This is due to DNA demethylation and thereby reactivation of the inactive X chromosome in female.
Collapse
Affiliation(s)
- Ying Zhou
- Department of Dermatology and Epigenetic Research Center, Second Xiangya Hospital, Central South University, 139 Renmin Middle Rd, Changsha, Hunan 410011, PR China
| | - Jun Yuan
- Department of Dermatology and Epigenetic Research Center, Second Xiangya Hospital, Central South University, 139 Renmin Middle Rd, Changsha, Hunan 410011, PR China
| | - Yujun Pan
- Arthritis and Immunology Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Yiping Fei
- Arthritis and Immunology Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
- Department of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Xiangning Qiu
- Department of Dermatology and Epigenetic Research Center, Second Xiangya Hospital, Central South University, 139 Renmin Middle Rd, Changsha, Hunan 410011, PR China
| | - Nan Hu
- Department of Dermatology and Epigenetic Research Center, Second Xiangya Hospital, Central South University, 139 Renmin Middle Rd, Changsha, Hunan 410011, PR China
| | - Yongqi Luo
- Department of Dermatology and Epigenetic Research Center, Second Xiangya Hospital, Central South University, 139 Renmin Middle Rd, Changsha, Hunan 410011, PR China
| | - Wenzhi Lei
- Department of Dermatology and Epigenetic Research Center, Second Xiangya Hospital, Central South University, 139 Renmin Middle Rd, Changsha, Hunan 410011, PR China
| | - Yaping Li
- Department of Dermatology and Epigenetic Research Center, Second Xiangya Hospital, Central South University, 139 Renmin Middle Rd, Changsha, Hunan 410011, PR China
| | - Hai Long
- Department of Dermatology and Epigenetic Research Center, Second Xiangya Hospital, Central South University, 139 Renmin Middle Rd, Changsha, Hunan 410011, PR China
| | - Amr H Sawalha
- Arthritis and Immunology Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
- Department of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Department of Veterans Affairs Medical Center, Oklahoma City, OK, USA
| | - Bruce Richardson
- Department of Medicine, University of Michigan, Ann Arbor, MI, USA
- Department of Medicine, Ann Arbor VA Medical Center, Ann Arbor, MI, USA
| | - Qianjin Lu
- Department of Dermatology and Epigenetic Research Center, Second Xiangya Hospital, Central South University, 139 Renmin Middle Rd, Changsha, Hunan 410011, PR China
| |
Collapse
|
30
|
Pyrovolaki K, Mavroudi I, Sidiropoulos P, Eliopoulos AG, Boumpas DT, Papadaki HA. Increased expression of CD40 on bone marrow CD34+ hematopoietic progenitor cells in patients with systemic lupus erythematosus: Contribution to Fas-mediated apoptosis. ACTA ACUST UNITED AC 2009; 60:543-52. [DOI: 10.1002/art.24257] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
|
31
|
Abstract
T-cell activation and differentiation depend on the signal strength received by the T-cell receptor and on signals provided by co-stimulatory molecules. The most prominent co-stimulatory molecule is CD28, which controls the activation of naïve and memory T cells by antigen presented on professional antigen-presenting cells. Blocking of the CD28-CD80/86 pathway has been an appealing strategy for inducing tolerance in autoimmune diseases where the disease-inducing autoantigens are not known. Although CD28 has maintained its unique position, the past decade has witnessed the recognition that a large number of regulatory molecules on T cells must be stimulated to generate a fully protective immune response. These regulatory receptors differ in their preferential expression on T-cell subsets, in the ligands that they recognize, and in the signaling pathways that they trigger. They have in common the fact that they provide information on the cellular environment in which the T-cell response occurs. By intercepting these signals, we may be able to influence disease-relevant T-cell responses in autoimmune diseases while potentially minimizing broad immunosuppression.
Collapse
Affiliation(s)
- Jörg J Goronzy
- Kathleen B and Mason I Lowance Center for Human Immunology and Rheumatology, Emory University, Woodruff Circle, Atlanta, Georgia 30322, USA.
| | | |
Collapse
|
32
|
Driver CB, Ishimori M, Weisman MH. The B cell in systemic lupus erythaematosus: a rational target for more effective therapy. Ann Rheum Dis 2008; 67:1374-81. [PMID: 17720723 DOI: 10.1136/ard.2007.076745] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
Current treatment options for systemic lupus erythaematosus (SLE) are diverse and poorly defined, and aggressive therapy can be associated with serious toxicity and tolerability issues. There is, therefore, a need for new and improved treatments to be studied thoroughly in well-designed controlled trials. B Cell dysfunction has emerged as a key pathophysiological component of SLE and is a prime target for the development of new agents for a wide range of lupus severity, including advanced disease. Although many current drugs appear to modify B cell function, the advent of new, targeted therapies offers the hope of improved efficacy and a better long-term tolerability profile.
Collapse
Affiliation(s)
- C B Driver
- Cedars-Sinai Medical Center, Los Angeles, California 90048, USA
| | | | | |
Collapse
|
33
|
Camous L, Melander C, Vallet M, Squalli T, Knebelmann B, Noël LH, Fakhouri F. Complete Remission of Lupus Nephritis With Rituximab and Steroids for Induction and Rituximab Alone for Maintenance Therapy. Am J Kidney Dis 2008; 52:346-52. [DOI: 10.1053/j.ajkd.2008.03.036] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2007] [Accepted: 03/06/2008] [Indexed: 01/29/2023]
|
34
|
Puliaev R, Puliaeva I, Welniak L, Ryan A, Haas M, Murphy W, Via CS. CTL-promoting effects of CD40 stimulation outweigh B cell-stimulatory effects resulting in B cell elimination and disease improvement in a murine model of lupus. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2008; 181:47-61. [PMID: 18566369 PMCID: PMC2613003 DOI: 10.4049/jimmunol.181.1.47] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
CD40/CD40L signaling promotes both B cell and CTL responses in vivo, the latter being beneficial in tumor models. Because CTL may also limit autoreactive B cell expansion in lupus, we asked whether an agonist CD40 mAb would exacerbate lupus due to B cell stimulation or would improve lupus due to CTL promotion. These studies used an induced model of lupus, the parent-into-F1 model in which transfer of DBA/2 splenocytes into B6D2F1 mice induces chronic lupus-like graft-vs-host disease (GVHD). Although agonist CD40 mAb treatment of DBA-->F1 mice initially exacerbated B cell expansion, it also strongly promoted donor CD8 T cell engraftment and cytolytic activity such that by 10 days host B cells were eliminated consistent with an accelerated acute GVHD. CD40 stimulation bypassed the requirement for CD4 T cell help for CD8 CTL possibly by licensing dendritic cells (DC) as shown by the following: 1) greater initial activation of donor CD8 T cells, but not CD4 T cells; 2) earlier activation of host DC; 3) host DC expansion that was CD8 dependent and CD4 independent; and 4) induction of acute GVHD using CD4-depleted purified DBA CD8+ T cells. A single dose of CD40 mAb improved lupus-like renal disease at 12 wk, but may not suffice for longer periods consistent with a need for continuing CD8 CTL surveillance. These results demonstrate that in the setting of lupus-like CD4 T cell-driven B cell hyperactivity, CTL promotion is both feasible and beneficial and the CTL-promoting properties of CD40 stimulation outweigh the B cell-stimulatory properties.
Collapse
Affiliation(s)
- Roman Puliaev
- Pathology Department, Uniformed Services University of Health Sciences, Bethesda, MD 20814, and Research Service, Baltimore VA Medical Center, and Division of Rheumatology and Clinical Immunology, University of Maryland School of Medicine, Baltimore MD 21201
| | - Irina Puliaeva
- Pathology Department, Uniformed Services University of Health Sciences, Bethesda, MD 20814, and Research Service, Baltimore VA Medical Center, and Division of Rheumatology and Clinical Immunology, University of Maryland School of Medicine, Baltimore MD 21201
| | - Lisbeth Welniak
- Department of Microbiology and Immunology, University of Nevada School of Medicine, Reno, NV, 89557
| | - Abigail Ryan
- Pathology Department, Uniformed Services University of Health Sciences, Bethesda, MD 20814, and Research Service, Baltimore VA Medical Center, and Division of Rheumatology and Clinical Immunology, University of Maryland School of Medicine, Baltimore MD 21201
| | - Mark Haas
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21287
| | - William Murphy
- Department of Microbiology and Immunology, University of Nevada School of Medicine, Reno, NV, 89557
| | - Charles S. Via
- Pathology Department, Uniformed Services University of Health Sciences, Bethesda, MD 20814, and Research Service, Baltimore VA Medical Center, and Division of Rheumatology and Clinical Immunology, University of Maryland School of Medicine, Baltimore MD 21201
| |
Collapse
|
35
|
Abstract
Platelets are an abundant source of CD40 ligand (CD154), an immunomodulatory and proinflammatory molecule implicated in the onset and progression of several inflammatory diseases, including systemic lupus erythematosus (SLE), diabetes, and cardiovascular disease. Heretofore considered largely restricted to activated T cells, we initiated studies to investigate the source and regulation of platelet-associated CD154. We found that CD154 is abundantly expressed in platelet precursor cells, megakaryocytes. We show that CD154 is expressed in primary human CD34+ and murine hematopoietic precursor cells only after cytokine-driven megakaryocyte differentiation. Furthermore, using several established megakaryocyte-like cells lines, we performed promoter analysis of the CD154 gene and found that NFAT, a calcium-dependent transcriptional regulator associated with activated T cells, mediated both differentiation-dependent and inducible megakaryocyte-specific CD154 expression. Overall, these data represent the first investigation of the regulation of a novel source of CD154 and suggests that platelet-associated CD154 can be biochemically modulated.
Collapse
|
36
|
Westerweel PE, Luyten RKMAC, Koomans HA, Derksen RHWM, Verhaar MC. Premature atherosclerotic cardiovascular disease in systemic lupus erythematosus. ACTA ACUST UNITED AC 2007; 56:1384-96. [PMID: 17469095 DOI: 10.1002/art.22568] [Citation(s) in RCA: 96] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
|
37
|
Blanco P, Pellegrin JL, Moreau JF, Viallard JF. Physiopathologie du lupus érythémateux systémique. Presse Med 2007; 36:825-34. [PMID: 17449371 DOI: 10.1016/j.lpm.2006.11.017] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2006] [Indexed: 01/08/2023] Open
Abstract
Innate immunity is directly implicated in the pathophysiology of lupus through the dendritic cell system and the activation by immune complexes of some toll-like receptors (TLR). Interferon-alpha plays a key role in the pathophysiology of lupus and represents a promising target for immune therapy. Dendritic cells are activated and able to capture large quantities of nuclear antigen-containing bodies to stimulate specific adaptive immune response.
Collapse
Affiliation(s)
- Patrick Blanco
- Clinique de médecine interne, Hôpital Haut-Lévêque, Pessac, France.
| | | | | | | |
Collapse
|
38
|
O'Hara RM, Benoit SE, Groves CJ, Collins M. Cell-surface and cytokine biomarkers in autoimmune and inflammatory diseases. Drug Discov Today 2007; 11:342-7. [PMID: 16580976 DOI: 10.1016/j.drudis.2006.02.009] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2005] [Revised: 01/09/2006] [Accepted: 02/20/2006] [Indexed: 11/20/2022]
Abstract
Increasing emphasis is being placed on biomarkers as indicators of disease states in patients with autoimmune and inflammatory disorders, such as rheumatoid arthritis, multiple sclerosis and systemic lupus erythematosus. Careful description of the expression of cell-surface markers and cytokines produced by T and B lymphocytes can lead to a more complete characterization of disease activity in patient populations, and serve as an indicator of the patient's response to therapy.
Collapse
Affiliation(s)
- Richard M O'Hara
- Department of Inflammation, Wyeth Research, 200 CambridgePark Drive, Cambridge, MA 02140, USA.
| | | | | | | |
Collapse
|
39
|
Browning JL. B cells move to centre stage: novel opportunities for autoimmune disease treatment. Nat Rev Drug Discov 2006; 5:564-76. [PMID: 16816838 DOI: 10.1038/nrd2085] [Citation(s) in RCA: 208] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
The B-cell arm of the immune system has long been appreciated for its crucial role in pathogen resistance, but in the study of many autoimmune diseases, T cells have dominated the limelight for decades. However, the development of the B-cell-depleting antibody rituximab as a lymphoma therapy has provided a tool to probe the contribution made by B cells in several immune disorders. Recently, the success of B-cell depletion with rituximab in the treatment of rheumatoid arthritis has stimulated investigation of its effects in several other immune disorders, and considerable interest in the potential of drugs that can modulate B-cell function for the treatment of such diseases in general. This article discusses the role of B cells in a range of autoimmune disorders, including rheumatoid arthritis and systemic lupus erythematosus, and analyses approaches to therapeutic B-cell manipulation.
Collapse
Affiliation(s)
- Jeffrey L Browning
- Department of Immunobiology, Biogen Idec, 12 Cambridge Center, Cambridge, Massachusetts 02445, USA.
| |
Collapse
|
40
|
Abstract
In the past 40 years, prognosis for patients with systemic lupus erythematosus (SLE) has improved, with 10-year survival now approximately 90%. This is due probably to a combination of earlier disease diagnosis and diagnosis of milder disease, due in part to availability of multiple serological tests for SLE, use of steroids and other immunosuppressive agents, and availability of renal dialysis and transplantation. Despite this, however, the potential for significant morbidity and mortality remains in the group of patients with partially responsive or treatment resistant disease. More recently, advancements in the understanding of molecular mechanisms involved in the pathogenesis of SLE have translated to the development of novel therapies, offering possible alternatives to this patient cohort. Discussion of these pharmacological options and ongoing research forms the basis of this review.
Collapse
Affiliation(s)
- F Goldblatt
- Centre for Rheumatology, The Middlesex Hospital, University College London, Arthur Stanley House, 40-50 Tottenham Street, London W1T 4NJ, UK.
| | | |
Collapse
|
41
|
Ye X, Zhu T, Bastacky S, McHale T, Li J, Xiao X. Prevention and reversal of lupus in NZB/NZW mice by costimulatory blockade with adeno-associated virus-mediated gene transfer. ACTA ACUST UNITED AC 2005; 52:3975-86. [PMID: 16329128 DOI: 10.1002/art.21417] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
OBJECTIVE To investigate the potency of costimulatory blockade with adeno-associated virus (AAV)-mediated gene transfer in the prevention and reversal of lupus in a murine model. METHODS AAV vectors expressing CTLA-4Ig or CD40Ig were injected into NZB/NZW mice. Serum levels of transgene expression and autoantibody titers were determined by enzyme-linked immunosorbent assay. The therapeutic effects on proteinuria, renal pathologic features, and survival rate were evaluated. Splenic T cell phenotypes were analyzed by flow cytometry. The humoral immune response to a foreign antigen was also examined in treated mice. RESULTS A single injection of AAV serotype 8 (AAV8)-CTLA-4Ig in neonatal NZB/NZW mice before the onset of lupus effectively delayed and inhibited autoantibody production, proteinuria, and kidney damage and prolonged their lifespan. In addition, coinjection of AAV8-CTLA-4Ig and AAV8-CD40Ig vectors into neonatal mice achieved a synergistic effect and the best efficacy. The preventive effects were attributed to suppression of CD4+ T cell activation and the transition from naive to memory T cells. Moreover, coinjection of these 2 vectors in adult mice reversed the existing autoantibody levels, suppressed the development of proteinuria, and prolonged their lifespan. The therapeutic effects were found to be dependent on the vector dose. In addition, AAV-mediated long-term gene expression did not severely suppress the host humoral response to foreign antigen. CONCLUSION Our findings show that delivery of costimulatory inhibitor transgenes by AAV vectors could prevent and reverse lupus in this murine model, suggesting the potential of AAV-mediated gene transfer as an alternative treatment for lupus.
Collapse
Affiliation(s)
- Xiaojing Ye
- University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 12561, USA
| | | | | | | | | | | |
Collapse
|