201
|
Abstract
Novel effective immunotherapies are needed for patients with multiple myeloma (MM), since disease recurrence remains a major obstacle. B-cell maturation antigen (BCMA), a cell surface protein universally expressed on malignant plasma cells , has emerged as a very selective antigen to be targeted in novel treatments for MM. We here first review BCMA-related biology, and then highlight the recent clinical development of a novel afucosylated anti-BCMA monoclonal antibody conjugated with monomethyl auristatin F via noncleavable linker (GSK2857916). Chimeric antigen receptor-expressing T cells targeting BCMA may also induce specific and durable anti-MM responses by patients' own effector cells. Clinical trials testing these two approaches (NCT02064387, NCT02215967) are currently ongoing in relapsed and refractory MM patients.
Collapse
Affiliation(s)
- Yu-Tzu Tai
- Jerome Lipper Multiple Myeloma Center, Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, 450 Brookline Ave, Boston, MA 02215, USA
| | - Kenneth C Anderson
- Jerome Lipper Multiple Myeloma Center, Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, 450 Brookline Ave, Boston, MA 02215, USA
| |
Collapse
|
202
|
Wang Y, Li YJ, Jiang WQ, Rao HL, Huang JJ, Xia Y, Bi X, Sun P, Huang HQ, Lin TY, Guan ZZ, Li ZM. Expression of BAFF-R, but not BAFF, is an independent prognostic factor in diffuse large B-cell lymphoma patients treated with R-CHOP. Ann Hematol 2015; 94:1865-73. [PMID: 26327569 DOI: 10.1007/s00277-015-2490-0] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2015] [Accepted: 08/24/2015] [Indexed: 12/23/2022]
Abstract
B-cell activating factor (BAFF) and BAFF-receptor (BAFF-R) play crucial roles in the progression of malignant B-cells. The aim of the present study was to evaluate the expression profiles and the clinical significance of BAFF and BAFF-R in diffuse large B-cell lymphoma (DLBCL). Paraffin-embedded specimens from 136 patients with newly diagnosed DLBCL, treated with rituximab plus cyclophosphamide, doxorubicin, vincristine, and prednisone chemotherapy (R-CHOP), were examined for BAFF and BAFF-R expression by immunohistochemistry. BAFF and BAFF-R were expressed in 72.1 % (98/136) and 47.1 % (64/136) of the DLBCL tissues, respectively. Negative BAFF-R expression was significantly correlated with elevated serum lactate dehydrogenase (LDH) levels (P = 0.036), an International Prognostic Index (IPI) score of 2 or higher (P < 0.001), and a poor revised IPI (R-IPI) risk score (P = 0.043). The complete response rate after R-CHOP was higher in patients with positive BAFF-R expression than in those with negative BAFF-R expression (73.4 vs. 56.9 %, P = 0.045). Negative expression of BAFF-R, but not of BAFF, was significantly associated with inferior progression-free survival (PFS; P = 0.020) and overall survival (OS; P = 0.028). Only negative BAFF-R expression was correlated with inferior PFS and OS in multivariate analysis (P = 0.049 and 0.040, respectively). Taken together, our results showed that the majority and approximate one-half of patients with DLBCL were positive for BAFF and BAFF-R, respectively. Negative expression of BAFF-R, but not of BAFF, could be an independent risk factor for PFS and OS in patients with DLBCL treated with standard R-CHOP.
Collapse
Affiliation(s)
- Yu Wang
- State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China.,Department of Medical Oncology, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China
| | - Ya-Jun Li
- Department of Lymphoma and Hematology, Hunan Provincial Tumor Hospital, Changsha, Hunan, 410013, China.,The Affiliated Tumor Hospital of Xiangya Medical School, Central South University, Changsha, Hunan, 410013, China
| | - Wen-Qi Jiang
- State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China.,Department of Medical Oncology, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China
| | - Hui-Lan Rao
- State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China.,Department of Pathology, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China
| | - Jia-Jia Huang
- State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China.,Department of Medical Oncology, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China
| | - Yi Xia
- State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China.,Department of Medical Oncology, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China
| | - Xiwen Bi
- State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China.,Department of Medical Oncology, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China
| | - Peng Sun
- State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China.,Department of Medical Oncology, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China
| | - Hui-Qiang Huang
- State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China.,Department of Medical Oncology, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China
| | - Tong-Yu Lin
- State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China.,Department of Medical Oncology, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China
| | - Zhong-Zhen Guan
- State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China.,Department of Medical Oncology, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China
| | - Zhi-Ming Li
- State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China. .,Department of Medical Oncology, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China.
| |
Collapse
|
203
|
Furie RA, Leon G, Thomas M, Petri MA, Chu AD, Hislop C, Martin RS, Scheinberg MA. A phase 2, randomised, placebo-controlled clinical trial of blisibimod, an inhibitor of B cell activating factor, in patients with moderate-to-severe systemic lupus erythematosus, the PEARL-SC study. Ann Rheum Dis 2015; 74:1667-75. [PMID: 24748629 DOI: 10.1136/annrheumdis-2013-205144] [Citation(s) in RCA: 110] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2014] [Accepted: 03/23/2014] [Indexed: 11/04/2022]
Abstract
OBJECTIVE To evaluate the efficacy and safety of subcutaneous blisibimod, an inhibitor of B cell activating factor, in patients with systemic lupus erythematosus (SLE) in a dose-ranging Phase 2b clinical trial. METHODS 547 patients with SLE with anti-double stranded DNA or antinuclear antibodies and Safety of Estrogens in Lupus Erythematosus National Assessment-SLE Disease Activity Index (SELENA-SLEDAI) score ≥6 at baseline were randomised to receive placebo or blisibimod at one of 3 dose levels. The primary end point, measured at Week 24, was the SLE Responder Index-5 (SRI-5, meeting established SRI criteria but with ≥5 point improvement in SELENA-SLEDAI). RESULTS Although SRI-5 response rates were not significantly improved in the pooled blisibimod groups compared with placebo, they were higher in subjects randomised to the highest dose of blisibimod (200 mg once-weekly (QW)) compared with pooled placebo, from Week 16 to Week 24, reaching statistical significance at Week 20 (p=0.02). SRI response rates compared with placebo were higher still in subjects who attained SELENA-SLEDAI improvements of ≥8, and in a subgroup of patients with severe disease (SELENA-SLEDAI ≥10 and receiving corticosteroids at baseline). In subjects with protein:creatine ratios of 1-6 at baseline, significant reductions in proteinuria were observed with blisibimod. Significant (p<0.01) changes in anti-double stranded DNA antibodies, complement C3 and C4, and reductions in B cells were observed with blisibimod.No imbalances in serious adverse events or infections (4/280 and 3/266), deaths (4/280 and 3/266) and malignancies (2/280 and 2/266) were reported for blisibimod compared with placebo. CONCLUSIONS This study successfully identified a safe, effective and convenient dose, study population and end point for evaluation of blisibimod effect in Phase 3. TRIAL REGISTRATION NUMBER NCT01162681.
Collapse
Affiliation(s)
- R A Furie
- North Shore-Long Island Jewish Health System, Great Neck, New York, USA
| | - G Leon
- Rheumatology Gynecology & Reproduction Institute, Lima, Peru
| | - M Thomas
- Health and Research Centre, Trivandrum, Kerala, India
| | - M A Petri
- Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - A D Chu
- Anthera Pharmaceuticals, Hayward, California, USA
| | - C Hislop
- Anthera Pharmaceuticals, Hayward, California, USA
| | - R S Martin
- Anthera Pharmaceuticals, Hayward, California, USA
| | - M A Scheinberg
- Rheumatology Hospital Abreu Sodre Pesquisa Clínica, São Paulo, Brazil
| |
Collapse
|
204
|
Jordan NP, D'Cruz DP. Efficacy, pharmacokinetic and pharmacodynamic profile of belimumab for systemic lupus erythematosus. Expert Opin Drug Metab Toxicol 2015; 11:1635-45. [PMID: 26327145 DOI: 10.1517/17425255.2015.1077808] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
INTRODUCTION Systemic lupus erythematosus (SLE) is a severe autoimmune disease and a sizeable proportion of patients remain refractory to currently available immunosuppressive agents. The burden of uncontrolled disease is significant as disease flare and persistent inflammation lead to long-term damage accrual and increased morbidity. Belimumab , a humanized monoclonal antibody that inhibits B-lymphocyte stimulator (BLyS), is the first drug to be approved by the US FDA for the treatment of SLE in 50 years. AREAS COVERED In this review, we provide an overview of novel therapeutic agents under development for the treatment of SLE, a description of the pharmacodynamic and pharmacokinetic properties of belimumab, evidence of efficacy of belimumab as demonstrated in clinical trials, safety and tolerability data and a summary of ongoing clinical trials of belimumab in SLE. This information was amassed following a comprehensive MEDLINE and Cochrane library search. Ongoing clinical trial information was obtained from ClinicalTrials.gov. EXPERT OPINION Two, large Phase III trials have demonstrated the clinical efficacy of belimumab in musculoskeletal, mucocutaneous, haematologic and constitutional features of SLE. Patients with severe disease manifestations such as lupus nephritis and CNS disease were excluded from these trials and hence the role of belimumab in the overall management of SLE has yet to be established. Studies in lupus nephritis are ongoing.
Collapse
Affiliation(s)
- Natasha P Jordan
- a 1 Cambridge University Hospitals NHS Foundation Trust, Cambridge Biomedical Campus, Rheumatology Department , Hills Road, Cambridge CB2 0QQ, UK
| | - David P D'Cruz
- b 2 Guy's Hospital, Louise Coote Lupus Unit , 4th Floor Tower Wing, London SE1 9RT, UK +004 420 718 897 56 ; +004 420 718 835 74 ; david.d'
| |
Collapse
|
205
|
Fiala GJ, Janowska I, Prutek F, Hobeika E, Satapathy A, Sprenger A, Plum T, Seidl M, Dengjel J, Reth M, Cesca F, Brummer T, Minguet S, Schamel WWA. Kidins220/ARMS binds to the B cell antigen receptor and regulates B cell development and activation. ACTA ACUST UNITED AC 2015; 212:1693-708. [PMID: 26324445 PMCID: PMC4577850 DOI: 10.1084/jem.20141271] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2014] [Accepted: 08/14/2015] [Indexed: 01/04/2023]
Abstract
Fiala et al. report that Kidins220/ARMS is a novel interactor of the B cell antigen receptor (BCR) and its deletion impairs B cell development and B cell functioning. B cell antigen receptor (BCR) signaling is critical for B cell development and activation. Using mass spectrometry, we identified a protein kinase D–interacting substrate of 220 kD (Kidins220)/ankyrin repeat–rich membrane-spanning protein (ARMS) as a novel interaction partner of resting and stimulated BCR. Upon BCR stimulation, the interaction increases in a Src kinase–independent manner. By knocking down Kidins220 in a B cell line and generating a conditional B cell–specific Kidins220 knockout (B-KO) mouse strain, we show that Kidins220 couples the BCR to PLCγ2, Ca2+, and extracellular signal-regulated kinase (Erk) signaling. Consequently, BCR-mediated B cell activation was reduced in vitro and in vivo upon Kidins220 deletion. Furthermore, B cell development was impaired at stages where pre-BCR or BCR signaling is required. Most strikingly, λ light chain–positive B cells were reduced sixfold in the B-KO mice, genetically placing Kidins220 in the PLCγ2 pathway. Thus, our data indicate that Kidins220 positively regulates pre-BCR and BCR functioning.
Collapse
Affiliation(s)
- Gina J Fiala
- Department of Molecular Immunology, BioIII, Faculty of Biology, University of Freiburg and Max Planck Institute of Immunobiology and Epigenetics, 79104 Freiburg, Germany Centre for Biological Signaling Studies (BIOSS), Spemann Graduate School of Biology and Medicine (SGBM), Centre of Chronic Immunodeficiency (CCI), Department of Dermatology, Center for Biological Systems Analysis (ZBSA), Institute of Molecular Medicine and Cell Research, Comprehensive Cancer Centre Freiburg, and Institute of Pathology, University Medical Center Freiburg, University of Freiburg, 79104 Freiburg, Germany Centre for Biological Signaling Studies (BIOSS), Spemann Graduate School of Biology and Medicine (SGBM), Centre of Chronic Immunodeficiency (CCI), Department of Dermatology, Center for Biological Systems Analysis (ZBSA), Institute of Molecular Medicine and Cell Research, Comprehensive Cancer Centre Freiburg, and Institute of Pathology, University Medical Center Freiburg, University of Freiburg, 79104 Freiburg, Germany Centre for Biological Signaling Studies (BIOSS), Spemann Graduate School of Biology and Medicine (SGBM), Centre of Chronic Immunodeficiency (CCI), Department of Dermatology, Center for Biological Systems Analysis (ZBSA), Institute of Molecular Medicine and Cell Research, Comprehensive Cancer Centre Freiburg, and Institute of Pathology, University Medical Center Freiburg, University of Freiburg, 79104 Freiburg, Germany
| | - Iga Janowska
- Department of Molecular Immunology, BioIII, Faculty of Biology, University of Freiburg and Max Planck Institute of Immunobiology and Epigenetics, 79104 Freiburg, Germany Centre for Biological Signaling Studies (BIOSS), Spemann Graduate School of Biology and Medicine (SGBM), Centre of Chronic Immunodeficiency (CCI), Department of Dermatology, Center for Biological Systems Analysis (ZBSA), Institute of Molecular Medicine and Cell Research, Comprehensive Cancer Centre Freiburg, and Institute of Pathology, University Medical Center Freiburg, University of Freiburg, 79104 Freiburg, Germany Centre for Biological Signaling Studies (BIOSS), Spemann Graduate School of Biology and Medicine (SGBM), Centre of Chronic Immunodeficiency (CCI), Department of Dermatology, Center for Biological Systems Analysis (ZBSA), Institute of Molecular Medicine and Cell Research, Comprehensive Cancer Centre Freiburg, and Institute of Pathology, University Medical Center Freiburg, University of Freiburg, 79104 Freiburg, Germany
| | - Fabiola Prutek
- Department of Molecular Immunology, BioIII, Faculty of Biology, University of Freiburg and Max Planck Institute of Immunobiology and Epigenetics, 79104 Freiburg, Germany Centre for Biological Signaling Studies (BIOSS), Spemann Graduate School of Biology and Medicine (SGBM), Centre of Chronic Immunodeficiency (CCI), Department of Dermatology, Center for Biological Systems Analysis (ZBSA), Institute of Molecular Medicine and Cell Research, Comprehensive Cancer Centre Freiburg, and Institute of Pathology, University Medical Center Freiburg, University of Freiburg, 79104 Freiburg, Germany Centre for Biological Signaling Studies (BIOSS), Spemann Graduate School of Biology and Medicine (SGBM), Centre of Chronic Immunodeficiency (CCI), Department of Dermatology, Center for Biological Systems Analysis (ZBSA), Institute of Molecular Medicine and Cell Research, Comprehensive Cancer Centre Freiburg, and Institute of Pathology, University Medical Center Freiburg, University of Freiburg, 79104 Freiburg, Germany
| | - Elias Hobeika
- Department of Molecular Immunology, BioIII, Faculty of Biology, University of Freiburg and Max Planck Institute of Immunobiology and Epigenetics, 79104 Freiburg, Germany Centre for Biological Signaling Studies (BIOSS), Spemann Graduate School of Biology and Medicine (SGBM), Centre of Chronic Immunodeficiency (CCI), Department of Dermatology, Center for Biological Systems Analysis (ZBSA), Institute of Molecular Medicine and Cell Research, Comprehensive Cancer Centre Freiburg, and Institute of Pathology, University Medical Center Freiburg, University of Freiburg, 79104 Freiburg, Germany Institute of Immunology, University Hospital Ulm, 89081 Ulm, Germany
| | - Annyesha Satapathy
- Center of Synaptic Neuroscience, Italian Institute of Technology, 16163 Genova, Italy
| | - Adrian Sprenger
- Centre for Biological Signaling Studies (BIOSS), Spemann Graduate School of Biology and Medicine (SGBM), Centre of Chronic Immunodeficiency (CCI), Department of Dermatology, Center for Biological Systems Analysis (ZBSA), Institute of Molecular Medicine and Cell Research, Comprehensive Cancer Centre Freiburg, and Institute of Pathology, University Medical Center Freiburg, University of Freiburg, 79104 Freiburg, Germany Centre for Biological Signaling Studies (BIOSS), Spemann Graduate School of Biology and Medicine (SGBM), Centre of Chronic Immunodeficiency (CCI), Department of Dermatology, Center for Biological Systems Analysis (ZBSA), Institute of Molecular Medicine and Cell Research, Comprehensive Cancer Centre Freiburg, and Institute of Pathology, University Medical Center Freiburg, University of Freiburg, 79104 Freiburg, Germany Centre for Biological Signaling Studies (BIOSS), Spemann Graduate School of Biology and Medicine (SGBM), Centre of Chronic Immunodeficiency (CCI), Department of Dermatology, Center for Biological Systems Analysis (ZBSA), Institute of Molecular Medicine and Cell Research, Comprehensive Cancer Centre Freiburg, and Institute of Pathology, University Medical Center Freiburg, University of Freiburg, 79104 Freiburg, Germany
| | - Thomas Plum
- Department of Molecular Immunology, BioIII, Faculty of Biology, University of Freiburg and Max Planck Institute of Immunobiology and Epigenetics, 79104 Freiburg, Germany Centre for Biological Signaling Studies (BIOSS), Spemann Graduate School of Biology and Medicine (SGBM), Centre of Chronic Immunodeficiency (CCI), Department of Dermatology, Center for Biological Systems Analysis (ZBSA), Institute of Molecular Medicine and Cell Research, Comprehensive Cancer Centre Freiburg, and Institute of Pathology, University Medical Center Freiburg, University of Freiburg, 79104 Freiburg, Germany Centre for Biological Signaling Studies (BIOSS), Spemann Graduate School of Biology and Medicine (SGBM), Centre of Chronic Immunodeficiency (CCI), Department of Dermatology, Center for Biological Systems Analysis (ZBSA), Institute of Molecular Medicine and Cell Research, Comprehensive Cancer Centre Freiburg, and Institute of Pathology, University Medical Center Freiburg, University of Freiburg, 79104 Freiburg, Germany
| | - Maximilian Seidl
- Centre for Biological Signaling Studies (BIOSS), Spemann Graduate School of Biology and Medicine (SGBM), Centre of Chronic Immunodeficiency (CCI), Department of Dermatology, Center for Biological Systems Analysis (ZBSA), Institute of Molecular Medicine and Cell Research, Comprehensive Cancer Centre Freiburg, and Institute of Pathology, University Medical Center Freiburg, University of Freiburg, 79104 Freiburg, Germany Centre for Biological Signaling Studies (BIOSS), Spemann Graduate School of Biology and Medicine (SGBM), Centre of Chronic Immunodeficiency (CCI), Department of Dermatology, Center for Biological Systems Analysis (ZBSA), Institute of Molecular Medicine and Cell Research, Comprehensive Cancer Centre Freiburg, and Institute of Pathology, University Medical Center Freiburg, University of Freiburg, 79104 Freiburg, Germany
| | - Jörn Dengjel
- Centre for Biological Signaling Studies (BIOSS), Spemann Graduate School of Biology and Medicine (SGBM), Centre of Chronic Immunodeficiency (CCI), Department of Dermatology, Center for Biological Systems Analysis (ZBSA), Institute of Molecular Medicine and Cell Research, Comprehensive Cancer Centre Freiburg, and Institute of Pathology, University Medical Center Freiburg, University of Freiburg, 79104 Freiburg, Germany Centre for Biological Signaling Studies (BIOSS), Spemann Graduate School of Biology and Medicine (SGBM), Centre of Chronic Immunodeficiency (CCI), Department of Dermatology, Center for Biological Systems Analysis (ZBSA), Institute of Molecular Medicine and Cell Research, Comprehensive Cancer Centre Freiburg, and Institute of Pathology, University Medical Center Freiburg, University of Freiburg, 79104 Freiburg, Germany Centre for Biological Signaling Studies (BIOSS), Spemann Graduate School of Biology and Medicine (SGBM), Centre of Chronic Immunodeficiency (CCI), Department of Dermatology, Center for Biological Systems Analysis (ZBSA), Institute of Molecular Medicine and Cell Research, Comprehensive Cancer Centre Freiburg, and Institute of Pathology, University Medical Center Freiburg, University of Freiburg, 79104 Freiburg, Germany
| | - Michael Reth
- Department of Molecular Immunology, BioIII, Faculty of Biology, University of Freiburg and Max Planck Institute of Immunobiology and Epigenetics, 79104 Freiburg, Germany Centre for Biological Signaling Studies (BIOSS), Spemann Graduate School of Biology and Medicine (SGBM), Centre of Chronic Immunodeficiency (CCI), Department of Dermatology, Center for Biological Systems Analysis (ZBSA), Institute of Molecular Medicine and Cell Research, Comprehensive Cancer Centre Freiburg, and Institute of Pathology, University Medical Center Freiburg, University of Freiburg, 79104 Freiburg, Germany
| | - Fabrizia Cesca
- Center of Synaptic Neuroscience, Italian Institute of Technology, 16163 Genova, Italy
| | - Tilman Brummer
- Centre for Biological Signaling Studies (BIOSS), Spemann Graduate School of Biology and Medicine (SGBM), Centre of Chronic Immunodeficiency (CCI), Department of Dermatology, Center for Biological Systems Analysis (ZBSA), Institute of Molecular Medicine and Cell Research, Comprehensive Cancer Centre Freiburg, and Institute of Pathology, University Medical Center Freiburg, University of Freiburg, 79104 Freiburg, Germany Centre for Biological Signaling Studies (BIOSS), Spemann Graduate School of Biology and Medicine (SGBM), Centre of Chronic Immunodeficiency (CCI), Department of Dermatology, Center for Biological Systems Analysis (ZBSA), Institute of Molecular Medicine and Cell Research, Comprehensive Cancer Centre Freiburg, and Institute of Pathology, University Medical Center Freiburg, University of Freiburg, 79104 Freiburg, Germany Centre for Biological Signaling Studies (BIOSS), Spemann Graduate School of Biology and Medicine (SGBM), Centre of Chronic Immunodeficiency (CCI), Department of Dermatology, Center for Biological Systems Analysis (ZBSA), Institute of Molecular Medicine and Cell Research, Comprehensive Cancer Centre Freiburg, and Institute of Pathology, University Medical Center Freiburg, University of Freiburg, 79104 Freiburg, Germany
| | - Susana Minguet
- Department of Molecular Immunology, BioIII, Faculty of Biology, University of Freiburg and Max Planck Institute of Immunobiology and Epigenetics, 79104 Freiburg, Germany Centre for Biological Signaling Studies (BIOSS), Spemann Graduate School of Biology and Medicine (SGBM), Centre of Chronic Immunodeficiency (CCI), Department of Dermatology, Center for Biological Systems Analysis (ZBSA), Institute of Molecular Medicine and Cell Research, Comprehensive Cancer Centre Freiburg, and Institute of Pathology, University Medical Center Freiburg, University of Freiburg, 79104 Freiburg, Germany
| | - Wolfgang W A Schamel
- Department of Molecular Immunology, BioIII, Faculty of Biology, University of Freiburg and Max Planck Institute of Immunobiology and Epigenetics, 79104 Freiburg, Germany Centre for Biological Signaling Studies (BIOSS), Spemann Graduate School of Biology and Medicine (SGBM), Centre of Chronic Immunodeficiency (CCI), Department of Dermatology, Center for Biological Systems Analysis (ZBSA), Institute of Molecular Medicine and Cell Research, Comprehensive Cancer Centre Freiburg, and Institute of Pathology, University Medical Center Freiburg, University of Freiburg, 79104 Freiburg, Germany Centre for Biological Signaling Studies (BIOSS), Spemann Graduate School of Biology and Medicine (SGBM), Centre of Chronic Immunodeficiency (CCI), Department of Dermatology, Center for Biological Systems Analysis (ZBSA), Institute of Molecular Medicine and Cell Research, Comprehensive Cancer Centre Freiburg, and Institute of Pathology, University Medical Center Freiburg, University of Freiburg, 79104 Freiburg, Germany
| |
Collapse
|
206
|
Jacob CO, Yu N, Sindhava V, Cancro MP, Pawar RD, Putterman C, Stohl W. Differential Development of Systemic Lupus Erythematosus in NZM 2328 Mice Deficient in Discrete Pairs of BAFF Receptors. Arthritis Rheumatol 2015; 67:2523-35. [DOI: 10.1002/art.39210] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2014] [Accepted: 05/14/2015] [Indexed: 01/02/2023]
Affiliation(s)
- Chaim O. Jacob
- University of Southern California Keck School of Medicine; Los Angeles
| | - Ning Yu
- University of Southern California Keck School of Medicine; Los Angeles
| | | | | | | | | | - William Stohl
- University of Southern California Keck School of Medicine; Los Angeles
| |
Collapse
|
207
|
Sun Y, Sun L. CsBAFF, a Teleost B Cell Activating Factor, Promotes Pathogen-Induced Innate Immunity and Vaccine-Induced Adaptive Immunity. PLoS One 2015; 10:e0136015. [PMID: 26295165 PMCID: PMC4546598 DOI: 10.1371/journal.pone.0136015] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2015] [Accepted: 07/30/2015] [Indexed: 12/02/2022] Open
Abstract
B cell activating factor (BAFF) is a member of the tumor necrosis factor family that is known to play an important role in B cell activation, proliferation, and differentiation in mammals. However, studies of BAFF in teleosts are very limited and its function, in particular that under in vivo conditions, is essentially unknown. In this study, we conducted in vivo as well as in vitro functional analyses of a BAFF homologue (CsBAFF) from the teleost fish tongue sole (Cynoglossus semilaevis). CsBAFF is composed of 261 residues and shares moderate sequence identities with known BAFFs of other teleosts. CsBAFF expression was most abundant in immune organs and was upregulated during bacterial infection. Purified recombinant CsBAFF (rCsBAFF) bound to tongue sole lymphocytes and promoted cellular proliferation and survival. The results of an in vivo study showed that CsBAFF overexpression in tongue sole significantly enhanced macrophage activation and reduced bacterial infection in fish tissues, whereas knockdown of CsBAFF expression resulted in increased bacterial dissemination and colonization in fish tissues. Furthermore, vaccination studies showed that CsBAFF enhanced the immunoprotection of a DNA vaccine and augmented the production of specific serum antibodies. Taken together, these results provide the first in vivo evidence to indicate that teleost BAFF is an immunostimulator that significantly contributes to the innate antibacterial immune response and vaccine-induced adaptive immune response.
Collapse
Affiliation(s)
- Yun Sun
- Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, 266071, China
- State Key Laboratory Breeding Base for Sustainable Exploitation of Tropical Biotic Resources, College of Marine Science, Hainan University, Haikou, 570228, China
| | - Li Sun
- Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, 266071, China
- Function Laboratory for Marine Biology and Biotechnology, Qingdao National Oceanography Laboratory, Qingdao, China
- * E-mail:
| |
Collapse
|
208
|
Stohl W, Merrill JT, Looney RJ, Buyon J, Wallace DJ, Weisman MH, Ginzler EM, Cooke B, Holloway D, Kaliyaperumal A, Kuchimanchi KR, Cheah TC, Rasmussen E, Ferbas J, Belouski SS, Tsuji W, Zack DJ. Treatment of systemic lupus erythematosus patients with the BAFF antagonist "peptibody" blisibimod (AMG 623/A-623): results from randomized, double-blind phase 1a and phase 1b trials. Arthritis Res Ther 2015; 17:215. [PMID: 26290435 PMCID: PMC4545922 DOI: 10.1186/s13075-015-0741-z] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2015] [Accepted: 08/05/2015] [Indexed: 12/28/2022] Open
Abstract
INTRODUCTION Blisibimod is a potent B cell-activating factor (BAFF) antagonist that binds to both cell membrane-expressed and soluble BAFF. The goal of these first-in-human studies was to characterize the safety, tolerability, and pharmacokinetic and pharmacodynamic profiles of blisibimod in subjects with systemic lupus erythematosus (SLE). METHODS SLE subjects with mild disease that was stable/inactive at baseline received either a single dose of blisibimod (0.1, 0.3, 1, or 3 mg/kg subcutaneous [SC] or 1, 3, or 6 mg/kg intravenous [IV]) or placebo (phase 1a; N = 54), or four weekly doses of blisibimod (0.3, 1, or 3 mg/kg SC or 6 mg/kg IV) or placebo (phase 1b; N = 63). Safety and tolerability measures were collected, and B cell subset measurements and pharmacokinetic analyses were performed. RESULTS All subjects (93 % female; mean age 43.7 years) carried the diagnosis of SLE for ≥ 1 year. Single- and multiple-dose treatment with blisibimod produced a decrease in the number of naïve B cells (24-76 %) and a transient relative increase in the memory B cell compartment, with the greatest effect on IgD(-)CD27+; there were no notable changes in T cells or natural killer cells. With time, memory B cells reverted to baseline, leading to a calculated 30 % reduction in total B cells by approximately 160 days after the first dose. In both the single- and multiple-dosing SC cohorts, the pharmacokinetic profile indicated slow absorption, dose-proportional exposure from 0.3 through 3.0 mg/kg SC and 1 through 6 mg/kg IV, linear pharmacokinetics across the dose range of 1.0-6.0 mg/kg, and accumulation ratios ranging from 2.21 to 2.76. The relative increase in memory B cells was not associated with safety signals, and the incidence of adverse events, anti-blisibimod antibodies, and clinical laboratory abnormalities were comparable between blisibimod- and placebo-treated subjects. CONCLUSIONS Blisibimod changed the constituency of the B cell pool and single and multiple doses of blisibimod exhibited approximate dose-proportional pharmacokinetics across the dose range 1.0-6.0 mg/kg. The safety and tolerability profile of blisibimod in SLE was comparable with that of placebo. These findings support further studies of blisibimod in SLE and other B cell-mediated diseases. TRIAL REGISTRATION Clinicaltrials.gov NCT02443506 . Registered 11 May 2015. NCT02411136 Registered 7 April 2015.
Collapse
Affiliation(s)
- William Stohl
- Los Angeles County and University of Southern California Medical Center and University of Southern California Keck School of Medicine, 1975 Zonal Ave., Los Angeles, CA, 90033, USA.
- Division of Rheumatology, Los Angeles County + University of Southern California Medical Center and Keck School of Medicine of the University of Southern California, 2011 Zonal Ave., Los Angeles, CA, 90033, USA.
| | - Joan T Merrill
- Oklahoma Medical Research Foundation, 825 NE 13th St., Oklahoma City, OK, 73104, USA.
| | - R John Looney
- University of Rochester, 252 Elmwood Ave., Rochester, NY, 14627, USA.
| | - Jill Buyon
- Hospital for Joint Disease, 301 E 17th St., New York, NY, 10003, USA.
| | - Daniel J Wallace
- Cedars-Sinai Medical Center, 8700 Beverly Blvd., Los Angeles, CA, 90048, USA.
| | - Michael H Weisman
- Cedars-Sinai Medical Center, 8700 Beverly Blvd., Los Angeles, CA, 90048, USA.
| | - Ellen M Ginzler
- SUNY Downstate Medical Center, 450 Clarkson Ave., Brooklyn, NY, 11203, USA.
| | - Blaire Cooke
- Amgen Inc., 1 Amgen Center Dr., Thousand Oaks, CA, 91320, USA.
| | - Donna Holloway
- Formerly of Amgen, Inc., 1 Amgen Center Dr., Thousand Oaks, CA, 91320, USA.
| | | | | | | | - Erik Rasmussen
- Amgen Inc., 1 Amgen Center Dr., Thousand Oaks, CA, 91320, USA.
| | - John Ferbas
- Amgen Inc., 1 Amgen Center Dr., Thousand Oaks, CA, 91320, USA.
| | - Shelley S Belouski
- Formerly of Amgen, Inc., 1 Amgen Center Dr., Thousand Oaks, CA, 91320, USA.
| | - Wayne Tsuji
- Amgen Inc., 1 Amgen Center Dr., Thousand Oaks, CA, 91320, USA.
| | - Debra J Zack
- Formerly of Amgen, Inc., 1 Amgen Center Dr., Thousand Oaks, CA, 91320, USA.
| |
Collapse
|
209
|
De Paepe B, Zschüntzsch J. Scanning for Therapeutic Targets within the Cytokine Network of Idiopathic Inflammatory Myopathies. Int J Mol Sci 2015; 16:18683-713. [PMID: 26270565 PMCID: PMC4581266 DOI: 10.3390/ijms160818683] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2015] [Revised: 07/13/2015] [Accepted: 07/15/2015] [Indexed: 12/17/2022] Open
Abstract
The idiopathic inflammatory myopathies (IIM) constitute a heterogeneous group of chronic disorders that include dermatomyositis (DM), polymyositis (PM), sporadic inclusion body myositis (IBM) and necrotizing autoimmune myopathy (NAM). They represent distinct pathological entities that, most often, share predominant inflammation in muscle tissue. Many of the immunopathogenic processes behind the IIM remain poorly understood, but the crucial role of cytokines as essential regulators of the intramuscular build-up of inflammation is undisputed. This review describes the extensive cytokine network within IIM muscle, characterized by strong expression of Tumor Necrosis Factors (TNFα, LTβ, BAFF), Interferons (IFNα/β/γ), Interleukins (IL-1/6/12/15/18/23) and Chemokines (CXCL9/10/11/13, CCL2/3/4/8/19/21). Current therapeutic strategies and the exploration of potential disease modifying agents based on manipulation of the cytokine network are provided. Reported responses to anti-TNFα treatment in IIM are conflicting and new onset DM/PM has been described after administration of anti-TNFα agents to treat other diseases, pointing to the complex effects of TNFα neutralization. Treatment with anti-IFNα has been shown to suppress the IFN type 1 gene signature in DM/PM patients and improve muscle strength. Beneficial effects of anti-IL-1 and anti-IL-6 therapy have also been reported. Cytokine profiling in IIM aids the development of therapeutic strategies and provides approaches to subtype patients for treatment outcome prediction.
Collapse
Affiliation(s)
- Boel De Paepe
- Neuromuscular Reference Center, Laboratory for Neuropathology, 10K12E, Ghent University Hospital, 9000 Ghent, Belgium.
| | - Jana Zschüntzsch
- Department of Neurology, University Medical Centre, Göttingen University, 37075 Göttingen, Germany.
| |
Collapse
|
210
|
Jackson DA, Smith TD, Amarsaikhan N, Han W, Neil MS, Boi SK, Vrabel AM, Tolosa EJ, Almada LL, Fernandez-Zapico ME, Elsawa SF. Modulation of the IL-6 Receptor α Underlies GLI2-Mediated Regulation of Ig Secretion in Waldenström Macroglobulinemia Cells. THE JOURNAL OF IMMUNOLOGY 2015; 195:2908-16. [PMID: 26238488 DOI: 10.4049/jimmunol.1402974] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/26/2014] [Accepted: 07/14/2015] [Indexed: 12/15/2022]
Abstract
Ig secretion by terminally differentiated B cells is an important component of the immune response to foreign pathogens. Its overproduction is a defining characteristic of several B cell malignancies, including Waldenström macroglobulinemia (WM), where elevated IgM is associated with significant morbidity and poor prognosis. Therefore, the identification and characterization of the mechanisms controlling Ig secretion are of great importance for the development of future therapeutic approaches for this disease. In this study, we define a novel pathway involving the oncogenic transcription factor GLI2 modulating IgM secretion by WM malignant cells. Pharmacological and genetic inhibition of GLI2 in WM malignant cells resulted in a reduction in IgM secretion. Screening for a mechanism identified the IL-6Rα (gp80) subunit as a downstream target of GLI2 mediating the regulation of IgM secretion. Using a combination of expression, luciferase, and chromatin immunoprecipitation assays we demonstrate that GLI2 binds to the IL-6Rα promoter and regulates its activity as well as the expression of this receptor. Additionally, we were able to rescue the reduction in IgM secretion in the GLI2 knockdown group by overexpressing IL-6Rα, thus defining the functional significance of this receptor in GLI2-mediated regulation of IgM secretion. Interestingly, this occurred independent of Hedgehog signaling, a known regulator of GLI2, as manipulation of Hedgehog had no effect on IgM secretion. Given the poor prognosis associated with elevated IgM in WM patients, components of this new signaling axis could be important therapeutic targets.
Collapse
Affiliation(s)
- David A Jackson
- Department of Biological Sciences, Northern Illinois University, DeKalb, IL 60115; and
| | - Timothy D Smith
- Department of Biological Sciences, Northern Illinois University, DeKalb, IL 60115; and
| | - Nansalmaa Amarsaikhan
- Department of Biological Sciences, Northern Illinois University, DeKalb, IL 60115; and
| | - Weiguo Han
- Department of Biological Sciences, Northern Illinois University, DeKalb, IL 60115; and
| | - Matthew S Neil
- Department of Biological Sciences, Northern Illinois University, DeKalb, IL 60115; and
| | - Shannon K Boi
- Department of Biological Sciences, Northern Illinois University, DeKalb, IL 60115; and
| | - Anne M Vrabel
- Schulze Center for Novel Therapeutics, Mayo Clinic, Rochester, MN 55905
| | - Ezequiel J Tolosa
- Schulze Center for Novel Therapeutics, Mayo Clinic, Rochester, MN 55905
| | - Luciana L Almada
- Schulze Center for Novel Therapeutics, Mayo Clinic, Rochester, MN 55905
| | | | - Sherine F Elsawa
- Department of Biological Sciences, Northern Illinois University, DeKalb, IL 60115; and
| |
Collapse
|
211
|
Blockade of B-cell activating factor with TACI-IgG effectively reduced Th1 and Th17 cells but not memory T cells in experimental allergic encephalomyelitis mice. Cent Eur J Immunol 2015; 40:142-8. [PMID: 26557026 PMCID: PMC4637387 DOI: 10.5114/ceji.2015.52826] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2015] [Accepted: 04/09/2015] [Indexed: 12/15/2022] Open
Abstract
B-cell activating factor (BAFF) is regarded as a new therapeutic target in autoimmune diseases such as systemic lupus erythematosus (SLE) and multiple sclerosis (MS). Along with other researchers, we have demonstrated that BAFF inhibitor atacicept (TACI-IgG) suppresses lupus and experimental allergic encephalomyelitis (EAE) by reducing the mature B-cell number but not memory B cells. It is however unclear whether TACI-Ig affects pathogenic T cells and memory T cells. In the present study, we found that blocking BAFF with TACI-IgG effectively reduces the pathogenic Th1 and Th17 cells in EAE mice. However, TACI-IgG did not reduce memory CD62L+CD44hiCD4+ and CD62L+CD44hiCD8+ T cells in EAE mice. When interleukin (IL)-15 was neutralized, memory CD62L+CD44hi T cells were significantly reduced in TACI-IgG-treated EAE mice. These results suggest that TACI-IgG is effective in effective controlling Th1 and Th17 cells, but it also increases IL-15 to upregulate memory T cells in EAE mice. The study provides hints for the clinical application of the combination of BAFF- and IL-15-specific therapeutic agents.
Collapse
|
212
|
Morais SA, Vilas-Boas A, Isenberg DA. B-cell survival factors in autoimmune rheumatic disorders. Ther Adv Musculoskelet Dis 2015; 7:122-51. [PMID: 26288664 PMCID: PMC4530383 DOI: 10.1177/1759720x15586782] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Autoimmune rheumatic disorders have complex etiopathogenetic mechanisms in which B cells play a central role. The importance of factors stimulating B cells, notably the B-cell activating factor (BAFF) and A proliferation inducing ligand (APRIL) axis is now recognized. BAFF and APRIL are cytokines essential for B-cell proliferation and survival from the immature stages to the development of plasma cells. Their levels are increased in some subsets of patients with autoimmune disorders. Several recent biologic drugs have been developed to block this axis, namely belimumab [already licensed for systemic lupus erythematosus (SLE) treatment], tabalumab, atacicept and blisibimod. Many clinical trials to evaluate the safety and efficacy of these drugs in several autoimmune disorders are ongoing, or have been completed recently. This review updates the information on the use of biologic agents blocking BAFF/APRIL for patients with SLE, rheumatoid arthritis, Sjögren's syndrome and myositis.
Collapse
Affiliation(s)
- Sandra A Morais
- Internal Medicine Department, Hospital Pedro Hispano, Matosinhos, Portugal
| | - Andreia Vilas-Boas
- Internal Medicine Department, Hospital Pedro Hispano, Matosinhos, Portugal
| | - David A Isenberg
- Centre for Rheumatology, University College London, Room 424, 4th Floor Rayne Building, 5 University Street, London WC1E 6JF, UK
| |
Collapse
|
213
|
Sonar S, Lal G. Role of Tumor Necrosis Factor Superfamily in Neuroinflammation and Autoimmunity. Front Immunol 2015; 6:364. [PMID: 26257732 PMCID: PMC4507150 DOI: 10.3389/fimmu.2015.00364] [Citation(s) in RCA: 76] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2015] [Accepted: 07/05/2015] [Indexed: 12/18/2022] Open
Abstract
Tumor necrosis factor superfamily (TNFSF) molecules play an important role in the activation, proliferation, differentiation, and migration of immune cells into the central nervous system (CNS). Several TNF superfamily molecules are known to control alloimmunity, autoimmunity, and immunity. Development of transgenic and gene knockout animals, and monoclonal antibodies against TNFSF molecules have increased our understanding of individual receptor-ligand interactions, and their intracellular signaling during homeostasis and neuroinflammation. A strong clinical association has been observed between TNFSF members and CNS autoimmunity such as multiple sclerosis and also in its animal model experimental autoimmune encephalomyelitis. Therefore, they are promising targets for alternative therapeutic options to control autoimmunity. Although, TNFSF ligands are widely distributed and have diverse functions, we have restricted the discussions in this review to TNFSF receptor-ligand interactions and their role in the pathogenesis of neuroinflammation and CNS autoimmunity.
Collapse
|
214
|
Zollars E, Bienkowska J, Czerkowicz J, Allaire N, Ranger AM, Magder L, Petri M. BAFF (B cell activating factor) transcript level in peripheral blood of patients with SLE is associated with same-day disease activity as well as global activity over the next year. Lupus Sci Med 2015; 2:e000063. [PMID: 26113988 PMCID: PMC4477150 DOI: 10.1136/lupus-2014-000063] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2014] [Revised: 01/09/2015] [Accepted: 02/08/2015] [Indexed: 12/13/2022]
Abstract
Objectives Quantitating gene expression is a potential method of developing biomarkers in systemic lupus erythematosus (SLE). Because of the known pathological role of B cell activating factor (BAFF) in SLE, we explored the association between BAFF gene expression and clinical activity in SLE. Methods A total of 275 patients with SLE completed this phase of a prospective observational study. At entry into the study, the BAFF gene expression levels were determined in peripheral blood RNA. Serum concentration of BAFF protein was also measured. We then determined clinical associations with SLE disease history, SLE activity on the same day and SLE activity over the course of the next year. Results Elevated BAFF gene expression was associated with a history of more leucopenia and serologically with more autoantibodies (anti-dsDNA, anti-Sm, anti-Ro, anti-La and anti-RNP) and low complement. Patients with higher amounts of BAFF transcript had higher measured levels of clinical disease activity. Initial high levels of BAFF gene expression also predicted increased disease activity over the course of the next year. In contrast, serum concentration of BAFF protein was not strongly associated with same-day global disease activity or with future disease activity. Conclusions BAFF gene expression level is associated with clinical and serological SLE activity on the same day and predictive of clinical activity over the next year. BAFF gene expression is a better measure and predictor of SLE disease activity than the serum BAFF protein level.
Collapse
Affiliation(s)
- Eric Zollars
- Division of Rheumatology , Johns Hopkins University School of Medicine , Baltimore, Maryland , USA
| | | | | | | | | | - Laurence Magder
- Epidemiology and Public Health, University of Maryland , Baltimore, Maryland , USA
| | - Michelle Petri
- Division of Rheumatology , Johns Hopkins University School of Medicine , Baltimore, Maryland , USA
| |
Collapse
|
215
|
Updating the clinical evidence on belimumab role modulating B-cell response and treatment of systemic lupus erythematosus. ACTA ACUST UNITED AC 2015. [DOI: 10.4155/cli.15.16] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
|
216
|
Emerah AA, Mohamed KF, Elbadawy NE, Rashad MH. Effects of interleukin-10 gene polymorphism on clinical diversity and activity of systemic lupus erythematosus. EGYPTIAN RHEUMATOLOGY AND REHABILITATION 2015. [DOI: 10.4103/1110-161x.157855] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
|
217
|
Li DH, Yang Q, Zhou JS, Zhang ZW, Miao MY, Yang SS, Xu WD. Regulatory role of B-cell maturation antigen on the toxic effect of chromium ions on human SaOS-2 osteoblasts. Biotechnol Appl Biochem 2015; 64:638-646. [PMID: 26011700 DOI: 10.1002/bab.1401] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2014] [Accepted: 05/21/2015] [Indexed: 01/07/2023]
Abstract
Metal prostheses of artificial joints undergo wear, producing numerous metal particles and ions, such as Cr3+ . Cr3+ is considered a key factor leading to aseptic loosening. Many studies focus on the effect of Cr3+ on osteoblasts; however, little is known about the effect of Cr3+ on the B-cell maturation antigen (BCMA) in the osteoblasts. In this study, we first demonstrated the BCMA expressed in human SaOS-2 osteoblasts through reverse transcriptase-PCR, Western blot, and immunocytochemical analyses. Cr3+ decreased alkaline phosphatase (ALP), osteocalcin (OC), cell mineralization, and collagen type I mRNA and protein expression. Moreover, Cr3+ has an inhibitive effect on the expression of the BCMA in human SaOS-2 osteoblasts. However, after we upregulated the expression of the BCMA, ALP, OC, cell mineralization, and collagen type I mRNA and protein expression were increased. Overall, this study demonstrates that the BCMA is involved in human SaOS-2 osteoblast osteogenetic metabolism and plays a regulatory role on the toxic effect of chromium ions on human SaOS-2 osteoblasts.
Collapse
Affiliation(s)
- D H Li
- Department of Orthopedics, Changhai Hospital Affiliated to the Second Military Medical University, Yangpu, Shanghai, People's Republic of China
| | - Q Yang
- Department of Orthopedics, Changhai Hospital Affiliated to the Second Military Medical University, Yangpu, Shanghai, People's Republic of China
| | - J S Zhou
- Department of Biochemistry and Molecular Biology, Second Military Medical University, Yangpu, Shanghai, People's Republic of China
| | - Z W Zhang
- Department of Biochemistry and Molecular Biology, Second Military Medical University, Yangpu, Shanghai, People's Republic of China
| | - M Y Miao
- Department of Biochemistry and Molecular Biology, Second Military Medical University, Yangpu, Shanghai, People's Republic of China
| | - S S Yang
- Department of Biochemistry and Molecular Biology, Second Military Medical University, Yangpu, Shanghai, People's Republic of China
| | - W D Xu
- Department of Orthopedics, Changhai Hospital Affiliated to the Second Military Medical University, Yangpu, Shanghai, People's Republic of China
| |
Collapse
|
218
|
Coquery CM, Loo WM, Wade NS, Bederman AG, Tung KS, Lewis JE, Hess H, Erickson LD. BAFF regulates follicular helper t cells and affects their accumulation and interferon-γ production in autoimmunity. Arthritis Rheumatol 2015; 67:773-84. [PMID: 25385309 DOI: 10.1002/art.38950] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2014] [Accepted: 11/04/2014] [Indexed: 01/06/2023]
Abstract
OBJECTIVE Follicular helper T (Tfh) cells are critical for the development of protective antibodies via germinal center (GC) B cell responses; however, uncontrolled Tfh cell expansion activates autoreactive B cells to produce antibodies that cause autoimmunity. The mechanisms that control Tfh cell homeostasis remain largely unknown. The aim of this study was to determine the contribution of BAFF to Tfh cell responses in autoimmunity. METHODS We analyzed the properties of Tfh cells in lupus-prone mice sufficient or deficient in BCMA. Adoptive transfer studies and mixed bone marrow chimeras were used to test BCMA signaling in T cells. We assessed BAFF stimulation of Tfh cells through in vitro cell cocultures and in vivo depletion studies using flow cytometry. RESULTS In Nba2 mice, Tfh cells expressed the BAFF receptors BCMA and B lymphocyte stimulator receptor 3 (BR-3) and accumulated in the spleen when BCMA was absent. BCMA deficiency in T cells promoted the expansion of Tfh cells, GC formation, autoantibody production, and interferon-γ (IFNγ) production by Tfh cells through BR-3. IFNγ-producing Tfh cells increased BAFF expression in dendritic cells. Blocking BAFF or IFNγ in vivo reduced Tfh cell accumulation and reduced autoimmunity in BCMA-deficient animals. Moreover, circulating Tfh-like cells that expressed BR-3 (but not BCMA) were elevated in patients with systemic lupus erythematosus, and this correlated with serum BAFF and IFNγ levels. CONCLUSION In Nba2 mice, BCMA negatively regulates Tfh cell expansion, while BAFF signaling through BR-3 promotes Tfh cell accumulation. Our findings suggest that the balance between BCMA and BR-3 signaling in Tfh cells serves as a checkpoint of immune tolerance.
Collapse
|
219
|
Lin W, Seshasayee D, Lee WP, Caplazi P, McVay S, Suto E, Nguyen A, Lin Z, Sun Y, DeForge L, Balazs M, Martin F, Zarrin AA. Dual B cell immunotherapy is superior to individual anti-CD20 depletion or BAFF blockade in murine models of spontaneous or accelerated lupus. Arthritis Rheumatol 2015; 67:215-24. [PMID: 25303150 PMCID: PMC4312898 DOI: 10.1002/art.38907] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2014] [Accepted: 10/02/2014] [Indexed: 12/18/2022]
Abstract
Objective To determine whether a combination of B cell depletion and BAFF blockade is more effective than monotherapy in treating models of spontaneous or accelerated systemic lupus erythematosus (SLE) in (NZB × NZW)F1 mice. Methods Clinical parameters such as disease progression–free survival, proteinuria, and renal injury were assessed in models of spontaneous, interferon-α (IFNα)–accelerated, or pristane-accelerated lupus in (NZB × NZW)F1 mice. Treatment arms included anti-CD20 (B cell depletion), B lymphocyte stimulator receptor 3 fusion protein (BR-3-Fc) (BAFF blockade), the combination of anti-CD20 and BR-3-Fc, isotype control, or cyclophosphamide. In models of spontaneous, IFNα-accelerated, or pristane-accelerated lupus, mice were treated for 24 weeks, 8 weeks, or 12 weeks, respectively. Peripheral and resident B cell subsets and various autoantibodies were examined. Results Compared to B cell depletion or BAFF blockade alone, combined therapy significantly improved disease manifestations in all 3 lupus models. In addition, marginal zone B cells, plasmablasts, and circulating and tissue plasma cells were decreased more effectively. Dual B cell immunotherapy also reduced multiple classes of pathogenic autoantibodies, consistent with its observed effectiveness in reducing immune complex–mediated renal injury. Conclusion Dual immunotherapy via B cell depletion and BAFF blockade is more efficacious than single agent immunotherapy in murine SLE models, and this combination treatment is predicted to be an effective strategy for immunotherapy in human SLE.
Collapse
Affiliation(s)
- WeiYu Lin
- Genentech, Inc., South San Francisco, California
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
220
|
Biswas G, Kinoshita S, Kono T, Hikima JI, Sakai M. Evolutionary evidence of tumor necrosis factor super family members in the Japanese pufferfish (Takifugu rubripes): Comprehensive genomic identification and expression analysis. Mar Genomics 2015; 22:25-36. [PMID: 25792259 DOI: 10.1016/j.margen.2015.03.003] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2015] [Revised: 03/05/2015] [Accepted: 03/05/2015] [Indexed: 12/12/2022]
Abstract
Tumor necrosis factor (TNF) and its superfamily (TNFSF) members are important inflammatory cytokines. Although fish have fourteen TNFSF genes, their genomic location and existence are yet to be described and confirmed in the Japanese pufferfish (Fugu) (Takifugu rubripes). Therefore, we conducted in silico identification, synteny analysis of TNFSF genes from Fugu with that of zebrafish and human TNFSF loci and their expression analysis in various tissues. We identified ten novel TNFSF genes, viz. TNFSF5 (CD40L), TNFSF6 (FasL), three TNFSF10 (TRAIL) (-1, 2 and 3), TNFSF11 (RANKlg), TNFSF12 (TWEAK), two TNFSF13B (BAFF) (1 and 2) and TNFSF14 (LIGHT) belonging to seven TNFSFs in Fugu. Several features such as existence of TNF family signature, conservation of genes in TNF loci with human and zebrafish chromosomes and phylogenetic clustering with other teleost TNFSF orthologs confirmed their identity. Fugu TNFSF genes were constitutively expressed in all eight different tissues with most of them expressed highly in liver. Fugu TNFSF10 gene has three homologs present on chromosomes 10 (TNFSF10-1), 8 (TNFSF10-2) and 2 (TNFSF10-3). Moreover, a phylogenetic analysis containing all available vertebrate (mammals, birds, reptiles, amphibians and fish) TNFSF10 orthologs showed that Fugu TNFSF10-1 and 10-3 are present in all vertebrates, whereas TNFSF10-2 was not related to any mammalian and avian orthologs. Viral double-stranded RNA mimic poly (I:C) caused an elevated expression of three Fugu TNFSF10 genes in head kidney cells at 4h indicating probable role of these genes to induce apoptosis in virus-infected cells. In conclusion, Fugu possesses genes belonging to nine TNFSFs including the newly identified seven and previously reported two, TNFSF New (TNF-N) and TNFSF2 (TNF-α). Our findings would add up information to TNFSF evolution among vertebrates.
Collapse
Affiliation(s)
- Gouranga Biswas
- Interdisciplinary Graduate School of Agriculture and Engineering, University of Miyazaki, 1-1 Gakuenkibanadai-nishi, Miyazaki 889-2192, Japan
| | - Shunsuke Kinoshita
- Department of Biochemistry and Applied Biosciences, Faculty of Agriculture, University of Miyazaki, 1-1 Gakuenkibanadai-nishi, Miyazaki 889-2192, Japan
| | - Tomoya Kono
- Department of Biochemistry and Applied Biosciences, Faculty of Agriculture, University of Miyazaki, 1-1 Gakuenkibanadai-nishi, Miyazaki 889-2192, Japan
| | - Jun-ichi Hikima
- Department of Biochemistry and Applied Biosciences, Faculty of Agriculture, University of Miyazaki, 1-1 Gakuenkibanadai-nishi, Miyazaki 889-2192, Japan
| | - Masahiro Sakai
- Department of Biochemistry and Applied Biosciences, Faculty of Agriculture, University of Miyazaki, 1-1 Gakuenkibanadai-nishi, Miyazaki 889-2192, Japan.
| |
Collapse
|
221
|
Effect of TACI signaling on humoral immunity and autoimmune diseases. J Immunol Res 2015; 2015:247426. [PMID: 25866827 PMCID: PMC4381970 DOI: 10.1155/2015/247426] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2015] [Accepted: 03/04/2015] [Indexed: 02/02/2023] Open
Abstract
Transmembrane activator and calcium-modulating cyclophilin ligand interactor (TACI) is one of the receptors of B cell activating factor of the tumor necrosis factor family (BAFF) and a proliferation-inducing ligand (APRIL). TACI is a regulator in the immune responses. TACI inhibits B cell expansion and promotes the differentiation and survival of plasma cells. The mechanisms underlying these effects probably involve changed expressions of some crucial molecules, such as B lymphocyte induced maturation protein-1 (Blimp-1) and inducible T-cell costimulator ligand (ICOSL) in B cells and/or plasma cells. However, abnormal TACI signaling may relate to autoimmune disorders. Common variable immune deficiency (CVID) patients with heterozygous mutations in TACI alleles increase susceptibility to autoimmune diseases. Taci−/− mice and BAFF transgenic mice both develop signs of human SLE. These findings that indicate inappropriate levels of TACI signaling may disrupt immune system balance, thereby promoting the development of autoimmune diseases. In this review, we summarize the basic characteristics of the TACI ligands BAFF and APRIL, and detail the research findings on the role of TACI in humoral immunity. We also discuss the possible mechanisms underlying the susceptibility of CVID patients with TACI mutations to autoimmune diseases and the role of TACI in the pathogenesis of SLE.
Collapse
|
222
|
Cogollo E, Cogollo E, Silva MA, Isenberg D. Profile of atacicept and its potential in the treatment of systemic lupus erythematosus. DRUG DESIGN DEVELOPMENT AND THERAPY 2015; 9:1331-9. [PMID: 25834391 PMCID: PMC4357613 DOI: 10.2147/dddt.s71276] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
The importance of B cell activating factors in the generation of autoantibodies in patients with systemic lupus erythematosus (SLE) is now recognized. The two key factors, known as BAFF and APRIL, produced by a variety of cells including monocytes, dendritic cells and T cells, also help to regulate B cell maturation, function and survival. Biologic agents that block these factors have now been developed and tried out in large scale clinical trials in SLE patients. Benlysta which blocks BAFF has met some of its end points in clinical trials and is approved for use in patients with skin and joint disease who have failed conventional drugs. In contrast, clinical trials using atacicept which blocks both BAFF and APRIL have been more challenging to interpret. An early study in lupus nephritis was, mistakenly, abandoned due to serious infections thought to be linked to the biologic when in fact the dramatic fall in the immunoglobulin levels took place when the patients were given mycophenolate, prior to the introduction of the atacicept. Likewise the higher dose arm (150 mgm) of a flare prevention study was terminated prematurely when 2 deaths occurred. However, the mortality rate in this study was identical to that seen in the Benlysta studies and a post hoc analysis found a highly significant benefit for the 150mgm arm compared to the lower dose (75 mgm) and placebo arms. Other trials with both Benlysta and atacicept are on-going.
Collapse
Affiliation(s)
| | - Estafania Cogollo
- Department of Internal Medicine, Hospital Principe de Asturias, Alcala de Henares, Madrid, Spain
| | - Marta Amaral Silva
- Department of Internal Medicine, Hospital Distrital da Figueira da Foz, Coimbra, Portugal
| | - David Isenberg
- Centre for Rheumatology, Department of Medicine, University College London, London, UK
| |
Collapse
|
223
|
Weldon AJ, Moldovan I, Cabling MG, Hernandez EA, Hsu S, Gonzalez J, Parra A, Benitez A, Daoud N, Colburn K, Payne KJ. Surface APRIL Is Elevated on Myeloid Cells and Is Associated with Disease Activity in Patients with Rheumatoid Arthritis. J Rheumatol 2015; 42:749-59. [PMID: 25729037 DOI: 10.3899/jrheum.140630] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/14/2015] [Indexed: 12/25/2022]
Abstract
OBJECTIVE To assess surface APRIL (a proliferation-inducing ligand; CD256) expression by circulating myeloid cells in rheumatoid arthritis (RA) and to determine its relationship to disease activity. METHODS Peripheral blood mononuclear cells (PBMC) and plasma were obtained from patients with RA and healthy donors. PBMC were stained for flow cytometry to detect surface APRIL and blood cell markers to identify circulating myeloid cell subsets. Based on CD14 and CD16 phenotypes, monocyte subsets described as classical (CD14+CD16-), intermediate (CD14+CD16+), and nonclassical (CD14loCD16+) were identified. Levels of surface APRIL expression were measured by flow cytometry and median fluorescence intensity was used for comparisons. Levels of soluble APRIL in the plasma were determined by ELISA. Disease activity was measured by the Disease Activity Score in 28 joints. RESULTS In patients with RA, total myeloid cells showed expression of surface APRIL that correlated with disease activity and with plasma APRIL levels observed in these patients. In healthy donors, classical monocytes were composed of > 80% of circulating monocytes. However, in patients with RA, the intermediate and nonclassical subsets were elevated and made up the majority of circulating monocytes. In contrast to healthy donors, where high levels of surface APRIL were only observed in nonclassical monocytes, patients with RA showed high levels of surface APRIL expression by all circulating monocyte subsets. CONCLUSION Surface APRIL is elevated in circulating myeloid cells in patients with RA where it is highly correlated with disease activity. Patients with RA also showed skewing of monocytes toward subsets associated with secretion of tumor necrosis factor-α and/or interleukin 1β.
Collapse
Affiliation(s)
- Abby Jones Weldon
- From the Center for Health Disparities and Molecular Medicine, Department of Microbiology and Molecular Genetics, Department of Medicine, Department of Pharmaceutical and Administrative Sciences, School of Pharmacy, Department of Pathology and Human Anatomy, Loma Linda University, Loma Linda; Division of Rheumatology, Beaver Medical Group, Redlands, California, USA.A.J. Weldon, MS; A. Benitez, PhD, Center for Health Disparities and Molecular Medicine, and Department of Microbiology and Molecular Genetics, Loma Linda University; I. Moldovan, MD, Department of Medicine, Loma Linda University, and Division of Rheumatology, Beaver Medical Group; M.G. Cabling, MD; S. Hsu, MD; N. Daoud, MD; K. Colburn, MD, Department of Medicine, Loma Linda University; E.A. Hernandez, PhD, Department of Pharmaceutical and Administrative Sciences, School of Pharmacy, Loma Linda University; J. Gonzalez, BS; A. Parra, BS, Center for Health Disparities and Molecular Medicine, Loma Linda University; K.J. Payne, PhD, Center for Health Disparities and Molecular Medicine, and Department of Pathology and Human Anatomy, Loma Linda University.
| | - Ioana Moldovan
- From the Center for Health Disparities and Molecular Medicine, Department of Microbiology and Molecular Genetics, Department of Medicine, Department of Pharmaceutical and Administrative Sciences, School of Pharmacy, Department of Pathology and Human Anatomy, Loma Linda University, Loma Linda; Division of Rheumatology, Beaver Medical Group, Redlands, California, USA.A.J. Weldon, MS; A. Benitez, PhD, Center for Health Disparities and Molecular Medicine, and Department of Microbiology and Molecular Genetics, Loma Linda University; I. Moldovan, MD, Department of Medicine, Loma Linda University, and Division of Rheumatology, Beaver Medical Group; M.G. Cabling, MD; S. Hsu, MD; N. Daoud, MD; K. Colburn, MD, Department of Medicine, Loma Linda University; E.A. Hernandez, PhD, Department of Pharmaceutical and Administrative Sciences, School of Pharmacy, Loma Linda University; J. Gonzalez, BS; A. Parra, BS, Center for Health Disparities and Molecular Medicine, Loma Linda University; K.J. Payne, PhD, Center for Health Disparities and Molecular Medicine, and Department of Pathology and Human Anatomy, Loma Linda University
| | - Marven G Cabling
- From the Center for Health Disparities and Molecular Medicine, Department of Microbiology and Molecular Genetics, Department of Medicine, Department of Pharmaceutical and Administrative Sciences, School of Pharmacy, Department of Pathology and Human Anatomy, Loma Linda University, Loma Linda; Division of Rheumatology, Beaver Medical Group, Redlands, California, USA.A.J. Weldon, MS; A. Benitez, PhD, Center for Health Disparities and Molecular Medicine, and Department of Microbiology and Molecular Genetics, Loma Linda University; I. Moldovan, MD, Department of Medicine, Loma Linda University, and Division of Rheumatology, Beaver Medical Group; M.G. Cabling, MD; S. Hsu, MD; N. Daoud, MD; K. Colburn, MD, Department of Medicine, Loma Linda University; E.A. Hernandez, PhD, Department of Pharmaceutical and Administrative Sciences, School of Pharmacy, Loma Linda University; J. Gonzalez, BS; A. Parra, BS, Center for Health Disparities and Molecular Medicine, Loma Linda University; K.J. Payne, PhD, Center for Health Disparities and Molecular Medicine, and Department of Pathology and Human Anatomy, Loma Linda University
| | - Elvin A Hernandez
- From the Center for Health Disparities and Molecular Medicine, Department of Microbiology and Molecular Genetics, Department of Medicine, Department of Pharmaceutical and Administrative Sciences, School of Pharmacy, Department of Pathology and Human Anatomy, Loma Linda University, Loma Linda; Division of Rheumatology, Beaver Medical Group, Redlands, California, USA.A.J. Weldon, MS; A. Benitez, PhD, Center for Health Disparities and Molecular Medicine, and Department of Microbiology and Molecular Genetics, Loma Linda University; I. Moldovan, MD, Department of Medicine, Loma Linda University, and Division of Rheumatology, Beaver Medical Group; M.G. Cabling, MD; S. Hsu, MD; N. Daoud, MD; K. Colburn, MD, Department of Medicine, Loma Linda University; E.A. Hernandez, PhD, Department of Pharmaceutical and Administrative Sciences, School of Pharmacy, Loma Linda University; J. Gonzalez, BS; A. Parra, BS, Center for Health Disparities and Molecular Medicine, Loma Linda University; K.J. Payne, PhD, Center for Health Disparities and Molecular Medicine, and Department of Pathology and Human Anatomy, Loma Linda University
| | - Sheri Hsu
- From the Center for Health Disparities and Molecular Medicine, Department of Microbiology and Molecular Genetics, Department of Medicine, Department of Pharmaceutical and Administrative Sciences, School of Pharmacy, Department of Pathology and Human Anatomy, Loma Linda University, Loma Linda; Division of Rheumatology, Beaver Medical Group, Redlands, California, USA.A.J. Weldon, MS; A. Benitez, PhD, Center for Health Disparities and Molecular Medicine, and Department of Microbiology and Molecular Genetics, Loma Linda University; I. Moldovan, MD, Department of Medicine, Loma Linda University, and Division of Rheumatology, Beaver Medical Group; M.G. Cabling, MD; S. Hsu, MD; N. Daoud, MD; K. Colburn, MD, Department of Medicine, Loma Linda University; E.A. Hernandez, PhD, Department of Pharmaceutical and Administrative Sciences, School of Pharmacy, Loma Linda University; J. Gonzalez, BS; A. Parra, BS, Center for Health Disparities and Molecular Medicine, Loma Linda University; K.J. Payne, PhD, Center for Health Disparities and Molecular Medicine, and Department of Pathology and Human Anatomy, Loma Linda University
| | - Jennifer Gonzalez
- From the Center for Health Disparities and Molecular Medicine, Department of Microbiology and Molecular Genetics, Department of Medicine, Department of Pharmaceutical and Administrative Sciences, School of Pharmacy, Department of Pathology and Human Anatomy, Loma Linda University, Loma Linda; Division of Rheumatology, Beaver Medical Group, Redlands, California, USA.A.J. Weldon, MS; A. Benitez, PhD, Center for Health Disparities and Molecular Medicine, and Department of Microbiology and Molecular Genetics, Loma Linda University; I. Moldovan, MD, Department of Medicine, Loma Linda University, and Division of Rheumatology, Beaver Medical Group; M.G. Cabling, MD; S. Hsu, MD; N. Daoud, MD; K. Colburn, MD, Department of Medicine, Loma Linda University; E.A. Hernandez, PhD, Department of Pharmaceutical and Administrative Sciences, School of Pharmacy, Loma Linda University; J. Gonzalez, BS; A. Parra, BS, Center for Health Disparities and Molecular Medicine, Loma Linda University; K.J. Payne, PhD, Center for Health Disparities and Molecular Medicine, and Department of Pathology and Human Anatomy, Loma Linda University
| | - Andrea Parra
- From the Center for Health Disparities and Molecular Medicine, Department of Microbiology and Molecular Genetics, Department of Medicine, Department of Pharmaceutical and Administrative Sciences, School of Pharmacy, Department of Pathology and Human Anatomy, Loma Linda University, Loma Linda; Division of Rheumatology, Beaver Medical Group, Redlands, California, USA.A.J. Weldon, MS; A. Benitez, PhD, Center for Health Disparities and Molecular Medicine, and Department of Microbiology and Molecular Genetics, Loma Linda University; I. Moldovan, MD, Department of Medicine, Loma Linda University, and Division of Rheumatology, Beaver Medical Group; M.G. Cabling, MD; S. Hsu, MD; N. Daoud, MD; K. Colburn, MD, Department of Medicine, Loma Linda University; E.A. Hernandez, PhD, Department of Pharmaceutical and Administrative Sciences, School of Pharmacy, Loma Linda University; J. Gonzalez, BS; A. Parra, BS, Center for Health Disparities and Molecular Medicine, Loma Linda University; K.J. Payne, PhD, Center for Health Disparities and Molecular Medicine, and Department of Pathology and Human Anatomy, Loma Linda University
| | - Abigail Benitez
- From the Center for Health Disparities and Molecular Medicine, Department of Microbiology and Molecular Genetics, Department of Medicine, Department of Pharmaceutical and Administrative Sciences, School of Pharmacy, Department of Pathology and Human Anatomy, Loma Linda University, Loma Linda; Division of Rheumatology, Beaver Medical Group, Redlands, California, USA.A.J. Weldon, MS; A. Benitez, PhD, Center for Health Disparities and Molecular Medicine, and Department of Microbiology and Molecular Genetics, Loma Linda University; I. Moldovan, MD, Department of Medicine, Loma Linda University, and Division of Rheumatology, Beaver Medical Group; M.G. Cabling, MD; S. Hsu, MD; N. Daoud, MD; K. Colburn, MD, Department of Medicine, Loma Linda University; E.A. Hernandez, PhD, Department of Pharmaceutical and Administrative Sciences, School of Pharmacy, Loma Linda University; J. Gonzalez, BS; A. Parra, BS, Center for Health Disparities and Molecular Medicine, Loma Linda University; K.J. Payne, PhD, Center for Health Disparities and Molecular Medicine, and Department of Pathology and Human Anatomy, Loma Linda University
| | - Nasim Daoud
- From the Center for Health Disparities and Molecular Medicine, Department of Microbiology and Molecular Genetics, Department of Medicine, Department of Pharmaceutical and Administrative Sciences, School of Pharmacy, Department of Pathology and Human Anatomy, Loma Linda University, Loma Linda; Division of Rheumatology, Beaver Medical Group, Redlands, California, USA.A.J. Weldon, MS; A. Benitez, PhD, Center for Health Disparities and Molecular Medicine, and Department of Microbiology and Molecular Genetics, Loma Linda University; I. Moldovan, MD, Department of Medicine, Loma Linda University, and Division of Rheumatology, Beaver Medical Group; M.G. Cabling, MD; S. Hsu, MD; N. Daoud, MD; K. Colburn, MD, Department of Medicine, Loma Linda University; E.A. Hernandez, PhD, Department of Pharmaceutical and Administrative Sciences, School of Pharmacy, Loma Linda University; J. Gonzalez, BS; A. Parra, BS, Center for Health Disparities and Molecular Medicine, Loma Linda University; K.J. Payne, PhD, Center for Health Disparities and Molecular Medicine, and Department of Pathology and Human Anatomy, Loma Linda University
| | - Keith Colburn
- From the Center for Health Disparities and Molecular Medicine, Department of Microbiology and Molecular Genetics, Department of Medicine, Department of Pharmaceutical and Administrative Sciences, School of Pharmacy, Department of Pathology and Human Anatomy, Loma Linda University, Loma Linda; Division of Rheumatology, Beaver Medical Group, Redlands, California, USA.A.J. Weldon, MS; A. Benitez, PhD, Center for Health Disparities and Molecular Medicine, and Department of Microbiology and Molecular Genetics, Loma Linda University; I. Moldovan, MD, Department of Medicine, Loma Linda University, and Division of Rheumatology, Beaver Medical Group; M.G. Cabling, MD; S. Hsu, MD; N. Daoud, MD; K. Colburn, MD, Department of Medicine, Loma Linda University; E.A. Hernandez, PhD, Department of Pharmaceutical and Administrative Sciences, School of Pharmacy, Loma Linda University; J. Gonzalez, BS; A. Parra, BS, Center for Health Disparities and Molecular Medicine, Loma Linda University; K.J. Payne, PhD, Center for Health Disparities and Molecular Medicine, and Department of Pathology and Human Anatomy, Loma Linda University
| | - Kimberly J Payne
- From the Center for Health Disparities and Molecular Medicine, Department of Microbiology and Molecular Genetics, Department of Medicine, Department of Pharmaceutical and Administrative Sciences, School of Pharmacy, Department of Pathology and Human Anatomy, Loma Linda University, Loma Linda; Division of Rheumatology, Beaver Medical Group, Redlands, California, USA.A.J. Weldon, MS; A. Benitez, PhD, Center for Health Disparities and Molecular Medicine, and Department of Microbiology and Molecular Genetics, Loma Linda University; I. Moldovan, MD, Department of Medicine, Loma Linda University, and Division of Rheumatology, Beaver Medical Group; M.G. Cabling, MD; S. Hsu, MD; N. Daoud, MD; K. Colburn, MD, Department of Medicine, Loma Linda University; E.A. Hernandez, PhD, Department of Pharmaceutical and Administrative Sciences, School of Pharmacy, Loma Linda University; J. Gonzalez, BS; A. Parra, BS, Center for Health Disparities and Molecular Medicine, Loma Linda University; K.J. Payne, PhD, Center for Health Disparities and Molecular Medicine, and Department of Pathology and Human Anatomy, Loma Linda University
| |
Collapse
|
224
|
Kwun J, Page E, Hong JJ, Gibby A, Yoon J, Farris AB, Villinger F, Knechtle S. Neutralizing BAFF/APRIL with atacicept prevents early DSA formation and AMR development in T cell depletion induced nonhuman primate AMR model. Am J Transplant 2015; 15:815-22. [PMID: 25675879 PMCID: PMC5504528 DOI: 10.1111/ajt.13045] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2014] [Revised: 09/24/2014] [Accepted: 09/28/2014] [Indexed: 01/25/2023]
Abstract
Depletional strategies directed toward achieving tolerance induction in organ transplantation have been associated with an increased incidence and risk of antibody-mediated rejection (AMR) and graft injury. Our clinical data suggest correlation of increased serum B cell activating factor/survival factor (BAFF) with increased risk of antibody-mediated rejection in alemtuzumab treated patients. In the present study, we tested the ability of BAFF blockade (TACI-Ig) in a nonhuman primate AMR model to prevent alloantibody production and prolong allograft survival. Three animals received the AMR inducing regimen (CD3-IT/alefacept/tacrolimus) with TACI-Ig (atacicept), compared to five control animals treated with the AMR inducing regimen only. TACI-Ig treatment lead to decreased levels of DSA in treated animals at 2 and 4 weeks posttransplantation (p < 0.05). In addition, peripheral B cell numbers were significantly lower at 6 weeks posttransplantation. However, it provided only a marginal increase in graft survival (59 ± 22 vs. 102 ± 47 days; p = 0.11). Histological analysis revealed a substantial reduction in findings typically associated with humoral rejection with atacicept treatment. More T cell rejection findings were observed with increased graft T cell infiltration in atacicept treatment, likely secondary to the graft prolongation. We show that BAFF/APRIL blockade using concomitant TACI-Ig treatment reduced the humoral portion of rejection in our depletion-induced preclinical AMR model.
Collapse
Affiliation(s)
- J. Kwun
- Department of Surgery, Emory Transplant Center, Emory University School of Medicine, Atlanta, GA
| | - E. Page
- Department of Surgery, Emory Transplant Center, Emory University School of Medicine, Atlanta, GA
| | - J. J. Hong
- Department of Pathology, Emory University School of Medicine, Atlanta, GA,Division of Pathology, Yerkes National Primate Research Center, Emory University, Atlanta, GA
| | - A. Gibby
- Department of Surgery, Emory Transplant Center, Emory University School of Medicine, Atlanta, GA
| | - J. Yoon
- Department of Surgery, Emory Transplant Center, Emory University School of Medicine, Atlanta, GA
| | - A. B. Farris
- Department of Pathology, Emory University School of Medicine, Atlanta, GA
| | - F. Villinger
- Department of Pathology, Emory University School of Medicine, Atlanta, GA,Division of Pathology, Yerkes National Primate Research Center, Emory University, Atlanta, GA
| | - S. Knechtle
- Department of Surgery, Emory Transplant Center, Emory University School of Medicine, Atlanta, GA,Corresponding author Stuart J. Knechtle,
| |
Collapse
|
225
|
Stohl W. Editorial: The BAFFling Immunology of Systemic Lupus Erythematosus: Beyond B Cells. Arthritis Rheumatol 2015; 67:612-5. [DOI: 10.1002/art.38951] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2014] [Accepted: 11/04/2014] [Indexed: 01/07/2023]
|
226
|
Zhan Y, Carrington EM, Ko HJ, Vikstrom IB, Oon S, Zhang JG, Vremec D, Brady JL, Bouillet P, Wu L, Huang DCS, Wicks IP, Morand EF, Strasser A, Lew AM. Bcl-2 Antagonists Kill Plasmacytoid Dendritic Cells From Lupus-Prone Mice and Dampen Interferon-α Production. Arthritis Rheumatol 2015; 67:797-808. [DOI: 10.1002/art.38966] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2014] [Accepted: 11/13/2014] [Indexed: 02/06/2023]
Affiliation(s)
- Yifan Zhan
- The Walter & Eliza Hall Institute of Medical Research and University of Melbourne, Parkville; Victoria Australia
| | - Emma M. Carrington
- The Walter & Eliza Hall Institute of Medical Research and University of Melbourne, Parkville; Victoria Australia
| | - Hyun-Ja Ko
- The Walter & Eliza Hall Institute of Medical Research and University of Melbourne, Parkville; Victoria Australia
| | - Ingela B. Vikstrom
- The Walter & Eliza Hall Institute of Medical Research and University of Melbourne, Parkville; Victoria Australia
| | - Shereen Oon
- The Walter & Eliza Hall Institute of Medical Research and University of Melbourne, Parkville; Victoria Australia
| | - Jian-Guo Zhang
- The Walter & Eliza Hall Institute of Medical Research and University of Melbourne, Parkville; Victoria Australia
| | - David Vremec
- The Walter & Eliza Hall Institute of Medical Research, Parkville; Victoria Australia
| | - Jamie L. Brady
- The Walter & Eliza Hall Institute of Medical Research, Parkville; Victoria Australia
| | - Philippe Bouillet
- The Walter & Eliza Hall Institute of Medical Research and University of Melbourne, Parkville; Victoria Australia
| | - Li Wu
- Tsinghua University and Peking University Joint Center for Life Sciences and Tsinghua University School of Medicine; Beijing China
| | - David C. S. Huang
- The Walter & Eliza Hall Institute of Medical Research and University of Melbourne, Parkville; Victoria Australia
| | - Ian P. Wicks
- The Walter & Eliza Hall Institute of Medical Research and University of Melbourne, Parkville; Victoria Australia
| | - Eric F. Morand
- Centre for Inflammatory Diseases, Monash University, Melbourne; Victoria Australia
| | - Andreas Strasser
- Centre for Inflammatory Diseases, Monash University, Melbourne; Victoria Australia
| | - Andrew M. Lew
- The Walter & Eliza Hall Institute of Medical Research and University of Melbourne, Parkville; Victoria Australia
| |
Collapse
|
227
|
Wu DH, Xu L, Wen CP, Xie GQ, Ji JJ, Pan JL, Jiao YF, Fan YS. The effects of Jieduquyuzishen prescription-treated rat serum on the BAFF/BAFF-R signal pathway. PLoS One 2015; 10:e0118462. [PMID: 25689512 PMCID: PMC4331425 DOI: 10.1371/journal.pone.0118462] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2014] [Accepted: 01/17/2015] [Indexed: 01/28/2023] Open
Abstract
Systemic lupus erythematosus (SLE) is a chronic inflammatory disease mainly characterized by B cell hyperactivity. Glucocorticoid (GC) is widely used in SLE for its potent anti-inflammatory and immunosuppressive effects. Despite its important clinical efficacy, high-dose or long-term use of GC can cause severe side effects, such as osteoporosis, osteonecrosis, cataracts, hyperglycemia, coronary heart disease and cognitive impairment. Our early clinical studies have shown that Jieduquyuzishen prescription (JP) can effectively reduce the adverse effects and improve the curative effect of GC in the treatment of SLE. The BAFF/BAFF-R signaling pathway plays an important role in the development of SLE and has been regarded as a potential target for the therapy of SLE. In this study, we attempt to investigate the effect of JP on the BAFF/BAFF-R signaling pathway to explore the mechanism of JP in reducing the toxicity and enhancing the efficacy of GC. YAC-1 cells, isolated rat peripheral blood lymphocytes, polymorphonuclear neutrophils and spleen lymphocytes were treated with drug-containing serum. The results of RT-PCR, Western blot and dual-luciferase reporter gene assays indicate that either JP or GC can inhibit the mBAFF-induced up-regulation of BAFF, BAFF-R, Bcl-2, IL-10 and NF-κB in YAC-1 cells and WEHI-231 cells. Furthermore, MTS, flow cytometry and CFSE results reveal that the proliferation and survival of lymphocytes activated by mBAFF are suppressed by JP, GC and their combination. Contrary to GC, JP can reduce the apoptosis and raise the survival of polymorphonuclear neutrophils and can’t increase the apoptosis of the peripheral blood lymphocytes and spleen lymphocytes. Therefore, it is possible that JP can down-regulate the BAFF/BAFF-R signaling pathway as effectively as GC, which may result in the dosage reduction of GC, thus decreasing the toxicity and improving the efficacy of GC-based treatment of SLE.
Collapse
Affiliation(s)
- De-hong Wu
- First Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Li Xu
- College of Basic Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| | - Cheng-ping Wen
- College of Basic Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| | - Guan-qun Xie
- College of Basic Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| | - Jin-jun Ji
- College of Basic Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| | - Jie-li Pan
- College of Basic Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| | - Yi-feng Jiao
- College of Basic Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| | - Yong-sheng Fan
- First Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China
- * E-mail:
| |
Collapse
|
228
|
Chang Y, Sun X, Jia X, Xu S, Wei F, Yang X, Wei W. Expression and effects of B-lymphocyte stimulator and its receptors in T cell-mediated autoimmune arthritis. Int Immunopharmacol 2015; 24:451-457. [DOI: 10.1016/j.intimp.2015.01.001] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2014] [Revised: 12/25/2014] [Accepted: 01/05/2015] [Indexed: 02/05/2023]
|
229
|
Gorbacheva V, Ayasoufi K, Fan R, Baldwin WM, Valujskikh A. B cell activating factor (BAFF) and a proliferation inducing ligand (APRIL) mediate CD40-independent help by memory CD4 T cells. Am J Transplant 2015; 15:346-57. [PMID: 25496308 PMCID: PMC4304935 DOI: 10.1111/ajt.12984] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2014] [Revised: 08/07/2014] [Accepted: 08/12/2014] [Indexed: 01/25/2023]
Abstract
Donor-reactive memory T cells undermine organ transplant survival and are poorly controlled by immunosuppression or costimulatory blockade. Memory CD4 T cells provide CD40-independent help for the generation of donor-reactive effector CD8 T cells and alloantibodies (alloAbs) that rapidly mediate allograft rejection. The goal of this study was to investigate the role of B cell activating factor (BAFF) and a proliferation-inducing ligand (APRIL) in alloresponses driven by memory CD4 T cells. The short-term neutralization of BAFF alone or BAFF plus APRIL synergized with anti-CD154 mAb to prolong heart allograft survival in recipients containing donor-reactive memory CD4 T cells. The prolongation was associated with reduction in antidonor alloAb responses and with inhibited reactivation and helper functions of memory CD4 T cells. Additional depletion of CD8 T cells did not enhance the prolonged allograft survival suggesting that donor-reactive alloAbs mediate late graft rejection in these recipients. This is the first report that targeting the BAFF cytokine network inhibits both humoral and cellular immune responses induced by memory CD4 T cells. Our results suggest that reagents neutralizing BAFF and APRIL may be used to enhance the efficacy of CD40/CD154 costimulatory blockade and improve allograft survival in T cell-sensitized recipients.
Collapse
Affiliation(s)
- Victoria Gorbacheva
- Glickman Urological Institute and Department of Immunology, Cleveland Clinic, Cleveland, Ohio 44195, USA
| | - Katayoun Ayasoufi
- Glickman Urological Institute and Department of Immunology, Cleveland Clinic, Cleveland, Ohio 44195, USA
| | - Ran Fan
- Glickman Urological Institute and Department of Immunology, Cleveland Clinic, Cleveland, Ohio 44195, USA
| | - William M. Baldwin
- Glickman Urological Institute and Department of Immunology, Cleveland Clinic, Cleveland, Ohio 44195, USA
| | - Anna Valujskikh
- Glickman Urological Institute and Department of Immunology, Cleveland Clinic, Cleveland, Ohio 44195, USA
| |
Collapse
|
230
|
Zhao Y, Hao X, Feng J, Shen B, Wei J, Sun J. The comparison of BLyS-binding peptides from phage display library and computer-aided design on BLyS–TACI interaction. Int Immunopharmacol 2015; 24:219-223. [DOI: 10.1016/j.intimp.2014.12.013] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2014] [Revised: 11/30/2014] [Accepted: 12/09/2014] [Indexed: 10/24/2022]
|
231
|
Kitabatake M, Soma M, Zhang T, Kuwahara K, Fukushima Y, Nojima T, Kitamura D, Sakaguchi N. JNK regulatory molecule G5PR induces IgG autoantibody-producing plasmablasts from peritoneal B1a cells. THE JOURNAL OF IMMUNOLOGY 2015; 194:1480-8. [PMID: 25601926 DOI: 10.4049/jimmunol.1401127] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Peritoneal B1a cells expressing CD5 and CD11b generate autoantibody-producing precursors in autoimmune-prone mice. Previous studies show reduced JNK signaling in peritoneal B1a cells of female New Zealand Black mice and an abnormal increase of protein phosphatase 2A subunit G5PR that regulates BCR-mediated JNK signaling as a cause of autoimmunity. To investigate the mechanism regulating B1a differentiation into autoantibody-secreting plasmablasts (PBs), we applied an in vitro culture system that supports long-term growth of germinal center (GC) B cells (iGB) with IL-4, CD40L, and BAFF. Compared with spleen B2 cells, B1a cells differentiated into GC-like B cells, but more markedly into PBs, and underwent class switching toward IgG1. During iGB culture, B1a cells expressed GC-associated aicda, g5pr, and bcl6, and markedly PB-associated prdm1, irf4, and xbp1. B1a-derived iGB cells from New Zealand Black × New Zealand White F1 mice highly differentiated into autoantibody-secreting PBs in vitro and localized to the GC area in vivo. In iGB culture, JNK inhibitor SP600125 augmented the differentiation of C57BL/6 B1a cells into PBs. Furthermore, B1a cells from G5PR transgenic mice markedly differentiated into IgM and IgG autoantibody-secreting PBs. In conclusion, JNK regulation is critical to suppress autoantibody-secreting PBs from peritoneal B1a cells.
Collapse
Affiliation(s)
- Masahiro Kitabatake
- Department of Immunology, Graduate School of Medical Sciences, Kumamoto University, Kumamoto 860-8556, Japan
| | - Miho Soma
- Department of Immunology, Graduate School of Medical Sciences, Kumamoto University, Kumamoto 860-8556, Japan
| | - Tianli Zhang
- Department of Immunology, Graduate School of Medical Sciences, Kumamoto University, Kumamoto 860-8556, Japan
| | - Kazuhiko Kuwahara
- Department of Immunology, Graduate School of Medical Sciences, Kumamoto University, Kumamoto 860-8556, Japan
| | - Yoshimi Fukushima
- Department of Immunology, Graduate School of Medical Sciences, Kumamoto University, Kumamoto 860-8556, Japan
| | - Takuya Nojima
- Division of Molecular Biology, Research Institute for Biomedical Sciences, Tokyo University of Science, Chiba 278-0022, Japan; and
| | - Daisuke Kitamura
- Division of Molecular Biology, Research Institute for Biomedical Sciences, Tokyo University of Science, Chiba 278-0022, Japan; and
| | - Nobuo Sakaguchi
- Department of Immunology, Graduate School of Medical Sciences, Kumamoto University, Kumamoto 860-8556, Japan;
| |
Collapse
|
232
|
A mutation in caspase-9 decreases the expression of BAFFR and ICOS in patients with immunodeficiency and lymphoproliferation. Genes Immun 2015; 16:151-61. [PMID: 25569260 DOI: 10.1038/gene.2014.74] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2014] [Revised: 10/09/2014] [Accepted: 11/17/2014] [Indexed: 01/25/2023]
Abstract
Lymphocyte apoptosis is mainly induced by either death receptor-dependent activation of caspase-8 or mitochondria-dependent activation of caspase-9. Mutations in caspase-8 lead to autoimmunity/lymphoproliferation and immunodeficiency. This work describes a heterozygous H237P mutation in caspase-9 that can lead to similar disorders. H237P mutation was detected in two patients: Pt1 with autoimmunity/lymphoproliferation, severe hypogammaglobulinemia and Pt2 with mild hypogammaglobulinemia and Burkitt lymphoma. Their lymphocytes displayed defective caspase-9 activity and decreased apoptotic and activation responses. Transfection experiments showed that mutant caspase-9 display defective enzyme and proapoptotic activities and a dominant-negative effect on wild-type caspase-9. Ex vivo analysis of the patients' lymphocytes and in vitro transfection experiments showed that the expression of mutant caspase-9 correlated with a downregulation of BAFFR (B-cell-activating factor belonging to the TNF family (BAFF) receptor) in B cells and ICOS (inducible T-cell costimulator) in T cells. Both patients carried a second inherited heterozygous mutation missing in the relatives carrying H237P: Pt1 in the transmembrane activator and CAML interactor (TACI) gene (S144X) and Pt2 in the perforin (PRF1) gene (N252S). Both mutations have been previously associated with immunodeficiencies in homozygosis or compound heterozygosis. Taken together, these data suggest that caspase-9 mutations may predispose to immunodeficiency by cooperating with other genetic factors, possibly by downregulating the expression of BAFFR and ICOS.
Collapse
|
233
|
Lenert A, Lenert P. Current and emerging treatment options for ANCA-associated vasculitis: potential role of belimumab and other BAFF/APRIL targeting agents. DRUG DESIGN DEVELOPMENT AND THERAPY 2015; 9:333-47. [PMID: 25609919 PMCID: PMC4294650 DOI: 10.2147/dddt.s67264] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Antineutrophil cytoplasmic antibody (ANCA)-associated vasculitis (AAV) comprises several clinical entities with diverse clinical presentations, outcomes, and nonunifying pathogenesis. AAV has a clear potential for relapses, and shows unpredictable response to treatment. Cyclophosphamide-based therapies have remained the hallmark of induction therapy protocols for more than four decades. Recently, B-cell depleting therapy with the anti-CD20 antibody rituximab has proved beneficial in AAV, leading to Food and Drug Administration approval of rituximab in combination with corticosteroids for the treatment of AAV in adults. Rituximab for ANCA-associated vasculitis and other clinical trials provided clear evidence that rituximab was not inferior to cyclophosphamide for remission induction, and rituximab appeared even more beneficial in patients with relapsing disease. This raised hopes that other B-cell-targeted therapies directed either against CD19, CD20, CD22, or B-cell survival factors, B-cell activating factor of the tumor necrosis factor family (BAFF) and a proliferation-inducing ligand could also be beneficial for the management of AAV. BAFF neutralization with the fully humanized monoclonal antibody belimumab has already shown success in human systemic lupus erythematosus and, along with another anti-BAFF reagent blisibimod, is currently undergoing Phase II and III clinical trials in AAV. Local production of BAFF in granulomatous lesions and elevated levels of serum BAFF in AAV provide a rationale for BAFF-targeted therapies not only in AAV but also in other forms of vasculitis such as Behcet’s disease, large-vessel vasculitis, or cryoglobulinemic vasculitis secondary to chronic hepatitis C infection. BAFF-targeted therapies have a very solid safety profile, and may have an additional benefit of preferentially targeting newly arising autoreactive B cells over non-self-reactive B cells.
Collapse
Affiliation(s)
- Aleksander Lenert
- Division of Rheumatology, University of Kentucky, Kentucky Clinic, Lexington, KY, USA
| | - Petar Lenert
- Division of Immunology, Department of Internal Medicine, The University of Iowa, Iowa City, IA, USA
| |
Collapse
|
234
|
Siebert S, Tsoukas A, Robertson J, McInnes I. Cytokines as therapeutic targets in rheumatoid arthritis and other inflammatory diseases. Pharmacol Rev 2015; 67:280-309. [PMID: 25697599 DOI: 10.1124/pr.114.009639] [Citation(s) in RCA: 242] [Impact Index Per Article: 24.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/21/2025] Open
Abstract
The human immune system involves highly complex and coordinated processes in which small proteins named cytokines play a key role. Cytokines have been implicated in the pathogenesis of a number of inflammatory and autoimmune diseases. Cytokines are therefore attractive therapeutic targets in these conditions. Anticytokine therapy for inflammatory diseases became a clinical reality with the introduction of tumor necrosis factor (TNF) inhibitors for the treatment of severe rheumatoid arthritis. Although these therapies have transformed the treatment of patients with severe inflammatory arthritis, there remain significant limiting factors: treatment failure is commonly seen in the clinic; safety concerns remain; there is uncertainty regarding the relevance of immunogenicity; the absence of biomarkers to direct therapy decisions and high drug costs limit availability in some healthcare systems. In this article, we provide an overview of the key efficacy and safety trials for currently approved treatments in rheumatoid arthritis and review the major lessons learned from a decade of use in clinical practice, focusing mainly on anti-TNF and anti-interleukin (IL)-6 agents. We also describe the clinical application of anticytokine therapies for other inflammatory diseases, particularly within the spondyloarthritis spectrum, and highlight differential responses across diseases. Finally, we report on the current state of trials for newer therapeutic targets, focusing mainly on the IL-17 and IL-23 pathways.
Collapse
MESH Headings
- Animals
- Anti-Inflammatory Agents/adverse effects
- Anti-Inflammatory Agents/therapeutic use
- Anti-Inflammatory Agents, Non-Steroidal/adverse effects
- Anti-Inflammatory Agents, Non-Steroidal/therapeutic use
- Antirheumatic Agents/adverse effects
- Antirheumatic Agents/therapeutic use
- Arthritis, Psoriatic/drug therapy
- Arthritis, Psoriatic/immunology
- Arthritis, Psoriatic/metabolism
- Arthritis, Rheumatoid/drug therapy
- Arthritis, Rheumatoid/immunology
- Arthritis, Rheumatoid/metabolism
- Crohn Disease/drug therapy
- Crohn Disease/immunology
- Crohn Disease/metabolism
- Cytokines/antagonists & inhibitors
- Cytokines/metabolism
- Drugs, Investigational/adverse effects
- Drugs, Investigational/therapeutic use
- Humans
- Interleukin-1/antagonists & inhibitors
- Interleukin-1/metabolism
- Interleukin-6/antagonists & inhibitors
- Interleukin-6/metabolism
- Models, Biological
- Molecular Targeted Therapy/adverse effects
- Psoriasis/drug therapy
- Psoriasis/immunology
- Psoriasis/metabolism
- Spondylitis, Ankylosing/drug therapy
- Spondylitis, Ankylosing/immunology
- Spondylitis, Ankylosing/metabolism
- Tumor Necrosis Factor Inhibitors
- Tumor Necrosis Factors/metabolism
Collapse
Affiliation(s)
- Stefan Siebert
- Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, United Kingdom (S.S., J.R., I.M.); and Division of Rheumatology, McGill University Health Centre, Montreal, Quebec, Canada (A.T.)
| | - Alexander Tsoukas
- Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, United Kingdom (S.S., J.R., I.M.); and Division of Rheumatology, McGill University Health Centre, Montreal, Quebec, Canada (A.T.)
| | - Jamie Robertson
- Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, United Kingdom (S.S., J.R., I.M.); and Division of Rheumatology, McGill University Health Centre, Montreal, Quebec, Canada (A.T.)
| | - Iain McInnes
- Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, United Kingdom (S.S., J.R., I.M.); and Division of Rheumatology, McGill University Health Centre, Montreal, Quebec, Canada (A.T.)
| |
Collapse
|
235
|
Jordan N, D’Cruz DP. Belimumab for the treatment of systemic lupus erythematosus. Expert Rev Clin Immunol 2014; 11:195-204. [DOI: 10.1586/1744666x.2015.996550] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
236
|
Peng QL, Shu XM, Wang DX, Wang Y, Lu X, Wang GC. B-cell activating factor as a serological biomarker for polymyositis and dermatomyositis. Biomark Med 2014; 8:395-403. [PMID: 24712431 DOI: 10.2217/bmm.13.124] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
AIM To investigate serum levels of B-cell activating factor (BAFF) in the patients with polymyositis (PM) and dermatomyositis (DM), and to systematically examine the association between serum BAFF levels and disease activity in PM/DM patients. PATIENTS & METHODS A cross-sectional analysis included 92 PM/DM patients and 25 healthy control subjects. A longitudinal study followed 24 patients. Serum BAFF concentrations were detected by the ELISA method. RESULTS Serum BAFF levels in PM/DM patients were significantly higher than those in healthy controls. A cross-sectional assessment revealed a modest correlation between serum BAFF levels and global disease activity and a mild correlation between serum BAFF levels and muscle disease activity. The longitudinal study showed that serum BAFF levels modestly correlated with global disease activity and muscle disease activity. CONCLUSION Resulting data showed high serum BAFF levels in PM/DM patients and suggested BAFF as a serological biomarker for PM/DM disease activity.
Collapse
Affiliation(s)
- Qing-Lin Peng
- Department of Rheumatology, China-Japan Friendship Hospital, Ying Hua East Road, Chao Yang District, Beijing 100029, China
| | | | | | | | | | | |
Collapse
|
237
|
Mackern-Oberti JP, Vega F, Llanos C, Bueno SM, Kalergis AM. Targeting dendritic cell function during systemic autoimmunity to restore tolerance. Int J Mol Sci 2014; 15:16381-417. [PMID: 25229821 PMCID: PMC4200801 DOI: 10.3390/ijms150916381] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2014] [Revised: 08/29/2014] [Accepted: 09/05/2014] [Indexed: 12/11/2022] Open
Abstract
Systemic autoimmune diseases can damage nearly every tissue or cell type of the body. Although a great deal of progress has been made in understanding the pathogenesis of autoimmune diseases, current therapies have not been improved, remain unspecific and are associated with significant side effects. Because dendritic cells (DCs) play a major role in promoting immune tolerance against self-antigens (self-Ags), current efforts are focusing at generating new therapies based on the transfer of tolerogenic DCs (tolDCs) during autoimmunity. However, the feasibility of this approach during systemic autoimmunity has yet to be evaluated. TolDCs may ameliorate autoimmunity mainly by restoring T cell tolerance and, thus, indirectly modulating autoantibody development. In vitro induction of tolDCs loaded with immunodominant self-Ags and subsequent cell transfer to patients would be a specific new therapy that will avoid systemic immunosuppression. Herein, we review recent approaches evaluating the potential of tolDCs for the treatment of systemic autoimmune disorders.
Collapse
Affiliation(s)
- Juan P Mackern-Oberti
- Millennium Institute of Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Portugal 49, Santiago 8330025, Chile.
| | - Fabián Vega
- Departamento de Inmunología Clínica y Reumatología, Escuela de Medicina, Pontificia Universidad Católica de Chile, Marcoleta 350, Santiago 8330033, Chile.
| | - Carolina Llanos
- Departamento de Inmunología Clínica y Reumatología, Escuela de Medicina, Pontificia Universidad Católica de Chile, Marcoleta 350, Santiago 8330033, Chile.
| | - Susan M Bueno
- Millennium Institute of Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Portugal 49, Santiago 8330025, Chile.
| | - Alexis M Kalergis
- Millennium Institute of Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Portugal 49, Santiago 8330025, Chile.
| |
Collapse
|
238
|
High APRIL but not BAFF serum levels are associated with poor outcome in patients with follicular lymphoma. Ann Hematol 2014; 94:79-88. [DOI: 10.1007/s00277-014-2173-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2014] [Accepted: 07/16/2014] [Indexed: 10/24/2022]
|
239
|
Kamal A, Khamashta M. The efficacy of novel B cell biologics as the future of SLE treatment: a review. Autoimmun Rev 2014; 13:1094-101. [PMID: 25149393 DOI: 10.1016/j.autrev.2014.08.020] [Citation(s) in RCA: 88] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2014] [Accepted: 05/28/2014] [Indexed: 01/01/2023]
Abstract
Systemic lupus erythematosus (SLE) is a chronic autoimmune inflammatory disease with wide ranging multi-systemic effects. Current understanding centralises B cells in SLE pathogenesis with clinical features resulting from autoantibody formation, immune complex deposition, antigen presentation and cytokine activation. Existing standard of care therapies generates adverse side effects; secondary to corticosteroid use and untargeted immunosuppression. The inability to uphold remission and abolish the disease process, in addition to the increasing numbers of patients seen with refractory disease with these therapies, has provoked the development of novel B cell biologics targeting specific pathogenic pathways fundamental to the SLE disease process. Current evidence highlighting the efficacy of Rituximab, Ocrelizumab and Epratuzumab in inducing B cell depletion and achieving disease amelioration through specific B cell surface receptor antagonism is discussed. We review the efficacy of Atacicept, Briobacept and Belimumab in antagonising B lymphocyte stimulator (BLyS) and A proliferation inducing ligand (APRIL), two stimulatory cytokines crucial to B cell survival, growth and function. Two large multicentre randomised controlled trials, BLISS-52 and BLISS-76, have led to FDA approval of Belimumab. Following this breakthrough, other anti-BLyS therapies, Blisibimod and Tabalumab, are currently under Phase III evaluation. Similarly, murine models and Phase I/II trials have demonstrated significant efficacy of Rituximab, Epratuzumab, Briobacept and Atacicept as potential future therapies and we now eagerly await results from Phase III trials. Future research must compare the efficacy of different biologics amongst different patient subpopulations and SLE manifestations, in order to develop clinically and cost effective therapies.
Collapse
Affiliation(s)
- Ameer Kamal
- King's College London, The Rayne Institute, 4th Floor Lambeth Wing, St Thomas' Hospital, Westminster Bridge Road, SE1 7EH London, UK
| | - Munther Khamashta
- Graham Hughes Lupus Research Laboratory, Division of Women's Health, King's College London, The Rayne Institute, Lambeth Wing, 4th Floor, St Thomas' Hospital, London SE1 7EH, UK.
| |
Collapse
|
240
|
Yasuda K, Watkins AA, Kochar GS, Wilson GE, Laskow B, Richez C, Bonegio RG, Rifkin IR. Interferon regulatory factor-5 deficiency ameliorates disease severity in the MRL/lpr mouse model of lupus in the absence of a mutation in DOCK2. PLoS One 2014; 9:e103478. [PMID: 25076492 PMCID: PMC4116215 DOI: 10.1371/journal.pone.0103478] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2014] [Accepted: 07/01/2014] [Indexed: 11/18/2022] Open
Abstract
Interferon regulatory factor 5 (IRF5) polymorphisms are strongly associated with an increased risk of developing the autoimmune disease systemic lupus erythematosus. In mouse lupus models, IRF5-deficiency was shown to reduce disease severity consistent with an important role for IRF5 in disease pathogenesis. However these mouse studies were confounded by the recent demonstration that the IRF5 knockout mouse line contained a loss-of-function mutation in the dedicator of cytokinesis 2 (DOCK2) gene. As DOCK2 regulates lymphocyte trafficking and Toll-like receptor signaling, this raised the possibility that some of the protective effects attributed to IRF5 deficiency in the mouse lupus models may instead have been due to DOCK2 deficiency. We have therefore here evaluated the effect of IRF5-deficiency in the MRL/lpr mouse lupus model in the absence of the DOCK2 mutation. We find that IRF5-deficient (IRF5−/−) MRL/lpr mice develop much less severe disease than their IRF5-sufficient (IRF5+/+) littermates. Despite markedly lower serum levels of anti-nuclear autoantibodies and reduced total splenocyte and CD4+ T cell numbers, IRF5−/− MRL/lpr mice have similar numbers of all splenic B cell subsets compared to IRF5+/+ MRL/lpr mice, suggesting that IRF5 is not involved in B cell development up to the mature B cell stage. However, IRF5−/− MRL/lpr mice have greatly reduced numbers of spleen plasmablasts and bone marrow plasma cells. Serum levels of B lymphocyte stimulator (BLyS) were markedly elevated in the MRL/lpr mice but no effect of IRF5 on serum BLyS levels was seen. Overall our data demonstrate that IRF5 contributes to disease pathogenesis in the MRL/lpr lupus model and that this is due, at least in part, to the role of IRF5 in plasma cell formation. Our data also suggest that combined therapy targeting both IRF5 and BLyS might be a particularly effective therapeutic approach in lupus.
Collapse
Affiliation(s)
- Kei Yasuda
- Renal Section, Department of Medicine, Boston University School of Medicine, Boston, Massachusetts, United States of America
- * E-mail: (KY); (IRR)
| | - Amanda A. Watkins
- Renal Section, Department of Medicine, Boston University School of Medicine, Boston, Massachusetts, United States of America
| | - Guneet S. Kochar
- Renal Section, Department of Medicine, Boston University School of Medicine, Boston, Massachusetts, United States of America
| | - Gabriella E. Wilson
- Renal Section, Department of Medicine, Boston University School of Medicine, Boston, Massachusetts, United States of America
| | - Bari Laskow
- Renal Section, Department of Medicine, Boston University School of Medicine, Boston, Massachusetts, United States of America
| | - Christophe Richez
- Renal Section, Department of Medicine, Boston University School of Medicine, Boston, Massachusetts, United States of America
| | - Ramon G. Bonegio
- Renal Section, Department of Medicine, Boston University School of Medicine, Boston, Massachusetts, United States of America
| | - Ian R. Rifkin
- Renal Section, Department of Medicine, Boston University School of Medicine, Boston, Massachusetts, United States of America
- * E-mail: (KY); (IRR)
| |
Collapse
|
241
|
Coquery CM, Wade NS, Loo WM, Kinchen JM, Cox KM, Jiang C, Tung KS, Erickson LD. Neutrophils contribute to excess serum BAFF levels and promote CD4+ T cell and B cell responses in lupus-prone mice. PLoS One 2014; 9:e102284. [PMID: 25010693 PMCID: PMC4092127 DOI: 10.1371/journal.pone.0102284] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2014] [Accepted: 06/16/2014] [Indexed: 12/31/2022] Open
Abstract
Despite increased frequencies of neutrophils found in autoimmune diseases such as systemic lupus erythematosus (SLE), how they contribute to disease pathogenesis and the mechanisms that affect the accumulation of neutrophils are poorly understood. The aim of this study was to identify factors in autoantibody-mediated autoimmunity that controls the accumulation of spleen resident neutrophils and to determine whether neutrophils contribute to abnormal B cell responses. Increased levels of the cytokine BAFF have been linked to loss of B cell tolerance in autoimmunity, but the cellular source responsible for excess BAFF is unknown. B cell maturation antigen (BCMA) is a receptor for BAFF and is critical for the survival of bone marrow plasma cells. Paradoxically, BCMA deficiency exacerbates the formation of autoantibody-secreting plasma cells in spleens of lupus-prone mice and the reasons for this effect are not understood. Here we analyzed the phenotype, localization and function of neutrophils in spleens of healthy mice and congenic lupus-prone mice, and compared mice sufficient or deficient in BCMA expression. Neutrophils were found to be significantly increased in frequency and activation status in spleens of lupus-prone mice when BCMA was absent. Furthermore, neutrophils localized within T cell zones and enhanced CD4+ T cell proliferation and IFNγ production through the production of BAFF. Reduced BAFF and IFNγ serum levels, decreased frequencies of IFNγ-producing T cells, germinal center B cells, and autoantibody production after neutrophil depletion indicated the involvement of neutrophils in these autoimmune traits. Thus, we have identified a novel role for BCMA to control excess BAFF production in murine lupus through restraining the accumulation of BAFF-producing neutrophils. Our data suggests that devising therapeutic strategies to reduce neutrophils in autoimmunity may decrease BAFF levels and ameliorate disease.
Collapse
Affiliation(s)
- Christine M. Coquery
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, Virginia, United States of America
- Beirne B. Carter Center for Immunology Research, University of Virginia, Charlottesville, Virginia, United States of America
| | - Nekeithia S. Wade
- Beirne B. Carter Center for Immunology Research, University of Virginia, Charlottesville, Virginia, United States of America
| | - William M. Loo
- Beirne B. Carter Center for Immunology Research, University of Virginia, Charlottesville, Virginia, United States of America
| | - Jason M. Kinchen
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, Virginia, United States of America
- Beirne B. Carter Center for Immunology Research, University of Virginia, Charlottesville, Virginia, United States of America
- Center for Cell Clearance, University of Virginia, Charlottesville, Virginia, United States of America
| | - Kelly M. Cox
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, Virginia, United States of America
- Beirne B. Carter Center for Immunology Research, University of Virginia, Charlottesville, Virginia, United States of America
| | - Chao Jiang
- Division of Extramural Research, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Kenneth S. Tung
- Beirne B. Carter Center for Immunology Research, University of Virginia, Charlottesville, Virginia, United States of America
- Department of Pathology, University of Virginia, Charlottesville, Virginia, United States of America
| | - Loren D. Erickson
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, Virginia, United States of America
- Beirne B. Carter Center for Immunology Research, University of Virginia, Charlottesville, Virginia, United States of America
- * E-mail:
| |
Collapse
|
242
|
López P, Scheel-Toellner D, Rodríguez-Carrio J, Caminal-Montero L, Gordon C, Suárez A. Interferon-α-induced B-lymphocyte stimulator expression and mobilization in healthy and systemic lupus erthymatosus monocytes. Rheumatology (Oxford) 2014; 53:2249-58. [PMID: 24942493 DOI: 10.1093/rheumatology/keu249] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
OBJECTIVE The aim of this study was to investigate the cellular populations and regulatory factors responsible for B-lymphocyte stimulator (BLyS) overexpression in SLE patients. METHODS Surface and intracellular BLyS levels were quantified by flow cytometry in healthy and SLE monocytes cultured in the presence of TNF-α, IFN-α, IFN-γ, GM-CSF and SLE immune complexes (SLE-ICs), while soluble BLyS was measured by ELISA. Also, both surface and intracellular BLyS expression by different cell subsets was determined in 23 SLE patients and 16 healthy controls. Disease activity was assessed using classic BILAG index. RESULTS In vitro experiments using healthy monocytes showed that IFN-α and SLE-ICs induced a progressive increase in surface-bound BLyS with respect to the intracellular stores. IFN-α-treated SLE monocytes, especially from patients with high anti-dsDNA levels or disease activity, exhibited higher intracellular levels of BLyS that was mobilized to the membrane more rapidly and subsequently released. Furthermore, ex vivo analysis of SLE patients revealed up-regulated BLyS expression in B cells, myeloid and plasmacytoid dendritic cells (DCs), whereas active patients had an increased surface:intracellular BLyS ratio in monocytes and myeloid DCs. CONCLUSION Monocyte BLyS induction and mobilization from intra- to extracellular compartments seems to be influenced by IFN-α and disease activity or anti-dsDNA levels. Accordingly, monocytes and myeloid DCs from active patients presented the highest membrane-bound:intracellular BLyS ratio. In addition, expression levels in several blood cells support the existence of generalized immune stimulation in SLE patients.
Collapse
Affiliation(s)
- Patricia López
- Department of Functional Biology, Immunology Area, Faculty of Medicine, University of Oviedo, Oviedo, Spain, Rheumatology Research Group, MRC Centre for Immune Regulation, Institute for Biomedical Research, School of Immunity and Infection, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK and Department of Internal Medicine, Hospital Universitario Central de Asturias, Oviedo, Spain. Department of Functional Biology, Immunology Area, Faculty of Medicine, University of Oviedo, Oviedo, Spain, Rheumatology Research Group, MRC Centre for Immune Regulation, Institute for Biomedical Research, School of Immunity and Infection, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK and Department of Internal Medicine, Hospital Universitario Central de Asturias, Oviedo, Spain.
| | - Dagmar Scheel-Toellner
- Department of Functional Biology, Immunology Area, Faculty of Medicine, University of Oviedo, Oviedo, Spain, Rheumatology Research Group, MRC Centre for Immune Regulation, Institute for Biomedical Research, School of Immunity and Infection, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK and Department of Internal Medicine, Hospital Universitario Central de Asturias, Oviedo, Spain
| | - Javier Rodríguez-Carrio
- Department of Functional Biology, Immunology Area, Faculty of Medicine, University of Oviedo, Oviedo, Spain, Rheumatology Research Group, MRC Centre for Immune Regulation, Institute for Biomedical Research, School of Immunity and Infection, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK and Department of Internal Medicine, Hospital Universitario Central de Asturias, Oviedo, Spain
| | - Luis Caminal-Montero
- Department of Functional Biology, Immunology Area, Faculty of Medicine, University of Oviedo, Oviedo, Spain, Rheumatology Research Group, MRC Centre for Immune Regulation, Institute for Biomedical Research, School of Immunity and Infection, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK and Department of Internal Medicine, Hospital Universitario Central de Asturias, Oviedo, Spain
| | - Caroline Gordon
- Department of Functional Biology, Immunology Area, Faculty of Medicine, University of Oviedo, Oviedo, Spain, Rheumatology Research Group, MRC Centre for Immune Regulation, Institute for Biomedical Research, School of Immunity and Infection, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK and Department of Internal Medicine, Hospital Universitario Central de Asturias, Oviedo, Spain
| | - Ana Suárez
- Department of Functional Biology, Immunology Area, Faculty of Medicine, University of Oviedo, Oviedo, Spain, Rheumatology Research Group, MRC Centre for Immune Regulation, Institute for Biomedical Research, School of Immunity and Infection, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK and Department of Internal Medicine, Hospital Universitario Central de Asturias, Oviedo, Spain
| |
Collapse
|
243
|
Zhu XJ, Shi Y, Zhang F, Yao QM, Liu YX, Shan NN, Wang D, Peng J, Xu J, Hou M. Reduced tumour necrosis factor receptor superfamily 13C inversely correlated with tumour necrosis factor superfamily 13B in patients with immune thrombocytopenia. Br J Haematol 2014; 166:783-91. [PMID: 24889407 DOI: 10.1111/bjh.12958] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2014] [Accepted: 04/01/2014] [Indexed: 01/06/2023]
Affiliation(s)
- Xiao-juan Zhu
- Department of Haematology; Provincial Hospital affiliated to Shandong University; Jinan China
| | - Yan Shi
- Department of Haematology; Qilu Hospital, Shandong University; Jinan China
- The Key Laboratory of Cardiovascular Remodeling and Function Research; Chinese Ministry of Education and Chinese Ministry of Health; Jinan China
| | - Feng Zhang
- Department of Haematology; Provincial Hospital affiliated to Shandong University; Jinan China
| | - Qing-min Yao
- Department of Haematology; Provincial Hospital affiliated to Shandong University; Jinan China
| | - Yan-xia Liu
- Department of Haematology; Provincial Hospital affiliated to Shandong University; Jinan China
| | - Ning-ning Shan
- Department of Haematology; Provincial Hospital affiliated to Shandong University; Jinan China
| | - Dan Wang
- Department of Research; Provincial Hospital affiliated to Shandong University; Jinan China
| | - Jun Peng
- Department of Haematology; Qilu Hospital, Shandong University; Jinan China
- The Key Laboratory of Cardiovascular Remodeling and Function Research; Chinese Ministry of Education and Chinese Ministry of Health; Jinan China
| | - Jian Xu
- Department of Haematology; Provincial Hospital affiliated to Shandong University; Jinan China
| | - Ming Hou
- Department of Haematology; Qilu Hospital, Shandong University; Jinan China
- The Key Laboratory of Cardiovascular Remodeling and Function Research; Chinese Ministry of Education and Chinese Ministry of Health; Jinan China
| |
Collapse
|
244
|
Liu XG, Hou M. Immune thrombocytopenia and B-cell-activating factor/a proliferation-inducing ligand. Semin Hematol 2014; 50 Suppl 1:S89-99. [PMID: 23664525 DOI: 10.1053/j.seminhematol.2013.03.021] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Primary immune thrombocytopenia (ITP) is an organ-specific autoimmune disorder characterized by autoantibody-mediated enhanced platelet destruction and dysmegakaryocytopoiesis. B cells have been demonstrated to play critical roles in the pathophysiology of ITP. B-cell-activating factor (BAFF) and a proliferation-inducing ligand (APRIL) are crucial cytokines supporting survival and differentiation of B cells, and dysregulation of BAFF/APRIL is involved in the pathogenesis of B-cell related autoimmune diseases including ITP. Currently ongoing clinical trials using BAFF and/or APRIL-blocking agents have yielded positive results in human systemic lupus erythematosus (SLE) and rheumatoid arthritis (RA), further confirming the pathological role of BAFF/APRIL in autoimmunity. This review will describe the function of BAFF/APRIL and address the feasibility of BAFF/APRIL inhibition in the management of ITP.
Collapse
Affiliation(s)
- Xin-guang Liu
- Department of Hematology, Qilu Hospital, Shandong University, Jinan, PR China
| | | |
Collapse
|
245
|
Abstract
B cell maturation antigen (BCMA) is a tumor necrosis family receptor (TNFR) member that is predominantly expressed on terminally differentiated B cells and, upon binding to its ligands B cell activator of the TNF family (BAFF) and a proliferation inducing ligand (APRIL), delivers pro-survival cell signals. Thus, BCMA is mostly known for its functional activity in mediating the survival of plasma cells that maintain long-term humoral immunity. The expression of BCMA has also been linked to a number of cancers, autoimmune disorders, and infectious diseases that suggest additional roles for BCMA activity. Despite recent advances in our understanding of the roles for the related TNFR members BAFF-R and transmembrane activator and calcium-modulator and cyclophilin ligand interactor (TACI), the signaling pathway used by BCMA for mediating plasma cell survival as well as its putative function in certain disease states are not well understood. By examining the expression, regulation, and signaling targets of BCMA, we may gain further insight into this receptor and how it operates within cells in both health and disease. This information is important for identifying new therapeutic targets that may be relevant in treating diseases that involve the BAFF/APRIL cytokine network.
Collapse
Affiliation(s)
- Christine M Coquery
- Department of Microbiology, Immunology, & Cancer Biology, University of Virginia School of Medicine, Charlottesville, VA 22908, USA
| | | |
Collapse
|
246
|
Chen SS, Chiorazzi N. Murine genetically engineered and human xenograft models of chronic lymphocytic leukemia. Semin Hematol 2014; 51:188-205. [PMID: 25048783 DOI: 10.1053/j.seminhematol.2014.05.001] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Chronic lymphocytic leukemia (CLL) is a genetically complex disease, with multiple factors having an impact on onset, progression, and response to therapy. Genetic differences/abnormalities have been found in hematopoietic stem cells from patients, as well as in B lymphocytes of individuals with monoclonal B-cell lymphocytosis who may develop the disease. Furthermore, after the onset of CLL, additional genetic alterations occur over time, often causing disease worsening and altering patient outcomes. Therefore, being able to genetically engineer mouse models that mimic CLL or at least certain aspects of the disease will help us understand disease mechanisms and improve treatments. This notwithstanding, because neither the genetic aberrations responsible for leukemogenesis and progression nor the promoting factors that support these are likely identical in character or influences for all patients, genetically engineered mouse models will only completely mimic CLL when all of these factors are precisely defined. In addition, multiple genetically engineered models may be required because of the heterogeneity in susceptibility genes among patients that can have an effect on genetic and environmental characteristics influencing disease development and outcome. For these reasons, we review the major murine genetically engineered and human xenograft models in use at the present time, aiming to report the advantages and disadvantages of each.
Collapse
Affiliation(s)
- Shih-Shih Chen
- The Feinstein Institute for Medical Research, North Shore-LIJ Health System, Manhasset, New York.
| | - Nicholas Chiorazzi
- The Feinstein Institute for Medical Research, North Shore-LIJ Health System, Manhasset, New York; Departments of Medicine and Molecular Medicine, Hofstra North Shore-LIJ School of Medicine, Manhasset, New York.
| |
Collapse
|
247
|
Reduced BAFF-R and increased TACI expression in common variable immunodeficiency. J Clin Immunol 2014; 34:573-83. [PMID: 24809296 DOI: 10.1007/s10875-014-0047-y] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2014] [Accepted: 04/15/2014] [Indexed: 10/25/2022]
Abstract
PURPOSE B-cell survival and differentiation critically depend on the interaction of BAFF-R and TACI with their ligands, BAFF and APRIL. Mature B-cell defects lead to Common Variable Immunodeficiency (CVID), which is associated with elevated serum levels of BAFF and APRIL. Nevertheless, BAFF-R and TACI expression in CVID and their relationship with ligand availability remain poorly understood. METHODS AND RESULTS We found that BAFF-R expression was dramatically reduced on B cells of CVID patients, relative to controls. BAFF-R levels inversely correlated with serum BAFF concentration both in CVID and healthy subjects. We also found that recombinant BAFF stimulation reduced BAFF-R expression on B cells without decreasing transcript levels. On the other hand, CVID subjects had increased TACI expression on B cells in direct association with serum BAFF but not APRIL levels. Moreover, splenomegaly was associated with higher TACI expression, suggesting that perturbations of TACI function may underlie lymphoproliferation in CVID. CONCLUSIONS Our results indicate that availability of BAFF determines BAFF-R and TACI expression on B cells, and that BAFF-R expression is controlled by BAFF binding. Identification of the factors governing BAFF-R and TACI is crucial to understanding CVID pathogenesis, and B-cell biology in general, as well as to explore their potential as therapeutic targets.
Collapse
|
248
|
Boghdadi G, Elewa EA. Increased serum APRIL differentially correlates with distinct cytokine profiles and disease activity in systemic lupus erythematosus patients. Rheumatol Int 2014; 34:1217-23. [PMID: 24748505 DOI: 10.1007/s00296-014-3020-4] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2014] [Accepted: 04/05/2014] [Indexed: 11/27/2022]
Abstract
Cytokines play an important role in the pathogenesis of systemic lupus erythematosus (SLE). Among the cytokines that regulate B cell homeostasis is a proliferation-inducing ligand (APRIL). This study aimed to determine whether serum levels of APRIL are raised in patients with SLE and correlate with disease activity or proinflammatory cytokines production, or both. Serum APRIL, interleukin-17 (IL-17), IL-4 and interferon gamma (IFN-γ) levels were measured in forty patients with SLE and 30 healthy controls. Disease activity was assessed by SLE disease activity index (SLEDAI), and results were correlated with serum APRIL levels. Serum APRIL levels were significantly higher in patients with SLE than in healthy controls. Positive correlation was found between serum APRIL levels and total SLEDAI score and anti-dsDNA antibody titers. Moreover, serum APRIL levels was significantly higher in patients with arthritis, mucocutaneous manifestations and proteinuria. APRIL is increased in patients with active SLE accompanying the increase of IL-17 and IFN-γ. Significant positive correlations between serum levels of APRIL and IL-17 and IFN-γ and a negative correlation between serum levels of APRIL and IL-4 were found. The results suggest that APRIL may be an important marker of disease activity in patients with SLE. We provide the analyses of APRIL levels in patients with SLE, suggesting new tools for the diagnosis, prognosis and possible therapeutic management of SLE.
Collapse
Affiliation(s)
- Ghada Boghdadi
- Immunology Research Lab, Department of Microbiology and Immunology, Faculty of Medicine, Zagazig University, Zagazig, Egypt,
| | | |
Collapse
|
249
|
Ma N, Xiao H, Marrero B, Xing C, Wang X, Zheng M, Han G, Chen G, Hou C, Shen B, Li Y, Wang R, Jiang Z. Combination of TACI-IgG and anti-IL-15 treats murine lupus by reducing mature and memory B cells. Cell Immunol 2014; 289:140-4. [PMID: 24791699 DOI: 10.1016/j.cellimm.2014.03.017] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2013] [Revised: 11/04/2013] [Accepted: 03/31/2014] [Indexed: 12/11/2022]
Abstract
Clinical trials suggest that BAFF inhibitors such as atacicept (TACI-IgG) and belimumab (anti-BAFF antibody) could not reduce memory B-cell numbers, although they reduced the numbers of CD20(+) naïve B cells and activated B cells. In the present study, we explored the way to reduce memory B-cell numbers. First, we used TACI-IgG to treat murine lupus. We found that TACI-IgG was effective in reducing mature B cell numbers. Accordingly it controlled the level of the anti-dsDNA antibody in lupus-like mice. In addition, TACI-IgG up-regulated memory B cells in murine lupus. Furthermore, we found that TACI-IgG up-regulated IL-15 expression in lupus-like mice. Thus, the combination of TACI-IgG and anti-IL-15 antibodies was explored to understand their effects on the treatment of murine lupus. Compared to treatments with TACI-IgG or anti-IL-15 alone, the combination of TACI-IgG and anti-IL-15 antibodies efficiently ameliorated murine lupus phenotypes. The study provides hints for the clinical application of BAFF- and IL-15-specific therapeutic agents.
Collapse
Affiliation(s)
- Ning Ma
- Department of Rheumatology, First Hospital of Jilin University, Changchun 130021, China; Laboratory of Immunology, Institute of Basic Medical Sciences, Beijing 100850, China
| | - He Xiao
- Laboratory of Immunology, Institute of Basic Medical Sciences, Beijing 100850, China
| | - Bernadette Marrero
- Molecular Immunology Section, Laboratory of Immunology, National Eye Institute, National Institutes of Health, Bethesda, MD 20892-1857, USA
| | - Chen Xing
- Laboratory of Immunology, Institute of Basic Medical Sciences, Beijing 100850, China
| | - Xiaoqian Wang
- Laboratory of Immunology, Institute of Basic Medical Sciences, Beijing 100850, China
| | - Mingke Zheng
- Department of Immunology, Medical College of Henan University, Kaifeng 475001, China
| | - Gencheng Han
- Laboratory of Immunology, Institute of Basic Medical Sciences, Beijing 100850, China
| | - Guojiang Chen
- Laboratory of Immunology, Institute of Basic Medical Sciences, Beijing 100850, China
| | - Chunmei Hou
- Laboratory of Immunology, Institute of Basic Medical Sciences, Beijing 100850, China
| | - Beifen Shen
- Laboratory of Immunology, Institute of Basic Medical Sciences, Beijing 100850, China
| | - Yan Li
- Laboratory of Immunology, Institute of Basic Medical Sciences, Beijing 100850, China
| | - Renxi Wang
- Laboratory of Immunology, Institute of Basic Medical Sciences, Beijing 100850, China
| | - Zhenyu Jiang
- Department of Rheumatology, First Hospital of Jilin University, Changchun 130021, China.
| |
Collapse
|
250
|
Scheinberg MA, Srinivasan D, Martin RS. The potential role of blisibimod for the treatment of systemic lupus erythematosus. ACTA ACUST UNITED AC 2014. [DOI: 10.2217/ijr.14.7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|