1
|
Cao L, Zhang R, Wang Y, Hu X, Yong L, Li B, Ge H, Chen W, Zhen Q, Yu Y, Mao Y, Li Z, Fan W, Sun L. Fine Mapping Analysis of the MHC Region to Identify Variants Associated With Chinese Vitiligo and SLE and Association Across These Diseases. Front Immunol 2022; 12:758652. [PMID: 35082778 PMCID: PMC8784546 DOI: 10.3389/fimmu.2021.758652] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2021] [Accepted: 12/20/2021] [Indexed: 11/13/2022] Open
Abstract
The important role of MHC in the pathogenesis of vitiligo and SLE has been confirmed in various populations. To map the most significant MHC variants associated with the risk of vitiligo and SLE, we conducted fine mapping analysis using 1117 vitiligo cases, 1046 SLE cases and 1693 healthy control subjects in the Han-MHC reference panel and 1000 Genomes Project phase 3. rs113465897 (P=1.03×10-13, OR=1.64, 95%CI =1.44–1.87) and rs3129898 (P=4.21×10-17, OR=1.93, 95%CI=1.66–2.25) were identified as being most strongly associated with vitiligo and SLE, respectively. Stepwise conditional analysis revealed additional independent signals at rs3130969(p=1.48×10-7, OR=0.69, 95%CI=0.60–0.79), HLA-DPB1*03:01 (p=1.07×10-6, OR=1.94, 95%CI=1.49–2.53) being linked to vitiligo and HLA-DQB1*0301 (P=4.53×10-7, OR=0.62, 95%CI=0.52-0.75) to SLE. Considering that epidemiological studies have confirmed comorbidities of vitiligo and SLE, we used the GCTA tool to analyse the genetic correlation between these two diseases in the HLA region, the correlation coefficient was 0.79 (P=5.99×10-10, SE=0.07), confirming their similar genetic backgrounds. Our findings highlight the value of the MHC region in vitiligo and SLE and provide a new perspective for comorbidities among autoimmune diseases.
Collapse
Affiliation(s)
- Lu Cao
- Department of Dermatology, The First Affiliated Hospital of Anhui Medical University, Hefei, China.,Institute of Dermatology, Anhui Medical University, Hefei, China.,Key Laboratory of Dermatology, Anhui Medical University, Ministry of Education, Hefei, China.,Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Medical University, Hefei, China.,Anhui Provincial Institute of Translational Medicine, Anhui Medical University, Hefei, China
| | - Ruixue Zhang
- Department of Dermatology, The First Affiliated Hospital of Anhui Medical University, Hefei, China.,Institute of Dermatology, Anhui Medical University, Hefei, China.,Key Laboratory of Dermatology, Anhui Medical University, Ministry of Education, Hefei, China.,Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Medical University, Hefei, China.,Anhui Provincial Institute of Translational Medicine, Anhui Medical University, Hefei, China
| | - Yirui Wang
- Department of Dermatology, The First Affiliated Hospital of Anhui Medical University, Hefei, China.,Institute of Dermatology, Anhui Medical University, Hefei, China.,Key Laboratory of Dermatology, Anhui Medical University, Ministry of Education, Hefei, China.,Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Medical University, Hefei, China.,Anhui Provincial Institute of Translational Medicine, Anhui Medical University, Hefei, China
| | - Xia Hu
- Department of Dermatology, The First Affiliated Hospital of Anhui Medical University, Hefei, China.,Institute of Dermatology, Anhui Medical University, Hefei, China.,Key Laboratory of Dermatology, Anhui Medical University, Ministry of Education, Hefei, China.,Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Medical University, Hefei, China.,Anhui Provincial Institute of Translational Medicine, Anhui Medical University, Hefei, China
| | - Liang Yong
- Department of Dermatology, The First Affiliated Hospital of Anhui Medical University, Hefei, China.,Institute of Dermatology, Anhui Medical University, Hefei, China.,Key Laboratory of Dermatology, Anhui Medical University, Ministry of Education, Hefei, China.,Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Medical University, Hefei, China.,Anhui Provincial Institute of Translational Medicine, Anhui Medical University, Hefei, China
| | - Bao Li
- The Comprehensive Lab, College of Basic Medicine, Anhui Medical University, Hefei, China
| | - Huiyao Ge
- Department of Dermatology, The First Affiliated Hospital of Anhui Medical University, Hefei, China.,Institute of Dermatology, Anhui Medical University, Hefei, China.,Key Laboratory of Dermatology, Anhui Medical University, Ministry of Education, Hefei, China.,Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Medical University, Hefei, China.,Anhui Provincial Institute of Translational Medicine, Anhui Medical University, Hefei, China
| | - Weiwei Chen
- Department of Dermatology, The First Affiliated Hospital of Anhui Medical University, Hefei, China.,Institute of Dermatology, Anhui Medical University, Hefei, China.,Key Laboratory of Dermatology, Anhui Medical University, Ministry of Education, Hefei, China.,Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Medical University, Hefei, China.,Anhui Provincial Institute of Translational Medicine, Anhui Medical University, Hefei, China
| | - Qi Zhen
- Department of Dermatology, The First Affiliated Hospital of Anhui Medical University, Hefei, China.,Institute of Dermatology, Anhui Medical University, Hefei, China.,Key Laboratory of Dermatology, Anhui Medical University, Ministry of Education, Hefei, China.,Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Medical University, Hefei, China.,Anhui Provincial Institute of Translational Medicine, Anhui Medical University, Hefei, China
| | - Yafen Yu
- Department of Dermatology, The First Affiliated Hospital of Anhui Medical University, Hefei, China.,Institute of Dermatology, Anhui Medical University, Hefei, China.,Key Laboratory of Dermatology, Anhui Medical University, Ministry of Education, Hefei, China.,Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Medical University, Hefei, China.,Anhui Provincial Institute of Translational Medicine, Anhui Medical University, Hefei, China
| | - Yiwen Mao
- Department of Dermatology, The First Affiliated Hospital of Anhui Medical University, Hefei, China.,Institute of Dermatology, Anhui Medical University, Hefei, China.,Key Laboratory of Dermatology, Anhui Medical University, Ministry of Education, Hefei, China.,Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Medical University, Hefei, China.,Anhui Provincial Institute of Translational Medicine, Anhui Medical University, Hefei, China
| | - Zhuo Li
- Department of Dermatology, The First Affiliated Hospital of Anhui Medical University, Hefei, China.,Institute of Dermatology, Anhui Medical University, Hefei, China.,Key Laboratory of Dermatology, Anhui Medical University, Ministry of Education, Hefei, China.,Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Medical University, Hefei, China.,Anhui Provincial Institute of Translational Medicine, Anhui Medical University, Hefei, China
| | - Wencheng Fan
- Department of Dermatology, The First Affiliated Hospital of Anhui Medical University, Hefei, China.,Institute of Dermatology, Anhui Medical University, Hefei, China.,Key Laboratory of Dermatology, Anhui Medical University, Ministry of Education, Hefei, China.,Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Medical University, Hefei, China.,Anhui Provincial Institute of Translational Medicine, Anhui Medical University, Hefei, China
| | - Liangdan Sun
- Department of Dermatology, The First Affiliated Hospital of Anhui Medical University, Hefei, China.,Institute of Dermatology, Anhui Medical University, Hefei, China.,Key Laboratory of Dermatology, Anhui Medical University, Ministry of Education, Hefei, China.,Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Medical University, Hefei, China.,Anhui Provincial Institute of Translational Medicine, Anhui Medical University, Hefei, China
| |
Collapse
|
2
|
Zhao Z, Anderson AN, Kannapell CC, Kwok WW, Gaskin F, Fu SM. HLA-DR3 restricted environmental epitopes from the bacterium Clostridium tetani have T cell cross-reactivity to the SLE-related autoantigen SmD. Front Immunol 2022; 13:928374. [PMID: 36389825 PMCID: PMC9659850 DOI: 10.3389/fimmu.2022.928374] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Accepted: 10/14/2022] [Indexed: 11/23/2022] Open
Abstract
HLA-DR3 (DR3) is one of the dominant HLA-DR alleles associated with systemic lupus erythematosus (SLE) susceptibility. Our previous studies showed multiple intramolecular DR3 restricted T cell epitopes in the Smith D (SmD) protein, from which we generated a non-homologous, bacterial epitope mimics library. From this library we identified ABC247-261 Mimic as one new DR3 restricted bacterial T cell epitope from the ABC transporter ATP-binding protein in Clostridium tetani. It activated and induced autoreactive SmD66-80-specific T cells and induced autoantibodies to lupus-related autoantigens in vivo. Compared to healthy donors, SLE patients have a greater percentage of cross-reactive T cells to ABC247-261 Mimic and SmD66-80. In addition, we analyzed the ability of single DR3 restricted Tetanus toxoid (TT) T cell epitopes to induce autoimmune T cells. We found that the immunodominant TT epitope TT826-845 stimulated SmD66-80 reactive T cells but failed to induce persistent anti-SmD autoantibodies compared to the ABC247-261 Mimic. Thus, exposure to the ABC247-261 Mimic epitope may contribute to autoimmunity in susceptible DR3 individuals.
Collapse
Affiliation(s)
- Zhenhuan Zhao
- Division of Rheumatology, Department of Medicine, University of Virginia, Charlottesville, VA, United States.,Center for Immunity, Inflammation and Regenerative Medicine, Division of Nephrology, Department of Medicine, University of Virginia, Charlottesville, VA, United States
| | - Ashley N Anderson
- Center for Immunity, Inflammation and Regenerative Medicine, Division of Nephrology, Department of Medicine, University of Virginia, Charlottesville, VA, United States
| | - Carol C Kannapell
- Division of Rheumatology, Department of Medicine, University of Virginia, Charlottesville, VA, United States.,Center for Immunity, Inflammation and Regenerative Medicine, Division of Nephrology, Department of Medicine, University of Virginia, Charlottesville, VA, United States
| | - William W Kwok
- Benaroya Research Institute, Virginia Mason, Seattle, WA, United States
| | - Felicia Gaskin
- Department of Psychiatry and Neurobehavioral Sciences, University of Virginia, Charlottesville, VA, United States
| | - Shu Man Fu
- Division of Rheumatology, Department of Medicine, University of Virginia, Charlottesville, VA, United States.,Center for Immunity, Inflammation and Regenerative Medicine, Division of Nephrology, Department of Medicine, University of Virginia, Charlottesville, VA, United States
| |
Collapse
|
3
|
Kheyri Z, Laripour A, Ala M. Peritonitis as the first presentation of systemic lupus erythematous: a case report. J Med Case Rep 2021; 15:611. [PMID: 34953487 PMCID: PMC8710242 DOI: 10.1186/s13256-021-03216-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Accepted: 11/22/2021] [Indexed: 12/27/2022] Open
Abstract
Background Systemic lupus erythematosus is an inflammatory disease affecting several organs. Serositis is one of the systemic lupus erythematosus presentations, but peritonitis is a relatively rare presentation. Particularly, it is extremely rare to observe peritonitis as the first presentation of systemic lupus erythematosus. Case presentation Here, we present a case of peritonitis without other symptoms of systemic lupus erythematosus, in a patient who was finally diagnosed with systemic lupus erythematosus. Our patient was a 27-year-old Persian/Caucasian male with fatigue, weakness, weight loss, abdominal distension, massive ascites, and normocytic hemolytic anemia. He did not mention any prior medical conditions and did not use any drugs. There were no signs of thyroid dysfunction, cardiac dysfunction, cancers, infectious diseases, hepatitis, kidney diseases, or other diseases. Low-gradient, high-protein ascites fluid, and positive antinuclear antibody and anti-double stranded DNA were in favor of systemic lupus erythematosus. Corticosteroid pulse therapy led to resolution of ascites, and the patient was discharged with prednisolone and hydroxychloroquine. Conclusion Peritonitis is a rare presentation of systemic lupus erythematosus, particularly as the first presentation and in the absence of other signs and symptoms; however, systemic lupus erythematosus should be considered as one the differential diagnoses for peritonitis when other etiologies have been ruled out.
Collapse
Affiliation(s)
- Zahedin Kheyri
- Baharloo Hospital, Tehran University of Medical Sciences (TUMS), Tehran, Iran.,Department of Gastroenterology, Tehran University of Medical Sciences (TUMS), Tehran, Iran
| | - Amirreza Laripour
- Students' Research Committee, Arak University of Medical Sciences, Arak, Iran
| | - Moein Ala
- School of Medicine, Tehran University of Medical Sciences (TUMS), Tehran, Iran.
| |
Collapse
|
4
|
Nogueira Almeida L, Clauder AK, Meng L, Ehlers M, Arce S, Manz RA. MHC haplotype and B cell autoimmunity: Correlation with pathogenic IgG autoantibody subclasses and Fc glycosylation patterns. Eur J Immunol 2021; 52:197-203. [PMID: 34609741 DOI: 10.1002/eji.202149279] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 08/20/2021] [Accepted: 09/28/2021] [Indexed: 12/21/2022]
Abstract
Genome-wide association studies (GWAS) have identified many genes that are associated with the development of certain autoimmune disorders, but the MHC haplotypes still represent the most prevalent genetic risk factor for many autoimmune diseases. The mechanisms by which MHC-associated genetic susceptibility translates into B cell autoimmunity and the development of autoimmune diseases are complex. There is increasing evidence that the MHC haplotype modulates autoreactive B cell responses in multiple ways. Instead of merely inhibiting the production of IgG autoantibodies and mediating complete immunological tolerance, the non-permitting MHC haplotypes seem to facilitate the production of IgG autoantibodies exhibiting Fc glycosylation patterns that are associated with reduced pathogenicity and a protective cytokine profile of T follicular helper (Tfh) cells. Here, we discuss mechanisms linking MHC haplotypes to the production of pathogenic IgG autoantibodies, which could be relevant for the development of improved diagnosis, particularly in the context of individual medicine.
Collapse
Affiliation(s)
| | - Ann-Katrin Clauder
- Institute for Systemic Inflammation Research, University of Lübeck, Lübeck, Germany
| | - Lingzhang Meng
- Center for Systemic Inflammation Research (CSIR), School of Preclinical Medicine, Youjiang Medical University for Nationalities, Baise City (Bose), Guangxi Zhuang, Autonomous Region, China
| | - Marc Ehlers
- Laboratories of Immunology and Antibody Glycan Analysis, Institute for Nutritional Medicine, University of Lübeck and University Medical Center Schleswig-Holstein, Lübeck, Germany
| | - Sergio Arce
- Department of Biomedical Sciences, University of South Carolina School of Medicine-Greenville, Greenville, SC, USA
| | - Rudolf Armin Manz
- Institute for Systemic Inflammation Research, University of Lübeck, Lübeck, Germany
| |
Collapse
|
5
|
Bachmann MP, Bartsch T, Bippes CC, Bachmann D, Puentes-Cala E, Bachmann J, Bartsch H, Arndt C, Koristka S, Loureiro LR, Kegler A, Laube M, Gross JK, Gross T, Kurien BT, Scofield RH, Farris AD, James JA, Schmitz M, Feldmann A. T Cell Mediated Conversion of a Non-Anti-La Reactive B Cell to an Autoreactive Anti-La B Cell by Somatic Hypermutation. Int J Mol Sci 2021; 22:1198. [PMID: 33530489 PMCID: PMC7865296 DOI: 10.3390/ijms22031198] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Revised: 01/20/2021] [Accepted: 01/22/2021] [Indexed: 12/18/2022] Open
Abstract
Since the first description of nuclear autoantigens in the late 1960s and early 1970s, researchers, including ourselves, have found it difficult to establish monoclonal antibodies (mabs) against nuclear antigens, including the La/SS-B (Sjögrens' syndrome associated antigen B) autoantigen. To date, only a few anti-La mabs have been derived by conventional hybridoma technology; however, those anti-La mabs were not bona fide autoantibodies as they recognize either human La specific, cryptic, or post-translationally modified epitopes which are not accessible on native mouse La protein. Herein, we present a series of novel murine anti-La mabs including truly autoreactive ones. These mabs were elicited from a human La transgenic animal through adoptive transfer of T cells from non-transgenic mice immunized with human La antigen. Detailed epitope and paratope analyses experimentally confirm the hypothesis that somatic hypermutations that occur during T cell dependent maturation can lead to autoreactivity to the nuclear La/SS-B autoantigen.
Collapse
Affiliation(s)
- Michael P. Bachmann
- Department of Radioimmunology, Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), 03128 Dresden, Germany; (T.B.); (E.P.-C.); (C.A.); (S.K.); (L.R.L.); (A.K.); (M.L.); (A.F.)
- University Cancer Center (UCC), Tumor Immunology, University Hospital Carl Gustav Carus Dresden, Technical University Dresden, 01307 Dresden, Germany; (D.B.); (J.B.)
- National Center for Tumor Diseases (NCT), 01307 Dresden, Germany
- German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Tabea Bartsch
- Department of Radioimmunology, Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), 03128 Dresden, Germany; (T.B.); (E.P.-C.); (C.A.); (S.K.); (L.R.L.); (A.K.); (M.L.); (A.F.)
| | - Claudia C. Bippes
- Institute of Immunology, Medical Faculty Carl Gustav Carus Dresden, Technical University Dresden, 01307 Dresden, Germany; (C.C.B.); (H.B.); (M.S.)
| | - Dominik Bachmann
- University Cancer Center (UCC), Tumor Immunology, University Hospital Carl Gustav Carus Dresden, Technical University Dresden, 01307 Dresden, Germany; (D.B.); (J.B.)
| | - Edinson Puentes-Cala
- Department of Radioimmunology, Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), 03128 Dresden, Germany; (T.B.); (E.P.-C.); (C.A.); (S.K.); (L.R.L.); (A.K.); (M.L.); (A.F.)
- Corporación para la Investigación de la Corrosión (CIC), Piedecuesta, Santander 681011, Colombia
| | - Jennifer Bachmann
- University Cancer Center (UCC), Tumor Immunology, University Hospital Carl Gustav Carus Dresden, Technical University Dresden, 01307 Dresden, Germany; (D.B.); (J.B.)
| | - Holger Bartsch
- Institute of Immunology, Medical Faculty Carl Gustav Carus Dresden, Technical University Dresden, 01307 Dresden, Germany; (C.C.B.); (H.B.); (M.S.)
| | - Claudia Arndt
- Department of Radioimmunology, Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), 03128 Dresden, Germany; (T.B.); (E.P.-C.); (C.A.); (S.K.); (L.R.L.); (A.K.); (M.L.); (A.F.)
| | - Stefanie Koristka
- Department of Radioimmunology, Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), 03128 Dresden, Germany; (T.B.); (E.P.-C.); (C.A.); (S.K.); (L.R.L.); (A.K.); (M.L.); (A.F.)
| | - Liliana R. Loureiro
- Department of Radioimmunology, Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), 03128 Dresden, Germany; (T.B.); (E.P.-C.); (C.A.); (S.K.); (L.R.L.); (A.K.); (M.L.); (A.F.)
| | - Alexandra Kegler
- Department of Radioimmunology, Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), 03128 Dresden, Germany; (T.B.); (E.P.-C.); (C.A.); (S.K.); (L.R.L.); (A.K.); (M.L.); (A.F.)
| | - Markus Laube
- Department of Radioimmunology, Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), 03128 Dresden, Germany; (T.B.); (E.P.-C.); (C.A.); (S.K.); (L.R.L.); (A.K.); (M.L.); (A.F.)
| | - Joanne K. Gross
- The Arthritis and Clinical Immunology Program, Oklahoma Medical Research Foundation and University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; (J.K.G.); (T.G.); (B.T.K.); (R.H.S.); (A.D.F.); (J.A.J.)
| | - Tim Gross
- The Arthritis and Clinical Immunology Program, Oklahoma Medical Research Foundation and University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; (J.K.G.); (T.G.); (B.T.K.); (R.H.S.); (A.D.F.); (J.A.J.)
| | - Biji T. Kurien
- The Arthritis and Clinical Immunology Program, Oklahoma Medical Research Foundation and University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; (J.K.G.); (T.G.); (B.T.K.); (R.H.S.); (A.D.F.); (J.A.J.)
| | - R. Hal Scofield
- The Arthritis and Clinical Immunology Program, Oklahoma Medical Research Foundation and University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; (J.K.G.); (T.G.); (B.T.K.); (R.H.S.); (A.D.F.); (J.A.J.)
| | - A. Darise Farris
- The Arthritis and Clinical Immunology Program, Oklahoma Medical Research Foundation and University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; (J.K.G.); (T.G.); (B.T.K.); (R.H.S.); (A.D.F.); (J.A.J.)
| | - Judith A. James
- The Arthritis and Clinical Immunology Program, Oklahoma Medical Research Foundation and University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; (J.K.G.); (T.G.); (B.T.K.); (R.H.S.); (A.D.F.); (J.A.J.)
| | - Marc Schmitz
- National Center for Tumor Diseases (NCT), 01307 Dresden, Germany
- German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
- Institute of Immunology, Medical Faculty Carl Gustav Carus Dresden, Technical University Dresden, 01307 Dresden, Germany; (C.C.B.); (H.B.); (M.S.)
| | - Anja Feldmann
- Department of Radioimmunology, Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), 03128 Dresden, Germany; (T.B.); (E.P.-C.); (C.A.); (S.K.); (L.R.L.); (A.K.); (M.L.); (A.F.)
| |
Collapse
|
6
|
Pan Q, Xiao H, Shi L, He Y, Cai J, Wu J, Li A, Ye L, Yang C, Liu HF. IgG4 Autoantibodies Attenuate Systemic Lupus Erythematosus Progression by Suppressing Complement Consumption and Inflammatory Cytokine Production. Front Immunol 2020; 11:1047. [PMID: 32625200 PMCID: PMC7311789 DOI: 10.3389/fimmu.2020.01047] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Accepted: 04/30/2020] [Indexed: 01/08/2023] Open
Abstract
Pathogenic autoantibodies can cause inflammation and tissue injury in systemic lupus erythematosus (SLE). Although IgG4 is considered non-inflammatory owing to the unique structure of its hinge region, the role of IgG4 autoantibodies in SLE remains largely unknown. The titers of serum anti-nuclear-IgG antibodies (ANA-IgG) and anti-nuclear-IgG4 antibodies (ANA-IgG4) in newly diagnosed SLE patients were detected. The effects of IgG4 purified from SLE patients (SLE IgG4) and healthy controls on complement consumption and inflammatory cytokine production were evaluated in vitro. The therapeutic effects of mouse IgG1 (functionally resembles human IgG4) purified from lupus-prone MRL-lpr/lpr mice (lupus IgG1) and control mice on disease progression were examined in MRL-lpr/lpr mice. The results showed that SLE patients with equal titers of total serum ANA-IgG (1:3,200) were divided into group I with lower ANA-IgG4 titers (≤ 1:10) and group II with higher ANA-IgG4 titers (≥ 1:100), and disease activity, inflammatory cytokine production, complement consumption, and renal-function parameters in group I SLE patients were more severe than those in group II. Further, compared with control IgG4, SLE IgG4 inhibited complement consumption by autoantibody-autoantigen immune complexes, and also inhibited inflammatory cytokines production by SLE PBMCs in vitro. Moreover, compared with control IgG1, lupus IgG1 exhibited a therapeutic effect on lupus by attenuating disease progression in MRL-lpr/lpr mice. These findings, for the first time, suggest that IgG4 autoantibodies can attenuate SLE progression by suppressing complement consumption and inflammatory cytokine production. Hence, this study may provide novel therapeutic strategies against SLE and other autoimmune diseases.
Collapse
Affiliation(s)
- Qingjun Pan
- Key Laboratory of Prevention and Management of Chronic Kidney Disease of Zhanjiang City, Institute of Nephrology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Haiyan Xiao
- Department of Cellular Biology and Anatomy, Augusta University, Augusta, GA, United States
| | - Lei Shi
- Key Laboratory of Prevention and Management of Chronic Kidney Disease of Zhanjiang City, Institute of Nephrology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Yiming He
- Key Laboratory of Prevention and Management of Chronic Kidney Disease of Zhanjiang City, Institute of Nephrology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Jun Cai
- Key Laboratory of Prevention and Management of Chronic Kidney Disease of Zhanjiang City, Institute of Nephrology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Jing Wu
- Key Laboratory of Prevention and Management of Chronic Kidney Disease of Zhanjiang City, Institute of Nephrology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Aifen Li
- Key Laboratory of Prevention and Management of Chronic Kidney Disease of Zhanjiang City, Institute of Nephrology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Lin Ye
- Key Laboratory of Prevention and Management of Chronic Kidney Disease of Zhanjiang City, Institute of Nephrology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Chen Yang
- Key Laboratory of Prevention and Management of Chronic Kidney Disease of Zhanjiang City, Institute of Nephrology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Hua-Feng Liu
- Key Laboratory of Prevention and Management of Chronic Kidney Disease of Zhanjiang City, Institute of Nephrology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| |
Collapse
|
7
|
Zhao Z, Ren J, Dai C, Kannapell CC, Wang H, Gaskin F, Fu SM. Nature of T cell epitopes in lupus antigens and HLA-DR determines autoantibody initiation and diversification. Ann Rheum Dis 2019; 78:380-390. [PMID: 30254034 PMCID: PMC6377330 DOI: 10.1136/annrheumdis-2018-214125] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2018] [Revised: 08/14/2018] [Accepted: 08/19/2018] [Indexed: 02/02/2023]
Abstract
OBJECTIVES The generation of systemic lupus erythematosus (SLE)-related autoantibodies have been shown to be T cell dependent and antigen driven with HLA-DR restriction. In this study, the initiating antigen(s) and the mechanism of autoantibody diversification were investigated. METHODS T cell epitopes (T-epitopes) of SmD1 (SmD) were mapped by T-T hybridomas generated from DR3+AE0 mice immunised with SmD and with SmD overlapping peptides. TCRs from the reactive hybridomas were sequenced. The core epitopes were determined. Bacterial mimics were identified by bioinformatics. Sera from DR3+AE0 mice immunised with SmD peptides and their mimics were analysed for their reactivity by ELISA and immunohistochemistry. Samples of blood donors were analysed for HLA-DR and autoantibody specificities. RESULTS Multiple HLA-DR3 restricted T-epitopes within SmD were identified. Many T-T hybridomas reacted with more than one epitope. Some of them were cross-reactive with other snRNP peptides and with proteins in the Ro60/La/Ro52 complex. The reactive hybridomas used unique TCRs. Multiple T-epitope mimics were identified in commensal and environmental bacteria. Certain bacterial mimics shared both T and B cell epitopes with the related SmD peptide. Bacterial mimics induced autoantibodies to lupus-related antigens and to different tissues. HLA-DR3+ blood donors made significantly more SLE-related autoantibodies. CONCLUSIONS The unique antigenic structures of the lupus-related autoantigens provide the basis for being targeted and for T and B cell epitope spreading and autoantibody diversification with unique patterns. SLE-related autoantibodies are likely generated from responses to commensal and/or environmental microbes due to incomplete negative selection for autoreactive T cells. The production of SLE-related antibodies is inevitable in normal individuals. The findings in this investigation have significant implications in autoimmunity in general.
Collapse
Affiliation(s)
- Zhenhuan Zhao
- Division of Rheumatology, Department of Medicine, University of Virginia, Charlottesville, Virginia, USA
| | - Jiling Ren
- Division of Rheumatology, Department of Medicine, University of Virginia, Charlottesville, Virginia, USA
| | - Chao Dai
- Division of Rheumatology, Department of Medicine, University of Virginia, Charlottesville, Virginia, USA
| | - Carol C Kannapell
- Division of Rheumatology, Department of Medicine, University of Virginia, Charlottesville, Virginia, USA
| | - Hongyang Wang
- Division of Rheumatology, Department of Medicine, University of Virginia, Charlottesville, Virginia, USA
| | - Felicia Gaskin
- Department of Psychiatry and Neurobehavioral Sciences, University of Virginia, Charlottesville, Virginia, USA
| | - Shu Man Fu
- Division of Rheumatology, Department of Medicine, University of Virginia, Charlottesville, Virginia, USA
- Department of Microbiology, Immunology and Cancer Biology, University of Virginia, Charlottesville, Virginia, USA
| |
Collapse
|
8
|
Bundhun PK, Soogund MZS, Huang F. Arterial/venous thrombosis, fetal loss and stillbirth in pregnant women with systemic lupus erythematosus versus primary and secondary antiphospholipid syndrome: a systematic review and meta-analysis. BMC Pregnancy Childbirth 2018; 18:212. [PMID: 29879927 PMCID: PMC5992885 DOI: 10.1186/s12884-018-1850-x] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2018] [Accepted: 05/25/2018] [Indexed: 01/17/2023] Open
Abstract
BACKGROUND We aimed to systematically compare arterial/venous thrombosis, fetal loss and stillbirth in pregnant women with systemic lupus erythematosus (SLE), primary anti-phospholipid syndrome (PAPS) and secondary anti-phospholipid syndrome (SAPS). METHODS Online databases were carefully searched for relevant publications comparing SLE with PAPS and/or SAPS in pregnancy. Studies were included if: they compared SLE with APS [SLE versus PAPS or SLE versus SAPS or SLE versus PAPS and SAPS respectively] in pregnant women; and they reported specific adverse outcomes as their clinical endpoints including arterial/venous thrombosis, fetal loss and stillbirth. Risk ratios (RR) with 95% confidence intervals (CIs) were used as statistical parameters and the analysis was carried out by the RevMan 5.3 software. RESULTS A total number of 941 pregnant women were included: 556 were candidates of SLE; 200 were candidates of PAPS; and 185 were candidates of SAPS. APS was associated with a significantly higher risk of fetal loss (RR: 4.49, 95% CI: 2.09-9.64; P = 0.0001). In addition, stillbirth and arterial/venous thrombosis were also significantly increased with APS (RR: 6.65, 95% CI: 2.14-20.60; P = 0.001) and (RR: 3.95, 95% CI: 1.28-12.16; P = 0.02) respectively. When patients with PAPS were compared with patients who suffered from SLE alone, fetal loss and arterial/venous thrombosis were still significantly higher with the former. When SAPS were compared with SLE (without anti-phospholipid antibodies), arterial/venous thrombosis, stillbirth and fetal loss were still significantly higher with SAPS. However, no significant difference was observed in arterial/venous thrombosis and fetal loss between PAPS and SAPS. CONCLUSIONS PAPS and SAPS were associated with significantly higher arterial/venous thrombosis, fetal loss and stillbirth in comparison to SLE. However, no significant difference was observed when PAPS was compared to SAPS.
Collapse
Affiliation(s)
- Pravesh Kumar Bundhun
- Department of Internal Medicine, the First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021 People’s Republic of China
| | | | - Feng Huang
- Institute of Cardiovascular Diseases and Guangxi Key Laboratory Base of Precision Medicine in Cardio-cerebrovascular Diseases Control and Prevention, the First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021 People’s Republic of China
| |
Collapse
|
9
|
Li X, Li H, Hu Q, Lin J, Zhang Q, Li Y, Li J, Chen T, Zhang Q, Qiu Y. Detection of epitopes in systemic lupus erythematosus using peptide microarray. Mol Med Rep 2018. [PMID: 29532871 PMCID: PMC5928640 DOI: 10.3892/mmr.2018.8710] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Systemic lupus erythematosus (SLE) is a common autoimmune disease, which features the secretion of antibodies directed against autoantigens in vivo. In the present study, a peptide microarray was developed to detect the epitopes recognized by autoantibodies in patients with SLE for an effective method of diagnosis. SLE-associated epitopes in 14 autoantigens were predicted using the antigenic epitope prediction software DNA star. Peptides were synthesized based on the predicted antigenic epitopes and immobilized on a slide surface and developed into a peptide microarray. Using this peptide microarray the autoantibodies in 120 patients with SLE and 110 healthy subjects were detected. A total of 73 potential antigenic epitopes in 14 autoantigens were predicted and screened. The peptide microarray based on the 73 epitopes was used to detect the autoantibodies in patients with SLE. A total of 14 epitopes with potential diagnostic values were screened out. The sensitivity and specificity of the 14 epitopes for the diagnosis of SLE were 71.6 and 85.8%, respectively. An optimal set of epitopes for SLE diagnosis was obtained. As individual patients had a specific autoantibody spectrum it was possible to detect autoantibodies in SLE and perform the diagnosis of SLE using the peptide microarray.
Collapse
Affiliation(s)
- Xin Li
- Clinical Laboratory of Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Haixia Li
- Clinical Laboratory of Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Qiongdan Hu
- Department of Nephrology, The Traditional Chinese Medicine Hospital, Affiliated to Southwest Medical University, Luzhou, Sichuan 646000, P.R. China
| | - Jinfei Lin
- South China Institute of Microbial Ecology and Health, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Qiong Zhang
- Clinical Laboratory of Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Yao Li
- Clinical Laboratory of Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Juan Li
- Department of Rheumatology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Tao Chen
- South China Institute of Microbial Ecology and Health, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Qiong Zhang
- Department of Nephrology, The Traditional Chinese Medicine Hospital, Affiliated to Southwest Medical University, Luzhou, Sichuan 646000, P.R. China
| | - Yurong Qiu
- Clinical Laboratory of Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| |
Collapse
|
10
|
Hachicha H, Kammoun A, Mahfoudh N, Marzouk S, Feki S, Fakhfakh R, Fourati H, Haddouk S, Frikha F, Gaddour L, Hakim F, Bahloul Z, Makni H, Masmoudi H. Human leukocyte antigens-DRB1*03 is associated with systemic lupus erythematosus and anti-SSB production in South Tunisia. Int J Health Sci (Qassim) 2018; 12:21-27. [PMID: 29623013 PMCID: PMC5870315] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022] Open
Abstract
INTRODUCTION Systemic lupus erythematosus (SLE) is an autoimmune disease with various presentations. This variation is due to the interaction of hormonal, environmental, and genetic factors. Associations between human leukocyte antigens and SLE have long been recognized in different ethnic populations and have been suggested to represent the most important association. OBJECTIVES The objectives of this paper were to determine susceptibility and protection human leukocyte antigens (HLA) Class II markers for SLE and to highlight, for the first time, associations between HLA alleles and clinical and serological features in South Tunisia. METHODS We conducted a case-control study on 75 SLE patients and 123 healthy controls. The HLA Class II DRB1/DQB1 of all patients and controls was genotyped using polymerase chain reaction-sequence specific primer technique. Statistical analysis was performed using SPSS software. RESULTS HLA-DRB1*03 was the principal Class II allele associated with the genetic susceptibility to SLE (pc = 0.02; OR = 2.57; CI = [1.39-4.75]; this allele was also associated with anti-SSB production (P = 0.016; OR = 4.00; CI = [1.24-12.96]). HLA-DRB1*01 was significantly more expressed in SLE patients with neurologic disorders (P = 0.013; OR = 20.25; CI = [1.87-219.21]). No allele was found to be protective against SLE in our study group. CONCLUSION Our results show that in South Tunisia SLE is associated with HLA-DRB1*03 and that some clinical features of SLE may be influenced by specific DRB1 and DQB1 alleles.
Collapse
Affiliation(s)
- Hend Hachicha
- Department of Immunology, Habib Bourguiba University, Hospital of Sfax, Sfax, Tunisia
- UR12SP14, Habib Bourguiba University, Hospital of Sfax, Sfax, Tunisia
- Department of Basic Sciences B, Faculty of Medicine of Sfax, Sfax, Tunisia
| | - Arwa Kammoun
- Department of Histocompatibility, Hedi Chaker University Hospital, University of Sfax, Tunisia
| | - Nadia Mahfoudh
- Department of Histocompatibility, Hedi Chaker University Hospital, University of Sfax, Tunisia
| | - Sameh Marzouk
- Department of Internal Medicine, Hedi Chaker University Hospital, University of Sfax, Tunisia
| | - Sawsan Feki
- Department of Immunology, Habib Bourguiba University, Hospital of Sfax, Sfax, Tunisia
| | - Raouia Fakhfakh
- UR12SP14, Habib Bourguiba University, Hospital of Sfax, Sfax, Tunisia
| | - Hajer Fourati
- UR12SP14, Habib Bourguiba University, Hospital of Sfax, Sfax, Tunisia
| | - Samy Haddouk
- Department of Immunology, Habib Bourguiba University, Hospital of Sfax, Sfax, Tunisia
- UR12SP14, Habib Bourguiba University, Hospital of Sfax, Sfax, Tunisia
| | - Faten Frikha
- Department of Internal Medicine, Hedi Chaker University Hospital, University of Sfax, Tunisia
| | - Lilia Gaddour
- Department of Histocompatibility, Hedi Chaker University Hospital, University of Sfax, Tunisia
| | - Feiza Hakim
- Department of Histocompatibility, Hedi Chaker University Hospital, University of Sfax, Tunisia
| | - Zouheir Bahloul
- Department of Internal Medicine, Hedi Chaker University Hospital, University of Sfax, Tunisia
| | - Hafedh Makni
- Department of Histocompatibility, Hedi Chaker University Hospital, University of Sfax, Tunisia
| | - Hatem Masmoudi
- Department of Immunology, Habib Bourguiba University, Hospital of Sfax, Sfax, Tunisia
- UR12SP14, Habib Bourguiba University, Hospital of Sfax, Sfax, Tunisia
| |
Collapse
|
11
|
Role of vitamin D deficiency in systemic lupus erythematosus incidence and aggravation. AUTOIMMUNITY HIGHLIGHTS 2017; 9:1. [PMID: 29280010 PMCID: PMC5743852 DOI: 10.1007/s13317-017-0101-x] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/07/2017] [Accepted: 12/01/2017] [Indexed: 12/31/2022]
Abstract
Vitamin D is one of the main groups of sterols; playing an important role in phospho-calcic metabolism. The conversion of 7-dehydrocholesterol to pre- vitamin D3 in the skin, through solar ultraviolet B radiation, is the main source of vitamin D. Since lupus patients are usually photosensitive, the risk of developing vitamin D deficiency in is high in this population. Although evidences showed the connotation between systemic lupus erythematosus (SLE) and vitamin D through which SLE can lead to lower vitamin D levels, it is also important to consider the possibility that vitamin D deficiency may have a causative role in SLE etiology. This paper analyzes existing data from various studies to highlight the role of vitamin D deficiency in SLE occurrence and aggravation and the probable efficacy of vitamin D supplementation on SLE patients. We searched “Science Direct” and “Pub Med” using “Vitamin D” and “SLE” for finding the studies focusing on the association between vitamin D deficiency and SLE incidence and consequences. Evidences show that vitamin D plays an important role in the pathogenesis and progression of SLE and vitamin D supplementation seems to ameliorate inflammatory and hemostatic markers; so, can improve clinical subsequent.
Collapse
|
12
|
Borba HHL, Funke A, Wiens A, Utiyama SRDR, Perlin CM, Pontarolo R. Update on Biologic Therapies for Systemic Lupus Erythematosus. Curr Rheumatol Rep 2017; 18:44. [PMID: 27299782 DOI: 10.1007/s11926-016-0589-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Systemic lupus erythematosus (SLE) is a chronic multisystemic autoimmune disease driven by genetic, hormonal, and environmental factors. Despite the advances in diagnostic and therapeutic approaches in the last decades, SLE still leads to significant morbidity and increased mortality. Although a cure for SLE is still unknown, treatment is required to control acute disease exacerbation episodes (flares), decrease the frequency and severity of subsequent lupus flares, address comorbidities, and prevent end-organ damage. While conventional SLE pharmacotherapy may exhibit suboptimal efficacy and substantial toxicity, a growing knowledge of the disease pathogenesis enabled the research on novel therapeutic agents directed at specific disease-related targets. In this paper, we review the recent progress in the clinical investigation of biologic agents targeting B cells, T cells, cytokines, innate immunity, and other immunologic or inflammatory pathways. Although many investigational agents exhibited insufficient efficacy or inadequate safety in clinical trials, one of them, belimumab, fulfilled the efficacy and safety regulatory requirements and was approved for the treatment of SLE in Europe and the USA, which confirms that, despite all difficulties, advances in this field are possible.
Collapse
Affiliation(s)
- Helena Hiemisch Lobo Borba
- Department of Pharmacy, Pharmaceutical Sciences Postgraduate Research Program, Federal University of Parana, Campus III, Av. Pref. Lothario Meissner, 632, Jardim Botanico, Curitiba, PR, 80210-170, Brazil
| | - Andreas Funke
- Rheumatology Service, Hospital de Clinicas, Federal University of Parana, Curitiba, PR, Brazil
| | - Astrid Wiens
- Department of Pharmacy, Pharmaceutical Sciences Postgraduate Research Program, Federal University of Parana, Campus III, Av. Pref. Lothario Meissner, 632, Jardim Botanico, Curitiba, PR, 80210-170, Brazil
| | - Shirley Ramos da Rosa Utiyama
- Department of Pharmacy, Pharmaceutical Sciences Postgraduate Research Program, Federal University of Parana, Campus III, Av. Pref. Lothario Meissner, 632, Jardim Botanico, Curitiba, PR, 80210-170, Brazil
| | - Cássio Marques Perlin
- Department of Pharmacy, Pharmaceutical Sciences Postgraduate Research Program, Federal University of Parana, Campus III, Av. Pref. Lothario Meissner, 632, Jardim Botanico, Curitiba, PR, 80210-170, Brazil
| | - Roberto Pontarolo
- Department of Pharmacy, Pharmaceutical Sciences Postgraduate Research Program, Federal University of Parana, Campus III, Av. Pref. Lothario Meissner, 632, Jardim Botanico, Curitiba, PR, 80210-170, Brazil.
| |
Collapse
|
13
|
Wang L, Hao C, Deng Y, Liu Y, Hu S, Peng Y, He M, Fu J, Liu M, Chen J, Chen X. Screening epitopes on systemic lupus erythematosus autoantigens with a peptide array. Oncotarget 2017; 8:85559-85567. [PMID: 29156741 PMCID: PMC5689631 DOI: 10.18632/oncotarget.20994] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2017] [Accepted: 08/28/2017] [Indexed: 11/29/2022] Open
Abstract
Systemic lupus erythematosus (SLE) is a common autoimmune disease. Many autoantibodies are closely associated with SLE. However, the specific epitopes recognized and bound by these autoantibodies are still unclear. This study screened the binding epitopes of SLE-related autoantibodies using a high-throughput screening method. Epitope prediction on 12 SLE-related autoantigens was performed using the Immune Epitope Database and Analysis Resource (IEDB) software. The predicted epitopes were synthesized into peptides and developed into a peptide array. Serum IgG from 50 SLE patients and 25 healthy controls was detected using the peptide array. The results were then validated using an enzyme-linked immunosorbent assay (ELISA). The diagnostic efficiency of each epitope was analyzed using a ROC curve. Seventy-three potential epitopes were screened for using the IEDB software after the epitopes on the 12 SLE-related autoantigens were analyzed. Peptide array screening revealed that the levels of the autoantibodies recognized and bound by 4 peptide antigens were significantly upregulated in the serum of SLE patients (P < 0.05). The ELISA results showed that the 4 antigens with significantly increased serum autoantibodies levels in SLE patients were acidic ribosomal phosphoprotein (P0)-4, acidic ribosomal phosphoprotein (P0)-11, DNA topoisomerase 1 (full length)-1, and U1-SnRNP 68/70 KDa-1 (P < 0.05), and the areas under the ROC curve for diagnosing SLE on the basis of these peptides were 0.91, 0.90, 0.93, and 0.91, respectively. Many autoantibodies specifically expressed in the serum of patients with SLE can be detected by specific peptide fragments and may be used as markers in clinical auxiliary diagnoses.
Collapse
Affiliation(s)
- Lin Wang
- Department of Rheumatology, Shaoyang Central Hospital, 422000 Shaoyang, China
| | - Chenjun Hao
- Obstetrics and gynecology, Guangzhou Panyu Hexian Memorial Hospital, 511400 Guangzhou, China
| | - Yongqiu Deng
- Obstetrics and gynecology, Guangzhou Panyu Hexian Memorial Hospital, 511400 Guangzhou, China
| | - Yanbo Liu
- Department of Rheumatology, Shaoyang Central Hospital, 422000 Shaoyang, China
| | - Shiliang Hu
- Department of Rheumatology, Shaoyang Central Hospital, 422000 Shaoyang, China
| | - Yangang Peng
- Department of Rheumatology, Shaoyang Central Hospital, 422000 Shaoyang, China
| | - Manna He
- Department of Rheumatology, Shaoyang Central Hospital, 422000 Shaoyang, China
| | - Jinhu Fu
- Department of Rheumatology, Shaoyang Central Hospital, 422000 Shaoyang, China
| | - Ming Liu
- Department of Rheumatology, Shaoyang Central Hospital, 422000 Shaoyang, China
| | - Jia Chen
- Department of Rheumatology, Shaoyang Central Hospital, 422000 Shaoyang, China
| | - Xiaoming Chen
- Department of Rheumatology, Shaoyang Central Hospital, 422000 Shaoyang, China
| |
Collapse
|
14
|
Wang J, Shen H, Zhu Y, Zhu Y, Cai L, Wang Z, Shi Q, Qiu Y. Characterization of a PRISTANE-induced lupus-associated model in the non-human primate cynomolgus monkey. J Med Primatol 2017; 47:18-28. [PMID: 28573661 DOI: 10.1111/jmp.12280] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/26/2017] [Indexed: 11/28/2022]
Abstract
BACKGROUND Lupus is an autoimmune disease with complex syndrome. Rodent models have limitations for recapitulating the spectrum of the disease. A more powerful translational model is desirable. METHOD Lupus-associated model in cynomolgus monkeys was induced by two intraperitoneal injections of 2, 6, 10, 14-tetramethylpentadecane (PRISTANE). Lupus-specific biomarkers and manifestations over a 246-day period were observed at multilevel. To visualize and quantify kidney function in real time, contrast-enhanced ultrasound was used. RESULTS The indicative biomarkers and manifestations fulfilled major diagnosis criteria according to the "Criteria of Lupus" of the American College of Rheumatology. Significant changes in time-intensity curve parameters were observed, indicating impaired renal function and the method as a feasible, non-invasive diagnostic method in primate model. CONCLUSIONS We successfully induced lupus-associated model with systemic lupus syndrome. This primate model can be a valuable translational model for further pathogenesis and symptomology studies and for exploring therapeutic candidates.
Collapse
Affiliation(s)
- Jing Wang
- Department of Immunology, Medical College, Soochow University, Jiangsu, China.,Laboratory Animal Center, Soochow University, Jiangsu, China
| | - Hui Shen
- Central Laboratory, The First hospital of Jiaxing, Zhejiang, China
| | - Yuqiang Zhu
- Department of Immunology, Medical College, Soochow University, Jiangsu, China
| | - Ying Zhu
- Center for Clinical Laboratory, First Affiliated Hospital of Soochow University, Jiangsu, China
| | - Lei Cai
- Department of Immunology, Medical College, Soochow University, Jiangsu, China
| | - Zhiyao Wang
- Department of Immunology, Medical College, Soochow University, Jiangsu, China
| | - Qin Shi
- Department of Orthopedics, First Affiliated Hospital of Soochow University, Jiangsu, China
| | - Yuhua Qiu
- Department of Immunology, Medical College, Soochow University, Jiangsu, China
| |
Collapse
|
15
|
Passos MEP, Alves HHO, Momesso CM, Faria FG, Murata G, Cury-Boaventura MF, Hatanaka E, Massao-Hirabara S, Gorjão R. Differential effects of palmitoleic acid on human lymphocyte proliferation and function. Lipids Health Dis 2016; 15:217. [PMID: 27964715 PMCID: PMC5154135 DOI: 10.1186/s12944-016-0385-2] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2016] [Accepted: 12/02/2016] [Indexed: 02/13/2023] Open
Abstract
BACKGROUND Palmitoleic acid (PA) is a n-7 monounsaturated fatty acid (MUFA) secreted by adipose tissue and related to decreased insulin resistance in peripheral tissues. Evidences have been shown that PA also decreased proinflammatory cytokine expression in cultured macrophages. Although studies have shown that other fatty acids (FAs) modulate several lymphocyte functions, the specific effect of PA on these cells is unknown. The aim of the present study was to evaluate the possible influence of PA on activation and differentiation of human lymphocytes in comparison to oleic acid (OA). METHODS Human lymphocytes were isolated from peripheral blood of health men and cultured in the presence of growing concentrations of PA or OA (5 to 200 μM), for 24 h. After that, cells were collected and cytotoxicity evaluated by flow cytometry. Then, we analyzed proliferative capacity in lymphocytes treated with non toxic concentrations of PA and OA (25 and 50 μM, respectively), in the presence or absence of concanavalin A (ConA). The Th1/Th2/Th17 cytokine production was determined by the Cytometric Bead Array. CD28 and CD95 surface expression and T regulatory cell percentage were determined by flow cytometry. RESULTS We observed that PA is toxic to lymphocytes above 50 μM. PA promoted a decrease of lymphocyte proliferation stimulated by ConA in both concentrations. PA also decreased CD28 externalization and increased CD95. On the other hand, OA did not alter these parameters. In the same way, PA reduced IL6, IFN-gamma, TNF-alpha and IL17A production in both concentration and IL2 only at 50 μM (in the presence of ConA). OA promoted IFN-gamma reduction in both concentrations and an increase of IL-2, IL4 and IL10 at 25 μM. Both fatty acids decreased the percentage of T regulatory cells. CONCLUSION In conclusion, PA promoted a suppressive effect on lymphocyte proliferation characterized by a decrease of Th1 and Th17 response, and co-stimulatory molecule (CD28). However, OA increased lymphocyte proliferation through IL2 production and Th2 response. These results also show a more suppressive effect of PA on lymphocytes in comparison to OA.
Collapse
Affiliation(s)
- M E P Passos
- Institute of Physical Activity and Sport Sciences, Interdisciplinary Post-graduate Program in Health Sciences, Cruzeiro do Sul University, Rua Galvão Bueno, 868, Liberdade, CEP: 01506 000, São Paulo, SP, Brazil.
| | - H H O Alves
- Institute of Physical Activity and Sport Sciences, Interdisciplinary Post-graduate Program in Health Sciences, Cruzeiro do Sul University, Rua Galvão Bueno, 868, Liberdade, CEP: 01506 000, São Paulo, SP, Brazil
| | - C M Momesso
- Institute of Physical Activity and Sport Sciences, Interdisciplinary Post-graduate Program in Health Sciences, Cruzeiro do Sul University, Rua Galvão Bueno, 868, Liberdade, CEP: 01506 000, São Paulo, SP, Brazil
| | - F G Faria
- Institute of Physical Activity and Sport Sciences, Interdisciplinary Post-graduate Program in Health Sciences, Cruzeiro do Sul University, Rua Galvão Bueno, 868, Liberdade, CEP: 01506 000, São Paulo, SP, Brazil
| | - G Murata
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - M F Cury-Boaventura
- Institute of Physical Activity and Sport Sciences, Interdisciplinary Post-graduate Program in Health Sciences, Cruzeiro do Sul University, Rua Galvão Bueno, 868, Liberdade, CEP: 01506 000, São Paulo, SP, Brazil
| | - E Hatanaka
- Institute of Physical Activity and Sport Sciences, Interdisciplinary Post-graduate Program in Health Sciences, Cruzeiro do Sul University, Rua Galvão Bueno, 868, Liberdade, CEP: 01506 000, São Paulo, SP, Brazil
| | - S Massao-Hirabara
- Institute of Physical Activity and Sport Sciences, Interdisciplinary Post-graduate Program in Health Sciences, Cruzeiro do Sul University, Rua Galvão Bueno, 868, Liberdade, CEP: 01506 000, São Paulo, SP, Brazil
| | - R Gorjão
- Institute of Physical Activity and Sport Sciences, Interdisciplinary Post-graduate Program in Health Sciences, Cruzeiro do Sul University, Rua Galvão Bueno, 868, Liberdade, CEP: 01506 000, São Paulo, SP, Brazil.
| |
Collapse
|
16
|
He YY, Yan Y, Zhang HF, Lin YH, Chen YC, Yan Y, Wu P, Fang JS, Yang SH, Du GH. Methyl salicylate 2- O-β-d-lactoside alleviates the pathological progression of pristane-induced systemic lupus erythematosus-like disease in mice via suppression of inflammatory response and signal transduction. DRUG DESIGN DEVELOPMENT AND THERAPY 2016; 10:3183-3196. [PMID: 27729775 PMCID: PMC5047741 DOI: 10.2147/dddt.s114501] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Systemic lupus erythematosus (SLE), with a high incidence rate and insufficient therapy worldwide, is a complex disease involving multiple organs characterized primarily by inflammation due to deposition of immunocomplexes formed by production of autoantibodies. The mechanism of SLE remains unclear, and the disease still cannot be cured. We used pristane to induce SLE in female BALB/c mice. Methyl salicylate 2-O-β-d-lactoside (MSL; 200, 400, and 800 mg/kg) was orally administered 45 days after pristane injection for 4.5 months. The results showed that MSL antagonized the increasing levels of multiple types of antibodies and cytokines in lupus mice. MSL was found to suppress joint swelling and have potent inhibitory effect on arthritis-like symptoms. MSL also significantly decreased the spleen index and expression of inflammatory markers in the lupus mice. MSL protected the kidneys of lupus mice from injury through inhibiting the expression of inflammatory cytokines and reducing the IgG and C3 immunocomplex deposits. Further Western blot assays revealed that the downregulation of the intracellular inflammatory signals of NFκB and JAK/STAT3 might be the potential molecular mechanisms of the pharmacological activity of MSL against SLE in vivo. These findings may demonstrate that MSL has the potential to be a useful and highly effective treatment for SLE.
Collapse
Affiliation(s)
- Yang-Yang He
- Beijing Key Laboratory of Drug Target Identification and Drug Screening, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing; State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing
| | - Yu Yan
- Beijing Key Laboratory of Drug Target Identification and Drug Screening, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing; State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing
| | - Hui-Fang Zhang
- Beijing Key Laboratory of Drug Target Identification and Drug Screening, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing; State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing
| | - Yi-Huang Lin
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing
| | - Yu-Cai Chen
- Beijing Key Laboratory of Drug Target Identification and Drug Screening, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing; State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing
| | - Yi Yan
- Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai
| | - Ping Wu
- Beijing Key Laboratory of Drug Target Identification and Drug Screening, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing; State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing
| | - Jian-Song Fang
- Institute of Clinical Pharmacology, Guangzhou University of Traditional Chinese Medicine, Guangzhou, People's Republic of China
| | - Shu-Hui Yang
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing
| | - Guan-Hua Du
- Beijing Key Laboratory of Drug Target Identification and Drug Screening, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing; State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing
| |
Collapse
|
17
|
Raj P, Rai E, Song R, Khan S, Wakeland BE, Viswanathan K, Arana C, Liang C, Zhang B, Dozmorov I, Carr-Johnson F, Mitrovic M, Wiley GB, Kelly JA, Lauwerys BR, Olsen NJ, Cotsapas C, Garcia CK, Wise CA, Harley JB, Nath SK, James JA, Jacob CO, Tsao BP, Pasare C, Karp DR, Li QZ, Gaffney PM, Wakeland EK. Regulatory polymorphisms modulate the expression of HLA class II molecules and promote autoimmunity. eLife 2016; 5:e12089. [PMID: 26880555 PMCID: PMC4811771 DOI: 10.7554/elife.12089] [Citation(s) in RCA: 94] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2015] [Accepted: 02/13/2016] [Indexed: 12/15/2022] Open
Abstract
Targeted sequencing of sixteen SLE risk loci among 1349 Caucasian cases and controls produced a comprehensive dataset of the variations causing susceptibility to systemic lupus erythematosus (SLE). Two independent disease association signals in the HLA-D region identified two regulatory regions containing 3562 polymorphisms that modified thirty-seven transcription factor binding sites. These extensive functional variations are a new and potent facet of HLA polymorphism. Variations modifying the consensus binding motifs of IRF4 and CTCF in the XL9 regulatory complex modified the transcription of HLA-DRB1, HLA-DQA1 and HLA-DQB1 in a chromosome-specific manner, resulting in a 2.5-fold increase in the surface expression of HLA-DR and DQ molecules on dendritic cells with SLE risk genotypes, which increases to over 4-fold after stimulation. Similar analyses of fifteen other SLE risk loci identified 1206 functional variants tightly linked with disease-associated SNPs and demonstrated that common disease alleles contain multiple causal variants modulating multiple immune system genes.
Collapse
Affiliation(s)
- Prithvi Raj
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, United States
| | - Ekta Rai
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, United States
- School of Biotechnology, Shri Mata Vaishno Devi University, Katra, India
| | - Ran Song
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, United States
| | - Shaheen Khan
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, United States
| | - Benjamin E Wakeland
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, United States
| | - Kasthuribai Viswanathan
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, United States
| | - Carlos Arana
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, United States
| | - Chaoying Liang
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, United States
| | - Bo Zhang
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, United States
| | - Igor Dozmorov
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, United States
| | - Ferdicia Carr-Johnson
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, United States
| | - Mitja Mitrovic
- Department of Neurology, Yale School of Medicine, New Haven, United States
| | - Graham B Wiley
- Arthritis and Clinical Immunology Program, Oklahoma Medical Research Foundation, Oklahoma City, United States
| | - Jennifer A Kelly
- Arthritis and Clinical Immunology Program, Oklahoma Medical Research Foundation, Oklahoma City, United States
| | - Bernard R Lauwerys
- Pole de pathologies rhumatismales, Institut de Recherche Expérimentale et Clinique, Université catholique de Louvain, Bruxelles, Belgium
| | - Nancy J Olsen
- Division of Rheumatology, Department of Medicine, Penn State Medical School, Hershey, United States
| | - Chris Cotsapas
- Department of Neurology, Yale School of Medicine, New Haven, United States
| | - Christine K Garcia
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, United States
- Eugene McDermott Center for Human Growth and Development, University of Texas Southwestern Medical Center, Dallas, United States
| | - Carol A Wise
- Eugene McDermott Center for Human Growth and Development, University of Texas Southwestern Medical Center, Dallas, United States
- Department of Orthopaedic Surgery, University of Texas Southwestern Medical Center, Dallas, United States
- Sarah M. and Charles E. Seay Center for Musculoskeletal Research, Texas Scottish Rite Hospital for Children, Dallas, United States
- Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, United States
| | - John B Harley
- Cincinnati VA Medical Center, Cincinnati, United States
- Cincinnati Children's Hospital Medical Center, Cincinnati, United States
| | - Swapan K Nath
- Arthritis and Clinical Immunology Program, Oklahoma Medical Research Foundation, Oklahoma City, United States
| | - Judith A James
- Arthritis and Clinical Immunology Program, Oklahoma Medical Research Foundation, Oklahoma City, United States
| | - Chaim O Jacob
- Department of Medicine, University of Southern California, Los Angeles, United States
| | - Betty P Tsao
- Department of Medicine, University of California, Los Angeles, Los Angeles, United States
| | - Chandrashekhar Pasare
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, United States
| | - David R Karp
- Rheumatic Diseases Division, Department of Medicine, University of Texas Southwestern Medical Center, Dallas, United States
| | - Quan Zhen Li
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, United States
| | - Patrick M Gaffney
- Arthritis and Clinical Immunology Program, Oklahoma Medical Research Foundation, Oklahoma City, United States
| | - Edward K Wakeland
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, United States
| |
Collapse
|
18
|
Carvalheiro T, Gomes D, Pinto LA, Inês L, Lopes A, Henriques A, Pedreiro S, Martinho A, Trindade H, Young HA, da Silva JAP, Paiva A. Sera from patients with active systemic lupus erythematosus patients enhance the toll-like receptor 4 response in monocyte subsets. JOURNAL OF INFLAMMATION-LONDON 2015; 12:38. [PMID: 26038677 PMCID: PMC4451730 DOI: 10.1186/s12950-015-0083-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/11/2015] [Accepted: 05/18/2015] [Indexed: 01/08/2023]
Abstract
Background Systemic Lupus Erythematosus (SLE) is an auto-immune disease whose complex pathogenesis remains unraveled. Here we aim to explore the inflammatory ability of SLE patients’ sera upon peripheral blood (PB) monocyte subsets and myeloid dendritic cells (mDCs) obtained from healthy donors. Methods In this study we included 11 SLE patients with active disease (ASLE), 11 with inactive disease (ISLE) and 10 healthy controls (HC). PB from healthy donors was stimulated with patients’ sera, toll-like receptor (TLR) 4 ligand – lipopolysaccharide or both. The intracellular production of TNF-α was evaluated in classical, non-classical monocytes and mDCs, using flow cytometry. TNF-α mRNA expression was assessed in all these purified cells, after sera treatment. Results We found that sera of SLE patients did not change spontaneous TNF-α production by monocytes or dendritic cells. However, upon stimulation of TLR4, the presence of sera from ASLE patients, but not ISLE, significantly increased the intracellular expression of TNF-α in classical and non-classical monocytes. This ability was related to titers anti-double stranded DNA antibodies in the serum. High levels of anti-TNF-α in the patients’ sera were associated with increased TNF-α expression by co-cultured mDCs. No relationship was found with the levels of a wide variety of other pro-inflammatory cytokines. A slight increase of TNF-α mRNA expression was observed in these purified cells when they were cultured only in the presence of SLE serum. Conclusions Our data suggest that SLE sera induce an abnormal in vitro TLR4 response in classical and non-classical monocytes, reflected by a higher TNF-α intracellular expression. These effects may be operative in the pathogenesis of SLE.
Collapse
Affiliation(s)
- Tiago Carvalheiro
- Blood and Transplantation Center of Coimbra, Portuguese Institute of Blood and Transplantation, Quinta da Vinha Moura, São Martinho do Bispo, 3041-861 Coimbra, Portugal
| | - Diane Gomes
- College of Health Technology of Coimbra, Rua 5 de Outubro, São Martinho do Bispo, 3046-854 Coimbra, Portugal
| | - Ligia A Pinto
- HPV Immunology Laboratory, Frederick National Laboratory for Cancer Research, Building 469, 21702 Frederick, MD USA
| | - Luis Inês
- Rheumatology Department, Centro Hospitalar e Universitário de Coimbra, Praceta Prof. Mota Pinto, 3000-075 Coimbra, Portugal ; Faculty of Medicine, University of Coimbra, Azinhaga de Santa Comba, Celas, 3000-548 Coimbra, Portugal ; School of Health Sciences, University of Beira Interior, Avenida Infante D. Henrique, 6200-506 Covilhã, Portugal
| | - Ana Lopes
- Blood and Transplantation Center of Coimbra, Portuguese Institute of Blood and Transplantation, Quinta da Vinha Moura, São Martinho do Bispo, 3041-861 Coimbra, Portugal
| | - Ana Henriques
- Faculty of Medicine, University of Coimbra, Azinhaga de Santa Comba, Celas, 3000-548 Coimbra, Portugal
| | - Susana Pedreiro
- Blood and Transplantation Center of Coimbra, Portuguese Institute of Blood and Transplantation, Quinta da Vinha Moura, São Martinho do Bispo, 3041-861 Coimbra, Portugal
| | - António Martinho
- Blood and Transplantation Center of Coimbra, Portuguese Institute of Blood and Transplantation, Quinta da Vinha Moura, São Martinho do Bispo, 3041-861 Coimbra, Portugal
| | - Hélder Trindade
- Blood and Transplantation Center of Coimbra, Portuguese Institute of Blood and Transplantation, Quinta da Vinha Moura, São Martinho do Bispo, 3041-861 Coimbra, Portugal
| | - Howard A Young
- Laboratory of Experimental Immunology, Cancer and Inflammation Program, National Cancer Institute at Frederick, Building 560, 21702-1201 Frederick, MD USA
| | - José António Pereira da Silva
- Rheumatology Department, Centro Hospitalar e Universitário de Coimbra, Praceta Prof. Mota Pinto, 3000-075 Coimbra, Portugal ; Faculty of Medicine, University of Coimbra, Azinhaga de Santa Comba, Celas, 3000-548 Coimbra, Portugal
| | - Artur Paiva
- Blood and Transplantation Center of Coimbra, Portuguese Institute of Blood and Transplantation, Quinta da Vinha Moura, São Martinho do Bispo, 3041-861 Coimbra, Portugal ; College of Health Technology of Coimbra, Rua 5 de Outubro, São Martinho do Bispo, 3046-854 Coimbra, Portugal
| |
Collapse
|
19
|
Abstract
Systemic lupus erythematosus (SLE) is a multisystem autoimmune disorder that has a broad spectrum of effects on the majority of organs, including the kidneys. Approximately 40-70% of patients with SLE will develop lupus nephritis. Renal assault during SLE is initiated by genes that breach immune tolerance and promote autoantibody production. These genes might act in concert with other genetic factors that augment innate immune signalling and IFN-I production, which in turn can generate an influx of effector leucocytes, inflammatory mediators and autoantibodies into end organs, such as the kidneys. The presence of cognate antigens in the glomerular matrix, together with intrinsic molecular abnormalities in resident renal cells, might further accentuate disease progression. This Review discusses the genetic insights and molecular mechanisms for key pathogenic contributors in SLE and lupus nephritis. We have categorized the genes identified in human studies of SLE into one of four pathogenic events that lead to lupus nephritis. We selected these categories on the basis of the cell types in which these genes are expressed, and the emerging paradigms of SLE pathogenesis arising from murine models. Deciphering the molecular basis of SLE and/or lupus nephritis in each patient will help physicians to tailor specific therapies.
Collapse
|
20
|
Silva-Sanchez A, Liu CR, Vale AM, Khass M, Kapoor P, Elgavish A, Ivanov II, Ippolito GC, Schelonka RL, Schoeb TR, Burrows PD, Schroeder HW. Violation of an evolutionarily conserved immunoglobulin diversity gene sequence preference promotes production of dsDNA-specific IgG antibodies. PLoS One 2015; 10:e0118171. [PMID: 25706374 PMCID: PMC4338297 DOI: 10.1371/journal.pone.0118171] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2014] [Accepted: 01/08/2015] [Indexed: 11/24/2022] Open
Abstract
Variability in the developing antibody repertoire is focused on the third complementarity determining region of the H chain (CDR-H3), which lies at the center of the antigen binding site where it often plays a decisive role in antigen binding. The power of VDJ recombination and N nucleotide addition has led to the common conception that the sequence of CDR-H3 is unrestricted in its variability and random in its composition. Under this view, the immune response is solely controlled by somatic positive and negative clonal selection mechanisms that act on individual B cells to promote production of protective antibodies and prevent the production of self-reactive antibodies. This concept of a repertoire of random antigen binding sites is inconsistent with the observation that diversity (DH) gene segment sequence content by reading frame (RF) is evolutionarily conserved, creating biases in the prevalence and distribution of individual amino acids in CDR-H3. For example, arginine, which is often found in the CDR-H3 of dsDNA binding autoantibodies, is under-represented in the commonly used DH RFs rearranged by deletion, but is a frequent component of rarely used inverted RF1 (iRF1), which is rearranged by inversion. To determine the effect of altering this germline bias in DH gene segment sequence on autoantibody production, we generated mice that by genetic manipulation are forced to utilize an iRF1 sequence encoding two arginines. Over a one year period we collected serial serum samples from these unimmunized, specific pathogen-free mice and found that more than one-fifth of them contained elevated levels of dsDNA-binding IgG, but not IgM; whereas mice with a wild type DH sequence did not. Thus, germline bias against the use of arginine enriched DH sequence helps to reduce the likelihood of producing self-reactive antibodies.
Collapse
Affiliation(s)
- Aaron Silva-Sanchez
- Department of Medicine, Division of Clinical Immunology and Rheumatology, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Cun Ren Liu
- Department of Medicine, Division of Clinical Immunology and Rheumatology, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Andre M. Vale
- Department of Medicine, Division of Clinical Immunology and Rheumatology, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
- Program in Immunobiology, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Mohamed Khass
- Department of Medicine, Division of Clinical Immunology and Rheumatology, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
- Genetic Engineering Division, National Research Center of Egypt, Ad Doqi, Egypt
| | - Pratibha Kapoor
- Department of Medicine, Division of Clinical Immunology and Rheumatology, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Ada Elgavish
- Department of Medicine, Division of Clinical Immunology and Rheumatology, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Ivaylo I. Ivanov
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Gregory C. Ippolito
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Robert L. Schelonka
- Department of Pediatrics, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Trenton R. Schoeb
- Department of Genetics, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Peter D. Burrows
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
- Department of Genetics, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Harry W. Schroeder
- Department of Medicine, Division of Clinical Immunology and Rheumatology, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
- Department of Genetics, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
- * E-mail:
| |
Collapse
|
21
|
Dai C, Deng Y, Quinlan A, Gaskin F, Tsao BP, Fu SM. Genetics of systemic lupus erythematosus: immune responses and end organ resistance to damage. Curr Opin Immunol 2014; 31:87-96. [PMID: 25458999 DOI: 10.1016/j.coi.2014.10.004] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2014] [Revised: 10/10/2014] [Accepted: 10/10/2014] [Indexed: 01/13/2023]
Abstract
Systemic lupus erythematosus (SLE) is a prototypic systemic autoimmune disorder. Considerable progress has been made to delineate the genetic control of this complex disorder. In this review, selected aspects of human and mouse genetics related to SLE are reviewed with emphasis on genes that contribute to both innate and adaptive immunity and to genes that contribute directly to susceptibility to end organ damage. It is concluded that the interactions among these two major pathways will provide further insight into the pathogenesis of SLE. An interactive model of the two major pathways is proposed without emphasis on the importance of breaking tolerance to autoantigens.
Collapse
Affiliation(s)
- Chao Dai
- Division of Rheumatology, Center of Inflammation, Immunity and Regenerative Medicine, Department of Medicine, University of Virginia School of Medicine, Charlottesville, VA 22908, United States
| | - Yun Deng
- Division of Rheumatology, Department of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, United States
| | - Aaron Quinlan
- Department of Public Health Sciences, Center for Public Health Genomics, University of Virginia, Charlottesville, VA 22908, United States
| | - Felicia Gaskin
- Department of Psychiatry and Neurobehavioral Sciences, University of Virginia School of Medicine, Charlottesville, VA 22908, United States
| | - Betty P Tsao
- Division of Rheumatology, Department of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, United States.
| | - Shu Man Fu
- Division of Rheumatology, Center of Inflammation, Immunity and Regenerative Medicine, Department of Medicine, University of Virginia School of Medicine, Charlottesville, VA 22908, United States.
| |
Collapse
|
22
|
Bon J, Kahloon R, Zhang Y, Xue J, Fuhrman CR, Tan J, Burger M, Kass DJ, Csizmadia E, Otterbein L, Chandra D, Bhargava A, Pilewski JM, Roodman GD, Sciurba FC, Duncan SR. Autoreactivity to glucose regulated protein 78 links emphysema and osteoporosis in smokers. PLoS One 2014; 9:e105066. [PMID: 25216103 PMCID: PMC4162538 DOI: 10.1371/journal.pone.0105066] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2014] [Accepted: 07/17/2014] [Indexed: 02/06/2023] Open
Abstract
Rationale Emphysema and osteoporosis are epidemiologically associated diseases of cigarette smokers. The causal mechanism(s) linking these illnesses is unknown. We hypothesized autoimmune responses may be involved in both disorders. Objectives To discover an antigen-specific autoimmune response associated with both emphysema and osteoporosis among smokers. Methods Replicate nonbiased discovery assays indicated that autoimmunity to glucose regulated protein 78 (GRP78), an endoplasmic reticulum chaperone and cell surface signaling receptor, is present in many smokers. Subject assessments included spirometry, chest CT scans, dual x-ray absorptiometry, and immunoblots for anti-GRP78 IgG. Anti-GRP78 autoantibodies were isolated from patient plasma by affinity chromatography, leukocyte functions assessed by flow cytometry, and soluble metabolites and mediators measured by immunoassays. Measurements and Main Results Circulating anti-GRP78 IgG autoantibodies were detected in plasma specimens from 86 (32%) of the 265 smoking subjects. Anti-GRP78 autoantibodies were singularly prevalent among subjects with radiographic emphysema (OR 3.1, 95%CI 1.7–5.7, p = 0.003). Anti-GRP78 autoantibodies were also associated with osteoporosis (OR 4.7, 95%CI 1.7–13.3, p = 0.002), and increased circulating bone metabolites (p = 0.006). Among emphysematous subjects, GRP78 protein was an autoantigen of CD4 T-cells, stimulating lymphocyte proliferation (p = 0.0002) and IFN-gamma production (p = 0.03). Patient-derived anti-GRP78 autoantibodies had avidities for osteoclasts and macrophages, and increased macrophage NFkB phosphorylation (p = 0.005) and productions of IL-8, CCL-2, and MMP9 (p = 0.005, 0.007, 0.03, respectively). Conclusions Humoral and cellular GRP78 autoimmune responses in smokers have numerous biologically-relevant pro-inflammatory and other deleterious actions, and are associated with emphysema and osteoporosis. These findings may have relevance for the pathogenesis of smoking-associated diseases, and development of biomarker immunoassays and/or novel treatments for these disorders.
Collapse
Affiliation(s)
- Jessica Bon
- Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Rehan Kahloon
- Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Yingze Zhang
- Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
- * E-mail: (YZ); (SRD)
| | - Jianmin Xue
- Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Carl R. Fuhrman
- Department of Radiology, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Jiangning Tan
- Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Mathew Burger
- Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Daniel J. Kass
- Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Eva Csizmadia
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Leo Otterbein
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Divay Chandra
- Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Arpit Bhargava
- Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Joseph M. Pilewski
- Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - G. David Roodman
- Department of Medicine, Indiana School of Medicine, Indianapolis, Indiana, United States of America
| | - Frank C. Sciurba
- Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Steven R. Duncan
- Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
- * E-mail: (YZ); (SRD)
| |
Collapse
|
23
|
Pestka JJ, Vines LL, Bates MA, He K, Langohr I. Comparative effects of n-3, n-6 and n-9 unsaturated fatty acid-rich diet consumption on lupus nephritis, autoantibody production and CD4+ T cell-related gene responses in the autoimmune NZBWF1 mouse. PLoS One 2014; 9:e100255. [PMID: 24945254 PMCID: PMC4063768 DOI: 10.1371/journal.pone.0100255] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2014] [Accepted: 05/23/2014] [Indexed: 12/23/2022] Open
Abstract
Mortality from systemic lupus erythematosus (SLE), a prototypical autoimmune disease, correlates with the onset and severity of kidney glomerulonephritis. There are both preclinical and clinical evidence that SLE patients may benefit from consumption of n-3 polyunsaturated fatty acids (PUFA) found in fish oil, but the mechanisms remain unclear. Here we employed the NZBWF1 SLE mouse model to compare the effects of dietary lipids on the onset and severity of autoimmune glomerulonephritis after consuming: 1) n-3 PUFA-rich diet containing docosahexaenoic acid-enriched fish oil (DFO), 2) n-6 PUFA-rich Western-type diet containing corn oil (CRN) or 3) n-9 monounsaturated fatty acid (MUFA)-rich Mediterranean-type diet containing high oleic safflower oil (HOS). Elevated plasma autoantibodies, proteinuria and glomerulonephritis were evident in mice fed either the n-6 PUFA or n-9 MUFA diets, however, all three endpoints were markedly attenuated in mice that consumed the n-3 PUFA diet until 34 wk of age. A focused PCR array was used to relate these findings to the expression of 84 genes associated with CD4+ T cell function in the spleen and kidney both prior to and after the onset of the autoimmune nephritis. n-3 PUFA suppression of autoimmunity in NZBWF1 mice was found to co-occur with a generalized downregulation of CD4+ T cell-related genes in kidney and/or spleen at wk 34. These genes were associated with the inflammatory response, antigen presentation, T cell activation, B cell activation/differentiation and leukocyte recruitment. Quantitative RT-PCR of representative affected genes confirmed that n-3 PUFA consumption was associated with reduced expression of CD80, CTLA-4, IL-10, IL-18, CCL-5, CXCR3, IL-6, TNF-α and osteopontin mRNAs in kidney and/or spleens as compared to mice fed n-6 PUFA or n-9 MUFA diets. Remarkably, many of the genes identified in this study are currently under consideration as biomarkers and/or biotherapeutic targets for SLE and other autoimmune diseases.
Collapse
Affiliation(s)
- James J. Pestka
- Department of Food Science and Human Nutrition, Diagnostic Center for Population and Animal Health, Michigan State University, East Lansing, Michigan, United States of America
- Center for Integrative Toxicology, Diagnostic Center for Population and Animal Health, Michigan State University, East Lansing, Michigan, United States of America
- Department of Microbiology and Molecular Genetics, Diagnostic Center for Population and Animal Health, Michigan State University, East Lansing, Michigan, United States of America
| | - Laura L. Vines
- Department of Food Science and Human Nutrition, Diagnostic Center for Population and Animal Health, Michigan State University, East Lansing, Michigan, United States of America
- Center for Integrative Toxicology, Diagnostic Center for Population and Animal Health, Michigan State University, East Lansing, Michigan, United States of America
| | - Melissa A. Bates
- Department of Food Science and Human Nutrition, Diagnostic Center for Population and Animal Health, Michigan State University, East Lansing, Michigan, United States of America
- Center for Integrative Toxicology, Diagnostic Center for Population and Animal Health, Michigan State University, East Lansing, Michigan, United States of America
| | - Kaiyu He
- Center for Integrative Toxicology, Diagnostic Center for Population and Animal Health, Michigan State University, East Lansing, Michigan, United States of America
- Department of Microbiology and Molecular Genetics, Diagnostic Center for Population and Animal Health, Michigan State University, East Lansing, Michigan, United States of America
| | - Ingeborg Langohr
- Division of Anatomic Pathology, Diagnostic Center for Population and Animal Health, Michigan State University, East Lansing, Michigan, United States of America
| |
Collapse
|
24
|
Vigato-Ferreira ICC, Toller-Kawahisa JE, Pancoto JAT, Mendes-Junior CT, Martinez EZ, Donadi EA, Louzada-Júnior P, Del Lama JEC, Marzocchi-Machado CM. FcγRIIa and FcγRIIIb polymorphisms and associations with clinical manifestations in systemic lupus erythematosus patients. Autoimmunity 2014; 47:451-8. [DOI: 10.3109/08916934.2014.921809] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
25
|
Bai Y, Ni M, Cooper B, Wei Y, Fury W. Inference of high resolution HLA types using genome-wide RNA or DNA sequencing reads. BMC Genomics 2014; 15:325. [PMID: 24884790 PMCID: PMC4035057 DOI: 10.1186/1471-2164-15-325] [Citation(s) in RCA: 84] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2013] [Accepted: 04/04/2014] [Indexed: 12/03/2022] Open
Abstract
BACKGROUND Accurate HLA typing at amino acid level (four-digit resolution) is critical in hematopoietic and organ transplantations, pathogenesis studies of autoimmune and infectious diseases, as well as the development of immunoncology therapies. With the rapid adoption of genome-wide sequencing in biomedical research, HLA typing based on transcriptome and whole exome/genome sequencing data becomes increasingly attractive due to its high throughput and convenience. However, unlike targeted amplicon sequencing, genome-wide sequencing often employs a reduced read length and coverage that impose great challenges in resolving the highly homologous HLA alleles. Though several algorithms exist and have been applied to four-digit typing, some deliver low to moderate accuracies, some output ambiguous predictions. Moreover, few methods suit diverse read lengths and depths, and both RNA and DNA sequencing inputs. New algorithms are therefore needed to leverage the accuracy and flexibility of HLA typing at high resolution using genome-wide sequencing data. RESULTS We have developed a new algorithm named PHLAT to discover the most probable pair of HLA alleles at four-digit resolution or higher, via a unique integration of a candidate allele selection and a likelihood scoring. Over a comprehensive set of benchmarking data (a total of 768 HLA alleles) from both RNA and DNA sequencing and with a broad range of read lengths and coverage, PHLAT consistently achieves a high accuracy at four-digit (92%-95%) and two-digit resolutions (96%-99%), outcompeting most of the existing methods. It also supports targeted amplicon sequencing data from Illumina Miseq. CONCLUSIONS PHLAT significantly leverages the accuracy and flexibility of high resolution HLA typing based on genome-wide sequencing data. It may benefit both basic and applied research in immunology and related fields as well as numerous clinical applications.
Collapse
Affiliation(s)
- Yu Bai
- Regeneron Pharmaceuticals, Inc, Tarrytown, New York USA
| | - Min Ni
- Regeneron Pharmaceuticals, Inc, Tarrytown, New York USA
| | - Blerta Cooper
- Regeneron Pharmaceuticals, Inc, Tarrytown, New York USA
| | - Yi Wei
- Regeneron Pharmaceuticals, Inc, Tarrytown, New York USA
| | - Wen Fury
- Regeneron Pharmaceuticals, Inc, Tarrytown, New York USA
| |
Collapse
|
26
|
Miyazaki Y, Shiozawa S. In vivo cell transfer assay to detect autoreactive T cell subsets. Methods Mol Biol 2014; 1142:49-53. [PMID: 24706274 DOI: 10.1007/978-1-4939-0404-4_6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/16/2023]
Abstract
Among the methods used in molecular biology, in vitro biochemical assays are more common, whereas in vivo assays, including the use of animal models, are less widely employed. In our studies on systemic lupus erythematosus (SLE), we have identified a novel T cell subtype termed "autoantibody-inducing CD4 T cells" (aiCD4 T cell) that is responsible for the development of autoimmunity. In order to identify and isolate these cells, we developed a new technique that involves the transfer of candidate T cell subpopulations into naïve mice and assaying for the development of autoantibodies in the recipient mice. We have previously described an experimental system in which mice not normally prone to autoimmune diseases can be induced to develop experimental SLE. In this experimental system, autoantibody-inducing CD4 T (aiCD4 T) cells are generated via de novo T cell receptor revision at peripheral lymphoid organs. These aiCD4 T cells not only induce a variety of autoantibodies but also promote the final differentiation of CD8 T cells into cytotoxic T lymphocytes, resulting in a pathology identical to SLE. We needed to develop a new methodology to isolate this subpopulation of aiCD4 T cells. Here we describe an in vivo assay to detect and isolate aiCD4 T cells by transferring the candidate cells into naïve recipient mice and monitoring the production of the appropriate antibody or cytokine.
Collapse
Affiliation(s)
- Yumi Miyazaki
- Department of Rheumatology, Kyushu University Beppu Hospital, 4546 Tsurumihara, Beppu, 874-0838, Japan,
| | | |
Collapse
|
27
|
Yang CY, Leung PSC, Adamopoulos IE, Gershwin ME. The implication of vitamin D and autoimmunity: a comprehensive review. Clin Rev Allergy Immunol 2014; 45:217-26. [PMID: 23359064 DOI: 10.1007/s12016-013-8361-3] [Citation(s) in RCA: 194] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Historically, vitamin D has been associated with the regulation of bone metabolism. However, increasing evidence demonstrates a strong association between vitamin D signaling and many biological processes that regulate immune responses. The discovery of the vitamin D receptor in multiple immune cell lineages, such as monocytes, dendritic cells, and activated T cells credits vitamin D with a novel role in modulating immunological functions and its subsequent role in the development or prevention of autoimmune diseases. In this review we, discuss five major areas in vitamin D biology of high immunological significance: (1) the metabolism of vitamin D; (2) the significance of vitamin D receptor polymorphisms in autoimmune diseases, such as multiple sclerosis, type 1 diabetes mellitus, and systemic lupus erythematosus; (3) vitamin D receptor transcriptional regulation of immune cell lineages, including Th1, Th17, Th2, regulatory T, and natural killer T cells; (4) the prevalence of vitamin D insufficiency/deficiency in patients with multiple sclerosis, type 1 diabetes mellitus, and systemic lupus erythematosus; and finally, (5) the therapeutic effects of vitamin D supplementation on disease severity and progression.
Collapse
Affiliation(s)
- Chen-Yen Yang
- Division of Rheumatology, Allergy and Clinical Immunology, University of California at Davis School of Medicine, 451 Health Sciences Drive, Suite 6510, Davis, CA, 95616, USA
| | | | | | | |
Collapse
|
28
|
Szymula A, Rosenthal J, Szczerba BM, Bagavant H, Fu SM, Deshmukh US. T cell epitope mimicry between Sjögren's syndrome Antigen A (SSA)/Ro60 and oral, gut, skin and vaginal bacteria. Clin Immunol 2014; 152:1-9. [PMID: 24576620 DOI: 10.1016/j.clim.2014.02.004] [Citation(s) in RCA: 108] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2014] [Accepted: 02/06/2014] [Indexed: 12/13/2022]
Abstract
This study was undertaken to test the hypothesis that Sjogren's syndrome Antigen A (SSA)/Ro60-reactive T cells are activated by peptides originating from oral and gut bacteria. T cell hybridomas generated from HLA-DR3 transgenic mice recognized 3 regions on Ro60, with core epitopes mapped to amino acids 228-238, 246-256 and 371-381. BLAST analysis identified several mimicry peptides, originating from human oral, intestinal, skin and vaginal bacteria, as well as environmental bacteria. Amongst these, a peptide from the von Willebrand factor type A domain protein (vWFA) from the oral microbe Capnocytophaga ochracea was the most potent activator. Further, Ro60-reactive T cells were activated by recombinant vWFA protein and whole Escherichia coli expressing this protein. These results demonstrate that peptides derived from normal human microbiota can activate Ro60-reactive T cells. Thus, immune responses to commensal microbiota and opportunistic pathogens should be explored as potential triggers for initiating autoimmunity in SLE and Sjögren's syndrome.
Collapse
Affiliation(s)
- Agnieszka Szymula
- Center for Immunity Inflammation and Regenerative Medicine, Division of Nephrology, Department of Medicine, University of Virginia, Charlottesville, VA, USA
| | - Jacob Rosenthal
- Center for Immunity Inflammation and Regenerative Medicine, Division of Nephrology, Department of Medicine, University of Virginia, Charlottesville, VA, USA
| | - Barbara M Szczerba
- Center for Immunity Inflammation and Regenerative Medicine, Division of Nephrology, Department of Medicine, University of Virginia, Charlottesville, VA, USA
| | - Harini Bagavant
- Center for Immunity Inflammation and Regenerative Medicine, Division of Nephrology, Department of Medicine, University of Virginia, Charlottesville, VA, USA; Department of Pharmacology, University of Virginia, Charlottesville, VA, USA
| | - Shu Man Fu
- Center for Immunity Inflammation and Regenerative Medicine, Division of Nephrology, Department of Medicine, University of Virginia, Charlottesville, VA, USA; Division of Rheumatology, Department of Medicine, University of Virginia, Charlottesville, VA, USA; Department of Microbiology, Immunology and Cancer Biology, University of Virginia, Charlottesville, VA, USA
| | - Umesh S Deshmukh
- Center for Immunity Inflammation and Regenerative Medicine, Division of Nephrology, Department of Medicine, University of Virginia, Charlottesville, VA, USA; Department of Pharmacology, University of Virginia, Charlottesville, VA, USA.
| |
Collapse
|
29
|
Zhu H, Sun X, Zhu L, Hu F, Shi L, Fan C, Li Z, Su Y. Different expression patterns and clinical significance of mAxl and sAxl in systemic lupus erythematosus. Lupus 2014; 23:624-34. [PMID: 24474706 DOI: 10.1177/0961203314520839] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2013] [Accepted: 12/23/2013] [Indexed: 11/17/2022]
Abstract
Axl is one of the TAM family members that downregulates activated immune responses to maintain immune homeostasis. We analyzed the expression and clinical relevance of Axl on the surface of CD14+ monocytes/macrophages (mAxl, membrane Axl) and in the plasma (sAxl, soluble Axl) from patients with systemic lupus erythematosus (SLE). Compared to healthy subjects, the concentrations of sAxl were significantly elevated in plasma from SLE patients, while the mAxl expression on CD14+ monocytes/macrophages from SLE patients was significantly downregulated. A series of severe disease clinical manifestations and laboratory features such as presence of autoantibodies, 24-hour proteinuria excretion or SLEDAI ≥10 were associated with decreased mAxl expression on monocytes/macrophages but elevated sAxl levels in plasma. The plasma level of Gas6, the main ligand of Axl, was slightly decreased in SLE patients, and was negatively correlated with anti-dsDNA antibodies and C-reactive protein. SLE patients with SLEDAI ≥10 showed significantly lower Gas6 levels. Our study suggests that abnormal mAxl and sAxl expression may be involved in the imbalance of immune regulation in SLE.
Collapse
Affiliation(s)
- H Zhu
- Department of Rheumatology and Immunology, Clinical Immunology Center, Peking University People’s Hospital, Beijing, China
| | - X Sun
- Department of Rheumatology and Immunology, Clinical Immunology Center, Peking University People’s Hospital, Beijing, China
| | - L Zhu
- Department of Rheumatology and Immunology, Clinical Immunology Center, Peking University People’s Hospital, Beijing, China
| | - F Hu
- Department of Rheumatology and Immunology, Clinical Immunology Center, Peking University People’s Hospital, Beijing, China
| | - L Shi
- Department of Rheumatology and Immunology, Clinical Immunology Center, Peking University People’s Hospital, Beijing, China
| | - C Fan
- Department of Laboratory Medicine, Peking University People’s Hospital, Beijing, China
| | - Z Li
- Department of Rheumatology and Immunology, Clinical Immunology Center, Peking University People’s Hospital, Beijing, China
| | - Y Su
- Department of Rheumatology and Immunology, Clinical Immunology Center, Peking University People’s Hospital, Beijing, China
| |
Collapse
|
30
|
Aguzzi A, Kranich J, Krautler NJ. Follicular dendritic cells: origin, phenotype, and function in health and disease. Trends Immunol 2013; 35:105-13. [PMID: 24315719 DOI: 10.1016/j.it.2013.11.001] [Citation(s) in RCA: 112] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2013] [Revised: 10/31/2013] [Accepted: 11/07/2013] [Indexed: 01/15/2023]
Abstract
Follicular dendritic cells (FDCs) were originally identified by their specific morphology and by their ability to trap immune-complexed antigen in B cell follicles. By virtue of the latter as well as the provision of chemokines, adhesion molecules, and trophic factors, FDCs participate in the shaping of B cell responses. Importantly, FDCs also supply tingible body macrophages (TBMs) with the eat-me-signaling molecule milk fat globule-EGF factor 8 (Mfge8), thereby enabling the disposal of apoptotic B cells. Recent studies have provided fundamental insights into the multiple functions of FDCs in both physiological and pathophysiological contexts and into their origin. Here we review these findings, and discuss current concepts related to FDC histogenesis both in lymphoid organs and in inflammatory lymphoneogenesis.
Collapse
Affiliation(s)
- Adriano Aguzzi
- Institute of Neuropathology, University of Zurich, Zurich, Switzerland.
| | - Jan Kranich
- Institute for Immunology, Ludwig Maximilians University, Munich, Germany
| | - Nike Julia Krautler
- Garvan Institute of Medical Research, Darlinghurst, New South Wales, Sydney, Australia.
| |
Collapse
|
31
|
Perricone C, Colafrancesco S, Mazor RD, Soriano A, Agmon-Levin N, Shoenfeld Y. Autoimmune/inflammatory syndrome induced by adjuvants (ASIA) 2013: Unveiling the pathogenic, clinical and diagnostic aspects. J Autoimmun 2013; 47:1-16. [PMID: 24238833 DOI: 10.1016/j.jaut.2013.10.004] [Citation(s) in RCA: 164] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2013] [Accepted: 10/21/2013] [Indexed: 12/23/2022]
Abstract
In 2011 a new syndrome termed 'ASIA Autoimmune/Inflammatory Syndrome Induced by Adjuvants' was defined pointing to summarize for the first time the spectrum of immune-mediated diseases triggered by an adjuvant stimulus such as chronic exposure to silicone, tetramethylpentadecane, pristane, aluminum and other adjuvants, as well as infectious components, that also may have an adjuvant effect. All these environmental factors have been found to induce autoimmunity by themselves both in animal models and in humans: for instance, silicone was associated with siliconosis, aluminum hydroxide with post-vaccination phenomena and macrophagic myofasciitis syndrome. Several mechanisms have been hypothesized to be involved in the onset of adjuvant-induced autoimmunity; a genetic favorable background plays a key role in the appearance on such vaccine-related diseases and also justifies the rarity of these phenomena. This paper will focus on protean facets which are part of ASIA, focusing on the roles and mechanisms of action of different adjuvants which lead to the autoimmune/inflammatory response. The data herein illustrate the critical role of environmental factors in the induction of autoimmunity. Indeed, it is the interplay of genetic susceptibility and environment that is the major player for the initiation of breach of tolerance.
Collapse
Affiliation(s)
- Carlo Perricone
- The Zabludowicz Center for Autoimmune Diseases, Sheba Medical Center, Tel-Hashomer, Israel; Reumatologia, Dipartimento di Medicina Interna e Specialità Mediche, Sapienza Università di Roma, Rome, Italy
| | | | | | | | | | | |
Collapse
|
32
|
Costimulatory pathways: physiology and potential therapeutic manipulation in systemic lupus erythematosus. Clin Dev Immunol 2013; 2013:245928. [PMID: 24000287 PMCID: PMC3755444 DOI: 10.1155/2013/245928] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2013] [Accepted: 07/08/2013] [Indexed: 02/07/2023]
Abstract
System lupus erythematosus (SLE) is an immune-complex-mediated autoimmune condition with protean immunological and clinical manifestation. While SLE has classically been advocated as a B-cell or T-cell disease, it is unlikely that a particular cell type is more pathologically predominant than the others. Indeed, SLE is characterized by an orchestrated interplay amongst different types of immunopathologically important cells participating in both innate and adaptive immunity including the dendritic cells, macrophages, neutrophils and lymphocytes, as well as traditional nonimmune cells such as endothelial, epithelial, and renal tubular cells. Amongst the antigen-presenting cells and lymphocytes, and between lymphocytes, the costimulatory pathways which involve mutual exchange of information and signalling play an essential role in initiating, perpetuating, and, eventually, attenuating the proinflammatory immune response. In this review, advances in the knowledge of established costimulatory pathways such as CD28/CTLA-4-CD80/86, ICOS-B7RP1, CD70-CD27, OX40-OX40L, and CD137-CD137L as well as their potential roles involved in the pathophysiology of SLE will be discussed. Attempts to target these costimulatory pathways therapeutically will pave more potential treatment avenues for patients with SLE. Preliminary laboratory and clinical evidence of the potential therapeutic value of manipulating these costimulatory pathways in SLE will also be discussed in this review.
Collapse
|
33
|
Selmi C, Gershwin ME. The long-term marriage between autoimmunity and internal medicine: a homage to Manuel Carlos Dias. Clin Rev Allergy Immunol 2013; 43:207-10. [PMID: 22826113 DOI: 10.1007/s12016-012-8333-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Our understanding of autoimmune diseases results from the perfect combination of basic and clinical scientific research, and the figure that is closest to the proposed autoimmunology specialist is certainly the internist. The role of B cells in rheumatoid arthritis, the immunological mechanisms to fibrosis or to tissue specific damage, the classification of Bechet's syndrome, the clinical outcomes of antiphospholid syndrome, and new biomarkers for vascular complications in systemic sclerosis constitute, among others, are ideal examples of this combination. For these reason, this issue includes comprehensive reviews in all these areas and is dedicated to Dr. Manuel Carlos Dias and his career in the perfectioning and teaching of the clinical skills necessary to manage autoimmune disease. We are convinced that these discussions are likely of interest to basic scientists and clinicians alike for the proposed translational applications.
Collapse
Affiliation(s)
- Carlo Selmi
- Division of Rheumatology, Allergy, and Clinical Immunology, University of California Davis, Davis, CA 95616, USA.
| | | |
Collapse
|
34
|
Xue J, Kass DJ, Bon J, Vuga L, Tan J, Csizmadia E, Otterbein L, Soejima M, Levesque MC, Gibson KF, Kaminski N, Pilewski JM, Donahoe M, Sciurba FC, Duncan SR. Plasma B lymphocyte stimulator and B cell differentiation in idiopathic pulmonary fibrosis patients. THE JOURNAL OF IMMUNOLOGY 2013; 191:2089-95. [PMID: 23872052 DOI: 10.4049/jimmunol.1203476] [Citation(s) in RCA: 132] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
We hypothesized B cells are involved in the pathogenesis of idiopathic pulmonary fibrosis (IPF), a progressive, restrictive lung disease that is refractory to glucocorticoids and other nonspecific therapies, and almost invariably lethal. Accordingly, we sought to identify clinically associated B cell-related abnormalities in these patients. Phenotypes of circulating B cells were characterized by flow cytometry. Intrapulmonary processes were evaluated by immunohistochemistry. Plasma B lymphocyte stimulating factor (BLyS) was assayed by ELISA. Circulating B cells of IPF subjects were more Ag differentiated, with greater plasmablast proportions (3.1 ± 0.8%) than in normal controls (1.3 ± 0.3%) (p < 0.03), and the extent of this differentiation correlated with IPF patient lung volumes (r = 0.44, p < 0.03). CD20(+) B cell aggregates, diffuse parenchymal and perivascular immune complexes, and complement depositions were all prevalent in IPF lungs, but much less prominent or absent in normal lungs. Plasma concentrations of BLyS, an obligate factor for B cell survival and differentiation, were significantly greater (p < 0.0001) in 110 IPF (2.05 ± 0.05 ng/ml) than among 53 normal (1.40 ± 0.04 ng/ml) and 90 chronic obstructive pulmonary disease subjects (1.59 ± 0.05 ng/ml). BLyS levels were uniquely correlated among IPF patients with pulmonary artery pressures (r = 0.58, p < 0.0001). The 25% of IPF subjects with the greatest BLyS values also had diminished 1-y survival (46 ± 11%), compared with those with lesser BLyS concentrations (81 ± 5%) (hazard ratio = 4.0, 95% confidence interval = 1.8-8.7, p = 0.0002). Abnormalities of B cells and BLyS are common in IPF patients, and highly associated with disease manifestations and patient outcomes. These findings have implications regarding IPF pathogenesis and illuminate the potential for novel treatment regimens that specifically target B cells in patients with this lung disease.
Collapse
Affiliation(s)
- Jianmin Xue
- Department of Medicine, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Abstract
The elevated cardiovascular morbidity in rheumatoid arthritis, systemic lupus erythematosus, and the antiphospholipid syndrome is well known, as well as the pulmonary involvement observed in these conditions and to a major extent in systemic sclerosis. These manifestations constitute a major challenge for clinicians involved in patient management. Moreover, several issues regarding the link between autoimmune rheumatic diseases and cardio pulmonary morbidity remain largely enigmatic. The mechanistic role of certain autoantibodies frequently observed in association with heart and lung diseases or the pathogenetic link between chronic inflammation and the pathways leading to atherosclerosis or pulmonary vascular changes are yet to be elucidated. As such, these questions as well as treatment strategies are of common interest to rheumatologists, immunologist, pulmonologists, and cardiologists and thus call for an interdisciplinary approach. This paradigm has been well established for rare conditions such as the Churg-Strauss syndrome. Nowadays, it seems that this approach should be expanded to encompass more common conditions such as coronary heart disease, pulmonary arterial hypertension or dilated cardiomyopathy. The present issue of Clinical Reviews in Allergy and Immunology addresses the new knowledge and concepts of autoimmune-related cardiopulmonary diseases. The issue derives from the 2010 International Autoimmunity Meeting held in Ljubljana, Slovenia and is thus timely and dedicated to the latest developments in this new multidisciplinary field.
Collapse
|
36
|
Kahloon RA, Xue J, Bhargava A, Csizmadia E, Otterbein L, Kass DJ, Bon J, Soejima M, Levesque MC, Lindell KO, Gibson KF, Kaminski N, Banga G, Oddis CV, Pilewski JM, Sciurba FC, Donahoe M, Zhang Y, Duncan SR. Patients with idiopathic pulmonary fibrosis with antibodies to heat shock protein 70 have poor prognoses. Am J Respir Crit Care Med 2013; 187:768-75. [PMID: 23262513 DOI: 10.1164/rccm.201203-0506oc] [Citation(s) in RCA: 142] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
RATIONALE Diverse autoantibodies are present in most patients with idiopathic pulmonary fibrosis (IPF). We hypothesized that specific autoantibodies may associate with IPF manifestations. OBJECTIVES To identify clinically relevant, antigen-specific immune responses in patients with IPF. METHODS Autoantibodies were detected by immunoblots and ELISA. Intrapulmonary immune processes were evaluated by immunohistochemistry. Anti-heat shock protein 70 (HSP70) IgG was isolated from plasma by immunoaffinity. Flow cytometry was used for leukocyte functional studies. MEASUREMENTS AND MAIN RESULTS HSP70 was identified as a potential IPF autoantigen in discovery assays. Anti-HSP70 IgG autoantibodies were detected by immunoblots in 3% of 60 control subjects versus 25% of a cross-sectional IPF cohort (n = 122) (P = 0.0004), one-half the patients with IPF who died (P = 0.008), and 70% of those with acute exacerbations (P = 0.0005). Anti-HSP70 autoantibodies in patients with IPF were significantly associated with HLA allele biases, greater subsequent FVC reductions (P = 0.0004), and lesser 1-year survival (40 ± 10% vs. 80 ± 5%; hazard ratio = 4.2; 95% confidence interval, 2.0-8.6; P < 0.0001). HSP70 protein, antigen-antibody complexes, and complement were prevalent in IPF lungs. HSP70 protein was an autoantigen for IPF CD4 T cells, inducing lymphocyte proliferation (P = 0.004) and IL-4 production (P = 0.01). IPF anti-HSP70 autoantibodies activated monocytes (P = 0.009) and increased monocyte IL-8 production (P = 0.049). ELISA confirmed the association between anti-HSP70 autoreactivity and IPF outcome. Anti-HSP70 autoantibodies were also found in patients with other interstitial lung diseases but were not associated with their clinical progression. CONCLUSIONS Patients with IPF with anti-HSP70 autoantibodies have more near-term lung function deterioration and mortality. These findings suggest antigen-specific immunoassays could provide useful clinical information in individual patients with IPF and may have implications for understanding IPF progression.
Collapse
Affiliation(s)
- Rehan A Kahloon
- Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Miyazaki Y, Tsumiyama K, Yamane T, Ito M, Shiozawa S. Self-Organized Criticality Theory and the Expansion of PD-1-Positive Effector CD4 T Cells: Search for Autoantibody-Inducing CD4 T Cells. Front Immunol 2013; 4:87. [PMID: 23596442 PMCID: PMC3622253 DOI: 10.3389/fimmu.2013.00087] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2013] [Accepted: 03/30/2013] [Indexed: 11/30/2022] Open
Affiliation(s)
- Yumi Miyazaki
- Department of Medicine, Kyushu University Beppu Hospital Beppu, Japan
| | | | | | | | | |
Collapse
|
38
|
Epigenetic considerations and the clinical reevaluation of the overlap syndrome between primary biliary cirrhosis and autoimmune hepatitis. J Autoimmun 2013. [DOI: 10.1016/j.jaut.2012.10.004] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|
39
|
Lewis JE, Fu SM, Gaskin F. Autoimmunity, end organ damage, and the origin of autoantibodies and autoreactive T cells in systemic lupus erythematosus. DISCOVERY MEDICINE 2013; 15:85-92. [PMID: 23449110 PMCID: PMC3725807] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Indexed: 06/01/2023]
Abstract
Systemic lupus erythematosus (SLE) is a prototype of systemic autoimmunity affecting many systems. Both antibodies and autoreactive T cells play significant roles in its pathogenesis. Experimental data and clinical observations indicate that autoimmunity and end organ damage are under separate genetic controls and that there are significant interactions between these two pathways. Experimental evidence has been obtained to support the hypothesis that autoantibodies and autoreactive T effector cells may be initiated by environmental factors through molecular mimicry and the inherent polyreactive nature of antigen receptors. A unified hypothesis has been postulated for the pathogenesis of SLE that has practical implications.
Collapse
Affiliation(s)
- Janet E Lewis
- Division of Rheumatology, Department of Medicine, University of Virginia School of Medicine, Charlottesville, Virginia 22908, USA
| | | | | |
Collapse
|
40
|
Castiello L, Mossoba M, Viterbo A, Sabatino M, Fellowes V, Foley JE, Winterton M, Halverson DC, Civini S, Jin P, Fowler DH, Stroncek DF. Differential gene expression profile of first-generation and second-generation rapamycin-resistant allogeneic T cells. Cytotherapy 2013; 15:598-609. [PMID: 23352462 DOI: 10.1016/j.jcyt.2012.12.008] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2012] [Revised: 11/14/2012] [Accepted: 12/28/2012] [Indexed: 11/18/2022]
Abstract
BACKGROUND AIMS We completed a phase II clinical trial evaluating rapamycin-resistant allogeneic T cells (T-rapa) and now have evaluated a T-rapa product manufactured in 6 days (T-rapa(6)) rather than 12 days (T-Rapa(12)). METHODS Using gene expression microarrays, we addressed our hypothesis that the two products would express a similar phenotype. The products had similar phenotypes using conventional comparison methods of cytokine secretion and surface markers. RESULTS Unsupervised analysis of 34,340 genes revealed that T-rapa(6) and T-rapa(12) products clustered together, distinct from culture input CD4(+) T cells. Statistical analysis of T-rapa(6) products revealed differential expression of 19.3% of genes (n = 6641) compared with input CD4(+) cells; similarly, 17.8% of genes (n = 6147) were differentially expressed between T-rapa(12) products and input CD4(+) cells. Compared with input CD4(+) cells, T-rapa(6) and T-rapa(12) products were similar in terms of up-regulation of major gene families (cell cycle, stress response, glucose catabolism, DNA metabolism) and down-regulation (inflammatory response, immune response, apoptosis, transcriptional regulation). However, when directly compared, T-rapa(6) and T-rapa(12) products showed differential expression of 5.8% of genes (n = 1994; T-rapa(6) vs. T-rapa(12)). CONCLUSIONS Second-generation T-rapa(6) cells possess a similar yet distinct gene expression profile relative to first-generation T-rapa(12) cells and may mediate differential effects after adoptive transfer.
Collapse
Affiliation(s)
- Luciano Castiello
- Department of Transfusion Medicine, Clinical Center, National Institutes of Health, Bethesda, MD 20892-1288, USA.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Abstract
The mechanisms leading to the onset and perpetuation of systemic and tissue-specific autoimmune diseases are complex, and numerous hypotheses have been proposed or confirmed over the past 12 months. It is particularly of note that the number of articles published during 2011 in the major immunology and autoimmunity journals increased by 3 % compared to the previous year. The present article is dedicated to a brief review of the reported data and, albeit not comprehensive of all articles, is aimed at identifying common and future themes. First, clinical researchers were particularly dedicated to defining refractory forms of diseases and to discuss the use and switch of therapeutic monoclonal antibodies in everyday practice. Second, following the plethora of genome-wide association studies reported in most multifactorial diseases, it became clear that genomics cannot fully explain the individual susceptibility and additional environmental or epigenetic factors are necessary. Both these components were widely investigated, both in organ-specific (i.e., type 1 diabetes) and systemic (i.e., systemic lupus erythematosus) diseases. Third, a large number of 2011 works published in the autoimmunity area are dedicated to dissect pathogenetic mechanisms of tolerance breakdown in general or in specific conditions. While our understanding of T regulatory and Th17 cells has significantly increased in 2011, it is of note that most of the proposed lines of evidence identify potential targets for future treatments and should not be overlooked.
Collapse
|
42
|
Achenza MIS, Meda F, Brunetta E, Selmi C. Serum autoantibodies for the diagnosis and management of autoimmune liver diseases. Expert Rev Gastroenterol Hepatol 2012; 6:717-29. [PMID: 23237257 DOI: 10.1586/egh.12.58] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The spectrum of autoimmune liver diseases (AILD) includes primary biliary cirrhosis, primary sclerosing cholangitis and autoimmune hepatitis. The immunological mechanisms triggering the initiation and perpetuation of AILD remains unknown, while autoantigens are now recognized in most cases, and are generally nontraditional in their widespread distribution. Sensitive and specific methods for the detection of serum autoantibodies in patients affected by AILD represent a challenge for researchers and clinicians who desire to obtain an early and certain diagnosis as well as markers of disease control. To this regard, the use and interpretation of serum autoantibodies in AILD may be seen as paradigmatic for the large gaps in our knowledge based on the lack of true population-based studies. The present review article will critically discuss the available evidence on the use of autoantibody findings in the diagnosis or management of autoimmune liver disease.
Collapse
Affiliation(s)
- Maria I S Achenza
- Rheumatology and Clinical Immunology, Humanitas Clinical and Research Center, Rozzano, Milan, Italy
| | | | | | | |
Collapse
|
43
|
Gatto M, Zen M, Ghirardello A, Bettio S, Bassi N, Iaccarino L, Punzi L, Doria A. Emerging and critical issues in the pathogenesis of lupus. Autoimmun Rev 2012; 12:523-36. [PMID: 23000207 DOI: 10.1016/j.autrev.2012.09.003] [Citation(s) in RCA: 96] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2012] [Accepted: 09/10/2012] [Indexed: 01/10/2023]
Abstract
Systemic lupus erythematosus (SLE) is a multisystemic, autoimmune disease, encompassing either mild or severe manifestations. SLE was originally labeled as being an immune complex-mediated disease, but further knowledge suggested its pathogenesis is motlier than that, involving complex interactions between predisposed individuals and their environment. People affected with SLE have their immune system skewed toward aberrant self-recognition usually after encountering a triggering agent. Defeats in early and late immune checkpoints contribute to tolerance breakdown and further generation and expansion of autoreactive cell-clones. B and T cells play a master role in SLE, however clues are emerging about other cell types and new light is being shed on SLE autoantibodies, since some of them display really harmful potential (pathogenic antibodies), while others are just connected with disease development (pathological antibodies) and may even be protective. Autoantibody generation is elicited by abnormal apoptosis and inefficient clearance of cellular debris causing intracellular autoantigens (e.g. nucleosomes) to persist in the extracellular environment, being further recognized by autoreactive cells. Here we explore the complexity of SLE pathogenesis through five core issues, i.e. genetic predisposition, B and T cell abnormalities, abnormal autoantigen availability, autoantibody generation and organ damage, relying on current knowledge and recent insights into SLE development.
Collapse
Affiliation(s)
- Mariele Gatto
- Division of Rheumatology, Department of Medicine, University of Padova, Via Giustiniani 2, 35128 Padova, Italy
| | | | | | | | | | | | | | | |
Collapse
|
44
|
Borchers AT, Leibushor N, Naguwa SM, Cheema GS, Shoenfeld Y, Gershwin ME. Lupus nephritis: a critical review. Autoimmun Rev 2012; 12:174-94. [PMID: 22982174 DOI: 10.1016/j.autrev.2012.08.018] [Citation(s) in RCA: 168] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/12/2012] [Indexed: 01/18/2023]
Abstract
Lupus nephritis remains one of the most severe manifestations of systemic lupus erythematosus associated with considerable morbidity and mortality. A better understanding of the pathogenesis of lupus nephritis is an important step in identifying more targeted and less toxic therapeutic approaches. Substantial research has helped define the pathogenetic mechanisms of renal manifestations and, in particular, the complex role of type I interferons is increasingly recognized; new insights have been gained into the contribution of immune complexes containing endogenous RNA and DNA in triggering the production of type I interferons by dendritic cells via activation of endosomal toll-like receptors. At the same time, there have been considerable advances in the treatment of lupus nephritis. Corticosteroids have long been the cornerstone of therapy, and the addition of cyclophosphamide has contributed to renal function preservation in patients with severe proliferative glomerulonephritis, though at the cost of serious adverse events. More recently, in an effort to minimize drug toxicity and achieve equal effectiveness, other immunosuppressive agents, including mycophenolate mofetil, have been introduced. Herein, we provide a detailed review of the trials that established the equivalency of these agents in the induction and/or maintenance therapy of lupus nephritis, culminating in the recent publication of new treatment guidelines by the American College of Rheumatology. Although newer biologics have been approved and continue to be a focus of research, they have, for the most part, been relatively disappointing compared to the effectiveness of biologics in other autoimmune diseases. Early diagnosis and treatment are essential for renal preservation.
Collapse
Affiliation(s)
- Andrea T Borchers
- Division of Rheumatology, Allergy and Clinical Immunology, University of California at Davis School of Medicine, Davis, CA 95616, United States
| | | | | | | | | | | |
Collapse
|
45
|
Current world literature. Curr Opin Rheumatol 2012; 24:586-94. [PMID: 22871955 DOI: 10.1097/bor.0b013e32835793df] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
46
|
Pierre Youinou: when intuition and determination meet autoimmunity. J Autoimmun 2012; 39:117-20. [PMID: 22863520 DOI: 10.1016/j.jaut.2012.05.004] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2012] [Accepted: 05/17/2012] [Indexed: 12/16/2022]
|
47
|
Winchester R, Wiesendanger M, Zhang HZ, Steshenko V, Peterson K, Geraldino-Pardilla L, Ruiz-Vazquez E, D'Agati V. Immunologic characteristics of intrarenal T cells: trafficking of expanded CD8+ T cell β-chain clonotypes in progressive lupus nephritis. ACTA ACUST UNITED AC 2012; 64:1589-600. [PMID: 22130908 DOI: 10.1002/art.33488] [Citation(s) in RCA: 80] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
OBJECTIVE To better define the immunologic character of the T cell infiltrate in lupus nephritis. METHODS We performed double immunohistochemical staining and clonotypic T cell receptor (TCR) β-chain sequencing in multiple anatomic regions isolated by laser-capture microdissection from renal biopsy samples. RESULTS Systemic lupus erythematosus (SLE) kidneys have a variably patterned and often extensive infiltrate of predominantly clonally expanded T cells of CD4 and CD8 lineages. CD4+ T cells were prominent in nearly two-thirds of SLE biopsy samples and were distributed as broad periglomerular aggregates or intermixed with CD8+ T cells forming periglomerular caps. Sequencing of the TCR from periglomerular regions showed a predominance of clonally expanded T cells. The CD8+ T cells, which were present in all biopsy samples, often adhered to Bowman's capsule and infiltrated the tubular epithelium. They exhibited features that suggest participation in an adaptive immune response: differentiation into CD28(null) memory-effector phenotype, trafficking of the same expanded clonotype to different regions of the kidney and to the peripheral blood, and clonal persistence for years in repeat biopsy samples. CD8+ T cell tubulitis was especially associated with progressive changes. CONCLUSION The immunologic characteristics of the infiltrating CD4+ and CD8+ T cells in the lupus kidney indicate that they have the potential to mediate injury, which may be relevant to development of progressive renal failure. Whereas the oligoclonality of the CD4+ T cell infiltrate is consistent with the paradigm of SLE as a class II major histocompatibility complex-associated autoimmune disease, the finding of CD8+ T cell clonality and trafficking implies participation in a distinct systemic adaptive immune response.
Collapse
Affiliation(s)
- Robert Winchester
- College of Physicians and Surgeons, Columbia University Medical Center, P&S Building, Room 10-432, 630 West 168th Street, New York, NY 10032, USA.
| | | | | | | | | | | | | | | |
Collapse
|
48
|
Beneficial effect of Bupleurum polysaccharides on autoimmune-prone MRL-lpr mice. Clin Dev Immunol 2012; 2012:842928. [PMID: 22701502 PMCID: PMC3372380 DOI: 10.1155/2012/842928] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2012] [Revised: 02/28/2012] [Accepted: 04/10/2012] [Indexed: 12/19/2022]
Abstract
Systemic lupus erythematosus (SLE) is a chronic systemic autoimmune disease leading to inflammatory tissue damage in multiple organs. The crude polysaccharides (BPs) isolated from the roots of Bupleurum smithii var. parvifolium have anticomplementary activity and immunomodulatory functions on macrophages. To study its potential benefit on SLE, we examined effects of BPs on MRL-lpr mice, which have similar disease features to human SLE. MRL-lpr mice were treated orally with BPs 15, 30, or 60 mg kg−1 day−1 for 12 weeks and their SLE characteristics were evaluated. The results revealed that BPs elongated life span, improved kidney function, delayed lymphadenopathy, and reduced autoantibodies. It seemed to be mediated by inhibition of complement and macrophages activation and suppression of interferon-γ (IFN-γ) and interleukin-6 (IL-6) gene expression in the kidney. These results implicate that BPs may be an immunomodulator for the treatment of autoimmune diseases like SLE.
Collapse
|
49
|
Kang HK, Chiang MY, Ecklund D, Zhang L, Ramsey-Goldman R, Datta SK. Megakaryocyte progenitors are the main APCs inducing Th17 response to lupus autoantigens and foreign antigens. THE JOURNAL OF IMMUNOLOGY 2012; 188:5970-80. [PMID: 22561152 DOI: 10.4049/jimmunol.1200452] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
In search of autoantigen-presenting cells that prime the pathogenic autoantibody-inducing Th cells of lupus, we found that CD41(+)CD151(+) cells among Lineage(-) (Lin(-)) CD117(+) (c-Kit(+)) CX3CR1(-) splenocytes depleted of known APCs were most proficient in presenting nuclear autoantigens from apoptotic cells to induce selectively an autoimmune Th17 response in different lupus-prone mouse strains. The new APCs have properties resembling megakaryocyte and/or bipotent megakaryocyte/erythroid progenitors of bone marrow, hence they are referred to as MM cells in this study. The MM cells produce requisite cytokines, but they require contact for optimal Th17 induction upon nucleosome feeding, and can induce Th17 only before undergoing differentiation to become c-Kit(-)CD41(+) cells. The MM cells expand up to 10-fold in peripheral blood of lupus patients and 49-fold in spleens of lupus mice preceding disease activity; they accelerate lupus in vivo and break tolerance in normal mice, inducing autoimmune Th17 cells. MM cells also cause Th17 skewing to foreign Ag in normal mice without Th17-polarizing culture conditions. Several molecules in MM cells are targets for blocking of autoimmunization. This study advances our understanding of lupus pathogenesis and Th17 differentiation biology by characterizing a novel category of APC.
Collapse
Affiliation(s)
- Hee-Kap Kang
- Division of Rheumatology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | | | | | | | | | | |
Collapse
|
50
|
Iron levels in polarized macrophages: regulation of immunity and autoimmunity. Autoimmun Rev 2012; 11:883-9. [PMID: 22449938 DOI: 10.1016/j.autrev.2012.03.003] [Citation(s) in RCA: 104] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2012] [Accepted: 03/08/2012] [Indexed: 02/06/2023]
Abstract
Although the hallmark of autoimmune diseases remains the generation of autoantigen-specific lynfocytic cell response, growing evidence is showing a key role for macrophages in a number of autoimmune diseases. Macrophages are characterized by phenotypical and functional heterogeneity. Different immunological signals, coming from systemic blood circulation or from microenvironment, polarize macrophages to classical (M1) or alternative (M2) phenotypes. Iron accumulation in M1 macrophages is a well known bacteriostatic mechanism and one of the mechanisms at the basis of anemia associated to chronic inflammation. Moreover, some recent data suggest that iron accumulation in macrophages can directly activate macrophages to pro-inflammatory M1 phenotype, highlighting a putative role of macrophage iron retention in the pathogenesis of chronic inflammatory and autoimmune diseases. Conversely, iron content is low in M2 macrophages, principally due to increased iron release, and increased availability of iron in the extracellular milieu supported by M2 macrophages could influence the growth rate of adjacent cell and thus play an important role in tumor growth and tissue remodeling. In this review we summarize the molecular mechanisms sustaining differential iron metabolism in polarized macrophages, discuss the relevance of this metabolic signature in chronic inflammatory and autoimmune diseases, and finally focus on potential therapeutic implications rising from a better understanding of underlying molecular mechanisms.
Collapse
|